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:

Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells

Abstract

Post-transcriptional modification of RNA nucleosides occurs in all living organisms. Pseudouridine, the most abundant modified nucleoside in non-coding RNAs1, enhances the function of transfer RNA and ribosomal RNA by stabilizing the RNA structure2,3,4,5,6,7,8. Messenger RNAs were not known to contain pseudouridine, but artificial pseudouridylation dramatically affects mRNA function—it changes the genetic code by facilitating non-canonical base pairing in the ribosome decoding centre9,10. However, without evidence of naturally occurring mRNA pseudouridylation, its physiological relevance was unclear. Here we present a comprehensive analysis of pseudouridylation in Saccharomyces cerevisiae and human RNAs using Pseudo-seq, a genome-wide, single-nucleotide-resolution method for pseudouridine identification. Pseudo-seq accurately identifies known modification sites as well as many novel sites in non-coding RNAs, and reveals hundreds of pseudouridylated sites in mRNAs. Genetic analysis allowed us to assign most of the new modification sites to one of seven conserved pseudouridine synthases, Pus1–4, 6, 7 and 9. Notably, the majority of pseudouridines in mRNA are regulated in response to environmental signals, such as nutrient deprivation in yeast and serum starvation in human cells. These results suggest a mechanism for the rapid and regulated rewiring of the genetic code through inducible mRNA modifications. Our findings reveal unanticipated roles for pseudouridylation and provide a resource for identifying the targets of pseudouridine synthases implicated in human disease11,12,13.

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: Genome-wide pseudouridine sequencing with single-nucleotide resolution.
Figure 2: Yeast mRNAs and ncRNAs are inducibly pseudouridylated.
Figure 3: Mechanisms of mRNA pseudouridylation.
Figure 4: Regulated pseudouridylation of human RNAs.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Data have been deposited in NCBI’s Gene Expression Omnibus (GEO), accession number GSE58200.

References

  1. Davis, F. F. & Allen, F. W. Ribonucleic acids from yeast which contain a fifth nucleotide. J. Biol. Chem. 227, 907–915 (1957)

    CAS  PubMed  Google Scholar 

  2. Arnez, J. G. & Steitz, T. A. Crystal structure of unmodified tRNAGln complexed with glutaminyl-tRNA synthetase and ATP suggests a possible role for pseudo-uridines in stabilization of RNA structure. Biochemistry 33, 7560–7567 (1994)

    Article  CAS  PubMed  Google Scholar 

  3. Charette, M. & Gray, M. W. Pseudouridine in RNA: what, where, how, and why. IUBMB Life 49, 341–351 (2000)

    Article  CAS  PubMed  Google Scholar 

  4. Davis, D. R. & Poulter, C. D. 1H–15N NMR studies of Escherichia coli tRNAPhe from hisT mutants: a structural role for pseudouridine. Biochemistry 30, 4223–4231 (1991)

    Article  CAS  PubMed  Google Scholar 

  5. Davis, D. R., Veltri, C. A. & Nielsen, L. An RNA model system for investigation of pseudouridine stabilization of the codon-anticodon interaction in tRNALys, tRNAHis and tRNATyr. J. Biomol. Struct. Dyn. 15, 1121–1132 (1998)

    Article  CAS  PubMed  Google Scholar 

  6. Hall, K. B. & Mclaughlin, L. W. Properties of a U1/mRNA 5′ splice site duplex containing pseudouridine as measured by thermodynamic and NMR methods. Biochemistry 30, 1795–1801 (1991)

    Article  CAS  PubMed  Google Scholar 

  7. Hudson, G. A., Bloomingdale, R. & Znosko, B. Thermodynamic contribution and nearest-neighbor parameters of pseudouridine-adenosine base pairs in oligoribonucleotides. RNA 19, 1474–1482 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yarian, C. S. et al. Structural and functional roles of the N1- and N3-protons of psi at tRNA’s position 39. Nucleic Acids Res. 27, 3543–3549 (1999)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Fernández, I. S. et al. Unusual base pairing during the decoding of a stop codon by the ribosome. Nature 500, 107–110 (2013)

