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:

Genetically encoded chemical crosslinking of RNA in vivo

Abstract

Protein–RNA interactions regulate RNA fate and function, and defects can lead to various disorders. Such interactions have mainly been studied by nucleoside-based UV crosslinking methods, which lack broad in vivo compatibility and the ability to resolve specific amino acids. In this study we genetically encoded latent bioreactive unnatural amino acids into proteins to react with bound RNA by proximity-enabled reactivity and demonstrated genetically encoded chemical crosslinking of proteins with target RNA (GECX-RNA) in vivo. Applying GECX-RNA to the RNA chaperone Hfq in Escherichia coli identified target RNAs with amino acid specificity. Combining GECX-RNA with immunoprecipitation and high-throughput sequencing of an N6-methyladenosine reader protein in mammalian cells allowed the in vivo identification of unknown N6-methyladenosine on RNA with single-nucleotide resolution throughout the transcriptome. GECX-RNA thus affords resolution at the nucleotide and amino acid level for interrogating protein–RNA interactions in vivo. It also enables the precise engineering of covalent linkages between a protein and RNA, which will inspire innovative solutions for RNA-related research and therapeutics.

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: GECX-RNA enables FSY-incorporated dPsCas13b to crosslink target RNA in vitro.
Fig. 2: GECX-RNA enables FSY-incorporated Hfq proteins to crosslink target RNA in E. coli.
Fig. 3: GECX-RNA enables FSY-incorporated dPsCas13b proteins to crosslink target RNA in mammalian cells.
Fig. 4: Genetically encoding SFY allows crosslinking of His, Tyr and Lys residues in protein and of RNA in cells.
Fig. 5: Design of GRIP-seq for in vivo detection of m6A on RNA with single-nucleotide resolution.
Fig. 6: GRIP-seq in vivo detected m6A on RNA with single-nucleotide resolution in mammalian cells.

Similar content being viewed by others

Data availability

All GRIP-seq data are available in the Sequence Read Archive through accession number PRJNA797913. All other data generated or analysed in this study are available within the article and its Supplementary Information. Source data are provided with this paper.

Code availability

The custom code used in this study is available at https://github.com/Shall-We-Dance/GRIP-seq.

References

  1. Gerstberger, S., Hafner, M. & Tuschl, T. A census of human RNA-binding proteins. Nat. Rev. Genet. 15, 829–845 (2014).

    Article  CAS  Google Scholar 

  2. Castello, A., Fischer, B., Hentze, M. W. & Preiss, T. RNA-binding proteins in Mendelian disease. Trends Genet. 29, 318–327 (2013).

    Article  CAS  Google Scholar 

  3. Nussbacher, J. K., Batra, R., Lagier-Tourenne, C. & Yeo, G. W. RNA-binding proteins in neurodegeneration: Seq and you shall receive. Trends Neurosci. 38, 226–236 (2015).

    Article  CAS  Google Scholar 

  4. Castello, A. et al. Comprehensive identification of RNA-binding domains in human cells. Mol. Cell 60, 696–710 (2016).

    Article  Google Scholar 

  5. Benhalevy, D., Anastasakis, D. G. & Hafner, M. Proximity-CLIP provides a snapshot of protein-occupied RNA elements in subcellular compartments. Nat. Methods 15, 1074–1082 (2018).

    Article  CAS  Google Scholar 

  6. Hentze, M. W., Castello, A., Schwarzl, T. & Preiss, T. A brave new world of RNA-binding proteins. Nat. Rev. Mol. Cell Biol. 19, 327–341 (2018).

    Article  CAS  Google Scholar 

  7. Müller-McNicoll, M. & Neugebauer, K. M. How cells get the message: dynamic assembly and function of mRNA–protein complexes. Nat. Rev. Genet. 14, 275–287 (2013).

    Article  Google Scholar 

  8. Wagenmakers, A. J. M., Reinders, R. J. & van Venrooij, W. J. Cross‐linking of mRNA to proteins by irradiation of intact cells with ultraviolet light. Eur. J. Biochem. 112, 323–330 (1980).

