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:

Towards a comprehensive catalogue of validated and target-linked human enhancers

Abstract

The human gene catalogue is essentially complete, but we lack an equivalently vetted inventory of bona fide human enhancers. Hundreds of thousands of candidate enhancers have been nominated via biochemical annotations; however, only a handful of these have been validated and confidently linked to their target genes. Here we review emerging technologies for discovering, characterizing and validating human enhancers at scale. We furthermore propose a new framework for operationally defining enhancers that accommodates the heterogeneous and complementary results that are emerging from reporter assays, biochemical measurements and CRISPR screens.

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: Approaches for identifying, validating and characterizing enhancers.
Fig. 2: CRISPR-based approaches for perturbing enhancers.
Fig. 3: CRISPR-based screens of enhancer–gene links.
Fig. 4: A tiered framework to describe the level of support for the enhancer candidacy of a non-coding sequence.
Fig. 5: The blind men and the elephant of human enhancer biology.

Similar content being viewed by others

References

  1. Jacob, F. & Monod, J. Genetic regulatory mechanisms in the synthesis of proteins. J. Mol. Biol. 3, 318–356 (1961).

    CAS  PubMed  Google Scholar 

  2. Ptashne, M. Specific binding of the λ phage repressor to λ DNA. Nature 214, 232–234 (1967).

    CAS  PubMed  Google Scholar 

  3. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  5. Stalder, J. et al. Tissue-specific DNA cleavages in the globin chromatin domain introduced by DNAase I. Cell 20, 451–460 (1980).

    CAS  PubMed  Google Scholar 

  6. Moreau, P. et al. The SV40 72 base repair repeat has a striking effect on gene expression both in SV40 and other chimeric recombinants. Nucleic Acids Res. 9, 6047–6068 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Banerji, J., Rusconi, S. & Schaffner, W. Expression of a β-globin gene is enhanced by remote SV40 DNA sequences. Cell 27, 299–308 (1981). The first episomal observation of in vitro enhancer activity, this work coined the term ‘enhancer’.

    CAS  PubMed  Google Scholar 

  8. Mercola, M., Wang, X., Olsen, J. & Calame, K. Transcriptional enhancer elements in the mouse immunoglobulin heavy chain locus. Science 221, 663–665 (1983).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  10. Gillies, S. D., Morrison, S. L., Oi, V. T. & Tonegawa, S. A tissue-specific transcription enhancer element is located in the major intron of a rearranged immunoglobulin heavy chain gene. Cell 33, 717–728 (1983).

    CAS  PubMed  Google Scholar 

  11. Hanahan, D. Heritable formation of pancreatic β-cell tumours in transgenic mice expressing recombinant insulin/simian virus 40 oncogenes. Nature 315, 115–122 (1985).

    CAS  PubMed  Google Scholar 

  12. Tuan, D., Solomon, W., Li, Q. & London, I. M. The ‘β-like-globin’ gene domain in human erythroid cells. Proc. Natl Acad. Sci. 82, 6384–6388 (1985).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Gross, D. S. & Garrard, W. T. Nuclease hypersensitive sites in chromatin. Annu. Rev. Biochem. 57, 159–197 (1988).

    CAS  PubMed  Google Scholar 

  14. Hebbes, T. R., Thorne, A. W. & Crane-Robinson, C. A direct link between core histone acetylation and transcriptionally active chromatin. EMBO J. 7, 1395–1402 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Lee, D. Y., Hayes, J. J., Pruss, D. & Wolffe, A. P. A positive role for histone acetylation in transcription factor access to nucleosomal DNA. Cell 72, 73–84 (1993).

    CAS  PubMed  Google Scholar 

  16. Hebbes, T. R., Clayton, A. L., Thorne, A. W. & Crane-Robinson, C. Core histone hyperacetylation co-maps with generalized DNase I sensitivity in the chicken β-globin chromosomal domain. EMBO J. 13, 1823–1830 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Serfling, E., Jasin, M. & Schaffner, W. Enhancers and eukaryotic gene transcription. Trends Genet. 1, 224–230 (1985).

    CAS  Google Scholar 

  18. Ikuta, T. & Kan, Y. W. In vivo protein–DNA interactions at the β-globin gene locus. Proc. Natl Acad. Sci. USA 88, 10188–10192 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Forsberg, M. & Westin, G. Enhancer activation by a single type of transcription factor shows cell type dependence. EMBO J. 10, 2543–2551 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Ptashne, M. Gene regulation by proteins acting nearby and at a distance. Nature 322, 697–701 (1986).

    CAS  PubMed  Google Scholar 

  21. Müeller-Storm, H. P., Sogo, J. M. & Schaffner, W. An enhancer stimulates transcription in trans when attached to the promoter via a protein bridge. Cell 58, 767–777 (1989).

    PubMed  Google Scholar 

  22. Van der Ploeg, L. H. T. et al. γ-β-Thalassaemia studies showing that deletion of the γ- and δ-genes influences β-globin gene expression in man. Nature 283, 637–642 (1980).

    PubMed  Google Scholar 

  23. Kioussis, D., Vanin, E., deLange, T., Flavell, R. A. & Grosveld, F. G. β-Globin gene inactivation by DNA translocation in γβ-thalassaemia. Nature 306, 662–666 (1983).

    CAS  PubMed  Google Scholar 

  24. Driscoll, M. C., Dobkin, C. S. & Alter, B. P. γδβ-Thalassemia due to a de novo mutation deleting the 5ʹ β-globin gene activation-region hypersensitive sites. Proc. Natl Acad. Sci. 86, 7470–7474 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Philipsen, S., Talbot, D., Fraser, P. & Grosveld, F. The β-globin dominant control region: hypersensitive site 2. EMBO J. 9, 2159–2167 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Talbot, D., Philipsen, S., Fraser, P. & Grosveld, F. Detailed analysis of the site 3 region of the human β-globin dominant control region. EMBO J. 9, 2169–2177 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Grosveld, F. et al. The regulation of human globin gene switching. Philos. Trans. R. Soc. Lond. B Biol. Sci. 339, 183–191 (1993).

    CAS  PubMed  Google Scholar 

  28. Moon, A. M. & Ley, T. J. Conservation of the primary structure, organization, and function of the human and mouse β-globin locus-activating regions. Proc. Natl Acad. Sci. USA 87, 7693–7697 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Margot, J. B., Demers, G. W. & Hardison, R. C. Complete nucleotide sequence of the rabbit β-like globin gene cluster: analysis of intergenic sequences and comparison with the human β-like globin gene cluster. J. Mol. Biol. 205, 15–40 (1989).

    CAS  PubMed  Google Scholar 

  30. Li, Q., Zhou, B., Powers, P., Enver, T. & Stamatoyannopoulos, G. Primary structure of the goat β-globin locus control region. Genomics 9, 488–499 (1991).

    CAS  PubMed  Google Scholar 

  31. Reitman, M. & Felsenfeld, G. Developmental regulation of topoisomerase II sites and DNase I-hypersensitive sites in the chicken β-globin locus. Mol. Cell. Biol. 10, 2774–2786 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Dunham, I. et al. The DNA sequence of human chromosome 22. Nature 402, 489–495 (1999).

    CAS  PubMed  Google Scholar 

  33. International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Google Scholar 

  34. Hardison, R. C., Oeltjen, J. & Miller, W. Long human–mouse sequence alignments reveal novel regulatory elements: a reason to sequence the mouse genome. Genome Res. 7, 959–966 (1997).

    CAS  PubMed  Google Scholar 

  35. Loots, G. G. et al. Identification of a coordinate regulator of interleukins 4, 13, and 5 by cross-species sequence comparisons. Science 288, 136–140 (2000). This study identifies the non-coding regions regulating several interleukin genes by comparing 1 Mb of mouse–human orthologous sequences. The global application of this strategy was one of the key motivations for sequencing of the mouse genome.

    CAS  PubMed  Google Scholar 

  36. Hardison, R. C. Conserved noncoding sequences are reliable guides to regulatory elements. Trends Genet. 16, 369–372 (2000).

    CAS  PubMed  Google Scholar 

  37. Pennacchio, L. A. & Rubin, E. M. Genomic strategies to identify mammalian regulatory sequences. Nat. Rev. Genet. 2, 100–109 (2001).

    CAS  PubMed  Google Scholar 

  38. Nobrega, M. A., Ovcharenko, I., Afzal, V. & Rubin, E. M. Scanning human gene deserts for long-range enhancers. Science 302, 413 (2003).

    CAS  PubMed  Google Scholar 

  39. Pennacchio, L. A. et al. In vivo enhancer analysis of human conserved non-coding sequences. Nature 444, 499–502 (2006).

    CAS  PubMed  Google Scholar 

  40. Mouse Genome Sequencing Consortium et al. Initial sequencing and comparative analysis of the mouse genome. Nature 420, 520–562 (2002).

    Google Scholar 

  41. Dermitzakis, E. T. et al. Numerous potentially functional but non-genic conserved sequences on human chromosome 21. Nature 420, 578–582 (2002).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 489, 75–82 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Laurent, L. et al. Dynamic changes in the human methylome during differentiation. Genome Res. 20, 320–331 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lister, R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Cokus, S. J. et al. Shotgun bisulphite sequencing of the Arabidopsis genome reveals DNA methylation patterning. Nature 452, 215–219 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    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. Nat. Genet. 39, 311–318 (2007). This study provides one of the earliest uses of genome-wide biochemical annotation datasets to annotate candidate enhancers.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  54. Visel, A. et al. ChIP-seq accurately predicts tissue-specific activity of enhancers. Nature 457, 854–858 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Shendure, J. et al. DNA sequencing at 40: past, present and future. Nature 550, 345–353 (2017).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455–461 (2014). This article reports an integrated analysis of cap analysis of gene expression (CAGE) datasets across hundreds of cell types and tissues performed to generate biochemical annotations of thousands of cell type-specific enhancers by their signatures of bidirectional transcription.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Core, L. J. et al. Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers. Nat. Genet. 46, 1311–1320 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012). This report summarizes the wealth of data generated by the ENCODE Consortium. Hundreds of genome-wide datasets are used across hundreds of cell types and tissues to ascribe a function to the majority of the genome via biochemical annotation.

    Google Scholar 

  61. Roadmap Epigenomics Consortium et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    PubMed Central  Google Scholar 

  62. Stunnenberg, H. G., International Human Epigenome Consortium & Hirst, M. The International Human Epigenome Consortium: A Blueprint for Scientific Collaboration and Discovery. Cell 167, 1897 (2016).

    CAS  PubMed  Google Scholar 

  63. Arner, E. et al. Transcribed enhancers lead waves of coordinated transcription in transitioning mammalian cells. Science 347, 1010–1014 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. ENCODE Project. SCREEN: search candidate regulatory elements by ENCODE. ENCODE Project http://screen.encodeproject.org/index/about (2019).

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

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Contributors to Wikimedia projects. Blind Men and An Elephant—Wikipedia (Wikimedia Foundation, Inc., 2006).

  67. Patwardhan, R. P. et al. Massively parallel functional dissection of mammalian enhancers in vivo. Nat. Biotechnol. 30, 265–270 (2012). Along with Melnikov et al. (2012), this study is the first application of an MPRA to enhancer sequences.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  70. Blackwood, E. M. & Kadonaga, J. T. Going the distance: a current view of enhancer action. Science 281, 60–63 (1998).

    CAS  PubMed  Google Scholar 

  71. Li, L. & Wunderlich, Z. An enhancer’s length and composition are shaped by its regulatory task. Front. Genet. 8, 63 (2017).

    PubMed  PubMed Central  Google Scholar 

  72. Gasperini, M. et al. A genome-wide framework for mapping gene regulation via cellular genetic screens. Cell 176, 377–390 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Smedley, D. et al. A whole-genome analysis framework for effective identification of pathogenic regulatory variants in Mendelian disease. Am. J. Hum. Genet. 99, 595–606 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Corradin, O. & Scacheri, P. C. Enhancer variants: evaluating functions in common disease. Genome Med. 6, 85 (2014).

    PubMed  PubMed Central  Google Scholar 

  75. Gjoneska, E. et al. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature 518, 365–369 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Wang, X. et al. High-resolution genome-wide functional dissection of transcriptional regulatory regions and nucleotides in human. Nat. Commun. 9, 5380 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Ghavi-Helm, Y. et al. Highly rearranged chromosomes reveal uncoupling between genome topology and gene expression. Nat. Genet. 51, 1272–1282 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Zabidi, M. A. et al. Enhancer-core-promoter specificity separates developmental and housekeeping gene regulation. Nature 518, 556–559 (2015). STARR-seq utilizing different classes of promoters in D. melanogaster shows that enhancers often do not work in a promoter-generic fashion.

    CAS  PubMed  Google Scholar 

  80. Dorighi, K. M. et al. Mll3 and Mll4 facilitate enhancer RNA synthesis and transcription from promoters independently of H3K4 monomethylation. Mol. Cell 66, 568–576.e4 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Rickels, R. et al. Histone H3K4 monomethylation catalyzed by Trr and mammalian COMPASS-like proteins at enhancers is dispensable for development and viability. Nat. Genet. 49, 1647–1653 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Eagen, K. P. Principles of chromosome architecture revealed by Hi-C. Trends Biochem. Sci. 43, 469–478 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Inoue, F. & Ahituv, N. Decoding enhancers using massively parallel reporter assays. Genomics 106, 159–164 (2015).

    CAS  PubMed  Google Scholar 

  86. Klein, J. C., Chen, W., Gasperini, M. & Shendure, J. Identifying novel enhancer elements with CRISPR-based screens. ACS Chem. Biol. 13, 326–332 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Hnisz, D., Day, D. S. & Young, R. A. Insulated neighborhoods: structural and functional units of mammalian gene control. Cell 167, 1188–1200 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Maricque, B. B., Chaudhari, H. G. & Cohen, B. A. A massively parallel reporter assay dissects the influence of chromatin structure on cis-regulatory activity. Nat. Biotechnol. 37, 90–95 (2019).

    CAS  Google Scholar 

  89. Heinz, S., Romanoski, C. E., Benner, C. & Glass, C. K. The selection and function of cell type-specific enhancers. Nat. Rev. Mol. Cell Biol. 16, 144–154 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature 518, 331–336 (2015). This study shows compelling use of Hi-C contact maps to study the dynamics of enhancer–promoter interactions through cellular differentiation.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Thanos, D. & Maniatis, T. Virus induction of human IFNβ gene expression requires the assembly of an enhanceosome. Cell 83, 1091–1100 (1995).

    CAS  PubMed  Google Scholar 

  92. Simeonov, D. R. et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Ray, J. et al. Chromatin conformation remains stable upon extensive transcriptional changes driven by heat shock. Proc. Natl Acad. Sci. USA 116, 19431–19439 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Fuda, N. J., Ardehali, M. B. & Lis, J. T. Defining mechanisms that regulate RNA polymerase II transcription in vivo. Nature 461, 186–192 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Juven-Gershon, T., Cheng, S. & Kadonaga, J. T. Rational design of a super core promoter that enhances gene expression. Nat. Methods 3, 917–922 (2006).

    CAS  PubMed  Google Scholar 

  96. Chen, F. X. et al. PAF1 regulation of promoter-proximal pause release via enhancer activation. Science 357, 1294–1298 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Engreitz, J. M. et al. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539, 452–455 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Murakami, S., Nagari, A. & Kraus, W. L. Dynamic assembly and activation of estrogen receptor α enhancers through coregulator switching. Genes. Dev. 31, 1535–1548 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Gosselin, D. et al. An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

    PubMed  PubMed Central  Google Scholar 

  100. Vihervaara, A. et al. Transcriptional response to stress is pre-wired by promoter and enhancer architecture. Nat. Commun. 8, 255 (2017).

    PubMed  PubMed Central  Google Scholar 

  101. Iwafuchi-Doi, M. et al. The pioneer transcription factor foxa maintains an accessible nucleosome configuration at enhancers for tissue-specific gene activation. Mol. Cell 62, 79–91 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Furlong, E. E. M. & Levine, M. Developmental enhancers and chromosome topology. Science 361, 1341–1345 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  104. Lindblad-Toh, K. et al. A high-resolution map of human evolutionary constraint using 29 mammals. Nature 478, 476–482 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Fulco, C. P. et al. Systematic mapping of functional enhancer-promoter connections with CRISPR interference. Science 354, 769–773 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Henriques, T. et al. Widespread transcriptional pausing and elongation control at enhancers. Genes Dev. 32, 26–41 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Kwasnieski, J. C., Fiore, C., Chaudhari, H. G. & Cohen, B. A. High-throughput functional testing of ENCODE segmentation predictions. Genome Res. 24, 1595–1602 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Muerdter, F. et al. Resolving systematic errors in widely used enhancer activity assays in human cells. Nat. Methods 15, 141–149 (2018).

    CAS  PubMed  Google Scholar 

  109. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Weintraub, A. S. et al. YY1 Is a structural regulator of enhancer-promoter loops. Cell 171, 1573–1588.e28 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Williamson, I. et al. Anterior–posterior differences in HoxD chromatin topology in limb development. Development 139, 3157–3167 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Ghavi-Helm, Y. et al. Enhancer loops appear stable during development and are associated with paused polymerase. Nature 512, 96–100 (2014).

    CAS  PubMed  Google Scholar 

  113. Benabdallah, N. S. et al. Decreased enhancer-promoter proximity accompanying enhancer activation. Mol. Cell 76, 473–484 (2019).

    Google Scholar 

  114. Alexander, J. M. et al. Live-cell imaging reveals enhancer-dependent Sox2 transcription in the absence of enhancer proximity. eLife 8, e41769 (2019).

    PubMed  PubMed Central  Google Scholar 

  115. Osterwalder, M. et al. Enhancer redundancy provides phenotypic robustness in mammalian development. Nature 554, 239–243 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Levings, P. P. & Bungert, J. The human β-globin locus control region. Eur. J. Biochem. 269, 1589–1599 (2002).

    CAS  PubMed  Google Scholar 

  117. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    CAS  PubMed  Google Scholar 

  118. Lettice, L. A. et al. 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).

    CAS  PubMed  Google Scholar 

  119. Bahr, C. et al. Author correction: A Myc enhancer cluster regulates normal and leukaemic haematopoietic stem cell hierarchies. Nature 558, E4 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  121. Cho, S. W. et al. Promoter of lncRNA Gene PVT1 Is a tumor-suppressor DNA boundary element. Cell 173, 1398–1412.e22 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Cinghu, S. et al. Intragenic enhancers attenuate host gene expression. Mol. Cell 68, 104–117.e6 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Blow, M. J. et al. ChIP-seq identification of weakly conserved heart enhancers. Nat. Genet. 42, 806–810 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Schmidt, D. et al. Five-vertebrate ChIP-seq reveals the evolutionary dynamics of transcription factor binding. Science 328, 1036–1040 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. King, M. C. & Wilson, A. C. Evolution at two levels in humans and chimpanzees. Science 188, 107–116 (1975).

    CAS  PubMed  Google Scholar 

  126. Danko, C. G. et al. Dynamic evolution of regulatory element ensembles in primate CD4+ T cells. Nat. Ecol. Evol. 2, 537–548 (2018).

    PubMed  PubMed Central  Google Scholar 

  127. Kulakovskiy, I. V. et al. HOCOMOCO: a comprehensive collection of human transcription factor binding sites models. Nucleic Acids Res. 41, D195–D202 (2013).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  129. Van Loo, P. & Marynen, P. Computational methods for the detection of cis-regulatory modules. Brief. Bioinform. 10, 509–524 (2009).

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Worsley Hunt, R. & Wasserman, W. W. Non-targeted transcription factors motifs are a systemic component of ChIP-seq datasets. Genome Biol. 15, 412 (2014).

    PubMed  PubMed Central  Google Scholar 

  132. Jain, D., Baldi, S., Zabel, A., Straub, T. & Becker, P. B. Active promoters give rise to false positive ‘phantom peaks’ in ChIP-seq experiments. Nucleic Acids Res. 43, 6959–6968 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Diao, Y. et al. A new class of temporarily phenotypic enhancers identified by CRISPR/Cas9-mediated genetic screening. Genome Res. 26, 397–405 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Pliner, H. A. et al. Cicero predicts cis-regulatory DNA Interactions from single-cell chromatin accessibility data. Mol. Cell 71, 858–871.e8 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  136. GTEx Consortium et al. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    PubMed Central  Google Scholar 

  137. Strober, B. J. et al. Dynamic genetic regulation of gene expression during cellular differentiation. Science 364, 1287–1290 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. van der Wijst, M. G. P. et al. Single-cell RNA sequencing identifies celltype-specific cis-eQTLs and co-expression QTLs. Nat. Genet. 50, 493–497 (2018).

    PubMed  PubMed Central  Google Scholar 

  139. Ptashne, M. How eukaryotic transcriptional activators work. Nature 335, 683–689 (1988).

    CAS  PubMed  Google Scholar 

  140. Schleif, R. DNA looping. Annu. Rev. Biochem. 61, 199–223 (1992).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Dekker, J., Rippe, K., Dekker, M. & Kleckner, N. Capturing chromosome conformation. Science 295, 1306–1311 (2002).

    CAS  PubMed  Google Scholar 

  143. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  145. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  147. Fullwood, M. J. & Ruan, Y. ChIP-based methods for the identification of long-range chromatin interactions. J. Cell. Biochem. 107, 30–39 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Mumbach, M. R. et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Fang, R. et al. Mapping of long-range chromatin interactions by proximity ligation-assisted ChIP-seq. Cell Res. 26, 1345–1348 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Ma, W. et al. Fine-scale chromatin interaction maps reveal the cis-regulatory landscape of human lincRNA genes. Nat. Methods 12, 71–78 (2015).

    PubMed  Google Scholar 

  151. Chen, H. et al. Dynamic interplay between enhancer-promoter topology and gene activity. Nat. Genet. 50, 1296–1303 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Schmitt, A. D. et al. A compendium of chromatin contact maps reveals spatially active regions in the human genome. Cell Rep. 17, 2042–2059 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Williamson, I., Lettice, L. A., Hill, R. E. & Bickmore, W. A. Shh and ZRS enhancer colocalisation is specific to the zone of polarising activity. Development 143, 2994–3001 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Gu, B. et al. Transcription-coupled changes in nuclear mobility of mammalian cis-regulatory elements. Science 359, 1050–1055 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320.e24 (2017). Rapid cohesin depletion in cultured cells results in widespread loss of the genome’s 3D organization, with minimal changes in gene expression. This has caused the field to question the importance of genome organization (that is, stable enhancer–promoter loops as they had been conceptualized).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Nora, E. P. et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169, 930–944.e22 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Trapnell, C. Defining cell types and states with single-cell genomics. Genome Res. 25, 1491–1498 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Novick, A. & Weiner, M. Enzyme induction as an all-or-none phenomenon. Proc. Natl Acad. Sci. USA 43, 553–566 (1957).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Bonn, S. et al. Tissue-specific analysis of chromatin state identifies temporal signatures of enhancer activity during embryonic development. Nat. Genet. 44, 148–156 (2012).

    CAS  PubMed  Google Scholar 

  160. Cusanovich, D. A. et al. A single-cell atlas of in vivo mammalian chromatin accessibility. Cell 174, 1309–1324.e18 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Cusanovich, D. A. et al. The cis-regulatory dynamics of embryonic development at single-cell resolution. Nature 555, 538–542 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Lai, B. et al. Publisher correction: principles of nucleosome organization revealed by single-cell micrococcal nuclease sequencing. Nature 564, E17 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Ramani, V. et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Flyamer, I. M. et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 544, 110–114 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Nagano, T. et al. Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature 547, 61–67 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  167. Tan, L., Xing, D., Chang, C.-H., Li, H. & Xie, X. S. Three-dimensional genome structures of single diploid human cells. Science 361, 924–928 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Lee, D. S. et al. Simultaneous profiling of 3D genome structure and DNA methylation in single human cells. Nat. Methods 16, 999–1006 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Rotem, A. et al. Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol. 33, 1165–1172 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Hainer, S. J., Bošković, A., McCannell, K. N., Rando, O. J. & Fazzio, T. G. Profiling of pluripotency factors in single cells and early embryos. Cell 177, 1319–1329 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Benabdallah, N. S. et al. Decreased enhancer–promoter proximity accompanying enhancer activation. Mol. Cell 76, 473–484.e7 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Fukaya, T., Lim, B. & Levine, M. Enhancer control of transcriptional bursting. Cell 166, 358–368 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Patwardhan, R. P. et al. High-resolution analysis of DNA regulatory elements by synthetic saturation mutagenesis. Nat. Biotechnol. 27, 1173–1175 (2009). This study provides the first demonstration of an MPRA coupled to sequencing-based readout, used here to evaluate all possible single-nucleotide variants of bacteriophage and mammalian core promoters.

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Arnold, C. D. et al. Genome-wide quantitative enhancer activity maps identified by STARR-seq. Science 339, 1074–1077 (2013). This is the first article to describe STARR-seq, as well as the first whole-genome shotgun MPRA. Quantitative, genome-wide maps of enhancer potential generated in two different D. melanogaster cell lines provide insight into the general characteristics of enhancers and enable analysis of cell-type specificity.

    CAS  PubMed  Google Scholar 

  175. Gasperini, M., Starita, L. & Shendure, J. The power of multiplexed functional analysis of genetic variants. Nat. Protoc. 11, 1782–1787 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Vockley, C. M. et al. Direct GR binding sites potentiate clusters of TF binding across the human genome. Cell 166, 1269–1281.e19 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Vanhille, L. et al. High-throughput and quantitative assessment of enhancer activity in mammals by CapStarr-seq. Nat. Commun. 6, 6905 (2015).

    CAS  PubMed  Google Scholar 

  178. Klein, J. C., Keith, A., Agarwal, V., Durham, T. & Shendure, J. Functional characterization of enhancer evolution in the primate lineage. Genome Biol. 19, 99 (2018).

    PubMed  PubMed Central  Google Scholar 

  179. Ulirsch, J. C. et al. Systematic functional dissection of common genetic variation affecting red blood cell traits. Cell 165, 1530–1545 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Tewhey, R. et al. Direct identification of hundreds of expression-modulating variants using a multiplexed reporter assay. Cell 172, 1132–1134 (2018).

    CAS  PubMed  Google Scholar 

  181. Vockley, C. M. et al. Massively parallel quantification of the regulatory effects of noncoding genetic variation in a human cohort. Genome Res. 25, 1206–1214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Klein, J. C. et al. Functional testing of thousands of osteoarthritis-associated variants for regulatory activity. Nat. Commun. 10, 2434 (2019).

    PubMed  PubMed Central  Google Scholar 

  183. Liu, Y. et al. Functional assessment of human enhancer activities using whole-genome STARR-sequencing. Genome Biol. 18, 219 (2017).

    PubMed  PubMed Central  Google Scholar 

  184. Melnikov, A. et al. Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay. Nat. Biotechnol. 30, 271 (2012). Along with Patwardhan et al. (2012), this study shows the first application of an MPRA to variants of enhancer sequences, in addition to coining the term ‘massively parallel reporter assay’.

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Kircher, M. et al. Saturation mutagenesis of twenty disease-associated regulatory elements at single base-pair resolution. Nat. Commun. 10, 3583 (2019).

    PubMed  PubMed Central  Google Scholar 

  186. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  188. van Arensbergen, J. et al. Genome-wide mapping of autonomous promoter activity in human cells. Nat. Biotechnol. 35, 145–153 (2017).

    PubMed  Google Scholar 

  189. Gilbert, N. & Allan, J. Supercoiling in DNA and chromatin. Curr. Opin. Genet. Dev. 25, 15–21 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Inoue, F. et al. A systematic comparison reveals substantial differences in chromosomal versus episomal encoding of enhancer activity. Genome Res. 27, 38–52 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  192. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    CAS  PubMed  Google Scholar 

  193. Zhou, Y. et al. High-throughput screening of a CRISPR/Cas9 library for functional genomics in human cells. Nature 509, 487–491 (2014).

    CAS  PubMed  Google Scholar 

  194. Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR–Cas9 system. Science 343, 80–84 (2014).

    CAS  PubMed  Google Scholar 

  195. Canver, M. C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015). This study is the first CRISPR-based screen to identify functional non-coding sequence within an enhancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  196. van Overbeek, M. et al. DNA repair profiling reveals nonrandom outcomes at Cas9-mediated breaks. Mol. Cell 63, 633–646 (2016).

    PubMed  Google Scholar 

  197. Chen, W. et al. Massively parallel profiling and predictive modeling of the outcomes of CRISPR/Cas9-mediated double-strand break repair. Nucleic Acids Res. 47, 7989– 8003 (2019).

    PubMed  PubMed Central  Google Scholar 

  198. Vierstra, J. et al. Functional footprinting of regulatory DNA. Nat. Methods 12, 927–930 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Wright, J. B. & Sanjana, N. E. CRISPR screens to discover functional noncoding elements. Trends Genet. 32, 526–529 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Korkmaz, G. et al. Functional genetic screens for enhancer elements in the human genome using CRISPR–Cas9. Nat. Biotechnol. 34, 192–198 (2016).

    CAS  PubMed  Google Scholar 

  201. Rajagopal, N. et al. High-throughput mapping of regulatory DNA. Nat. Biotechnol. 34, 167–174 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Sanjana, N. E. et al. High-resolution interrogation of functional elements in the noncoding genome. Science 353, 1545–1549 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Diao, Y. et al. A tiling-deletion-based genetic screen for cis-regulatory element identification in mammalian cells. Nat. Methods 14, 629–635 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Gasperini, M. et al. CRISPR/Cas9-mediated scanning for regulatory elements required for HPRT1 expression via thousands of large, programmed genomic deletions. Am. J. Hum. Genet. 101, 192–205 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Aparicio-Prat, E. et al. DECKO: Single-oligo, dual-CRISPR deletion of genomic elements including long non-coding RNAs. BMC Genomics 16, 846 (2015).

    PubMed  PubMed Central  Google Scholar 

  206. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765–771 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Thakore, P. I. et al. Highly specific epigenome editing by CRISPR–Cas9 repressors for silencing of distal regulatory elements. Nat. Methods 12, 1143–1149 (2015). This study provides the first demonstration that nuclease-inactivated Cas9 tethered to the KRAB repressor and targeted to an enhancer can mediate repression of a target gene.

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Klann, T. S. et al. CRISPR–Cas9 epigenome editing enables high-throughput screening for functional regulatory elements in the human genome. Nat. Biotechnol. 35, 561 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Fulco, C. P. et al. Activity-by-contact model of enhancer–promoter regulation from thousands of CRISPR perturbations. Nat. Genet. 51, 1664–1669 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Kearns, N. A. et al. Functional annotation of native enhancers with a Cas9-histone demethylase fusion. Nat. Methods 12, 401–403 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Kwon, D. Y., Zhao, Y.-T., Lamonica, J. M. & Zhou, Z. Locus-specific histone deacetylation using a synthetic CRISPR–Cas9-based HDAC. Nat. Commun. 8, 15315 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Lei, Y. et al. Targeted DNA methylation in vivo using an engineered dCas9–MQ1 fusion protein. Nat. Commun. 8, 16026 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Vojta, A. et al. Repurposing the CRISPR–Cas9 system for targeted DNA methylation. Nucleic Acids Res. 44, 5615–5628 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247.e17 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Huang, Y.-H. et al. DNA epigenome editing using CRISPR–cas suntag-directed DNMT3A. Genome Biol. 18, 176 (2017).

    PubMed  PubMed Central  Google Scholar 

  216. Xie, S., Duan, J., Li, B., Zhou, P. & Hon, G. C. Multiplexed engineering and analysis of combinatorial enhancer activity in single cells. Mol. Cell 66, 285–299.e5 (2017). This study reports the first use of single-cell RNA-seq to phenotype pools of candidate enhancer perturbations in a ‘whole-transcriptome’ screen.

    CAS  PubMed  Google Scholar 

  217. Hong, J.-W., Hendrix, D. A. & Levine, M. S. Shadow enhancers as a source of evolutionary novelty. Science 321, 1314 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

    PubMed  PubMed Central  Google Scholar 

  219. 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).

    CAS  PubMed  Google Scholar 

  220. Lupiáñez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene–enhancer interactions. Cell 161, 1012–1025 (2015). This article demonstrates that rearrangement across TAD boundaries can cause a pathogenic phenotype in humans; the study also uses CRISPR–Cas9 to effectively reconstruct human patient genome rearrangements in mouse models.

    PubMed  PubMed Central  Google Scholar 

  221. Dickel, D. E. et al. Ultraconserved enhancers are required for normal development. Cell 172, 491–499.e15 (2018). This report demonstrates that the knockout of ultraconserved enhancers in a mouse model had subtle but consequential effects on organismal development that were not readily detected by gross phenotyping.

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Zeng, W., Wu, M. & Jiang, R. Prediction of enhancer–promoter interactions via natural language processing. BMC Genomics 19, 84 (2018).

    PubMed  PubMed Central  Google Scholar 

  223. Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Gill, L. L., Karjalainen, K. & Zaninetta, D. A transcriptional enhancer of the mouse T cell receptor δ gene locus. Eur. J. Immunol. 21, 807–810 (1991).

    CAS  PubMed  Google Scholar 

  225. Greaves, D. R., Wilson, F. D., Lang, G. & Kioussis, D. Human CD2 3ʹ-flanking sequences confer high-level, T cell-specific, position-independent gene expression in transgenic mice. Cell 56, 979–986 (1989).

    CAS  PubMed  Google Scholar 

  226. Raab, J. R. & Kamakaka, R. T. Insulators and promoters: closer than we think. Nat. Rev. Genet. 11, 439–446 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  227. Mikhaylichenko, O. et al. The degree of enhancer or promoter activity is reflected by the levels and directionality of eRNA transcription. Genes. Dev. 32, 42–57 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Ngoc, L. V., Wang, Y.-L., Kassavetis, G. A. & Kadonaga, J. T. The punctilious RNA polymerase II core promoter. Genes Dev. 31, 1289–1301 (2017).

    CAS  Google Scholar 

  230. Jayavelu, N. D., Jajodia, A., Mishra, A. & Hawkins, R. An atlas of silencer elements for the human and mouse genomes. Preprint at bioRxiv https://doi.org/10.1101/252304 (2018).

    Article  Google Scholar 

  231. West, A. G., Gaszner, M. & Felsenfeld, G. Insulators: many functions, many mechanisms. Genes. Dev. 16, 271–288 (2002).

    PubMed  Google Scholar 

  232. Stranger, B. E. et al. Patterns of cis regulatory variation in diverse human populations. PLoS Genet. 8, e1002639 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  233. Pollard, K. S., Hubisz, M. J., Rosenbloom, K. R. & Siepel, A. Detection of nonneutral substitution rates on mammalian phylogenies. Genome Res. 20, 110–121 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  234. Siepel, A. et al. Evolutionarily conserved elements in vertebrate, insect, worm, and yeast genomes. Genome Res. 15, 1034–1050 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  235. Khan, A. et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46, D1284 (2018).

    PubMed  Google Scholar 

  236. Kulakovskiy, I. V. et al. HOCOMOCO: towards a complete collection of transcription factor binding models for human and mouse via large-scale ChIP-Seq analysis. Nucleic Acids Res. 46, D252–D259 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Mahat, D. B. et al. Base-pair-resolution genome-wide mapping of active RNA polymerases using precision nuclear run-on (PRO-seq). Nat. Protoc. 11, 1455–1476 (2016).

    PubMed  PubMed Central  Google Scholar 

  240. Tome, J. M., Tippens, N. D. & Lis, J. T. Single-molecule nascent RNA sequencing identifies regulatory domain architecture at promoters and enhancers. Nat. Genet. 50, 1533–1541 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Ku, W. L. et al. Single-cell chromatin immunocleavage sequencing (scChIC-seq) to profile histone modification. Nat. Methods 16, 323–325 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  242. Skene, P. J. & Henikoff, S. An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. eLife 6, e21856 (2017).

    PubMed  PubMed Central  Google Scholar 

  243. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012). This study reports a computational framework for utilizing ChIP–seq of histone modifications to classify regions of the genome by their likely biological function (for example, strong enhancer or weak promoter).

    CAS  PubMed  PubMed Central  Google Scholar 

  244. Hoffman, M. M. et al. Unsupervised pattern discovery in human chromatin structure through genomic segmentation. Nat. Methods 9, 473–476 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank S. Kim, C. Trapnell and S. Domcke, as well as other members of the Shendure Lab, for helpful discussions. J.S. is an investigator for the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

M.G. and J.S. wrote the initial manuscript. M.G., J.T. and J.S. contributed to researching content for the article, discussing the content and reviewing/editing the manuscript before submission.

Corresponding author

Correspondence to Jay Shendure.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Glossary

Transcription factors

(TFs). Proteins that bind DNA, typically consisting of specific DNA sequences or motifs, and contribute to the regulation of RNA transcription.

Open chromatin

A nucleosome-loose packaging state of DNA that is permissive for transcription-factor binding.

Episomal reporter vector

Plasmid DNA that can be synthetically delivered, is autonomous from genomic DNA and includes a reporter gene, typically downstream of a candidate regulatory element (for example, an enhancer adjacent to a minimal promoter).

Expressed sequence tag

(EST). In the early days of genomics, shotgun sequencing of cDNA was used as an efficient strategy for discovering genes, and subsequently to quantify their relative abundance.

Open reading frame

The portion of a gene that is translatable by a ribosome; these are relatively straightforward to annotate by sequence alone, due to the required start and stop codons.

Regulatory element

A functional non-coding DNA sequence that regulates transcription; classes of regulatory elements include enhancers, promoters, silencers and insulators (further defined in Box 2).

Chromosome conformation capture

(3C). Methods that map the 3D positioning, looping and spatial organization of DNA within the nucleus, often relative to other segments of DNA.

CRISPR

Clustered regularly interspaced short palindromic repeats. A system that consists of the components of a bacterial immune system that have been adopted for synthetic genetic perturbation. The term is most often used in reference to the Type II Cas9 endonuclease version, which can introduce a double-stranded break into genomic DNA as directed by a synthetic guide RNA.

Topologically associating domains

(TADs). Broad regions of genomic DNA that are physically packaged together in the nucleus in 3D space, typically at a scale from hundreds of kilobases to several megabases.

Pioneer factor

A TF that can directly interact with compact, closed chromatin; this class of TFs are thought to initiate (‘pioneer’) chromatin remodelling events.

Linkage disequilibrium

The population genetics phenomenon by which genetic variants are nonrandomly associated within a population. Variants are said to be in ‘linkage disequilibrium’ if they are found to reside on a haplotype more frequently than one would expect by completely random assortment; variants in linkage disequilibrium are nearby on a genomic locus and hence are co-inherited because they are rarely separated through meiotic recombination.

Simpson’s paradox

A phenomenon in statistics in which different trends may exist in subgroups of a dataset but are undetectable when the groups are analysed as a whole.

Saturation mutagenesis

A molecular biology technique in which all possible sequence changes are generated from a parental sequence (for example, all possible amino acids in an open reading frame, or all possible single-nucleotide variants in an enhancer).

Protospacer-adjacent motif

(PAM). In the original CRISPR bacterial immune system, fragments of previously encountered viral DNA are preserved in the bacterial genome; these ‘remembered’ sequences are processed into RNAs that guide the CRISPR nuclease to destroy newly invading viral DNA. But, to prevent the nuclease from destroying the matching ‘remembered’ sequence in the bacteria’s own genome, a motif (the PAM) is required next to the target sequence in the viral genome. When genome editing is performed in eukaryotic cells, the presence of this sequence is still required by CRISPR nucleases.

Shadow enhancers

Redundant enhancers, often located far away from their target gene; enhancer redundancy is thought to enable robust buffered expression of the target gene and to provide a versatile platform for the evolution of new regulatory functions.

ENCODE-4

The fourth generation of projects funded by the Encyclopedia of DNA Elements (ENCODE) Consortium, begun in 2017 and including a new component focused on the implementation of high-throughput functional assays.

Human Cell Atlas

An international scientific community to coordinate the generation of human single-cell datasets, with the goal of generating a reference map of every cell type in the human body.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gasperini, M., Tome, J.M. & Shendure, J. Towards a comprehensive catalogue of validated and target-linked human enhancers. Nat Rev Genet 21, 292–310 (2020). https://doi.org/10.1038/s41576-019-0209-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-019-0209-0

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