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:

Characterization of noncoding regulatory DNA in the human genome

Abstract

Genetic variants associated with common diseases are usually located in noncoding parts of the human genome. Delineation of the full repertoire of functional noncoding elements, together with efficient methods for probing their biological roles, is therefore of crucial importance. Over the past decade, DNA accessibility and various epigenetic modifications have been associated with regulatory functions. Mapping these features across the genome has enabled researchers to begin to document the full complement of putative regulatory elements. High-throughput reporter assays to probe the functions of regulatory regions have also been developed but these methods separate putative regulatory elements from the chromosome so that any effects of chromatin context and long-range regulatory interactions are lost. Definitive assignment of function(s) to putative cis-regulatory elements requires perturbation of these elements. Genome-editing technologies are now transforming our ability to perturb regulatory elements across entire genomes. Interpretation of high-throughput genetic screens that incorporate genome editors might enable the construction of an unbiased map of functional noncoding elements in the human genome.

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: Genome-wide identification of candidate regulatory regions.

Debbie Maizels/Springer Nature

Figure 2: High-throughput measurements of enhancer activity using exogenous assays.

Debbie Maizels/Springer Nature

Figure 3: Elucidation of functional variants in regulatory elements.
Figure 4: Inference of enhancer-promoter links.

Debbie Maizels/Springer Nature

Figure 5: Mapping physical interactions between putative enhancers and promoters.

Debbie Maizels/Springer Nature

Figure 6: Functional screens for DNA regulatory elements using genome-editing tools.

Debbie Maizels/Springer Nature

Similar content being viewed by others

References

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

  2. Cech, T.R. & Steitz, J.A. The noncoding RNA revolution—trashing old rules to forge new ones. Cell 157, 77–94 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Deniz, E. & Erman, B. Long noncoding RNA (lincRNA), a new paradigm in gene expression control. Funct. Integr. Genomics 17, 135–143 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Deplancke, B., Alpern, D. & Gardeux, V. The genetics of transcription factor DNA binding variation. Cell 166, 538–554 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Manolio, T.A., Brooks, L.D. & Collins, F.S. A HapMap harvest of insights into the genetics of common disease. J. Clin. Invest. 118, 1590–1605 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Welter, D. et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 42, D1001–D1006 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Huang, Q. Genetic study of complex diseases in the post-GWAS era. J. Genet. Genomics 42, 87–98 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. 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 

  9. Turner, B.M. Defining an epigenetic code. Nat. Cell Biol. 9, 2–6 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Song, L. & Crawford, G.E. DNase-seq: a high-resolution technique for mapping active gene regulatory elements across the genome from mammalian cells. Cold Spring Harb. Protoc. 2010, http://dx.doi.org/doi:1101/pdb.prot5384 (2010).

  11. Simon, J.M., Giresi, P.G., Davis, I.J. & Lieb, J.D. Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat. Protoc. 7, 256–267 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wang, Z. et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat. Genet. 40, 897–903 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Cheng, C. et al. Understanding transcriptional regulation by integrative analysis of transcription factor binding data. Genome Res. 22, 1658–1667 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hoffman, M.M. et al. Integrative annotation of chromatin elements from ENCODE data. Nucleic Acids Res. 41, 827–841 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Romanoski, C.E., Glass, C.K., Stunnenberg, H.G., Wilson, L. & Almouzni, G. Epigenomics: roadmap for regulation. Nature 518, 314–316 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Fisher, W.W. et al. DNA regions bound at low occupancy by transcription factors do not drive patterned reporter gene expression in Drosophila. Proc. Natl. Acad. Sci. USA 109, 21330–21335 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. Lizio, M. et al. Mapping mammalian cell-type-specific transcriptional regulatory networks using KD-CAGE and ChIP-seq data in the TC-YIK cell line. Front. Genet. 6, 331 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  37. 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 

