1887

Abstract

The herpes simplex virus 1 (HSV1) virion host shutoff (vhs) protein is an endoribonuclease that regulates the translational environment of the infected cell, by inducing the degradation of host mRNA via cellular exonuclease activity. To further understand the relationship between translational shutoff and mRNA decay, we have used ectopic expression to compare HSV1 vhs (vhsH) to its homologues from four other alphaherpesviruses – varicella zoster virus (vhsV), bovine herpesvirus 1 (vhsB), equine herpesvirus 1 (vhsE) and Marek’s disease virus (vhsM). Only vhsH, vhsB and vhsE induced degradation of a reporter luciferase mRNA, with poly(A)+ hybridization indicating a global depletion of cytoplasmic poly(A)+ RNA and a concomitant increase in nuclear poly(A)+ RNA and the polyA tail binding protein PABPC1 in cells expressing these variants. By contrast, vhsV and vhsM failed to induce reporter mRNA decay and poly(A)+ depletion, but rather, induced cytoplasmic G3BP1 and poly(A)+ mRNA- containing granules and phosphorylation of the stress response proteins eIF2α and protein kinase R. Intriguingly, regardless of their apparent endoribonuclease activity, all vhs homologues induced an equivalent general blockade to translation as measured by single-cell puromycin incorporation. Taken together, these data suggest that the activities of translational arrest and mRNA decay induced by vhs are separable and we propose that they represent sequential steps of the vhs host interaction pathway.

Funding
This study was supported by the:
  • MRC (Award MR/T001038/1)
    • Principle Award Recipient: GillianElliott
  • BBSRC (Award BB/T007923/1)
    • Principle Award Recipient: GillianElliott
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001976
2024-04-04
2024-04-30
Loading full text...

Full text loading...

/deliver/fulltext/jgv/105/4/jgv001976.html?itemId=/content/journal/jgv/10.1099/jgv.0.001976&mimeType=html&fmt=ahah

References

  1. Gaucherand L, Porter BK, Levene RE, Price EL, Schmaling SK et al. The influenza A virus endoribonuclease PA-X usurps host mRNA processing machinery to limit host gene expression. Cell Rep 2019; 27:776–792 [View Article] [PubMed]
    [Google Scholar]
  2. Jagger BW, Wise HM, Kash JC, Walters KA, Wills NM et al. An overlapping protein-coding region in influenza A virus segment 3 modulates the host response. Science 2012; 337:199–204 [View Article] [PubMed]
    [Google Scholar]
  3. Glaunsinger B, Ganem D. Lytic KSHV infection inhibits host gene expression by accelerating global mRNA turnover. Mol Cell 2004; 13:713–723 [View Article] [PubMed]
    [Google Scholar]
  4. Read GS. Virus-encoded endonucleases: expected and novel functions. Wiley Interdisciplinary Reviews RNA 2013; 4693–708 [View Article] [PubMed]
    [Google Scholar]
  5. Bhardwaj K, Guarino L, Kao CC. The severe acute respiratory syndrome coronavirus Nsp15 protein is an endoribonuclease that prefers manganese as a cofactor. J Virol 2004; 78:12218–12224 [View Article] [PubMed]
    [Google Scholar]
  6. Covarrubias S, Richner JM, Clyde K, Lee YJ, Glaunsinger BA. Host shutoff is a conserved phenotype of gammaherpesvirus infection and is orchestrated exclusively from the cytoplasm. J Virol 2009; 83:9554–9566 [View Article] [PubMed]
    [Google Scholar]
  7. Read GS, Frenkel N. Herpes simplex virus mutants defective in the virion-associated shutoff of host polypeptide synthesis and exhibiting abnormal synthesis of alpha (immediate early) viral polypeptides. J Virol 1983; 46:498–512 [View Article] [PubMed]
    [Google Scholar]
  8. Kwong AD, Frenkel N. The herpes simplex virus virion host shutoff function. J Virol 1989; 63:4834–4839 [View Article] [PubMed]
    [Google Scholar]
  9. Read GS, Karr BM, Knight K. Isolation of a herpes simplex virus type 1 mutant with a deletion in the virion host shutoff gene and identification of multiple forms of the vhs (UL41) polypeptide. J Virol 1993; 67:7149–7160 [View Article] [PubMed]
    [Google Scholar]
  10. Matis J, Kúdelová M. Early shutoff of host protein synthesis in cells infected with herpes simplex viruses. Acta Virol 2001; 45:269–277 [PubMed]
    [Google Scholar]
  11. Jones FE, Smibert CA, Smiley JR. Mutational analysis of the herpes simplex virus virion host shutoff protein: evidence that vhs functions in the absence of other viral proteins. J Virol 1995; 69:4863–4871 [View Article] [PubMed]
    [Google Scholar]
  12. Everly DN, Read GS. Mutational analysis of the virion host shutoff gene (UL41) of herpes simplex virus (HSV): characterization of HSV type 1 (HSV-1)/HSV-2 chimeras. J Virol 1997; 71:7157–7166 [View Article] [PubMed]
    [Google Scholar]
  13. Elliott G, Pheasant K, Ebert-Keel K, Stylianou J, Franklyn A et al. Multiple posttranscriptional strategies to regulate the herpes simplex virus 1 vhs endoribonuclease. J Virol 2018; 92:e00818-18 [View Article] [PubMed]
    [Google Scholar]
  14. Strand SS, Leib DA. Role of the VP16-binding domain of vhs in viral growth, host shutoff activity, and pathogenesis. J Virol 2004; 78:13562–13572 [View Article] [PubMed]
    [Google Scholar]
  15. Oroskar AA, Read GS. Control of mRNA stability by the virion host shutoff function of herpes simplex virus. J Virol 1989; 63:1897–1906 [View Article] [PubMed]
    [Google Scholar]
  16. Elgadi MM, Hayes CE, Smiley JR. The herpes simplex virus vhs protein induces endoribonucleolytic cleavage of target RNAs in cell extracts. J Virol 1999; 73:7153–7164 [View Article] [PubMed]
    [Google Scholar]
  17. Taddeo B, Roizman B. The virion host shutoff protein (UL41) of herpes simplex virus 1 is an endoribonuclease with a substrate specificity similar to that of RNase A. J Virol 2006; 80:9341–9345 [View Article] [PubMed]
    [Google Scholar]
  18. Everly DN, Feng P, Mian IS, Read GS. mRNA degradation by the virion host shutoff (Vhs) protein of herpes simplex virus: genetic and biochemical evidence that Vhs is a nuclease. J Virol 2002; 76:8560–8571 [View Article] [PubMed]
    [Google Scholar]
  19. Feng P, Everly DN, Read GS. mRNA decay during herpes simplex virus (HSV) infections: protein-protein interactions involving the HSV virion host shutoff protein and translation factors eIF4H and eIF4A. J Virol 2005; 79:9651–9664 [View Article] [PubMed]
    [Google Scholar]
  20. Page HG, Read GS. The virion host shutoff endonuclease (UL41) of herpes simplex virus interacts with the cellular cap-binding complex eIF4F. J Virol 2010; 84:6886–6890 [View Article] [PubMed]
    [Google Scholar]
  21. Doepker RC, Hsu WL, Saffran HA, Smiley JR. Herpes simplex virus virion host shutoff protein is stimulated by translation initiation factors eIF4B and eIF4H. J Virol 2004; 78:4684–4699 [View Article] [PubMed]
    [Google Scholar]
  22. Covarrubias S, Gaglia MM, Kumar GR, Wong W, Jackson AO et al. Coordinated destruction of cellular messages in translation complexes by the gammaherpesvirus host shutoff factor and the mammalian exonuclease Xrn1. PLoS Pathog 2011; 7:e1002339 [View Article] [PubMed]
    [Google Scholar]
  23. Kumar GR, Glaunsinger BA. Nuclear import of cytoplasmic poly(A) binding protein restricts gene expression via hyperadenylation and nuclear retention of mRNA. Mol Cell Biol 2010; 30:4996–5008 [View Article] [PubMed]
    [Google Scholar]
  24. Pheasant K, Möller-Levet CS, Jones J, Depledge D, Breuer J et al. Nuclear-cytoplasmic compartmentalization of the herpes simplex virus 1 infected cell transcriptome is co-ordinated by the viral endoribonuclease vhs and cofactors to facilitate the translation of late proteins. PLoS Pathog 2018; 14:e1007331 [View Article] [PubMed]
    [Google Scholar]
  25. Afonina E, Stauber R, Pavlakis GN. The human poly(A)-binding protein 1 shuttles between the nucleus and the cytoplasm. J Biol Chem 1998; 273:13015–13021 [View Article] [PubMed]
    [Google Scholar]
  26. Burgess HM, Richardson WA, Anderson RC, Salaun C, Graham SV et al. Nuclear relocalisation of cytoplasmic poly(A)-binding proteins PABP1 and PABP4 in response to UV irradiation reveals mRNA-dependent export of metazoan PABPs. J Cell Sci 2011; 124:3344–3355 [View Article] [PubMed]
    [Google Scholar]
  27. Gilbertson S, Federspiel JD, Hartenian E, Cristea IM, Glaunsinger B. Changes in mRNA abundance drive shuttling of RNA binding proteins, linking cytoplasmic RNA degradation to transcription. Elife 2018; 7:e37663 [View Article] [PubMed]
    [Google Scholar]
  28. Dauber B, Saffran HA, Smiley JR. The herpes simplex virus host shutoff (vhs) RNase limits accumulation of double stranded RNA in infected cells: Evidence for accelerated decay of duplex RNA. PLoS Pathog 2019; 15:e1008111 [View Article] [PubMed]
    [Google Scholar]
  29. Dauber B, Pelletier J, Smiley JR. The herpes simplex virus 1 vhs protein enhances translation of viral true late mRNAs and virus production in a cell type-dependent manner. J Virol 2011; 85:5363–5373 [View Article] [PubMed]
    [Google Scholar]
  30. Esclatine A, Taddeo B, Roizman B. Herpes simplex virus 1 induces cytoplasmic accumulation of TIA-1/TIAR and both synthesis and cytoplasmic accumulation of tristetraprolin, two cellular proteins that bind and destabilize AU-rich RNAs. J Virol 2004; 78:8582–8592 [View Article] [PubMed]
    [Google Scholar]
  31. Anderson P, Kedersha N. RNA granules. J Cell Biol 2006; 172:803–808 [View Article] [PubMed]
    [Google Scholar]
  32. Berthomme H, Jacquemont B, Epstein A. The pseudorabies virus host-shutoff homolog gene: nucleotide sequence and comparison with alphaherpesvirus protein counterparts. Virology 1993; 193:1028–1032 [View Article] [PubMed]
    [Google Scholar]
  33. Everly DN, Read GS. Site-directed mutagenesis of the virion host shutoff gene (UL41) of herpes simplex virus (HSV): analysis of functional differences between HSV type 1 (HSV-1) and HSV-2 alleles. J Virol 1999; 73:9117–9129 [View Article] [PubMed]
    [Google Scholar]
  34. Lin HW, Chang YY, Wong ML, Lin JW, Chang TJ. Functional analysis of virion host shutoff protein of pseudorabies virus. Virology 2004; 324:412–418 [View Article] [PubMed]
    [Google Scholar]
  35. Ma W, Wang H, He H. Bovine herpesvirus 1 tegument protein UL41 suppresses antiviral innate immune response via directly targeting STAT1. Vet Microbiol 2019; 239:108494 [View Article] [PubMed]
    [Google Scholar]
  36. Hinkley S, Ambagala AP, Jones CJ, Srikumaran S. A vhs-like activity of bovine herpesvirus-1. Arch Virol 2000; 145:2027–2046 [View Article] [PubMed]
    [Google Scholar]
  37. Dai H, Wu J, Yang H, Guo Y, Di H et al. Construction of BHV-1 UL41 defective virus using the CRISPR/Cas9 system and analysis of viral replication properties. Front Cell Infect Microbiol 2022; 12:942987 [View Article] [PubMed]
    [Google Scholar]
  38. Lin HW, Hsu WL, Chang YY, Jan MS, Wong ML et al. Role of the UL41 protein of pseudorabies virus in host shutoff, pathogenesis and induction of TNF-α expression. J Vet Med Sci 2010; 72:1179–1187 [View Article] [PubMed]
    [Google Scholar]
  39. Gimeno I, Silva RF. Deletion of the Marek’s disease virus UL41 gene (vhs) has no measurable effect on latency or pathogenesis. Virus Genes 2008; 36:499–507 [View Article] [PubMed]
    [Google Scholar]
  40. Mao W, Niikura M, Silva RF, Cheng HH. Quantitative evaluation of viral fitness due to a single nucleotide polymorphism in the Marek’s disease virus UL41 gene via an in vitro competition assay. J Virol Methods 2008; 148:125–131 [View Article] [PubMed]
    [Google Scholar]
  41. Feng X, Thompson YG, Lewis JB, Caughman GB. Expression and function of the equine herpesvirus 1 virion-associated host shutoff homolog. J Virol 1996; 70:8710–8718 [View Article] [PubMed]
    [Google Scholar]
  42. Sato H, Callanan LD, Pesnicak L, Krogmann T, Cohen JI. Varicella-zoster virus (VZV) ORF17 protein induces RNA cleavage and is critical for replication of VZV at 37 degrees C but not 33 degrees C. J Virol 2002; 76:11012–11023 [View Article] [PubMed]
    [Google Scholar]
  43. Desloges N, Rahaus M, Wolff MH. The varicella-zoster virus-mediated delayed host shutoff: open reading frame 17 has no major function, whereas immediate-early 63 protein represses heterologous gene expression. Microbes Infect 2005; 7:1519–1529 [View Article] [PubMed]
    [Google Scholar]
  44. Wise EL, Samolej J, Elliott G. Herpes simplex virus 1 expressing GFP-tagged virion host shutoff (vhs) protein uncouples the activities of RNA degradation and differential nuclear retention of the virus transcriptome. J Virol 2022; 96:e0192621 [View Article] [PubMed]
    [Google Scholar]
  45. Taddeo B, Sciortino MT, Zhang W, Roizman B. 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 2007; 104:12163–12168 [View Article] [PubMed]
    [Google Scholar]
  46. Burgess HM, Gray NK. An integrated model for the nucleo-cytoplasmic transport of cytoplasmic poly(A)-binding proteins. Commun Integr Biol 2012; 5:243–247 [View Article] [PubMed]
    [Google Scholar]
  47. Kedersha N, Stoecklin G, Ayodele M, Yacono P, Lykke-Andersen J et al. Stress granules and processing bodies are dynamically linked sites of mRNP remodeling. J Cell Biol 2005; 169:871–884 [View Article] [PubMed]
    [Google Scholar]
  48. Lam Q, Smibert CA, Koop KE, Lavery C, Capone JP et al. Herpes simplex virus VP16 rescues viral mRNA from destruction by the virion host shutoff function. EMBO J 1996; 15:2575–2581 [PubMed]
    [Google Scholar]
  49. Finnen RL, Hay TJM, Dauber B, Smiley JR, Banfield BW. The herpes simplex virus 2 virion-associated ribonuclease vhs interferes with stress granule formation. J Virol 2014; 88:12727–12739 [View Article] [PubMed]
    [Google Scholar]
  50. Dauber B, Poon D, Dos Santos T, Duguay BA, Mehta N et al. The herpes simplex virus virion host shutoff protein enhances translation of viral true late mRNAs independently of suppressing protein kinase R and stress granule formation. J Virol 2016; 90:6049–6057 [View Article] [PubMed]
    [Google Scholar]
  51. Burgess HM, Mohr I. Defining the role of stress granules in innate immune suppression by the herpes simplex virus 1 endoribonuclease VHS. J Virol 2018; 92:e00829-18 [View Article] [PubMed]
    [Google Scholar]
  52. Reineke LC, Kedersha N, Langereis MA, van Kuppeveld FJM, Lloyd RE. Stress granules regulate double-stranded RNA-dependent protein kinase activation through a complex containing G3BP1 and Caprin1. mBio 2015; 6:e02486 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001976
Loading
/content/journal/jgv/10.1099/jgv.0.001976
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error