Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Viral hijacking of the TENT4–ZCCHC14 complex protects viral RNAs via mixed tailing

Abstract

TENT4 enzymes generate ‘mixed tails’ of diverse nucleotides at 3′ ends of RNAs via nontemplated nucleotide addition to protect messenger RNAs from deadenylation. Here we discover extensive mixed tailing in transcripts of hepatitis B virus (HBV) and human cytomegalovirus (HCMV), generated via a similar mechanism exploiting the TENT4–ZCCHC14 complex. TAIL-seq on HBV and HCMV RNAs revealed that TENT4A and TENT4B are responsible for mixed tailing and protection of viral poly(A) tails. We find that the HBV post-transcriptional regulatory element (PRE), specifically the CNGGN-type pentaloop, is critical for TENT4-dependent regulation. HCMV uses a similar pentaloop, an interesting example of convergent evolution. This pentaloop is recognized by the sterile alpha motif domain–containing ZCCHC14 protein, which in turn recruits TENT4. Overall, our study reveals the mechanism of action of PRE, which has been widely used to enhance gene expression, and identifies the TENT4–ZCCHC14 complex as a potential target for antiviral therapeutics.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Extensive mixed tailing of viral RNAs.
Fig. 2: HBV RNAs are the major mRNA substrates of TENT4 via the PRE of HBV.
Fig. 3: Stem-loop structure of PRE is necessary but may not be sufficient for TENT4-dependent RNA tailing of HBV mRNA.
Fig. 4: Stem-loop structure of HCMV RNA2.7 is the cis-acting RNA element responsible for TENT4-dependent regulation.
Fig. 5: SAM domain–containing proteins, including ZCCHC14, bind to the stem-loop structure and regulate HBV mRNAs.
Fig. 6: Cytoplasmic ZCCHC14 recruits TENT4 to protect RNAs with the stem-loop structure.

Similar content being viewed by others

Data availability

TAIL-seq and fCLIP-seq data are available at the GEO with accession number GSE146602. The mass spectrometry proteomics data are available via ProteomeXchange with identifier PXD018061. Source data for Figs. 1 and 36 and Extended Data Figs. 16 are available online.