  38. Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Léveillé, N. et al. Genome-wide profiling of p53-regulated enhancer RNAs uncovers a subset of enhancers controlled by a lncRNA. Nat. Commun. 6, 6520 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Wu, H. et al. Tissue-specific RNA expression marks distant-acting developmental enhancers. PLoS Genet. 10, e1004610 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shiraki, T. et al. Cap analysis gene expression for high-throughput analysis of transcriptional starting point and identification of promoter usage. Proc. Natl. Acad. Sci. USA 100, 15776–15781 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hah, N., Murakami, S., Nagari, A., Danko, C.G. & Kraus, W.L. Enhancer transcripts mark active estrogen receptor binding sites. Genome Res. 23, 1210–1223 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Melo, C.A. et al. eRNAs are required for p53-dependent enhancer activity and gene transcription. Mol. Cell 49, 524–535 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455–461 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, D. et al. Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature 474, 390–394 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Koch, F. & Andrau, J.C. Initiating RNA polymerase II and TIPs as hallmarks of enhancer activity and tissue-specificity. Transcription 2, 263–268 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Koch, F. et al. Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat. Struct. Mol. Biol. 18, 956–963 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Melo, C.A., Léveillé, N. & Agami, R. eRNAs reach the heart of transcription. Cell Res. 23, 1151–1152 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. Muerdter, F., Bory´n, L.M. & Arnold, C.D. STARR-seq—principles and applications. Genomics 106, 145–150 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kheradpour, P. et al. Systematic dissection of regulatory motifs in 2000 predicted human enhancers using a massively parallel reporter assay. Genome Res. 23, 800–811 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Birnbaum, R.Y. et al. Systematic dissection of coding exons at single nucleotide resolution supports an additional role in cell-specific transcriptional regulation. PLoS Genet. 10, e1004592 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Patwardhan, R.P. et al. Massively parallel functional dissection of mammalian enhancers in vivo. Nat. Biotechnol. 30, 265–270 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. White, M.A., Myers, C.A., Corbo, J.C. & Cohen, B.A. Massively parallel in vivo enhancer assay reveals that highly local features determine the cis-regulatory function of ChIP-seq peaks. Proc. Natl. Acad. Sci. USA 110, 11952–11957 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  57. Patwardhan, R.P. et al. High-resolution analysis of DNA regulatory elements by synthetic saturation mutagenesis. Nat. Biotechnol. 27, 1173–1175 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kinney, J.B., Murugan, A., Callan, C.G. Jr. & Cox, E.C. Using deep sequencing to characterize the biophysical mechanism of a transcriptional regulatory sequence. Proc. Natl. Acad. Sci. USA 107, 9158–9163 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Shen, S.Q. et al. Massively parallel cis-regulatory analysis in the mammalian central nervous system. Genome Res. 26, 238–255 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. International HapMap Consortium. A haplotype map of the human genome. Nature 437, 1299–1320 (2005).

  64. Manolio, T.A. Bringing genome-wide association findings into clinical use. Nat. Rev. Genet. 14, 549–558 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. McCarthy, M.I. et al. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat. Rev. Genet. 9, 356–369 (2008).

    Article  CAS  PubMed  Google Scholar 

  66. Schaub, M.A., Boyle, A.P., Kundaje, A., Batzoglou, S. & Snyder, M. Linking disease associations with regulatory information in the human genome. Genome Res. 22, 1748–1759 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cowper-Sallari, R. et al. Breast cancer risk-associated SNPs modulate the affinity of chromatin for FOXA1 and alter gene expression. Nat. Genet. 44, 1191–1198 (2012).

    Article  CAS  Google Scholar 

  68. Karczewski, K.J. et al. Systematic functional regulatory assessment of disease-associated variants. Proc. Natl. Acad. Sci. USA 110, 9607–9612 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Whitington, T. et al. Gene regulatory mechanisms underpinning prostate cancer susceptibility. Nat. Genet. 48, 387–397 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Degner, J.F. et al. DNase I sensitivity QTLs are a major determinant of human expression variation. Nature 482, 390–394 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. McVicker, G. et al. Identification of genetic variants that affect histone modifications in human cells. Science 342, 747–749 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Tehranchi, A.K. et al. Pooled ChIP-seq links variation in transcription factor binding to complex disease risk. Cell 165, 730–741 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Musunuru, K. et al. From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature 466, 714–719 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Smemo, S. et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507, 371–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Farh, K.K. et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337–343 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Grubert, F. et al. Genetic control of chromatin states in humans involves local and distal chromosomal interactions. Cell 162, 1051–1065 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kasowski, M. et al. Variation in transcription factor binding among humans. Science 328, 232–235 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. McDaniell, R. et al. Heritable individual-specific and allele-specific chromatin signatures in humans. Science 328, 235–239 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Reddy, T.E. et al. Effects of sequence variation on differential allelic transcription factor occupancy and gene expression. Genome Res. 22, 860–869 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Wu, T.D. & Nacu, S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics 26, 873–881 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Rozowsky, J. et al. AlleleSeq: analysis of allele-specific expression and binding in a network framework. Mol. Syst. Biol. 7, 522 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. van de Geijn, B., McVicker, G., Gilad, Y. & Pritchard, J.K. WASP: allele-specific software for robust molecular quantitative trait locus discovery. Nat. Methods 12, 1061–1063 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Romanel, A., Lago, S., Prandi, D., Sboner, A. & Demichelis, F. ASEQ: fast allele-specific studies from next-generation sequencing data. BMC Med. Genomics 8, 9 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Harvey, C.T. et al. QuASAR: quantitative allele-specific analysis of reads. Bioinformatics 31, 1235–1242 (2015).

    Article  PubMed  Google Scholar 

  85. Gilad, Y., Rifkin, S.A. & Pritchard, J.K. Revealing the architecture of gene regulation: the promise of eQTL studies. Trends Genet. 24, 408–415 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Nica, A.C. et al. Candidate causal regulatory effects by integration of expression QTLs with complex trait genetic associations. PLoS Genet. 6, e1000895 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Nicolae, D.L. et al. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 6, e1000888 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Bahcall, O.G. Human genetics: GTEx pilot quantifies eQTL variation across tissues and individuals. Nat. Rev. Genet. 16, 375 (2015).

    Article  CAS  PubMed  Google Scholar 

  89. GTEx Consortium. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  90. Fairfax, B.P. et al. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Parker, S.C. et al. Chromatin stretch enhancer states drive cell-specific gene regulation and harbor human disease risk variants. Proc. Natl. Acad. Sci. USA 110, 17921–17926 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Kilpinen, H. et al. Coordinated effects of sequence variation on DNA binding, chromatin structure, and transcription. Science 342, 744–747 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Skelly, D.A., Johansson, M., Madeoy, J., Wakefield, J. & Akey, J.M. A powerful and flexible statistical framework for testing hypotheses of allele-specific gene expression from RNA-seq data. Genome Res. 21, 1728–1737 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Pickrell, J.K. et al. Understanding mechanisms underlying human gene expression variation with RNA sequencing. Nature 464, 768–772 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Montgomery, S.B. et al. Transcriptome genetics using second generation sequencing in a Caucasian population. Nature 464, 773–777 (2010).

    Article  CAS  PubMed  Google Scholar 

  96. de Wit, E. & de Laat, W. A decade of 3C technologies: insights into nuclear organization. Genes Dev. 26, 11–24 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Dekker, J., Marti-Renom, M.A. & Mirny, L.A. Exploring the three-dimensional organization of genomes: interpreting chromatin interaction data. Nat. Rev. Genet. 14, 390–403 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. 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 

  102. Jin, F. et al. A high-resolution map of the three-dimensional chromatin interactome in human cells. Nature 503, 290–294 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Tang, Z. et al. CTCF-Mediated Human 3D Genome architecture reveals chromatin topology for transcription. Cell 163, 1611–1627 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Lupiáñez, D.G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161, 1012–1025 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science 351, 1454–1458 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Katainen, R. et al. CTCF/cohesin-binding sites are frequently mutated in cancer. Nat. Genet. 47, 818–821 (2015).

    Article  CAS  PubMed  Google Scholar 

  107. Flavahan, W.A. et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529, 110–114 (2016).

    Article  CAS  PubMed  Google Scholar 

  108. Grimmer, M.R. & Costello, J.F. Cancer: oncogene brought into the loop. Nature 529, 34–35 (2016).

    Article  CAS  PubMed  Google Scholar 

  109. Dryden, N.H. et al. Unbiased analysis of potential targets of breast cancer susceptibility loci by Capture Hi-C. Genome Res. 24, 1854–1868 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Mifsud, B. et al. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C. Nat. Genet. 47, 598–606 (2015).

    Article  CAS  PubMed  Google Scholar 

  111. Fullwood, M.J. et al. An oestrogen-receptor-alpha-bound human chromatin interactome. Nature 462, 58–64 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. 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 

  113. 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 

  114. 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 

  115. Mahfouz, M.M., Piatek, A. & Stewart, C.N. Jr. Genome engineering via TALENs and CRISPR/Cas9 systems: challenges and perspectives. Plant Biotechnol. J. 12, 1006–1014 (2014).

    Article  CAS  PubMed  Google Scholar 

  116. Gaj, T., Gersbach, C.A. & Barbas, C.F., III. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31, 397–405 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hsu, P.D., Lander, E.S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sander, J.D. & Joung, J.K. CRISPR–Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347–355 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Kim, H. & Kim, J.S. A guide to genome engineering with programmable nucleases. Nat. Rev. Genet. 15, 321–334 (2014).

    Article  CAS  PubMed  Google Scholar 

  120. Wei, C. et al. TALEN or Cas9—rapid, efficient and specific choices for genome modifications. J. Genet. Genomics 40, 281–289 (2013).

    Article  CAS  PubMed  Google Scholar 

  121. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009).

    Article  CAS  PubMed  Google Scholar 

  122. Moscou, M.J. & Bogdanove, A.J. A simple cipher governs DNA recognition by TAL effectors. Science 326, 1501 (2009).

    Article  CAS  PubMed  Google Scholar 

  123. Spisák, S. et al. CAUSEL: an epigenome- and genome-editing pipeline for establishing function of noncoding GWAS variants. Nat. Med. 21, 1357–1363 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Soldner, F. et al. Parkinson-associated risk variant in distal enhancer of a-synuclein modulates target gene expression. Nature 533, 95–99 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  127. Parnas, O. et al. A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell 162, 675–686 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Canver, M.C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  132. Guo, Y. et al. CRISPR inversion of CTCF sites alters genome topology and enhancer/promoter function. Cell 162, 900–910 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Fanucchi, S., Shibayama, Y., Burd, S., Weinberg, M.S. & Mhlanga, M.M. Chromosomal contact permits transcription between coregulated genes. Cell 155, 606–620 (2013).

    Article  CAS  PubMed  Google Scholar 

  134. Dominguez, A.A., Lim, W.A. & Qi, L.S. Beyond editing: repurposing CRISPR-Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell Biol. 17, 5–15 (2016).

    Article  CAS  PubMed  Google Scholar 

  135. Kungulovski, G. & Jeltsch, A. Epigenome editing: state of the art, concepts, and perspectives. Trends Genet. 32, 101–113 (2016).

    Article  CAS  PubMed  Google Scholar 

  136. 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 

  137. Maeder, M.L. et al. Targeted DNA demethylation and activation of endogenous genes using programmable TALE-TET1 fusion proteins. Nat. Biotechnol. 31, 1137–1142 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Perez-Pinera, P. et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat. Methods 10, 973–976 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Maeder, M.L. et al. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 10, 977–979 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

  141. Tanenbaum, M.E., Gilbert, L.A., Qi, L.S., Weissman, J.S. & Vale, R.D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Gao, X. et al. Comparison of TALE designer transcription factors and the CRISPR/dCas9 in regulation of gene expression by targeting enhancers. Nucleic Acids Res. 42, e155 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gilbert, L.A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Hilton, I.B. et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 33, 510–517 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Hsu, P.D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Tsai, S.Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  PubMed  Google Scholar 

  153. Kleinstiver, B.P. et al. Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol. 33, 1293–1298 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Kleinstiver, B.P. et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523, 481–485 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Haeussler, M. & Concordet, J.P. Genome editing with CRISPR-Cas9: can it get any better? J. Genet. Genomics 43, 239–250 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Kim, D. et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat. Methods 12, 237–243 (2015).

    Article  CAS  PubMed  Google Scholar 

  157. Plank, J.L. & Dean, A. Enhancer function: mechanistic and genome-wide insights come together. Mol. Cell. 55, 5–14 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Boyle, A.P. et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 22, 1790–1797 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Li, G. et al. ChIA-PET tool for comprehensive chromatin interaction analysis with paired-end tag sequencing. Genome Biol. 11, R22 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Crawford, G.E. et al. Genome-wide mapping of DNase hypersensitive sites using massively parallel signature sequencing (MPSS). Genome Res. 16, 123–131 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. 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 

  162. 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 

  163. Li, W., Notani, D. & Rosenfeld, M.G. Enhancers as noncoding RNA transcription units: recent insights and future perspectives. Nat. Rev. Genet. 17, 207–223 (2016).

    Article  CAS  PubMed  Google Scholar 

  164. Danko, C.G. et al. Identification of active transcriptional regulatory elements from GRO-seq data. Nat. Methods 12, 433–438 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Zhang, F. et al. Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat. Biotechnol. 29, 149–153 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, e82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Miller, J.C. et al. A TALE nuclease architecture for efficient genome editing. Nat. Biotechnol. 29, 143–148 (2011).

    Article  CAS  PubMed  Google Scholar 

  168. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

R.E. is a Faculty Fellow of the Edmond J. Safra Center for Bioinformatics at Tel Aviv University. This work was supported by funds from the EEF-CEC/EU ERC-AdV and NWO (NGI 93512001/2012) programs to R.A. and from the Israel Cancer Association to R.E.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Ran Elkon or Reuven Agami.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elkon, R., Agami, R. Characterization of noncoding regulatory DNA in the human genome. Nat Biotechnol 35, 732–746 (2017). https://doi.org/10.1038/nbt.3863

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3863

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