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:

Tracking endogenous proteins based on RNA editing-mediated genetic code expansion

Abstract

Protein labeling approaches are important to study proteins in living cells, and genome editing tools make it possible to tag endogenous proteins to address the concerns associated with overexpression. Here we established RNA editing-mediated noncanonical amino acids (ncAAs) protein tagging (RENAPT) to site-specifically label endogenous proteins with ncAAs in living cells. RENAPT labels protein in a temporary and nonheritable manner and is not restricted by protospacer adjacent motif sequence. Using a fluorescent ncAA or ncAA with a bio-orthogonal reaction handle for subsequent dye labeling, we demonstrated that a variety of endogenous proteins can be imaged at their specific subcellular locations. In addition, two proteins can be tagged individually and simultaneously using two different ncAAs. Furthermore, endogenous ion channels and neuron-specific proteins can be real-time labeled in primary neurons. Thus, RENAPT presents a promising platform with broad applicability for tagging endogenous proteins in living cells to study their localization and functions.

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: Graphical representation showing the principle of RENAPT and evaluation of RNA editing efficiency.
Fig. 2: Site-specific post-transcriptional incorporation of ncAAs into proteins encoded by endogenous genes using RENAPT.
Fig. 3: Subcellular localization of different endogenous proteins in U2OS cells by RENAPT.
Fig. 4: Dual labeling of target proteins by RENAPT.
Fig. 5: Applications of RENAPT in super-resolution imaging.
Fig. 6: Labeling of endogenous proteins in primary mouse hippocampal neurons by RENAPT.

Similar content being viewed by others

Data availability

The authors declare that the main data supporting the study are provided within this article and its Supplementary Information. Source data are provided with this paper.

References

  1. Kim, J. & Do Heo, W. Synergistic ensemble of optogenetic actuators and dynamic indicators in cell biology. Mol. Cells 41, 809–817 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Peng, T. & Hang, H. C. Site-specific bioorthogonal labeling for fluorescence imaging of intracellular proteins in living cells. J. Am. Chem. Soc. 138, 14423–14433 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lee, K. J., Kang, D. & Park, H.-S. Site-specific labeling of proteins using unnatural amino acids. Mol. Cells 42, 386–396 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Gao, Y. et al. Plug-and-play protein modification using homology-independent universal genome engineering. Neuron 103, 583–597 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cheng, Z., Kuru, E., Sachdeva, A. & Vendrell, M. Fluorescent amino acids as versatile building blocks for chemical biology. Nat. Rev. Chem. 4, 275–290 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Los, G. V. et al. HaloTag: a novel protein labeling technology for cell imaging and protein analysis. ACS Chem. Biol. 3, 373–382 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Keppler, A. et al. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat. Biotechnol. 21, 86–89 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Gautier, A. et al. An engineered protein tag for multiprotein labeling in living cells. Chem. Biol. 15, 128–136 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Calloway, N. T. et al. Optimized fluorescent trimethoprim derivatives for in vivo protein labeling. ChemBioChem 8, 767–774 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Gallagher, S. S., Sable, J. E., Sheetz, M. P. & Cornish, V. W. An in vivo covalent TMP-tag based on proximity-induced reactivity. ACS Chem. Biol. 4, 547–556 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Adams, S. R. et al. New biarsenical ligands and tetracysteine motifs for protein labeling in vitro and in vivo: synthesis and biological applications. J. Am. Chem. Soc. 124, 6063–6076 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Martin, B. R., Giepmans, B. N., Adams, S. R. & Tsien, R. Y. Mammalian cell-based optimization of the biarsenical-binding tetracysteine motif for improved fluorescence and affinity. Nat. Biotechnol. 23, 1308–1314 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Zhou, Z. et al. Genetically encoded short peptide tags for orthogonal protein labeling by Sfp and AcpS phosphopantetheinyl transferases. ACS Chem. Biol. 2, 337–346 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Chatterjee, A., Guo, J., Lee, H. S. & Schultz, P. G. A genetically encoded fluorescent probe in mammalian cells. J. Am. Chem. Soc. 135, 12540–12543 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Arsić, A., Hagemann, C., Stajković, N., Schubert, T. & Nikić-Spiegel, I. Minimal genetically encoded tags for fluorescent protein labeling in living neurons. Nat. Commun. 13, 314 (2022).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  16. Zhang, G., Zheng, S., Liu, H. & Chen, P. R. Illuminating biological processes through site-specific protein labeling. Chem. Soc. Rev. 44, 3405–3417 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Shiraiwa, K., Cheng, R., Nonaka, H., Tamura, T. & Hamachi, I. Chemical tools for endogenous protein labeling and profiling. Cell Chem. Biol. 27, 970–985 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Bukhari, H. & Müller, T. Endogenous fluorescence tagging by CRISPR. Trends Cell Biol. 29, 912–928 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Kamiyama, R. et al. Cell-type-specific, multicolor labeling of endogenous proteins with split fluorescent protein tags in Drosophila. Proc. Natl Acad. Sci. USA 118, e2024690118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Suzuki, K. et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 540, 144–149 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhong, H. et al. High-fidelity, efficient, and reversible labeling of endogenous proteins using CRISPR-based designer exon insertion. eLife 10, e64911 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schwinn, M. K. et al. CRISPR-mediated tagging of endogenous proteins with a luminescent peptide. ACS Chem. Biol. 13, 467–474 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Uttamapinant, C. et al. Genetic code expansion enables live-cell and super-resolution imaging of site-specifically labeled cellular proteins. J. Am. Chem. Soc. 137, 4602–4605 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nikić, I. et al. Minimal tags for rapid dual‐color live‐cell labeling and super‐resolution microscopy. Angew. Chem. Int. Ed. 53, 2245–2249 (2014).

    Article  Google Scholar 

  25. Werther, P. et al. Bio-orthogonal red and far-red fluorogenic probes for wash-free live-cell and super-resolution microscopy. ACS Cent. Sci. 7, 1561–1571 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Alamudi, S. H. et al. Development of background-free tame fluorescent probes for intracellular live cell imaging. Nat. Commun. 7, 11964 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Montiel-Gonzalez, M. F., Quiroz, J. F. D. & Rosenthal, J. J. Current strategies for site-directed RNA editing using ADARs. Methods 156, 16–24 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Terns, M. P. CRISPR-based technologies: impact of RNA-targeting systems. Mol. Cell 72, 404–412 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Qu, L. et al. Programmable RNA editing by recruiting endogenous ADAR using engineered RNAs. Nat. Biotechnol. 37, 1059–1069 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Merkle, T. et al. Precise RNA editing by recruiting endogenous ADARs with antisense oligonucleotides. Nat. Biotechnol. 37, 133–138 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Abudayyeh, O. O. et al. A cytosine deaminase for programmable single-base RNA editing. Science 365, 382–386 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Huang, X. et al. Programmable C‐to‐U RNA editing using the human APOBEC 3A deaminase. EMBO J. 39, e104741 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Xu, C. et al. Programmable RNA editing with compact CRISPR–Cas13 systems from uncultivated microbes. Nat. Methods 18, 499–506 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Özcan, A. et al. Programmable RNA targeting with the single-protein CRISPR effector Cas7-11. Nature 597, 720–725 (2021).

    Article  ADS  PubMed  Google Scholar 

  36. Tong, H. et al. High-fidelity Cas13 variants for targeted RNA degradation with minimal collateral effects. Nat. Biotechnol. 41, 108–119 (2023).

    Article  CAS  PubMed  Google Scholar 

  37. Neubert, F. et al. Bioorthogonal click chemistry enables site‐specific fluorescence labeling of functional NMDA receptors for super‐resolution imaging. Angew. Chem. Int. Ed. 57, 16364–16369 (2018).