References

  1. Schmidt, K. & Butler, J. S. Nuclear RNA surveillance: role of TRAMP in controlling exosome specificity. Wiley Interdiscip. Rev. RNA 4, 217–231 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  2. Boele, J. et al. PAPD5-mediated 3′ adenylation and subsequent degradation of miR-21 is disrupted in proliferative disease. Proc. Natl Acad. Sci. USA 111, 11467–11472 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chang, H., Lim, J., Ha, M. & Kim, V. N. TAIL-seq: genome-wide determination of poly(A) tail length and 3′ end modifications. Mol. Cell 53, 1044–1052 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Lim, J. et al. Uridylation by TUT4 and TUT7 marks mRNA for degradation. Cell 159, 1365–1376 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Shukla, S., Schmidt, J. C., Goldfarb, K. C., Cech, T. R. & Parker, R. Inhibition of telomerase RNA decay rescues telomerase deficiency caused by dyskerin or PARN defects. Nat. Struct. Mol. Biol. 23, 286–292 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Lim, J. et al. Mixed tailing by TENT4A and TENT4B shields mRNA from rapid deadenylation. Science 361, 701–704 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Son, A., Park, J. E. & Kim, V. N. PARN and TOE1 constitute a 3′ end maturation module for nuclear non-coding RNAs. Cell Rep. 23, 888–898 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Warkocki, Z., Liudkovska, V., Gewartowska, O., Mroczek, S. & Dziembowski, A. Terminal nucleotidyl transferases (TENTs) in mammalian RNA metabolism. Philos. Trans. R. Soc. Lond. B Biol. Sci. 373, 20180162 (2018).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Chang, H. et al. Terminal uridylyltransferases execute programmed clearance of maternal transcriptome in vertebrate embryos. Mol. Cell 70, 72–82.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Yeo, J. & Kim, V. N. U-tail as a guardian against invading RNAs. Nat. Struct. Mol. Biol. 25, 903–905 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Le Pen, J. et al. Terminal uridylyltransferases target RNA viruses as part of the innate immune system. Nat. Struct. Mol. Biol. 25, 778–786 (2018).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Batra, R. et al. RNA-binding protein CPEB1 remodels host and viral RNA landscapes. Nat. Struct. Mol. Biol. 23, 1101–1110 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Mueller, H. et al. A novel orally available small molecule that inhibits hepatitis B virus expression. J. Hepatol. 68, 412–420 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Mueller, H. et al. PAPD5/7 are host factors that are required for hepatitis B virus RNA stabilization. Hepatology 69, 1398–1411 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Sells, M. A., Chen, M. L. & Acs, G. Production of hepatitis B virus particles in Hep G2 cells transfected with cloned hepatitis B virus DNA. Proc. Natl Acad. Sci. USA 84, 1005–1009 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kim, B. & Kim, V. N. fCLIP-seq for transcriptomic footprinting of dsRNA-binding proteins: lessons from DROSHA. Methods 152, 3–11 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Ricci, E. P. et al. Staufen1 senses overall transcript secondary structure to regulate translation. Nat. Struct. Mol. Biol. 21, 26–35 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Lamontagne, R. J., Bagga, S. & Bouchard, M. J. Hepatitis B virus molecular biology and pathogenesis. Hepatoma Res. 2, 163–186 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Huang, J. & Liang, T. J. A novel hepatitis B virus (HBV) genetic element with Rev response element-like properties that is essential for expression of HBV gene products. Mol. Cell Biol. 13, 7476–7486 (1993).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Huang, Z. M. & Yen, T. S. Hepatitis B virus RNA element that facilitates accumulation of surface gene transcripts in the cytoplasm. J. Virol. 68, 3193–3199 (1994).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Donello, J. E., Beeche, A. A., Smith, G. J. III, Lucero, G. R. & Hope, T. J. The hepatitis B virus posttranscriptional regulatory element is composed of two subelements. J. Virol. 70, 4345–4351 (1996).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Donello, J. E., Loeb, J. E. & Hope, T. J. Woodchuck hepatitis virus contains a tripartite posttranscriptional regulatory element. J. Virol. 72, 5085–5092 (1998).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Zufferey, R., Donello, J. E., Trono, D. & Hope, T. J. Woodchuck hepatitis virus posttranscriptional regulatory element enhances expression of transgenes delivered by retroviral vectors. J. Virol. 73, 2886–2892 (1999).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Huang, Z. M., Zang, W. Q. & Yen, T. S. Cellular proteins that bind to the hepatitis B virus posttranscriptional regulatory element. Virology 217, 573–581 (1996).

    Article  CAS  PubMed  Google Scholar 

  25. Zang, W. Q., Fieno, A. M., Grant, R. A. & Yen, T. S. Identification of glyceraldehyde-3-phosphate dehydrogenase as a cellular protein that binds to the hepatitis B virus posttranscriptional regulatory element. Virology 248, 46–52 (1998).

    Article  CAS  PubMed  Google Scholar 

  26. Heise, T., Guidotti, L. G. & Chisari, F. V. La autoantigen specifically recognizes a predicted stem-loop in hepatitis B virus RNA. J. Virol. 73, 5767–5776 (1999).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Zang, W. Q., Li, B., Huang, P. Y., Lai, M. M. & Yen, T. S. Role of polypyrimidine tract binding protein in the function of the hepatitis B virus posttranscriptional regulatory element. J. Virol. 75, 10779–10786 (2001).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Ehlers, I. et al. Functional characterization of the interaction between human La and hepatitis B virus RNA. J. Biol. Chem. 279, 43437–43447 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Chi, B. et al. A sub-element in PRE enhances nuclear export of intronless mRNAs by recruiting the TREX complex via ZC3H18. Nucleic Acids Res. 42, 7305–7318 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Zhou, T. et al. HBsAg mRNA degradation induced by a dihydroquinolizinone compound depends on the HBV posttranscriptional regulatory element. Antiviral Res. 149, 191–201 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Schwalbe, M. et al. Solution structure of stem-loop alpha of the hepatitis B virus post-transcriptional regulatory element. Nucleic Acids Res. 36, 1681–1689 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Green, J. B., Gardner, C. D., Wharton, R. P. & Aggarwal, A. K. RNA recognition via the SAM domain of smaug. Mol. Cell 11, 1537–1548 (2003).

