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:

Spatial imaging of glycoRNA in single cells with ARPLA

Abstract

Little is known about the biological roles of glycosylated RNAs (glycoRNAs), a recently discovered class of glycosylated molecules, because of a lack of visualization methods. We report sialic acid aptamer and RNA in situ hybridization-mediated proximity ligation assay (ARPLA) to visualize glycoRNAs in single cells with high sensitivity and selectivity. The signal output of ARPLA occurs only when dual recognition of a glycan and an RNA triggers in situ ligation, followed by rolling circle amplification of a complementary DNA, which generates a fluorescent signal by binding fluorophore-labeled oligonucleotides. Using ARPLA, we detect spatial distributions of glycoRNAs on the cell surface and their colocalization with lipid rafts as well as the intracellular trafficking of glycoRNAs through SNARE protein-mediated secretory exocytosis. Studies in breast cell lines suggest that surface glycoRNA is inversely associated with tumor malignancy and metastasis. Investigation of the relationship between glycoRNAs and monocyte–endothelial cell interactions suggests that glycoRNAs may mediate cell–cell interactions during the immune response.

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: Schematic illustration of glycoRNA in situ imaging using ARPLA.
Fig. 2: Evaluation of ARPLA for glycoRNA imaging in HeLa cells.
Fig. 3: Spatial distributions of glycoRNAs revealed by ARPLA in HL-60 cells.
Fig. 4: Visualization of glycoRNAs in malignant transformation using ARPLA.
Fig. 5: Visualization of glycoRNA levels during THP-1 differentiation and activation by LPS.

Similar content being viewed by others

Data availability

The data generated and analyzed during the current study are available at https://figshare.com/projects/Spatial_Imaging_of_GlycoRNA_in_single_Cells_with_ARPLA/164113. Source data are provided with this paper.

Code availability

The code generated and used for data analysis during the current study are available at https://figshare.com/projects/Spatial_Imaging_of_GlycoRNA_in_single_Cells_with_ARPLA/164113.

References

  1. Reily, C., Stewart, T. J., Renfrow, M. B. & Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. 15, 346–366 (2019).

    PubMed  PubMed Central  Google Scholar 

  2. Flynn, R. A. et al. Small RNAs are modified with N-glycans and displayed on the surface of living cells. Cell 184, 3109–3124 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Caldwell, R. M. & Flynn, R. A. Discovering glycoRNA: traditional and non-canonical approaches to studying RNA modifications. Isr. J. Chem. 63, e202200059 (2022).

  4. Suzuki, T. The expanding world of tRNA modifications and their disease relevance. Nat. Rev. Mol. Cell Biol. 22, 375–392 (2021).

    CAS  PubMed  Google Scholar 

  5. Tyagi, W., Pandey, V. & Pokharel, Y. R. Membrane linked RNA glycosylation as new trend to envision epi-transcriptome epoch. Cancer Gene Ther. https://doi.org/10.1038/s41417-022-00430-z (2022).

    Article  PubMed  Google Scholar 

  6. Helm, M. & Motorin, Y. Detecting RNA modifications in the epitranscriptome: predict and validate. Nat. Rev. Genet. 18, 275–291 (2017).

    CAS  PubMed  Google Scholar 

  7. Li, Y. et al. N6-Methyladenosine co-transcriptionally directs the demethylation of histone H3K9me2. Nat. Genet. 52, 870–877 (2020).

    CAS  PubMed  Google Scholar 

  8. Yu, S. & Kim, V. N. A tale of non-canonical tails: gene regulation by post-transcriptional RNA tailing. Nat. Rev. Mol. Cell Biol. 21, 542–556 (2020).

    CAS  PubMed  Google Scholar 

  9. Dunn, M. R., Jimenez, R. M. & Chaput, J. C. Analysis of aptamer discovery and technology. Nat. Rev. Chem. 1, 0076 (2017).

    CAS  Google Scholar 

  10. Sunbul, M. et al. Super-resolution RNA imaging using a rhodamine-binding aptamer with fast exchange kinetics. Nat. Biotechnol. 39, 686–690 (2021).

    CAS  PubMed  Google Scholar 

  11. Jou, A. F., Chou, Y. T., Willner, I. & Ho, J. A. Imaging of cancer cells and dictated cytotoxicity using aptamer-guided hybridization chain reaction (HCR)-generated G-quadruplex chains. Angew. Chem. Int. Ed. Engl. 60, 21673–21678 (2021).

    CAS  PubMed  Google Scholar 

  12. Dey, S. K. et al. Repurposing an adenine riboswitch into a fluorogenic imaging and sensing tag. Nat. Chem. Biol. 18, 180–190 (2022).

    CAS  PubMed  Google Scholar 

  13. Coonahan, E. S. et al. Structure-switching aptamer sensors for the specific detection of piperaquine and mefloquine. Sci. Transl. Med. 13, eabe1535 (2021).

    CAS  PubMed  Google Scholar 

  14. Peinetti, A. S. et al. Direct detection of human adenovirus or SARS-CoV-2 with ability to inform infectivity using DNA aptamer-nanopore sensors. Sci. Adv. 7, eabh2848 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Wu, L. et al. Aptamer-based detection of circulating targets for precision medicine. Chem. Rev. 121, 12035–12105 (2021).

    CAS  PubMed  Google Scholar 

  16. Arroyo-Curras, N. et al. Real-time measurement of small molecules directly in awake, ambulatory animals. Proc. Natl Acad. Sci. USA 114, 645–650 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Nakatsuka, N. et al. Aptamer-field-effect transistors overcome Debye length limitations for small-molecule sensing. Science 362, 319–324 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Cheung, Y. W. et al. Evolution of abiotic cubane chemistries in a nucleic acid aptamer allows selective recognition of a malaria biomarker. Proc. Natl Acad. Sci. USA 117, 16790–16798 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, Z. et al. High-affinity dimeric aptamers enable the rapid electrochemical detection of wild-type and B.1.1.7 SARS-CoV-2 in unprocessed saliva. Angew. Chem. Int. Ed. Engl. 60, 24266–24274 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Gunaratne, R. et al. Combination of aptamer and drug for reversible anticoagulation in cardiopulmonary bypass. Nat. Biotechnol. 36, 606–613 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Valero, J. et al. A serum-stable RNA aptamer specific for SARS-CoV-2 neutralizes viral entry. Proc. Natl Acad. Sci. USA 118, e2112942118 (2021).

    PubMed  PubMed Central  Google Scholar 

  22. Zhang, Y. et al. Immunotherapy for breast cancer using EpCAM aptamer tumor-targeted gene knockdown. Proc. Natl Acad. Sci. USA 118, e2022830118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Schmitz, A. et al. A SARS-CoV-2 spike binding DNA aptamer that inhibits pseudovirus infection by an RBD-independent mechanism*. Angew. Chem. Int. Ed. Engl. 60, 10279–10285 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Soderberg, O. et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000 (2006).

    PubMed  Google Scholar 

  25. Tavoosidana, G. et al. Multiple recognition assay reveals prostasomes as promising plasma biomarkers for prostate cancer. Proc. Natl Acad. Sci. USA 108, 8809–8814 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Jalili, R., Horecka, J., Swartz, J. R., Davis, R. W. & Persson, H. H. J. Streamlined circular proximity ligation assay provides high stringency and compatibility with low-affinity antibodies. Proc. Natl Acad. Sci. USA 115, E925–E933 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Dore, K. et al. SYNPLA, a method to identify synapses displaying plasticity after learning. Proc. Natl Acad. Sci. USA 117, 3214–3219 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Weibrecht, I. et al. In situ detection of individual mRNA molecules and protein complexes or post-translational modifications using padlock probes combined with the in situ proximity ligation assay. Nat. Protoc. 8, 355–372 (2013).

    CAS  PubMed  Google Scholar 

  29. Gao, X. & Hannoush, R. N. Single-cell in situ imaging of palmitoylation in fatty-acylated proteins. Nat. Protoc. 9, 2607–2623 (2014).

    CAS  PubMed  Google Scholar 

  30. tom Dieck, S. et al. Direct visualization of newly synthesized target proteins in situ. Nat. Methods 12, 411–414 (2015).

    CAS  PubMed  Google Scholar 

  31. Robinson, P. V., Tsai, C. T., de Groot, A. E., McKechnie, J. L. & Bertozzi, C. R. Glyco-seek: ultrasensitive detection of protein-specific glycosylation by proximity ligation polymerase chain reaction. J. Am. Chem. Soc. 138, 10722–10725 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Frei, A. P. et al. Highly multiplexed simultaneous detection of RNAs and proteins in single cells. Nat. Methods 13, 269–275 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Duckworth, A. D. et al. Multiplexed profiling of RNA and protein expression signatures in individual cells using flow or mass cytometry. Nat. Protoc. 14, 901–920 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Ohata, J. et al. An activity-based methionine bioconjugation approach to developing proximity-activated imaging reporters. ACS Cent. Sci. 6, 32–40 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Ren, X. et al. Single-cell imaging of m6A modified RNA using m6A-specific in situ hybridization mediated proximity ligation assay (m6AISH-PLA). Angew. Chem. Int. Ed. Engl. 60, 22646–22651 (2021).

    CAS  PubMed  Google Scholar 

  36. Yue, H. et al. Systematic screening and optimization of single-stranded DNA aptamer specific for N-acetylneuraminic acid: a comparative study. Sens. Actuators B Chem. 344, 130270 (2021).

    CAS  Google Scholar 

  37. Cummings, R. D. & Etzler, M. E. In Essentials of Glycobiology 2nd edn (eds Cummings, V. A. et al.) Ch 45 (Cold Spring Harbor Laboratory Press, 2009).

  38. Tommasone, S. et al. The challenges of glycan recognition with natural and artificial receptors. Chem. Soc. Rev. 48, 5488–5505 (2019).

    CAS  PubMed  Google Scholar 

  39. Huang, N. et al. Natural display of nuclear-encoded RNA on the cell surface and its impact on cell interaction. Genome Biol. 21, 225 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Sezgin, E., Levental, I., Mayor, S. & Eggeling, C. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat. Rev. Mol. Cell Biol. 18, 361–374 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Manders, E. M. M., Verbeek, F. J. & Aten, J. A. Measurement of co-localization of objects in dual-colour confocal images. J. Microsc. 169, 375–382 (1993).

    CAS  PubMed  Google Scholar 

  42. Hong, W. SNAREs and traffic. Biochim. Biophys. Acta 1744, 120–144 (2005).

    CAS  PubMed  Google Scholar 

  43. Potapenko, I. O. et al. Glycan-related gene expression signatures in breast cancer subtypes; relation to survival. Mol. Oncol. 9, 861–876 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Pinho, S. S. & Reis, C. A. Glycosylation in cancer: mechanisms and clinical implications. Nat. Rev. Cancer 15, 540–555 (2015).

    CAS  PubMed  Google Scholar 

  45. Munkley, J. & Elliott, D. J. Hallmarks of glycosylation in cancer. Oncotarget 7, 35478–35489 (2016).

    PubMed  PubMed Central  Google Scholar 

  46. Pearce, O. M. & Laubli, H. Sialic acids in cancer biology and immunity. Glycobiology 26, 111–128 (2016).

    CAS  PubMed  Google Scholar 

  47. Reis, C. A., Osorio, H., Silva, L., Gomes, C. & David, L. Alterations in glycosylation as biomarkers for cancer detection. J. Clin. Pathol. 63, 322–329 (2010).

    CAS  PubMed  Google Scholar 

  48. Ma, X. et al. Functional roles of sialylation in breast cancer progression through miR-26a/26b targeting ST8SIA4. Cell Death Dis. 7, e2561 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhang, T., de Waard, A. A., Wuhrer, M. & Spaapen, R. M. The role of glycosphingolipids in immune cell functions. Front. Immunol. 10, 90 (2019).

    PubMed  PubMed Central  Google Scholar 

  50. Dusoswa, S. A. et al. Glioblastomas exploit truncated O-linked glycans for local and distant immune modulation via the macrophage galactose-type lectin. Proc. Natl Acad. Sci. USA 117, 3693–3703 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Bartish, M., Del Rincon, S. V., Rudd, C. E. & Saragovi, H. U. Aiming for the sweet spot: glyco-immune checkpoints and γδ T cells in targeted immunotherapy. Front. Immunol. 11, 564499 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Venkatakrishnan, V. et al. Glycan analysis of human neutrophil granules implicates a maturation-dependent glycosylation machinery. J. Biol. Chem. 295, 12648–12660 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Sewald, X. et al. Retroviruses use CD169-mediated trans-infection of permissive lymphocytes to establish infection. Science 350, 563–567 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Man, S. et al. CXCL12-induced monocyte-endothelial interactions promote lymphocyte transmigration across an in vitro blood–brain barrier. Sci. Transl. Med. 4, 119ra114 (2012).

    Google Scholar 

  55. Ren, X. et al. Reconstruction of cell spatial organization from single-cell RNA sequencing data based on ligand–receptor mediated self-assembly. Cell Res. 30, 763–778 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Li, F. et al. A covalent approach for site-specific RNA labeling in mammalian cells. Angew. Chem. Int. Ed. Engl. 54, 4597–4602 (2015).

