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.

  • Primer
  • Published:

Time-resolved single-cell RNA-seq using metabolic RNA labelling

Abstract

Single-cell RNA genomics technologies are revolutionizing biomedical science by profiling single cells with unprecedented resolution, providing fundamental insights into the role of different cellular states and intercellular heterogeneity in health and disease. The combination of single-cell RNA sequencing (scRNA-seq) with metabolic RNA labelling approaches now enables time-resolved monitoring of transcriptional responses for thousands of genes in thousands of individual cells in parallel. This facilitates and accelerates direct characterization of the temporal dimension of biological processes, which has been largely missing in current data. In this Primer, we provide an overview of the various metabolic RNA labelling approaches and their combination with currently available scRNA-seq and multi-omics platforms. We summarize the main challenges in the design of such experiments and discuss the various applications of time-resolved scRNA-seq in vitro and in vivo. We outline the computational tools and challenges to the analyses of the temporal dynamics of transcriptional responses at the single-cell level. We discuss the prospect of integrating data obtained by the respective time-resolved scRNA-seq approaches with complementary methods to elucidate gene regulatory networks that underlie molecular mechanisms. Finally, we discuss open questions and challenges in the field and give our thoughts for future development and applications.

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: Metabolic RNA labelling chemistries and approaches.
Fig. 2: Overview of workflows that allow recording of transcriptional activity at the single-cell level.
Fig. 3: scSLAM-seq data analysis and results.
Fig. 4: Applications of time-resolved scRNA-seq using metabolic RNA labelling across biological fields.
Fig. 5: Overview of methods compatible with RNA metabolic labelling amenable to multi-modal single-cell analysis.

Similar content being viewed by others

References

  1. Tanay, A. & Regev, A. Scaling single-cell genomics from phenomenology to mechanism. Nature 541, 331–338 (2017).

    ADS  Google Scholar 

  2. Saliba, A.-E., Westermann, A. J., Gorski, S. A. & Vogel, J. Single-cell RNA-seq: advances and future challenges. Nucleic Acids Res. 42, 8845–8860 (2014).

    Google Scholar 

  3. Regev, A. et al. The human cell atlas. eLife 6, e27041 (2017).

    Google Scholar 

  4. Elmentaite, R., Domínguez Conde, C., Yang, L. & Teichmann, S. A. Single-cell atlases: shared and tissue-specific cell types across human organs. Nat. Rev. Genet. 23, 395–410 (2022).

    Google Scholar 

  5. Shalek, A. K. et al. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature 510, 363–369 (2014).

    ADS  Google Scholar 

  6. Dölken, L. et al. High-resolution gene expression profiling for simultaneous kinetic parameter analysis of RNA synthesis and decay. RNA 14, 1959–1972 (2008). Dölken et al. exemplify the benefits of metabolic labelling for measuring short-term changes in RNA synthesis and decay in the interferon response of fibroblasts.

    Google Scholar 

  7. Friedel, C. C., Dölken, L., Ruzsics, Z., Koszinowski, U. H. & Zimmer, R. Conserved principles of mammalian transcriptional regulation revealed by RNA half-life. Nucleic Acids Res. 37, e115 (2009).

    Google Scholar 

  8. Windhager, L. et al. Ultrashort and progressive 4sU-tagging reveals key characteristics of RNA processing at nucleotide resolution. Genome Res. 22, 2031–2042 (2012).

    Google Scholar 

  9. Yang, E. et al. Decay rates of human mRNAs: correlation with functional characteristics and sequence attributes. Genome Res. 13, 1863–1872 (2003).

    Google Scholar 

  10. Rutkowski, A. J. et al. Widespread disruption of host transcription termination in HSV-1 infection. Nat. Commun. 6, 7126 (2015).

    ADS  Google Scholar 

  11. Erhard, F. et al. scSLAM-seq reveals core features of transcription dynamics in single cells. Nature 571, 419–423 (2019). This article introduces scSLAM-seq to perform 4sU RNA metabolic labelling at the single-cell level in plate-based format.

    Google Scholar 

  12. Cleary, M. D., Meiering, C. D., Jan, E., Guymon, R. & Boothroyd, J. C. Biosynthetic labeling of RNA with uracil phosphoribosyltransferase allows cell-specific microarray analysis of mRNA synthesis and decay. Nat. Biotechnol. 23, 232–237 (2005). This article introduces metabolic RNA labelling using 4tU and its activation by UPRT in eukaryotic cells coupled to microarray analysis.

    Google Scholar 

  13. Miller, M. R., Robinson, K. J., Cleary, M. D. & Doe, C. Q. TU-tagging: cell type-specific RNA isolation from intact complex tissues. Nat. Methods 6, 439–441 (2009).

