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:

Regulation of disease-associated gene expression in the 3D genome

Key Points

  • The human genome harbours a dizzying array of regulatory sequences, such as enhancers with a priori unpredictable, promiscuous and context-dependent behaviour.

  • More than 90% of disease-associated variants lie in non-coding DNA, accumulating in presumptive enhancers. Therefore, a picture is emerging in which diseases and traits are often the consequence of erroneous wiring of regulatory circuitry between enhancers and their target genes.

  • Enhancers communicate with distant target genes through chromatin loops. Such transcription regulatory loops are formed within topologically associated domains (TADs), the structural and functional units of the chromosomes.

  • Disrupting enhancer–promoter loops, as well as TAD boundaries, can lead to gene dysregulation and disease, the latter due to previously insulated genes and enhancers being released to form new regulatory chromatin loops.

  • Technological advances and the availability of increasingly detailed genomic contact maps generated by Hi-C are enabling the elucidation of the physical contacts between genes and non-coding sequences in different cell types, which greatly facilitates the assigning of function to disease-associated genetic variation.

  • We present a roadmap — from disease-associated genetic variants to molecular mechanisms causing disease — that proposes to integrate epigenetic data with detailed 3D genome information and haplotype-resolved expression analysis to help move results descriptive genetic association studies towards the discovery of the molecular mechanisms underlying disease.

Abstract

Genetic variation associated with disease often appears in non-coding parts of the genome. Understanding the mechanisms by which this phenomenon leads to disease is necessary to translate results from genetic association studies to the clinic. Assigning function to this type of variation is notoriously difficult because the human genome harbours a complex regulatory landscape with a dizzying array of transcriptional regulatory sequences, such as enhancers that have unpredictable, promiscuous and context-dependent behaviour. In this Review, we discuss how technological advances have provided increasingly detailed information on genome folding; for example, genome folding forms loops that bring enhancers and target genes into close proximity. We also now know that enhancers function within topologically associated domains, which are structural and functional units of chromosomes. Studying disease-associated mutations and chromosomal rearrangements in the context of the 3D genome will enable the identification of dysregulated target genes and aid the progression from descriptive genetic association results to discovering molecular mechanisms underlying disease.

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: Mechanism of gene expression regulation by enhancers.
Figure 2: TADs host de novo and pre-formed regulatory enhancer–promoter loops.
Figure 3: Erroneous regulatory wiring between enhancers and target genes causing disease.
Figure 4: Regulatory detours between TADs.
Figure 5: Disease variants associated with disruptions in TADs and the 3D regulatory landscape.

Similar content being viewed by others

References

  1. Pauling, L. et al. Sickle cell anemia a molecular disease. Science 110, 543–548 (1949).

    Article  CAS  PubMed  Google Scholar 

  2. Ingram, V. M. Gene mutations in human haemoglobin: the chemical difference between normal and sickle cell haemoglobin. Nature 180, 326–328 (1957).

    Article  CAS  PubMed  Google Scholar 

  3. Fritsch, E. F., Lawn, R. M. & Maniatis, T. Characterisation of deletions which affect the expression of fetal globin genes in man. Nature 279, 598–603 (1979).

    Article  CAS  PubMed  Google Scholar 

  4. Van der Ploeg, L. H. 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).

    Article  CAS  PubMed  Google Scholar 

  5. Banerji, J., Rusconi, S. & Schaffner, W. Expression of a β-globin gene is enhanced by remote SV40 DNA sequences. Cell 27, 299–308 (1981).

    Article  CAS  PubMed  Google Scholar 

  6. Benoist, C. & Chambon, P. In vivo sequence requirements of the SV40 early promotor region. Nature 290, 304–310 (1981).

    Article  CAS  PubMed  Google Scholar 

  7. 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).

    Article  CAS  PubMed  Google Scholar 

  8. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  10. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Venter, J. C. et al. The sequence of the human genome. Science 291, 1304–1351 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. 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). References 12 and 13 reveal that most of the disease-associated variations are located in regulatory sequences.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Ingram, V. M. Sickle-cell anemia hemoglobin: the molecular biology of the first “molecular disease” — the crucial importance of serendipity. Genetics 167, 1–7 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  15. 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 

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

  17. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Abecasis, G. R. et al. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  21. Fromm, M. & Berg, P. Simian virus 40 early- and late-region promoter functions are enhanced by the 72-base-pair repeat inserted at distant locations and inverted orientations. Mol. Cell. Biol. 3, 991–999 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  23. 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 

  24. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Allen, B. L. & Taatjes, D. J. The Mediator complex: a central integrator of transcription. Nat. Rev. Mol. Cell Biol. 16, 155–166 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Grosveld, F., van Assendelft, G. B., Greaves, D. R. & Kollias, G. Position-independent, high-level expression of the human beta-globin gene in transgenic mice. Cell 51, 975–985 (1987).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  29. Kellis, M. et al. Defining functional DNA elements in the human genome. Proc. Natl Acad. Sci. USA 111, 6131–6138 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Shin, H. Y. et al. Hierarchy within the mammary STAT5-driven Wap super-enhancer. Nat. Genet. 48, 904–911 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hnisz, D. et al. Convergence of developmental and oncogenic signaling pathways at transcriptional super-enhancers. Mol. Cell 58, 362–370 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bender, M. A. et al. Targeted deletion of 5′HS1 and 5′HS4 of the β-globin locus control region reveals additive activity of the DNaseI hypersensitive sites. Blood 98, 2022–2027 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. 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 

  35. 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).

    Article  CAS  PubMed  Google Scholar 

  36. Jeong, Y., El-Jaick, K., Roessler, E., Muenke, M. & Epstein, D. J. A functional screen for sonic hedgehog regulatory elements across a 1 Mb interval identifies long-range ventral forebrain enhancers. Development 133, 761–772 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Sagai, T. et al. A cluster of three long-range enhancers directs regional Shh expression in the epithelial linings. Development 136, 1665–1674 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Uslu, V. V. et al. Long-range enhancers regulating Myc expression are required for normal facial morphogenesis. Nat. Genet. 46, 753–758 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Marinic, M., Aktas, T., Ruf, S. & Spitz, F. An integrated holo-enhancer unit defines tissue and gene specificity of the Fgf8 regulatory landscape. Dev. Cell 24, 530–542 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Montavon, T. et al. A regulatory archipelago controls Hox genes transcription in digits. Cell 147, 1132–1145 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Andrey, G. et al. A switch between topological domains underlies HoxD genes collinearity in mouse limbs. Science 340, 1234167 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Khan, M., Vaes, E. & Mombaerts, P. Regulation of the probability of mouse odorant receptor gene choice. Cell 147, 907–921 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Tasic, B. et al. Promoter choice determines splice site selection in protocadherin α and γ pre-mRNA splicing. Mol. Cell 10, 21–33 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Lower, K. M. et al. Adventitious changes in long-range gene expression caused by polymorphic structural variation and promoter competition. Proc. Natl Acad. Sci. USA 106, 21771–21776 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Dillon, N., Trimborn, T., Strouboulis, J., Fraser, P. & Grosveld, F. The effect of distance on long-range chromatin interactions. Mol. Cell 1, 131–139 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. De Gobbi, M. et al. A regulatory SNP causes a human genetic disease by creating a new transcriptional promoter. Science 312, 1215–1217 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Wijgerde, M., Grosveld, F. & Fraser, P. Transcription complex stability and chromatin dynamics in vivo. Nature 377, 209–213 (1995).

    Article  CAS  PubMed  Google Scholar 

  48. Zabidi, M. A. et al. Enhancer-core-promoter specificity separates developmental and housekeeping gene regulation. Nature 518, 556–559 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. 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 

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

    Article  CAS  PubMed  Google Scholar 

  51. Sagai, T., Hosoya, M., Mizushina, Y., Tamura, M. & Shiroishi, T. Elimination of a long-range cis-regulatory module causes complete loss of limb-specific Shh expression and truncation of the mouse limb. Development 132, 797–803 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Nobrega, M. A., Zhu, Y., Plajzer-Frick, I., Afzal, V. & Rubin, E. M. Megabase deletions of gene deserts result in viable mice. Nature 431, 988–993 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Sturtevant, A. H. The effects of unequal crossing over at the bar locus in Drosophila. Genetics 10, 117–147 (1925).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Ruf, S. et al. Large-scale analysis of the regulatory architecture of the mouse genome with a transposon-associated sensor. Nat. Genet. 43, 379–386 (2011). This study comprehensively illustrates the importance of genomic context for enhancer function.

    Article  CAS  PubMed  Google Scholar 

  55. 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 

  56. Denker, A. & de Laat, W. The second decade of 3C technologies: detailed insights into nuclear organization. Genes Dev. 30, 1357–1382 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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 

  58. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012). References 57 and 58 demonstrate the presence of TADs in the mammalian genome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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 

  60. Bouwman, B. A. & de Laat, W. Getting the genome in shape: the formation of loops, domains and compartments. Genome Biol. 16, 154 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  62. Tolhuis, B., Palstra, R. J., Splinter, E., Grosveld, F. & de Laat, W. Looping and interaction between hypersensitive sites in the active β-globin locus. Mol. Cell 10, 1453–1465 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature 518, 331–336 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Deng, W. et al. Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 149, 1233–1244 (2012). This study provides direct evidence that chromatin loops between enhancers and promoters underlie transcription.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Deng, W. et al. Reactivation of developmentally silenced globin genes by forced chromatin looping. Cell 158, 849–860 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Phillips-Cremins, J. E. et al. Architectural protein subclasses shape 3D organization of genomes during lineage commitment. Cell 153, 1281–1295 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Palstra, R. J. et al. The β-globin nuclear compartment in development and erythroid differentiation. Nat. Genet. 35, 190–194 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Krijger, P. H. et al. Cell-of-origin-specific 3D genome structure acquired during somatic cell reprogramming. Cell Stem Cell 18, 597–610 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Beagan, J. A. et al. Local genome topology can exhibit an incompletely rewired 3D-folding state during somatic cell reprogramming. Cell Stem Cell 18, 611–624 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  73. Bickmore, W. A. & van Steensel, B. Genome architecture: domain organization of interphase chromosomes. Cell 152, 1270–1284 (2013).

    Article  CAS  PubMed  Google Scholar 

  74. Pope, B. D. et al. Topologically associating domains are stable units of replication-timing regulation. Nature 515, 402–405 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Symmons, O. et al. Functional and topological characteristics of mammalian regulatory domains. Genome Res. 24, 390–400 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Waszak, S. M. et al. Population variation and genetic control of modular chromatin architecture in humans. Cell 162, 1039–1050 (2015).

    Article  CAS  PubMed  Google Scholar 

  77. 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 

  78. Denker, A. & de Laat, W. A. Long-distance chromatin affair. Cell 162, 942–943 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Heath, H. et al. CTCF regulates cell cycle progression of αβ T cells in the thymus. EMBO J. 27, 2839–2850 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Chen, H., Tian, Y., Shu, W., Bo, X. & Wang, S. Comprehensive identification and annotation of cell type-specific and ubiquitous CTCF-binding sites in the human genome. PLoS ONE 7, e41374 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Ong, C. T. & Corces, V. G. CTCF: an architectural protein bridging genome topology and function. Nat. Rev. Genetics 15, 234–246 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Splinter, E. et al. CTCF mediates long-range chromatin looping and local histone modification in the beta-globin locus. Genes Dev. 20, 2349–2354 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Handoko, L. et al. CTCF-mediated functional chromatin interactome in pluripotent cells. Nat. Genet. 43, 630–638 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Yusufzai, T. M., Tagami, H., Nakatani, Y. & Felsenfeld, G. CTCF tethers an insulator to subnuclear sites, suggesting shared insulator mechanisms across species. Mol. Cell 13, 291–298 (2004).