    Article  CAS  PubMed  Google Scholar 

  33. Aviv, T. et al. The RNA-binding SAM domain of Smaug defines a new family of post-transcriptional regulators. Nat. Struct. Biol. 10, 614–621 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Aviv, T., Lin, Z., Ben-Ari, G., Smibert, C. A. & Sicheri, F. Sequence-specific recognition of RNA hairpins by the SAM domain of Vts1p. Nat. Struct. Mol. Biol. 13, 168–176 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Oberstrass, F. C. et al. Shape-specific recognition in the structure of the Vts1p SAM domain with RNA. Nat. Struct. Mol. Biol. 13, 160–167 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Johnson, P. E. & Donaldson, L. W. RNA recognition by the Vts1p SAM domain. Nat. Struct. Mol. Biol. 13, 177–178 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Youn, J. Y. et al. High-density proximity mapping reveals the subcellular organization of mRNA-associated granules and bodies. Mol. Cell 69, 517–532.e11 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Hyrina, A. et al. A genome-wide CRISPR screen identifies ZCCHC14 as a host factor required for hepatitis B surface antigen production. Cell Rep. 29, 2970–2978.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. Berndt, H. et al. Maturation of mammalian H/ACA box snoRNAs: PAPD5-dependent adenylation and PARN-dependent trimming. RNA 18, 958–972 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Schmid, M. et al. Rrp6p controls mRNA poly(A) tail length and its decoration with poly(A) binding proteins. Mol. Cell 47, 267–280 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Sudo, H., Nozaki, A., Uno, H., Ishida, Y. & Nagahama, M. Interaction properties of human TRAMP-like proteins and their role in pre-rRNA 5′ ETS turnover. FEBS Lett. 590, 2963–2972 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Boyraz, B. et al. Posttranscriptional manipulation of TERC reverses molecular hallmarks of telomere disease. J. Clin. Invest. 126, 3377–3382 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  43. Kilchert, C., Wittmann, S. & Vasiljeva, L. The regulation and functions of the nuclear RNA exosome complex. Nat. Rev. Mol. Cell Biol. 17, 227–239 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Yuen, M. F. et al. Hepatitis B virus infection. Nat. Rev. Dis. Primers 4, 18035 (2018).

    Article  PubMed  Google Scholar 

  45. Sunbul, M. Hepatitis B virus genotypes: global distribution and clinical importance. World J. Gastroenterol. 20, 5427–5434 (2014).

    Article  PubMed Central  PubMed  Google Scholar 

  46. Smibert, C. A., Wilson, J. E., Kerr, K. & Macdonald, P. M. smaug protein represses translation of unlocalized nanos mRNA in the Drosophila embryo. Genes Dev. 10, 2600–2609 (1996).