    Google Scholar 

  14. Miller, C. et al. Dynamic transcriptome analysis measures rates of mRNA synthesis and decay in yeast. Mol. Syst. Biol. 7, 458 (2011).

    Google Scholar 

  15. Hida, N. et al. EC-tagging allows cell type-specific RNA analysis. Nucleic Acids Res. 45, e138 (2017).

    Google Scholar 

  16. Kofoed, R. H., Betzer, C., Lykke-Andersen, S., Molska, E. & Jensen, P. H. Investigation of RNA synthesis using 5-bromouridine labelling and immunoprecipitation. J. Vis. Exp. 135, 57056 (2018).

    Google Scholar 

  17. Kawata, K. et al. Metabolic labeling of RNA using multiple ribonucleoside analogs enables the simultaneous evaluation of RNA synthesis and degradation rates. Genome Res. 30, 1481–1491 (2020).

    Google Scholar 

  18. Schwalb, B. et al. TT-seq maps the human transient transcriptome. Science 352, 1225–1228 (2016).

    ADS  Google Scholar 

  19. Gay, L. et al. Mouse TU tagging: a chemical/genetic intersectional method for purifying cell type-specific nascent RNA. Genes Dev. 27, 98–115 (2013).

    Google Scholar 

  20. Erickson, T. & Nicolson, T. Identification of sensory hair-cell transcripts by thiouracil-tagging in zebrafish. BMC Genomics 16, 842 (2015).

    Google Scholar 

  21. Erickson, T. & Nicolson, T. Cell type-specific transcriptomic analysis by thiouracil tagging in zebrafish. Methods Cell Biol. 135, 309–328 (2016).

    Google Scholar 

  22. Tallafuss, A. et al. Transcriptomes of post-mitotic neurons identify the usage of alternative pathways during adult and embryonic neuronal differentiation. BMC Genomics 16, 1100 (2015).

    Google Scholar 

  23. Ussuf, K. K., Anikumar, G. & Nair, P. M. Newly synthesised mRNA as a probe for identification of wound responsive genes from potatoes. Indian J. Biochem. 32, 78–83 (1995).

    Google Scholar 

  24. Herzog, V. A. et al. Thiol-linked alkylation of RNA to assess expression dynamics. Nat. Methods 14, 1198–1204 (2017). This article provides the conceptual basis for nucleotide conversion sequencing using iodoacetamide (IAA) to achieve a 4sU-to-C conversion.

    Google Scholar 

  25. Schofield, J. A., Duffy, E. E., Kiefer, L., Sullivan, M. C. & Simon, M. D. TimeLapse-seq: adding a temporal dimension to RNA sequencing through nucleoside recoding. Nat. Methods 15, 221–225 (2018). This article provides the conceptual basis for nucleotide conversion sequencing using oxidative-nucleophilic-aromatic substitution to achieve a 4sU-to-C conversion.

    Google Scholar 

  26. Riml, C. et al. Osmium-mediated transformation of 4-thiouridine to cytidine as key to study RNA dynamics by sequencing. Angew. Chem. Int. Ed.56, 13479–13483 (2017). This article provides the conceptual basis for nucleotide conversion sequencing using Osmium-mediated 4sU-to-C conversion.

    Google Scholar 

  27. Kiefer, L., Schofield, J. A. & Simon, M. D. Expanding the nucleoside recoding toolkit: revealing RNA population dynamics with 6-thioguanosine. J. Am. Chem. Soc. 140, 14567–14570 (2018).

    Google Scholar 

  28. Gasser, C. et al. Thioguanosine conversion enables mRNA-lifetime evaluation by RNA sequencing using double metabolic labeling (TUC-seq DUAL). Angew. Chem. Int. Ed. 59, 6881–6886 (2020).

    Google Scholar 

  29. Chen, Y. et al. Acrylonitrile-mediated nascent RNA sequencing for transcriptome-wide profiling of cellular RNA dynamics. Adv. Sci. 7, 1900997 (2020).

    Google Scholar 

  30. Maier, K. C., Gressel, S., Cramer, P. & Schwalb, B. Native molecule sequencing by nano-ID reveals synthesis and stability of RNA isoforms. Genome Res. 30, 1332–1344 (2020).

    Google Scholar 

  31. Baptista, M. A. P. & Dölken, L. RNA dynamics revealed by metabolic RNA labeling and biochemical nucleoside conversions. Nat. Methods 15, 171–172 (2018).

    Google Scholar 

  32. Jürges, C., Dölken, L. & Erhard, F. Dissecting newly transcribed and old RNA using GRAND-SLAM. Bioinformatics 34, i218–i226 (2018). GRAND-SLAM provides a computational framework that enables the proportion of old and new RNA to be estimated.

