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 efficiency of mRNAs is increased by antisense oligonucleotides targeting upstream open reading frames

Abstract

Increasing the levels of therapeutic proteins in vivo remains challenging. Antisense oligonucleotides (ASOs) are often used to downregulate gene expression1 or to modify RNA splicing2,3, but antisense technology has not previously been used to directly increase the production of selected proteins. Here we used a class of modified ASOs that bind to mRNA sequences in upstream open reading frames (uORFs) to specifically increase the amounts of protein translated from a downstream primary ORF (pORF). Using ASO treatment, we increased the amount of proteins expressed from four genes by 30–150% in a dose-dependent manner in both human and mouse cells. Notably, systemic treatment of mice with ASO resulted in an 80% protein increase of LRPPRC. The ASO-mediated increase in protein expression was sequence-specific, occurred at the level of translation and was dependent on helicase activity. We also found that the type of RNA modification and the position of modified nucleotides in ASOs affected translation of a pORF. ASOs are a useful class of therapeutic agents with broad utility.

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: Antisense ASOs targeting the uAUG region specifically increased protein production from the pORF.
Figure 2: ASO modification and length effects on RNase H1 protein level.
Figure 3: uORF ASOs increase translation in a helicase-dependent manner.
Figure 4: uORF ASOs can increase protein levels of different genes in different species or in mice.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Crooke, S.T., Vickers, T., Lima, W. & Wu, H.-J. Mechanisms of antisense drug action, an introduction. In Antisense Drug Technology - Principles, Strategies, and Application (ed. Crooke, S.T.) 3–46 (CRC Press, 2008).

  2. Hua, Y., Vickers, T.A., Baker, B.F., Bennett, C.F. & Krainer, A.R. Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol. 5, e73 (2007).

    Article  Google Scholar 

  3. Mourich, D.V. et al. Alternative splice forms of CTLA-4 induced by antisense mediated splice-switching influences autoimmune diabetes susceptibility in NOD mice. Nucleic Acid Ther. 24, 114–126 (2014).

    Article  CAS  Google Scholar 

  4. Pearson, S., Jia, H. & Kandachi, K. China approves first gene therapy. Nat. Biotechnol. 22, 3–4 (2004).

    Article  CAS  Google Scholar 

  5. Hoyng, S.A. et al. Gene therapy and peripheral nerve repair: a perspective. Front. Mol. Neurosci. 8, 32 (2015).

    Article  Google Scholar 

  6. Leader, B., Baca, Q.J. & Golan, D.E. Protein therapeutics: a summary and pharmacological classification. Nat. Rev. Drug Discov. 7, 21–39 (2008).

    Article  CAS  Google Scholar 

  7. Meng, L. et al. Towards a therapy for Angelman syndrome by targeting a long non-coding RNA. Nature 518, 409–412 (2015).

    Article  CAS  Google Scholar 

  8. Kozak, M. Pushing the limits of the scanning mechanism for initiation of translation. Gene 299, 1–34 (2002).

    Article  CAS  Google Scholar 

  9. Kozak, M. Evaluation of the “scanning model” for initiation of protein synthesis in eucaryotes. Cell 22, 7–8 (1980).

    Article  CAS  Google Scholar 

  10. Sonenberg, N. & Hinnebusch, A.G. Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136, 731–745 (2009).

    Article  CAS  Google Scholar 

  11. Kozak, M. Regulation of translation via mRNA structure in prokaryotes and eukaryotes. Gene 361, 13–37 (2005).

    Article  CAS  Google Scholar 

  12. Araujo, P.R. et al. Before it gets started: regulating translation at the 5′ UTR. Comp. Funct. Genomics 2012, 475731 (2012).

    Article  Google Scholar 

  13. Pichon, X. et al. RNA binding protein/RNA element interactions and the control of translation. Curr. Protein Pept. Sci. 13, 294–304 (2012).

    Article  CAS  Google Scholar 

  14. Xue, S. & Barna, M. Specialized ribosomes: a new frontier in gene regulation and organismal biology. Nat. Rev. Mol. Cell Biol. 13, 355–369 (2012).

    Article  CAS  Google Scholar 

  15. Wethmar, K., Smink, J.J. & Leutz, A. Upstream open reading frames: molecular switches in (patho)physiology. BioEssays: News and Reviews Mol. Cell. Dev. Biol. 32, 885–893 (2010).

    Article  CAS  Google Scholar 

  16. Barbosa, C., Peixeiro, I. & Romão, L. Gene expression regulation by upstream open reading frames and human disease. PLoS Genet. 9, e1003529 (2013).

    Article  CAS  Google Scholar 

  17. Calvo, S.E., Pagliarini, D.J. & Mootha, V.K. Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc. Natl. Acad. Sci. USA 106, 7507–7512 (2009).

    Article  CAS  Google Scholar 

  18. Lee, S. et al. Global mapping of translation initiation sites in mammalian cells at single-nucleotide resolution. Proc. Natl. Acad. Sci. USA 109, E2424–E2432 (2012).

    Article  CAS  Google Scholar 

  19. Suzuki, Y. et al. An upstream open reading frame and the context of the two AUG codons affect the abundance of mitochondrial and nuclear RNase H1. Mol. Cell. Biol. 30, 5123–5134 (2010).

    Article  CAS  Google Scholar 

  20. Liang, X.H., Sun, H., Shen, W. & Crooke, S.T. Identification and characterization of intracellular proteins that bind oligonucleotides with phosphorothioate linkages. Nucleic Acids Res. 43, 2927–2945 (2015).

    Article  CAS  Google Scholar 

  21. Deng, Y. et al. Therapeutic potentials of gene silencing by RNA interference: principles, challenges, and new strategies. Gene 538, 217–227 (2014).

    Article  CAS  Google Scholar 

  22. Freier, S.M. & Altmann, K.H. The ups and downs of nucleic acid duplex stability: structure-stability studies on chemically-modified DNA:RNA duplexes. Nucleic Acids Res. 25, 4429–4443 (1997).

    Article  CAS  Google Scholar 

  23. Seth, P.P. et al. Synthesis and biophysical evaluation of 2′,4′-constrained 2′O-methoxyethyl and 2′,4′-constrained 2′O-ethyl nucleic acid analogues. J. Org. Chem. 75, 1569–1581 (2010).

    Article  CAS  Google Scholar 

  24. Ling, J., Morley, S.J. & Traugh, J.A. Inhibition of cap-dependent translation via phosphorylation of eIF4G by protein kinase Pak2. EMBO J. 24, 4094–4105 (2005).

    Article  CAS  Google Scholar 

  25. Chiang, M.Y. et al. Antisense oligonucleotides inhibit intercellular adhesion molecule 1 expression by two distinct mechanisms. J. Biol. Chem. 266, 18162–18171 (1991).

    CAS  PubMed  Google Scholar 

  26. Pisareva, V.P., Pisarev, A.V., Komar, A.A., Hellen, C.U. & Pestova, T.V. Translation initiation on mammalian mRNAs with structured 5′UTRs requires DExH-box protein DHX29. Cell 135, 1237–1250 (2008).

    Article  CAS  Google Scholar 

  27. Koller, E. et al. Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes. Nucleic Acids Res. 39, 4795–4807 (2011).

    Article  CAS  Google Scholar 

  28. Wethmar, K. The regulatory potential of upstream open reading frames in eukaryotic gene expression. Wiley Interdiscip. Rev. RNA 5, 765–768 (2014).

    Article  CAS  Google Scholar 

  29. Postina, R. et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J. Clin. Invest. 113, 1456–1464 (2004).

    Article  CAS  Google Scholar 

  30. Liang, X.H., Liu, Q. & Fournier, M.J. rRNA modifications in an intersubunit bridge of the ribosome strongly affect both ribosome biogenesis and activity. Mol. Cell 28, 965–977 (2007).

    Article  CAS  Google Scholar 

  31. Liang, X.H., Hart, C.E. & Crooke, S.T. Transfection of siRNAs can alter miRNA levels and trigger non-specific protein degradation in mammalian cells. Biochim. Biophys. Acta 1829, 455–468 (2013).

    Article  CAS  Google Scholar 

  32. Bleriot, Y. et al. Synthesis and acid catalyzed hydrolysis of B2,5 type conformationally constrained glucopyranosides: incorporation into a cellobiose analog. Tetrahedron 60, 6813–6828 (2004).

    Article  CAS  Google Scholar 

  33. Liang, X.H., Liu, Q. & Michaeli, S. Small nucleolar RNA interference induced by antisense or double-stranded RNA in trypanosomatids. Proc. Natl. Acad. Sci. USA 100, 7521–7526 (2003).

    Article  CAS  Google Scholar 

  34. Liang, X.H., Vickers, T.A., Guo, S. & Crooke, S.T. Efficient and specific knockdown of small non-coding RNAs in mammalian cells and in mice. Nucleic Acids Res. 39, e13 (2011).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by Ionis Pharmaceuticals internal funding. We thank M. Ramesh for technique assistance; H. Murray and C.L. De Hoyos for help in the animal study; S. Damle for microarray data processing and deposition; T. Reigle for help in figure preparation; and F. Bennett, E. Swayze, W. Lima and S. Wang for discussions.

Author information

Authors and Affiliations

Authors

Contributions

X.L., W.S. and S.T.C. designed the research; X.L., W.S., H.S. and T.A.V. performed the experiments; and all authors analyzed the data. M.T.M. provided ASOs and measured Tm. X.L. and S.T.C. wrote the manuscript.

Corresponding author

Correspondence to Xue-hai Liang.

Ethics declarations

Competing interests

All authors are employees of Ionis Pharmaceuticals. Patents related to this study have been applied for (S.T.C., X.L., and W.S., patent application number: PCT/US2015/060938).

Integrated supplementary information

Supplementary Figure 1 ASOs targeting RNase H1 uORF can specifically increase RNase H1 protein levels and activity.

(a) qRT-PCR analyses for RNase H1 mRNA levels in HeLa cells transfected for 24 hr with different uORF ASOs, as used in Figure 1b. (b) qRT-PCR analyses for U16 RNA levels in HeLa cells treated for 24 hr with uORF ASO, followed by transfection of gapmer ASO462026 targeting U16 RNA for 4 hr. (c) qRT-PCR analyses for NCL1 mRNA levels in HeLa cells treated for 24 hr with uORF ASO, followed by transfection of gapmer ASO110074 targeting NCL1 mRNA for 4 hr, as in panel (b). The error bars for qRT-PCR panels are standard deviation of three independent experiments. (d) Western analysis of RNase H1 protein in HEK293 cells treated with ASO761909. The relative ratios of RNase H1 normalized to loading control protein Ku70 are listed below the lanes.

Supplementary Figure 2 The increase of RNase H1 protein is specific to the uORF ASO.

(a) qRT-PCR analyses for RNaseH1 mRNA levels in HeLa cells treated with or without uORF ASO761909 and complementary ASO761929, as used in Figure 1f. (b) Equal amount of S35-methionine labeled HeLa cell lysates treated with ASO761909 was analyzed by SDS-PAGE, and visualized by autoradiography. (c) Western analyses for the levels of eIF2a and phosphorylated eIF2a (Ser51) proteins in HeLa cells treated or not treated with 25 nM uORF ASOs for 16 hr. (d) Predicted off-target genes with imperfect base-pairing with ASO761909, as searched using Blast. (e) qRT-PCR results for the level of mRNAs in control cells or cells treated with 25 nM ASO761909 for 16 hr. Error bars indicate standard deviations of three independent experiments. (f) Western analysis for RNase H1, SPPL2B, and FGFR1 proteins in HeLa cells treated for different times with the uORF ASO. Ku70 served as a loading control.

Supplementary Figure 3 Effects of chemical modification on uORF ASO activity.

(a) Western for RNase H1 protein in HEK293 cells treated with uORF ASO for different times. (b) and (c) RNaseH1 protein levels in HEK293 cells tend to reduce by 16-mer and 20-mer PO/MOE ASOs, respectively, as determined by western analyses. The relative RNase H1 protein levels are listed below the lanes. (d) qRT-PCR analyses of RNase H1 mRNA levels in cells transfected with PO/MOE ASOs, as in panels (b) and (c). The error bars are standard deviations of three independent experiments.

Supplementary Figure 4 The RNase H1 uORF ASO did not act at mRNA level.

(a) qRT-PCR analysis for pre-mRNA levels of RNase H1 in cells treated with 25 nM ASOs for 16 hr. (b) The RNase H1 uORF ASO did not affect pre-mRNA splicing of RNase H1. RNase H1 mRNA in different test cells was detected by reverse-transcription-PCR, using primers specific to exon-exon junctions, followed by separation of PCR products in a 2% Agarose gel. PTEN mRNA was detected and served as a control for loading. The arrows indicate the specific PCR products. (c) qRT-PCR analyses for different RNAs in cytoplasmic and nuclear fractions prepared from cells treated with or without uORF ASO761909. Drosha and Malat1 RNAs served as control for cytoplasmic and nuclear RNAs, respectively. The error bars are standard deviations of three independent experiments. (d) Northern hybridization of RNase H1 mRNA after cleavage using specific oligonucleotides. The positions of oligonucleotides in RNase H1 3’ UTR region are depicted in upper panel. The arrows indicate the full length of the 3’ fragment after cleavage using different oligonucleotides. XL564 was used as probe for hybridization.

Supplementary Figure 5 RNase H1 mRNA shifted towards polysomes by uORF ASO treatment.

RNA was prepared from different fractions and subjected to qRT-PCR analysis, using primer probe sets specific to 28S rRNA (upper panel), RNase H1 mRNA (middle panel), and PTEN mRNA (lower panel), respectively. The percentages of each fraction are plotted. The error bars are standard deviation of 3 experiments. The 80S mono-ribosome region (Fractions 9-12) and polysome regions (fractions 15-25) are indicated by blue and red lines, respectively. The increased levels of RNase H1 mRNA in polysomes are marked with an arrow.

Supplementary Figure 6 RNase H1 uORF ASO can increase translation in a dual luciferase reporter assay.

(a) Schematic representation of the dual luciferase reporter system. The expression directions are indicated by arrows. (b) A uAUG→uUUG single nucleotide mutation. (c) A 7-nt mutation of the RNase H1 mRNA sequence and compensatory mutation of an ASO. The uAUG region is highlighted, and the mutated nucleotides are in red. (d) Luciferase activity assay for the effect of uAUG→uUUG mutation and ASO-binding site 7-nt mutations. The reporter constructs were transfected into HEK293 cells for 24 hr, and luciferase activity was measured. (e) uORF ASO761909 had no significant effect on luciferase activity for the uUUG reporter. The uUUG mutant reporter was transfected into HEK293 cells for 24 hr, followed by ASO treatment for an additional 16 hr. All the Luciferase experiments were repeated more than five times and representative results are shown. The error bars are standard deviation of three independent experiments. (f) Schematic depiction of the reporters with stepwise mutations (in red) and the ASO761909 base-pair patterns. The RNA mismatch nts with the ASO are underlined. (g) Luciferase activity assay for the effects of stepwise mutations in the reporters. The plasmids PXL59 (WT), PXL63 (M1), PXL64 (M2), PXL65 (M3), or PXL66 (M4) were transfected into HEK293 cells for 24 hr and ASO761909 was transfected at 40 nM for an additional 15 hr. The luciferase activity relative to non-ASO transfected control cells expressing the same reporter was plotted. The experiment with multiple duplicates was repeated five times and representative results are shown. The error bars are standard deviations of four independent experiments. (h) Schematic depiction of the base-pairing patterns between the reporters with stepwise mutations (in red) and ASO XL695. The blue letters in the ASO are compensatory mutations relative to ASO761909. The RNA mismatch nts with the ASO are underlined. (i) Luciferase activity assay for the reporters, as described in panel g, but treated with 40 nM ASO XL695 for 15 hr. The luciferase activity relative to non-ASO transfected control cells expressing the same reporter was plotted. The experiment with multiple duplicates was repeated five times and representative results are shown. The error bars represent standard deviation of four independent experiments. P-values were calculated based on unpaired t-test. *: P<0.05; **: P<0.01; ***: P<0.001; ****: P<0.0001. NS, not significant.

Supplementary Figure 7 uORF ASO effect on other targeted proteins.

(a) The uORF characteristics of four mRNAs of selected human and mouse genes. nt, nucleotide. aa, amino acid. (b) The 5’ UTR sequence of human SFXN3 mRNA. The uORF ASO position and the uORF sequence are shown. (c) qRT-PCR analyses of human SFXN3 mRNA in cells transfected with the uORF ASO. (d) An ASO with 2-nt mismatches (underlined) at the uAUG region significantly reduce the upregulation activity, as determined by western. Tubulin served as a loading control. The relative levels of SFXN3 are listed below the lanes.

Supplementary Figure 8 Mouse MRPL11 mRNA contains an uORF.

(a) The 5’ UTR sequence of mouse MRPL11 mRNA. The uORF ASO position is shown. (b) The mRNA levels of mouse MRPL11 in cells transfected with the uORF ASO were determined by qRT-PCR. The error bars are standard deviations of three independent experiments. (c) Luciferase activity assay for cells transiently transfected with a dual luciferase reporter construct, followed by treatment with different ASOs. The error bars are standard deviation of four independent experiments.

Supplementary Figure 9 Mouse LRPPRC protein was increased by uORF ASOs in vitro and in vivo.

(a) The 5’ UTR sequence of mouse LRPPRC mRNA. The position of the uORF ASO is shown. (b) Western analyses for LRPRPC protein levels in MHT cells treated for 10 hr with an 18-mer PO/Me ASO. ANXA2 served as a loading control. The relative LRPPRC protein levels are listed below the lanes. (c) qRT-PCR analyses for LRPPRC mRNA levels in MHT cells treated with uORF ASO761930. The error bars are standard deviations of three independent experiments. (d) Western analyses for LRPPRC protein in mouse liver homogenates treated with a control ASO or saline (N=3). hnRNP K served as a loading control. (e) Western analyses of LRPPRC in liver homogenates from mice (N=3) administrated twice with different doses of the uORF ASO. hnRNP K served as a loading control.

Supplementary Figure 10 Full length blot images presented in Figure 1.

The protein bands shown in Figure 1 are indicated by arrows.

Supplementary Figure 11 Full length blot images presented in Figure 2.

The protein bands shown in Figure 2 are indicated by arrows. The X in panel b indicates a duplicate sample that was not included in main figure.

Supplementary Figure 12 Full length blot images presented in Figure 3.

The protein bands shown in Figure 3 are indicated by arrows. The X in panel b indicates a positive control sample (CHX, cycloheximide treatment) that was not included in main figure.

Supplementary Figure 13 Full length blot images presented in Figure 4.

The protein bands shown in Figure 4 are indicated by arrows. The X in different panels indicates either different controls or at very high ASO doses that was not included in main figure, to increase conciseness. In panel c, the control sample used in main figure has higher level than the excluded control sample, thus the levels of increase for ASO treated samples are more conservative. In panel d, the untreated control was excluded since the cells in this sample were not mock transfected. In panel f, the LRPPRC band was confirmed using a different antibody.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13 and Supplementary Tables 1 and 2 (PDF 2606 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liang, Xh., Shen, W., Sun, H. et al. Translation efficiency of mRNAs is increased by antisense oligonucleotides targeting upstream open reading frames. Nat Biotechnol 34, 875–880 (2016). https://doi.org/10.1038/nbt.3589

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3589

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