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:

Transcriptional enhancers: from properties to genome-wide predictions

Key Points

  • The development of all organisms relies on differential gene expression, which is controlled by genomic regions called enhancers or cis-regulatory modules (CRMs). Recent studies highlight the importance of enhancers in evolution and disease; however, our understanding of their properties and functions remains incomplete.

  • Enhancers contain short DNA sequences, which are binding sites for transcription factors. In turn, transcription factors recruit cofactors, which modify the nearby chromatin and lead to transcriptional activation.

  • The location of putative enhancers can be predicted genome wide by assessing either the binding of transcription factors and cofactors or post-translational histone modifications by chromatin immunoprecipitation followed by deep sequencing (ChIP–seq). 'Open' chromatin with accessible DNA can be detected by DNase I hypersensitive site sequencing (DNase-seq), micrococcal nuclease sequencing (MNase-seq), formaldehyde-assisted isolation of regulatory elements followed by deep sequencing (FAIRE–seq) or assay for transposase-accessible chromatin using sequencing (ATAC-seq).

  • Distal enhancers can activate target gene expression by looping to promoters. Such spatial contacts can be detected by chromosome conformation capture (3C) assays and its variants circular chromosome conformation capture (4C), chromosome conformation capture carbon copy (5C) and Hi-C methods or by chromatin interaction analysis with paired-end tag sequencing (ChIA–PET, which is a combination of ChIP and various 3C-based methods).

  • The genome-wide prediction of enhancers based on characteristic chromatin features is powerful, but such results have to be interpreted with caution because none of the known features is perfectly predictive.

  • Enhancer activities of candidate sequences can be measured directly in a developmental context using image-based readouts or enhancer-FACS-seq. High-throughput parallel enhancer assays use either ectopic reporters to test thousands of candidates (which are based on DNA barcodes) or genome-wide screens (such as self-transcribing active regulatory region sequencing (STARR-seq)).

  • Our understanding of enhancer biology will be further accelerated by advances in genome editing methods (such as transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR)–Cas9 system), as well as by the development or improvements of methods to assess gene expression, chromatin state and structure in entire genomes and from increasingly few cells (such as thousands of reporters integrated in parallel (TRIP), single-cell RNA sequencing or ChIP–seq, and high-resolution Hi-C).

Abstract

Cellular development, morphology and function are governed by precise patterns of gene expression. These are established by the coordinated action of genomic regulatory elements known as enhancers or cis-regulatory modules. More than 30 years after the initial discovery of enhancers, many of their properties have been elucidated; however, despite major efforts, we only have an incomplete picture of enhancers in animal genomes. In this Review, we discuss how properties of enhancer sequences and chromatin are used to predict enhancers in genome-wide studies. We also cover recently developed high-throughput methods that allow the direct testing and identification of enhancers on the basis of their activity. Finally, we discuss recent technological advances and current challenges in the field of regulatory genomics.

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: Enhancers and their features.
Figure 2: Chromatin accessibility and histone marks at regulatory elements.
Figure 3: Genomic methods for predicting enhancers through the detection of transcription factor binding, 'open' chromatin, chromatin marks, or long-range contacts.
Figure 4: Novel approaches to study and manipulate endogenous regulatory activities.

Similar content being viewed by others

References

  1. Banerji, J., Rusconi, S. & Schaffner, W. Expression of a β-globin gene is enhanced by remote SV40 DNA sequences. Cell 27, 299–308 (1981). This paper reports the first sequence that can increase transcription levels from a given promoter, defines the term enhancer and describes many enhancer properties.

    Article  CAS  PubMed  Google Scholar 

  2. Banerji, J., Olson, L. & Schaffner, W. A lymphocyte-specific cellular enhancer is located downstream of the joining region in immunoglobulin heavy chain genes. 33, 729–740 (1983).

  3. Amano, T. et al. Chromosomal dynamics at the shh locus: limb bud-specific differential regulation of competence and active transcription. Dev. Cell 16, 47–57 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Arnone, M. I. & Davidson, E. H. The hardwiring of development: organization and function of genomic regulatory systems. Development 124, 1851–1864 (1997).

    CAS  PubMed  Google Scholar 

  5. Dawson, M. A. & Kouzarides, T. Cancer epigenetics: from mechanism to therapy. Cell 150, 12–27 (2012).

    CAS  PubMed  Google Scholar 

  6. Carroll, S. B. Evo-devo and an expanding evolutionary synthesis: a genetic theory of morphological evolution. Cell 134, 25–36 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Visel, A., Rubin, E. M. & Pennacchio, L. A. Genomic views of distant-acting enhancers. Nature 461, 199–205 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yáñez-Cuna, J. O., Kvon, E. Z. & Stark, A. Deciphering the transcriptional cis-regulatory code. Trends Genet. 29, 11–22 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Tomancak, P. et al. Systematic determination of patterns of gene expression during Drosophila embryogenesis. Genome Biol. 3, research0088-0088.14 (2002).

    Article  Google Scholar 

  10. Richardson, L. et al. EMAGE mouse embryo spatial gene expression database: 2010 update. Nucleic Acids Res. 38, D703–D709 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. International HapMap 3 Consortium. Integrating common and rare genetic variation in diverse human populations. Nature 467, 52–58 (2010).

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

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

  14. Tjian, R. The binding site on SV40 DNA for a T antigen-related protein. Cell 13, 165–179 (1978).

    Article  CAS  PubMed  Google Scholar 

  15. Giniger, E., Varnum, S. M. & Ptashne, M. Specific DNA binding of GAL4, a positive regulatory protein of yeast. Cell 40, 767–774 (1985).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. Kheradpour, P., Stark, A., Roy, S. & Kellis, M. Reliable prediction of regulator targets using 12 Drosophila genomes. Genome Res. 17, 1919–1931 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Del Bene, F. et al. In vivo validation of a computationally predicted conserved Ath5 target gene set. PLoS Genet. 3, 1661–1671 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Hallikas, O. et al. Genome-wide prediction of mammalian enhancers based on analysis of transcription-factor binding affinity. Cell 124, 47–59 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Sinha, S., van Nimwegen, E. & Siggia, E. D. A probabilistic method to detect regulatory modules. Bioinformatics 19, i292–i301 (2003).

    Article  PubMed  Google Scholar 

  21. Herrmann, C., Van de Sande, B., Potier, D. & Aerts, S. i-cisTarget: an integrative genomics method for the prediction of regulatory features and cis-regulatory modules. Nucleic Acids Res. 40, e114 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Warner, J. B. et al. Systematic identification of mammalian regulatory motifs' target genes and functions. Nature Methods 5, 347–353 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Aerts, S. Computational strategies for the genome-wide identification of cis-regulatory elements and transcriptional targets. Curr. Top. Dev. Biol. 98, 121–145 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Hardison, R. C. & Taylor, J. Genomic approaches towards finding cis-regulatory modules in animals. Nature Rev. Genet. 13, 469–483 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Wei, G.-H. et al. Genome-wide analysis of ETS-family DNA-binding in vitro and in vivo. EMBO J. 29, 2147–2160 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jolma, A. et al. DNA-binding specificities of human transcription factors. Cell 152, 327–339 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Noyes, M. B. et al. Analysis of homeodomain specificities allows the family-wide prediction of preferred recognition sites. Cell 133, 1277–1289 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yanez-Cuna, J. O., Dinh, H. Q., Kvon, E. Z., Shlyueva, D. & Stark, A. Uncovering cis-regulatory sequence requirements for context-specific transcription factor binding. Genome Res. 22, 2018–2030 (2012). This paper shows that transcription factor binding can be predicted by cell-type-specific combinations of transcription factor binding sequences for different partner transcription factors, which are shared across many binding sites.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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 

  30. Blow, M. J. et al. ChIP–seq identification of weakly conserved heart enhancers. Nature Genet. 42, 818–822 (2010).

    Article  CAS  Google Scholar 

  31. Meireles-Filho, A. C. A. & Stark, A. Comparative genomics of gene regulation-conservation and divergence of cis-regulatory information. Curr. Opin. Genet. Dev. 19, 565–570 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Kantorovitz, M. R. et al. Motif-blind, genome-wide discovery of cis-regulatory modules in Drosophila and mouse. Dev. Cell 17, 568–579 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Narlikar, L. et al. Genome-wide discovery of human heart enhancers. Genome Res. 20, 381–392 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Burzynski, G. M. et al. Systematic elucidation and in vivo validation of sequences enriched in hindbrain transcriptional control. Genome Res. 22, 2278–2289 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Robertson, G. et al. Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing. Nature Methods 4, 651–657 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. 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 

  38. van Steensel, B. & Henikoff, S. Identification of in vivo DNA targets of chromatin proteins using tethered Dam methyltransferase. Nature Biotech. 18, 424–428 (2000).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  40. Sandmann, T. et al. A core transcriptional network for early mesoderm development in Drosophila melanogaster. Genes Dev. 21, 436–449 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zeitlinger, J. et al. Whole-genome ChIP–chip analysis of Dorsal, Twist, and Snail suggests integration of diverse patterning processes in the Drosophila embryo. Genes Dev. 21, 385–390 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li, X.-Y. et al. Transcription factors bind thousands of active and inactive regions in the Drosophila blastoderm. PLoS Biol. 6, e27 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kvon, E. Z., Stampfel, G., Yáñez-Cuna, J. O., Dickson, B. J. & Stark, A. HOT regions function as patterned developmental enhancers and have a distinct cis-regulatory signature. Genes Dev. 26, 908–913 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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. 109, 21330–21335 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hammar, P. et al. The lac repressor displays facilitated diffusion in living cells. Science 336, 1595–1598 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. 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. 110, 18602–18607 (2013). This study shows that ChIP assays can lead to false-positive binding sites for transcription factors or even for non-DNA binding proteins (such as GFP), thus cautioning the interpretation of this widely used technique.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Poorey, K. et al. Measuring chromatin interaction dynamics on the second time scale at single-copy genes. Science 342, 369–372 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 (2013).

    Article  CAS  Google Scholar 

  49. 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 

  50. Visel, A. et al. ChIP–seq accurately predicts tissue-specific activity of enhancers. Nature 457, 854–858 (2009). This paper shows that p300 binding in the murine forebrain, hindbrain and limb can predict tissue-specific enhancers.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Heintzman, N. D. et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nature Genet. 39, 311–318 (2007). This study shows that human promoters and enhancers are marked by characteristic combinations of histone modifications that are predictive.

    Article  CAS  PubMed  Google Scholar 

  52. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Visel, A. et al. A high-resolution enhancer atlas of the developing telencephalon. Cell 152, 895–908 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. May, D. et al. Large-scale discovery of enhancers from human heart tissue. Nature Genet. 44, 89–93 (2012).

    Article  CAS  Google Scholar 

  55. Filion, G. J. et al. Systematic protein location mapping reveals five principal chromatin types in Drosophila cells. Cell 143, 212–224 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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 

  57. Ram, O. et al. Combinatorial patterning of chromatin regulators uncovered by genome-wide location analysis in human cells. Cell 147, 1628–1639 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Weintraub, H. & Groudine, M. Chromosomal subunits in active genes have an altered conformation. Science 193, 848–856 (1976).

    Article  CAS  PubMed  Google Scholar 

  59. Axel, R., Cedar, H. & Felsenfeld, G. Synthesis of globin ribonucleic acid from duck-reticulocyte chromatin in vitro. Proc. Natl. Acad. Sci. USA 70, 2029–2032 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Boyle, A. P. et al. High-resolution mapping and characterization of open chromatin across the genome. Cell 132, 311–322 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  62. Neph, S. et al. An expansive human regulatory lexicon encoded in transcription factor footprints. Nature 488, 83–90 (2012).

    Article  CAS  Google Scholar 

  63. 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 

  64. Bell, O., Tiwari, V. K., Thomä, N. H. & Schübeler, D. Determinants and dynamics of genome accessibility. Nature Rev. Genet. 12, 554–564 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. 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 

  66. Mirny, L. A. Nucleosome-mediated cooperativity between transcription factors. Proc. Natl Acad. Sci. 107, 22534–22539 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  67. 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 

  68. Pique-Regi, R. et al. Accurate inference of transcription factor binding from DNA sequence and chromatin accessibility data. Genome Res. 21, 447–455 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kaplan, T. et al. Quantitative models of the mechanisms that control genome-wide patterns of transcription factor binding during early Drosophila development. PLoS Genet. 7, e1001290 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Heintzman, N. D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 488, 75–82 (2013).

    Google Scholar 

  72. Arnold, C. D. et al. Genome-wide quantitative enhancer activity maps identified by STARR-seq. Science 339, 1074–1077 (2013). This paper introduces a method that allows the genome-wide identification of enhancers on the direct basis of their activity and that finds 'closed enhancers', which are silenced endogenously presumably at the chromatin level.

    Article  CAS  PubMed  Google Scholar 

  73. Xi, H. et al. Identification and characterization of cell type-specific and ubiquitous chromatin regulatory structures in the human genome. PLoS Genet. 3, e136 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Gray, S. & Levine, M. Transcriptional repression in development. Curr. Opin. Cell Biol. 8, 358–364 (1996).

    Article  CAS  PubMed  Google Scholar 

  75. Cochella, L. & Hobert, O. Embryonic priming of a miRNA locus predetermines postmitotic neuronal left/right asymmetry in C. elegans. Cell 151, 1229–1242 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    CAS  PubMed  Google Scholar 

  77. Roh, T.-Y., Cuddapah, S. & Zhao, K. Active chromatin domains are defined by acetylation islands revealed by genome-wide mapping. Genes Dev. 19, 542–552 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Bonn, S. et al. Tissue-specific analysis of chromatin state identifies temporal signatures of enhancer activity during embryonic development. Nature Genet. 44, 148–156 (2012). This study couples ChIP–seq with nuclear sorting to allow the cell-type-specific investigation of chromatin features.

    Article  CAS  PubMed  Google Scholar 

  79. Peters, A. H. F. M. et al. Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin. Nature Genet. 30, 77–80 (2002).

    Article  CAS  PubMed  Google Scholar 

  80. Simon, J. A. & Kingston, R. E. Mechanisms of Polycomb gene silencing: knowns and unknowns. Nature Rev. Mol. Cell. Biol. 10, 697–708 (2009).

    Article  CAS  Google Scholar 

  81. Wamstad, J. A. et al. Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 151, 206–220 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Shen, Y. et al. A map of the cis-regulatory sequences in the mouse genome. Nature 488, 116–120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kharchenko, P. V. et al. Comprehensive analysis of the chromatin landscape in Drosophila melanogaster. Nature 471, 480–485 (2012).

    Article  CAS  Google Scholar 

  84. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  86. Ostuni, R. et al. Latent enhancers activated by stimulation in differentiated cells. Cell 152, 157–171 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  88. Hah, N. et al. A rapid, extensive, and transient transcriptional response to estrogen signaling in breast cancer cells. Cell 145, 622–634 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Core, L. J. et al. Defining the status of RNA polymerase at promoters. Cell Rep. 2, 1025–1035 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Kim, T.-K. et al. Widespread transcription at neuronal activity-regulated enhancers. Nature 465, 182–187 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. De Santa, F. et al. A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoS Biol. 8, e1000384 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Lai, F. et al. Activating RNAs associate with Mediator to enhance chromatin architecture and transcription. Nature 494, 497–501 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Natoli, G. & Andrau, J.-C. Noncoding transcription at enhancers: general principles and functional models. Annu. Rev. Genet. 46, 1–19 (2012).

    Article  CAS  PubMed  Google Scholar 

  94. Ebisuya, M., Yamamoto, T., Nakajima, M. & Nishida, E. Ripples from neighbouring transcription. Nature Cell Biol. 10, 1106–1113 (2008).

    Article  CAS  PubMed  Google Scholar 

  95. Ponting, C. P., Oliver, P. L. & Reik, W. Evolution and functions of long noncoding RNAs. Cell 136, 629–641 (2009).

    Article  CAS  PubMed  Google Scholar 

  96. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nature Methods 9, 215–216 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Hödl, M. & Basler, K. Transcription in the absence of histone H3.2 and H3K4 methylation. Curr. Biol. 22, 2253–2257 (2012).

    Article  CAS  PubMed  Google Scholar 

  98. Pengelly, A. R., Copur, O., Jackle, H., Herzig, A. & Muller, J. A. Histone mutant reproduces the phenotype caused by loss of histone-modifying factor Polycomb. Science 339, 698–699 (2013). References 97 and 98 investigate the importance of histone modifications for gene transcription by mutating H3K4 and H3K27. H3-K27R mutants led to the derepression of Polycomb target genes but was otherwise compatible with gene transcription, as were H3K4 mutants that could not be methylated.

    Article  CAS  PubMed  Google Scholar 

  99. Hathaway, N. A. et al. Dynamics and memory of heterochromatin in living cells. Cell 149, 1447–1460 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Calo, E. & Wysocka, J. Modification of enhancer chromatin: what, how, and why? Mol. Cell 49, 825–837 (2013).

    Article  CAS  PubMed  Google Scholar 

  101. Kagey, M. H. et al. Mediator and cohesin connect gene expression and chromatin architecture. Nature 467, 430–435 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Schmidt, D. et al. A CTCF-independent role for cohesin in tissue-specific transcription. Genome Res. 20, 578–588 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. van Steensel, B. & Dekker, J. Genomics tools for unraveling chromosome architecture. Nature Biotech. 28, 1089–1095 (2010).

    Article  CAS  Google Scholar 

  105. Fullwood, M. J. et al. An oestrogen-receptor-α-bound human chromatin interactome. Nature 461, 58–64 (2009).

    Article  CAS  Google Scholar 

  106. Li, G. et al. Extensive promoter-centered chromatin interactions provide a topological basis for transcription regulation. Cell 148, 84–98 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Sexton, T. et al. Three-dimensional folding and functional organization principles of the Drosophila genome. Cell 148, 458–472 (2012).

    Article  CAS  PubMed  Google Scholar 

  109. Jin, F. et al. A high-resolution map of the three-dimensional chromatin interactome in human cells. Nature 503, 290–294 (2013). This paper significantly improves the resolution of Hi-C experiments and provides bulk evidence that implicates the interactions in gene expression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Gibcus, J. H. & Dekker, J. The hierarchy of the 3D genome. Mol. Cell 49, 773–782 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. de Laat, W. & Duboule, D. Topology of mammalian developmental enhancers and their regulatory landscapes. Nature 502, 499–506 (2013).

    Article  CAS  PubMed  Google Scholar 

  112. Sanyal, A., Lajoie, B. R., Jain, G. & Dekker, J. The long-range interaction landscape of gene promoters. Nature 489, 109–113 (2013).

    Article  CAS  Google Scholar 

  113. Lettice, L. A. A long-range Shh enhancer regulates expression in the developing limb and fin and is associated with preaxial polydactyly. Hum. Mol. Genet. 12, 1725–1735 (2003).

    Article  CAS  PubMed  Google Scholar 

  114. Sur, I. K. et al. Mice lacking a Myc enhancer that includes human SNP rs6983267 are resistant to intestinal tumors. Science 338, 1360–1363 (2012).

    Article  CAS  PubMed  Google Scholar 

  115. Huang, F. W. et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959 (2013). References 114 and 115 describe defined mutations in transcriptional regulatory regions (that is, promoters and enhancers) that are causally linked to the deregulation of MYC and telomerase reverse transcriptase (TERT), and cancer.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zeitlinger, J. & Stark, A. Developmental gene regulation in the era of genomics. Dev. Biol. 339, 230–239 (2010).

    Article  CAS  PubMed  Google Scholar 

  117. Crocker, J. & Stern, D. L. TALE-mediated modulation of transcriptional enhancers in vivo. Nature Methods 10, 762–767 (2013). This study recruits transcriptional activators and repressors to specific enhancers in D. melanogaster using TALE fusion proteins and thereby modulate target gene expression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Zhang, Y. et al. Chromatin connectivity maps reveal dynamic promoter-enhancer long-range associations. Nature 504, 306–310 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Kieffer-Kwon, K.-R. et al. Interactome maps of mouse gene regulatory domains reveal basic principles of transcriptional regulation. Cell 155, 1507–1520 (2013).

    Article  CAS  PubMed  Google Scholar 

  120. Zinzen, R. P., Girardot, C., Gagneur, J., Braun, M. & Furlong, E. E. M. Combinatorial binding predicts spatio-temporal cis-regulatory activity. Nature 462, 65–70 (2009).

    Article  CAS  PubMed  Google Scholar 

  121. Manning, L. et al. A resource for manipulating gene expression and analyzing cis-regulatory modules in the Drosophila CNS. Cell Rep. 2, 1002–1013 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Dupuy, D. A. First version of the Caenorhabditis elegans promoterome. Genome Res. 14, 2169–2175 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Visel, A., Minovitsky, S., Dubchak, I. & Pennacchio, L. A. VISTA enhancer browser — a database of tissue-specific human enhancers. Nucleic Acids Res. 35, D88–D92 (2007).

    Article  CAS  PubMed  Google Scholar 

  124. Gisselbrecht, S. S. et al. Highly parallel assays of tissue-specific enhancers in whole Drosophila embryos. Nature Methods 10, 774–780 (2013). This paper introduces enhancer-FACS-seq to parallelize in vivo enhancer testing in D. melanogaster.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Nam, J. & Davidson, E. H. Barcoded DNA-tag reporters for multiplex cis-regulatory analysis. PLoS ONE 7, e35934 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Melnikov, A. et al. Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay. Nature Biotech. 30, 271–277 (2012).

    Article  CAS  Google Scholar 

  127. Patwardhan, R. P. et al. Massively parallel functional dissection of mammalian enhancers in vivo. Nature Biotech. 30, 265–270 (2012).

    Article  CAS  Google Scholar 

  128. Kwasnieski, J. C., Mogno, I., Myers, C. A., Corbo, J. C. & Cohen, B. A. Complex effects of nucleotide variants in a mammalian cis-regulatory element. Proc. Natl. Acad. Sci. 109, 19498–19503 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Sharon, E. et al. Inferring gene regulatory logic from high-throughput measurements of thousands of systematically designed promoters. Nature Biotech. 30, 521–530 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  131. Gertz, J., Siggia, E. D. & Cohen, B. A. Analysis of combinatorial cis-regulation in synthetic and genomic promoters. Nature 457, 215–218 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. 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 

  133. Adli, M. & Bernstein, B. E. Whole-genome chromatin profiling from limited numbers of cells using nano-ChIP–seq. Nature Protoc. 6, 1656–1668 (2011).

    Article  CAS  Google Scholar 

  134. 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 

  135. 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 Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Deng, Q., Ramsköld, D., Reinius, B. & Sandberg, R. Single-cell RNA-seq reveals dynamic, random monoallelic gene expression in mammalian cells. Science 343, 193–196 (2014).

    Article  CAS  PubMed  Google Scholar 

  137. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  139. 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 

  140. Reyon, D. et al. FLASH assembly of TALENs for high-throughput genome editing. Nature Biotech. 30, 460–465 (2012).

    Article  CAS  Google Scholar 

  141. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012). This study shows that the bacterial Cas9 protein uses dual RNAs for sequence-specific DNA targeting and cleavage, and highlights the potential of the CRISPR–Cas9 system for genome editing.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Ruf, S. et al. Large-scale analysis of the regulatory architecture of the mouse genome with a transposon-associated sensor. Nature Genet. 43, 379–386 (2011).

    Article  CAS  PubMed  Google Scholar 

  144. Mollereau, B. et al. A green fluorescent protein enhancer trap screen in Drosophila photoreceptor cells. Mech. Dev. 93, 151–160 (2000).

    Article  CAS  PubMed  Google Scholar 

  145. Akhtar, W. et al. Chromatin position effects assayed by thousands of reporters integrated in parallel. Cell 154, 914–927 (2013).

    Article  CAS  PubMed  Google Scholar 

  146. Maurano, M. T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337, 1190–1195 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013). This paper defines super enhancers as exceptionally long genomic regions that are strongly bound by cofactors. The proximity to some oncogenes and the loss of bromodomain-containing protein 4 (BRD4) binding upon inhibition draws the attention of a broader medical community to such enhancers.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Shi, J. et al. Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev. 27, 2648–2662 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011). This study reports that inhibition of the broadly expressed transcriptional co-activator BRD4 has a specific effect on acute myeloid leukaemia cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Delmore, J. E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Knutson, S. K. et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nature Chem. Biol. 8, 890–896 (2012).

    Article  CAS  Google Scholar 

  152. Herrmann, H. et al. Small-molecule inhibition of BRD4 as a new potent approach to eliminate leukemic stem- and progenitor cells in acute myeloid leukemia AML. Oncotarget 3, 1588–1599 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  153. Stormo, G. D. & Zhao, Y. Determining the specificity of protein–DNA interactions. Nature Rev. Genet. 11, 751–760 (2010).

    Article  CAS  PubMed  Google Scholar 

  154. Roth, F. P., Hughes, J. D., Estep, P. W. & Church, G. M. Finding DNA regulatory motifs within unaligned noncoding sequences clustered by whole-genome mRNA quantitation. Nature Biotech. 16, 939–945 (1998).

    Article  CAS  Google Scholar 

  155. Kellis, M., Patterson, N., Endrizzi, M., Birren, B. & Lander, E. S. Sequencing and comparison of yeast species to identify genes and regulatory elements. Nature 423, 241–254 (2003).

    Article  CAS  PubMed  Google Scholar 

  156. Bosch, J. R., Benavides, J. A. & Cline, T. W. The TAGteam DNA motif controls the timing of Drosophila pre-blastoderm transcription. Development 133, 1967–1977 (2006).

    Article  CAS  PubMed  Google Scholar 

  157. Liang, H.-L. et al. The zinc-finger protein Zelda is a key activator of the early zygotic genome in Drosophila. Nature 456, 400–403 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Bulyk, M. L., Gentalen, E., Lockhart, D. J. & Church, G. M. Quantifying DNA–protein interactions by double-stranded DNA arrays. Nature Biotech. 17, 573–577 (1999).

    Article  CAS  Google Scholar 

  159. Tuerk, C. & Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249, 505–510 (1990).

    Article  CAS  PubMed  Google Scholar 

  160. Li, J. J. & Herskowitz, I. Isolation of ORC6, a component of the yeast origin recognition complex by a one-hybrid system. Science 262, 1870–1874 (1993).

    Article  CAS  PubMed  Google Scholar 

  161. Meng, X., Brodsky, M. H. & Wolfe, S. A. A bacterial one-hybrid system for determining the DNA-binding specificity of transcription factors. Nature Biotech. 23, 988–994 (2005).

    Article  CAS  Google Scholar 

  162. Matys, V. et al. TRANSFAC and its module TRANSCompel: transcriptional gene regulation in eukaryotes. Nucleic Acids Res. 34, D108–D110 (2006).

    Article  CAS  PubMed  Google Scholar 

  163. Portales-Casamar, E. et al. JASPAR 2010: the greatly expanded open-access database of transcription factor binding profiles. Nucleic Acids Res. 38, D105–D110 (2010).

    Article  CAS  PubMed  Google Scholar 

  164. Newburger, D. E. & Bulyk, M. L. UniPROBE: an online database of protein binding microarray data on protein-DNA interactions. Nucleic Acids Res. 37, D77–D82 (2009).

    Article  CAS  PubMed  Google Scholar 

  165. Mogno, I., Kwasnieski, J. C. & Cohen, B. A. Massively parallel synthetic promoter assays reveal the in vivo effects of binding site variants. Genome Res. 23, 1908–1915 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank O. Bell (Institute of Molecular Biotechnology (IMBA), Vienna, Austria) and members of A.S's group (Research Institute of Molecular Pathology (IMP), Vienna, Austria) for discussions. We apologize to all colleagues whose work could not be discussed or referenced owing to space limitations. D.S. is supported by a European Research Council (ERC) Starting grant (no. 242922) awarded to A.S., and A.S.'s group by the Austrian Science Fund (FWF): F4303-B09. Basic research at the IMP is supported by Boehringer Ingelheim GmbH.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alexander Stark.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

FURTHER INFORMATION

REDfly

Vienna Tiles Browser

VISTA

PowerPoint slides

Glossary

Transcription factor binding motif

(Also known as transcription factor sequence motif, transcription factor motif and transcription factor binding site motif.) A degenerate short (6–10-bp) DNA sequence pattern that summarizes the DNA sequence binding preference of a transcription factor. These motifs are usually represented either as consensus sequences in IUPAC code or by position weight matrices.

Transcription factor motif matches

(Also known as transcription factor motif instances, transcription factor motif occurrences and transcription factor binding sequences). Specific genomic sequences or positions that match transcription factor binding motifs and thus constitute potential transcription factor binding sites. These are also sometimes called transcription factor binding sites, although we prefer to reserve this term for experimentally determined ones.

Position weight matrices

(Also known as position-specific weight matrices or position-specific scoring matrices). Matrices that provide the frequencies at which individual nucleotides are found at the positions of the transcription factor binding motif.

Transcription factor binding sites

Genomic locations of transcription factor binding, typically in vivo. These sites can be determined experimentally (for example, using chromatin immunoprecipitation (ChIP)). ChIP experiments typically reveal that these binding sites and transcription factor motif matches often, but not always, coincide.

Insulator

A chromatin element that acts as a barrier against the influence of positive signals (from enhancers) or negative signals (from silencers and heterochromatin).

Silencers

DNA sequences that cause reduced expression of their target gene (or genes).

Global run-on sequencing

(GRO-seq). A genome-wide method that maps the position and amount of transcriptionally engaged RNA polymerase II.

Chromatin interaction analysis with paired-end tag sequencing

(ChIA–PET). A high-throughput method based on a combination of chromatin immunoprecipitation (ChIP) and chromatin proximity ligation assays to predict long-range chromatin interactions that are mediated by either RNA polymerase II or transcription factors.

Barcodes

Short and typically artificially designed DNA sequences that are used to uniquely identify DNA constructs (for example, those expressing short hairpin RNAs or reporter genes) or cell lines (for example, yeast knockouts). The uniqueness of the barcodes allows screening or testing in parallel using pooling.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shlyueva, D., Stampfel, G. & Stark, A. Transcriptional enhancers: from properties to genome-wide predictions. Nat Rev Genet 15, 272–286 (2014). https://doi.org/10.1038/nrg3682

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3682

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