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.

  • Analysis
  • Published:

The rules and impact of nonsense-mediated mRNA decay in human cancers

Abstract

Premature termination codons (PTCs) cause a large proportion of inherited human genetic diseases. PTC-containing transcripts can be degraded by an mRNA surveillance pathway termed nonsense-mediated mRNA decay (NMD). However, the efficiency of NMD varies; it is inefficient when a PTC is located downstream of the last exon junction complex (EJC). We used matched exome and transcriptome data from 9,769 human tumors to systematically elucidate the rules of NMD targeting in human cells. An integrated model incorporating multiple rules beyond the canonical EJC model explains approximately three-fourths of the non-random variance in NMD efficiency across thousands of PTCs. We also show that dosage compensation may sometimes mask the effects of NMD. Applying the NMD model identifies signatures of both positive and negative selection on NMD-triggering mutations in human tumors and provides a classification for tumor-suppressor genes.

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

Figure 1: Study overview.
Figure 2: A downstream EJC and proximity to the start codon are widespread signals for NMD.
Figure 3: Exon length, distance to the stop codon, mRNA decay rate and RNA-binding proteins influence NMD efficiency.
Figure 4: The identified NMD rules explain a large part of NMD efficiency.
Figure 5: Signatures of negative and positive selection on somatic nonsense mutations.
Figure 6: Model summarizing the rules governing NMD in human cells.

Similar content being viewed by others

References

  1. Maquat, L.E. When cells stop making sense: effects of nonsense codons on RNA metabolism in vertebrate cells. RNA 1, 453–465 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Frischmeyer, P.A. & Dietz, H.C. Nonsense-mediated mRNA decay in health and disease. Hum. Mol. Genet. 8, 1893–1900 (1999).

    Article  CAS  Google Scholar 

  4. Hall, G.W. & Thein, S. Nonsense codon mutations in the terminal exon of the β-globin gene are not associated with a reduction in β-mRNA accumulation: a mechanism for the phenotype of dominant β-thalassemia. Blood 83, 2031–2037 (1994).

    CAS  PubMed  Google Scholar 

  5. Kerr, T.P., Sewry, C.A., Robb, S.A. & Roberts, R.G. Long mutant dystrophins and variable phenotypes: evasion of nonsense-mediated decay? Hum. Genet. 109, 402–407 (2001).

    Article  CAS  Google Scholar 

  6. Zhang, J., Sun, X., Qian, Y., LaDuca, J.P. & Maquat, L.E. At least one intron is required for the nonsense-mediated decay of triosephosphate isomerase mRNA: a possible link between nuclear splicing and cytoplasmic translation. Mol. Cell. Biol. 18, 5272–5283 (1998).

    Article  CAS  Google Scholar 

  7. Thermann, R. et al. Binary specification of nonsense codons by splicing and cytoplasmic translation. EMBO J. 17, 3484–3494 (1998).

    Article  CAS  Google Scholar 

  8. Le Hir, H., Gatfield, D., Izaurralde, E. & Moore, M.J. The exon–exon junction complex provides a binding platform for factors involved in mRNA export and nonsense-mediated mRNA decay. EMBO J. 20, 4987–4997 (2001).

    Article  CAS  Google Scholar 

  9. Chamieh, H., Ballut, L., Bonneau, F. & Le Hir, H. NMD factors UPF2 and UPF3 bridge UPF1 to the exon junction complex and stimulate its RNA helicase activity. Nat. Struct. Mol. Biol. 15, 85–93 (2008).

    Article  CAS  Google Scholar 

  10. Brogna, S. & Wen, J. Nonsense-mediated mRNA decay (NMD) mechanisms. Nat. Struct. Mol. Biol. 16, 107–113 (2009).

    Article  CAS  Google Scholar 

  11. Wang, J., Gudikote, J.P., Olivas, O.R. & Wilkinson, M.F. Boundary-independent polar nonsense-mediated decay. EMBO Rep. 3, 274–279 (2002).