    Google Scholar 

  33. Muthmann, N., Hartstock, K. & Rentmeister, A. Chemo-enzymatic treatment of RNA to facilitate analyses. Wiley Interdiscip. Rev. RNA 11, e1561 (2020).

    Google Scholar 

  34. Singha, M., Spitalny, L., Nguyen, K., Vandewalle, A. & Spitale, R. C. Chemical methods for measuring RNA expression with metabolic labeling. Wiley Interdiscip. Rev. RNA 12, e1650 (2021).

    Google Scholar 

  35. Klöcker, N., Weissenboeck, F. P. & Rentmeister, A. Covalent labeling of nucleic acids. Chem. Soc. Rev. 49, 8749–8773 (2020).

    Google Scholar 

  36. Anhäuser, L. & Rentmeister, A. Enzyme-mediated tagging of RNA. Curr. Opin. Biotechnol. 48, 69–76 (2017).

    Google Scholar 

  37. Qu, D. et al. 5-Ethynylcytidine as a new agent for detecting RNA synthesis in live cells by ‘click’ chemistry. Anal. Biochem. 434, 128–135 (2013).

    Google Scholar 

  38. Haider, S. R., Juan, G., Traganos, F. & Darzynkiewicz, Z. Immunoseparation and immunodetection of nucleic acids labeled with halogenated nucleotides. Exp. Cell Res. 234, 498–506 (1997).

    Google Scholar 

  39. Kubota, M. et al. Expanding the scope of RNA metabolic labeling with vinyl nucleosides and inverse electron-demand diels-alder chemistry. ACS Chem. Biol. 14, 1698–1707 (2019).

    Google Scholar 

  40. Nainar, S. et al. An optimized chemical-genetic method for cell-specific metabolic labeling of RNA. Nat. Methods 17, 311–318 (2020).

    Google Scholar 

  41. Meng, L. et al. Metabolic RNA labeling for probing RNA dynamics in bacteria. Nucleic Acids Res. 48, 12566–12576 (2020).

    Google Scholar 

  42. Wang, D., Zhang, Y. & Kleiner, R. E. Cell- and polymerase-selective metabolic labeling of cellular RNA with 2′-azidocytidine. J. Am. Chem. Soc. 142, 14417–14421 (2020).

    Google Scholar 

  43. He, Z. et al. Metabolic labeling and imaging of cellular rna via bioorthogonal cyclopropene-tetrazine ligation. CCS Chem. 2, 89–97 (2020).

    Google Scholar 

  44. Beasley, S., Nguyen, K., Fazio, M. & Spitale, R. C. Protected pyrimidine nucleosides for cell-specific metabolic labeling of RNA. Tetrahedron Lett. 59, 3912–3915 (2018).

    Google Scholar 

  45. Nguyen, K. et al. Cell-selective bioorthogonal metabolic labeling of RNA. J. Am. Chem. Soc. 139, 2148–2151 (2017).

    Google Scholar 

  46. Moreno, S. et al. Synthesis of 4-thiouridines with prodrug functionalization for RNA metabolic labeling. RSC Chem. Biol. 3, 447 (2022).

    Google Scholar 

  47. Holler, K. et al. Spatio-temporal mRNA tracking in the early zebrafish embryo. Nat. Commun. 12, 3358 (2021). Holler et al. combine metabolic RNA labelling with spatially resolved transcriptomics to measure activation of cygotic transcription in the zebrafish embryo.

    ADS  Google Scholar 

  48. Tani, H. et al. Genome-wide determination of RNA stability reveals hundreds of short-lived noncoding transcripts in mammals. Genome Res. 22, 947–956 (2012).

    Google Scholar 

  49. Imamachi, N. et al. BRIC-seq: a genome-wide approach for determining RNA stability in mammalian cells. Methods 67, 55–63 (2014).

    Google Scholar 

  50. Paulsen, M. T. et al. Use of Bru-Seq and BruChase-Seq for genome-wide assessment of the synthesis and stability of RNA. Methods 67, 45–54 (2014).

    Google Scholar 

  51. Russo, J., Heck, A. M., Wilusz, J. & Wilusz, C. J. Metabolic labeling and recovery of nascent RNA to accurately quantify mRNA stability. Methods 120, 39–48 (2017).

    Google Scholar 

  52. Cleary, M. D. Uncovering cell type-specific complexities of gene expression and RNA metabolism by TU-tagging and EC-tagging. Wiley Interdiscip. Rev. Dev. Biol. 7, e315 (2018).

    Google Scholar 

  53. Duffy, E. E. et al. Tracking distinct RNA populations using efficient and reversible covalent chemistry. Mol. Cell 59, 858–866 (2015).

    Google Scholar 

  54. Michel, M. et al. TT-seq captures enhancer landscapes immediately after T-cell stimulation. Mol. Syst. Biol. 13, 920 (2017).

    Google Scholar 

  55. Gregersen, L. H., Mitter, R. & Svejstrup, J. Q. Using TTchem-seq for profiling nascent transcription and measuring transcript elongation. Nat. Protoc. 15, 604–627 (2020).

    Google Scholar 

  56. Nguyen, K. et al. Spatially restricting bioorthogonal nucleoside biosynthesis enables selective metabolic labeling of the mitochondrial transcriptome. ACS Chem. Biol. 13, 1474–1479 (2018).

    Google Scholar 

  57. Zajaczkowski, E. L. et al. Bioorthogonal metabolic labeling of nascent RNA in neurons improves the sensitivity of transcriptome-wide profiling. ACS Chem. Neurosci. 9, 1858–1865 (2018).

    Google Scholar 

  58. Zhang, Y. & Kleiner, R. E. A metabolic engineering approach to incorporate modified pyrimidine nucleosides into cellular RNA. J. Am. Chem. Soc. 141, 3347–3351 (2019).

    Google Scholar 

  59. Nainar, S. et al. Metabolic incorporation of azide functionality into cellular RNA. ChemBioChem 17, 2149–2152 (2016).

    Google Scholar 

  60. Su, L. et al. Addition-elimination mechanism-activated nucleotide transition sequencing for RNA dynamics profiling. Anal. Chem. 93, 13974–13980 (2021).

    Google Scholar 

  61. Schott, J. et al. Nascent Ribo-Seq measures ribosomal loading time and reveals kinetic impact on ribosome density. Nat. Methods 18, 1068–1074 (2021).

    Google Scholar 

  62. Boileau, E., Altmüller, J., Naarmann-de Vries, I. S. & Dieterich, C. A comparison of metabolic labeling and statistical methods to infer genome-wide dynamics of RNA turnover. Brief. Bioinform. 22, bbab219 (2021).

    Google Scholar 

  63. Qiu, Q. et al. Massively parallel and time-resolved RNA sequencing in single cells with scNT-seq. Nat. Methods 17, 991–1001 (2020). This article introduces scNT-seq for time-resolved scRNA sequencing using the TimeLapse-seq chemistry.

    Google Scholar 

  64. Mitter, M. et al. Conformation of sister chromatids in the replicated human genome. Nature 586, 139–144 (2020).

    ADS  Google Scholar 

  65. Mitter, M. et al. Sister chromatid–sensitive Hi-C to map the conformation of replicated genomes. Nat. Protoc. 17, 1486–1517 (2022).

    Google Scholar 

  66. Drexler, H. L., Choquet, K. & Churchman, L. S. Splicing kinetics and coordination revealed by direct nascent RNA sequencing through nanopores. Mol. Cell 77, 985–998.e8 (2020).

    Google Scholar 

  67. Drexler, H. L. et al. Revealing nascent RNA processing dynamics with nano-COP. Nat. Protoc. 16, 1343–1375 (2021).

    Google Scholar 

  68. Wagner, D. E. & Klein, A. M. Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet. 21, 410–427 (2020).

    Google Scholar 

  69. Saelens, W., Cannoodt, R., Todorov, H. & Saeys, Y. A comparison of single-cell trajectory inference methods. Nat. Biotechnol. 37, 547–554 (2019).

    Google Scholar 

  70. Olivares-Chauvet, P. & Junker, J. P. Inclusion of temporal information in single cell transcriptomics. Int. J. Biochem. Cell Biol. 122, 105745 (2020).

    Google Scholar 

  71. Hendriks, G.-J. et al. NASC-seq monitors RNA synthesis in single cells. Nat. Commun. 10, 3138 (2019). This article introduces NASC-seq to perform 4sU RNA metabolic labelling at the single-cell level in plate-based format.

    ADS  Google Scholar 

  72. Cao, J., Zhou, W., Steemers, F., Trapnell, C. & Shendure, J. Sci-fate characterizes the dynamics of gene expression in single cells. Nat. Biotechnol. 38, 980–988 (2020). This article introduces sci-fate to perform 4sU RNA metabolic labelling at the single-cell level using combinatorial indexing.

    Google Scholar 

  73. Phan, H. Van. et al. High-throughput RNA sequencing of paraformaldehyde-fixed single cells. Nat. Commun. 12, 5636 (2021).

    ADS  Google Scholar 

  74. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    ADS  Google Scholar 

  75. Cao, J. et al. Comprehensive single-cell transcriptional profiling of a multicellular organism. Science 357, 661–667 (2017).

