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:

NMD-degradome sequencing reveals ribosome-bound intermediates with 3′-end non-templated nucleotides

A Publisher Correction to this article was published on 23 October 2018

This article has been updated

Abstract

Nonsense-mediated messenger RNA decay (NMD) controls mRNA quality and degrades physiologic mRNAs to fine-tune gene expression in changing developmental or environmental milieus. NMD requires that its targets are removed from the translating pool of mRNAs. Since the decay steps of mammalian NMD remain unknown, we developed assays to isolate and sequence direct NMD decay intermediates transcriptome-wide based on their co-immunoprecipitation with phosphorylated UPF1, which is the active form of this essential NMD factor. We show that, unlike steady-state UPF1, phosphorylated UPF1 binds predominantly deadenylated mRNA decay intermediates and activates NMD cooperatively from 5′- and 3′-ends. We leverage method modifications to characterize the 3′-ends of NMD decay intermediates, show that they are ribosome-bound, and reveal that some are subject to the addition of non-templated nucleotide. Uridines are added by TUT4 and TUT7 terminal uridylyl transferases and removed by the Perlman syndrome-associated exonuclease DIS3L2. The addition of other non-templated nucleotides appears to inhibit decay.

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: Transcriptome-wide NMD-DegSeq maps decay intermediate 3′-ends, revealing heterogenous non-templated nucleotide additions.
Fig. 2: NMD-DegNAs identifies NMD degradative activities.
Fig. 3: NMD-DegTAs demonstrate that terminal uridylyl transferases function in NMD.
Fig. 4: p-UPF1 localizes diffusely throughout the cytoplasm and fails to co-immunoprecipitate with NMD targets when translation is inhibited.
Fig. 5: NMD-DegRPL5 and NMD-DegRibo show NMD decay intermediates are bound by ribosomes.
Fig. 6: Model for the decay steps of NMD.

Similar content being viewed by others

Data availability

Data have been deposited in the Gene Expression Omnibus with accession code GSE111818. Source Data for Figs. 2c,d, 3a,b,d, and 4b are available online. Additional data are available upon reasonable request

Change history

  • 23 October 2018

    In the version of this paper originally published, in the PDF references 48–55 appeared in the reference list for the Methods section although they should have been in the reference list for the main text. The error has been corrected in the PDF now available.

References

  1. Kurosaki, T. & Maquat, L. E. Nonsense-mediated mRNA decay in humans at a glance. J. Cell. Sci. 129, 461–467 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  2. Goetz, A. E. & Wilkinson, M. Stress and the nonsense-mediated RNA decay pathway. Cell. Mol. Life Sci. 74, 3509–3531 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  3. Lejeune, F. Nonsense-mediate mRNA decay at the crossroads of many cellular pathways. BMC Reports 50, 175–185 (2017).

    Article  CAS  Google Scholar 

  4. Nasif, S., Contu, L. & Mühlemann, O. Beyond quality control: the role of nonsense-mediated mRNA decay (NMD) in regulating gene expression. Semin. Cell. Dev. Biol. 75, 78–87 (2017).

    Article  Google Scholar 

  5. Popp, M. W. & Maquat, L. E. Nonsense-mediated mRNA decay and cancer. Curr. Opin. Genet. Dev. 48, 44–50 (2017).

    Article  Google Scholar 

  6. Kurosaki, T. et al. A post-translational regulatory switch on UPF1 controls targeted mRNA degradation. Genes Dev. 28, 1900–1916 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  7. Durand, S., Franks, T. M. & Lykke-Andersen, J. Hyperphosphorylation amplifies UPF1 activity to resolve stalls in nonsense-mediated mRNA decay. Nat. Commun. 7, 12434 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  8. Imamachi, N., Salam, K. A., Suzuki, Y. & Akimitsu, N. A GC-rich sequence feature in the 3′ UTR directs UPF1-dependent mRNA decay in mammalian cells. Genome Res. 27, 407–418 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  9. Eberle, A. B., Lykke-Andersen, S., Mühlemann, O. & Jensen, T. H. SMG6 promotes endonucleolytic cleavage of nonsense mRNA in human cells. Nat. Struct. Mol. Biol. 16, 49–55 (2009).