    CAS  PubMed  Google Scholar 

  57. Rabani, M. et al. Metabolic labeling of RNA uncovers principles of RNA production and degradation dynamics in mammalian cells. Nat. Biotechnol. 29, 436–442 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ouldridge, T. E., Louis, A. A. & Doye, J. P. K. Structural, mechanical, and thermodynamic properties of a coarse-grained DNA model. J. Chem. Phys. 134, 085101 (2011).

    PubMed  Google Scholar 

  59. Snodin, B. E. K. et al. Introducing improved structural properties and salt dependence into a coarse-grained model of DNA. J. Chem. Phys. 142, 234901 (2015).

    PubMed  Google Scholar 

  60. Bohlin, J. et al. Design and simulation of DNA, RNA and hybrid protein–nucleic acid nanostructures with oxView. Nat. Protoc. 17, 1762–1788 (2022).

    CAS  PubMed  Google Scholar 

  61. Baxter, E. W. et al. Standardized protocols for differentiation of THP-1 cells to macrophages with distinct M(IFNγ + LPS), M(IL-4) and M(IL-10) phenotypes. J. Immunol. Methods 478, 112721 (2020).

    CAS  PubMed  Google Scholar 

  62. Tarella, C., Ferrero, D., Gallo, E., Pagliardi, G. L. & Ruscetti, F. W. Induction of differentiation of HL-60 cells by dimethyl sulfoxide: evidence for a stochastic model not linked to the cell division cycle 1. Cancer Res. 42, 445–449 (1982).

    CAS  PubMed  Google Scholar 

  63. Millius, A. & Weiner, O. D. in Live Cell Imaging: Methods and Protocols (ed Papkovsky, D. B.) 147–158 (Humana Press, 2010).

  64. Lopez-Sambrooks, C. et al. Oligosaccharyltransferase inhibition induces senescence in RTK-driven tumor cells. Nat. Chem. Biol. 12, 1023–1030 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Elbein, A. D., Tropea, J. E., Mitchell, M. & Kaushal, G. P. Kifunensine, a potent inhibitor of the glycoprotein processing mannosidase I. J. Biol. Chem. 265, 15599–15605 (1990).

    CAS  PubMed  Google Scholar 

  66. Tulsiani, D. R., Harris, T. M. & Touster, O. Swainsonine inhibits the biosynthesis of complex glycoproteins by inhibition of Golgi mannosidase II. J. Biol. Chem. 257, 7936–7939 (1982).

    CAS  PubMed  Google Scholar 

  67. Poppleton, E. et al. Design, optimization and analysis of large DNA and RNA nanostructures through interactive visualization, editing and molecular simulation. Nucleic Acids Res. 48, e72 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by the US National Institutes of Health (GM141931 to Y.L. and GM133658 to S.S.Y.) and the Susan G. Komen Foundation (CCR19609287 to S.S.Y.). Additionally, the Robert A. Welch Foundation (grant F-0020 to Y. L.) supported the Lu group research programs at The University of Texas at Austin. We thank L. M. Mirica and J. Chan at the Department of Chemistry at University of Illinois Urbana–Champaign for providing SH-SY5Y and THP-1 cell lines and A. B. Baker at the Department of Biomedical Engineering at The University of Texas at Austin for providing the MDA-MB-231 cell line. We especially thank B. Belardi at the Department of Chemical Engineering and B. Xhemalce at the Department of Molecular Biosciences at The University of Texas at Austin for manuscript suggestions. Confocal imaging was performed at the Center for Biomedical Research Support Microscopy and Imaging Facility at The University of Texas at Austin (RRID: SCR_021756). We thank A. Webb and P. Oliphint at The University of Texas at Austin for providing advice on confocal imaging. We would like to thank Y. Wu, M. Banik and R. Yang for providing suggestions on the manuscript and for proofreading.

Author information

Authors and Affiliations

Authors

Contributions

Y.M., W.G. and Y.L. conceived and designed the study. Y.M. and W.G. performed the experiments and analyzed the data. Q.M. assisted in designing and validating the ARPLA method. Y.M., M.L., C.W. and V.G. performed RNA blotting experiments. Y.M., W.G., X.S., Z.Y. and W.L. performed cell experiments. L.K. performed the MD simulations. X.P. and S.S.Y. analyzed the data. The manuscript was written by Y.M., W.G. and Y.L. Y.L. supervised the project.

Corresponding author

Correspondence to Yi Lu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Matthew Disney, Ryan Flynn 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 MD simulation of the structure of the ARPLA system.

(a) A representation of the ARPLA system with different sites (site 1-6) chosen to analyze distances; (b) A representative structure from the simulation with oxDNA, including the glycan probe, the RNA binding probe, the connector 1, and the connector 2. The circle of the connector 1 and the connector 2 tends to form a triangle structure, with two sides formed by DNA helix and one side formed by the ssDNA region in the connector 1; (c) The distributions of the distance between interested sites. 3 sets of distances were calculated with oxDNA tool12, including the distances between the edges of the connector 1 ssDNA region (site 1 and site 2), between the ends of spacers of the Glycan probe and the RNA binding probe (site 3 and site 4), and between the predicted glycan binding site36 and the center of the RISH site (site 5 and site 6). The distance between site 1 and site 2 is in the range of 1-14 nm, with an average around 7 nm. The results agree with the B-form DNA length of the connector 1 ssDNA region (43 nt, around 14.3 nm), with the consideration of ssDNA bending and folding. The distance between site 3 and site 4 is in the range of 5-15 nm, with an average around 10 nm. The distance between site 5 and site 6 is in the range of 5-20 nm, with an average around 15 nm.

Extended Data Fig. 2 Verification of ARPLA method using HeLa as a model cell line.

(a) Thermogram for the ITC titration of 20 µM Neu5Ac aptamer titrated by 1 mM Neu5Ac in aptamer binding buffer; (b) Integrated heat of the ITC titration for Neu5Ac aptamer and Neu5Ac, the black line represents the binding curve fitted with the ‘one set of binding sites’ model; (c) Blotting of total RNA from HeLa cells after metabolic labeling with Ac4ManNAz, or HeLa cells without metabolic labeling; (d) ARPLA-mediated glycoRNA imaging on the surface of HeLa, HL-60, and THP-1 cells. Scale bar: 20 μm (HeLa), 10 μm (HL-60, THP-1); (e) Transmission-through dye microscopic image of HeLa cell. The membrane permeable dye (CellTracker Orange CMRA) and the membrane impermeant quencher acid blue 9 (AB9) were applied to the same cells. AB9 cannot enter the cell with intact membrane and thus cannot quench the membrane permeable dye, so the cells with intact membrane show bright fluorescence signals from CellTracker Orange CMRA. Leaky or damaged membrane after permeabilization treatment allows for the quencher to enter the cell, resulting in reduced or diminished fluorescence of the cell. Scale bar: 40 μm. All experiments were repeated independently three times with similar results.

Extended Data Fig. 3 Evaluation of the generality of ARPLA.

(a) CLSM images of glycoRNAs, utilizing ARPLA with Glycan probes with Neu5Ac aptamer, Tn antigen aptamer, and GalNAc aptamer; (b) Visualization of glycoRNAs with various RNA sequences, including U3, U8, U35a, and Y5; (c) CLSM images of U1 glycoRNA in different cell lines, such as SH-SY5Y, PANC-1, and HEK293T. Scale bars (b,c): 40 µm; (d) Blotting of total RNA from SH-SY5Y cells after metabolic labeling with Ac4ManNAz, or SH-SY5Y cells without metabolic labeling; (e) Blotting of total RNA from PANC-1 cells after metabolic labeling with Ac4ManNAz, or PANC-1 cells without metabolic labeling; (f) Blotting of total RNA from HEK293T cells after metabolic labeling with Ac4ManNAz, or HEK293T cells without metabolic labeling.

Extended Data Fig. 4 3D visualization of the spatial distributions of U1 glycoRNA and CT-B in HL-60 cells.

Z-stack images were collected with the staining of U1 glycoRNA by ARPLA (green) and lipid raft by CT-B (red). The images were shown in z-slices format (a), orthographic projection (b), and maximum intensity projection (c). Scale bar: 2 µm. The z-stack colocalization was repeated independently three times with similar results.

Extended Data Fig. 5 Visualization of glycoRNAs in malignant transformation using ARPLA, related with Fig. 4.

(a,b) CLSM images of MCF-10A, MCF-7, MDA-MB-231 cells in control groups using DNA with scrambled sequences. (c) Agarose gel electrophoresis image of total RNA from MCF-10A, MCF-7, MDA-MB-231 cells. These cells were treated with Ac4ManNAz for 48 h before RNA extraction. All experiments were repeated independently 3 times with similar results.

Extended Data Fig. 6 Fluorescence imaging of bulk sialic acid on the cell surface of MCF-10A, MCF-7, and MDA-MB-231 cells.

(a) Representative cell fluorescent images of bulk sialic acid. These cells were metabolically labeled by Ac4ManNAz for 24 h, followed by incubation with DBCO-PEG4-biotin and Cy5-streptavidin for fluorescence imaging. Scale bars for cell image: 50 μm. (b) Bar plot of the mean fluorescent intensities, the data were calculated from 3 biological replicates. The plot is shown in mean ± SD. Unpaired two-tailed Student’s t-test determines the statistical significance as (*) p = 0.0352, (ns) p = 0.0793, n = 3 independent replicates.

Extended Data Fig. 7 Visualization of glycoRNA level during THP-1 differentiation and activation by LPS, related with Fig. 5.

(a, b) CLSM images of THP-1 monocyte, resting M0 macrophage, and activated M0 macrophage by LPS, which are treated with DNA probe with scrambled sequence to replace aptamer in ARPLA. (c) Blotting of total RNA from THP-1 cells after metabolic labeling with Ac4ManNAz or THP-1 cells without metabolic labeling. (d) Agarose gel electrophoresis image of total RNA from THP-1 monocyte, resting M0 macrophage, and activated M0 macrophage by LPS. These cells were treated with Ac4ManNAz for 48 h before RNA extraction. All experiments were repeated independently three times with similar results.

Extended Data Fig. 8 Investigation of glycoRNA levels during HL-60 differentiation.

(a) CLSM images of U1, U3, and U8 glycoRNA levels evaluated by ARPLA in HL-60 and dHL-60 cells; (b) CLSM images of HL-60, dHL-60 cells in control groups using DNA with scrambled sequences; (c) Quantitative analysis for relative fluorescence intensity of ARPLA in (a) and (b). Data in (c) are representative of three independent experiments, n = 5 frames. Data are mean ±S.D. The statistical significance is determined by unpaired two-tailed Student’s t-test as (ns) not significant, (*) P < 0.05, (**) P < 0.01, and (***) P < 0.001. P (U1 HL60 vs. dHL60) = 0.0159, P (U3 HL60 vs. dHL60) = 0.0079, P (U8 HL60 vs. dHL60) = 0.0002.

Extended Data Fig. 9 Fluorescence imaging of bulk sialic acid on the cell surface of THP-1 monocyte, resting M0 macrophage (M0), LPS activated M0 macrophage (M0 + LPS).

(a) Representative images of total sialic acid. These cells were metabolically labeled by Ac4ManNAz for 24 h, followed by incubation with DBCO-PEG4-biotin and Cy5-streptavidin for fluorescence imaging. Scale bars for cell image: 50 μm. (b) Quantification of the mean fluorescent intensity of the images, n = 3 biological replicates. Data are mean ±S.D. Unpaired two-tailed Student’s t-test determines the statistical significance. (**) p = 0.0013, (*) p = 0.0469.

Extended Data Fig. 10 Cell attachment assay.

The average cell attachment levels in resting M0 macrophage, activated M0 macrophage by LPS, activated M0 macrophage after RNase treatment. Data are representative of three independent experiments, n = 6 technical repeats. Data are mean ±S.D. The statistical significance is determined by unpaired two-tailed Student’s t-test, (***) P < 0.0001, (**) P = 0.0051. Cell attachment assay was performed three times independently with similar results.

Supplementary information

Supplementary Information

Supplementary Table 1. DNA sequences used in the experiments (5′ to 3′). Supplementary Fig. 1. Quantification of ARPLA signal dots for individual cells using U1 glycoRNA in HeLa cells as an example. Supplementary Fig. 2. GlycoRNA gel blot in HeLa cell subfractions. Supplementary Fig. 3. Single-cell analysis of ARPLA dots in breast cell lines. Supplementary Fig. 4. Investigation of total glycoRNA levels during HL-60 differentiation by RNA blotting. Supplementary Fig. 5. Estimation of the limit of resolution.

Reporting Summary

Source data

Source data

Unprocessed gels and blots for all the gels and blots shown in the manuscript.

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

Ma, Y., Guo, W., Mou, Q. et al. Spatial imaging of glycoRNA in single cells with ARPLA. Nat Biotechnol 42, 608–616 (2024). https://doi.org/10.1038/s41587-023-01801-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-023-01801-z

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