    ADS  Google Scholar 

  76. Battich, N. et al. Sequencing metabolically labeled transcripts in single cells reveals mRNA turnover strategies. Science 367, 1151–1156 (2020). Battich et al. employ ethynyluridine labelling in single cells to estimate RNA synthesis and decay rates in thousands of intestinal organoid cells.

    ADS  Google Scholar 

  77. Marcinowski, L. et al. Real-time transcriptional profiling of cellular and viral gene expression during lytic cytomegalovirus infection. PLoS Pathog. 8, e1002908 (2012).

    Google Scholar 

  78. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Google Scholar 

  79. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Google Scholar 

  80. Luecken, M. D. & Theis, F. J. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol. Syst. Biol. 15, e8746 (2019).

    Google Scholar 

  81. Qiu, X. et al. Mapping transcriptomic vector fields of single cells. Cell 185, 690–711.e45 (2022). Qiu et al. introduce the analytical framework (Dynamo) to infer absolute RNA velocity and predict cell fates on the basis of metabolic RNA labelling in single cells.

    Google Scholar 

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

    Google Scholar 

  83. Petukhov, V. et al. dropEst: pipeline for accurate estimation of molecular counts in droplet-based single-cell RNA-seq experiments. Genome Biol 19, 78 (2018).

    Google Scholar 

  84. Kaminow, B., Yunusov, D. & Dobin, A. STARsolo: accurate, fast and versatile mapping/quantification of single-cell and single-nucleus RNA-seq data. Preprint at bioRxiv https://doi.org/10.1101/2021.05.05.442755 (2021).

    Article  Google Scholar 

  85. Melsted, P. et al. Modular and efficient pre-processing of single-cell RNA-seq. Preprint at bioRxiv https://doi.org/10.1101/673285 (2019).

    Article  Google Scholar 

  86. Melsted, P. et al. Modular, efficient and constant-memory single-cell RNA-seq preprocessing. Nat. Biotechnol. 39, 813–818 (2021).

    Google Scholar 

  87. Robert, F. & Pelletier, J. Exploring the impact of single-nucleotide polymorphisms on translation. Front. Genet. 9, 507 (2018).

    Google Scholar 

  88. Kang, H. M. et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol. 36, 89–94 (2018).

    Google Scholar 

  89. Koboldt, D. C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Google Scholar 

  90. Burger, K. et al. 4-thiouridine inhibits rRNA synthesis and causes a nucleolar stress response. RNA Biol. 10, 1623–1630 (2013).

    Google Scholar 

  91. Muhar, M. et al. SLAM-seq defines direct gene-regulatory functions of the BRD4-MYC axis. Science 360, 800–805 (2018).

    Google Scholar 

  92. McCarthy, D. J., Campbell, K. R., Lun, A. T. L. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).

    Google Scholar 

  93. Dye, M. J., Gromak, N. & Proudfoot, N. J. Exon tethering in transcription by RNA polymerase II. Mol. Cell 21, 849–859 (2006).

    Google Scholar 

  94. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018). The RNA velocity framework provides a time derivative of the gene expression state by distinguishing between unspliced and spliced mRNAs.

    ADS  Google Scholar 

  95. Ameur, A. et al. Total RNA sequencing reveals nascent transcription and widespread co-transcriptional splicing in the human brain. Nat. Struct. Mol. Biol. 18, 1435–1440 (2011).

    Google Scholar 

  96. Bresnahan, W. A. & Shenk, T. A subset of viral transcripts packaged within human cytomegalovirus particles. Science 288, 2373–2376 (2000).

    ADS  Google Scholar 

  97. Terhune, S. S. et al. RNAs are packaged into human cytomegalovirus virions in proportion to their intracellular concentration. J. Virol. 78, 10390–10398 (2004).

    Google Scholar 

  98. Geula, S. et al. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science 347, 1002–1006 (2015).

    ADS  Google Scholar 

  99. Batista, P. J. et al. M6A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 15, 707–719 (2014).

    Google Scholar 

  100. Junker, J. P. et al. Genome-wide RNA tomography in the zebrafish embryo. Cell 159, 662–675 (2014).

    Google Scholar 

  101. Holler, K. & Junker, J. P. RNA tomography for spatially resolved transcriptomics (tomo-seq). Methods Mol. Biol. 1920, 129–141 (2019).

    Google Scholar 

  102. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    ADS  Google Scholar 

  103. Afgan, E. et al. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2018 update. Nucleic Acids Res. 46, W537–W544 (2018).

    Google Scholar 

  104. Köster, J. et al. Sustainable data analysis with Snakemake. F1000Res. 10, 33 (2021).

    Google Scholar 

  105. Kluge, M., Friedl, M. S., Menzel, A. L. & Friedel, C. C. Watchdog 2.0: new developments for reusability, reproducibility, and workflow execution. Gigascience 9, giaa068 (2020).

    Google Scholar 

  106. Uvarovskii, A., Naarmann-De Vries, I. S. & Dieterich, C. On the optimal design of metabolic RNA labeling experiments. PLoS Comput. Biol. 15, e1007252 (2019).

    ADS  Google Scholar 

  107. Behm-Ansmant, I., Helm, M. & Motorin, Y. Use of specific chemical reagents for detection of modified nucleotides in RNA. J. Nucleic Acids 2011, 408053 (2011).

    Google Scholar 

  108. Mitchell, D., Assmann, S. M. & Bevilacqua, P. C. Probing RNA structure in vivo. Curr. Opin. Struct. Biol. 59, 151–158 (2019).

    Google Scholar 

  109. Ziff, E. B. & Fresco, J. R. Chemical transformation of 4-thiouracil nucleosides to uracil and cytosine counterparts. J. Am. Chem. Soc. 90, 7338–7342 (1968).

    Google Scholar 

  110. You, Y. et al. Benchmarking UMI-based single-cell RNA-seq preprocessing workflows. Genome Biol. 22, 339 (2021).

    Google Scholar 

  111. Hahaut, V. et al. Fast and highly sensitive full-length single-cell RNA sequencing using FLASH-seq. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01312-3 (2022).

    Article  Google Scholar 

  112. Replogle, J. M. et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing. Nat. Biotechnol. 38, 954–961 (2020).

    Google Scholar 

  113. Stuart, T. & Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 20, 257–272 (2019).

    Google Scholar 

  114. Gray Camp, J., Platt, R. & Treutlein, B. Mapping human cell phenotypes to genotypes with single-cell genomics. Science 365, 1401–1405 (2019).

    ADS  Google Scholar 

  115. Matsushima, W. et al. SLAM-ITseq: sequencing cell type-specific transcriptomes without cell sorting. Development 145, dev164640 (2018).

    Google Scholar 

  116. Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2021).

    Google Scholar 

  117. He, Z. et al. Lineage recording in human cerebral organoids. Nat. Methods 19, 90–99 (2022).

    Google Scholar 

  118. Macaulay, I. C. et al. G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat. Methods 12, 519–522 (2015).

    Google Scholar 

  119. Han, K. Y. et al. SIDR: simultaneous isolation and parallel sequencing of genomic DNA and total RNA from single cells. Genome Res. 28, 75–87 (2018).

    Google Scholar 

  120. Dey, S. S., Kester, L., Spanjaard, B., Bienko, M. & Van Oudenaarden, A. Integrated genome and transcriptome sequencing of the same cell. Nat. Biotechnol. 33, 285–289 (2015).

    Google Scholar 

  121. Rodriguez-Meira, A. et al. Unravelling intratumoral heterogeneity through high-sensitivity single-cell mutational analysis and parallel RNA sequencing. Mol. Cell 73, 1292–1305.e8 (2019).

    Google Scholar 

  122. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    Google Scholar 

  123. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Google Scholar 

  124. Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).

    ADS  Google Scholar 

  125. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Google Scholar 

  126. Chen, S., Lake, B. B. & Zhang, K. High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell. Nat. Biotechnol. 37, 1452–1457 (2019).

    Google Scholar 

  127. Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, 1380–1385 (2018).

    ADS  Google Scholar 

  128. Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).

    ADS  Google Scholar 

  129. Bartosovic, M., Kabbe, M. & Castelo-Branco, G. Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues. Nat. Biotechnol. 39, 825–835 (2021).

    Google Scholar 

  130. Wu, S. J. et al. Single-cell CUT&Tag analysis of chromatin modifications in differentiation and tumor progression. Nat. Biotechnol. 39, 819–824 (2021).

    Google Scholar 

  131. Carter, B. et al. Mapping histone modifications in low cell number and single cells using antibody-guided chromatin tagmentation (ACT-seq). Nat. Commun. 10, 3747 (2019).

    ADS  Google Scholar 

  132. Wang, Q. et al. CoBATCH for high-throughput single-cell epigenomic profiling. Mol. Cell 76, 206–216.e7 (2019).

    Google Scholar 

  133. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Google Scholar 

  134. Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).

    Google Scholar 

  135. Gerlach, J. P. et al. Combined quantification of intracellular (phospho-)proteins and transcriptomics from fixed single cells. Sci. Rep. 9, 1469 (2019).

    ADS  Google Scholar 

  136. VanInsberghe, M., van den Berg, J., Andersson-Rolf, A., Clevers, H. & van Oudenaarden, A. Single-cell Ribo-seq reveals cell cycle-dependent translational pausing. Nature 597, 561–565 (2021). Single-cell Ribo-seq allows translation dynamics to be measured at single-cell level. The authors generated ribosome profiling from rare primary mouse intestinal enteroendocrine cells.

    ADS  Google Scholar 

  137. Hou, Y. et al. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 26, 304–319 (2016).

    Google Scholar 

  138. Clark, S. J. et al. ScNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun. 9, 781 (2018).

    ADS  Google Scholar 

  139. Swanson, E. et al. Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using tea-seq. eLife 10, e63632 (2021).

    Google Scholar 

  140. Parker, M. T. et al. Nanopore direct RNA sequencing maps the complexity of arabidopsis mRNA processing and m6A modification. eLife 9, e49658 (2020).

    Google Scholar 

  141. Stein, D. F. et al. singlecellVR: interactive visualization of single-cell data in virtual reality. Front. Genet. 12, 764170 (2021).

    Google Scholar 

  142. Tunnacliffe, E. & Chubb, J. R. What is a transcriptional burst? Trends Genet. 36, 288–297 (2020).

    Google Scholar 

  143. Singh, A. & Bokes, P. Consequences of mRNA transport on stochastic variability in protein levels. Biophys. J. 103, 1087–1096 (2012).

    ADS  Google Scholar 

  144. Kim, J. K. & Marioni, J. C. Inferring the kinetics of stochastic gene expression from single-cell RNA-sequencing data. Genome Biol. 14, R7 (2013).

    Google Scholar 

  145. Larsson, A. J. M. M. et al. Genomic encoding of transcriptional burst kinetics. Nature 565, 251–254 (2019).

    ADS  Google Scholar 

  146. Furlan, M., De Pretis, S. & Pelizzola, M. Dynamics of transcriptional and post-transcriptional regulation. Brief. Bioinform. 22, bbaa389 (2021).

    Google Scholar 

  147. Dixit, A. et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866.e17 (2016).

    Google Scholar 

  148. Jaitin, D. A. et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896.e15 (2016).

    Google Scholar 

  149. Jin, X. et al. In vivo Perturb-Seq reveals neuronal and glial abnormalities associated with autism risk genes. Science 370, eaaz6063 (2020).

    Google Scholar 

  150. Schraivogel, D. et al. Targeted Perturb-seq enables genome-scale genetic screens in single cells. Nat. Methods 17, 629–635 (2020).

    Google Scholar 

  151. Hein, M. Y. & Weissman, J. S. Functional single-cell genomics of human cytomegalovirus infection. Nat. Biotechnol. 40, 391–401 (2022).

    Google Scholar 

  152. Frangieh, C. J. et al. Multimodal pooled Perturb-CITE-seq screens in patient models define mechanisms of cancer immune evasion. Nat. Genet. 53, 332–341 (2021).

    Google Scholar 

  153. Legut, M. et al. A genome-scale screen for synthetic drivers of T cell proliferation. Nature 603, 728–735 (2022).

    ADS  Google Scholar 

  154. Haukenes, G., Szilvay, A. M., Brokstad, K. A., Kanestrom, A. & Kalland, K. H. Labeling of RNA transcripts of eukaryotic cells in culture with BrUTP using a liposome transfection reagent (DOTAP). Biotechniques 22, 308–312 (1997).

    Google Scholar 

  155. Kageyama, S., Nagata, M. & Aoki, F. Isolation of nascent messenger RNA from mouse preimplantation embryos. Biol.Reprod. 71, 1948–1955 (2004).

    Google Scholar 

  156. Yamada, T. et al. 5′-bromouridine IP chase (BRIC)-seq to determine RNA half-lives. Methods Mol. Biol. 1720, 1–13 (2018).

    Google Scholar 

  157. Melvin, W. T., Milne, H. B., Slater, A. A., Allen, H. J. & Keir, H. M. Incorporation of 6-thioguanosine and 4-thiouridine into RNA. Application to isolation of newly synthesised RNA by affinity chromatography. Eur. J Biochem. 92, 373–379 (1978).

    Google Scholar 

  158. Jao, C. Y. & Salic, A. Exploring RNA transcription and turnover in vivo by using click chemistry. Proc. Natl Acad. Sci. USA 105, 15779–15784 (2008).