    Article  CAS  Google Scholar 

  12. Bühler, M., Paillusson, A. & Mühlemann, O. Efficient downregulation of immunoglobulin μ mRNA with premature translation-termination codons requires the 5′-half of the VDJ exon. Nucleic Acids Res. 32, 3304–3315 (2004).

    Article  Google Scholar 

  13. Eberle, A.B., Stalder, L., Mathys, H., Orozco, R.Z. & Mühlemann, O. Posttranscriptional gene regulation by spatial rearrangement of the 3′ untranslated region. PLoS Biol. 6, e92 (2008).

    Article  Google Scholar 

  14. Mangus, D.A., Evans, M.C. & Jacobson, A. Poly(A)-binding proteins: multifunctional scaffolds for the post-transcriptional control of gene expression. Genome Biol. 4, 223 (2003).

    Article  Google Scholar 

  15. Gatfield, D., Unterholzner, L., Ciccarelli, F.D., Bork, P. & Izaurralde, E. Nonsense-mediated mRNA decay in Drosophila: at the intersection of the yeast and mammalian pathways. EMBO J. 22, 3960–3970 (2003).

    Article  CAS  Google Scholar 

  16. Longman, D., Plasterk, R.H., Johnstone, I.L. & Cáceres, J.F. Mechanistic insights and identification of two novel factors in the C. elegans NMD pathway. Genes Dev. 21, 1075–1085 (2007).

    Article  CAS  Google Scholar 

  17. Zhang, J. & Maquat, L.E. Evidence that translation reinitiation abrogates nonsense-mediated mRNA decay in mammalian cells. EMBO J. 16, 826–833 (1997).

    Article  CAS  Google Scholar 

  18. Romão, L. et al. Nonsense mutations in the human β-globin gene lead to unexpected levels of cytoplasmic mRNA accumulation. Blood 96, 2895–2901 (2000).

    PubMed  Google Scholar 

  19. Silva, A.L., Ribeiro, P., Inácio, A., Liebhaber, S.A. & Romão, L. Proximity of the poly(A)-binding protein to a premature termination codon inhibits mammalian nonsense-mediated mRNA decay. RNA 14, 563–576 (2008).

    Article  CAS  Google Scholar 

  20. Lappalainen, T. et al. Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501, 506–511 (2013).

    Article  CAS  Google Scholar 

  21. Rivas, M.A. et al. Effect of predicted protein-truncating genetic variants on the human transcriptome. Science 348, 666–669 (2015).

    Article  CAS  Google Scholar 

  22. Martincorena, I. & Campbell, P.J. Somatic mutation in cancer and normal cells. Science 349, 1483–1489 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  24. Singh, G., Rebbapragada, I. & Lykke-Andersen, J. A competition between stimulators and antagonists of Upf complex recruitment governs human nonsense-mediated mRNA decay. PLoS Biol. 6, e111 (2008).

    Article  Google Scholar 

  25. 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  Google Scholar 

  26. Leek, J.T. & Storey, J.D. Capturing heterogeneity in gene expression studies by surrogate variable analysis. PLoS Genet. 3, 1724–1735 (2007).

    Article  CAS  Google Scholar 

  27. Fiorini, F., Bagchi, D., Le Hir, H. & Croquette, V. Human Upf1 is a highly processive RNA helicase and translocase with RNP remodelling activities. Nat. Commun. 6, 7581 (2015).

    Article  Google Scholar 

  28. Ray, D. et al. A compendium of RNA-binding motifs for decoding gene regulation. Nature 499, 172–177 (2013).

    Article  CAS  Google Scholar 

  29. Zhang, Z. et al. Nonsense-mediated decay targets have multiple sequence-related features that can inhibit translation. Mol. Syst. Biol. 6, 442 (2010).