    Article  CAS  Google Scholar 

  9. Saito, I. & Matsuura, T. Chemical aspects of UV-induced cross-linking of proteins to nucleic acids. Photoreactions with lysine and tryptophan. Acc. Chem. Res. 18, 134–141 (1985).

    Article  CAS  Google Scholar 

  10. Hafner, M. et al. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 141, 129–141 (2010).

    Article  CAS  Google Scholar 

  11. Baltz, A. G. et al. The mRNA-bound proteome and its global occupancy profile on protein-coding transcripts. Mol. Cell 46, 674–690 (2012).

    Article  CAS  Google Scholar 

  12. Licatalosi, D. D. et al. HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature 456, 464–469 (2008).

    Article  CAS  Google Scholar 

  13. König, J. et al. ICLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nat. Struct. Mol. Biol. 17, 909–915 (2010).

    Article  Google Scholar 

  14. Castello, A. et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell 149, 1393–1406 (2012).

    Article  CAS  Google Scholar 

  15. Lee, F. C. Y. & Ule, J. Advances in CLIP technologies for studies of protein–RNA Interactions. Mol. Cell 69, 354–369 (2018).

    Article  CAS  Google Scholar 

  16. Sugimoto, Y. et al. Analysis of CLIP and iCLIP methods for nucleotide-resolution studies of protein–RNA interactions. Genome Biol. 13, R67 (2012).

    Article  Google Scholar 

  17. Xiang, Z. et al. Adding an unnatural covalent bond to proteins through proximity-enhanced bioreactivity. Nat. Methods 10, 885–888 (2013).

    Article  CAS  Google Scholar 

  18. Wang, L. Genetically encoding new bioreactivity. N. Biotechnol. 38, 16–25 (2017).

    Article  Google Scholar 

  19. Coin, I. et al. Genetically encoded chemical probes in cells reveal the binding path of urocortin-I to CRF class B GPCR. Cell 155, 1258–1269 (2013).

    Article  CAS  Google Scholar 

  20. Yang, B. et al. Spontaneous and specific chemical cross-linking in live cells to capture and identify protein interactions. Nat. Commun. 8, 2240 (2017).

    Article  Google Scholar 

  21. Li, Q. et al. Developing covalent protein drugs via proximity-enabled reactive therapeutics. Cell 182, 85–97 (2020).

    Article  CAS  Google Scholar 

  22. Wang, N. et al. Genetically encoding fluorosulfate-l-tyrosine to react with lysine, histidine, and tyrosine via SuFEx in proteins in vivo. J. Am. Chem. Soc. 140, 4995–4999 (2018).

    Article  CAS  Google Scholar 

  23. Abudayyeh, O. O. et al. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science 353, aaf5573 (2016).

    Article  Google Scholar 

  24. Cox, D. B. T. et al. RNA editing with CRISPR-Cas13. Science 358, 1019–1027 (2017).

    Article  CAS  Google Scholar 

  25. Yang, L. Z. et al. Dynamic imaging of RNA in living cells by CRISPR-Cas13 systems. Mol. Cell 76, 981–997 (2019).

    Article  CAS  Google Scholar 

  26. Liu, L. et al. Two distant catalytic sites are responsible for C2c2 RNase activities. Cell 168, 121–134 (2017).

    Article  CAS  Google Scholar 

  27. Smargon, A. A. et al. Cas13b is a type VI-B CRISPR-associated RNA-guided RNase differentially regulated by accessory proteins Csx27 and Csx28. Mol. Cell 65, 618–630 (2017).

    Article  CAS  Google Scholar 

  28. Zhang, B. et al. Structural insights into Cas13b-guided CRISPR RNA maturation and recognition. Cell Res. 28, 1198–1201 (2018).

    Article  Google Scholar 

  29. Wilusz, C. J. & Wilusz, J. Eukaryotic Lsm proteins: lessons from bacteria. Nat. Struct. Mol. Biol. 12, 1031–1036 (2005).

