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:

Generating specificity in genome regulation through transcription factor sensitivity to chromatin

Abstract

Cell type-specific gene expression relies on transcription factors (TFs) binding DNA sequence motifs embedded in chromatin. Understanding how motifs are accessed in chromatin is crucial to comprehend differential transcriptional responses and the phenotypic impact of sequence variation. Chromatin obstacles to TF binding range from DNA methylation to restriction of DNA access by nucleosomes depending on their position, composition and modification. In vivo and in vitro approaches now enable the study of TF binding in chromatin at unprecedented resolution. Emerging insights suggest that TFs vary in their ability to navigate chromatin states. However, it remains challenging to link binding and transcriptional outcomes to molecular characteristics of TFs or the local chromatin substrate. Here, we discuss our current understanding of how TFs access DNA in chromatin and novel techniques and directions towards a better understanding of this critical step in genome regulation.

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: Chromatin features at active regulatory elements.
Fig. 2: Nucleosome sensitivity.
Fig. 3: A scheme illustrating the use of occupancy in the genome as an indicator of chromatin sensitivity.
Fig. 4: Initiating and maintaining open chromatin.

Similar content being viewed by others

References

  1. D’Haeseleer, P. What are DNA sequence motifs? Nat. Biotechnol. 24, 423–425 (2006).

    Article  PubMed  Google Scholar 

  2. Hansen, L., Mariño-Ramírez, L. & Landsman, D. Differences in local genomic context of bound and unbound motifs. Gene 506, 125–134 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Slattery, M. et al. Absence of a simple code: how transcription factors read the genome. Trends Biochem. Sci. 39, 381–399 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wunderlich, Z. & Mirny, L. A. Different gene regulation strategies revealed by analysis of binding motifs. Trends Genet. 25, 434–440 (2009). This work comparing bacterial and eukaryotic TF motifs reveals that they use different strategies to achieve specificity in their respective genomes. Whereas bacterial motifs have sufficient information content for TFs to bind specific sites, eukaryotic motifs require clustering of motifs to achieve the same level of specificity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Adams, C. C. & Workman, J. L. Binding of disparate transcriptional activators to nucleosomal DNA is inherently cooperative. Mol. Cell. Biol. 15, 1405–1421 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Domcke, S. et al. Competition between DNA methylation and transcription factors determines binding of NRF1. Nature 528, 575–579 (2015). This work using accessibility profiling in Dnmt1;Dnmt3a;Dnmt3b triple knockout cells demonstrates that NRF1 is highly sensitive to DNA methylation in vivo.

    Article  CAS  PubMed  Google Scholar 

  7. Meyer, A. et al. Giant lungfish genome elucidates the conquest of land by vertebrates. Nature 590, 284–289 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  Google Scholar 

  9. Adams, D. et al. BLUEPRINT to decode the epigenetic signature written in blood. Nat. Biotechnol. 30, 224–226 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Moorman, C. et al. Hotspots of transcription factor colocalization in the genome of Drosophila melanogaster. Proc. Natl Acad. Sci. USA 103, 12027–12032 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Li, X. Y. et al. The role of chromatin accessibility in directing the widespread, overlapping patterns of Drosophila transcription factor binding. Genome Biol. 12, R34 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Stadler, M. B. et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480, 490–495 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Zhou, V. W., Goren, A. & Bernstein, B. E. Charting histone modifications and the functional organization of mammalian genomes. Nat. Rev. Genet. 12, 7–18 (2011).

    Article  PubMed  Google Scholar 

  14. Narlikar, G. J., Sundaramoorthy, R. & Owen-Hughes, T. Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes. Cell 154, 490–503 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.21–21.29.29 (2015).

    Article  Google Scholar 

  16. Song, L. & Crawford, G. E. DNase-seq: a high-resolution technique for mapping active gene regulatory elements across the genome from mammalian cells. Cold Spring Harb. Protoc. https://doi.org/10.1101/pdb.prot5384 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Bird, A., Taggart, M., Frommer, M., Miller, O. J. & Macleod, D. A fraction of the mouse genome that is derived from islands of nonmethylated, CpG-rich DNA. Cell 40, 91–99 (1985).

    Article  CAS  PubMed  Google Scholar 

  18. Schmitz, R. J., Lewis, Z. A. & Goll, M. G. DNA methylation: shared and divergent features across eukaryotes. Trends Genet. 35, 818–827 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Schübeler, D. Epigenetic islands in a genetic ocean. Science 338, 756–757 (2012).

    Article  PubMed  Google Scholar 

  20. Okano, M., Xie, S. & Li, E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat. Genet. 19, 219–220 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Bestor, T., Laudano, A., Mattaliano, R. & Ingram, V. Cloning and sequencing of a cDNA encoding DNA methyltransferase of mouse cells: the carboxyl-terminal domain of the mammalian enzymes is related to bacterial restriction methyltransferases. J. Mol. Biol. 203, 971–983 (1988).

    Article  CAS  PubMed  Google Scholar 

  22. Leonhardt, H., Page, A. W., Weier, H. U. & Bestor, T. H. A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell 71, 865–873 (1992).