    Article  Google Scholar 

  30. Supek, F., Skunca, N., Repar, J., Vlahovicek, K. & Smuc, T. Translational selection is ubiquitous in prokaryotes. PLoS Genet. 6, e1001004 (2010).

    Article  Google Scholar 

  31. Rehwinkel, J., Raes, J. & Izaurralde, E. Nonsense-mediated mRNA decay: target genes and functional diversification of effectors. Trends Biochem. Sci. 31, 639–646 (2006).

    Article  CAS  Google Scholar 

  32. Malone, J.H. et al. Mediation of Drosophila autosomal dosage effects and compensation by network interactions. Genome Biol. 13, r28 (2012).

    Article  CAS  Google Scholar 

  33. Fehrmann, R.S. et al. Gene expression analysis identifies global gene dosage sensitivity in cancer. Nat. Genet. 47, 115–125 (2015).

    Article  CAS  Google Scholar 

  34. Ju, Y.S. et al. Origins and functional consequences of somatic mitochondrial DNA mutations in human cancer. eLife 3, e02935 (2014).

    Article  Google Scholar 

  35. Holbrook, J.A., Neu-Yilik, G., Hentze, M.W. & Kulozik, A.E. Nonsense-mediated decay approaches the clinic. Nat. Genet. 36, 801–808 (2004).

    Article  CAS  Google Scholar 

  36. Gutmann, D.H. et al. Haploinsufficiency for the neurofibromatosis 1 (NF1) tumor suppressor results in increased astrocyte proliferation. Oncogene 18, 4450–4459 (1999).

    Article  CAS  Google Scholar 

  37. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013).

    Article  CAS  Google Scholar 

  38. McFarland, C.D., Mirny, L.A. & Korolev, K.S. Tug-of-war between driver and passenger mutations in cancer and other adaptive processes. Proc. Natl. Acad. Sci. USA 111, 15138–15143 (2014).

    Article  CAS  Google Scholar 

  39. Forbes, S.A. et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 43, D805–D811 (2015).

    Article  CAS  Google Scholar 

  40. Hart, T., Brown, K.R., Sircoulomb, F., Rottapel, R. & Moffat, J. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol. Syst. Biol. 10, 733 (2014).

    Article  Google Scholar 

  41. Solimini, N.L. et al. Recurrent hemizygous deletions in cancers may optimize proliferative potential. Science 337, 104–109 (2012).

    Article  CAS  Google Scholar 

  42. Gonzàlez-Porta, M., Frankish, A., Rung, J., Harrow, J. & Brazma, A. Transcriptome analysis of human tissues and cell lines reveals one dominant transcript per gene. Genome Biol. 14, R70 (2013).

    Article  Google Scholar 

  43. Pirinen, M. et al. Assessing allele-specific expression across multiple tissues from RNA-seq read data. Bioinformatics 31, 2497–2504 (2015).

    Article  CAS  Google Scholar 

  44. Stegle, O., Parts, L., Durbin, R. & Winn, J. A Bayesian framework to account for complex non-genetic factors in gene expression levels greatly increases power in eQTL studies. PLoS Comput. Biol. 6, e1000770 (2010).

    Article  Google Scholar 

  45. Supek, F., Miñana, B., Valcárcel, J., Gabaldón, T. & Lehner, B. Synonymous mutations frequently act as driver mutations in human cancers. Cell 156, 1324–1335 (2014).

    Article  CAS  Google Scholar 

  46. Friedel, C.C., Dölken, L., Ruzsics, Z., Koszinowski, U.H. & Zimmer, R. Conserved principles of mammalian transcriptional regulation revealed by RNA half-life. Nucleic Acids Res. 37, e115 (2009).

    Article  Google Scholar 

  47. Tani, H. et al. Genome-wide determination of RNA stability reveals hundreds of short-lived noncoding transcripts in mammals. Genome Res. 22, 947–956 (2012).