    Article  CAS  Google Scholar 

  38. Schmied, W. H., Elsässer, S. J., Uttamapinant, C. & Chin, J. W. Efficient multisite unnatural amino acid incorporation in mammalian cells via optimized pyrrolysyl tRNA synthetase/tRNA expression and engineered eRF1. J. Am. Chem. Soc. 136, 15577–15583 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Usuki, F. et al. Specific inhibition of nonsense-mediated mRNA decay components, SMG-1 or Upf1, rescues the phenotype of Ullrich disease fibroblasts. Mol. Ther. 14, 351–360 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Kervestin, S. & Jacobson, A. NMD: a multifaceted response to premature translational termination. Nat. Rev. Mol. Cell Biol. 13, 700–712 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lasalde, C. et al. Identification and functional analysis of novel phosphorylation sites in the RNA surveillance protein Upf1. Nucleic Acids Res. 42, 1916–1929 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Yamashita, A. Role of SMG-1-mediated U pf1 phosphorylation in mammalian nonsense-mediated m RNA decay. Genes Cells 18, 161–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Nikić, I., Kang, J. H., Girona, G. E., Aramburu, I. V. & Lemke, E. A. Labeling proteins on live mammalian cells using click chemistry. Nat. Protoc. 10, 780–791 (2015).

    Article  PubMed  Google Scholar 

  44. Young, D. D. & Schultz, P. G. Playing with the molecules of life. ACS Chem. Biol. 13, 854–870 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ngo, J. T. & Tirrell, D. A. Noncanonical amino acids in the interrogation of cellular protein synthesis. Acc. Chem. Res. 44, 677–685 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lee, S., Kim, J. & Koh, M. J. Recent advances in fluorescence imaging by genetically encoded non-canonical amino acids. J. Mol. Biol. 434, 167248 (2022).

    Article  CAS  PubMed  Google Scholar 

  47. Bessa-Neto, D. et al. Bioorthogonal labeling of transmembrane proteins with non-canonical amino acids unveils masked epitopes in live neurons. Nat. Commun. 12, 6715 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Meineke, B., Heimgärtner, J., Eirich, J., Landreh, M. & Elsässer, S. J. Site-specific incorporation of two ncAAs for two-color bioorthogonal labeling and crosslinking of proteins on live mammalian cells. Cell Rep. 31, 107811 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Lang, K. & Chin, J. W. Cellular incorporation of unnatural amino acids and bioorthogonal labeling of proteins. Chem. Rev. 114, 4764–4806 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Lukinavičius, G. et al. A near-infrared fluorophore for live-cell super-resolution microscopy of cellular proteins. Nat. Chem. 5, 132–139 (2013).

    Article  PubMed  Google Scholar 

  51. Jang, C.-W. et al. Histone H3.3 maintains genome integrity during mammalian development. Genes Dev. 29, 1377–1392 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Stajković, N. et al. Direct fluorescent labeling of NF186 and NaV1.6 in living primary neurons using bioorthogonal click chemistry. J. Cell Sci. 136, jcs260600 (2023).

  53. Zuo, E. et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289–292 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lei, Z. et al. Detect-seq reveals out-of-protospacer editing and target-strand editing by cytosine base editors. Nat. Methods 18, 643–651 (2021).

    Article  CAS  PubMed  Google Scholar 

  55. Castro-Chavez, F. Most used codons per amino acid and per genome in the code of man compared to other organisms according to the rotating circular genetic code. Neuroquantology 9, 500 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Yi, Z. et al. Engineered circular ADAR-recruiting RNAs increase the efficiency and fidelity of RNA editing in vitro and in vivo. Nat. Biotechnol. 40, 946–955 (2022).

    Article  CAS  PubMed  Google Scholar 

  57. Wang, J. et al. AAV-delivered suppressor tRNA overcomes a nonsense mutation in mice. Nature 604, 343–348 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Akin, E. J., Solé, L., Dib-Hajj, S. D., Waxman, S. G. & Tamkun, M. M. Preferential targeting of Nav1. 6 voltage-gated Na+ channels to the axon initial segment during development. PLoS ONE 10, e0124397 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lambert, T. J. & Waters, J. C. Navigating challenges in the application of superresolution microscopy. J. Cell Biol. 216, 53–63 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chen, C. et al. Genetic-code-expanded cell-based therapy for treating diabetes in mice. Nat. Chem. Biol. 18, 47–55 (2022).

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  61. Liu, C. C. & Schultz, P. G. Adding new chemistries to the genetic code. Annu. Rev. Biochem. 79, 413–444 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Bryson, D. I. et al. Continuous directed evolution of aminoacyl-tRNA synthetases. Nat. Chem. Biol. 13, 1253–1260 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gao, Y. et al. Inhibition of miR-15a promotes BDNF expression and rescues dendritic maturation deficits in MeCP2-deficient neurons. Stem Cells 33, 1618–1629 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Smrt, R. D. et al. MicroRNA miR-137 regulates neuronal maturation by targeting ubiquitin ligase mind bomb-1. Stem Cells 28, 1060–1070 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. To, T.-L. et al. Rational design of a GFP-based fluorogenic caspase reporter for imaging apoptosis in vivo. Cell Chem. Biol. 23, 875–882 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kluesner, M. G. et al. MultiEditR: the first tool for the detection and quantification of RNA editing from Sanger sequencing demonstrates comparable fidelity to RNA-seq. Mol. Ther. Nucleic Acids 25, 515–523 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Huang, X. et al. Fast, long-term, super-resolution imaging with Hessian structured illumination microscopy. Nat. Biotechnol. 36, 451–459 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Zhao, W. et al. Sparse deconvolution improves the resolution of live-cell super-resolution fluorescence microscopy. Nat. Biotechnol. 40, 606–617 (2022).

    Article  CAS  PubMed  Google Scholar 

  69. Collins, T. J. ImageJ for microscopy. Biotechniques 43, S25–S30 (2007).

    Article  Google Scholar 

Download references

Acknowledgements

This work was financially supported by the National Key Research and Development Program of China (2022YFA0912400), the National Natural Science Foundation of China (92253301, 22325701, U22A20332, 92156025 and 82003681), the National Science and Technology Major Project Program (2021YFA0909900 and 2022YFC3400600), China Postdoctoral Science Foundation (2020M680255) and the Beijing Natural Science Foundation (JQ20034). We acknowledge X. Zhang for assistance with high-resolution protein mass spectrometry and X. Yuan and Z. Jiang for assisting on confocal and FACS at the State Key Laboratory of Natural and Biomimetic Drugs. We acknowledge T. Chi from the ShanghaiTech University School of Life Sciences and Technology for kindly providing the CURE system and Split GFP reporter system and H. Yang from the Institute of Neuroscience for kindly providing the Cas13X RNA editor. We thank Guangzhou Computational Super-resolution Biotech for live-cell imaging by using their commercial super-resolution microscope (HIS-SIM), data acquisition, SR image reconstruction, analysis and discussion.

Author information

Authors and Affiliations

Authors

Contributions

M.H., X.L. and T.L. designed the experiments and revised the manuscript. M.H. wrote the manuscript and performed most of the experiments. M.H. performed confocal imaging and super-resolution imaging and analyzed data. Y.S. performed primary mouse hippocampal neuron culture. M.H. and X.W. performed plasmid construction, RNA-seq analysis, FACS and western blotting. W.L. assisted in confocal imaging. L.C. assisted with plasmid construction. Z.Z. performed chemical synthesis. X.S. assisted with mass spectrometry data analysis. M.N. assisted with marker protein selection. L.C. assisted in checking all the images. All authors approved the final manuscript.

Corresponding author

Correspondence to Tao Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Shigeki Kiyonaka 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 Efficiency of RNA editing.

a. The split GFP reporter system for evaluating RNA editing efficiency. The reconstituted GFP can be detected by FACS. b. Representatives of FACS plot chromatograph. The gating was initially performed based on FSC/SSC for live cells. Subsequently, a gate was applied to distinguish between positive and negative cells using control cells (transfected with scramble RNA) that were treated in the same manner on the day of each experiment. c. Schematics of nonsense-mediated mRNA decay (NMD) knockdown for enhanced RNA editing. NMD is a mechanism that degrades aberrant mRNAs containing premature nonsense codons in eukaryotes. d. SiRNA-mediated knockdown of Upf1 or SMG-1 results in a reduction in the expression of Upf1 or SMG-1 mRNA. The relative gene expression was calculated using the threshold cycle (2-ΔΔCT) method with the GAPDH mRNA as internal control. Data are presented as the mean ± SD, n = 3 independent samples. Source data are provided as a source data file. The primers are listed in Supplementary Table 1.

Source data

Extended Data Fig. 2 Transfection efficiency and editing efficiency of different sites in endogenous proteins.

a. Transfection efficiency of different sites in GRP94. The gating was initially performed based on FSC/SSC for live cells. Subsequently, a gate was applied to distinguish between positive and negative cells using control cells (transfected with scramble RNA) that were treated in the same manner on the day of each experiment. b. Editing efficiency of different sites in GRP94. c. Percentage of labeling efficiency in transfected cells. d. Western blotting analysis of stopped versus incorporates ncAA. e. Western blot analysis was performed for GRP94. HEK293 cells were transfected with plasmids (MbPylRS-AF/tRNACUA, RESCUE-S, gRNA) and grown with 250 μM TCO-L-lysine for 48 hours. The TCO-L-lysine medium was removed and the cells were washed twice with fresh medium. Then, the cells were incubated with 10 μM tetrazine dye diluted in fresh medium for 10 min at 37 °C with 5% CO2. After that, the cells were washed twice with fresh medium. The cells were lysed and centrifuged at 15000g for 30 minutes to clear the lysate. GRP94 antibody was labeled with AF488, and β-tubulin was labeled with Cy3. The fluorescence signals were visualized using Typhoon fluorescence imaging system. Lane A represents the wild-type (WT) sample, while lane B represents the PENAPT labeled sample. A protein marker (SMOBIO Technology, cat. no. PM2511) was used for size reference. Data are presented as the mean ± SD, n = 3 independent samples. Scale bar: 100 pixels. Source data are provided as a source data file.

Source data

Extended Data Fig. 3 Label of different endogenous proteins in HEK293 cells by RENAPT.

a. The mutations introduced at the post-translational (mRNA) level include GRP94Q452UAG, STX6Q212UAG, ATP1A1Q383UAG, ATP5AQ439UAG, IGF2-RQ506UAG and H3.3AQ56UAG and the corresponding proteins are localized to the endoplasmic reticulum, Golgi, cell membrane, mitochondria, lysosomes and nucleus, respectively. b. RNA editing efficiency on different sites of different protein coding mRNA. Cells were stained with Mito-Tracker Red CMXRos, Lyso-Tracker Red, ER-Tracker Red, Cell Plasma Membrane Staining Kit with DiI, Golgi-Tracker Red and Hoechst 33342, respectively. Scale bar: 20 μm. Data are presented as the mean ± SD, n = 3 independent samples. Source data are provided as a source data file.

Source data

Extended Data Fig. 4 Optimization of reaction conditions.

a. The optimal RS for introducing TCO-K. b. Concentration optimization of SiR-Tz dye. The endogenous protein ATP5AQ439UAG was used for optimization of SiR-Tz dye concentration. Scale bar: 20 μm. c. Cytotoxicity of different ncAA concentrations; d. Cytotoxicity of different plasmids transfection. The cytotoxicity of RENAPT was analyzed using a CCK-8. HEK293 cells were seeded into 96-well plates at a density of 2 × 103 cells per well. The cells were collected at 48 h transfection and cultured with 10 μL CCK-8 reagent. After incubating for 2 h at 37 °C, the absorbance at 450 nm was tested using a Microplate Reader. Source data are provided as a source data file. Data are presented as the mean ± SD, n = 3 independent samples.

Source data

Extended Data Fig. 5 Validation of selectivity and specificity of RENAPT system.

a. Subcellular localization of endogenous proteins in HEK293 cells visualized using the SiR-Tz dye. The mutations introduced at the post-translational (mRNA) level include ATP5AQ439UAG and IGF2-RQ506UAG, and the corresponding proteins are localized to the mitochondria and lysosome. Scale bar: 20 μm. b. Validation of selectivity and specificity of RENAPT system. The selectivity and specificity of RENAPT system were verified by labeling endogenous ATP5A protein in U2OS cells with different conditions. To illustrate the co-localization, the nucleus and mitochondria were stained by Hoechst 33342 and Mitochondrial Tracker-Red. Scale bar: 5 μm.

