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.

  • Letter
  • Published:

Translation readthrough mitigation

Abstract

A fraction of ribosomes engaged in translation will fail to terminate when reaching a stop codon, yielding nascent proteins inappropriately extended on their C termini. Although such extended proteins can interfere with normal cellular processes, known mechanisms of translational surveillance1 are insufficient to protect cells from potential dominant consequences. Here, through a combination of transgenics and CRISPR–Cas9 gene editing in Caenorhabditis elegans, we demonstrate a consistent ability of cells to block accumulation of C-terminal-extended proteins that result from failure to terminate at stop codons. Sequences encoded by the 3′ untranslated region (UTR) were sufficient to lower protein levels. Measurements of mRNA levels and translation suggested a co- or post-translational mechanism of action for these sequences in C. elegans. Similar mechanisms evidently operate in human cells, in which we observed a comparable tendency for translated human 3′ UTR sequences to reduce mature protein expression in tissue culture assays, including 3′ UTR sequences from the hypomorphic ‘Constant Spring’ haemoglobin stop codon variant. We suggest that 3′ UTRs may encode peptide sequences that destabilize the attached protein, providing mitigation of unwelcome and varied translation errors.

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: Translation into 3′ UTRs results in substantial loss of protein expression.
Figure 2: Identification of determinants for product loss upon translation into the 3′ UTR.
Figure 3: Translation into the 3′ UTR at an endogenous locus decreases protein levels.
Figure 4: Translation into 3′ UTRs results in protein loss for several genes in humans.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

References

  1. Klauer, A. A. & van Hoof, A. Degradation of mRNAs that lack a stop codon: a decade of nonstop progress. Wiley Interdiscip. Rev. RNA 3, 649–660 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hamby, S. E., Thomas, N. S., Cooper, D. N. & Chuzhanova, N. A meta-analysis of single base-pair substitutions in translational termination codons (‘nonstop’ mutations) that cause human inherited disease. Hum. Genomics 5, 241–264 (2011)

    Article  PubMed  PubMed Central  Google Scholar 

  3. Williams, I., Richardson, J., Starkey, A. & Stansfield, I. Genome-wide prediction of stop codon readthrough during translation in the yeast Saccharomyces cerevisiae. Nucleic Acids Res. 32, 6605–6616 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Falini, B. et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N. Engl. J. Med. 352, 254–266 (2005)

    Article  CAS  PubMed  Google Scholar 

  5. Hollingsworth, T. J. & Gross, A. K. The severe autosomal dominant retinitis pigmentosa rhodopsin mutant Ter349Glu mislocalizes and induces rapid rod cell death. J. Biol. Chem. 288, 29047–29055 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vidal, R. et al. A stop-codon mutation in the BRI gene associated with familial British dementia. Nature 399, 776–781 (1999)

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Vidal, R. et al. A decamer duplication in the 3′ region of the BRI gene originates an amyloid peptide that is associated with dementia in a Danish kindred. Proc. Natl Acad. Sci. USA 97, 4920–4925 (2000)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Pang, S. et al. A novel nonstop mutation in the stop codon and a novel missense mutation in the type II 3beta-hydroxysteroid dehydrogenase (3beta-HSD) gene causing, respectively, nonclassic and classic 3β-HSD deficiency congenital adrenal hyperplasia. J. Clin. Endocrinol. Metab. 87, 2556–2563 (2002)

    CAS  PubMed  Google Scholar 

  9. Clegg, J. B., Weatherall, D. J. & Milner, P. F. Haemoglobin Constant Spring—a chain termination mutant? Nature 234, 337–340 (1971)

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Namy, O., Duchateau-Nguyen, G. & Rousset, J. P. Translational readthrough of the PDE2 stop codon modulates cAMP levels in Saccharomyces cerevisiae. Mol. Microbiol. 43, 641–652 (2002)

    Article  CAS  PubMed  Google Scholar 

  11. Inada, T. & Aiba, H. Translation of aberrant mRNAs lacking a termination codon or with a shortened 3′-UTR is repressed after initiation in yeast. EMBO J. 24, 1584–1595 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Shibata, N. et al. Degradation of stop codon read-through mutant proteins via the ubiquitin-proteasome system causes hereditary disorders. J. Biol. Chem. 290, 28428–28437 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Capone, J. P., Sharp, P. A. & RajBhandary, U. L. Amber, ochre and opal suppressor tRNA genes derived from a human serine tRNA gene. EMBO J. 4, 213–221 (1985)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ahier, A. & Jarriault, S. Simultaneous expression of multiple proteins under a single promoter in Caenorhabditis elegans via a versatile 2A-based toolkit. Genetics 196, 605–613 (2014)

    Article  CAS  PubMed  Google Scholar 

  15. Doronina, V. A. et al. Site-specific release of nascent chains from ribosomes at a sense codon. Mol. Cell. Biol. 28, 4227–4239 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jan, C. H., Friedman, R. C., Ruby, J. G. & Bartel, D. P. Formation, regulation and evolution of Caenorhabditis elegans 3’UTRs. Nature 469, 97–101 (2011)

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Arribere, J. A. et al. Efficient marker-free recovery of custom genetic modifications with CRISPR/Cas9 in Caenorhabditis elegans. Genetics 198, 837–846 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ingolia, N. T., Ghaemmaghami, S., Newman, J. R. S. & Weissman, J. S. Genome-wide analysis in vivo of translation with nucleotide resolution using ribosome profiling. Science 324, 218–223 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yen, H.-C. S., Xu, Q., Chou, D. M., Zhao, Z. & Elledge, S. J. Global protein stability profiling in mammalian cells. Science 322, 918–923 (2008)

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Liebhaber, S. A. & Kan, Y. W. Differentiation of the mRNA transcripts originating from the alpha 1- and alpha 2-globin loci in normals and alpha-thalassemics. J. Clin. Invest. 68, 439–446 (1981)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Torabi, N. & Kruglyak, L. Genetic basis of hidden phenotypic variation revealed by increased translational readthrough in yeast. PLoS Genet. 8, e1002546 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Steneberg, P. & Samakovlis, C. A novel stop codon readthrough mechanism produces functional Headcase protein in Drosophila trachea. EMBO Rep. 2, 593–597 (2001)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Freitag, J., Ast, J. & Bölker, M. Cryptic peroxisomal targeting via alternative splicing and stop codon read-through in fungi. Nature 485, 522–525 (2012)

    Article  ADS  CAS  PubMed  Google Scholar 

  24. Eswarappa, S. M. et al. Programmed translational readthrough generates antiangiogenic VEGF-Ax. Cell 157, 1605–1618 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. True, H. L. & Lindquist, S. L. A yeast prion provides a mechanism for genetic variation and phenotypic diversity. Nature 407, 477–483 (2000)

    Article  ADS  CAS  PubMed  Google Scholar 

  26. Waterston, R. H. A second informational suppressor, sup-7 X, in Caenorhabditis elegans. Genetics 97, 307–325 (1981)

    Article  CAS  PubMed  Google Scholar 

  27. Laski, F. A., Ganguly, S., Sharp, P. A., RajBhandary, U. L. & Rubin, G. M. Construction, stable transformation, and function of an amber suppressor tRNA gene in Drosophila melanogaster. Proc. Natl Acad. Sci. USA 86, 6696–6698 (1989)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hudziak, R. M., Laski, F. A., RajBhandary, U. L., Sharp, P. A. & Capecchi, M. R. Establishment of mammalian cell lines containing multiple nonsense mutations and functional suppressor tRNA genes. Cell 31, 137–146 (1982)