    Article  CAS  Google Scholar 

  48. Andreev, D.E. et al. Translation of 5′ leaders is pervasive in genes resistant to eIF2 repression. eLife 4, e03971 (2015).

    Article  Google Scholar 

  49. Huntley, R.P. et al. The GOA database: gene ontology annotation updates for 2015. Nucleic Acids Res. 43, D1057–D1063 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by a European Research Council (ERC) Consolidator grant (616434), the Spanish Ministry of Economy and Competitiveness (BFU2011-26206 and 'Centro de Excelencia Severo Ochoa 2013–2017' SEV-2012-0208), the AXA Research Fund, the Agencia de Gestio d'Ajuts Universitaris i de Recerca (AGAUR), FP7 project 4DCellFate (277899) and the EMBL-CRG Systems Biology Program. F.S. was also supported by FP7 grants MAESTRA (ICT-2013-612944) and InnoMol (FP7-REGPOT-2012-2013-1-316289).

Author information

Authors and Affiliations

Authors

Contributions

R.G.H.L. performed all analyses. R.G.H.L., F.S. and B.L. designed analyses, interpreted the data and wrote the manuscript. B.L. conceived the project.

Corresponding authors

Correspondence to Fran Supek or Ben Lehner.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Data sources and validation of known NMD rules in human genomic data sets.

(a) Overview of the data preprocessing pipeline. (b) Somatic nonsense mutation tally across cancer types in the TCGA. (c,d) The standard EJC model applied to frameshift somatic mutations in TCGA (c) and germline truncating variants in Geuvadis (d). (eh) The faux 3′ UTR model is not broadly supported in TCGA somatic nonsense variants when examining the last exon (e) or intronless genes (f), and similarly so in the TCGA frameshift data set (g) and in the Geuvadis germline variants (h). In all panels, the blue line is a fit using loess or generalized additive models (Online Methods) and the shaded area is its 95% confidence interval.

Supplementary Figure 2 Effects of start-proximal PTCs on NMD efficiency.

(a) In genes with 3′ UTR introns, the penultimate EJC becomes the NMD-inducing EJC, as observed in Geuvadis germline variants. (be) The start-proximal effect on NMD evasion is evident in additional data sets and is associated with downstream in-frame stop codons, demonstrated for somatic frameshifts (b,d) and for germline variants (c,e). (f,g) There is no clear effect of distance to the downstream in-frame start codon (f) or the Kozak sequence (g). (hk) There is no decrease in NMD efficiency for shorter distances between the PTC (h) and putative PABPC1-binding motifs (i–k) in a hypothetical looped mRNA conformation. In all panels, the blue line is a fit using loess or generalized additive models (Online Methods) and the shaded area is its 95% confidence interval.

Supplementary Figure 3 Other influences on measured NMD efficiency in human genomic data.

(a) Somatic mutations with high allelic frequency cause NMD estimates to appear more efficient. (be) Long exons and very large distances to the normal stop codon are associated with inefficient NMD in additional data sets consisting of TCGA somatic frameshift mutations (b,d) and Geuvadis germline truncating variants (c,e). (f,g) The standard EJC model and start-proximal NMD evasion have a dominant effect on NMD efficiency (f) and were thus factored out (g) using regression, facilitating discovery of further rules. (h,i) Rapid mRNA turnover attenuates the effects of NMD in somatic frameshifts (h) and germline variants (i). (j) NMD is slightly more efficient in highly expressed genes. (k) Reduced NMD efficiency in transcripts with short half-lives is also observed when using mRNA half-life measures from HeLa cells. The x axis shows the median mRNA half-life in minutes in each box plot. (l,m) The observed reduction in NMD efficiency in genes with fast mRNA turnover in B cells (l) is still observed upon explicitly factoring out gene expression levels from the NMD efficiency measure (m). In all panels with a continuous x axis, the axis was square root transformed. In all panels, the blue line is a fit using loess or generalized additive models (Online Methods) and the shaded area is its 95% confidence interval.