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  10. Karijolich, J. & Yu, Y.-T. Converting nonsense codons into sense codons by targeted pseudouridylation. Nature 474, 395–398 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bykhovskaya, Y., Casas, K., Mengesha, E., Inbal, A. & Fischel-Ghodsian, N. Missense mutation in pseudouridine synthase 1 (PUS1) causes mitochondrial myopathy and sideroblastic anemia (MLASA). Am. J. Hum. Genet. 74, 1303–1308 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Heiss, N. S. et al. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nature Genet. 19, 32–38 (1998)

    Article  CAS  PubMed  Google Scholar 

  13. Mei, Y.-P. et al. Small nucleolar RNA 42 acts as an oncogene in lung tumorigenesis. Oncogene 31, 2794–2804 (2012)

    Article  CAS  PubMed  Google Scholar 

  14. Cantara, W. A. et al. The RNA Modification Database, RNAMDB: 2011 update. Nucleic Acids Res. 39, D195–D201 (2011)

    Article  CAS  PubMed  Google Scholar 

  15. Chen, L. Characterization and comparison of human nuclear and cytosolic editomes. Proc. Natl Acad. Sci. USA 110, E2741–E2747 (2013)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  16. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012)

    Article  CAS  PubMed  ADS  Google Scholar 

  17. Li, J. B. et al. Genome-wide identification of human RNA editing sites by parallel DNA capturing and sequencing. Science 324, 1210–1213 (2009)

    Article  CAS  PubMed  ADS  Google Scholar 

  18. Meyer, K. D. et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 149, 1635–1646 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Squires, J. E. et al. Widespread occurrence of 5-methylcytosine in human coding and non-coding RNA. Nucleic Acids Res. 40, 5023–5033 (2012)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  20. Bakin, A. & Ofengand, J. Four newly located pseudouridylate residues in Escherichia coli 23S ribosomal RNA are all at the peptidyltransferase center: analysis by the application of a new sequencing technique. Biochemistry 32, 9754–9762 (1993)

    Article  CAS  PubMed  Google Scholar 

  21. Wu, G., Xiao, M., Yang, C. & Yu, Y.-T. U2 snRNA is inducibly pseudouridylated at novel sites by Pus7p and snR81 RNP. EMBO J. 30, 79–89 (2011)

    Article  PubMed  CAS  Google Scholar 

  22. Ansmant, I., Massenet, S., Grosjean, H., Motorin, Y. & Branlant, C. Identification of the Saccharomyces cerevisiae RNA:pseudouridine synthase responsible for formation of Ψ2819 in 21S mitochondrial ribosomal RNA. Nucleic Acids Res. 28, 1941–1946 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Arluison, V., Buckle, M. & Grosjean, H. Pseudouridine synthetase Pus1 of Saccharomyces cerevisiae: kinetic characterisation, tRNA structural requirement and real-time analysis of its complex with tRNA. J. Mol. Biol. 289, 491–502 (1999)

    Article  CAS  PubMed  Google Scholar 

  24. Becker, H. F., Motorin, Y., Sissler, M., Florentz, C. & Grosjean, H. Major identity determinants for enzymatic formation of ribothymidine and pseudouridine in the TΨ-loop of yeast tRNAs. J. Mol. Biol. 274, 505–518 (1997)

    Article  CAS  PubMed  Google Scholar 

  25. Behm-Ansmant, I. et al. The Saccharomyces cerevisiae U2 snRNA:pseudouridine-synthase Pus7p is a novel multisite-multisubstrate RNA:Ψ-synthase also acting on tRNAs. RNA 9, 1371–1382 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kudla, G., Murray, A. W., Tollervey, D. & Plotkin, J. B. Coding-sequence determinants of gene expression in Escherichia coli. Science 324, 255–258 (2009)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  27. Shah, P., Ding, Y., Niemczyk, M., Kudla, G. & Plotkin, J. B. Rate-limiting steps in yeast protein translation. Cell 153, 1589–1601 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Somogyi, P., Jenner, A. J., Brierley, I. & Inglis, S. C. Ribosomal pausing during translation of an RNA pseudoknot. Mol. Cell. Biol. 13, 6931–6940 (1993)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jambhekar, A. & Derisi, J. L. Cis-acting determinants of asymmetric, cytoplasmic RNA transport. RNA 13, 625–642 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tan, X. et al. Tiling genomes of pathogenic viruses identifies potent antiviral shRNAs and reveals a role for secondary structure in shRNA efficacy. Proc. Natl Acad. Sci. USA 109, 869–874 (2012)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  31. Longtine, M. S. et al. Additional modules for versatile and economical PCR-based gene deletion and modification in Saccharomyces cerevisiae. Yeast 14, 953–961 (1998)

    Article  CAS  PubMed  Google Scholar 

  32. Winzeler, E. A. et al. Functional characterization of the S. cerevisiae genome by gene deletion and parallel analysis. Science 285, 901–906 (1999)

    Article  CAS  PubMed  Google Scholar 

  33. Collart, M. A. & Oliviero, S. Preparation of yeast RNA. Curr. Prot. Mol. Biol. Ch. 13, Unit–13.12 (2001)

    Google Scholar 

  34. Sambrook, J. & Russell, D. W. Molecular Cloning (Cold Spring Harbor Laboratory Press, 2001)

    Google Scholar 

  35. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011)

    Article  Google Scholar 

  36. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009)

    PubMed  PubMed Central  Google Scholar 

  38. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Xu, Z. et al. Bidirectional promoters generate pervasive transcription in yeast. Nature 457, 1033–1037 (2009)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  40. Piekna-Przybylska, D., Decatur, W. A. & Fournier, M. J. New bioinformatic tools for analysis of nucleotide modifications in eukaryotic rRNA. RNA 13, 305–312 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Crooks, G. E., Hon, G., Chandonia, J.-M. & Brenner, S. E. WebLogo: a sequence logo generator. Genome Res. 14, 1188–1190 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Darty, K., Denise, A. & Ponty, Y. VARNA: interactive drawing and editing of the RNA secondary structure. Bioinformatics 25, 1974–1975 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Jühling, F. et al. tRNAdb 2009: compilation of tRNA sequences and tRNA genes. Nucleic Acids Res. 37, D159–D162 (2009)

    Article  PubMed  CAS  Google Scholar 

  44. Hunter, J. D. Matplotlib: A 2D graphics environment. Comput. Sci. Eng. 9, 90–95 (2007)

    Article  Google Scholar 

Download references

Acknowledgements

We thank I. Cheeseman, C. Burge, U. RajBhandary, D. Bartel and members of the Gilbert laboratory for comments and discussion. The sequencing was performed at the BioMicro Center under the direction of S. Levine. This work was supported by grants from The American Cancer Society – Robbie Sue Mudd Kidney Cancer Research Scholar Grant (RSG-13-396-01-RMC) and the National Institutes of Health (GM094303, GM081399) to W.V.G. T.M.C. was supported by the American Cancer Society New England Division (Ellison Foundation Postdoctoral Fellowship), and K.M.B. was supported by a Postdoctoral Fellowship (PF-13-319-01 – RMC) from the American Cancer Society. This work was supported in part by the NIH Pre-Doctoral Training Grant T32GM007287.

Author information

Authors and Affiliations

Authors

Contributions

T.M.C. and W.V.G. conceived and designed the experiments. T.M.C., M.F.R.-D., H.S., K.M.B. and W.V.G. performed the experiments. T.M.C., B.Z. and H.S. performed the bioinformatic analyses. T.M.C. and W.V.G. interpreted the results and wrote the paper with input from all authors.

Corresponding author

Correspondence to Wendy V. Gilbert.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Detection of specific snoRNA target sites by Pseudo-seq.

Pseudo-seq was performed on wild-type (n = 4), snr37Δ (n = 2), snr81Δ (n = 2), snr43Δ (n = 2) and snr49Δ (n = 2) yeast strains. Cultures were harvested at high density (a, b) or log phase (c, d). Ψs dependent on the deleted snoRNA are indicated in red. CMC-dependent peaks of reads are indicated with dashed red lines. Traces are representative of indicated number of biological replicates. a, Pseudo-seq reads in RDN25-1 (chrXII: 452221–452270) showing SNR37-dependence of 25S-Ψ2944. b, Pseudo-seq reads in RDN25-1 (chrXII: 454111–454160, left), and U2 snRNA (LSR1, chrII: 681791–681840, right) showing SNR81-dependence of 25S-Ψ1052 and U2-Ψ42. c, Pseudo-seq reads in RDN58-1 (chrXII: 455466–455515) showing SNR43-dependence of 5.8S-Ψ73. SNR43-dependent 25S-Ψ960 was not consistently detected in wild type owing to an overlapping CMC-independent reverse transcription stop. d, Pseudo-seq reads in RDN18-1 (chrXII: 457361–457610) showing SNR49-dependence of 18S-Ψ302, 18S-Ψ211, and 18S-Ψ120. 25S-Ψ990 was also detected as SNR49-dependent (data not shown).

Extended Data Figure 2 Technical variations of Pseudo-seq give similar results.

ad, MetaPsi plots (left), and ROC curves (right) for various library prep conditions n = 1 for each condition. CMC-treated samples (solid) and mock-treated samples (dashed) are indicated. a, Comparison of AMV-RT (orange) and SuperScript III (blue) (0.2 M CMC; 115–130 nt, 100–115 nt fragments respectively). b, Comparison of 115–130 nt (orange), and 130–145 nt (blue) RNA fragment sizes (AMV-RT; 0.2 M CMC). c, Comparison of 0.2 M CMC (blue), and 0.4 M CMC (orange) (AMV-RT; 115–130 nt RNA). d, Comparison of shorter (orange) and longer (blue) truncated reverse transcription fragment sizes (AMV-RT; 115–130 nt RNA; 0.2 M CMC).

Extended Data Figure 3 Identification of pseudouridines in lowly expressed genes using multiple replicates.

a, Growth curves for wild-type yeast were grown in YPD. An A600 nm of 12 is indicated by the horizontal dotted line. b, c, Pseudo-seq was performed on polyA+ RNA isolated from high-density wild-type yeast strains. CMC-dependent peaks of reads are indicated with dashed red lines. b, Pseudo-seq reads from n = 4 biological replicates in a CDC39 (chrIII: 286226–286445, 12.3 average RPKMs), and c, IQG1 (chrXVI: 90655–90955, 12.4 average RPKMs) showing CDC39-Ψ6223 and IQG1-Ψ4367, respectively.

Source data

Extended Data Figure 4 Codons affected by mRNA pseudouridylation.

Pseudouridylation of mRNA preferentially affects GUA codons. Numbers of pseudouridines observed at the first (dark blue), second (blue), and third positions (light blue) of each codon are indicated.

Source data

Extended Data Figure 5 Expression levels minimally affect identification of yeast mRNAs displaying regulated pseudouridylation.

A plot of log-transformed average RPKMs in high-density versus log-phase yeast for all coding genes with a Ψ identified by Pseudo-seq n = 4 biological replicates for each condition. All genes (grey), genes with a high density induced Ψ (blue), and genes with a log phase induced Ψ (red) are indicated.

Extended Data Figure 6 Inducible pseudouridylation of ncRNAs.

a, b, Pseudo-seq was performed on wild-type (n = 4), snr81Δ (n = 2), pus1Δ (n = 2) and pus7Δ (n = 2) yeast strains grown to high density. CMC dependent peaks of reads are indicated with a dashed red line. Traces are representative indicated number of biological replicates. a, Pseudo-seq reads in U2 snRNA (LSR1; chrII: 681751–681790, left; chrII: 681769–681818, right) showing SNR81-dependence of U2-Ψ93, and PUS1-dependence of U2-Ψ56. Both are dependent on growth to high density. b, Pseudo-seq reads in U3a snoRNA (SNR17A, chrXV: 780461–780560) showing snR17A-Ψ369 (PUS7-dependent), snR17A-Ψ380, snR17A-Ψ391 and snR17A-Ψ425 (PUS1-dependent). c, Summaries of the numbers of Ψs called in ncRNAs by Pseudo-seq. Indicated are constitutive Ψs (top), and inducible Ψs (bottom).

Extended Data Figure 7 Analysis of potential snoRNA targets.

ad, Pseudo-seq was performed on wild-type yeast in log phase, or grown to high density. Reads from n = 4 biological replicate libraries for each condition were pooled. bd, Indicated are the predicted snoRNA target site (black, dashed), and the expected peak of CMC-dependent reads (black, dotted). a, b, Results of analysis on sets of random Us. a, A histogram of the differences (+CMC − −CMC) in mean normalized reads at the +1 peak position for 10,000 randomizations for high density (orange) and log phase (blue). The normalized read values for the computationally predicted Ψs in exponential and high density samples are indicated by arrows. b, An averaged metaPsi plot for all randomizations. c, d, +CMC (c) and −CMC (d) MetaPsi plots for computationally predicted Ψs separated by base pairing. Sites with 8 or more (red), 9 or more (blue), and 10 or more (orange) base pairs are indicated. Data for high density (left), and log phase (right) are indicated. e, Pseudo-seq reads for computationally predicted Ψs, CAT2 (chrXII: 193995–19450, left), and AIM6 (chrIV: 31135–31550, right). Traces are representative of six biological replicates.

Extended Data Figure 8 Mechanisms of Pus-dependent pseudouridylation.

a, b, Summaries of the PUS-dependence of called Ψs using higher stringency cut-offs (10/14 libraries) (a) and lower stringency cut-offs (9/14 libraries) (b). c, f, CMC-dependent peaks of reads are indicated with dashed red lines. Traces are representative of n = 4 (wild type), and n = 2 (pusΔ) biological replicates. Pseudo-seq reads for RPL14A (a, chrXI: 431901–432200) and PDI1 (d, chrIII: 49401–48760) showing PUS1- and PUS7-dependency, respectively. Both are dependent on growth to high density. d, e, g, h, WebLogo 3.3 was used to generate motifs for PUS1 (d), PUS2 (e), PUS7 (g) and PUS4 (h).

Extended Data Figure 9 Positive controls for human RNA Pseudo-seq.

a, Pseudo-seq reads for RDN28S5 (1516–1765) containing five known Ψs (28S-Ψ1536, 28S-Ψ1582, 28S-Ψ1677, 28S-Ψ1683 and 28S-Ψ1744). CMC-dependent peaks of reads are indicated with dashed red lines. Traces are representative of n = 5 biological replicates. b, A metaPsi plot of mean normalized reads (left axis) for +CMC libraries (orange), and −CMC libraries (blue). The number of Ψs at each position in the metaPsi window is indicated (black, right axis). c, A ROC curve of the Pseudo-seq signal for all known Ψs in rRNA and snRNA.

Extended Data Figure 10 New pseudouridines in human RNAs.

a, A Venn diagram showing the overlap of mRNA pseudouridylation events between serum-fed and serum-starved HeLa cells. b, A plot of log-transformed average RPKMs in serum-starved versus serum-fed HeLa for all coding genes with a Ψ identified by Pseudo-seq. All genes with a Ψ (grey), genes with a Ψ induced in plus serum cells (blue), and genes with a Ψ induced in serum-starved cells (red) are indicated. c, Pseudo-seq reads for RDN18S5 (184–411) (top, left), RDN18S5 (1015–1210) (top, right), RDN28S5 (2713–3108) (bottom, left), and RDN28S5 (4461–4618) (bottom, right). CMC-dependent peaks of reads are indicated with dashed red lines, and highlighted Ψs are indicated by red boxes. Traces are representative of n = 4 biological replicates.

Supplementary information

Supplementary Tables

This file contains the data for Supplementary Tables 1–9 (see separate file for description of tables). (XLSX 367 kb)

Supplementary Information

This file contains the legends for Supplementary Tables 1–9 (see separate excel file for tables). (PDF 110 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Carlile, T., Rojas-Duran, M., Zinshteyn, B. et al. Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells. Nature 515, 143–146 (2014). https://doi.org/10.1038/nature13802

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13802

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