    Article  CAS  PubMed  Google Scholar 

  29. Brenner, S. The genetics of Caenorhabditis elegans. Genetics 77, 71–94 (1974)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Okkema, P. G., Harrison, S. W., Plunger, V., Aryana, A. & Fire, A. Sequence requirements for myosin gene expression and regulation in Caenorhabditis elegans. Genetics 135, 385–404 (1993)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Granato, M., Schnabel, H. & Schnabel, R. pha-1, a selectable marker for gene transfer in C. elegans. Nucleic Acids Res. 22, 1762–1763 (1994)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mello, C. C., Kramer, J. M., Stinchcomb, D. & Ambros, V. Efficient gene transfer in C. elegans: extrachromosomal maintenance and integration of transforming sequences. EMBO J. 10, 3959–3970 (1991)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Stinchcomb, D. T., Shaw, J. E., Carr, S. H. & Hirsh, D. Extrachromosomal DNA transformation of Caenorhabditis elegans. Mol. Cell. Biol. 5, 3484–3496 (1985)

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Mango, S. E., Lambie, E. J. & Kimble, J. The pha-4 gene is required to generate the pharyngeal primordium of Caenorhabditis elegans. Development 120, 3019–3031 (1994)

    Article  CAS  PubMed  Google Scholar 

  35. Zhong, M. et al. Genome-wide identification of binding sites defines distinct functions for Caenorhabditis elegans PHA-4/FOXA in development and environmental response. PLoS Genet. 6, e1000848 (2010)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Venolia, L. & Waterston, R. H. The unc-45 gene of Caenorhabditis elegans is an essential muscle-affecting gene with maternal expression. Genetics 126, 345–353 (1990)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ao, W. & Pilgrim, D. Caenorhabditis elegans UNC-45 is a component of muscle thick filaments and colocalizes with myosin heavy chain B, but not myosin heavy chain A. J. Cell Biol. 148, 375–384 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hodgkin, J., Papp, A., Pulak, R., Ambros, V. & Anderson, P. A new kind of informational suppression in the nematode Caenorhabditis elegans. Genetics 123, 301–313 (1989)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hodgkin, J. A. & Brenner, S. Mutations causing transformation of sexual phenotype in the nematode Caenorhabditis elegans. Genetics 86, 275–287 (1977)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mapes, J., Chen, J.-T., Yu, J.-S. & Xue, D. Somatic sex determination in Caenorhabditis elegans is modulated by SUP-26 repression of tra-2 translation. Proc. Natl Acad. Sci. USA 107, 18022–18027 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Anderson, P. & Brenner, S. A selection for myosin heavy chain mutants in the nematode Caenorhabditis elegans. Proc. Natl Acad. Sci. USA 81, 4470–4474 (1984)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Eide, D. & Anderson, P. The gene structures of spontaneous mutations affecting a Caenorhabditis elegans myosin heavy chain gene. Genetics 109, 67–79 (1985)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. ENCODE Project Consortium. The ENCODE (ENCyclopedia Of DNA Elements) project. Science 306, 636–640 (2004)

  44. Pédelacq, J.-D., Cabantous, S., Tran, T., Terwilliger, T. C. & Waldo, G. S. Engineering and characterization of a superfolder green fluorescent protein. Nat. Biotechnol. 24, 79–88 (2006)

    Article  PubMed  CAS  Google Scholar 

  45. Hopp, T. P. et al. A short polypeptide marker sequence useful for recombinant protein identification and purification. Nat. Biotechnol. 6, 1204–1210 (1988)

    Article  CAS  Google Scholar 

  46. Field, J. et al. Purification of a RAS-responsive adenylyl cyclase complex from Saccharomyces cerevisiae by use of an epitope addition method. Mol. Cell. Biol. 8, 2159–2165 (1988)

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Stadler, M. & Fire, A. Wobble base-pairing slows in vivo translation elongation in metazoans. RNA 17, 2063–2073 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Morlan, J. D., Qu, K. & Sinicropi, D. V. Selective depletion of rRNA enables whole transcriptome profiling of archival fixed tissue. PLoS One 7, e42882 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. Nagalakshmi, U. et al. The transcriptional landscape of the yeast genome defined by RNA sequencing. Science 320, 1344–1349 (2008)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Agarwal, V., Bell, G. W., Nam, J. W. & Bartel, D. P. Predicting effective microRNA target sites in mammalian mRNAs. eLife 4, 1–38 (2015)