Supplementary Figure 4 RNA-binding protein motifs associated with differential NMD efficiency in additional data sets.

(a) Each row corresponds to one motif, which was detected near (±100 nt) the PTC in the case of SRSF1, PABPN1 and SNRPB2 and within the original 3′ UTR for ACO1. The leftmost column corresponds to the discovery data set consisting of nonsense somatic mutations in TCGA tumors (red); the middle column is the validation data set with frameshift mutations in TCGA (blue); and the rightmost column is the independent data set with truncating germline variants in the Geuvadis cohort (green). The “FALSE” and “TRUE” labels correspond to the motif being absent or present, respectively, in the particular location of the transcript. P values were calculated by Mann–Whitney U tests, two-tailed. An association was considered validated in additional data sets (frameshift and germline variants) if the pooled P value across the three data sets was <0.005. (b) Data are presented as in a. Motifs shown here were significant in the discovery set with TCGA nonsense somatic mutations and in the validation data set with TCGA somatic frameshifts. In the Geuvadis validation set with germline truncating variants, we observed a trend in the correct direction for these motifs but significance was not reached (P > 0.10).

Supplementary Figure 5 Some RNA-binding protein motifs associated with differential NMD efficiency have a general effect on mRNA turnover.

(ac) Association of the RBP motifs in Supplementary Figure 4 with mRNA half-life measures from B cells (left box plot) and HeLa cells (right box plot). Motifs that were associated with NMD when located near a PTC were tested for a different mRNA half-life when the motif was detected near the wild-type stop codon, and the GATCAA motif was tested around the most 3′ exon junction. The remaining RBP motifs were tested in the location of the transcript where they also showed an association with differential NMD (Supplementary Fig. 4). The “FALSE” and “TRUE” labels correspond to the motif being absent or present, respectively, in the particular location of the transcript. P values were calculated by Mann–Whitney U tests, two-tailed. (d,e) A marginally significant difference in NMD efficiency is observed for the 20% most biased transcripts. Codon optimality bias was defined by counting the codon usage that corresponds to highly abundant tRNAs (Online Methods) in either the whole coding region (d) or the first 50 codons (e). Bins are equally populated and P values were calculated by Mann–Whitney U tests, two-tailed.

Supplementary Figure 6 Tumor-suppressor gene candidates examined for mechanisms of inactivation via NMD and copy number changes.

(a) Principal-components plot as in Figure 5g, with nine additional genes overlaid (blue points). The additional genes were proposed to be possible tumor suppressors in Davoli et al.37, on the basis of their enrichment for deleterious over benign somatic mutations (the TUSON Explorer method, FDR ≤ 20% for all except ZFHX3 (22%) and MKI67 (31%)). Examined genes have ≥50 nonsense mutations in our data set. (b) Relative frequencies of NMD-inducing versus NMD-evading nonsense mutations (“NMD” and “noNMD”, respectively) and of gene deletions versus absence thereof (“Del” and “noDel”, respectively). Importantly, in the predictor from Davoli et al., nonsense mutations were not subclassified by their putative effects on NMD nor were copy number alterations used as input.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Note. (PDF 3191 kb)

Supplementary Table 1

Importance of various sequence features for predicting NMD efficiency before adjusting for the prevalent NMD rules. (XLSX 122 kb)

Supplementary Table 2

Importance of various sequence features for predicting NMD efficiency after adjusting for the prevalent NMD rules. (XLSX 123 kb)

Supplementary Table 3

Significant RF features after adjusting for the prevalent NMD rules (permutation based P value < 0.05). (XLSX 8 kb)

Supplementary Table 4

Genes with >50 somatic nonsense mutations in TCGA. (XLSX 13 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lindeboom, R., Supek, F. & Lehner, B. The rules and impact of nonsense-mediated mRNA decay in human cancers. Nat Genet 48, 1112–1118 (2016). https://doi.org/10.1038/ng.3664

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3664

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer