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:

High-resolution digital profiling of the epigenome

Key Points

  • The rapid decrease in sequencing costs has led to the recent development of a plethora of epigenomic techniques that use short-read sequencing as a readout.

  • Enzymatic footprinting (using micrococcal nuclease (MNase), deoxyribonuclease (DNase), Tn5 transposase or methyltransferases) provides a means to assess the occupancy of both nucleosomes and non-histone proteins in a single experiment.

  • Enzymatic digestion of chromatin before or after chromatin immunoprecipitation (ChIP) greatly increases its resolution.

  • Mapping the last nucleotide added to a nascent RNA chain provides base-pair resolution maps of RNA polymerase occupancy.

Abstract

The widespread adoption of short-read DNA sequencing as a digital epigenomic readout platform has motivated the development of genome-wide tools that achieve base-pair resolution. New methods for footprinting and affinity purification of nucleosomes, RNA polymerases, chromatin remodellers and transcription factors have increased the resolution of epigenomic profiling by two orders of magnitude, leading to new insights into how the chromatin landscape affects gene regulation. These digital epigenomic tools have also been applied to directly profile both turnover kinetics and transcription in situ. In this Review, we describe how these new genome-wide tools allow interrogation of diverse aspects of the epigenome.

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

Figure 1: Methods for chromatin footprinting.
Figure 2: High-resolution ChIP methodologies.
Figure 3: Methods for analysing chromatin dynamics.
Figure 4: Profiling transcription.

Similar content being viewed by others

References

  1. Jiang, C. & Pugh, B. F. Nucleosome positioning and gene regulation: advances through genomics. Nature Rev. Genet. 10, 161–172 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Talbert, P. B. & Henikoff, S. Histone variants — ancient wrap artists of the epigenome. Nature Rev. Mol. Cell Biol. 11, 264–275 (2010).

    Article  CAS  Google Scholar 

  3. Zentner, G. E. & Henikoff, S. Regulation of nucleosome dynamics by histone modifications. Nature Struct. Mol. Biol. 20, 259–266 (2013).

    Article  CAS  Google Scholar 

  4. Henikoff, S. Nucleosome destabilization in the epigenetic regulation of gene expression. Nature Rev. Genet. 9, 15–26 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Smith, Z. D. & Meissner, A. DNA methylation: roles in mammalian development. Nature Rev. Genet. 14, 204–220 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Zaret, K. S. & Carroll, J. S. Pioneer transcription factors: establishing competence for gene expression. Genes Dev. 25, 2227–2241 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hargreaves, D. C. & Crabtree, G. R. ATP-dependent chromatin remodeling: genetics, genomics and mechanisms. Cell Res. 21, 396–420 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Flynn, R. A. & Chang, H. Y. Active chromatin and noncoding RNAs: an intimate relationship. Curr. Opin. Genet. Dev. 22, 172–178 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Stamatoyannopoulos, J. A. What does our genome encode? Genome Res. 22, 1602–1611 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bernstein, B. E. et al. The NIH Roadmap Epigenomics Mapping Consortium. Nature Biotech. 28, 1045–1048 (2010).

    Article  CAS  Google Scholar 

  11. Li, R. et al. De novo assembly of human genomes with massively parallel short read sequencing. Genome Res. 20, 265–272 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Barski, A. et al. High-resolution profiling of histone methylations in the human genome. Cell 129, 823–837 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Johnson, D. S., Mortazavi, A., Myers, R. M. & Wold, B. Genome-wide mapping of in vivo protein–DNA interactions. Science 316, 1497–1502 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. Brogaard, K., Xi, L., Wang, J.-P. & Widom, J. A map of nucleosome positions in yeast at base-pair resolution. Nature 486, 496–501 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Reeves, R. & Jones, A. Genomic transcriptional activity and the structure of chromatin. Nature 260, 495–500 (1976).

    Article  CAS  PubMed  Google Scholar 

  18. Weintraub, H. & Groudine, M. Chromosomal subunits in active genes have an altered conformation. Science 193, 848–856 (1976). References 17 and 18 describe the first uses of MNase and DNase, respectively, to map the accessibility of chromatin at specific genomic loci.

    Article  CAS  PubMed  Google Scholar 

  19. Sabo, P. J. et al. Genome-scale mapping of DNase I sensitivity in vivo using tiling DNA microarrays. Nature Meth. 3, 511–518 (2006).

    Article  CAS  Google Scholar 

  20. Crawford, G. E. et al. DNase-chip: a high-resolution method to identify DNase I hypersensitive sites using tiled microarrays. Nature Meth. 3, 503–509 (2006).

    Article  CAS  Google Scholar 

  21. Yuan, G.-C. et al. Genome-scale identification of nucleosome positions in S. cerevisiae. Science 309, 626–630 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Giresi, P. G., Kim, J., McDaniell, R. M., Iyer, V. R. & Lieb, J. D. FAIRE (formaldehyde-assisted isolation of regulatory elements) isolates active regulatory elements from human chromatin. Genome Res. 17, 877–885 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Auerbach, R. K. et al. Mapping accessible chromatin regions using Sono-seq. Proc. Natl Acad. Sci. USA 106, 14926–14931 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sulkowski, E. & Laskowski, M. Mechanism of action of micrococcal nuclease on deoxyribonucleic acid. J. Biol. Chem. 237, 2620–2625 (1962).

    CAS  PubMed  Google Scholar 

  25. Noll, M. Subunit structure of chromatin. Nature 251, 249–251 (1974).

    Article  CAS  PubMed  Google Scholar 

  26. Weber, C. M., Henikoff, J. G. & Henikoff, S. H2A.Z nucleosomes enriched over active genes are homotypic. Nature Struct. Mol. Biol. 17, 1500–1507 (2010).

    Article  CAS  Google Scholar 

  27. Teves, S. S. & Henikoff, S. Heat shock reduces stalled RNA polymerase II and nucleosome turnover genome-wide. Genes Dev. 25, 2387–2397 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kent, N. A., Adams, S., Moorhouse, A. & Paszkiewicz, K. Chromatin particle spectrum analysis: a method for comparative chromatin structure analysis using paired-end mode next-generation DNA sequencing. Nucleic Acids Res. 39, e26 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Henikoff, J. G., Belsky, J. A., Krassovsky, K., MacAlpine, D. M. & Henikoff, S. Epigenome characterization at single base-pair resolution. Proc. Natl Acad. Sci. USA 108, 18318–18323 (2011). References 28 and 29 show that mapping of a broad range of MNase-digested fragments gives precise information about positioning and occupancy of both nucleosomes and non-histone proteins in a single sample.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Krassovsky, K., Henikoff, J. G. & Henikoff, S. Tripartite organization of centromeric chromatin in budding yeast. Proc. Natl Acad. Sci. USA 109, 243–248 (2012).

    Article  PubMed  Google Scholar 

  31. Chung, H.-R. et al. The effect of micrococcal nuclease digestion on nucleosome positioning data. PLoS ONE 5, e15754 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Deniz, O. et al. Physical properties of naked DNA influence nucleosome positioning and correlate with transcription start and termination sites in yeast. BMC Genomics 12, 489 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Allan, J., Fraser, R. M., Owen-Hughes, T. & Keszenman-Pereyra, D. Micrococcal nuclease does not substantially bias nucleosome mapping. J. Mol. Biol. 417, 152–164 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Albert, I. et al. Translational and rotational settings of H2A.Z nucleosomes across the Saccharomyces cerevisiae genome. Nature 446, 572–576 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Hesselberth, J. R. et al. Global mapping of protein–DNA interactions in vivo by digital genomic footprinting. Nature Meth 6, 283–289 (2009).

    Article  CAS  Google Scholar 

  36. Vierstra, J., Wang, H., John, S., Sandstrom, R. & Stamatoyannopoulos, J. A. Coupling transcription factor occupancy to nucleosome architecture with DNase-FLASH. Nature Meth 11, 66–72 (2014).

    Article  CAS  Google Scholar 

  37. He, H. H. et al. Refined DNase-seq protocol and data analysis reveals intrinsic bias in transcription factor footprint identification. Nature Meth 11, 73–78 (2014).

    Article  CAS  Google Scholar 

  38. Boyle, A. P. et al. High-resolution genome-wide in vivo footprinting of diverse transcription factors in human cells. Genome Res. 21, 456–464 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Neph, S. et al. An expansive human regulatory lexicon encoded in transcription factor footprints. Nature 489, 83–90 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lazarides, E. & Lindberg, U. Actin is the naturally occurring inhibitor of deoxyribonuclease I. Proc. Natl Acad. Sci. USA 71, 4742–4746 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Grontved, L. et al. Rapid genome-scale mapping of chromatin accessibility in tissue. Epigenetics Chromatin 5, 10 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Adey, A. et al. Rapid, low-input, low-bias construction of shotgun fragment libraries by high-density in vitro transposition. Genome Biol. 11, R119 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gangadharan, S., Mularoni, L., Fain-Thornton, J., Wheelan, S. J. & Craig, N. L. DNA transposon Hermes inserts into DNA in nucleosome-free regions in vivo. Proc. Natl Acad. Sci. 107, 21966–21972 (2010). This study describes a rapid, simple procedure for epigenomic analysis based on transposition of sequencing adapters into chromatin.

    Article  PubMed  PubMed Central  Google Scholar 

  44. 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. Nature Meth. 10, 1213–1218 (2013).

    Article  CAS  Google Scholar 

  45. Henikoff, S. et al. The budding yeast Centromere DNA Element II wraps a stable Cse4 hemisome in either orientation in vivo. eLife 3, e01861 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pan, C. Q., Landgraf, R. & Sigman, D. S. Drosophila engrailed-1, 10-phenanthroline chimeras as probes of homeodomain-DNA complexes. Protein Sci. 4, 2279–2288 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pan, C. Q., Johnson, R. C. & Sigman, D. S. Identification of new fis binding sites by DNA scission with fis-1,10-phenanthroline-copper(I) chimeras. Biochemistry 35, 4326–4333 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Landgraf, R., Pan, C., Sutton, C., Pearson, L. & Sigman, D. S. Engineering of DNA binding proteins into site-specific cutters: reactivity of Trp repressor-1,10-phenanthroline chimeras. Protein Eng. 9, 603–610 (1996).

    Article  CAS  PubMed  Google Scholar 

  49. Izzo, A. et al. The genomic landscape of the somatic linker histone subtypes H1.1 to H1.5 in human cells. Cell Rep. 3, 2142–2154 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. van Bemmel, J. G. et al. A network model of the molecular organization of chromatin in Drosophila. Mol. Cell 49, 759–771 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Jessen, W. J. et al. Mapping chromatin structure in vivo using DNA methyltransferases. Methods 33, 68–80 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Kelly, T. K. et al. Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules. Genome Res. 22, 2497–2506 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gerstein, M. B. et al. Integrative analysis of the Caenorhabditis elegans genome by the modENCODE project. Science 330, 1775–1787 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. The modENCODE Consortium. Identification of functional elements and regulatory circuits by Drosophila modENCODE. Science 330, 1787–1797 (2010).

  55. Wang, J. et al. Factorbook.org: a Wiki-based database for transcription factor-binding data generated by the ENCODE consortium. Nucleic Acids Res. 41, D171–D176 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Schmidt, D. et al. ChIP–seq: using high-throughput sequencing to discover protein–DNA interactions. Methods 48, 240–248 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rhee, H. S. & Pugh, B. F. Comprehensive genome-wide protein–DNA interactions detected at single-nucleotide resolution. Cell 147, 1408–1419 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nakahashi, H. et al. A genome-wide map of CTCF multivalency redefines the CTCF code. Cell Rep. 3, 1678–1689 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yen, K., Vinayachandran, V., Batta, K., Koerber, R. T. & Pugh, B. F. Genome-wide nucleosome specificity and directionality of chromatin remodelers. Cell 149, 1461–1473 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chen, J. et al. Single-molecule dynamics of enhanceosome assembly in embryonic stem cells. Cell 156, 1274–1285 (2014). This study suggests that TFs find their binding sites through trial-and-error sampling of degenerate motifs, which provides a potential explanation for the prevalence of low-occupancy ChIP–seq peaks with weak motifs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Guo, Y., Mahony, S. & Gifford, D. K. High resolution genome wide binding event finding and motif discovery reveals transcription factor spatial binding constraints. PLoS Comput. Biol. 8, e1002638 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Serandour, A., Brown, G., Cohen, J. & Carroll, J. Development of an Illumina-based ChIP-exonuclease method provides insight into FoxA1-DNA binding properties. Genome Biol. 14, R147 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Skene, P. J., Hernandez, A. E., Groudine, M. & Henikoff, S. The nucleosomal barrier to promoter escape by RNA polymerase II is overcome by the chromatin remodeler Chd1. eLife 3, e02042 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Zentner, G. E., Tsukiyama, T. & Henikoff, S. ISWI and CHD chromatin remodelers bind promoters but act in gene bodies. PLOS Genet. 9, e1003317 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kasinathan, S., Orsi, G. A., Zentner, G. E., Ahmad, K. & Henikoff, S. High-resolution mapping of transcription factor binding sites on native chromatin. Nature Meth 11, 203–209 (2014).

    Article  CAS  Google Scholar 

  66. Zentner, G. E. & Henikoff, S. Mot1 redistributes TBP from TATA-containing to TATA-less promoters. Mol. Cell. Biol. 33, 4996–5004 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Orsi, G. A. et al. High-resolution mapping defines the cooperative architecture of Polycomb response elements. Genome Res. 24, 809–820 (2014). References 64–67 show that native ChIP is applicable to a wide range of non-histone proteins.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Voss, T. C. & Hager, G. L. Dynamic regulation of transcriptional states by chromatin and transcription factors. Nature Rev. Genet. 15, 69–81 (2014).

    Article  CAS  PubMed  Google Scholar 

  69. Ahmad, K. & Henikoff, S. The histone variant H3.3 marks active chromatin by replication-independent nucleosome assembly. Mol. Cell 9, 1191–1200 (2002).

    Article  CAS  PubMed  Google Scholar 

  70. Mito, Y., Henikoff, J. G. & Henikoff, S. Genome-scale profiling of histone H3.3 replacement patterns. Nature Genet. 37, 1090–1097 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Mito, Y., Henikoff, J. G. & Henikoff, S. Histone replacement marks the boundaries of cis-regulatory domains. Science 315, 1408–1411 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Chow, C.-M. et al. Variant histone H3.3 marks promoters of transcriptionally active genes during mammalian cell division. EMBO Rep. 6, 354–360 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Jin, C. et al. H3.3/H2A. Z double variant-containing nucleosomes mark 'nucleosome-free regions' of active promoters and other regulatory regions. Nature Genet. 41, 941–945 (2009).

    Article  CAS  PubMed  Google Scholar 

  74. Ooi, S. L., Henikoff, J. G. & Henikoff, S. A native chromatin purification system for epigenomic profiling in Caenorhabditis elegans. Nucleic Acids Res. 38, e26 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Dion, M. F. et al. Dynamics of replication-independent histone turnover in budding yeast. Science 315, 1405–1408 (2007).

    Article  CAS  PubMed  Google Scholar 

  76. Jamai, A., Imoberdorf, R. M. & Strubin, M. Continuous histone H2B and transcription-dependent histone H3 exchange in yeast cells outside of replication. Mol. Cell 25, 345–355 (2007).

    Article  CAS  PubMed  Google Scholar 

  77. van Werven, F. J., van Teeffelen, H. A. A. M., Holstege, F. C. P. & Timmers, H. T. M. Distinct promoter dynamics of the basal transcription factor TBP across the yeast genome. Nature Struct. Mol. Biol. 16, 1043–1048 (2009).

    Article  CAS  Google Scholar 

  78. Lickwar, C. R., Mueller, F., Hanlon, S. E., McNally, J. G. & Lieb, J. D. Genome-wide protein–DNA binding dynamics suggest a molecular clutch for transcription factor function. Nature 484, 251–255 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Kraushaar, D. et al. Genome-wide incorporation dynamics reveal distinct categories of turnover for the histone variant H3.3. Genome Biol. 14, R121 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Huang, C. et al. H3.3–H4 tetramer splitting events feature cell-type specific enhancers. PLoS Genet. 9, e1003558 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Deal, R. B., Henikoff, J. G. & Henikoff, S. Genome-wide kinetics of nucleosome turnover determined by metabolic labeling of histones. Science 328, 1161–1164 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Yang, F., Kemp, Christopher, J. & Henikoff, S. Doxorubicin enhances nucleosome turnover around promoters. Curr. Biol. 23, 782–787 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Gupta, P., Zlatanova, J. & Tomschik, M. Nucleosome assembly depends on the torsion in the DNA molecule: a magnetic tweezers study. Biophys. J. 97, 3150–3157 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Bermúdez, I., García-Martínez, J., Pérez-Ortín, J. E. & Roca, J. A method for genome-wide analysis of DNA helical tension by means of psoralen–DNA photobinding. Nucleic Acids Res. 38, e182 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Naughton, C. et al. Transcription forms and remodels supercoiling domains unfolding large-scale chromatin structures. Nature Struct. Mol. Biol. 20, 387–395 (2013).

    Article  CAS  Google Scholar 

  86. Kouzine, F. et al. Transcription-dependent dynamic supercoiling is a short-range genomic force. Nature Struct. Mol. Biol. 20, 396–403 (2013).

    Article  CAS  Google Scholar 

  87. Teves, S. S. & Henikoff, S. Transcription-generated torsional stress destabilizes nucleosomes. Nature Struct. Mol. Biol. 21, 88–94 (2014).

    Article  CAS  Google Scholar 

  88. Poorey, K. et al. Measuring chromatin interaction dynamics on the second time scale at single-copy genes. Science 342, 369–372 (2013). This study indicates that a single long formaldehyde crosslinking time is unsuitable for inference of the relative occupancy or dynamics of a chromatin-binding factor.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Kulaeva, O. I., Hsieh, F.-K., Chang, H.-W., Luse, D. S. & Studitsky, V. M. Mechanism of transcription through a nucleosome by RNA polymerase II. Biochim. Biophys. Acta 1829, 76–83 (2013).

    Article  CAS  PubMed  Google Scholar 

  90. Adelman, K. & Lis, J. T. Promoter-proximal pausing of RNA polymerase II: emerging roles in metazoans. Nature Rev. Genet. 13, 720–731 (2012).

    Article  CAS  PubMed  Google Scholar 

  91. Hansen, K. D., Brenner, S. E. & Dudoit, S. Biases in Illumina transcriptome sequencing caused by random hexamer priming. Nucleic Acids Res. 38, e131 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Ståhlberg, A., Håkansson, J., Xian, X., Semb, H. & Kubista, M. Properties of the reverse transcription reaction in mRNA quantification. Clin. Chem. 50, 509–515 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Churchman, L. S. & Weissman, J. S. Nascent transcript sequencing visualizes transcription at nucleotide resolution. Nature 469, 368–373 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. Gatehouse, J. & Thompson, A. in Plant Gene Transfer and Expression Protocols Vol. 49 Ch. 19, (ed. Jones, H.) 229–238 (Springer, 1995).

    Book  Google Scholar 

  95. Core, L. J., Waterfall, J. J. & Lis, J. T. Nascent RNA. Sequencing reveals widespread pausing and divergent initiation at human promoters. Science 322, 1845–1848 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Kwak, H., Fuda, N. J., Core, L. J. & Lis, J. T. Precise maps of RNA polymerase reveal how promoters direct initiation and pausing. Science 339, 950–953 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Wuarin, J. & Schibler, U. Physical isolation of nascent RNA chains transcribed by RNA polymerase II: evidence for cotranscriptional splicing. Mol. Cell. Biol. 14, 7219–7225 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Weber, Christopher, M., Ramachandran, S. & Henikoff, S. Nucleosomes are context-specific, H2A.Z-modulated barriers to RNA polymerase. Mol. Cell 53, 819–830 (2014).

    Article  CAS  PubMed  Google Scholar 

  99. Macaulay, I. C. & Voet, T. Single cell genomics: advances and future perspectives. PLoS Genet. 10, e1004126 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Tomlinson, M. J., Tomlinson, S., Yang, X. B. & Kirkham, J. Cell separation: terminology and practical considerations. J. Tissue Eng. http://dx.doi.org/10.1177/2041731412472690 (2013).

  101. Curran, S., McKay, J. A., McLeod, H. L. & Murray, G. I. Laser capture microscopy. Mol. Pathol. 53, 64–68 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Deal, R. B. & Henikoff, S. A simple method for gene expression and chromatin profiling of individual cell types within a tissue. Dev. Cell 18, 1030–1040 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Steiner, F. A., Talbert, P. B., Kasinathan, S., Deal, R. B. & Henikoff, S. Cell-type-specific nuclei purification from whole animals for genome-wide expression and chromatin profiling. Genome Res. 22, 766–777 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Henry, G. L., Davis, F. P., Picard, S. & Eddy, S. R. Cell type-specific genomics of Drosophila neurons. Nucleic Acids Res. 40, 9691–9704 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Schauer, T. et al. CAST-ChIP maps cell-type-specific chromatin states in the Drosophila central nervous system. Cell Rep. 5, 271–282 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Satterlee, J. S., Schubeler, D. & Ng, H.-H. Tackling the epigenome: challenges and opportunities for collaboration. Nature Biotech. 28, 1039–1044 (2010).

    Article  CAS  Google Scholar 

  107. Lampa, S., Dahlo, M., Olason, P., Hagberg, J. & Spjuth, O. Lessons learned from implementing a national infrastructure in Sweden for storage and analysis of next-generation sequencing data. GigaScience 2, 9 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Kodama, Y., Shumway, M. & Leinonen, R. The sequence read archive: explosive growth of sequencing data. Nucleic Acids Res. 40, D54–D56 (2012).

    Article  CAS  PubMed  Google Scholar 

  109. Zhao, R., Bodnar, M. S. & Spector, D. L. Nuclear neighborhoods and gene expression. Curr. Opin. Genet. Dev. 19, 172–179 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Unnikrishnan, A., Gafken, P. R. & Tsukiyama, T. Dynamic changes in histone acetylation regulate origins of DNA replication. Nature Struct. Mol. Biol. 17, 430–437 (2010).

    Article  CAS  Google Scholar 

  111. Kitamura, E., Blow, J. J. & Tanaka, T. U. Live-cell imaging reveals replication of individual replicons in eukaryotic replication factories. Cell 125, 1297–1308 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  113. Ay, F., Bailey, T. L. & Noble, W. S. Statistical confidence estimation for Hi-C data reveals regulatory chromatin contacts. Genome Res. 24, 999–1011 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Mendenhall, E. M. et al. Locus-specific editing of histone modifications at endogenous enhancers. Nature Biotech. 31, 1133–1136 (2013).

    Article  CAS  Google Scholar 

  115. Konermann, S. et al. Optical control of mammalian endogenous transcription and epigenetic states. Nature 500, 472–476 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Rusk, N. CRISPRs and epigenome editing. Nature Methods 11, 28 (2014).

    Article  CAS  PubMed  Google Scholar 

  117. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin-based degron system for the rapid depletion of proteins in nonplant cells. Nature Meth 6, 917–922 (2009).

    Article  CAS  Google Scholar 

  118. Haruki, H., Nishikawa, J. & Laemmli, U. K. The anchor-away technique: rapid, conditional establishment of yeast mutant phenotypes. Mol. Cell 31, 925–932 (2008).

    Article  CAS  PubMed  Google Scholar 

  119. Landt, S. G. et al. ChIP–seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Bailey, T. et al. Practical guidelines for the comprehensive analysis of ChIP–seq data. PLoS Comput. Biol. 9, e1003326 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Wade, J. T., Struhl, K., Busby, S. J. W. & Grainger, D. C. Genomic analysis of protein–DNA interactions in bacteria: insights into transcription and chromosome organization. Mol. Microbiol. 65, 21–26 (2007).

    Article  CAS  PubMed  Google Scholar 

  122. Biddie, S. C. et al. Transcription factor AP1 potentiates chromatin accessibility and glucocorticoid receptor binding. Mol. Cell 43, 145–155 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Biggin, Mark, D. Animal transcription networks as highly connected, quantitative continua. Dev. Cell 21, 611–626 (2011).

    Article  CAS  PubMed  Google Scholar 

  124. Fisher, W. W. et al. DNA regions bound at low occupancy by transcription factors do not drive patterned reporter gene expression in Drosophila. Proc. Natl Acad. Sci. USA 109, 21330–21335 (2012). This study shows that low-occupancy TF sites determined by ChIP–seq are often non-functional, which argues for cautious interpretation of such sites.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Marinov, G. K., Kundaje, A., Park, P. J. & Wold, B. J. Large-scale quality analysis of published ChIP–seq data. G3 4, 209–223 (2014). This analysis suggests that a substantial minority of published ChIP–seq data sets are of poor or intermediate quality.

  126. Teytelman, L., Thurtle, D. M., Rine, J. & van Oudenaarden, A. Highly expressed loci are vulnerable to misleading ChIP localization of multiple unrelated proteins. Proc. Natl Acad. Sci. USA 110, 18602–18607 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Park, D., Lee, Y., Bhupindersingh, G. & Iyer, V. R. Widespread misinterpretable ChIP–seq bias in yeast. PLoS ONE 8, e83506 (2013). References 123 and 124 describe biases in X-ChIP–seq experiments that could lead to artefactual results.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Fan, X. & Struhl, K. Where does Mediator bind in vivo? PLoS ONE 4, e5029 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Jeronimo, C. & Robert, F. Kin28 regulates the transient association of Mediator with core promoters. Nature Struct. Mol. Biol. 21, 449–455 (2014).

    Article  CAS  Google Scholar 

  130. Zhang, L., Zhang, K., Prändl, R. & Schöffl, F. Detecting DNA-binding of proteins in vivo by UV-crosslinking and immunoprecipitation. Biochem. Biophys. Res. Commun. 322, 705–711 (2004).

    Article  CAS  PubMed  Google Scholar 

  131. Vega, V. B., Cheung, E., Palanisamy, N. & Sung, W.-K. Inherent signals in sequencing-based chromatin-immunoprecipitation control libraries. PLoS ONE 4, e5241 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Teytelman, L. et al. Impact of chromatin structures on DNA processing for genomic analyses. PLoS ONE 4, e6700 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank S. Kasinathan and S. Ramachandran for critical reading of the manuscript and C. Weber for discussions. Work in the authors' laboratory is supported by the US National Institutes of Health grants 5U01 HG004274, U54 CA143862, and R01 ES020116 and by the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Steven Henikoff.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

FURTHER INFORMATION

CexoR

DNA sequencing costs

MACE

PowerPoint slides

Glossary

Digital epigenomic analysis

The use of methods with sequencing-based readouts to interrogate the epigenome.

Sequencing library

A collection of DNA fragments prepared for high-throughput sequencing by the addition of specific adapter sequences.

Fragment midpoint-versus-length plots

(V-plots). Representations of paired-end sequencing data in which a point corresponding to the midpoint of a paired-end read is plotted in two-dimensional space. The x coordinate of the point represents the distance of the read midpoint from a defined genomic feature, and its y coordinate represents the length of the fragment from which it was derived.

Affinity reagents

Antibodies or other molecules used to recover specific proteins from a complex mixture.

Tagmentation

Simultaneous fragmentation and incorporation of sequencing adapters into chromatin using Tn5 transposase.

Native ChIP

Chromatin immunoprecipitation (ChIP) using chromatin that has not been crosslinked with formaldehyde or any other crosslinking agent.

Nuclear run-on

A technique in which transcription is reinitated in isolated nuclei to determine the rates at which genes are transcribed.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zentner, G., Henikoff, S. High-resolution digital profiling of the epigenome. Nat Rev Genet 15, 814–827 (2014). https://doi.org/10.1038/nrg3798

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3798

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