    Article  Google Scholar 

  52. Miller, D. M. III, Ortiz, I., Berliner, G. C. & Epstein, H. F. Differential localization of two myosins within nematode thick filaments. Cell 34, 477–490 (1983)

    Article  CAS  PubMed  Google Scholar 

  53. Thompson, O. et al. The million mutation project: a new approach to genetics in Caenorhabditis elegans. Genome Res. 23, 1749–1762 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kyte, J. & Doolittle, R. F. A simple method for displaying the hydropathic character of a protein. J. Mol. Biol. 157, 105–132 (1982)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Fire Laboratory for critical reading of the manuscript, C. Frøkjær-Jensen and K. Artiles for technical expertise, and T. Schedl, T. Inada, C. Joazeiro, L. Ling, A. Nager, and N. Spies for discussions. A. Sapiro and B. Li were instrumental in developing the RNA-seq2 protocol. This work was supported by grants from NIH R01GM37706, T32HG000044 (G.T.H.), 1DP2HD084069-01 (M.C.B.), 5F32GM112474-02 (J.A.A.), Walter and Idun Berry Foundation (E.S.C.), and NSF DGE-114747 (C.H.L.).

Author information

Authors and Affiliations

Authors

Contributions

J.A.A., E.S.C., and A.Z.F. designed C. elegans experiments. J.A.A. and E.S.C. conducted C. elegans experiments. N.J. developed the RNA-seq2 protocol. J.A.A. performed computational analyses. J.A.A. conducted experiments in human cell lines, as designed and aided by J.A.A., G.T.H., C.H.L., M.C.B., and A.Z.F. J.A.A. and A.Z.F. wrote the paper with help from all authors.

Corresponding author

Correspondence to Andrew Z. Fire.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Sequencing data are available at Sequence Read Archive (SRP064516).

Reviewer Information Nature thanks J. S. Butler, M. Yarus and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Distribution of C-terminal extensions upon stop codon readthrough.

Annotations and genomes were as described in Supplementary Methods. Each 3′ UTR was translated starting one codon after the stop codon until the next in-frame stop codon. For metazoans, counting was performed in three different ways: including only genes for which exactly one 3′ UTR was annotated (blue), counting each annotated 3′ UTR separately (green), or counting each gene once and splitting gene counts with multiple 3′ UTRs equally amongst the 3′ UTR isoforms (red). ‘Nonstop’ denotes 3′ UTRs for which no stop codon was encountered before the poly(A) tail. For each species the distribution of next in-frame stop codons was calculated for 1,000 nucleotide shuffling of 3′ UTR sequences for genes with a single 3′ UTR annotated, and 95% confidence interval shown (yellow). A similar ‘randomized’ distribution was obtained upon shuffling 3′ UTR sequences and preserving dinucleotide frequency. The frequency of stops immediately after the annotated stop codon (amino acid length 0) is highlighted with a blue arrow in each species. The distribution of peptide lengths follows an exponential decay curve, where the slope is related to the probability of encountering a stop codon at each position. In the simplest model, the probability of encountering a stop codon is constant throughout the 3′ UTR, accounting for the roughly linear shape of each plot (previously noted2,3). Notable exceptions are a tendency towards second in-frame stops in E. coli (blue arrow), and a tendency towards peptides >60 amino acids in length in all species. In E. coli, the enrichment towards longer downstream peptides is at least partially explained by the operonic layout of genes.

Extended Data Figure 2 Example quantification of the GFP:mCherry fluorescence ratios of images.

Images were taken under a broad excitation and emission filter to allow for simultaneous capture of GFP and mCherry fluorescence. Intensities of each pixel in the red and green channels were extracted in python. Unfiltered pixel intensities are shown as black dots. Pixels were filtered, background subtracted, and linear regression performed (red dots and line, see Methods). For simplicity, the green–red intensities from 1,000 random pixels are shown. The GFP:mCherry fluorescence ratio was taken as the slope of the linear regression line. 10 × objective.