    Article  CAS  Google Scholar 

  10. Lykke-Andersen, S. et al. Human nonsense-mediated RNA decay initiates widely by endonucleolysis and targets snoRNA host genes. Genes Dev. 28, 2498–2517 (2014).

    Article  PubMed Central  Google Scholar 

  11. Schmidt, S. A. et al. Identification of SMG6 cleavage sites and a preferred RNA cleavage motif by global analysis of endogenous NMD targets in human cells. Nucleic Acids Res. 43, 309–323 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  12. Lejeune, F., Li, X. & Maquat, L. E. Nonsense-mediated mRNA decay in mammalian cells involves decapping, deadenylating, and exonucleolytic activities. Mol. Cell 12, 675–687 (2003).

    Article  CAS  PubMed Central  Google Scholar 

  13. Yamashita, A. et al. Concerted action of poly(A) nucleases and decapping enzyme in mammalian mRNA turnover. Nat. Struct. Mol. Biol. 12, 1054–1063 (2005).

    Article  CAS  PubMed Central  Google Scholar 

  14. Jonas, S., Weichenrieder, O. & Izaurralde, E. An unusual arrangement of two 14-3-3-like domains in the SMG5-SMG7 heterodimer is required for efficient nonsense-mediated mRNA decay. Genes Dev. 27, 211–225 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  15. Loh, B., Jonas, S. & Izaurralde, E. The SMG5-SMG7 heterodimer directly recruits the CCR4-NOT deadenylase complex to mRNAs containing nonsense codons via interaction with POP2. Genes Dev. 27, 2125–2138 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  16. Hu, W., Sweet, T. J., Chamnongpol, S., Baker, K. E. & Coller, J. Co-translational mRNA decay in Saccharomyces cerevisiae. Nature 461, 225–229 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  17. Hu, W., Petzold, C., Coller, J. & Baker, K. E. Nonsense-mediated mRNA decapping occurs on polyribosomes in Saccharomyces cerevisiae. Nat. Struct. Mol. Biol. 17, 244–247 (2010).

    Article  CAS  PubMed Central  Google Scholar 

  18. Astuti, D. et al. Germline mutations in DIS3L2 cause the Perlman syndrome of overgrowth and Wilms tumor susceptibility. Nat. Genet. 44, 277–284 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  19. Morris, M. R., Astuti, D. & Maher, E. R. Perlman syndrome: overgrowth, Wilms tumor predisposition and DIS3L2. Am. J. Med. Genet. Part C Semin. Med. Genet. 163, 106–113 (2013).

    Article  CAS  Google Scholar 

  20. Wegert, J. et al. Mutations in the SIX1/2 pathway and the DROSHA/DGCR8 miRNA microprocessor complex underlie high-risk blastemal type Wilms tumors. Cancer Cell. 27, 298–311 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  21. Song, M.-G. & Kiledjian, M. 3′ Terminal oligo U-tract-mediated stimulation of decapping. RNA 13, 2356–2365 (2007).

    Article  CAS  PubMed Central  Google Scholar 

  22. Rissland, O. S. & Norbury, C. J. Decapping is preceded by 3′ uridylation in a novel pathway of bulk mRNA turnover. Nat. Struct. Mol. Biol. 16, 616–623 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  23. Choi, Y. S., Patena, W., Leavitt, A. D. & McManus, M. T. Widespread RNA 3′-end oligouridylation in mammals. RNA 18, 394–401 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  24. Chang, H. M., Triboulet, R., Thornton, J. E. & Gregory, R. I. A role for the Perlman syndrome exonuclease Dis3l2 in the Lin28-let-7 pathway. Nature 497, 244–248 (2014).

    Article  Google Scholar 

  25. Faehnle, C. R., Walleshauser, J. & Joshua-Tor, L. Mechanism of Dis3l2 substrate recognition in the Lin28-let-7 pathway. Nature 514, 252–256 (2014).

    Article  CAS  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed Central  Google Scholar 

  27. Pirouz, M., Du, P., Munafò, M. & Gregory, R. I. Dis3l2-mediated decay is a quality control pathway for noncoding RNAs. Cell Rep. 16, 1861–1873 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  28. Ustianenko, D. et al. TUT-DIS3L2 is a mammalian surveillance pathway for aberrant structured non-coding RNAs. EMBO J. 35, 2179–2191 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  29. Morgan, M. et al. mRNA 3′ uridylation and poly(A) tail length sculpt the mammalian maternal transcriptome. Nature 548, 347–351 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  30. Kurosaki, T., Hoque, M. & Maquat, L. E. Identifying cellular nonsense-mediated mRNA decay (NMD) targets: Immunoprecipitation of phosphorylated UPF1 followed by RNA sequencing (p-UPF1 RIP-Seq). Methods Mol. Biol. 1720, 175–186 (2018).

    Article  Google Scholar 

  31. Cho, H., Kim, K. M. & Kim, Y. K. Human proline-rich nuclear receptor coregulatory protein 2 mediates an interaction between mRNA surveillance machinery and decapping complex. Mol. Cell 33, 75–86 (2009).

    Article  CAS  Google Scholar 

  32. Franks, T. M., Singh, G. & Lykke-Andersen, J. Upf1 ATPase-dependent mRNP disassembly is required for completion of nonsense-mediated mRNA decay. Cell 143, 938–950 (2010).

    Article  CAS  PubMed Central  Google Scholar 

  33. Cho, H. et al. SMG5-PNRC2 is functionally dominant compared with SMG5-SMG7 in mammalian nonsense-mediated mRNA decay. Nucleic Acids Res. 41, 1319–1328 (2013).

    Article  CAS  Google Scholar 

  34. Yamashita, A., Ohnishi, T., Kashima, I., Taya, Y. & Ohno, S. Human SMG-1, a novel phosphatidylinositol 3-kinase-related protein kinase, associates with components of the mRNA surveillance complex and is involved in the regulation of nonsense-mediated mRNA decay. Genes Dev. 15, 2215–2228 (2001).

    Article  CAS  PubMed Central  Google Scholar 

  35. Ohnishi, T. et al. Phosphorylation of hUPF1 induces formation of mRNA surveillance complexes containing hSMG-5 and hSMG-7. Mol. Cell 12, 1187–1200 (2003).

    Article  CAS  Google Scholar 

  36. Isken, O. et al. Upf1 phosphorylation triggers translational repression during nonsense-mediated mRNA decay. Cell 133, 314–327 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  37. Mendell, J. T., Sharifi, N. A., Meyers, J. L., Martinez-Murillo, F. & Dietz, H. C. Nonsense surveillance regulates expression of diverse classes of mammalian transcripts and mutes genomic noise. Nat. Genet. 36, 1073–1078 (2004).

    Article  CAS  Google Scholar 

  38. Viegas, M. H., Gehring, N. H., Breit, S., Hentze, M. W. & Kulozik, A. E. The abundance of RNPS1, a protein component of the exon junction complex, can determine the variability in efficiency of the nonsense mediated decay pathway. Nucleic Acids Res. 35, 4542–4551 (2007).

    Article  CAS  PubMed Central  Google Scholar 

  39. Tani, H. et al. Identification of hundreds of novel UPF1 target transcripts by direct determination of whole transcriptome stability. RNA. Biol. 9, 1370–1379 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  40. Lou, C.-H. et al. Nonsense-mediated RNA decay influences human embryonic stem cell fate. Stem Cell Reports 6, 844–857 (2016).

    Article  CAS  PubMed Central  Google Scholar 

  41. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Article  CAS  Google Scholar 

  42. Piovesan, A., Caracausi, M., Antonaros, F., Pelleri, M. C. & Vitale, L. GeneBase 1.1: a tool to summarize data from NCBI gene datasets and its application to an update of human gene statistics. Database. 2016, baw153 (2016).

    Article  PubMed Central  Google Scholar 

  43. 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  Google Scholar 

  44. Kurosaki, T. & Maquat, L. E. Rules that govern UPF1 binding to mRNA 3′ UTRs. Proc. Natl Acad. Sci. USA 110, 3357–3362 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  45. Brouwer, R. et al. Three novel components of the human exosome. J. Biol. Chem. 276, 6177–6184 (2001).

    Article  CAS  PubMed Central  Google Scholar 

  46. van Dijk, E. L., Schilders, G. & Pruijn, G. J. M. Human cell growth requires a functional cytoplasmic exosome, which is involved in various mRNA decay pathways. RNA 13, 1027–1035 (2007).

    Article  PubMed Central  Google Scholar 

  47. Staals, R. H. J. et al. Dis3-like 1: a novel exoribonuclease associated with the human exosome. EMBO J. 29, 2358–2367 (2010).

    Article  CAS  PubMed Central  Google Scholar 

  48. Kikin, O., D’Antonio, L. & Bagga, P. S. QGRS Mapper: a web-based server for predicting G-quadruplexes in nucleotide sequences. Nucleic Acids Res. 34, 676–682 (2006).

    Article  Google Scholar 

  49. Mullen, T. E. & Marzluff, W. F. Degradation of histone mRNA requires oligouridylation followed by decapping and simultaneous degradation of the mRNA both 5′ to 3′ and 3′ to 5′. Genes Dev. 22, 50–65 (2008).

    Article  CAS  PubMed Central  Google Scholar 

  50. Choi, Y. S., Patena, W., Leavitt, A. D. & McManus, M. T. Widespread RNA 3′-end oligouridylation in mammals. RNA 18, 394–401 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  51. Slevin, M. K. et al. Deep sequencing shows multiple oligouridylations are required for 3′ to 5′ degradation of histone mRNAs on polyribosomes. Mol. Cell 53, 1020–1030 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  52. Welch, J. D. et al. EnD-Seq and AppEnD: sequencing 3′ ends to identify nontemplated tails and degradation intermediates. RNA 21, 1375–1389 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  53. Schmidt, M.-J., West, S. & Norbury, C. J. The human cytoplasmic RNA terminal U-transferase ZCCHC11 targets histone mRNAs for degradation. RNA 17, 39–44 (2011).

    Article  CAS  PubMed Central  Google Scholar 

  54. Unterholzner, L. & Izaurralde, E. SMG7 acts as a molecular link between mRNA surveillance and mRNA decay. Mol. Cell 16, 587–596 (2004).

    Article  CAS  PubMed Central  Google Scholar 

  55. Fukuhara, N. et al. SMG7 is a 14-3-3-like adaptor in the nonsense-mediated mRNA decay pathway. Mol. Cell 17, 537–547 (2005).

    Article  CAS  PubMed Central  Google Scholar 

  56. Trcek, T., Sato, H., Singer, R. H. & Maquat, L. E. Temporal and spatial characterization of nonsense-mediated mRNA decay. Genes Dev. 27, 541–551 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  57. Ingolia, N. T., Lareau, L. F. & Weissman, J. S. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell 147, 789–802 (2011).

    Article  CAS  PubMed Central  Google Scholar 

  58. Okada-Katsuhata, Y. et al. N- and C-terminal Upf1 phosphorylations create binding platforms for SMG-6 and SMG-5:SMG-7 during NMD. Nucleic Acids Res. 40, 1251–1266 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  59. Chakrabarti, S., Bonneau, F., Schussler, S., Eppinger, E. & Conti, E. Phospho-dependent and phospho-independent interactions of the helicase UPF1 with the NMD factors SMG5-SMG7 and SMG6. Nucleic Acids Res. 42, 9447–9460 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  60. Nicholson, P., Josi, C., Kurosawa, H., Yamashita, A. & Mühlemann, O. A novel phosphorylation-independent interaction between SMG6 and UPF1 is essential for human NMD. Nucleic Acids Res. 42, 9217–9235 (2014).

    Article  CAS  PubMed Central  Google Scholar 

  61. Gong, C., Kim, Y. K., Woeller, C. F., Tang, Y. & Maquat, L. E. SMD and NMD are competitive pathways that contribute to myogenesis: effects on PAX3 and myogenin mRNAs. Genes Dev. 23, 54–66 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  62. Yamashita, A. et al. SMG-8 and SMG-9, two novel subunits of the SMG-1 complex, regulate remodeling of the mRNA surveillance complex during nonsense-mediated mRNA decay. Genes Dev. 23, 1091–1105 (2009).

    Article  CAS  PubMed Central  Google Scholar 

  63. Serin, G., Gersappe, A., Black, J. D., Aronoff, R. & Maquat, L. E. Identification and characterization of human orthologues to Saccharomyces cerevisiae Upf2 protein and Upf3 protein (Caenorhabditis elegans SMG-4). Mol. Cell. Biol. 21, 209–223 (2001).

    Article  CAS  PubMed Central  Google Scholar 

  64. Martin, M. CutAdapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J 17, 10–12 (2011).

    Article  Google Scholar 

  65. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2012).

    Article  PubMed Central  Google Scholar 

  66. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  Google Scholar 

  67. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome. Biol. 15, 550 (2014).

    Article  PubMed Central  Google Scholar 

  68. Huber, W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015).

    Article  CAS  PubMed Central  Google Scholar 

  69. Li, H. et al. The Sequence alignment/map (SAM) format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed Central  Google Scholar 

  70. Ferrari, F. et al. ‘Jump Start and Gain’ model for dosage compensation in Drosophila based on direct sequencing of nascent transcripts. Cell Rep 5, 629–636 (2013).

    Article  CAS  Google Scholar 

  71. Breese, M. R. & Liu, Y. NGSUtils: a software suite for analyzing and manipulating next-generation sequencing datasets. Bioinformatics 29, 494–496 (2013).

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Pelletier for hippuristanol, S. Ohno and A. Yamashita for anti-p-UPF1(S1078, S1096), D. Ermolenko for Biocomp Gradient Fractionator usage, the University of Rochester Genomics Research Center for assistance in NGS library design, construction, and sequencing support, the University of Rochester Confocal Microscopy Core Facility for technical advice regarding confocal microscopy, and B. Lucas, M. Popp, and X. Rambout for comments on the manuscript. This work was supported by the National Institutes of Health (NIH) grant number R01 GM59614 to L.E.M. Salary support for T.K. was derived in part from a post-doctoral FRAXA Research Foundation Fellowship. The NIH through grant number S10 OD021489-01A1 supported purchase of the Typhoon FLA 9500 Phosphorimager.

Author information

Authors and Affiliations

Authors

Contributions

T.K. and L.E.M. conceived the project, developed the methods, and analyzed the data. T.K. and K.M. performed the experiments. J.R.M. and T.K. performed computational analyses. T.K. and L.E.M. wrote the manuscript with help writing up the computational analyses from J.R.M.

Corresponding author

Correspondence to Lynne E. Maquat.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Transcriptome-wide NMD-Degradome Sequencing (NMD-DegSeq) maps decay intermediate (DI) 3′-ends, revealing the existence of not only homogeneous but also heterogeneous non-templated nucleotide additions.

a, Supplemental details of transcriptome-wide NMD-DegSeq library construction. b, Western blot prior to (−) or after immunoprecipitation using rIgG or anti(α)-p-UPF1. The leftmost five lanes represent threefold serial dilutions of lysate prior to immunoprecipitation. c, Control experiment showing that there was neither RNA degradation during p-UPF1 immunoprecipitation (IP) nor non-specific binding in the anti(α)-p-UPF1 immunoprecipitation using in vitro–synthesized and 32P-labeled mouse β-actin mRNA, which is not an NMD target and was added to lysates prior to p-UPF1 immunoprecipitation. d, Primer extension assay workflow. See the Methods for experimental procedures. e, Size distribution (blue histogram) and cumulative size distribution (red line) of p-UPF1-bound decay intermediates analyzed for NMD-DegSeq. f, Mean read density of exonic sequence reads ± 500 nucleotides (nt) relative to the transcription start site (TSS). Sequence reads from paired-end reads before (−) immunoprecipitation (dotted line) or after (solid line) p-UPF1 immunoprecipitation are shown. g, As in f, except for exonic sequence reads ± 500 nt relative to the translation start codon. h, As in Fig. 1h, but for DDIT4 mRNA, aligned to the coding strand of the DDIT4 gene. TSS, transcription start site; TES, transcription end site; Alt TES, alternative transcription end site. i, Scheme for streamlined NMD-DegSeq. See the Methods for details. j, Western blotting (WB) before (−) or after immunoprecipitation of lysates of HEK293T-cells, which were treated with okadaic acid prior to lysis, using anti(α) p-UPF1 or, as a negative control, rabbit (r)IgG. The five leftmost lanes represent threefold serial dilutions of lysate prior to immunoprecipitation. k, SYBR-Gold staining of the dRLUC-Gl PTC decay intermediate. l, Sequences of dRLUC-Gl PTC decay intermediates in streamlined NMD-DegSeq. The PTC (red) and exon 1–exon 2 junction are shown in the reference sequence (RefSeq). PCR primers (sense, blue arrows; antisense, green arrows) and non-templated nucleotide additions (pink) are provided in the decay intermediates, for which numbers to the left of blue arrows denote the percent (%) that each decay intermediate constitutes relative to all sequenced decay intermediates. m, As in k, but for GADD45A decay intermediates. n, As in l, but for GADD45A decay intermediates. Results are representative of two independently performed experiments.

Supplementary Figure 2 Defining degradative activities that generate NMD decay intermediates.

a, Western blotting of the samples analyzed in Fig. 2c,d. The five leftmost lanes represent threefold serial dilutions of lysate. Norm, normal; PTC, premature termination codon; Ctl, control. b, As in a, using multiple siRNAs where indicated. c, Western blotting (WB), SYBR-Gold-stained RT–PCR products of full-length (FL) dRLUC-Gl or MUP mRNA. d, As in c, but RT–PCR products of full-length GADD45A NMD target and β-actin mRNA. e, Sequences of dRLUC-Gl PTC decay intermediates (DIs) obtained in HEK293T cells transfected with the specified siRNAs. The PTC (red), exon 1–exon 2 junction, and putative G-quadruplex-forming sequences (underlined) are shown in the reference sequence (RefSeq). PCR primers (sense, blue arrows; antisense, green arrows) and non-templated nucleotide additions (pink) are provided in the decay intermediates, for which numbers in parentheses specify the number of clones sequenced and numbers to the left of the blue arrows denote the percent (%) that each decay intermediate constitutes relative to all sequenced decay intermediates. f, Essentially as in e but for endogenous GADD45A mRNA. Ter (red) specifies the normal termination codon. Short sequencing products (<10 nt) were excluded from the analysis. NMD-DegNAs results are representative of two independently performed experiments.

Supplementary Figure 3 NMD-target degradation involves terminal uridylyl transferases.

a, Western blotting of the HEK293T cell lysates analyzed in Fig. 3a–d. The four leftmost lanes represent threefold serial dilutions of lysate. Ctl, control. b, As in Fig. 3d, but for analyzing RNAs with 3′-terminal A after normalization to their level in total-cell RNAs. c, As in b, but for analyzing RNAs with 3′-terminal G. d, As in b, but for analyzing RNAs with 3′-terminal C. e, As in Fig. 3d, but analyzing RNAs ending with UUUU. *P < 0.05 and **P < 0.01 pertain to comparisons to Ctl siRNA or XRN1 siRNA samples (unpaired two-tailed t test); n = 3–4, showing means with s.d.

Supplementary Figure 4 p-UPF1 localizes diffusely throughout cytoplasm with ribosomal proteins and other NMD factors, and fails to co-immunoprecipitate with RPL5 when translation is inhibited.

a, Western blotting of lysates of HeLa cells (5 × 106/100-mm dish) transiently transfected with the specified siRNA. Ctl, control. b, Immunofluorescence microscopy of Ctl siRNA-treated HeLa cells (5 × 104/well of a 24-well plate) to localize anti(α)-UPF1, anti-p-UPF1(S1089) or anti-p-UPF1(S1116) reactivity, shown in green, relative to anti-α-tubulin reactivity, shown in red, and nuclei, stained blue using DAPI. Scale bar, 10 μm. c, As in b but using UPF1 siRNA-treated cells. d, Western blotting of nuclear and cytoplasmic fractions of HEK293T cells (8 × 107/150-mm dish). e, As in Fig. 4a, but using the specified antibody. f, Western blotting of lysates of HeLa cells (5 × 106/100-mm dish) that were or were not (−) exposed to 200 nM of okadaic acid for 3 h. g, Immunofluorescence microscopy of okadaic acid–treated HeLa cells (5 × 104/well using a 24-well plate) to localize anti(α)-UPF1, anti-p-UPF1(S1089) or anti-p-UPF1(S1116) reactivity, shown in green, relative to anti-Dcp1a reactivity, shown in red. Nuclei were stained blue using DAPI. Solid boxed regions are threefold magnifications of the dotted boxed regions. Scale bar, 10 μm. h, Western blotting of the samples assayed in Fig. 4b. Translation inhibition was evaluated by measuring the abundance of cell division cycle 6 (CDC6) protein, which has a half-life of 2–4 h (Genes Dev. 14, 2330–2343, 2000; Eur. J. Biochem. 269, 1040–1046, 2002). i, As in h, but immunoprecipitation used anti(α)-RPL5. j, Using fractions from polysome profile of HEK293T cell lysates shown (top), western blotting (WB) using the specified antibody and ethidium bromide staining (EB) to analyze 28S and 18S rRNAs.

Supplementary Figure 5 Sequences of NMD decay intermediates from NMD-DegRPL5 and NMD-DegRibo.

a, Sequences of the dRLUC-Gl PTC decay intermediates obtained in Fig. 5c. The PTC (red) and exon 1–exon 2 junction are shown in the reference sequence (RefSeq). PCR primers (sense, blue arrows; antisense, green arrows) and non-templated nucleotide additions (pink) are provided in the decay intermediates, for which numbers to the left of the blue arrows denote the percent (%) that each decay intermediate constitutes relative to all sequenced decay intermediates. b, As in a, but for the GADD45A decay intermediates obtained in Fig. 5d. Ter (red) specifies the normal termination codon. c, dRLUC-Gl PTC decay intermediates were obtained in Fig. 5f. RNA samples were generated using the scheme outlined for polysome fractionation shown in Fig. 5a. d, As in c, but for the GADD45A NMD target decay intermediates obtained in Fig. 5g.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5, Supplementary Tables 2 and 3, and Supplementary Notes 1–5

Reporting Summary

Supplementary Table 1

Differential expression of RNA fragments that derived from bona fide NMD targets

Supplementary Dataset 1

Supplementary Dataset 2

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kurosaki, T., Miyoshi, K., Myers, J.R. et al. NMD-degradome sequencing reveals ribosome-bound intermediates with 3′-end non-templated nucleotides. Nat Struct Mol Biol 25, 940–950 (2018). https://doi.org/10.1038/s41594-018-0132-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0132-7

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