Extended Data Fig. 6 Labeling of ATP5A and GRP94 by RENAPT and antibodies.

The cells were immunostained with anti-ATP5A or anti-GRP94 primary antibody, followed by CY3-conjugated secondary antibody. Scale bar: 5 μm.

Extended Data Fig. 7 Subcellular localization of different endogenous proteins by RENAPT.

The mutations introduced at the post-transcriptional (mRNA) level include STX6Q212UAG, H3.3AQ56UAG and RAB7AQ27UAG, and the corresponding proteins are localized to the Golgi, nucleus and cell membrane, respectively. The cells were stained with Golgi-Tracker Red, Hoechst 33342 and Cell Plasma Membrane Staining Kit with DiI, respectively. Scale bar: 5 μm.

Extended Data Fig. 8 Pearson correlation coefficient (PCC) was used for quantitative analysis of colocalization.

a. Endogenous protein PCC coefficient in U2OS cells. b. PCC coefficient for dual-labeling of histone H3.3a-mCherry and histone H3.3b-eGFP. c. PCC coefficient for dual-labeling of endogenous ATP5A and IGF2-R. d. PCC plot representing the colocalization between target protein and tracker. Data are presented as the mean ± SD, n = 3 independent samples. Source data are provided as a source data file.

Source data

Extended Data Fig. 9 RENAPT method for labeling different endogenous proteins in primary hippocampal neurons.

a. Labeling different endogenous proteins in primary hippocampal neurons. 4 days after plating, primary mouse hippocampal neurons were transfected with RESCUE-S-mCherry, gRNA and NES PylRS/tRNACUAPyl. After 2 days of incubation with Anap, neurons were imaged on the confocal scanning microscope. Scale bar: 20 μm. b. The specificity of genetic incorporation was confirmed by the control group (scramble gRNA), where no fluorescence signal of SiR-Tz was detected. 8 days after plating, primary mouse hippocampal neurons were transfected with RESCUE-S, gRNA and NES PylRS/tRNACUAPyl. After 3 days of incubation with TCO-K, the neurons were labeled with the dye (SiR-Tz) and subsequently imaged using a confocal scanning microscope with a 100× oil objective. Scale bar: 5 μm. c. Distribution of Nav1.6 in primary mouse hippocampal neurons transfected with eGFP and anti-Nav1.6 immunostaining. The neurons were transfected with RESCUE-S-eGFP, gRNA and NES PylRS/tRNACUAPyl. After 3 days of incubation with TCO-K, the neurons were labeled with the dye (SiR-Tz). Then, the neurons were labeled with Nav1.6 antibody and imaged on a confocal scanning microscope. Scale bar: 5 μm.

Extended Data Fig. 10 Relevant components and sizes of the plasmid.

Plasmid 1 represents the RNA editing system, comprising primarily dPspCas13b, ADAR2dd, and gRNA. Plasmid 2 contains the essential elements of the non-natural amino acid insertion system, including orthogonal TCOK-RS and tRNA.

Supplementary information

Supplementary Information

Supplementary Tables 1–3.

Reporting Summary

Supplementary Video 1

Mitochondrial Tracker Red.

Supplementary Video 2

Mitochondrial protein ATP5A.

Supplementary Video 3

Colocalization of ATP5A and Tracker.

Supplementary Video 4

Lysosome Tracker Red.

Supplementary Video 5

Lysosome protein IGF2-R.

Supplementary Video 6

Colocalization of IGF2-R and Tracker.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data of Extended Data Fig. 2a.

Source Data Extended Data Fig. 2

Unprocessed western blots of Extended Data Fig. 2d,e.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Hao, M., Ling, X., Sun, Y. et al. Tracking endogenous proteins based on RNA editing-mediated genetic code expansion. Nat Chem Biol (2024). https://doi.org/10.1038/s41589-023-01533-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-023-01533-w

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