    Article  CAS  Google Scholar 

  30. Bilusic, I., Popitsch, N., Rescheneder, P., Schroeder, R. & Lybecker, M. Revisiting the coding potential of the E. coli genome through Hfq co-immunoprecipitation. RNA Biol. 11, 641–654 (2014).

    Article  Google Scholar 

  31. Holmqvist, E. et al. Global RNA recognition patterns of post‐transcriptional regulators Hfq and CsrA revealed by UV crosslinking in vivo. EMBO J. 35, 991–1011 (2016).

    Article  CAS  Google Scholar 

  32. Chao, Y., Papenfort, K., Reinhardt, R., Sharma, C. M. & Vogel, J. An atlas of Hfq-bound transcripts reveals 3′ UTRs as a genomic reservoir of regulatory small RNAs. EMBO J. 31, 4005–4019 (2012).

    Article  CAS  Google Scholar 

  33. Wang, W., Wang, L., Wu, J., Gong, Q. & Shi, Y. Hfq-bridged ternary complex is important for translation activation of rpoS by DsrA. Nucleic Acids Res. 41, 5938–5948 (2013).

    Article  CAS  Google Scholar 

  34. Peng, Y., Curtis, J. E., Fang, X. & Woodson, S. A. Structural model of an mRNA in complex with the bacterial chaperone Hfq. Proc. Natl Acad. Sci. USA 111, 17134–17139 (2014).

    Article  CAS  Google Scholar 

  35. Tree, J. J., Granneman, S., McAteer, S. P., Tollervey, D. & Gally, D. L. Identification of bacteriophage-encoded anti-sRNAs in pathogenic Escherichia coli. Mol. Cell 55, 199–213 (2014).

    Article  CAS  Google Scholar 

  36. Schu, D. J., Zhang, A., Gottesman, S. & Storz, G. Alternative Hfq–sRNA interaction modes dictate alternative mRNA recognition. EMBO J. 34, 2557–2573 (2015).

    Article  CAS  Google Scholar 

  37. Hoppmann, C. & Wang, L. Proximity-enabled bioreactivity to generate covalent peptide inhibitors of p53–Mdm4. Chem. Commun. 52, 5140–5143 (2016).

    Article  CAS  Google Scholar 

  38. Liu, J. et al. Genetically encoding photocaged quinone methide to multitarget protein residues covalently in vivo. J. Am. Chem. Soc. 141, 9458–9462 (2019).

    Article  CAS  Google Scholar 

  39. Li, S. et al. Genetically encoded chemical crosslinking of carbohydrate. Nat. Chem. https://doi.org/10.1038/s41557-022-01059-z (2022).

  40. Nachtergaele, S. & He, C. Chemical modifications in the life of an mRNA transcript. Annu. Rev. Genet. 52, 349–372 (2018).

    Article  CAS  Google Scholar 

  41. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  43. Linder, B. et al. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat. Methods 12, 767–772 (2015).

    Article  CAS  Google Scholar 

  44. Xu, C. et al. Structural basis for the discriminative recognition of N6-methyladenosine RNA by the human YT521-B homology domain family of proteins. J. Biol. Chem. 290, 24902–24913 (2015).

    Article  CAS  Google Scholar 

  45. Meyer, K. D. DART-seq: an antibody-free method for global m6A detection. Nat. Methods 16, 1275–1280 (2019).

    Article  CAS  Google Scholar 

  46. Van Nostrand, E. L. et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat. Methods 13, 508–514 (2016).

    Article  Google Scholar 

  47. Lovci, M. T. et al. Rbfox proteins regulate alternative mRNA splicing through evolutionarily conserved RNA bridges. Nat. Struct. Mol. Biol. 20, 1434–1442 (2013).

    Article  CAS  Google Scholar 

  48. Tang, Y. et al. m6A-Atlas: a comprehensive knowledgebase for unraveling the N6-methyladenosine (m6A) epitranscriptome. Nucleic Acids Res. 49, D134–D143 (2020).

    Article  Google Scholar 

  49. Sanchez de Groot, N. et al. RNA structure drives interaction with proteins. Nat. Commun. 10, 3246 (2019).

    Article  Google Scholar 

  50. Siegfried, N. A., Busan, S., Rice, G. M., Nelson, J. A. E. & Weeks, K. M. RNA motif discovery by SHAPE and mutational profiling (SHAPE-MaP). Nat. Methods 11, 959–965 (2014).

    Article  CAS  Google Scholar 

  51. Gruber, A. R., Lorenz, R., Bernhart, S. H., Neuböck, R. & Hofacker, I. L. The Vienna RNA websuite. Nucleic Acids Res. 36, W70–W74 (2008).

    Article  CAS  Google Scholar 

  52. Hwang, H.-W. et al. PAPERCLIP identifies microRNA targets and a role of CstF64/64tau in promoting non-canonical poly(A) site usage. Cell Rep. 15, 423–435 (2016).

    Article  CAS  Google Scholar 

  53. Kini, H. K., Silverman, I. M., Ji, X., Gregory, B. D. & Liebhaber, S. A. Cytoplasmic poly(A) binding protein-1 binds to genomically encoded sequences within mammalian mRNAs. RNA 22, 61–74 (2016).

    Article  CAS  Google Scholar 

  54. Wang, L. Engineering the genetic code in cells and animals: biological considerations and impacts. Acc. Chem. Res. 50, 2767–2776 (2017).

    Article  CAS  Google Scholar 

  55. Mackereth, C. D. & Sattler, M. Dynamics in multi-domain protein recognition of RNA. Curr. Opin. Struct. Biol. 22, 287–296 (2012).

    Article  CAS  Google Scholar 

  56. Lunde, B. M., Moore, C. & Varani, G. RNA-binding proteins: Modular design for efficient function. Nat. Rev. Mol. Cell Biol. 8, 479–490 (2007).

    Article  CAS  Google Scholar 

  57. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  CAS  Google Scholar 

  58. Baek, M. et al. Accurate prediction of protein structures and interactions using a three-track neural network. Science 373, 871–876 (2021).

    Article  CAS  Google Scholar 

  59. McMahon, A. C. et al. TRIBE: hijacking an RNA-editing enzyme to identify cell-specific targets of RNA-binding proteins. Cell 165, 742–753 (2016).

    Article  CAS  Google Scholar 

  60. Brannan, K. W. et al. Robust single-cell discovery of RNA targets of RNA-binding proteins and ribosomes. Nat. Methods 18, 507–519 (2021).

    Article  CAS  Google Scholar 

  61. Cao, L. & Wang, L. New covalent bonding ability for proteins. Protein Sci. 31, 312–322 (2021).

    Article  Google Scholar 

  62. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  Google Scholar 

  63. Chen, S., Zhou, Y., Chen, Y. & Gu, J. Fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  65. Bailey, T. L. et al. MEME suite: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–W208 (2009).

    Article  CAS  Google Scholar 

  66. Olarerin-George, A. O. & Jaffrey, S. R. MetaPlotR: a Perl/R pipeline for plotting metagenes of nucleotide modifications and other transcriptomic sites. Bioinformatics 33, 1563–1564 (2017).

    Article  CAS  Google Scholar 

  67. Lorenz, R. et al. ViennaRNA Package 2.0. Algorithms Mol. Biol. 6, 26 (2011).

    Article  Google Scholar 

Download references

Acknowledgements

L.W acknowledges the support of the NIH (R01GM118384 and R01CA258300). Y.S. acknowledges the support of the NIH (R01AG057497 and R01EY027789).

Author information

Authors and Affiliations

Authors

Contributions

W.S. designed and conducted the experiments, analysed the data and wrote the manuscript; N.W. evolved SFYRS and characterized SFY incorporation and the crosslinking of proteins; H.L. conducted the data analysis of GRIP-seq; B.Y. synthesized FSY and SFY, performed the SFY reactions with NMPs in vitro and analysed the data; L.J. helped with the dCas13b target RNA crosslinking and enrichment in mammalian cells; X.R. and Y.S. helped with the GRIP-seq experiments; L.W. conceived, directed and supported the project and wrote the manuscript.

Corresponding author

Correspondence to Lei Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemistry thanks Ryan Flynn, Stephen Fried and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Identification of the positively charged residues of PsCas13b involved in pre-crRNA cleavage.

a) Multiple sequence alignment of Cas13b proteins from different species (Bzo: Bergeyella zoohelcum, Psp: Prevotella sp. P5-125, Pgu: Porphyromonas gingivalis, Pbu: Prevotella buccae, and Ran: Riemerella anatipestifer) for β-sheets 5 and 6 involved in pre-crRNA cleavage. The secondary structure of BzoCas13b is shown above the sequence28. Identical and similar residues are highlighted in red and white boxes, respectively. Positive charged catalytic residues in BzoCas13b involved in the pre-crRNA cleavage on β-sheets 5 and 6 (450 R, 452 K, 459 R) are marked with green stars on the bottom. Positive charged residues in PsCas13b located on β-sheets 5 and 6 (367 K, 370 K, 378 R, 380 R) are marked with purple squares. Multiple sequence alignment of full-length Cas13b proteins from different species is shown in Supplementary Fig. 1. b) Denaturing urea-PAGE demonstrating the pre-crRNA cleavage by dPsCas13b-WT and dPsCas13b-Ala-mutants speculatively involved in the pre-crRNA processing. dPsCas13b-WT and dPsCas13b-Ala-mutants were incubated with pre-crRNA and then separated on denaturing urea-PAGE. The Urea-gel was stained with SybrGold for fluorescent detection of RNA.

Source data

Extended Data Fig. 2 GRIP results demonstrate that site 25 of Hfq directly binds with (AAN)4 elements of rpoS RNA.

GRIP identified the binding sites of Tyr25 of Hfq on rpoS RNA in E. coli cells. Red triangles indicate cross-linking sites identified from GRIP for rpoS RNA from Hfq-25FSY expressing E. coli cells. Two examples of Sanger sequencing of clones from Hfq-25FSY sample were shown below.

Source data

Extended Data Fig. 3 Examples of GRIP-seq data for m6A identification.

Genome browser tracks of GRIP-seq data in JUN and DICER1 mRNA regions. Reverse-transcription-termination sites (RT-termination sites) from GRIP-seq were marked as yellow triangles. Known m6A sites from published datasets were marked as grey triangles.

Supplementary information

Supplementary Information

Supplementary Figs. 1–7, Tables 1–4 and source data files for the supplementary figures.

Reporting Summary

Supplementary Table 2

Sequencing statistics for GRIP-seq.

Supplementary Table 3

m6A sites identified from GRIP-seq (coordinates in hg19 human genome assembly).

Supplementary Data 1

Statistical source data for Supplementary Fig. 2b.

Supplementary Data 2

Statistical source data for Supplementary Fig. 4.

Supplementary Data 3

Statistical source data for Supplementary Fig. 5.

Supplementary Data 4

Source data for Sanger sequencing in Supplementary Fig. 2d (Ab1 files).

Supplementary Data 5

Source data for Sanger sequencing in Supplementary Fig. 2g (Ab1 files).

Supplementary Data 6

Source data for Sanger sequencing in Supplementary Fig. 7h (Ab1 files).

Source data

Source Data Fig. 1

Unprocessed gels.

Source Data Fig. 2

Unprocessed western blot and statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Uncropped images, unprocessed gels, unprocessed western blots and statistical source data.

Source Data Extended Data Fig. 1

Unprocessed gels.

Source Data Extended Data Fig. 2

Source data for Sanger sequencing (Ab1 files).

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, W., Wang, N., Liu, H. et al. Genetically encoded chemical crosslinking of RNA in vivo. Nat. Chem. 15, 21–32 (2023). https://doi.org/10.1038/s41557-022-01038-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-022-01038-4

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