    Article  CAS  PubMed  Google Scholar 

  23. Ooi, S. K. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Baubec, T. et al. Genomic profiling of DNA methyltransferases reveals a role for DNMT3B in genic methylation. Nature 520, 243–247 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Taylor, S. M. & Jones, P. A. Multiple new phenotypes induced in 10T1/2 and 3T3 cells treated with 5-azacytidine. Cell 17, 771–779 (1979).

    Article  CAS  PubMed  Google Scholar 

  26. Chong, S. et al. Modifiers of epigenetic reprogramming show paternal effects in the mouse. Nat. Genet. 39, 614–622 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, E., Beard, C. & Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature 366, 362–365 (1993).

    Article  CAS  PubMed  Google Scholar 

  28. Walsh, C. P., Chaillet, J. R. & Bestor, T. H. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat. Genet. 20, 116–117 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Panning, B. & Jaenisch, R. DNA hypomethylation can activate Xist expression and silence X-linked genes. Genes Dev. 10, 1991–2002 (1996).

    Article  CAS  PubMed  Google Scholar 

  30. Rao, S. et al. Systematic prediction of DNA shape changes due to CpG methylation explains epigenetic effects on protein–DNA binding. Epigenet. Chromatin 11, 6 (2018).

    Article  Google Scholar 

  31. Lewis, J. D. et al. Purification, sequence, and cellular localization of a novel chromosomal protein that binds to methylated DNA. Cell 69, 905–914 (1992).

    Article  CAS  PubMed  Google Scholar 

  32. Hendrich, B. & Bird, A. Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol. Cell. Biol. 18, 6538–6547 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Baubec, T., Ivánek, R., Lienert, F. & Schübeler, D. Methylation-dependent and -independent genomic targeting principles of the MBD protein family. Cell 153, 480–492 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Prokhortchouk, A. et al. The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev. 15, 1613–1618 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Quenneville, S. et al. In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol. Cell 44, 361–372 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Arita, K., Ariyoshi, M., Tochio, H., Nakamura, Y. & Shirakawa, M. Recognition of hemi-methylated DNA by the SRA protein UHRF1 by a base-flipping mechanism. Nature 455, 818–821 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Avvakumov, G. V. et al. Structural basis for recognition of hemi-methylated DNA by the SRA domain of human UHRF1. Nature 455, 822–825 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Hashimoto, H. et al. The SRA domain of UHRF1 flips 5-methylcytosine out of the DNA helix. Nature 455, 826–829 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nan, X. et al. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393, 386–389 (1998).

    Article  CAS  PubMed  Google Scholar 

  40. Ng, H. H. et al. MBD2 is a transcriptional repressor belonging to the MeCP1 histone deacetylase complex. Nat. Genet. 23, 58–61 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Ngo, T. T. M. et al. Effects of cytosine modifications on DNA flexibility and nucleosome mechanical stability. Nat. Commun. 7, 10813 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Choy, J. S. et al. DNA methylation increases nucleosome compaction and rigidity. J. Am. Chem. Soc. 132, 1782–1783 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Jimenez-Useche, I. & Yuan, C. The effect of DNA CpG methylation on the dynamic conformation of a nucleosome. Biophys. J. 103, 2502–2512 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lambert, S. A. et al. The human transcription factors. Cell 172, 650–665 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Mann, I. K. et al. CG methylated microarrays identify a novel methylated sequence bound by the CEBPB|ATF4 heterodimer that is active in vivo. Genome Res. 23, 988–997 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yin, Y. et al. Impact of cytosine methylation on DNA binding specificities of human transcription factors. Science 356, eaaj2239 (2017). This work using high-throughput SELEX demonstrates widespread methylation-sensitive binding of mammalian TFs.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Kribelbauer, J. F. et al. Quantitative analysis of the DNA methylation sensitivity of transcription factor complexes. Cell Rep. 19, 2383–2395 (2017). This comprehensive in vitro study shows that methylation sensitivity of a TF can be a function of binding motif sequence variants.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zuo, Z., Roy, B., Chang, Y. K., Granas, D. & Stormo, G. D. Measuring quantitative effects of methylation on transcription factor–DNA binding affinity. Sci. Adv. 3, eaao1799 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. O’Malley, R. C. et al. Cistrome and epicistrome features shape the regulatory DNA landscape. Cell 165, 1280–1292 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Schübeler, D. Function and information content of DNA methylation. Nature 517, 321–326 (2015).

    Article  PubMed  Google Scholar 

  51. Tsumura, A. et al. Maintenance of self-renewal ability of mouse embryonic stem cells in the absence of DNA methyltransferases Dnmt1, Dnmt3a and Dnmt3b. Genes Cells 11, 805–814 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Cusack, M. et al. Distinct contributions of DNA methylation and histone acetylation to the genomic occupancy of transcription factors. Genome Res. 30, 1393–1406 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Grand, R. S. et al. BANP opens chromatin and activates CpG-island-regulated genes. Nature 596, 133–137 (2021).

    Article  CAS  PubMed  Google Scholar 

  54. Zhang, J. et al. Highly enriched BEND3 prevents the premature activation of bivalent genes during differentiation. Science 375, 1053–1058 (2022).

    Article  CAS  PubMed  Google Scholar 

  55. Stielow, B. et al. The SAM domain-containing protein 1 (SAMD1) acts as a repressive chromatin regulator at unmethylated CpG islands. Sci. Adv. 7, eabf2229 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pappalardi, M. B. et al. Discovery of a first-in-class reversible DNMT1-selective inhibitor with improved tolerability and efficacy in acute myeloid leukemia. Nat. Cancer 2, 1002–1017 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Luger, K., Mäder, A. W., Richmond, R. K., Sargent, D. F. & Richmond, T. J. Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260 (1997).

    Article  CAS  PubMed  Google Scholar 

  58. Richmond, T. J. & Davey, C. A. The structure of DNA in the nucleosome core. Nature 423, 145–150 (2003).

    Article  CAS  PubMed  Google Scholar 

  59. Matsumoto, S. et al. DNA damage detection in nucleosomes involves DNA register shifting. Nature 571, 79–84 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Michael, A. K. & Thomä, N. H. Reading the chromatinized genome. Cell 184, 3599–3611 (2021).

    Article  CAS  PubMed  Google Scholar 

  61. Segal, E. & Widom, J. From DNA sequence to transcriptional behaviour: a quantitative approach. Nat. Rev. Genet. 10, 443–456 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Radman-Livaja, M. & Rando, O. J. Nucleosome positioning: how is it established, and why does it matter? Dev. Biol. 339, 258–266 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Schones, D. E. et al. Dynamic regulation of nucleosome positioning in the human genome. Cell 132, 887–898 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Li, Q. & Wrange, O. Translational positioning of a nucleosomal glucocorticoid response element modulates glucocorticoid receptor affinity. Genes Dev. 7, 2471–2482 (1993).

    Article  CAS  PubMed  Google Scholar 

  65. Soufi, A. et al. Pioneer transcription factors target partial DNA motifs on nucleosomes to initiate reprogramming. Cell 161, 555–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. McDaniel, S. L. et al. Continued activity of the pioneer factor zelda is required to drive zygotic genome activation. Mol. Cell 74, 185–195.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Hsu, H. T. et al. Recruitment of RNA polymerase II by the pioneer transcription factor PHA-4. Science 348, 1372–1376 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Cirillo, L. A. et al. Binding of the winged-helix transcription factor HNF3 to a linker histone site on the nucleosome. EMBO J. 17, 244–254 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Perlmann, T. & Wrange, O. Specific glucocorticoid receptor binding to DNA reconstituted in a nucleosome. EMBO J. 7, 3073–3079 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Clark, K. L., Halay, E. D., Lai, E. & Burley, S. K. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 364, 412–420 (1993).

    Article  CAS  PubMed  Google Scholar 

  71. Li, G. & Widom, J. Nucleosomes facilitate their own invasion. Nat. Struct. Mol. Biol. 11, 763–769 (2004). This work using measurements of conformational transitions of nucleosomes in vitro demonstrates an inherent property involving the transient partial unwrapping of the nucleosomal DNA, suggesting a means by which many factors might gain access.

    Article  CAS  PubMed  Google Scholar 

  72. Li, G., Levitus, M., Bustamante, C. & Widom, J. Rapid spontaneous accessibility of nucleosomal DNA. Nat. Struct. Mol. Biol. 12, 46–53 (2005).

    Article  CAS  PubMed  Google Scholar 

  73. Zhu, F. et al. The interaction landscape between transcription factors and the nucleosome. Nature 562, 76–81 (2018). This work using high-throughput SELEX demonstrates comprehensive engagement of nucleosomes by mammalian TFs via multiple binding modes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Morgunova, E. & Taipale, J. Structural insights into the interaction between transcription factors and the nucleosome. Curr. Opin. Struct. Biol. 71, 171–179 (2021).

    Article  CAS  PubMed  Google Scholar 

  75. Fernandez Garcia, M. et al. Structural features of transcription factors associating with nucleosome binding. Mol. Cell 75, 921–932.e6 (2019). This work presents a protein microarray screen for TFs that are able to engage with nucleosomal motifs.

    Article  CAS  PubMed  Google Scholar 

  76. Dodonova, S. O., Zhu, F., Dienemann, C., Taipale, J. & Cramer, P. Nucleosome-bound SOX2 and SOX11 structures elucidate pioneer factor function. Nature 580, 669–672 (2020). This work presents high-resolution structures of SOX2 and SOX11 TFs engaged at an internal SHL2 position on the nucleosome.

    Article  CAS  PubMed  Google Scholar 

  77. Michael, A. K. et al. Mechanisms of OCT4–SOX2 motif readout on nucleosomes. Science 368, 1460–1465 (2020). This work using high-throughput binding assays and structural determination demonstrates that OCT4–SOX2 can engage partial motifs at the entry–exit sites of a nucleosome.

    Article  CAS  PubMed  Google Scholar 

  78. Lowary, P. T. & Widom, J. New DNA sequence rules for high affinity binding to histone octamer and sequence-directed nucleosome positioning. J. Mol. Biol. 276, 19–42 (1998).

    Article  CAS  PubMed  Google Scholar 

  79. Tanaka, H. et al. Interaction of the pioneer transcription factor GATA3 with nucleosomes. Nat. Commun. 11, 4136 (2020). This work on the structure of GATA3 on the nucleosome demonstrates tandem motif engagement at high resolution.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. North, J. A. et al. Regulation of the nucleosome unwrapping rate controls DNA accessibility. Nucleic Acids Res. 40, 10215–10227 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Gaffney, D. J. et al. Controls of nucleosome positioning in the human genome. PLoS Genet. 8, e1003036 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Krietenstein, N. et al. Genomic nucleosome organization reconstituted with pure proteins. Cell 167, 709–721.e12 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Lancrey, A. et al. Nucleosome positioning on large tandem DNA repeats of the ‘601’ sequence engineered in Saccharomyces cerevisiae. J. Mol. Biol. 434, 167497 (2022).

    Article  CAS  PubMed  Google Scholar 

  84. Hertel, C. B., Längst, G., Hörz, W. & Korber, P. Nucleosome stability at the yeast PHO5 and PHO8 promoters correlates with differential cofactor requirements for chromatin opening. Mol. Cell. Biol. 25, 10755–10767 (2005). This study shows that nuclear factors can locally tune nucleosome positioning at yeast promoters.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Almer, A., Rudolph, H., Hinnen, A. & Hörz, W. Removal of positioned nucleosomes from the yeast PHO5 promoter upon PHO5 induction releases additional upstream activating DNA elements. EMBO J. 5, 2689–2696 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Lam, F. H., Steger, D. J. & O’Shea, E. K. Chromatin decouples promoter threshold from dynamic range. Nature 453, 246–250 (2008). This study dissects the interplay between affinity and proximity of TF binding sites relative to positioned nucleosomes and suggests a role for chromatin to modulate the transcriptional response to external signalling.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Takahata, S., Yu, Y. & Stillman, D. J. FACT and Asf1 regulate nucleosome dynamics and coactivator binding at the HO promoter. Mol. Cell 34, 405–415 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yarrington, R. M., Rudd, J. S. & Stillman, D. J. Spatiotemporal cascade of transcription factor binding required for promoter activation. Mol. Cell. Biol. 35, 688–698 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Johnson, T. A. et al. Conventional and pioneer modes of glucocorticoid receptor interaction with enhancer chromatin in vivo. Nucleic Acids Res. 46, 203–214 (2018).

    Article  CAS  PubMed  Google Scholar 

  90. Iwafuchi, M. et al. Gene network transitions in embryos depend upon interactions between a pioneer transcription factor and core histones. Nat. Genet. 52, 418–427 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Nishimura, M., Arimura, Y., Nozawa, K. & Kurumizaka, H. Linker DNA and histone contributions in nucleosome binding by p53. J. Biochem. 168, 669–675 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Musselman, C. A., Lalonde, M. E., Côté, J. & Kutateladze, T. G. Perceiving the epigenetic landscape through histone readers. Nat. Struct. Mol. Biol. 19, 1218–1227 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Andrews, F. H., Strahl, B. D. & Kutateladze, T. G. Insights into newly discovered marks and readers of epigenetic information. Nat. Chem. Biol. 12, 662–668 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Shimko, J. C., North, J. A., Bruns, A. N., Poirier, M. G. & Ottesen, J. J. Preparation of fully synthetic histone H3 reveals that acetyl-lysine 56 facilitates protein binding within nucleosomes. J. Mol. Biol. 408, 187–204 (2011).

    Article  CAS  PubMed  Google Scholar 

  95. Simon, M. et al. Histone fold modifications control nucleosome unwrapping and disassembly. Proc. Natl Acad. Sci. USA 108, 12711–12716 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Shoaib, M. et al. Histone H4 lysine 20 mono-methylation directly facilitates chromatin openness and promotes transcription of housekeeping genes. Nat. Commun. 12, 4800 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Klemm, S. L., Shipony, Z. & Greenleaf, W. J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20, 207–220 (2019).

    Article  CAS  PubMed  Google Scholar 

  98. Cirillo, L. A. et al. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289 (2002).

    Article  CAS  PubMed  Google Scholar 

  99. Zaret, K. S. Pioneer transcription factors initiating gene network changes. Annu. Rev. Genet. 54, 367–385 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Zaret, K. S. & Carroll, J. S. Pioneer transcription factors: establishing competence for gene expression. Genes Dev. 25, 2227–2241 (2011). This work presents a model of pioneer factor activity in the genome, whereby specialized TFs are the first in a hierarchy of factors that enable the activation of regulatory regions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Swinstead, E. E. et al. Steroid receptors reprogram FoxA1 occupancy through dynamic chromatin transitions. Cell 165, 593–605 (2016). This study demonstrates that chromatin-insensitive TFs may open chromatin independently at some sites, whereas at others they must cooperate with additional TFs to mutually occupy regulatory regions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Zaret, K. S., Lerner, J. & Iwafuchi-Doi, M. Chromatin scanning by dynamic binding of pioneer factors. Mol. Cell 62, 665–667 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Glont, S. E., Chernukhin, I. & Carroll, J. S. Comprehensive genomic analysis reveals that the pioneering function of FOXA1 is independent of hormonal signaling. Cell Rep. 26, 2558–2565.e3 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Paakinaho, V., Swinstead, E. E., Presman, D. M., Grøntved, L. & Hager, G. L. Meta-analysis of chromatin programming by steroid receptors. Cell Rep. 28, 3523–3534.e2 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Hansen, J. L., Loell, K. J. & Cohen, B. A. The pioneer factor hypothesis is not necessary to explain ectopic liver gene activation. eLife 11, e73358 (2022). This study shows that high expression of a TF that cannot bind to nucleosomal DNA nevertheless accesses closed chromatin, similar to a TF that can bind nucleosomal DNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Minderjahn, J. et al. Mechanisms governing the pioneering and redistribution capabilities of the non-classical pioneer PU.1. Nat. Commun. 11, 402 (2020). This study reveals that the PU.1 TF utilizes BRG1 to enable genomic binding and opening of chromatin in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. 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 

  108. Plasschaert, R. N. et al. CTCF binding site sequence differences are associated with unique regulatory and functional trends during embryonic stem cell differentiation. Nucleic Acids Res. 42, 774–789 (2014).

    Article  CAS  PubMed  Google Scholar 

  109. Xu, B. et al. Acute depletion of CTCF rewires genome-wide chromatin accessibility. Genome Biol. 22, 244 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Sönmezer, C. et al. Molecular co-occupancy identifies transcription factor binding cooperativity in vivo. Mol. Cell 81, 255–267.e6 (2021).

    Article  PubMed  Google Scholar 

  111. Fu, Y., Sinha, M., Peterson, C. L. & Weng, Z. The insulator binding protein CTCF positions 20 nucleosomes around its binding sites across the human genome. PLoS Genet. 4, e1000138 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  112. Sherwood, R. I. et al. Discovery of directional and nondirectional pioneer transcription factors by modeling DNase profile magnitude and shape. Nat. Biotechnol. 32, 171–178 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Yan, C., Chen, H. & Bai, L. Systematic study of nucleosome-displacing factors in budding yeast. Mol. Cell 71, 294–305.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Kribelbauer, J. F. et al. Context-dependent gene regulation by homeodomain transcription factor complexes revealed by shape-readout deficient proteins. Mol. Cell 78, 152–167.e11 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Kribelbauer, J. F., Rastogi, C., Bussemaker, H. J. & Mann, R. S. Low-affinity binding sites and the transcription factor specificity paradox in eukaryotes. Annu. Rev. Cell Dev. Biol. 35, 357–379 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Miller, J. A. & Widom, J. Collaborative competition mechanism for gene activation in vivo. Mol. Cell. Biol. 23, 1623–1632 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Slattery, M. et al. Cofactor binding evokes latent differences in DNA binding specificity between Hox proteins. Cell 147, 1270–1282 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Srivastava, D. & Mahony, S. Sequence and chromatin determinants of transcription factor binding and the establishment of cell type-specific binding patterns. Biochim. Biophys. Acta Gene Regul. Mech. 1863, 194443 (2020).

    Article  CAS  PubMed  Google Scholar 

  120. Fan, Y. et al. H1 linker histones are essential for mouse development and affect nucleosome spacing in vivo. Mol. Cell. Biol. 23, 4559–4572 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Healton, S. E. et al. H1 linker histones silence repetitive elements by promoting both histone H3K9 methylation and chromatin compaction. Proc. Natl Acad. Sci. USA 117, 14251–14258 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Blackledge, N. P., Rose, N. R. & Klose, R. J. Targeting Polycomb systems to regulate gene expression: modifications to a complex story. Nat. Rev. Mol. Cell Biol. 16, 643–649 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Pengelly, A. R., Copur, Ö., Jäckle, H., Herzig, A. & Müller, J. A histone mutant reproduces the phenotype caused by loss of histone-modifying factor Polycomb. Science 339, 698–699 (2013).

    Article  CAS  PubMed  Google Scholar 

  124. Minoux, M. et al. Gene bivalency at Polycomb domains regulates cranial neural crest positional identity. Science 355, eaal2913 (2017).

    Article  PubMed  Google Scholar 

  125. Nicetto, D. et al. H3K9me3-heterochromatin loss at protein-coding genes enables developmental lineage specification. Science 363, 294–297 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  127. Clapier, C. R. & Cairns, B. R. The biology of chromatin remodeling complexes. Annu. Rev. Biochem. 78, 273–304 (2009).

    Article  CAS  PubMed  Google Scholar 

  128. Marmorstein, R. & Trievel, R. C. Histone modifying enzymes: structures, mechanisms, and specificities. Biochim. Biophys. Acta 1789, 58–68 (2009).

    Article  CAS  PubMed  Google Scholar 

  129. Morgan, M. A. J. & Shilatifard, A. Reevaluating the roles of histone-modifying enzymes and their associated chromatin modifications in transcriptional regulation. Nat. Genet. 52, 1271–1281 (2020).

    Article  CAS  PubMed  Google Scholar 

  130. Côté, J., Quinn, J., Workman, J. L. & Peterson, C. L. Stimulation of GAL4 derivative binding to nucleosomal DNA by the yeast SWI/SNF complex. Science 265, 53–60 (1994).

    Article  PubMed  Google Scholar 

  131. Mivelaz, M. et al. Chromatin fiber invasion and nucleosome displacement by the Rap1 transcription factor. Mol. Cell 77, 488–500.e9 (2020). This work presents in vitro experiments demonstrating that the Rap1 TF engages nucleosomal motifs and inhibits inter-nucleosome contacts, but that it needs a remodeller to displace nucleosomes and achieve stable binding on DNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Kubik, S. et al. Sequence-directed action of RSC remodeler and general regulatory factors modulates +1 nucleosome position to facilitate transcription. Mol. Cell 71, 89–102.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  133. Fyodorov, D. V. & Kadonaga, J. T. Dynamics of ATP-dependent chromatin assembly by ACF. Nature 418, 897–900 (2002).

    Article  CAS  PubMed  Google Scholar 

  134. Lusser, A., Urwin, D. L. & Kadonaga, J. T. Distinct activities of CHD1 and ACF in ATP-dependent chromatin assembly. Nat. Struct. Mol. Biol. 12, 160–166 (2005).

    Article  CAS  PubMed  Google Scholar 

  135. Imbalzano, A. N., Schnitzler, G. R. & Kingston, R. E. Nucleosome disruption by human SWI/SNF is maintained in the absence of continued ATP hydrolysis. J. Biol. Chem. 271, 20726–20733 (1996).

    Article  CAS  PubMed  Google Scholar 

  136. Aoyagi, S. et al. Nucleosome remodeling by the human SWI/SNF complex requires transient global disruption of histone–DNA interactions. Mol. Cell. Biol. 22, 3653–3662 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. McKnight, J. N., Jenkins, K. R., Nodelman, I. M., Escobar, T. & Bowman, G. D. Extranucleosomal DNA binding directs nucleosome sliding by Chd1. Mol. Cell. Biol. 31, 4746–4759 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Li, M. et al. Dynamic regulation of transcription factors by nucleosome remodeling. eLife 4, e06249 (2015).

    Article  PubMed Central  Google Scholar 

  139. Wiechens, N. et al. The chromatin remodelling enzymes SNF2H and SNF2L position nucleosomes adjacent to CTCF and other transcription factors. PLoS Genet. 12, e1005940 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  140. King, H. W. & Klose, R. J. The pioneer factor OCT4 requires the chromatin remodeller BRG1 to support gene regulatory element function in mouse embryonic stem cells. eLife 6, e22631 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  141. Barisic, D., Stadler, M. B., Iurlaro, M. & Schübeler, D. Mammalian ISWI and SWI/SNF selectively mediate binding of distinct transcription factors. Nature 569, 136–140 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Teufel, D. P., Freund, S. M., Bycroft, M. & Fersht, A. R. Four domains of p300 each bind tightly to a sequence spanning both transactivation subdomains of p53. Proc. Natl Acad. Sci. USA 104, 7009–7014 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. He, J. et al. Structure of p300 bound to MEF2 on DNA reveals a mechanism of enhanceosome assembly. Nucleic Acids Res. 39, 4464–4474 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Dann, G. P. et al. ISWI chromatin remodellers sense nucleosome modifications to determine substrate preference. Nature 548, 607–611 (2017). This study shows that ISWI complexes utilize accessory subunits to interpret histone modifications in vitro, which affects remodeller activity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Mashtalir, N. et al. Chromatin landscape signals differentially dictate the activities of mSWI/SNF family complexes. Science 373, 306–315 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Kadoch, C. & Crabtree, G. R. Mammalian SWI/SNF chromatin remodeling complexes and cancer: mechanistic insights gained from human genomics. Sci. Adv. 1, e1500447 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Iurlaro, M. et al. Mammalian SWI/SNF continuously restores local accessibility to chromatin. Nat. Genet. 53, 279–287 (2021). This work targeting the SWI/SNF remodeller with rapid chemical inhibition demonstrates the continuous requirement for remodellers to maintain open chromatin in mouse cells.

    Article  CAS  PubMed  Google Scholar 

  148. Schick, S. et al. Acute BAF perturbation causes immediate changes in chromatin accessibility. Nat. Genet. 53, 269–278 (2021). This work on acute degradation of different subunits of the SWI/SNF remodeller complex and chemical inhibition demonstrates the continuous requirement for remodellers to maintain open chromatin in human cells.

    Article  CAS  PubMed  Google Scholar 

  149. Kubik, S. et al. Opposing chromatin remodelers control transcription initiation frequency and start site selection. Nat. Struct. Mol. Biol. 26, 744–754 (2019).

    Article  CAS  PubMed  Google Scholar 

  150. Kim, J. M. et al. Single-molecule imaging of chromatin remodelers reveals role of ATPase in promoting fast kinetics of target search and dissociation from chromatin. eLife 10, e69387 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Paakinaho, V. et al. Single-molecule analysis of steroid receptor and cofactor action in living cells. Nat. Commun. 8, 15896 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Hota, S. K. et al. Brahma safeguards canalization of cardiac mesoderm differentiation. Nature 602, 129–134 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Friman, E. T. et al. Dynamic regulation of chromatin accessibility by pluripotency transcription factors across the cell cycle. eLife 8, e50087 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  154. Ginno, P. A. et al. A genome-scale map of DNA methylation turnover identifies site-specific dependencies of DNMT and TET activity. Nat. Commun. 11, 2680 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Charlton, J. et al. TETs compete with DNMT3 activity in pluripotent cells at thousands of methylated somatic enhancers. Nat. Genet. 52, 819–827 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Voss, T. C. et al. Dynamic exchange at regulatory elements during chromatin remodeling underlies assisted loading mechanism. Cell 146, 544–554 (2011). This study shows that the binding of one TF facilitates another even though they compete for the same motif, suggesting a model of cooperativity termed assisted loading whereby TFs amplify each other’s binding via increased chromatin accessibility.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Madsen, M. S., Siersbæk, R., Boergesen, M., Nielsen, R. & Mandrup, S. Peroxisome proliferator-activated receptor γ and C/EBPα synergistically activate key metabolic adipocyte genes by assisted loading. Mol. Cell. Biol. 34, 939–954 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  158. Siersbæk, R. et al. IL6/STAT3 signaling hijacks estrogen receptor α enhancers to drive breast cancer metastasis. Cancer Cell 38, e419 (2020).

    Article  Google Scholar 

  159. Spitz, F. & Furlong, E. E. Transcription factors: from enhancer binding to developmental control. Nat. Rev. Genet. 13, 613–626 (2012).

    Article  CAS  PubMed  Google Scholar 

  160. Berman, B. P. et al. Exploiting transcription factor binding site clustering to identify cis-regulatory modules involved in pattern formation in the Drosophila genome. Proc. Natl Acad. Sci. USA 99, 757–762 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Chen, J. et al. Single-molecule dynamics of enhanceosome assembly in embryonic stem cells. Cell 156, 1274–1285 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Izeddin, I. et al. Single-molecule tracking in live cells reveals distinct target-search strategies of transcription factors in the nucleus. eLife 3, e02230 (2014).

    Article  PubMed Central  Google Scholar 

  163. Morisaki, T., Müller, W. G., Golob, N., Mazza, D. & McNally, J. G. Single-molecule analysis of transcription factor binding at transcription sites in live cells. Nat. Commun. 5, 4456 (2014).

    Article  CAS  PubMed  Google Scholar 

  164. Hu, Z., Killion, P. J. & Iyer, V. R. Genetic reconstruction of a functional transcriptional regulatory network. Nat. Genet. 39, 683–687 (2007).

    Article  CAS  PubMed  Google Scholar 

  165. Hay, D. et al. Genetic dissection of the α-globin super-enhancer in vivo. Nat. Genet. 48, 895–903 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Klein, J. C. et al. A systematic evaluation of the design and context dependencies of massively parallel reporter assays. Nat. Methods 17, 1083–1091 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Jaeger, M. G. & Winter, G. E. Fast-acting chemical tools to delineate causality in transcriptional control. Mol. Cell 81, 1617–1630 (2021).

    Article  CAS  PubMed  Google Scholar 

  169. Weingarten-Gabbay, S. et al. Systematic interrogation of human promoters. Genome Res. 29, 171–183 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Grossman, S. R. et al. Systematic dissection of genomic features determining transcription factor binding and enhancer function. Proc. Natl Acad. Sci. USA 114, E1291–E1300 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. King, D. M. et al. Synthetic and genomic regulatory elements reveal aspects of cis-regulatory grammar in mouse embryonic stem cells. eLife 9, e41279 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Smith, R. P. et al. Massively parallel decoding of mammalian regulatory sequences supports a flexible organizational model. Nat. Genet. 45, 1021–1028 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Hammelman, J., Krismer, K., Banerjee, B., Gifford, D. K. & Sherwood, R. I. Identification of determinants of differential chromatin accessibility through a massively parallel genome-integrated reporter assay. Genome Res. 30, 1468–1480 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Haberle, V. et al. Transcriptional cofactors display specificity for distinct types of core promoters. Nature 570, 122–126 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Alerasool, N., Leng, H., Lin, Z. Y., Gingras, A. C. & Taipale, M. Identification and functional characterization of transcriptional activators in human cells. Mol. Cell 82, 677–695.e7 (2022).

    Article  CAS  PubMed  Google Scholar 

  176. Arnold, C. D. et al. Genome-wide quantitative enhancer activity maps identified by STARR-seq. Science 339, 1074–1077 (2013).

    Article  CAS  PubMed  Google Scholar 

  177. Avsec, Ž. et al. Effective gene expression prediction from sequence by integrating long-range interactions. Nat. Methods 18, 1196–1203 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Fan, L. Z. & Lin, M. Z. Optical control of biological processes by light-switchable proteins. Wiley Interdiscip. Rev. Dev. Biol. 4, 545–554 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Lin, C. Y. et al. Transcriptional amplification in tumor cells with elevated c-Myc. Cell 151, 56–67 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Stengel, K. R., Ellis, J. D., Spielman, C. L., Bomber, M. L. & Hiebert, S. W. Definition of a small core transcriptional circuit regulated by AML1-ETO. Mol. Cell 81, 530–545.e5 (2021).

    Article  CAS  PubMed  Google Scholar 

  182. Liscovitch-Brauer, N. et al. Profiling the genetic determinants of chromatin accessibility with scalable single-cell CRISPR screens. Nat. Biotechnol. 39, 1270–1277 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Pierce, S. E., Granja, J. M. & Greenleaf, W. J. High-throughput single-cell chromatin accessibility CRISPR screens enable unbiased identification of regulatory networks in cancer. Nat. Commun. 12, 2969–2969 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Beliveau, B. J. et al. Single-molecule super-resolution imaging of chromosomes and in situ haplotype visualization using Oligopaint FISH probes. Nat. Commun. 6, 7147 (2015).

    Article  CAS  PubMed  Google Scholar 

  185. Bintu, B. et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  186. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. 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). This work shows that FiberSeq generates single-molecule data on TFs and nucleosomes that span kilobases using a footprinting and long-read sequencing technique.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  189. Spruijt, C. G. et al. Dynamic readers for 5-(hydroxy)methylcytosine and its oxidized derivatives. Cell 152, 1146–1159 (2013).

    Article  CAS  PubMed  Google Scholar 

  190. Sequeira, V. M. & Vermeulen, M. Identifying readers for (hydroxy)methylated DNA using quantitative interaction proteomics: advances and challenges ahead. J. Mol. Biol. 432, 1792–1800 (2019).

    Article  Google Scholar 

  191. Brinkman, A. B. et al. Sequential ChIP-bisulfite sequencing enables direct genome-scale investigation of chromatin and DNA methylation cross-talk. Genome Res. 22, 1128–1138 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Statham, A. L. et al. Bisulfite sequencing of chromatin immunoprecipitated DNA (BisChIP-seq) directly informs methylation status of histone-modified DNA. Genome Res. 22, 1120–1127 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Lhoumaud, P. et al. EpiMethylTag: simultaneous detection of ATAC-seq or ChIP-seq signals with DNA methylation. Genome Biol. 20, 248 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Meers, M. P., Janssens, D. H. & Henikoff, S. Pioneer factor-nucleosome binding events during differentiation are motif encoded. Mol. Cell 75, 562–575.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Gutin, J. et al. Fine-resolution mapping of TF binding and chromatin interactions. Cell Rep. 22, 2797–2807 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. 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 

  197. Pardo, C. E., Darst, R. P., Nabilsi, N. H., Delmas, A. L. & Kladde, M. P. Simultaneous single-molecule mapping of protein–DNA interactions and DNA methylation by MAPit. Curr. Protoc. Mol. Biol. 95, 21.22.1–21.22.18 (2011).

    Article  Google Scholar 

  198. Levo, M. et al. Systematic investigation of transcription factor activity in the context of chromatin using massively parallel binding and expression assays. Mol. Cell 65, 604–617.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  199. Krebs, A. R., Dessus-Babus, S., Burger, L. & Schübeler, D. High-throughput engineering of a mammalian genome reveals building principles of methylation states at CG rich regions. eLife 3, e04094 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

L.I. acknowledges the Australian National Health and Medical Research Council CJ Martin Fellowship APP1148380. R.S.G. acknowledges European Molecular Biology Organization (EMBO) Long-Term Fellowships. R.S.G. and L.I. acknowledge the European Union (EU) Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant (705354 to R.S.G. and 748760 to L.I.). D.S. acknowledges support from the Novartis Research Foundation, the Swiss National Science Foundation (310030B_176394) and the European Research Council under the EU Horizon 2020 research and innovation programme grant agreements (ReadMe-667951 and DNAaccess-884664). The authors thank N. Thomä, A. Michael, B. Cohen and members of the Schübeler laboratory for insightful discussions and critical feedback on the manuscript. They similarly thank the reviewers for their insightful comments.

Author information

Authors and Affiliations

Authors

Contributions

All authors substantially contributed to researching and discussing content for the article, writing the manuscript and reviewing/editing the manuscript before submission.

Corresponding author

Correspondence to Dirk Schübeler.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Genetics thanks Gordon Hager and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Chromatin sensitivity

The propensity of transcription factors (TFs) to be inhibited from binding motifs due to reversible chromatin features such as modification of DNA (CpG methylation) or presence and actions of chromatin proteins (such as nucleosomes).

Methylation-sensitive

Refers to transcription factor (TF) motifs that are bound with lower affinity when CpG dinucleotides within the motif are methylated.

Methylation-preferred

Refers to transcription factor (TF) motifs that are bound with higher affinity when CpG dinucleotides within the motif are methylated.

Micrococcal nuclease

(MNase). An enzyme that generates cuts preferentially within linker DNA between nucleosomes and in nucleosome-depleted regions. Coupling MNase digestion of chromatin with next-generation sequencing generates maps of nucleosome position and density.

DNase I hypersensitive site

A chromatin region with a high rate of cleavage by DNase I due to its preference for open chromatin. DNase I hypersensitivity generally reflects transcription factor (TF) binding and a local reduction in nucleosome occupancy.

Genomic footprinting

The use of an ectopically supplied enzyme that adds chemical groups to DNA but is itself sensitive to the factors binding DNA, such as transcription factors (TFs) or nuclesomes. Its activity can subsequently be read out by sequencing.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Isbel, L., Grand, R.S. & Schübeler, D. Generating specificity in genome regulation through transcription factor sensitivity to chromatin. Nat Rev Genet 23, 728–740 (2022). https://doi.org/10.1038/s41576-022-00512-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-022-00512-6

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