    ADS  Google Scholar 

  159. Hafner, M. et al. PAR-CliP-a method to identify transcriptome-wide the binding sites of RNA binding proteins. J. Vis. Exp. https://doi.org/10.3791/2034 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by the European Research Council (ERC-2016-CoG 721016–HERPES and ERC-2022-CoG 101041177–DecipherHSV to L.D.), the Deutsche Forschungsgemeinschaft (DFG) to F.E. (ER 927/2-1) and C.C.F. (FR 2938/9-1) and the Amar Foundation to B.K.P. A.-E.S. is supported by the Bundesministerium für Bildung und Forschung (BMBF, HOPARL (COMPLS4-025)) and NIH NHGRI R01. F.E., A.-E.S. and L.D. are jointly supported by the DFG CRC1525 (453989101) and by the FOR-COVID (Bayerisches Staatsministerium für Wissenschaft und Kunst). E.A.M. is supported by a Wellcome Trust Senior Investigator award (219475/Z/19/Z) and CRUK awards (C13474 and A27826). I.A. is an Eden and Steven Romick Professorial Chair, supported by Merck KGaA, Darmstadt, Germany, the Chan Zuckerberg Initiative (CZI), the HHMI International Scholar award, the ERC Consolidator Grant (ERC-COG) 724471 HemTree2.0, an SCA award of the Wolfson Foundation and Family Charitable Trust, the Helen and Martin Kimmel award for innovative investigation, the NeuroMac DFG/Transregional Collaborative Research Center Grant. This work was supported by the Austrian Science Fund FWF (P31691 and F8011-B to R.M.; P33936 and F8009-B to A.L.). The Helmholtz Institute for RNA-based Infection Research (HIRI) supported this work with a seed grant through funds from the Bavarian Ministry of Economic Affairs and Media, Energy and Technology (grant allocation nos. 0703/68674/5/2017 and 0703/89374/3/2017).

Author information

Authors and Affiliations

Authors

Contributions

Introduction (L.D., F.E., A.-E.S., A.L., T.H., B.K.P., C.C.F., I.A. and R.M.); Experimentation (L.D., F.E., A.-E.S., A.L. and R.M.); Results (L.D., F.E. and C.C.F.); Applications (L.D., F.E., A.-E.S., C.T., T.H., B.K.P., D.K., K.A., E.A.M. and I.A.); Reproducibility and data deposition (L.D., F.E. and C.C.F.); Limitations and optimizations (L.D., F.E., A.-E.S., A.L., C.T., D.K., K.A., E.A.M., C.C.F., I.A. and R.M.); Figures (R.M., F.E. and A.-E.S.); Outlook (L.D., F.E., A.-E.S., A.L., C.T., T.H., B.K.P., D.K., K.A., E.A.M., C.C.F., I.A. and R.M.); Overview of the Primer (L.D.).

Corresponding authors

Correspondence to Florian Erhard, Antoine-Emmanuel Saliba or Lars Dölken.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Methods Primers thanks Wei Chen, Xing Chen, Xiaohui Fan 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.

Related links

Jupyter Notebook: https://jupyter.org/

Zenodo: https://zenodo.org/

Glossary

Bulk RNA-seq

Transcriptomics analysis of pooled cell populations, tissue sections or biopsy samples.

Nucleotide salvage pathways

Used to recover bases and nucleosides that are formed during degradation of RNA and DNA.

Prodrugs

Medications or compounds that are metabolized in the body into pharmacologically active drugs.

Click chemistry

A growing class of biocompatible chemical reactions for bioconjugation that are high in yield, wide in scope and simple to perform in easily removable or benign solvents under ambient temperatures.

Pulse-labelling

Metabolic labelling of a biological target molecule (in this case, RNA) with a chemically modified compound by administering the compound to the cell culture medium or injecting it into a living organism.

Pulse-chasing

Monitoring active degradation of a target molecule (in this case, RNA) after pulse-labelling to determine its decay rate.

Paired-end sequencing

Sequencing the cloned complementary DNA (cDNA) fragments from both ends enables the detection and removal of sequencing errors in the overlapping sequence and is necessary for reliable identification of nucleotide conversions in metabolically labelled RNA molecules.

Unique molecular identifiers

Random barcode sequences introduced during reverse transcription to distinguish multiple copies of the same RNA from PCR duplicates, providing an absolute quantification of transcript numbers of a given gene per single cell.

BAM format

Binary and compressed file format to store genomic coordinates along with specific information on each read — sequence and quality scores — and its alignment with the reference sequence.

RNA velocity

Temporal information contained in intronic reads (nascent RNA) of single-cell RNA sequencing data to predict future cell states and infer cell trajectories.

Off–on switches

Initiation (off–on) or abrogation (on–off) of transcription in individual cells in response to a stimulus that can be differentiated from a general increase (up) or decrease (down) in transcriptional activity in all cells.

Transcriptional bursting

A fundamental property of genes in which transcription from DNA to RNA occurs in pulses (‘bursts’).

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

Erhard, F., Saliba, AE., Lusser, A. et al. Time-resolved single-cell RNA-seq using metabolic RNA labelling. Nat Rev Methods Primers 2, 77 (2022). https://doi.org/10.1038/s43586-022-00157-z

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43586-022-00157-z

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