Extended Data Figure 3 Readthrough regions confer a loss of superfolder GFP fluorescence.

Each of the indicated TerByP regions were inserted downstream of superfolder (sf) GFP, upstream of the let-858 3′ UTR. TerByP is the region after the annotated stop codon, up to and including the first in-frame stop codon in the 3′ UTR. Quantification was performed as described in Extended Data Fig. 2.

Extended Data Figure 4 Explanation of ‘shuffle’ sequences.

Trinucleotide codons from each TerByP region are colour-coded by gene (top). Codons were extracted and randomly shuffled in python. A codon was iteratively selected until a stop codon was encountered, defining shuffle1. The process was repeated twice more to define shuffle2 and shuffle3. The resulting shuffle peptides are a combination of all three TerByP regions. Lengths and colour-coding of codons for shuffle1–3 accurately reflect the sequences they are derived from.

Extended Data Figure 5 RNA-seq and ribo-seq from unc-54 mutants.

ac, RNA-seq (a) and ribosome footprint profiling (ribo-seq) (b) library mRNA counts, with summary counts (c) for the indicated strains and mRNAs. Libraries were prepared from L4 animals, as described Methods. ‘N2’ is wild type (PD1074, VC2010 (ref. 53)). unc-54(cc3389) bears a TAA (stop) to AAT (Asn) mutation, unc-54(TerByP). unc-54(e1301) has a GGA (Gly387) to AGA (Arg387) point mutation that confers a temperature-sensitive Unc phenotype with minimal discernible effects on UNC-54 protein levels. unc-54(e1301) was included as a control for the Unc phenotype of unc-54(cc3389), though e1301 confers a less severe Unc phenotype than cc3389. Values for unc-54 mRNA (blue) are highlighted throughout, and for comparison, three additional transcripts known to be at least partly expressed in the body-wall muscles are also highlighted: unc-87 (pink), unc-15 (green), and unc-22 (red).

Extended Data Figure 6 Ribo-seq of unc-54(cc3389) shows an unexceptional progression of ribosomes in the readthrough region.

a, Raw ribo-seq reads for unc-54(+) (blue) and unc-54(cc3389) (green) animals, plotted as read pile-ups. Mismatched bases are indicated with black bars. Location of the normal stop codon and the first in-frame stop codon are indicated with ‘TAA’ and dotted lines. The extension in unc-54(cc3389) is 30 amino acids. b, The number of ribo-seq reads in the last 30 codons, compared to the previous 30 codons, for all mRNAs. Linear regression was performed on all points (solid line), and twofold difference shown (dashed lines). c, The distribution of ribo-seq reads in the last 30 codons (90 nt) of unc-54(cc3389) is shown in green, and the 95% confidence interval (CI) for all open reading frames in dashed lines. d, The fraction of in-frame ribo-seq reads in the last 30 codons is plotted as a function of read counts in the last 30 codons, and unc-54(cc3389) highlighted. e, The distribution of read lengths in the last 30 codons of unc-54(cc3389), and all open reading frames (95% confidence interval, dashed lines). For bd, reads were restricted to 28, 29, 30 nt lengths. For be, a 12 nt offset was performed for the ribosomal P-site, and read counts were derived solely from the unc-54(cc3389) ribo-seq library. For c and e, a minimum 15 read counts was imposed to obtain the 95% confidence interval from ‘all genes’.

Extended Data Figure 7 Lack of general conservation of coding potential downstream of stop codons in Caenorhabditis.

Whole-genome alignment of six nematode species with C. elegans genome assembly ce10/WS220 was obtained from the UCSC genome browser. For each annotated transcript, the aligned bases from the multiple species alignment were extracted and compared to the reference (C. elegans) genome. The left plot shows summary information of the alignment centred on annotated stop codons; the right plot shows the same centred on the first in-frame stop codon in 3′ UTRs. In red is the substitution frequency, that is, the number of mismatched bases divided by the number of aligned bases at a given position. The enrichment of ‘wobble’ position mutations is apparent as an increase in substitutions at the third position of each codon in the CDS. In green is the synonymous substitution frequency, that is, for codons beginning at a given position, the fraction of mutations that yield a synonymous substitution divided by all mutations at that position (synonymous and non-synonymous). The tendency to conserve amino acids in the CDS is apparent as a green spike at every in-frame codon. The change in substitution frequency and synonymous substitution frequency about the first in-frame stop codon (right plot) is due to a tendency for NTR codons to be conserved, and for AAN/AGN/GAN codons to not be conserved in 3′ UTRs, regardless of frame.

Extended Data Figure 8 Nucleotide and amino acid composition of readthrough regions (C. elegans).

CDSs and 3′ UTRs were analysed for various sequence properties. For simplicity, only genes and 3′ UTRs for which a single 3′ UTR was annotated were considered. Similar results were obtained with genes with multiple 3′ UTRs. a, Nucleotide frequency of CDS, 3′ UTR, and TerByP (region between annotated stop codon and first in-frame stop codon). b, Frequency of amino acids in all three possible frames for the TerByP region. 3′ UTRs were translated one codon past the stop codon of the CDS until the next in-frame stop codon, with nonstop 3′ UTRs ignored. Highlighted are codons with high G content (GGN, Gly) and high T content (TTY, Phe). c, TerByP regions tend to be hydrophobic, regardless of frame. Kyte–Doolittle score was used as a measure of hydrophobicity54. To reduce noise, only TerByP regions at least 10 amino acids long were considered. P value is from Kolmogorov–Smirnov test comparing CDSs and TerByP sequences (each frame has P value < 10e-293 for this comparison). As the TerByP sequences are shorter than CDSs on average, the distribution of TerByP hydrophobicity scores will tend to have higher variance than CDSs. Random portions of CDSs were taken, length-matched to TerByP frame zero peptide lengths. This was repeated 100 times, and the 95% confidence interval is shown (dashed lines, ‘CDS rands’). d, Hydrophobicity of the inserts is correlated with a negative effect on GFP fluorescence. The GFP:mCherry fluorescence ratio (Fig. 2b) was plotted against the maximum Kyte–Doolittle score in a six amino acid window for each insert. (Similar results were obtained using the Kyte–Doolittle score averaged across the entire sequence.) Mean (circle) and s.d. (bars) are shown. 3′-UTR-derived sequences are in blue, and non-3′-UTR-derived sequences are in red. To avoid redundancy or skewing of the data, in cases where multiple constructs were present with the same peptide sequence (for example, unc-54(TerByP), unc-54(TerByP, syn1), and unc-54(TerByP, syn2)), only the first of these was used. e, Hydrophobicity analysis of the TerByP extensions obtained by CRISPR–Cas9 engineering at the unc-22 and unc-54 loci. ‘+1/-1 TerByP’ denotes the gain or loss of a nucleotide, generating a late frameshift and allowing translation to proceed past the annotated stop codon out of frame with the upstream open reading frame. In each case, Kyte–Doolittle hydropathy was used to analyse the C-terminal appendage. The least phenotypically affected strain of the three is shown in bold.

Extended Data Figure 9 Nucleotide and amino acid composition of readthrough regions (H. sapiens).

a, b, Similar analysis of hydrophobicity as in Extended Data Fig. 8c, d, performed in humans.

Extended Data Table 1 Translation into 3′ UTRs at endogenous loci tends to yield hypomorphs

Supplementary information

Supplementary Table 1

This table contains a list of strains used in the study. (XLSX 14 kb)

Supplementary Table 2

This table contains a list of plasmids used in the study. (XLSX 17 kb)

Supplementary Table 3

This table contains DNA oligos for rRNA digestion by RNaseH. (XLSX 7 kb)

Supplementary Information

This file contains supplementary text and gel source data for figure 3d. (PDF 704 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arribere, J., Cenik, E., Jain, N. et al. Translation readthrough mitigation. Nature 534, 719–723 (2016). https://doi.org/10.1038/nature18308

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18308

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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