1887

Abstract

HPV16 late L1 mRNAs encode a short central exon that is located between HPV16 3′-splice site SA3358 and HPV16 5′-splice site SD3632. While SA3358 is used to produce both HPV16 early mRNAs encoding the E6 and E7 oncogenes, and late mRNAs encoding E4, L1 and L2, SD3632 is used exclusively to produce late L1 mRNA. We have previously identified an 8-nucleotide regulatory RNA element that is required for inclusion of the exon between SA3358 and SD3632 to produce L1 mRNAs at the expense of mRNAs polyadenylated at the HPV16 early polyadenylation signal pAE. Here we show that this HPV16 8-nucleotide splicing enhancer interacts with hnRNP G. Binding of hnRNP G to this element prevents inclusion of the exon between SA3358 and SD3632 on the HPV16 late L1 mRNAs. We concluded that hnRNP G has a splicing inhibitory role and that hnRNP G can control HPV16 mRNA splicing.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001019
2018-03-01
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/99/3/328.html?itemId=/content/journal/jgv/10.1099/jgv.0.001019&mimeType=html&fmt=ahah

References

  1. Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA et al. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 1999; 189:12–19 [View Article][PubMed]
    [Google Scholar]
  2. Syrjänen S, Rautava J, Syrjänen K. HPV in Head and Neck Cancer-30 Years of History. Recent Results Cancer Res 2017; 206:3–25 [View Article][PubMed]
    [Google Scholar]
  3. Ramqvist T, Grün N, Dalianis T. Human papillomavirus and tonsillar and base of tongue cancer. Viruses 2015; 7:1332–1343 [View Article][PubMed]
    [Google Scholar]
  4. Schiffman M, Doorbar J, Wentzensen N, de Sanjosé S, Fakhry C et al. Carcinogenic human papillomavirus infection. Nat Rev Dis Primers 2016; 2:16086 [View Article][PubMed]
    [Google Scholar]
  5. Mighty KK, Laimins LA. The role of human papillomaviruses in oncogenesis. Recent Results Cancer Res 2014; 193:135–148 [View Article][PubMed]
    [Google Scholar]
  6. Chow LT, Broker TR, Steinberg BM. The natural history of human papillomavirus infections of the mucosal epithelia. APMIS 2010; 118:422–449 [View Article][PubMed]
    [Google Scholar]
  7. Johansson C, Schwartz S. Regulation of human papillomavirus gene expression by splicing and polyadenylation. Nat Rev Microbiol 2013; 11:239–251 [View Article][PubMed]
    [Google Scholar]
  8. Wu C, Kajitani N, Schwartz S. Splicing and polyadenylation of human papillomavirus type 16 mRNAs. Int J Mol Sci 2017; 18:366 [View Article][PubMed]
    [Google Scholar]
  9. Graham SV, Faizo AAA. Control of human papillomavirus gene expression by alternative splicing. Virus Res 2017; 231:83–95 [View Article][PubMed]
    [Google Scholar]
  10. Jia R, Zheng ZM. Regulation of bovine papillomavirus type 1 gene expression by RNA processing. Front Biosci 2009; 14:1270–1282 [View Article][PubMed]
    [Google Scholar]
  11. Doorbar J, Quint W, Banks L, Bravo IG, Stoler M et al. The biology and life-cycle of human papillomaviruses. Vaccine 2012; 30:F55–F70 [View Article][PubMed]
    [Google Scholar]
  12. Hong S, Laimins LA. Regulation of the life cycle of HPVs by differentiation and the DNA damage response. Future Microbiol 2013; 8:1547–1557 [View Article][PubMed]
    [Google Scholar]
  13. Stanley MA. Epithelial cell responses to infection with human papillomavirus. Clin Microbiol Rev 2012; 25:215–222 [View Article][PubMed]
    [Google Scholar]
  14. McBride AA. The papillomavirus E2 proteins. Virology 2013; 445:57–79 [View Article][PubMed]
    [Google Scholar]
  15. Johansson C, Somberg M, Li X, Backström Winquist E, Fay J et al. HPV-16 E2 contributes to induction of HPV-16 late gene expression by inhibiting early polyadenylation. EMBO J 2012; 31:3212–3227 [View Article][PubMed]
    [Google Scholar]
  16. Graham SV. Human papillomavirus E2 protein: linking replication, transcription, and RNA processing. J Virol 2016; 90:8384–8388 [View Article][PubMed]
    [Google Scholar]
  17. Pentland I, Parish JL. Targeting CTCF to control virus gene expression: a common theme amongst diverse DNA viruses. Viruses 2015; 7:3574–3585 [View Article][PubMed]
    [Google Scholar]
  18. Bernard HU. Regulatory elements in the viral genome. Virology 2013; 445:197–204 [View Article][PubMed]
    [Google Scholar]
  19. Thierry F. Transcriptional regulation of the papillomavirus oncogenes by cellular and viral transcription factors in cervical carcinoma. Virology 2009; 384:375–379 [View Article][PubMed]
    [Google Scholar]
  20. Kajitani N, Schwartz S. RNA binding proteins that control human papillomavirus gene expression. Biomolecules 2015; 5:758–774 [View Article][PubMed]
    [Google Scholar]
  21. Kajitani N, Glahder J, Wu C, Yu H, Nilsson K et al. hnRNP L controls HPV16 RNA polyadenylation and splicing in an Akt kinase-dependent manner. Nucleic Acids Res 2017; 45:9654–9678 [View Article][PubMed]
    [Google Scholar]
  22. Schwartz S. Papillomavirus transcripts and posttranscriptional regulation. Virology 2013; 445:187–196 [View Article][PubMed]
    [Google Scholar]
  23. van Doorslaer K, Tan Q, Xirasagar S, Bandaru S, Gopalan V et al. The Papillomavirus Episteme: a central resource for papillomavirus sequence data and analysis. Nucleic Acids Res 2013; 41:D571–D578 [View Article][PubMed]
    [Google Scholar]
  24. Rush M, Zhao X, Schwartz S. A splicing enhancer in the E4 coding region of human papillomavirus type 16 is required for early mRNA splicing and polyadenylation as well as inhibition of premature late gene expression. J Virol 2005; 79:12002–12015 [View Article][PubMed]
    [Google Scholar]
  25. Somberg M, Schwartz S. Multiple ASF/SF2 sites in the human papillomavirus type 16 (HPV-16) E4-coding region promote splicing to the most commonly used 3'-splice site on the HPV-16 genome. J Virol 2010; 84:8219–8230 [View Article][PubMed]
    [Google Scholar]
  26. Li X, Johansson C, Cardoso Palacios C, Mossberg A, Dhanjal S et al. Eight nucleotide substitutions inhibit splicing to HPV-16 3'-splice site SA3358 and reduce the efficiency by which HPV-16 increases the life span of primary human keratinocytes. PLoS One 2013; 8:e72776 [View Article][PubMed]
    [Google Scholar]
  27. Li X, Johansson C, Glahder J, Mossberg AK, Schwartz S. Suppression of HPV-16 late L1 5′-splice site SD3632 by binding of hnRNP D proteins and hnRNP A2/B1 to upstream AUAGUA RNA motifs. Nucleic Acids Res 2013; 41:10488–10508 [View Article][PubMed]
    [Google Scholar]
  28. Dhanjal S, Kajitani N, Glahder J, Mossberg AK, Johansson C et al. Heterogeneous nuclear ribonucleoprotein C proteins interact with the human papillomavirus type 16 (HPV16) Early 3'-untranslated region and alleviate suppression of HPV16 late L1 mRNA splicing. J Biol Chem 2015; 290:13354–13371 [View Article][PubMed]
    [Google Scholar]
  29. Collier B, Oberg D, Zhao X, Schwartz S. Specific inactivation of inhibitory sequences in the 5′ end of the human papillomavirus type 16 L1 open reading frame results in production of high levels of L1 protein in human epithelial cells. J Virol 2002; 76:2739–2752 [View Article][PubMed]
    [Google Scholar]
  30. Zhao X, Rush M, Schwartz S. Identification of an hnRNP A1-dependent splicing silencer in the human papillomavirus type 16 L1 coding region that prevents premature expression of the late L1 gene. J Virol 2004; 78:10888–10905 [View Article][PubMed]
    [Google Scholar]
  31. Cartegni L, Wang J, Zhu Z, Zhang MQ, Krainer AR. ESEfinder: A web resource to identify exonic splicing enhancers. Nucleic Acids Res 2003; 31:3568–3571 [View Article][PubMed]
    [Google Scholar]
  32. Somberg M, Li X, Johansson C, Orru B, Chang R et al. Serine/arginine-rich protein 30c activates human papillomavirus type 16 L1 mRNA expression via a bimodal mechanism. J Gen Virol 2011; 92:2411–2421 [View Article][PubMed]
    [Google Scholar]
  33. Heinrich B, Zhang Z, Raitskin O, Hiller M, Benderska N et al. Heterogeneous nuclear ribonucleoprotein G regulates splice site selection by binding to CC(A/C)-rich regions in pre-mRNA. J Biol Chem 2009; 284:14303–14315 [View Article][PubMed]
    [Google Scholar]
  34. Nasim MT, Chernova TK, Chowdhury HM, Yue BG, Eperon IC. HnRNP G and Tra2beta: opposite effects on splicing matched by antagonism in RNA binding. Hum Mol Genet 2003; 12:1337–1348 [View Article][PubMed]
    [Google Scholar]
  35. Kanhoush R, Beenders B, Perrin C, Moreau J, Bellini M et al. Novel domains in the hnRNP G/RBMX protein with distinct roles in RNA binding and targeting nascent transcripts. Nucleus 2010; 1:109–122 [View Article][PubMed]
    [Google Scholar]
  36. Moursy A, Allain FH, Cléry A. Characterization of the RNA recognition mode of hnRNP G extends its role in SMN2 splicing regulation. Nucleic Acids Res 2014; 42:6659–6672 [View Article][PubMed]
    [Google Scholar]
  37. Hofmann Y, Wirth B. hnRNP-G promotes exon 7 inclusion of survival motor neuron (SMN) via direct interaction with Htra2-beta1. Hum Mol Genet 2002; 11:2037–2049 [View Article][PubMed]
    [Google Scholar]
  38. Venables JP, Elliott DJ, Makarova OV, Makarov EM, Cooke HJ et al. RBMY, a probable human spermatogenesis factor, and other hnRNP G proteins interact with Tra2beta and affect splicing. Hum Mol Genet 2000; 9:685–694 [View Article][PubMed]
    [Google Scholar]
  39. Young PJ, Didonato CJ, Hu D, Kothary R, Androphy EJ et al. SRp30c-dependent stimulation of survival motor neuron (SMN) exon 7 inclusion is facilitated by a direct interaction with hTra2 beta 1. Hum Mol Genet 2002; 11:577–587 [View Article][PubMed]
    [Google Scholar]
  40. Cléry A, Jayne S, Benderska N, Dominguez C, Stamm S et al. Molecular basis of purine-rich RNA recognition by the human SR-like protein Tra2-β1. Nat Struct Mol Biol 2011; 18:443–450 [View Article][PubMed]
    [Google Scholar]
  41. Tsuda K, Someya T, Kuwasako K, Takahashi M, He F et al. Structural basis for the dual RNA-recognition modes of human Tra2-β RRM. Nucleic Acids Res 2011; 39:1538–1553 [View Article][PubMed]
    [Google Scholar]
  42. Hirschfeld M, Ouyang YQ, Jaeger M, Erbes T, Orlowska-Volk M et al. HNRNP G and HTRA2-BETA1 regulate estrogen receptor alpha expression with potential impact on endometrial cancer. BMC Cancer 2015; 15:86 [View Article][PubMed]
    [Google Scholar]
  43. Shin KH, Kang MK, Kim RH, Christensen R, Park NH. Heterogeneous nuclear ribonucleoprotein G shows tumor suppressive effect against oral squamous cell carcinoma cells. Clin Cancer Res 2006; 12:3222–3228 [View Article][PubMed]
    [Google Scholar]
  44. Shin KH, Kim RH, Kang MK, Kim RH, Kim SG et al. p53 promotes the fidelity of DNA end-joining activity by, in part, enhancing the expression of heterogeneous nuclear ribonucleoprotein G. DNA Repair 2007; 6:830–840 [View Article][PubMed]
    [Google Scholar]
  45. Shin KH, Kim RH, Yu B, Kang MK, Elashoff D et al. Expression and mutation analysis of heterogeneous nuclear ribonucleoprotein G in human oral cancer. Oral Oncol 2011; 47:1011–1016 [View Article][PubMed]
    [Google Scholar]
  46. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P et al. Proteomics. Tissue-based map of the human proteome. Science 2015; 347:1260419 [View Article][PubMed]
    [Google Scholar]
  47. Adamson B, Smogorzewska A, Sigoillot FD, King RW, Elledge SJ. A genome-wide homologous recombination screen identifies the RNA-binding protein RBMX as a component of the DNA-damage response. Nat Cell Biol 2012; 14:318–328 [View Article][PubMed]
    [Google Scholar]
  48. Dignam JD, Lebovitz RM, Roeder RG. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res 1983; 11:1475–1489 [View Article][PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001019
Loading
/content/journal/jgv/10.1099/jgv.0.001019
Loading

Data & Media loading...

Supplements

Supplementary File 1

PDF
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