    Article  CAS  PubMed  Google Scholar 

  47. Zaessinger, S., Busseau, I. & Simonelig, M. Oskar allows nanos mRNA translation in Drosophila embryos by preventing its deadenylation by Smaug/CCR4. Development 133, 4573–4583 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Amadei, G. et al. A Smaug2-based translational repression complex determines the balance between precursor maintenance versus differentiation during mammalian neurogenesis. J. Neurosci. 35, 15666–15681 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Baez, M. V. & Boccaccio, G. L. Mammalian Smaug is a translational repressor that forms cytoplasmic foci similar to stress granules. J. Biol. Chem. 280, 43131–43140 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Kim, B., Jeong, K. & Kim, V. N. Genome-wide mapping of DROSHA cleavage sites on primary microRNAs and noncanonical substrates. Mol. Cell 66, 258–269.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Zhang, J., Kobert, K., Flouri, T. & Stamatakis, A. PEAR: a fast and accurate Illumina Paired-End reAd mergeR. Bioinformatics 30, 614–620 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Uren, P. J. et al. Site identification in high-throughput RNA–protein interaction data. Bioinformatics 28, 3013–3020 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Hodges, J. L. & Lehmann, E. L. Estimates of location based on rank-tests. Ann. Math. Stats 34, 598–611 (1963).

    Article  Google Scholar 

  56. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Bazzini, A. A., Lee, M. T. & Giraldez, A. J. Ribosome profiling shows that miR-430 reduces translation before causing mRNA decay in zebrafish. Science 336, 233–237 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. The UniProt Consortium. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 45, D158–D169 (2017).

  59. Edgar, R. C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797 (2004).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  60. Anisimova, M. & Gascuel, O. Approximate likelihood-ratio test for branches: a fast, accurate, and powerful alternative. Syst. Biol. 55, 539–552 (2006).

    Article  PubMed  Google Scholar 

  61. Chevenet, F., Brun, C., Banuls, A. L., Jacq, B. & Christen, R. TreeDyn: towards dynamic graphics and annotations for analyses of trees. BMC Bioinforma. 7, 439 (2006).

    Article  CAS  Google Scholar 

  62. Dereeper, A. et al. Phylogeny.fr: robust phylogenetic analysis for the non-specialist. Nucleic Acids Res. 36, W465–W469 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Wisniewski, J. R., Zougman, A., Nagaraj, N. & Mann, M. Universal sample preparation method for proteome analysis. Nat. Methods 6, 359–362 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

  65. Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).

    Article  CAS  PubMed  Google Scholar 

  66. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of our laboratory for discussion and technical help, especially B. Kim for fCLIP-seq experiment, S. Lee for RNA pulldown, S.-C. Kwon for antibody generation and technical advice and E. Kim for technical assistance. We also thank J. Kim and Y. Choi for support with the mass spectrometry, E. Ko and H.-W. Seo for their advice about handling HepG2.2.15, and T. Shenk (Princeton University) for providing HCMV Toledo BAC DNAs. This work was supported by grant no. IBS-R008-D1 from the Institute for Basic Science from the Ministry of Science, ICT and Future Planning of Korea (D.K., Y.-s.L., S.-J.J., J.Y., J.J.S., Y.-Y.L., J.L., H.C., J.S., J.h.Y., J.-S.K., K.A., and V.N.K.), BK21 Research Fellowships from the Ministry of Education of Korea (D.K., J.J.S., Y.-Y.L., and J.S.) and the NRF-2018-Global PhD Fellowship Program from the National Research Foundation of Korea (Y.-Y.L.).

Author information

Authors and Affiliations

Authors

Contributions

D.K., Y.-s.L., S.-J.J., J.Y. and V.N.K. designed the experiments. D.K., S.-J.J., J.Y., J.J.S., Y.-Y.L. and J.L. performed the biochemical and cell biological experiments. Y.-s.L. carried out computational analyses. H.C. supported TAIL-seq analysis. J.S. and K.A. provided HCMV-infected primary HFF cells. J.h.Y. made the TENT4A and TENT4B knockout cells and laboratory-made TENT4A and TENT4B antibodies. J.-S.K. helped with the LC–MS/MS experiment. G.J. provided the HepG2.2.15 cell line as well as valuable comments and facilities. D.K., Y.-s.L., S.-J.J., J.Y. and V.N.K. wrote the manuscript.