    Article  CAS  PubMed  Google Scholar 

  85. Hadjur, S. et al. Cohesins form chromosomal cis-interactions at the developmentally regulated IFNG locus. Nature 460, 410–413 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Parelho, V. et al. Cohesins functionally associate with CTCF on mammalian chromosome arms. Cell 132, 422–433 (2008).

    Article  CAS  PubMed  Google Scholar 

  88. Wendt, K. S. et al. Cohesin mediates transcriptional insulation by CCCTC-binding factor. Nature 451, 796–801 (2008).

    Article  CAS  PubMed  Google Scholar 

  89. Watrin, E., Kaiser, F. J. & Wendt, K. S. Gene regulation and chromatin organization: relevance of cohesin mutations to human disease. Curr. Opin. Genet. Dev. 37, 59–66 (2016).

    Article  CAS  PubMed  Google Scholar 

  90. Vietri Rudan, M. et al. Comparative Hi-C reveals that CTCF underlies evolution of chromosomal domain architecture. Cell Rep. 10, 1297–1309 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. de Wit, E. et al. CTCF binding polarity determines chromatin looping. Mol. Cell 60, 676–684 (2015).

    Article  CAS  PubMed  Google Scholar 

  92. 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 

  93. Sanborn, A. L. et al. Chromatin extrusion explains key features of loop and domain formation in wild-type and engineered genomes. Proc. Natl Acad. Sci. USA 112, E6456–E6465 (2015). References 59, 91–93 show that TADs are formed between convergent CTCF sites.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Gomez-Marin, C. et al. Evolutionary comparison reveals that diverging CTCF sites are signatures of ancestral topological associating domains borders. Proc. Natl Acad. Sci. USA 112, 7542–7547 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Alipour, E. & Marko, J. F. Self-organization of domain structures by DNA-loop-extruding enzymes. Nucleic Acids Res. 40, 11202–11212 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Fudenberg, G. et al. Formation of chromosomal domains by loop extrusion. Cell Rep. 15, 2038–2049 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Merkenschlager, M. & Nora, E. P. CTCF and cohesin in genome folding and transcriptional gene regulation. Annu. Rev. Genomics Hum. Genet. 31, 17–43 (2016).

    Article  CAS  Google Scholar 

  98. Narendra, V. et al. CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation. Science 347, 1017–1021 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Dowen, J. M. et al. Control of cell identity genes occurs in insulated neighborhoods in mammalian chromosomes. Cell 159, 374–387 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Ji, X. et al. 3D chromosome regulatory landscape of human pluripotent cells. Cell Stem Cell 18, 262–275 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kind, J. & van Steensel, B. Genome-nuclear lamina interactions and gene regulation. Curr. Opin. Cell Biol. 22, 320–325 (2010).

    Article  CAS  PubMed  Google Scholar 

  103. de Wit, E. et al. The pluripotent genome in three dimensions is shaped around pluripotency factors. Nature 501, 227–231 (2013).

    Article  CAS  PubMed  Google Scholar 

  104. Bantignies, F. et al. Polycomb-dependent regulatory contacts between distant Hox loci in Drosophila. Cell 144, 214–226 (2011).

    Article  CAS  PubMed  Google Scholar 

  105. Denholtz, M. et al. Long-range chromatin contacts in embryonic stem cells reveal a role for pluripotency factors and polycomb proteins in genome organization. Cell Stem Cell 13, 602–616 (2013).

    Article  CAS  PubMed  Google Scholar 

  106. Wijchers, P. J. et al. Cause and consequence of tethering a SubTAD to different nuclear compartments. Mol. Cell 61, 461–473 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Krijger, P. H. & de Laat, W. Identical cells with different 3D genomes; cause and consequences? Curr. Opin. Genet. Dev. 23, 191–196 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  109. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Therizols, P. et al. Chromatin decondensation is sufficient to alter nuclear organization in embryonic stem cells. Science 346, 1238–1242 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. 1000 Genomes Project Consortium et al. A global reference for human genetic variation. Nature 526, 68–74 (2015).

  112. Tak, Y. G. & Farnham, P. J. Making sense of GWAS: using epigenomics and genome engineering to understand the functional relevance of SNPs in non-coding regions of the human genome. Epigenetics Chromatin 8, 57 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Kleinjan, D. J. & Coutinho, P. Cis-ruption mechanisms: disruption of cis-regulatory control as a cause of human genetic disease. Brief. Funct. Genomics Proteomic 8, 317–332 (2009).

    Article  CAS  Google Scholar 

  114. Benko, S. et al. Highly conserved non-coding elements on either side of SOX9 associated with Pierre Robin sequence. Nat. Genet. 41, 359–364 (2009).

    Article  CAS  PubMed  Google Scholar 

  115. Fukami, M. et al. Complex genomic rearrangement in the SOX9 5′ region in a patient with Pierre Robin sequence and hypoplastic left scapula. Am. J. Med. Genet. A 158A, 1529–1534, (2012).

    Article  CAS  PubMed  Google Scholar 

  116. Taub, R. et al. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc. Natl Acad. Sci. USA 79, 7837–7841 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Dalla-Favera, R. et al. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc. Natl Acad. Sci. USA 79, 7824–7827 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428–434 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. 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 

  120. Oldridge, D. A. et al. Genetic predisposition to neuroblastoma mediated by a LMO1 super-enhancer polymorphism. Nature 528, 418–421 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Mansour, M. R. et al. Oncogene regulation. An oncogenic super-enhancer formed through somatic mutation of a noncoding intergenic element. Science 346, 1373–1377 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Wieczorek, D. et al. A specific mutation in the distant sonic hedgehog (SHH) cis-regulator (ZRS) causes Werner mesomelic syndrome (WMS) while complete ZRS duplications underlie Haas type polysyndactyly and preaxial polydactyly (PPD) with or without triphalangeal thumb. Hum. Mutat. 31, 81–89 (2010).

    Article  CAS  PubMed  Google Scholar 

  123. Lettice, L. A. et al. Opposing functions of the ETS factor family define Shh spatial expression in limb buds and underlie polydactyly. Dev. Cell 22, 459–467 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Zhang, X. et al. Identification of focally amplified lineage-specific super-enhancers in human epithelial cancers. Nat. Genet. 48, 176–182 (2016).

    Article  CAS  PubMed  Google Scholar 

  125. van de Werken, H. J. et al. Robust 4C-seq data analysis to screen for regulatory DNA interactions. Nat. Methods 9, 969–972 (2012).

    Article  CAS  PubMed  Google Scholar 

  126. Gavrilov, A. A., Chetverina, H. V., Chermnykh, E. S., Razin, S. V. & Chetverin, A. B. Quantitative analysis of genomic element interactions by molecular colony technique. Nucleic Acids Res. 42, e36 (2014).

    Article  CAS  PubMed  Google Scholar 

  127. Jiang, T. et al. Identification of multi-loci hubs from 4C-seq demonstrates the functional importance of simultaneous interactions. Nucleic Acids Res. http://dx.doi.org/10.1093/nar/gkw568 (2016).

  128. Fuss, S. H., Omura, M. & Mombaerts, P. Local and cis effects of the H element on expression of odorant receptor genes in mouse. Cell 130, 373–384 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. Montavon, T., Thevenet, L. & Duboule, D. Impact of copy number variations (CNVs) on long-range gene regulation at the HoxD locus. Proc. Natl Acad. Sci. USA 109, 20204–20211 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Lupianez, D. G., Spielmann, M. & Mundlos, S. Breaking TADs: how alterations of chromatin domains result in disease. Trends Genet. 32, 225–237 (2016).

    Article  CAS  PubMed  Google Scholar 

  131. Kandoth, C. et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Filippova, G. N. et al. A widely expressed transcription factor with multiple DNA sequence specificity, CTCF, is localized at chromosome segment 16q22.1 within one of the smallest regions of overlap for common deletions in breast and prostate cancers. Genes Chromosomes Cancer 22, 26–36 (1998).

    Article  CAS  PubMed  Google Scholar 

  133. Kon, A. et al. Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms. Nat. Genet. 45, 1232–1237 (2013).

    Article  CAS  PubMed  Google Scholar 

  134. Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

  135. Kemp, C. J. et al. CTCF haploinsufficiency destabilizes DNA methylation and predisposes to cancer. Cell Rep. 7, 1020–1029 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Mullenders, J. et al. Cohesin loss alters adult hematopoietic stem cell homeostasis, leading to myeloproliferative neoplasms. J. Exp. Med. 212, 1833–1850 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Viny, A. D. et al. Dose-dependent role of the cohesin complex in normal and malignant hematopoiesis. J. Exp. Med. 212, 1819–1832 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Corces, M. R. & Corces, V. G. The three-dimensional cancer genome. Curr. Opin. Genet. Dev. 36, 1–7 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  140. 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 

  141. Bell, A. C. & Felsenfeld, G. Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature 405, 482–485 (2000).

    Article  CAS  PubMed  Google Scholar 

  142. Wang, H. et al. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res. 22, 1680–1688 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Maurano, M. T. et al. Large-scale identification of sequence variants influencing human transcription factor occupancy in vivo. Nat. Genet. 47, 1393–1401 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  145. Visser, M., Kayser, M. & Palstra, R. J. HERC2 rs12913832 modulates human pigmentation by attenuating chromatin-loop formation between a long-range enhancer and the OCA2 promoter. Genome Res. 22, 446–455 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Groschel, S. et al. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia. Cell 157, 369–381 (2014).

    Article  CAS  PubMed  Google Scholar 

  147. Lupianez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161, 1012–1025 (2015). References 146 and 147 illustrate how chromatin contact profiles help uncover the functional consequences of chromosomal rearrangements and in the discovery of molecular mechanisms underlying disease.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Fischer, J. et al. Inactivation of the Fto gene protects from obesity. Nature 458, 894–898 (2009).

    Article  CAS  PubMed  Google Scholar 

  149. 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 

  150. Claussnitzer, M. et al. FTO obesity variant circuitry and adipocyte browning in humans. N. Engl. J. Med. 373, 895–907 (2015). References 149 and 150 illustrate how the implementation of both epigenetic and 3D genome maps can greatly assist in the identification of the target gene of disease-associated variants.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. de Vree, P. J. et al. Targeted sequencing by proximity ligation for comprehensive variant detection and local haplotyping. Nat. Biotechnol. 32, 1019–1025 (2014).

    Article  CAS  PubMed  Google Scholar 

  152. Selvaraj, S., Schmitt, A. D., Dixon, J. R. & Ren, B. Complete haplotype phasing of the MHC and KIR loci with targeted HaploSeq. BMC Genomics 16, 900 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Splinter, E. et al. The inactive X chromosome adopts a unique three-dimensional conformation that is dependent on Xist RNA. Genes Dev. 25, 1371–1383 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Simonis, M. et al. Nuclear organization of active and inactive chromatin domains uncovered by chromosome conformation capture-on-chip (4C). Nat. Genet. 38, 1348–1354 (2006).

    Article  CAS  PubMed  Google Scholar 

  155. Hughes, J. R. et al. Analysis of hundreds of cis-regulatory landscapes at high resolution in a single, high-throughput experiment. Nat. Genet. 46, 205–212 (2014).

    Article  CAS  PubMed  Google Scholar 

  156. Davies, J. O. et al. Multiplexed analysis of chromosome conformation at vastly improved sensitivity. Nat. Methods 13, 74–80 (2016).

    Article  CAS  PubMed  Google Scholar 

  157. 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 

  158. Sahlen, P. et al. Genome-wide mapping of promoter-anchored interactions with close to single-enhancer resolution. Genome Biol. 16, 156 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Dostie, J. et al. Chromosome Conformation Capture Carbon Copy (5C): a massively parallel solution for mapping interactions between genomic elements. Genome Res. 16, 1299–1309 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Li, M. J., Wang, L. Y., Xia, Z., Sham, P. C. & Wang, J. GWAS3D: detecting human regulatory variants by integrative analysis of genome-wide associations, chromosome interactions and histone modifications. Nucleic Acids Res. 41, W150–W158 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

W.d.L's research is supported by a Netherlands Organisation for Scientific Research-Chemical Sciences (NWO/CW) TOP grant (714.012.002), an NWO-Innovational Research Incentives Scheme (VICI) grant (724.012.003), a European Union grant (2010–259743; Modelling Hepatocellular Carcinoma (MODHEP)) and a grant from The Leducq Foundation (FP058566-01-PR).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wouter de Laat.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Thalassaemias

Inherited blood disorders characterized by the abnormal formation of haemoglobin and caused by the reduced production of globin proteins.

Enhancers

Regulatory DNA elements that can upregulate the transcriptional output of a target gene.

Topologically associated domains

(TADs). Boundary-insulated chromosomal segments within which sequences preferentially contact each other.

Cofactor p300

An enhancer-associated protein with histone acetyltransferase activity that recruits transcriptional-activating proteins and can increase gene expression.

STARR-seq

Self-transcribing active regulatory region sequencing. A massively parallel analysis plasmid based method to interrogate the sequence-intrinsic enhancer capacity of genomic segments.

Housekeeping gene promoter

Promoter of a gene that is expressed in all tissues.

Minimal promoter

The minimal portion of the promoter (the core promoter) required to initiate transcription. When integrated in the genome, it depends on enhancers for detectable levels of transcription.

Enhancer trap assay

Assay in which transcriptional output of a genomic integrated reporter gene under the control of a minimal promoter is used to identify regulatory elements in a given genomic context.

Structural variations

Changes in the linear order of multiple base pairs as a consequence of a chromosomal rearrangement (for example, translocation, inversion, deletion, insertion or duplication).

Nuclear compartments

Nuclear sub-volumes in which chromosomal regions with similar chromatin composition spatially aggregate.

Haplotype

A genomic region that is inherited from one parent that has a sequence composition with genetic variations such as single-nucleotide polymorphisms (SNPs) that can be different from those found in the corresponding genomic region inherited by the other parent.

Chromocentres

Sites in the nuclear interior where heterochromatic pericentromeric regions of different chromosomes cluster.

Polycomb

A family of proteins that remodel chromatin to repress transcription.

SNPs

Single-nucleotide polymorphisms causing genetic variation.

Indels

Short DNA sequence insertions or deletions.

Copy number variations

Changes in the number of copies of a given chromosomal segment in a cell caused by their duplication or deletion.

Drivers

Risk-associated variants that cause disease.

Passengers

Risk-associated variants that do not cause disease.

Linkage disequilibrium

Alleles of SNPs are in linkage disequilibrium when their frequency of association in the population is not random, which indicates that they are physically and/or functionally linked.

Chromosomal breakpoints

The sites where chromosomes were incidentally broken and, as consequence of erroneous DNA repair, now flank a chromosomal rearrangement.

Haploinsufficient

A phenotypic state observed despite a diploid organism still carrying one functional gene.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Krijger, P., de Laat, W. Regulation of disease-associated gene expression in the 3D genome. Nat Rev Mol Cell Biol 17, 771–782 (2016). https://doi.org/10.1038/nrm.2016.138

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm.2016.138

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