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.

  • Review Article
  • Published:

Navigating the pitfalls of mapping DNA and RNA modifications

Abstract

Chemical modifications to nucleic acids occur across the kingdoms of life and carry important regulatory information. Reliable high-resolution mapping of these modifications is the foundation of functional and mechanistic studies, and recent methodological advances based on next-generation sequencing and long-read sequencing platforms are critical to achieving this aim. However, mapping technologies may have limitations that sometimes lead to inconsistent results. Some of these limitations are technical in nature and specific to certain types of technology. Here, however, we focus on common (yet not always widely recognized) pitfalls that are shared among frequently used mapping technologies and discuss strategies to help technology developers and users mitigate their effects. Although the emphasis is primarily on DNA modifications, RNA modifications are also discussed.

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: DNA and RNA modification mapping methods based on next-generation sequencing and long-read sequencing technologies.
Fig. 2: Experimental pitfalls that can lead to false positive calls during modification mapping.
Fig. 3: Analytical pitfalls that can lead to false positive calls during modification mapping.
Fig. 4: Overview of pitfalls that can lead to false negatives during modification mapping.
Fig. 5: Mitigating false positive mapping calls.

Similar content being viewed by others

References

  1. Greenberg, M. V. C. & Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Michalak, E. M., Burr, M. L., Bannister, A. J. & Dawson, M. A. The roles of DNA, RNA and histone methylation in ageing and cancer. Nat. Rev. Mol. Cell Biol. 20, 573–589 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Jiang, X. et al. The role of m6A modification in the biological functions and diseases. Signal Transduct. Target. Ther. 6, 74 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sánchez-Romero, M. A. & Casadesús, J. The bacterial epigenome. Nat. Rev. Microbiol. 18, 7–20 (2020). This review summarizes epigenetic regulation by bacterial DNA methylation and its contribution to phenotypic heterogeneity in bacterial populations.

    Article  PubMed  Google Scholar 

  5. Luo, C., Hajkova, P. & Ecker, J. R. Dynamic DNA methylation: in the right place at the right time. Science 361, 1336–1340 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wu, X. & Zhang, Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nat. Rev. Genet. 18, 517–534 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19, 371–384 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Beaulaurier, J., Schadt, E. E. & Fang, G. Deciphering bacterial epigenomes using modern sequencing technologies. Nat. Rev. Genet. 20, 157–172 (2019). This review discusses the potential of currently available methods, especially LRS technologies, for mapping and characterizing bacterial methylomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Blow, M. J. et al. The epigenomic landscape of prokaryotes. PLoS Genet. 12, 1–28 (2016).

    Article  Google Scholar 

  10. Boulias, K. & Greer, E. L. Means, mechanisms and consequences of adenine methylation in DNA. Nat. Rev. Genet. https://doi.org/10.1038/s41576-022-00456-x (2022).

    Article  PubMed  Google Scholar 

  11. Fang, G. et al. Genome-wide mapping of methylated adenine residues in pathogenic Escherichia coli using single-molecule real-time sequencing. Nat. Biotechnol. 30, 1232–1239 (2012). This paper applies SMRT sequencing to map 6mA in a bacterium at genome-wide scale and highlights the importance of FDR evaluation.

    Article  CAS  PubMed  Google Scholar 

  12. Beaulaurier, J. et al. Metagenomic binning and association of plasmids with bacterial host genomes using DNA methylation. Nat. Biotechnol. 36, 61–69 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Kumar, S. & Mohapatra, T. Deciphering epitranscriptome: modification of mRNA bases provides a new perspective for post-transcriptional regulation of gene expression. Front. Cell Dev. Biol. 9, 1–22 (2021).

    Article  Google Scholar 

  14. Barbieri, I. & Kouzarides, T. Role of RNA modifications in cancer. Nat. Rev. Cancer 20, 303–322 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Helm, M. & Motorin, Y. Detecting RNA modifications in the epitranscriptome: predict and validate. Nat. Rev. Genet. 18, 275–291 (2017). This review discusses the principles, advantages and drawbacks of new high-throughput methods for characterizing RNA modifications.

    Article  CAS  PubMed  Google Scholar 

  16. Frye, M., Jaffrey, S. R., Pan, T., Rechavi, G. & Suzuki, T. RNA modifications: what have we learned and where are we headed? Nat. Rev. Genet. 17, 365–372 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Roundtree, I. A., Evans, M. E., Pan, T. & He, C. Dynamic RNA modifications in gene expression regulation. Cell 169, 1187–1200 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Grozhik, A. V. & Jaffrey, S. R. Distinguishing RNA modifications from noise in epitranscriptome maps. Nat. Chem. Biol. 14, 215–225 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Saletore, Y. et al. The birth of the epitranscriptome: deciphering the function of RNA modifications. Genome Biol. 13, 175 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zaccara, S., Ries, R. J. & Jaffrey, S. R. Reading, writing and erasing mRNA methylation. Nat. Rev. Mol. Cell Biol. 20, 608–624 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. He, C. Grand challenge commentary: RNA epigenetics? Nat. Chem. Biol. 6, 863–865 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Xue, C. et al. Role of main RNA modifications in cancer: N6-methyladenosine, 5-methylcytosine, and pseudouridine. Signal Transduct. Target. Ther. 7, 142 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Tretyakova, N., Villalta, P. W. & Kotapati, S. Mass spectrometry of structurally modified DNA. Chem. Rev. 113, 2395–2436 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Boulias, K. & Greer, E. L. in DNA Modifications: Methods and Protocols (eds Ruzov, A. & Gering, M.) 79–90 (Springer US, 2021).

  26. Eid, J. et al. Real-time DNA sequencing from single polymerase molecules. Science 323, 133–138 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Flusberg, B. A. et al. Direct detection of DNA methylation during single-molecule, real-time sequencing. Nat. Methods 7, 461–465 (2010). This paper provides an early description of direct mapping of 5mC, 5hmC and 6mA using SMRT sequencing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Deamer, D., Akeson, M. & Branton, D. Three decades of nanopore sequencing. Nat. Biotechnol. 34, 518–524 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wang, Y., Zhao, Y., Bollas, A., Wang, Y. & Au, K. F. Nanopore sequencing technology, bioinformatics and applications. Nat. Biotechnol. 39, 1348–1365 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Laszlo, A. H. et al. Detection and mapping of 5-methylcytosine and 5-hydroxymethylcytosine with nanopore MspA. Proc. Natl Acad. Sci. USA 110, 18904–18909 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Amarasinghe, S. L. et al. Opportunities and challenges in long-read sequencing data analysis. Genome Biol. 21, 1–16 (2020).

    Article  Google Scholar 

  32. Krueger, F., Kreck, B., Franke, A. & Andrews, S. R. DNA methylome analysis using short bisulfite sequencing data. Nat. Methods 9, 145–151 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Darst, R. P., Pardo, C. E., Ai, L., Brown, K. D. & Kladde, M. P. Bisulfite sequencing of DNA. Curr. Protoc. Mol. Biol. https://doi.org/10.1002/0471142727.mb0709s91 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Shi, D. Q., Ali, I., Tang, J. & Yang, W. C. New insights into 5hmC DNA modification: generation, distribution and function. Front. Genet. 8, 1–11 (2017).

    Article  Google Scholar 

  35. Amente, S. et al. Genome-wide mapping of genomic DNA damage: methods and implications. Cell. Mol. Life Sci. 78, 6745–6762 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rybin, M. J. et al. Emerging technologies for genome-wide profiling of DNA breakage. Front. Genet. 11, 610386 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Zhao, L. Y., Song, J., Liu, Y., Song, C. X. & Yi, C. Mapping the epigenetic modifications of DNA and RNA. Protein Cell 11, 792–808 (2020). This review summarizes high-throughput NGS-based methods for mapping five forms of DNA modifications and eight forms of RNA modifications, and also summarizes biological discoveries made using these methods.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. O’Brown, Z. K. et al. Sources of artifact in measurements of 6mA and 4mC abundance in eukaryotic genomic DNA. BMC Genomics 20, 1–15 (2019). This article reports sources of artefacts in measurements of 6mA and 4mC abundance in eukaryotic gDNA, including both quantification methods and mapping methods.

    Article  Google Scholar 

  39. Kong, Y. et al. Critical assessment of DNA adenine methylation in eukaryotes using quantitative deconvolution. Science 375, 515–522 (2022). This paper describes a machine learning method that can quantitatively deconvolve 6mA events into eukaryotic species of interest and other sources, and warns about bacterial contamination in the study of 6mA in eukaryotic samples.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Patil, V. et al. Human mitochondrial DNA is extensively methylated in a non-CpG context. Nucleic Acids Res. 47, 10072–10085 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bellizzi, D. et al. The control region of mitochondrial DNA shows an unusual CpG and non-CpG methylation pattern. DNA Res. 20, 537–547 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dou, X. et al. The strand-biased mitochondrial DNA methylome and its regulation by DNMT3A. Genome Res. 29, 1622–1634 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sharma, N., Pasala, M. S. & Prakash, A. Mitochondrial DNA: epigenetics and environment. Environ. Mol. Mutagen. 60, 668–682 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Owa, C., Poulin, M., Yan, L. & Shioda, T. Technical adequacy of bisulfite sequencing and pyrosequencing for detection of mitochondrial DNA methylation: sources and avoidance of false-positive detection. PLoS ONE 13, 1–19 (2018).

    Article  Google Scholar 

  45. Bicci, I., Calabrese, C., Golder, Z. J., Gomez-Duran, A. & Chinnery, P. F. Single-molecule mitochondrial DNA sequencing shows no evidence of CpG methylation in human cells and tissues. Nucleic Acids Res. 49, 12757–12768 (2021). This paper describes the bias and technical concerns related to BS-seq in mapping 5mC in mtDNA and reports more reliable 5mC levels estimated by machine learning modelling of nanopore data.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mechta, M., Ingerslev, L. R., Fabre, O., Picard, M. & Barrès, R. Evidence suggesting absence of mitochondrial DNA methylation. Front. Genet. 8, 1–9 (2017).

    Article  Google Scholar 

  47. Matsuda, S. et al. Accurate estimation of 5-methylcytosine in mammalian mitochondrial DNA. Sci. Rep. 8, 1–13 (2018).

    Article  Google Scholar 

  48. Hong, E. E., Okitsu, C. Y., Smith, A. D. & Hsieh, C.-L. Regionally specific and genome-wide analyses conclusively demonstrate the absence of CpG methylation in human mitochondrial DNA. Mol. Cell. Biol. 33, 2683–2690 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cao, K., Feng, Z., Gao, F., Zang, W. & Liu, J. Mitoepigenetics: an intriguing regulatory layer in aging and metabolic-related diseases. Free. Radic. Biol. Med. 177, 337–346 (2021).

    Article  CAS  PubMed  Google Scholar 

  50. Stoccoro, A. & Coppedè, F. Mitochondrial DNA methylation and human diseases. Int. J. Mol. Sci. 22, 1–27 (2021).

    Article  Google Scholar 

  51. Lopes, A. F. C. Mitochondrial metabolism and DNA methylation: a review of the interaction between two genomes. Clin. Epigenetics 12, 1–13 (2020).

    Article  Google Scholar 

  52. Zhang, G. et al. N6-Methyladenine DNA modification in Drosophila. Cell 161, 893–906 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Wu, T. P. et al. DNA methylation on N6-adenine in mammalian embryonic stem cells. Nature 532, 329–333 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Greer, E. L. et al. DNA methylation on N6-adenine in C. elegans. Cell 161, 868–878 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhou, C. et al. Identification and analysis of adenine N6-methylation sites in the rice genome. Nat. Plants 4, 554–563 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. Hao, Z. et al. N6-Deoxyadenosine methylation in mammalian mitochondrial DNA. Mol. Cell https://doi.org/10.1016/j.molcel.2020.02.018 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Wang, Y., Chen, X., Sheng, Y., Liu, Y. & Gao, S. N6-Adenine DNA methylation is associated with the linker DNA of H2A.Z-containing well-positioned nucleosomes in Pol II-transcribed genes in Tetrahymena. Nucleic Acids Res. 45, 11594–11606 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Fu, Y. et al. N6-Methyldeoxyadenosine marks active transcription start sites in Chlamydomonas. Cell 161, 879–892 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Xie, Q. et al. N6-Methyladenine DNA modification in glioblastoma. Cell 175, 1228–1243.e20 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Xiao, C. L. et al. N6-Methyladenine DNA modification in the human genome. Mol. Cell 71, 306–318.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  61. Lentini, A. et al. A reassessment of DNA-immunoprecipitation-based genomic profiling. Nat. Methods 15, 499–504 (2018). This paper demonstrates systematic off-target binding of antibodies to unmodified short tandem repeats in DIP-seq studies and highlights the importance of using matched IgG negative controls.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Douvlataniotis, K., Bensberg, M., Lentini, A., Gylemo, B. & Nestor, C. E. No evidence for DNA N6-methyladenine in mammals. Sci. Adv. 6, 1–10 (2020). This article reports that DIP-seq libraries can be contaminated with mammalian mRNA, and that high abundance of m6A in mRNA can contribute to false positive 6mA peaks in DIP-seq.

    Article  Google Scholar 

  63. Musheev, M. U., Baumgärtner, A., Krebs, L. & Niehrs, C. The origin of genomic N6-methyl-deoxyadenosine in mammalian cells. Nat. Chem. Biol. 16, 630–634 (2020).

    Article  CAS  PubMed  Google Scholar 

  64. Foox, J. et al. The SEQC2 epigenomics quality control (EpiQC) study. Genome Biol. 22, 1–30 (2021).

    Google Scholar 

  65. Skvortsova, K. et al. Comprehensive evaluation of genome-wide 5-hydroxymethylcytosine profiling approaches in human DNA. Epigenetics Chromatin 10, 1–20 (2017).

    Article  Google Scholar 

  66. Branco, M. R., Ficz, G. & Reik, W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat. Rev. Genet. 13, 7–13 (2012).

    Article  CAS  Google Scholar 

  67. Olova, N. et al. Comparison of whole-genome bisulfite sequencing library preparation strategies identifies sources of biases affecting DNA methylation data. Genome Biol. 19, 1–19 (2018).

    Article  Google Scholar 

  68. Ramsahoye, B. H. et al. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc. Natl Acad. Sci. USA 97, 5237–5242 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Patil, V., Ward, R. L. & Hesson, L. B. The evidence for functional non-CpG methylation in mammalian cells. Epigenetics 9, 823–828 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Jang, H. S., Shin, W. J., Lee, J. E. & Do, J. T. CpG and non-CpG methylation in epigenetic gene regulation and brain function. Genes 8, 2–20 (2017).

    Article  Google Scholar 

  71. Zhang, Y. et al. Large-scale comparative epigenomics reveals hierarchical regulation of non-CG methylation in Arabidopsis. Proc. Natl Acad. Sci. USA 115, E1069–E1074 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Morrison, J. et al. Evaluation of whole-genome DNA methylation sequencing library preparation protocols. Epigenetics Chromatin 14, 1–15 (2021).

    Article  Google Scholar 

  73. Robertson, K. D. DNA methylation and human disease. Nat. Rev. Genet. 6, 597–610 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Suzuki, M. M. & Bird, A. DNA methylation landscapes: provocative insights from epigenomics. Nat. Rev. Genet. 9, 465–476 (2008).

    Article  CAS  PubMed  Google Scholar 

  75. Schaefer, M., Pollex, T., Hanna, K. & Lyko, F. RNA cytosine methylation analysis by bisulfite sequencing. Nucleic Acids Res 37, e12 (2009).

    Article  PubMed  Google Scholar 

  76. Legrand, C. et al. Statistically robust methylation calling for whole transcriptome bisulfite sequencing reveals distinct methylation patterns for mouse RNAs. Genome Res. 27, 1589–1596 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Huang, T., Chen, W., Liu, J., Gu, N. & Zhang, R. Genome-wide identification of mRNA 5-methylcytosine in mammals. Nat. Struct. Mol. Biol. 26, 380–388 (2019).

    Article  CAS  PubMed  Google Scholar 

  78. Schaefer, M. RNA 5-methylcytosine analysis by bisulfite sequencing. Methods Enzymol. 560, 297–329 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Trixl, L., Rieder, D., Amort, T. & Lusser, A. in Epitranscriptomics (eds Wajapeyee, N. & Gupta, R.) 1–21 (Springer, 2019).

  80. Carlile, T. M. et al. Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells. Nature 515, 143–146 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Li, X. et al. Chemical pulldown reveals dynamic pseudouridylation of the mammalian transcriptome. Nat. Chem. Biol. 11, 592–597 (2015).

    Article  CAS  PubMed  Google Scholar 

  82. Wiener, D. & Schwartz, S. The epitranscriptome beyond m6A. Nat. Rev. Genet. 22, 119–131 (2021). This Perspective discusses the technical and analytical challenges that led to inconsistent conclusions regarding the abundance and distribution of six forms of RNA modifications beyond m6A.

    Article  CAS  PubMed  Google Scholar 

  83. Luo, G. Z. et al. Characterization of eukaryotic DNA N6-methyladenine by a highly sensitive restriction enzyme-assisted sequencing. Nat. Commun. 7, 2–7 (2016).

    Article  Google Scholar 

  84. Cohen-Karni, D. et al. The MspJI family of modification-dependent restriction endonucleases for epigenetic studies. Proc. Natl Acad. Sci. USA 108, 11040–11045 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Sun, Z. et al. A sensitive approach to map genome-wide 5-hydroxymethylcytosine and 5-formylcytosine at single-base resolution. Mol. Cell 57, 750–761 (2015).

    Article  CAS  PubMed  Google Scholar 

  86. Roberts, R. J., Vincze, T., Posfai, J. & Macelis, D. REBASE — a database for DNA restriction and modification: enzymes, genes and genomes. Nucleic Acids Res. 43, D298–D299 (2015).

    Article  CAS  PubMed  Google Scholar 

  87. Zhang, Z. et al. Single-base mapping of m6A by an antibody-independent method. Sci. Adv. 5, 1–12 (2019).

    Article  Google Scholar 

  88. Garcia-Campos, M. A. et al. Deciphering the “m6A Code” via antibody-independent quantitative profiling. Cell 178, 731–747.e16 (2019).

    Article  CAS  PubMed  Google Scholar 

  89. Zhang, Z. et al. Systematic calibration of epitranscriptomic maps using a synthetic modification-free RNA library. Nat. Methods 18, 1213–1222 (2021).

    Article  CAS  PubMed  Google Scholar 

  90. Dominissini, D. et al. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature 530, 441–446 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Safra, M. et al. The m1A landscape on cytosolic and mitochondrial mRNA at single-base resolution. Nature 551, 251–255 (2017).

    Article  CAS  PubMed  Google Scholar 

  92. Jin, H., Huo, C., Zhou, T. & Xie, S. m1A RNA modification in gene expression regulation. Genes 13, 910 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Wei, J. et al. Differential m6A, m6Am, and m1A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol. Cell 71, 973–985.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Cui, X. et al. 5-Methylcytosine RNA methylation in Arabidopsis thaliana. Mol. Plant. 10, 1387–1399 (2017).

    Article  CAS  PubMed  Google Scholar 

  95. Ma, J. et al. M5C-Atlas: a comprehensive database for decoding and annotating the 5-methylcytosine (m5C) epitranscriptome. Nucleic Acids Res. 50, D196–D203 (2022).

    Article  CAS  PubMed  Google Scholar 

  96. Lin, S., Choe, J., Du, P., Triboulet, R. & Gregory, R. I. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell 62, 335–345 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Gokhale, N. S. et al. N6-Methyladenosine in flaviviridae viral RNA genomes regulates infection. Cell Host Microbe 20, 654–665 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Delatte, B. et al. Transcriptome-wide distribution and function of RNA hydroxymethylcytosine. Science 351, 282–285 (2016).

    Article  CAS  PubMed  Google Scholar 

  101. Wu, H. et al. Genome-wide analysis of 5-hydroxymethylcytosine distribution reveals its dual function in transcriptional regulation in mouse embryonic stem cells. Genes Dev. 25, 679–684 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Li, W., Li, X., Ma, X., Xiao, W. & Zhang, J. Mapping the m1A, m5C, m6A and m7G methylation atlas in zebrafish brain under hypoxic conditions by MeRIP-seq. BMC Genomics 23, 1–19 (2022).

    Google Scholar 

  103. Song, C. X., Yi, C. & He, C. Mapping recently identified nucleotide variants in the genome and transcriptome. Nat. Biotechnol. 30, 1107–1116 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Arango, D. et al. Acetylation of cytidine in mRNA promotes translation efficiency. Cell 175, 1872–1886.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Sas-Chen, A. et al. Dynamic RNA acetylation revealed by quantitative cross-evolutionary mapping. Nature 583, 638–643 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Thalalla Gamage, S., Sas-Chen, A., Schwartz, S. & Meier, J. L. Quantitative nucleotide resolution profiling of RNA cytidine acetylation by ac4C-seq. Nat. Protoc. 16, 2286–2307 (2021).

    Article  CAS  PubMed  Google Scholar 

  107. Grozhik, A. V. et al. Antibody cross-reactivity accounts for widespread appearance of m1A in 5′UTRs. Nat. Commun. 10, 1–13 (2019).

    Article  CAS  Google Scholar 

  108. Zhu, S. et al. Mapping and characterizing N6-methyladenine in eukaryotic genomes using single-molecule real-time sequencing. Genome Res. 28, 1067–1078 (2018). This article demonstrates that sequencing technologies tend to make false positive 6mA calls in eukaryotes, and highlights the importance of FDR evaluation for low-abundance modifications.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Liu, X. et al. N6-Methyladenine is incorporated into mammalian genome by DNA polymerase. Cell Res. 31, 94–97 (2021).

    Article  CAS  PubMed  Google Scholar 

  110. Liu, B., Liu, X., Lai, W. & Wang, H. Metabolically generated stable isotope-labeled deoxynucleoside code for tracing DNA N6-methyladenine in human cells. Anal. Chem. 89, 6202–6209 (2017).

    Article  CAS  PubMed  Google Scholar 

  111. O’Brown, Z. K. & Greer, E. L. in DNA methyltransferases — Role and Function (eds Jeltsch, A. & Jurkowska, R. Z.) 213–246 (Springer International, 2016).

  112. Schiffers, S. et al. Quantitative LC-MS provides no evidence for m6dA or m4dC in the genome of mouse embryonic stem cells and tissues. Angew. Chem. — Int. Ed. 56, 11268–11271 (2017).

    Article  CAS  Google Scholar 

  113. Tan, S. C. & Yiap, B. C. DNA, RNA, and protein extraction: the past and the present. J. Biomed. Biotechnol. 2009, 574398 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Edelheit, S., Schwartz, S., Mumbach, M. R., Wurtzel, O. & Sorek, R. Transcriptome-wide mapping of 5-methylcytidine RNA modifications in bacteria, archaea, and yeast reveals m5C within archaeal mRNAs. PLoS Genet 9, e1003602 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Thu, K. L. et al. Methylated DNA immunoprecipitation. J. Vis. Exp. https://doi.org/10.3791/935 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Amort, T. & Lusser, A. in RNA Methylation (ed. Lusser, A.) 107–121 (Springer, 2017).

  117. Meyer, C. A. & Liu, X. S. Identifying and mitigating bias in next-generation sequencing methods for chromatin biology. Nat. Rev. Genet. 15, 709–721 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sims, D., Sudbery, I., Ilott, N. E., Heger, A. & Ponting, C. P. Sequencing depth and coverage: key considerations in genomic analyses. Nat. Rev. Genet. 15, 121–132 (2014).

    Article  CAS  PubMed  Google Scholar 

  119. McIntyre, A. B. R. et al. Limits in the detection of m6A changes using MeRIP/m6A-seq. Sci. Rep. 10, 1–15 (2020).

    Article  Google Scholar 

  120. Beaulaurier, J. et al. Single molecule-level detection and long read-based phasing of epigenetic variations in bacterial methylomes. Nat. Commun. 6, 7438 (2015).

    Article  CAS  PubMed  Google Scholar 

  121. Schadt, E. E. et al. Modeling kinetic rate variation in third generation DNA sequencing data to detect putative modifications to DNA bases. Genome Res. 23, 129–141 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Vilfan, I. D. et al. Analysis of RNA base modification and structural rearrangement by single-molecule real-time detection of reverse transcription. J. Nanobiotechnol. 11, 1 (2013).

    Article  Google Scholar 

  123. Abdi, E., Latifi-Navid, S. & Latifi-Navid, H. Long noncoding RNA polymorphisms and colorectal cancer risk: progression and future perspectives. Environ. Mol. Mutagen. 63, 98–112 (2022).

    Article  CAS  PubMed  Google Scholar 

  124. Pratanwanich, P. N. et al. Identification of differential RNA modifications from nanopore direct RNA sequencing with xPore. Nat. Biotechnol. 39, 1394–1402 (2021).

    Article  CAS  PubMed  Google Scholar 

  125. Jenjaroenpun, P. et al. Decoding the epitranscriptional landscape from native RNA sequences. Nucleic Acids Res. 49, 1–13 (2021).

    Article  Google Scholar 

  126. Price, A. M. et al. Direct RNA sequencing reveals m6A modifications on adenovirus RNA are necessary for efficient splicing. Nat. Commun. 11, 6016 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Parker, M. T. et al. Nanopore direct RNA sequencing maps the complexity of arabidopsis mRNA processing and m6A modification. Elife 9, 1–35 (2020).

    Article  Google Scholar 

  128. Liu, H. et al. Accurate detection of m6A RNA modifications in native RNA sequences. Nat. Commun. 10, 1–9 (2019).

    Google Scholar 

  129. Kimura, S., Dedon, P. C. & Waldor, M. K. Comparative tRNA sequencing and RNA mass spectrometry for surveying tRNA modifications. Nat. Chem. Biol. 16, 964–972 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Ameur, A., Kloosterman, W. P. & Hestand, M. S. Single-molecule sequencing: towards clinical applications. Trends Biotechnol. 37, 72–85 (2019).

    Article  CAS  PubMed  Google Scholar 

  131. Fang, L. et al. DeepRepeat: direct quantification of short tandem repeats on signal data from nanopore sequencing. Genome Biol. 23, 1–27 (2022).

    Article  Google Scholar 

  132. Shi, H., Wei, J. & He, C. Where, when, and how: context-dependent functions of RNA methylation writers, readers, and erasers. Mol. Cell 74, 640–650 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Huang, Y. et al. The behaviour of 5-hydroxymethylcytosine in bisulfite sequencing. PLoS ONE 5, 1–9 (2010).

    Google Scholar 

  134. Liu, Y. et al. DNA methylation-calling tools for Oxford Nanopore sequencing: a survey and human epigenome-wide evaluation. Genome Biol. 22, 295 (2021). This paper benchmarks computational methods for detecting 5mC in mammalian genomes using nanopore sequencing, and demonstrates that 5hmC levels may contribute to the discrepancy between BS-seq and nanopore sequencing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Clark, T. A., Spittle, K. E., Turner, S. W. & Korlach, J. Direct detection and sequencing of damaged DNA bases. Genome Integr. 2, 10 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Mauer, J. et al. Reversible methylation of m6Am in the 5′ cap controls mRNA stability. Nature 541, 371–375 (2017).

    Article  CAS  PubMed  Google Scholar 

  137. Sun, H. et al. m6Am-seq reveals the dynamic m6Am methylation in the human transcriptome. Nat. Commun. 12, 1–12 (2021).

    Google Scholar 

  138. Guiblet, W. M. et al. Long-read sequencing technology indicates genome-wide effects of non-B DNA on polymerization speed and error rate. Genome Res. 28, 1767–1778 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Kwok, C. K., Tang, Y., Assmann, S. M. & Bevilacqua, P. C. The RNA structurome: transcriptome-wide structure probing with next-generation sequencing. Trends Biochem. Sci. 40, 221–232 (2015).

    Article  CAS  PubMed  Google Scholar 

  140. Mortimer, S. A., Kidwell, M. A. & Doudna, J. A. Insights into RNA structure and function from genome-wide studies. Nat. Rev. Genet. 15, 469–479 (2014).

    Article  CAS  PubMed  Google Scholar 

  141. Wan, Y., Kertesz, M., Spitale, R. C., Segal, E. & Chang, H. Y. Understanding the transcriptome through RNA structure. Nat. Rev. Genet. 12, 641–655 (2011).

    Article  CAS  PubMed  Google Scholar 

  142. Talkish, J., May, G., Lin, Y., Woolford, J. L. & McManus, C. J. Mod-seq: high-throughput sequencing for chemical probing of RNA structure. Rna 20, 713–720 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Weeks, K. M. Advances in RNA structure analysis by chemical probing. Curr. Opin. Struct. Biol. 20, 295–304 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Helm, M., Lyko, F. & Motorin, Y. Limited antibody specificity compromises epitranscriptomic analyses. Nat. Commun. 10, 9–11 (2019).

    Article  Google Scholar 

  145. Zaringhalam, M. & Papavasiliou, F. N. Pseudouridylation meets next-generation sequencing. Methods 107, 63–72 (2016).

    Article  CAS  PubMed  Google Scholar 

  146. Palazzo, A. F. & Lee, E. S. Non-coding RNA: what is functional and what is junk? Front. Genet. 5, 1–11 (2015).

    Google Scholar 

  147. Stark, R., Grzelak, M. & Hadfield, J. RNA sequencing: the teenage years. Nat. Rev. Genet. 20, 631–656 (2019).

    Article  CAS  PubMed  Google Scholar 

  148. Ozsolak, F. & Milos, P. M. RNA sequencing: advances, challenges and opportunities. Nat. Rev. Genet. 12, 87–98 (2011).

    Article  CAS  PubMed  Google Scholar 

  149. Hussain, S., Aleksic, J., Blanco, S., Dietmann, S. & Frye, M. Characterizing 5-methylcytosine in the mammalian epitranscriptome. Genome Biol. 14, 1–10 (2013).

    Article  Google Scholar 

  150. Oliveira, P. H. et al. Epigenomic characterization of Clostridioides difficile finds a conserved DNA methyltransferase that mediates sporulation and pathogenesis. Nat. Microbiol. 5, 166–180 (2020).

    Article  CAS  PubMed  Google Scholar 

  151. Hong, T. et al. Selective detection of N6-methyladenine in DNA via metal ion-mediated replication and rolling circle amplification. Chem. Sci. 8, 200–205 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  152. Engel, J. D. & Von Hippel, P. H. Effects of methylation on the stability of nucleic acid conformations. Studies at the polymer level. J. Biol. Chem. 253, 927–934 (1978).

    Article  CAS  PubMed  Google Scholar 

  153. Clark, T. A. et al. Enhanced 5-methylcytosine detection in single-molecule, real-time sequencing via Tet1 oxidation. BMC Biol. 11, 4 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Chaveza, L. et al. Simultaneous sequencing of oxidized methylcytosines produced by TET/JBP dioxygenases in Coprinopsis cinerea. Proc. Natl Acad. Sci. USA 111, E5149–E5158 (2014).

    Google Scholar 

  155. Simpson, J. T. et al. Detecting DNA cytosine methylation using nanopore sequencing. Nat. Methods 14, 407–410 (2017). This paper describes the use of synthetically methylated DNA to train a hidden Markov model for distinguishing 5mC from unmethylated cytosine in mammalian genomes using nanopore data.

    Article  CAS  PubMed  Google Scholar 

  156. Rand, A. C. et al. Mapping DNA methylation with high-throughput nanopore sequencing. Nat. Methods 14, 411–413 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Tourancheau, A., Mead, E. A., Zhang, X. S. & Fang, G. Discovering multiple types of DNA methylation from bacteria and microbiome using nanopore sequencing. Nat. Methods 18, 491–498 (2021). This paper reports that the nanopore sequencing signal displays complex heterogeneity across methylation events of the same type, and develops a method for de novo detection of three forms of bacterial DNA methylation across diverse sequence contexts.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Ni, P. et al. Genome-wide detection of cytosine methylations in plant from nanopore data using deep learning. Nat. Commun. 12, 1–11 (2021).

    Article  Google Scholar 

  159. Liu, Y. et al. Bisulfite-free direct detection of 5-methylcytosine and 5-hydroxymethylcytosine at base resolution. Nat. Biotechnol. 37, 424–429 (2019).

    Article  CAS  PubMed  Google Scholar 

  160. Liu, Y. et al. Accurate targeted long-read DNA methylation and hydroxymethylation sequencing with TAPS. Genome Biol. 21, 1–9 (2020).

    Article  Google Scholar 

  161. Song, C. X. et al. Sensitive and specific single-molecule sequencing of 5-hydroxymethylcytosine. Nat. Methods 9, 75–77 (2012).

    Article  CAS  Google Scholar 

  162. Xiong, X., Li, X., Wang, K. & Yi, C. Perspectives on topology of the human m1A methylome at single nucleotide resolution. Rna 24, 1437–1442 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Li, X. et al. Transcriptome-wide mapping reveals reversible and dynamic N1-methyladenosine methylome. Nat. Chem. Biol. 12, 311–316 (2016).

    Article  CAS  PubMed  Google Scholar 

  164. Gallego Romero, I., Pai, A. A., Tung, J. & Gilad, Y. RNA-seq: impact of RNA degradation on transcript quantification. BMC Biol. 12, 1–13 (2014).

    Article  Google Scholar 

  165. Schuierer, S. et al. A comprehensive assessment of RNA-seq protocols for degraded and low-quantity samples. BMC Genomics 18, 1–13 (2017).

    Article  Google Scholar 

  166. Siwek, W., Czapinska, H., Bochtler, M., Bujnicki, J. M. & Skowronek, K. Crystal structure and mechanism of action of the N6-methyladenine-dependent type IIM restriction endonuclease R.DpnI. Nucleic Acids Res. 40, 7563–7572 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Mierzejewska, K. et al. Structural basis of the methylation specificity of R.DpnI. Nucleic Acids Res. 42, 8745–8754 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Kazrani, A. A., Kowalska, M., Czapinska, H. & Bochtler, M. Crystal structure of the 5hmC specific endonuclease PvuRts1I. Nucleic Acids Res. 42, 5929–5936 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Ni, P. et al. DeepSignal: detecting DNA methylation state from nanopore sequencing reads using deep-learning. Bioinformatics 35, 4586–4595 (2019).

    Article  CAS  PubMed  Google Scholar 

  170. Liu, Q. et al. Detection of DNA base modifications by deep recurrent neural network on Oxford Nanopore sequencing data. Nat. Commun. 10, 2449 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  171. McIntyre, A. B. R. et al. Single-molecule sequencing detection of N6-methyladenine in microbial reference materials. Nat. Commun. 10, 1–11 (2019).

    Article  Google Scholar 

  172. Yuen, Z. W. S. et al. Systematic benchmarking of tools for CpG methylation detection from nanopore sequencing. Nat. Commun. 12, 1–12 (2021).

    Article  Google Scholar 

  173. Zhang, Y. Z. et al. On the application of BERT models for nanopore methylation detection. Proc. 2021 IEEE Int. Conf. Bioinforma. Biomed. BIBM 2021 https://doi.org/10.1109/BIBM52615.2021.9669841 (2021).

    Article  Google Scholar 

  174. Liu, Q., Georgieva, D. C., Egli, D. & Wang, K. NanoMod: a computational tool to detect DNA modifications using nanopore long-read sequencing data. BMC Genomics 20, 78 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Tse, O. Y. O. et al. Genome-wide detection of cytosine methylation by single molecule real-time sequencing. Proc. Natl Acad. Sci. USA 118, 1–11 (2021).

    Article  Google Scholar 

  176. Ni, P. et al. DNA 5-methylcytosine detection and methylation phasing using PacBio circular consensus sequencing. Preprint at bioRxiv https://doi.org/10.1101/2022.02.26.482074 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  177. Garalde, D. R. et al. Highly parallel direct RN A sequencing on an array of nanopores. Nat. Methods 15, 201–206 (2018). This paper provides an early description of direct RNA sequencing for detecting RNA modifications using the nanopore platform.

    Article  CAS  PubMed  Google Scholar 

  178. Xu, L. & Seki, M. Recent advances in the detection of base modifications using the nanopore sequencer. J. Hum. Genet. 65, 25–33 (2020).

    Article  CAS  PubMed  Google Scholar 

  179. Begik, O. et al. Quantitative profiling of pseudouridylation dynamics in native RNAs with nanopore sequencing. Nat. Biotechnol. 39, 1278–1291 (2021).

    Article  CAS  PubMed  Google Scholar 

  180. Lorenz, D. A., Sathe, S., Einstein, J. M. & Yeo, G. W. Direct RNA sequencing enables m6A detection in endogenous transcript isoforms at base-specific resolution. RNA 26, 19–28 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Gao, Y. et al. Quantitative profiling of N6-methyladenosine at single-base resolution in stem-differentiating xylem of Populus trichocarpa using nanopore direct RNA sequencing. Genome Biol. 22, 1–17 (2021).

    Article  Google Scholar 

  182. Qin, H. et al. DENA: training an authentic neural network model using nanopore sequencing data of Arabidopsis transcripts for detection and quantification of N6-methyladenosine on RNA. Genome Biol. 23, 1–23 (2022).

    Article  CAS  Google Scholar 

  183. Leger, A. et al. RNA modifications detection by comparative nanopore direct RNA sequencing. Nat. Commun. 12, 1–17 (2021).

    Article  Google Scholar 

  184. Whalen, S., Schreiber, J., Noble, W. S. & Pollard, K. S. Navigating the pitfalls of applying machine learning in genomics. Nat. Rev. Genet. 23, 169–181 (2022).

    Article  CAS  PubMed  Google Scholar 

  185. Schutsky, E. K. et al. Nondestructive, base-resolution sequencing of 5-hydroxymethylcytosine using a DNA deaminase. Nat. Biotechnol. 36, 1083–1090 (2018).

    Article  CAS  Google Scholar 

  186. Zhang, W., Qian, Y. & Jia, G. The detection and functions of RNA modification m6A based on m6A writers and erasers. J. Biol. Chem. 297, 100973 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Tavakoli, S. et al. Semi-quantitative detection of pseudouridine modifications and type I/II hypermodifications in human mRNAs using direct and long-read sequencing. Preprint at bioRxiv https://doi.org/10.1101/2021.11.03.467190 (2022).

    Article  Google Scholar 

  188. Makhamreh, A. et al. Messenger-RNA modification standards and machine learning models facilitate absolute site-specific pseudouridine quantification. Preprint at bioRxiv https://doi.org/10.1101/2022.05.06.490948 (2022).

    Article  Google Scholar 

  189. Plongthongkum, N., Diep, D. H. & Zhang, K. Advances in the profiling of DNA modifications: cytosine methylation and beyond. Nat. Rev. Genet. 15, 647–661 (2014). This review discusses the principles as well as experimental and analytical challenges in mapping 5mC and its oxidation derivatives with BS-seq and other NGS-based methods.

    Article  CAS  PubMed  Google Scholar 

  190. Lio, C. W. J. & Rao, A. TET enzymes and 5hMC in adaptive and innate immune systems. Front. Immunol. 10, 1–13 (2019).

    Article  CAS  Google Scholar 

  191. Lai, W., Lyu, C. & Wang, H. Vertical ultrafiltration-facilitated DNA digestion for rapid and sensitive UHPLC-MS/MS detection of DNA modifications. Anal. Chem. 90, 6859–6866 (2018).

    Article  CAS  PubMed  Google Scholar 

  192. Liu, X., Lai, W., Zhang, N. & Wang, H. Predominance of N6-methyladenine-specific DNA fragments enriched by multiple immunoprecipitation. Anal. Chem. 90, 5546–5551 (2018).

    Article  CAS  PubMed  Google Scholar 

  193. Debo, B. M., Mallory, B. & Stergachis, A. B. Evaluation of N6-adenine DNA-immunoprecipitation-based genomic profiling in eukaryotes. Preprint at bioRxiv https://doi.org/10.1101/2022.03.02.482749 (2022).

    Article  Google Scholar 

  194. Liang, Z. et al. DNA N6-adenine methylation in Arabidopsis thaliana. Dev. Cell 45, 406–416.e3 (2018).

    Article  CAS  PubMed  Google Scholar 

  195. Luo, G. Z. et al. N6-Methyldeoxyadenosine directs nucleosome positioning in Tetrahymena DNA. Genome Biol. 19, 1–12 (2018).

    Article  Google Scholar 

  196. Mondo, S. J. et al. Widespread adenine N6-methylation of active genes in fungi. Nat. Genet. https://doi.org/10.1038/ng.3859 (2017).

    Article  PubMed  Google Scholar 

  197. Vaisvila, R. et al. Enzymatic methyl sequencing detects DNA methylation at single-base resolution from picograms of DNA. Genome Res. 31, 1280–1289 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  198. Sun, Z. et al. Nondestructive enzymatic deamination enables single-molecule long-read amplicon sequencing for the determination of 5-methylcytosine and 5-hydroxymethylcytosine at single-base resolution. Genome Res. 31, 291–300 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  199. Feng, S., Zhong, Z., Wang, M. & Jacobsen, S. E. Efficient and accurate determination of genome-wide DNA methylation patterns in Arabidopsis thaliana with enzymatic methyl sequencing. Epigenetics Chromatin 13, 1–17 (2020).

    Article  Google Scholar 

  200. Wang, Y. et al. A distinct class of eukaryotic MT-A70 methyltransferases maintain symmetric DNA N6-adenine methylation at the ApT dinucleotides as an epigenetic mark associated with transcription. Nucleic Acids Res. 47, 11771–11789 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Beh, L. Y. et al. Identification of a DNA N6-adenine methyltransferase complex and its impact on chromatin organization. Cell 177, 1781–1796.e25 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Rodriguez, F., Yushenova, I. A., DiCorpo, D. & Arkhipova, I. R. Bacterial N4-methylcytosine as an epigenetic mark in eukaryotic DNA. Nat. Commun. 13, 1–17 (2022).

    Article  Google Scholar 

  203. Dietzsch, J., Feineis, D. & Höbartner, C. Chemoselective labeling and site-specific mapping of 5-formylcytosine as a cellular nucleic acid modification. FEBS Lett. 592, 2032–2047 (2018).

    Article  CAS  PubMed  Google Scholar 

  204. Song, C. X. et al. Genome-wide profiling of 5-formylcytosine reveals its roles in epigenetic priming. Cell 153, 678–691 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Zhu, C. et al. Single-cell 5-formylcytosine landscapes of mammalian early embryos and ESCs at single-base resolution. Cell Stem Cell 20, 720–731.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  206. Shen, L. et al. Genome-wide analysis reveals TET- and TDG-dependent 5-methylcytosine oxidation dynamics. Cell 153, 692–706 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Neri, F., Incarnato, D., Krepelova, A., Parlato, C. & Oliviero, S. Methylation-assisted bisulfite sequencing to simultaneously map 5fC and 5caC on a genome-wide scale for DNA demethylation analysis. Nat. Protoc. 11, 1191–1205 (2016).

    Article  CAS  PubMed  Google Scholar 

  208. Yu, M. et al. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome. Cell 149, 1368–1380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Booth, M. J., Marsico, G., Bachman, M., Beraldi, D. & Balasubramanian, S. Quantitative sequencing of 5-formylcytosine in DNA at single-base resolution. Nat. Chem. 6, 435–440 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Booth, M. J. et al. Quantitative sequencing of 5-methylcytosine and 5-hydroxymethylcytosine at single-base resolution. Science 336, 934–937 (2012).

    Article  CAS  PubMed  Google Scholar 

  211. Füllgrabe, J. et al. Accurate simultaneous sequencing of genetic and epigenetic bases in DNA. Preprint at bioRxiv https://doi.org/10.1101/2022.07.08.499285 (2022).

    Article  Google Scholar 

  212. Shen, L., Song, C. X., He, C. & Zhang, Y. Mechanism and function of oxidative reversal of DNA and RNA methylation. Annu. Rev. Biochem. 83, 585–614 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Wescoe, Z. L., Schreiber, J. & Akeson, M. Nanopores discriminate among five C5-cytosine variants in DNA. J. Am. Chem. Soc. 136, 16582–16587 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  215. Krutyhołowa, R., Zakrzewski, K. & Glatt, S. Charging the code — tRNA modification complexes. Curr. Opin. Struct. Biol. 55, 138–146 (2019).

    Article  PubMed  Google Scholar 

  216. Sloan, K. E. et al. Tuning the ribosome: the influence of rRNA modification on eukaryotic ribosome biogenesis and function. RNA Biol. 14, 1138–1152 (2017).

    Article  PubMed  Google Scholar 

  217. Decatur, W. A. & Fournier, M. J. rRNA modifications and ribosome function. Trends Biochem. Sci. 27, 344–351 (2002).

    Article  CAS  PubMed  Google Scholar 

  218. Sergiev, P. V., Aleksashin, N. A., Chugunova, A. A., Polikanov, Y. S. & Dontsova, O. A. Structural and evolutionary insights into ribosomal RNA methylation. Nat. Chem. Biol. 14, 226–235 (2018).

    Article  CAS  PubMed  Google Scholar 

  219. Anreiter, I., Mir, Q., Simpson, J. T., Janga, S. C. & Soller, M. New twists in detecting mRNA modification dynamics. Trends Biotechnol. 39, 72–89 (2021).

    Article  CAS  PubMed  Google Scholar 

  220. Michaela, F., Bryan, T. H., Mikaela, B. & Chuan, H. RNA modifications modulate gene expression during development. Science 361, 1346–1349 (2018).

    Article  Google Scholar 

  221. Stephenson, W. et al. Direct detection of RNA modifications and structure using single-molecule nanopore sequencing. Cell Genomics 2, 100097 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Yu, F. et al. CFEA: a cell-free epigenome atlas in human diseases. Nucleic Acids Res. 48, D40–D44 (2020).

    Article  CAS  PubMed  Google Scholar 

  223. Li, W. et al. 5-Hydroxymethylcytosine signatures in circulating cell-free DNA as diagnostic biomarkers for human cancers. Cell Res. 27, 1243–1257 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Tian, X. et al. Circulating tumor DNA 5-hydroxymethylcytosine as a novel diagnostic biomarker for esophageal cancer. Cell Res. 28, 597–600 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Siejka-Zielińska, P. et al. Cell-free DNA TAPS provides multimodal information for early cancer detection. Sci. Adv. 7, eabh0534 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  226. Johnson, P., Zhou, Q., Dao, D. Y. & Lo, Y. M. D. Circulating biomarkers in the diagnosis and management of hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 19, 670–681 (2022).

    Article  PubMed  Google Scholar 

  227. Poetsch, A. R. The genomics of oxidative DNA damage, repair, and resulting mutagenesis. Comput. Struct. Biotechnol. J. 18, 207–219 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Chatterjee, N. & Walker, G. C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 58, 235–263 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Huang, R. & Zhou, P. K. DNA damage repair: historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 6, 254 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Abdulhay, N. J. et al. Massively multiplex single-molecule oligonucleosome footprinting. eLife 9, 1–23 (2020).

    Article  Google Scholar 

  231. Shipony, Z. et al. Long-range single-molecule mapping of chromatin accessibility in eukaryotes. Nat. Methods 17, 319–327 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Wang, Y. et al. Single-molecule long-read sequencing reveals the chromatin basis of gene expression. Genome Res. 29, 1329–1342 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Stergachis, A. B., Debo, B. M., Haugen, E., Churchman, L. S. & Stamatoyannopoulos, J. A. Single-molecule regulatory architectures captured by chromatin fiber sequencing. Science 368, 1449–1454 (2020).

    Article  CAS  PubMed  Google Scholar 

  234. Altemose, N. et al. DiMeLo-seq: a long-read, single-molecule method for mapping protein–DNA interactions genome wide. Nat. Methods 19, 711–723 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Ghut, J. et al. Determination of isoform-specific RNA structure with nanopore long reads. Nat. Biotechnol. 39, 336–346 (2020).

    Google Scholar 

  236. Weber, M. et al. Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nat. Genet. 37, 853–862 (2005).

    Article  CAS  PubMed  Google Scholar 

  237. Pomraning, K. R., Smith, K. M. & Freitag, M. Genome-wide high throughput analysis of DNA methylation in eukaryotes. Methods 47, 142–150 (2009).

    Article  CAS  PubMed  Google Scholar 

  238. Ficz, G. et al. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature 473, 398–404 (2011).

    Article  CAS  PubMed  Google Scholar 

  239. Xu, Y. et al. Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells. Mol. Cell 42, 451–464 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Liu, J. et al. Abundant DNA 6mA methylation during early embryogenesis of zebrafish and pig. Nat. Commun. 7, 1–7 (2016).

    Article  CAS  Google Scholar 

  241. Koziol, M. J. et al. Identification of methylated deoxyadenosines in vertebrates reveals diversity in DNA modifications. Nat. Struct. Mol. Biol. 23, 24–30 (2016).

    Article  CAS  PubMed  Google Scholar 

  242. Ando, Y. & Hayashizaki, Y. Restriction landmark genomic scanning. Nat. Protoc. 1, 2774–2783 (2007).

    Article  Google Scholar 

  243. Koike, K., Matsuyama, T. & Ebisuzaki, T. Epigenetics: application of virtual image restriction landmark genomic scanning (Vi-RLGS). FEBS J. 275, 1608–1616 (2008).

    Article  CAS  PubMed  Google Scholar 

  244. Suzuki, M. & Greally, J. M. DNA methylation profiling using HpaII tiny fragment enrichment by ligation-mediated PCR (HELP). Methods 52, 218–222 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Oda, M. & Greally, J. M. in DNA Methylation (ed. Tost, J.) 77–87 (Humana, 2009).

  246. Sun, Z. et al. High-resolution enzymatic mapping of genomic 5-hydroxymethylcytosine in mouse embryonic stem cells. Cell Rep. 3, 567–576 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Jia, Z. et al. A 5-mC dot blot assay quantifying the DNA methylation level of chondrocyte dedifferentiation in vitro. J. Vis. Exp. 2017, 5–9 (2017).

    Google Scholar 

  248. Jin, S. G., Wu, X., Li, A. X. & Pfeifer, G. P. Genomic mapping of 5-hydroxymethylcytosine in the human brain. Nucleic Acids Res. 39, 5015–5024 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Inoue, A., Shen, L., Dai, Q., He, C. & Zhang, Y. Generation and replication-dependent dilution of 5fC and 5caC during mouse preimplantation development. Cell Res. 21, 1670–1676 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Wang, Y. et al. Naphthalimide derivatives as multifunctional molecules for detecting 5-formylpyrimidine by both PAGE analysis and dot-blot assays. Chem. Commun. 54, 1497–1500 (2018).

    Article  CAS  Google Scholar 

  251. Wheldon, L. M. et al. Transient accumulation of 5-carboxylcytosine indicates involvement of active demethylation in lineage specification of neural stem cells. Cell Rep. 7, 1353–1361 (2014).

    Article  CAS  PubMed  Google Scholar 

  252. Shen, L., Liang, Z. & Yu, H. Dot blot analysis of N6-methyladenosine RNA modification levels. Bio-Protoc. 7, 4–8 (2017).

    Article  Google Scholar 

  253. Nagarajan, A., Janostiak, R. & Wajapeyee, N. in Epitranscriptomics (eds Wajapeyee, N. & Gupta, R.) 263–271 (Springer, 2019).

  254. Mishima, E. et al. Immuno-northern blotting: detection of RNA modifications by using antibodies against modified nucleosides. PLoS ONE 10, 1–17 (2015).

    Article  Google Scholar 

  255. Shen, C., Wang, K., Deng, X. & Chen, J. DNA N6-methyldeoxyadenosine in mammals and human disease. Trends Genet. 38, 454–467 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Yu, M. et al. Base-resolution detection of N4-methylcytosine in genomic DNA using 4mC-Tet-assisted-bisulfitesequencing. Nucleic Acids Res. 43, 1–10 (2015).

    CAS  Google Scholar 

  257. Faulk, C. in Toxicoepigenetics (eds McCullough, S. D. & Dolinoy, D. C.) 153–171 (Academic Press, 2018).

Download references

Acknowledgements

The work was funded by R01 HG011095 (G.F.) and R35 GM139655 (G.F.) from the National Institutes of Health (NIH).

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for the article, made substantial contributions to discussions of the content and reviewed and edited the manuscript before submission. Y.K. and G.F. wrote the article.

Corresponding author

Correspondence to Gang Fang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Genetics thanks the anonymous reviewers 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.

Glossary

Endogenous DNA and RNA modifications

DNA and RNA modifications generated during metabolic processes in living organisms, usually catalysed by specific enzymes within the cells.

Exogenous DNA and RNA modifications

DNA and RNA modifications generated by exogenous factors that originated outside the cells, such as externally modified nucleotides that are randomly incorporated during DNA replication, or events that are directly catalysed by exogenous DNA methyltransferases in vitro or in vivo.

False discovery rates

(FDRs). These mathematical concepts describe the expected ratio of the number of false positive classifications to the total number of positive classifications. In the context of mapping DNA or RNA modifications, the FDR refers to the probability of false positive calls among the detected modification events by a mapping method.

False negative

This mathematical concept describes when a test result incorrectly indicates the absence of a condition. In the context of mapping DNA or RNA modifications, it refers to the case whereby an authentic modification event of interest is classified as either unmodified or as a different type of modification.

False negative rate

This mathematical concept describes the probability of making a false negative call for a particular test. In the context of mapping DNA or RNA modifications, it refers to the proportion of false negative calls among all the authentic modifications of interest by a mapping method.

False positive

This mathematical concept describes when a test result incorrectly indicates the presence of a condition. In the context of mapping DNA or RNA modifications, it refers to the case whereby a base is called as modified even though it is not, or a different type of modification is called as the specific modification type of interest.

False positive rate

(FPR). This mathematical concept describes the probability of making false positive calls with a particular test. In the context of mapping DNA or RNA modifications, it refers to the proportion of false positive modification calls among unmodified bases (or modified bases of other types) by a mapping method.

Heteroplasmic

The presence of more than one type of organellar genome (mitochondrial DNA (mtDNA) or plastid DNA) within a cell or individual.

Inter-pulse duration ratio

(IPD ratio). The IPD ratio is the deviation of an observed IPD (the time length between emission pulses associated with base incorporation events) from the expected IPD associated with modification-free DNA with the same flanking sequence context. The IPD ratio reflects the presence of a chemical modification of a nucleotide or its neighbouring nucleotides.

Methylation motifs

Short sequence patterns (usually 2 ~ 10 bp) that are enriched for a certain type of DNA or RNA methylation event in an organism, and which are often determined by the recognition preference of DNA or RNA methyltransferases. For example, >95% of adenines at GATC sites are methylated (N6-methyladenine (6mA)) in γ-proteobacteria, and >80% of cytosines at CpG sites are methylated (5-methylcytosine (5mC)) in the human genome.

Modification quality value

The –log10 transformed p value. In single-molecule, real-time (SMRT) sequencing, the modification quality value describes the significance of the observed inter-pulse duration (IPD) deviation from the expected level (free of modification).

Multiple hypothesis testing

In statistics, the multiple testing problem occurs when a set of statistical inferences based on the observed values are considered simultaneously. The more inferences are made, the more likely erroneous inferences become.

Restriction-modification systems

Rudimentary immune systems found in bacteria and other prokaryotic organisms, which provide defence against foreign DNA. They include a restriction enzyme, which cuts specific unmethylated DNA sequences, and the methyltransferase, which protects the same DNA sequences.

Sensitivity

Also known as the recall or true positive rate, this mathematical concept describes the probability that a positive test is truly positive. In the context of mapping DNA or RNA modifications, it refers to the probability that true modification events are successfully detected as such by a mapping method.

Specificity

Also known as the true negative rate, this mathematical concept describes the probability that a negative test is truly negative, and is expressed as specificity = 1 – false positive rate (FPR). In the context of mapping DNA or RNA modifications, it refers to the probability that a modified event detected by a mapping method truly belongs to the modification type of interest.

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

Kong, Y., Mead, E.A. & Fang, G. Navigating the pitfalls of mapping DNA and RNA modifications. Nat Rev Genet 24, 363–381 (2023). https://doi.org/10.1038/s41576-022-00559-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-022-00559-5

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