Corresponding author

Correspondence to V. Narry Kim.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Extensive 3′ end tail modification of viral RNAs.

a, HBV mRNAs are highly mixed tailed (G:guanylation, U:uridylation, C:cytidylation) in HepG2.2.15 cells. Fraction of each modification includes their respective terminal and internal modifications. Fraction of cytidylated and uridylated tails of cellular and viral mRNAs were calculated for those with poly(A) tail length ≥25 nt (n = 2 TAIL-seq experiments). b, HCMV RNAs are also highly mixed tailed in HCMV-infected HFF cells. HCMV RNA2.7 (VRNA2.7) is substantially mixed tailed in particular. (n = 1 TAIL-seq experiment) c, Gene-level analysis of guanylated tails of cellular (grey) and viral (yellow) mRNAs in HCMV-infected HFF cells. d, siRNA knockdown confirmation by RT-qPCR. e, Decrease in the fraction of mixed tail of HBV mRNAs in TENT4-depleted HepG2.2.15 cells. f, TENT4A/B expression is induced in HCMV-infected HFF cells. TENT4A and TENT4B RNA levels in HCMV-infected HFF cells were measured by RT-qPCR (n = 1). g, Decrease in the fraction of mixed tail of HCMV RNA2.7 in TENT4-depleted HCMV-infected HFF cells. h, HBV mRNAs exhibit shorter poly(A) tail length in TENT4-depleted HepG2.2.15 cells. The medians are marked by dashed vertical lines and shown in parentheses. i, Global poly(A) tail length distribution of viral mRNAs after TENT4 depletion in HCMV-infected HFF cells. j, Poly(A) tail length distribution of HCMV RNA2.7 after TENT4 depletion. k, HBV transcripts and their respective CDS regions are shown. Bars indicate the position of RT-qPCR amplicons (#1–3). l, Tail modifications of HBV mRNAs across poly(A) tail length in TENT4-depleted HepG2.2.15 cells. m, Half-life of HCMV RNA2.7 was measured by RT-qPCR (n = 3 independent experiments) in HCMV-infected HFF cells after TENT4 depletion. GAPDH mRNA was used for normalization. Mean was calculated. Data for graphs in a-b,d-g,m are available as source data.

Source data

Extended Data Fig. 2 Analysis and confirmation of TENT4A/B and HBV mRNA interaction.

a, TENT4B-bound RNA fragments are again enriched within the PRE region of HBV (highlighted in yellow). Standardized read coverage of fCLIP-seq libraries across the HBV genome (NCBI:U95551.1). X-axis scale is the same as in Fig. 2a. b, Immunoprecipitation with anti-TENT4A and anti-TENT4B followed by RT-qPCR was used to measure the enrichment of TENT4-bound RNA in HepG2.2.15 cells (n = 3 independent experiments). Immunoprecipitation with normal mouse IgG was used for normalization. Mean was calculated and Error bars represent SEM. **P < 0.01, ****P < 0.0001; two-sided t test. Data for graphs in b is available as source data.

Source data

Extended Data Fig. 3 Confirmation of TENT4-dependent regulation of PRE reporters.

a, Knockout confirmation by western blotting. GAPDH was used as a loading control. b, RNA levels of firefly reporters harboring subregions of PRE were measured by RT-qPCR (PRE:PREα:PREβ, n = 4; αΔ1, n = 2; αΔ2, n = 1 independent experiment). mRNA levels of both renilla and control vector were used for normalization. Mean was calculated and Error bars represent SEM. *P < 0.05; two-sided t test. c, siRNA knockdown confirmation by western blotting in HEK293T cells. GAPDH was used as a loading control. d, Firefly luciferase activity of PRE reporter constructs in TENT4-depleted HEK293T cells (PRE:PREα:PREβ, n = 7; αΔ1, n = 6; αΔ2, n = 5 independent experiments). Luciferase activities of both renilla and control vector were used for normalization. Mean was calculated and Error bars represent SEM. *P < 0.05, **P < 0.01, ***P < 0.001; two-sided t test. e, Over-expression confirmation by western blotting in KO cells. GAPDH was used as a loading control. f, RNA levels of firefly reporters used in rescue experiments in TENT4 KO cells with ectopically expressed TENT4 wild-type or mutant (n = 3 independent experiments) were measured by RT-qPCR (n = 3 independent experiments). mRNA levels of both renilla and control vector were used for normalization. Mean was calculated and Error bars represent SEM. *P < 0.05, **P < 0.01; two-sided t test. Uncropped blots for panel a,c,e and data for graphs in b,d,f are available as source data.

Source data

Extended Data Fig. 4 Confirmation of TENT4-dependent regulation of RNA2.7 reporters.

a, Immunoprecipitation with anti-TENT4A was confirmed by western blotting in RNA2.7-transfected HEK293T cells. Normal mouse IgG (NMG) was used as a negative control. RNA2.7 transcript is shown with bar indicating the position of RT-qPCR amplicon. b-e, RNA levels of firefly reporters harboring b, subregions of RNA2.7 (n = 3 independent experiments), c, deletion constructs of fragment 1 (1–513) of RNA2.7 (n = 3 independent experiments), d, construct 1D (314-413), 1E (414–513) and stem-loop mutant 1E (n = 3 independent experiments) and e, construct SL2.7 (414–463) and controls (n = 3 independent experiments) in parental and TENT4 KO cells. mRNA levels of both renilla and control vector were used for normalization. Mean was calculated and Error bars represent SEM. *P < 0.05, **P < 0.01; two-sided t test. Uncropped blots for panel a and data for graphs in b-e are available as source data.

Source data

Extended Data Fig. 5 Confirmation of ZCCHC14 and SAMD4A/B knockdown.

siRNA knockdown confirmation by RT-qPCR (n = 4 independent experiments) and western blotting in HepG2.2.15 cells. Mean was calculated and Error bars represent SEM. ****P < 0.0001; two-sided t test. GAPDH was used as a loading control. Uncropped blots and data for graphs are available as source data.

Source data

Extended Data Fig. 6 Confirmation of ZCCHC14 knockdown, stem-loop-dependent tail regulation, and immunoprecipitation.

a, siRNA knockdown confirmation by western blotting in ZCCHC14-depleted HEK293T cells. GAPDH was used as a loading control. The dashed line indicates discontinuous lanes from the same gel. b, Stem-loop structures of WPREα and their respective mutants. c, Distributions of poly(A) tail length of reporter RNAs measured by Hire-PAT assay. The reporter constructs were transfected into HEK293T cells after ZCCHC14 depletion. d, Immunoprecipitation with anti-ZCCHC14 was confirmed by western blotting in HepG2.2.15 cells. Normal rabbit IgG (NRG) was used as a negative control. e, Immunoprecipitation with anti-TENT4A and anti-TENT4B followed by western blotting was used to measure the physical interaction between TENT4A/B and ZCCHC14 in HepG2.2.15 and primary HFF cells. Cell extracts were treated with RNase A. Normal mouse IgG (NMG) was used as a negative control. The asterisks indicate cross-reacting bands. Uncropped blots for panel a,d-e are available as source data.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

The information about genomic sequences of TENT4 and oligonucleotide sequences used in this study.

Source data

Source Data Fig. 1

Statistical source data

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Fig. 5

Unprocessed western blots

Source Data Fig. 6

Statistical source data

Source Data Fig. 6

Unprocessed western blots

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 3

Statistical source data

Source Data Extended Data Fig. 3

Unprocessed western blots

Source Data Extended Data Fig. 4

Statistical source data

Source Data Extended Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 6

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, D., Lee, Ys., Jung, SJ. et al. Viral hijacking of the TENT4–ZCCHC14 complex protects viral RNAs via mixed tailing. Nat Struct Mol Biol 27, 581–588 (2020). https://doi.org/10.1038/s41594-020-0427-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-020-0427-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing