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:

Understanding 3D genome organization by multidisciplinary methods

Abstract

Understanding how chromatin is folded in the nucleus is fundamental to understanding its function. Although 3D genome organization has been historically difficult to study owing to a lack of relevant methodologies, major technological breakthroughs in genome-wide mapping of chromatin contacts and advances in imaging technologies in the twenty-first century considerably improved our understanding of chromosome conformation and nuclear architecture. In this Review, we discuss methods of 3D genome organization analysis, including sequencing-based techniques, such as Hi-C and its derivatives, Micro-C, DamID and others; microscopy-based techniques, such as super-resolution imaging coupled with fluorescence in situ hybridization (FISH), multiplex FISH, in situ genome sequencing and live microscopy methods; and computational and modelling approaches. We describe the most commonly used techniques and their contribution to our current knowledge of nuclear architecture and, finally, we provide a perspective on up-and-coming methods that open possibilities for future major discoveries.

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: Multiple levels of genome organization and the methods to study them.
Fig. 2: Main C-based methods for interrogation of 3D genome organization.
Fig. 3: Main ligation-independent methods for interrogation of 3D genome organization.
Fig. 4: Microscopy and FISH-based methods for 3D genome investigation.
Fig. 5: Computational modelling of the 3D genome.
Fig. 6: Live microscopy using CRISPR–dCas9 to study the 4D genome.

Similar content being viewed by others

References

  1. Flemming, W. Zellsubstanz, Kern und Zelltheilung (F. C. W. Vogel, 1882).

  2. Heitz, E. Das heterochromatin der moose. Jahrbücher Wissenschaftliche Bot. 69, 762–818 (1928).

    Google Scholar 

  3. Boveri, T. Die Blastomerenkerne von Ascaris megalocephala und die Theorie der Chromosomenindividualität (Engelmann, 1909).

  4. Cremer, T. et al. Analysis of chromosome positions in the interphase nucleus of Chinese hamster cells by laser-UV-microirradiation experiments. Hum. Genet. 62, 201–209 (1982).

    Article  CAS  PubMed  Google Scholar 

  5. Manuelidis, L. Individual interphase chromosome domains revealed by in situ hybridization. Hum. Genet. 71, 288–293 (1985).

    Article  CAS  PubMed  Google Scholar 

  6. Schardin, M., Cremer, T., Hager, H. D. & Lang, M. Specific staining of human chromosomes in Chinese hamster × man hybrid cell lines demonstrates interphase chromosome territories. Hum. Genet. 71, 281–287 (1985).

    Article  CAS  PubMed  Google Scholar 

  7. Cremer, M. et al. Multicolor 3D fluorescence in situ hybridization for imaging interphase chromosomes. Methods Mol. Biol. 463, 205–239 (2012).

    Article  Google Scholar 

  8. Branco, M. R. & Pombo, A. Intermingling of chromosome territories in interphase suggests role in translocations and transcription-dependent associations. PLoS Biol. 4, e138 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Chambeyron, S. & Bickmore, W. A. Chromatin decondensation and nuclear reorganization of the HoxB locus upon induction of transcription. Genes Dev. 18, 1119–1130 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ferrai, C. et al. Poised transcription factories prime silent uPA gene prior to activation. PLoS Biol. 8, e1000270 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Rosin, L. F., Nguyen, S. C. & Joyce, E. F. Condensin II drives large-scale folding and spatial partitioning of interphase chromosomes in Drosophila nuclei. PLOS Genet. 14, e1007393 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Su, J. H., Zheng, P., Kinrot, S. S., Bintu, B. & Zhuang, X. Genome-scale imaging of the 3D organization and transcriptional activity of chromatin. Cell 182, 1641–1659 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fritz, A. J., Sehgal, N., Pliss, A., Xu, J. & Berezney, R. Chromosome territories and the global regulation of the genome. Genes Chromosom. Cancer 58, 407–426 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Nguyen, H. Q. et al. 3D mapping and accelerated super-resolution imaging of the human genome using in situ sequencing. Nat. Methods 17, 822–832 (2020). This article is the first mention of OligoFISSEQ, showcasing its multiplexing possibilities by imaging 249 loci simultaneously.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Takei, Y. et al. Integrated spatial genomics reveals global architecture of single nuclei. Nature 590, 344–350 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Payne, A. C. et al. In situ genome sequencing resolves DNA sequence and structure in intact biological samples. Science 371, eaay3446 (2020).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  17. Cullen, K. E., Kladde, M. P. & Seyfred, M. A. Interaction between transcription regulatory regions of prolactin chromatin. Science 261, 203–206 (1993).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. 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 

  21. Schoenfelder, S. et al. The pluripotent regulatory circuitry connecting promoters to their long-range interacting elements. Genome Res. 25, 582–597 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schoenfelder, S. et al. Polycomb repressive complex PRC1 spatially constrains the mouse embryonic stem cell genome. Nat. Genet. 47, 1179–1186 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. 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 

  25. Davies, J. O. J. et al. Multiplexed analysis of chromosome conformation at vastly improved sensitivity. Nat. Methods 13, 74–80 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Jäger, R. et al. Capture Hi-C identifies the chromatin interactome of colorectal cancer risk loci. Nat. Commun. 6, 6178 (2015).

    Article  PubMed  CAS  Google Scholar 

  27. 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 

  28. Duan, Z. et al. A three-dimensional model of the yeast genome. Nature 465, 363–367 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sati, S. & Cavalli, G. Chromosome conformation capture technologies and their impact in understanding genome function. Chromosoma 126, 33–44 (2017).

    Article  PubMed  Google Scholar 

  30. Horike, S. I., Cai, S., Miyano, M., Cheng, J. F. & Kohwi-Shigematsu, T. Loss of silent-chromatin looping and impaired imprinting of DLX5 in Rett syndrome. Nat. Genet. 37, 31–40 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Fullwood, M. J., Wei, C. L., Liu, E. T. & Ruan, Y. Next-generation DNA sequencing of paired-end tags (PET) for transcriptome and genome analyses. Genome Res. 19, 521–532 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, Y. et al. Mapping 3D genome organization relative to nuclear compartments using TSA-seq as a cytological ruler. J. Cell Biol. 217, 4025–4048 (2018). This article introduces TSA-seq, a cytological ruler for nuclear speckles and lamina, which is the first genomics method enabling transforming sequencing reads into physical distances in the nucleus.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Quinodoz, S. A. et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 174, 744–757 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Arrastia, M. V. et al. A single-cell method to map higher-order 3D genome organization in thousands of individual cells reveals structural heterogeneity in mouse ES cells. Preprint at bioRxiv https://doi.org/10.1101/2020.08.11.242081 (2020).

    Article  Google Scholar 

  37. Quinodoz, S. A. et al. RNA promotes the formation of spatial compartments in the nucleus. Preprint at bioRxiv https://doi.org/10.1101/2020.08.25.267435 (2020).

    Article  Google Scholar 

  38. Van Steensel, B. & Henikoff, S. Identification of in vivo DNA targets of chromatin proteins using tethered Dam methyltransferase. Nat. Biotechnol. 14, 424–428 (2000). This article introduces DamID, which is used for spatial positioning of heterochromatin protein 1 on chromatin in vitro and in vivo in D. melanogaster.

    Article  CAS  Google Scholar 

  39. Vogel, M. J., Peric-Hupkes, D. & van Steensel, B. Detection of in vivo protein–DNA interactions using DamID in mammalian cells. Nat. Protoc. 2, 1467–1478 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Guelen, L. et al. Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions. Nature 453, 948–951 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Beagrie, R. A. et al. Complex multi-enhancer contacts captured by genome architecture mapping. Nature 543, 519–524 (2017). This article introduces the GAM method and the discovery of a richness of multiway contacts in the nucleus.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang, L. et al. TSA-seq reveals a largely conserved genome organization relative to nuclear speckles with small position changes tightly correlated with gene expression changes. Genome Res. 31, 251–264 (2021).

    Article  PubMed Central  Google Scholar 

  43. Szabo, Q. et al. TADs are 3D structural units of higher-order chromosome organization in Drosophila. Sci. Adv. 4, eaar8082 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Bintu, B. et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 362, eaau1783 (2018). This study uses high-throughput oligopaint technology to study chromatin dynamics and shows that TADs are highly stochastic in single cells, but predictable at the population level.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Cardozo Gizzi, A. M. et al. Microscopy-based chromosome conformation capture enables simultaneous visualization of genome organization and transcription in intact organisms. Mol. Cell 74, 212–222.e5 (2019). This article presents highly multiplexed sequential oligopaint FISH (RNA and DNA) based on the conjunction of microscopy and microfluidics in D. melanogaster embryos.

    Article  CAS  PubMed  Google Scholar 

  46. Nir, G. et al. Walking along chromosomes with super-resolution imaging, contact maps, and integrative modeling. PLoS Genet. 14, e1007872 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Mateo, L. J. et al. Visualizing DNA folding and RNA in embryos at single-cell resolution. Nature 568, 49–54 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sawh, A. N. et al. Lamina-dependent stretching and unconventional chromosome compartments in early C. elegans embryos. Mol. Cell 78, 96–111 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shaban, H. A. & Seeber, A. Monitoring the spatio-temporal organization and dynamics of the genome. Nucleic Acids Res. 48, 3423–3434 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Barth, R., Bystricky, K. & Shaban, H. A. Coupling chromatin structure and dynamics by live super-resolution imaging. Sci. Adv. 6, eaaz2196 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Vangala, P. et al. High-resolution mapping of multiway enhancer–promoter interactions regulating pathogen detection. Mol. Cell 80, 359–373 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

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

    Article  CAS  PubMed  Google Scholar 

  55. 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 

  56. Kalhor, R. et al. Developmental barcoding of whole mouse via homing CRISPR. Science 361, eaat9804 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Szabo, Q. et al. Regulation of single-cell genome organization into TADs and chromatin nanodomains. Nat. Genet. 52, 1151–1157 (2020). This article is the first description of chromatin nanodomains in mammals using oligopaints in conjunction with SIM.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wutz, G. et al. Topologically associating domains and chromatin loops depend on cohesin and are regulated by CTCF, WAPL, and PDS5 proteins. EMBO J. 36, 3573–3599 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Schwarzer, W. et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature 551, 51–56 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  62. 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 

  63. Haarhuis, J. H. I. et al. The cohesin release factor WAPL restricts chromatin loop extension. Cell 169, 693–707 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bonev, B. et al. Multiscale 3D genome rewiring during mouse article multiscale 3D genome rewiring during mouse neural development. Cell 171, 557–572 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ulianov, S. V. et al. Active chromatin and transcription play a key role in chromosome partitioning into topologically associating domains. Genome Res. 26, 70–84 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  67. 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, 6456–6465 (2015).

    Article  CAS  Google Scholar 

  68. Nuebler, J., Fudenberg, G., Imakaev, M., Abdennur, N. & Mirny, L. A. Chromatin organization by an interplay of loop extrusion and compartmental segregation. Proc. Natl Acad. Sci. USA 115, 6697–6706 (2018).

    Article  CAS  Google Scholar 

  69. Rowley, M. J. et al. Evolutionarily conserved principles predict 3D chromatin organization. Mol. Cell 67, 837–852 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Benedetti, F., Racko, D., Dorier, J., Burnier, Y. & Stasiak, A. Transcription-induced supercoiling explains formation of self-interacting chromatin domains in S. pombe. Nucleic Acids Res. 45, 9850–9859 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Szabo, Q., Bantignies, F. & Cavalli, G. Principles of genome folding into topologically associating domains. Sci. Adv. 5, eaaw1668 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jerković, I., Szabo, Q., Bantignies, F. & Cavalli, G. Higher-order chromosomal structures mediate genome function. J. Mol. Biol. 432, 676–681 (2020).

    Article  PubMed  CAS  Google Scholar 

  73. Oomen, M. E., Hedger, A. K., Watts, J. K. & Dekker, J. Detecting chromatin interactions between and along sister chromatids with SisterC. Nat. Methods 17, 1002–1009 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mitter, M. et al. Conformation of sister chromatids in the replicated human genome. Nature 586, 139–144 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. AlHaj Abed, J. et al. Highly structured homolog pairing reflects functional organization of the Drosophila genome. Nat. Commun. 10, 1–14 (2019).

    Article  CAS  Google Scholar 

  76. Franke, M. et al. Formation of new chromatin domains determines pathogenicity of genomic duplications. Nature 538, 265–269 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  78. Despang, A. et al. Functional dissection of the Sox9–Kcnj2 locus identifies nonessential and instructive roles of TAD architecture. Nat. Genet. 51, 1263–1271 (2019).

    Article  CAS  PubMed  Google Scholar 

  79. Andrey, G. & Mundlos, S. The three-dimensional genome: regulating gene expression during pluripotency and development. Development 144, 3646–3658 (2017).

    Article  CAS  PubMed  Google Scholar 

  80. Weischenfeldt, J. et al. Pan-cancer analysis of somatic copy-number alterations implicates IRS4 and IGF2 in enhancer hijacking. Nat. Genet. 49, 65–74 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Liu, X. S. et al. Rescue of fragile X syndrome neurons by DNA methylation editing of the FMR1 gene. Cell 172, 979–992 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Bruneau, B. G. & Nora, E. P. Chromatin domains go on repeat in disease. Cell 175, 38–40 (2018).

    Article  CAS  PubMed  Google Scholar 

  83. Akdemir, K. C. et al. Somatic mutation distributions in cancer genomes vary with three-dimensional chromatin structure. Nat. Genet. 52, 1178–1188 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Valton, A. L. & Dekker, J. TAD disruption as oncogenic driver. Curr. Opin. Genet. Dev. 36, 34–40 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. 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). This paper showcases the first use of the in situ Hi-C protocol and introduces HICCUPS, a contact caller dedicated to chromatin loops.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Nagano, T. et al. Comparison of Hi-C results using in-solution versus in-nucleus ligation. Genome Biol. 16, 175 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  87. 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 

  88. Ma, W. et al. Fine-scale chromatin interaction maps reveal the cis-regulatory landscape of human lincRNA genes. Nat. Methods 12, 71–78 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Ramani, V. et al. Mapping 3D genome architecture through in situ DNase Hi-C. Nat. Protoc. 11, 2104–2121 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Deng, X. et al. Bipartite structure of the inactive mouse X chromosome. Genome Biol. 16, 152 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Hsieh, T. H. S. et al. Mapping nucleosome resolution chromosome folding in yeast by Micro-C. Cell 162, 108–119 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Hsieh, T.-H. S. et al. Resolving the 3D landscape of transcription-linked mammalian chromatin folding. Mol. Cell 78, 539–553 (2020). This article presents high-resolution Micro-C performed in mouse embryonic stem cells with concomitant analysis of nucleosome positioning.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Krietenstein, N. et al. Ultrastructural details of mammalian chromosome architecture. Mol. Cell 78, 554–565 (2020). This article presents high-resolution Micro-C performed in human embryonic stem cells and fibroblasts with concomitant analysis of nucleosome positioning.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Goel, V. Y. & Hansen, A. S. The macro and micro of chromosome conformation capture. WIREs Dev. Biol. https://doi.org/10.1002/wdev.395 (2020).

    Article  Google Scholar 

  95. Akgol Oksuz, B. et al. Systematic evaluation of chromosome conformation capture assays. Preprint at bioRxiv https://doi.org/10.1101/2020.12.26.424448 (2020).

    Article  Google Scholar 

  96. Baranello, L., Kouzine, F., Sanford, S. & Levens, D. ChIP bias as a function of cross-linking time. Chromosom. Res. 24, 175–181 (2016).

    Article  CAS  Google Scholar 

  97. Gavrilov, A., Razin, S. V. & Cavalli, G. In vivo formaldehyde cross-linking: it is time for black box analysis. Brief. Funct. Genomics 14, 163–165 (2015).

    Article  CAS  PubMed  Google Scholar 

  98. Kempfer, R. & Pombo, A. Methods for mapping 3D chromosome architecture. Nat. Rev. Genet. 21, 207–226 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Kind, J. et al. Genome-wide maps of nuclear lamina interactions in single human cells. Cell 163, 134–147 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Redolfi, J. et al. DamC reveals principles of chromatin folding in vivo without crosslinking and ligation. Nat. Struct. Mol. Biol. 26, 471–480 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Li, X. et al. Long-read ChIA-PET for base-pair-resolution mapping of haplotype-specific chromatin interactions. Nat. Protoc. 12, 899–915 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Andrey, G. et al. Characterization of hundreds of regulatory landscapes in developing limbs reveals two regimes of chromatin folding. Genome Res. 27, 223–233 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Rooijers, K. et al. Simultaneous quantification of protein–DNA contacts and transcriptomes in single cells. Nat. Biotechnol. 37, 766–772 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Sahl, S. J., Hell, S. W. & Jakobs, S. Fluorescence nanoscopy in cell biology. Nat. Rev. Mol. Cell Biol. 18, 685–701 (2017).

    Article  CAS  PubMed  Google Scholar 

  106. Liu, Z., Lavis, L. D. & Betzig, E. Imaging live-cell dynamics and structure at the single-molecule level. Mol. Cell 58, 644–659 (2015).

    Article  CAS  PubMed  Google Scholar 

  107. Heintzmann, R. & Cremer, C. G. Laterally modulated excitation microscopy: improvement of resolution by using a diffraction grating. Proc. SPIE 3568, 185–196 (1999).

    Article  Google Scholar 

  108. Gustafsson, M. G. L. Surpassing the lateral resolution limit by a factor of two using structured illumination microscopy. J. Microsc. 198, 82–87 (2000).

    Article  CAS  PubMed  Google Scholar 

  109. Gustafsson, M. G. L. et al. Three-dimensional resolution doubling in wide-field fluorescence microscopy by structured illumination. Biophys. J. 94, 4957–4970 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Schermelleh, L. et al. Subdiffraction multicolor imaging of the nuclear periphery with 3D structured illumination microscopy. Science 320, 1332–1336 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Miron, E. et al. Chromatin arranges in chains of mesoscale domains with nanoscale functional topography independent of cohesin. Sci. Adv. 6, eaba8811 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Rust, M. J., Bates, M. & Zhuang, X. Sub-diffraction-limit imaging by stochastic optical reconstruction microscopy (STORM). Nat. Methods 3, 793–795 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Betzig, E. et al. Imaging intracellular fluorescent proteins at nanometer resolution. Science 313, 1642–1645 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Hess, S. T., Girirajan, T. P. K. & Mason, M. D. Ultra-high resolution imaging by fluorescence photoactivation localization microscopy. Biophys. J. 91, 4258–4272 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Huang, B., Babcock, H. & Zhuang, X. Breaking the diffraction barrier: super-resolution imaging of cells. Cell 143, 1047–1058 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Ricci, M. A., Manzo, C., Lakadamyali, M. & Cosma, M. P. Chromatin fibers are formed by heterogeneous groups of nucleosomes in vivo. Cell 160, 1145–1158 (2015).

    Article  CAS  PubMed  Google Scholar 

  117. Nozaki, T. et al. Dynamic organization of chromatin domains revealed by super-resolution live-cell imaging. Mol. Cell 67, 282–293 (2017).

    Article  CAS  PubMed  Google Scholar 

  118. Hell, S. W. & Wichmann, J. Breaking the diffraction resolution limit by stimulated emission: stimulated-emission-depletion fluorescence microscopy. Opt. Lett. 19, 780–782 (1994).

    Article  CAS  PubMed  Google Scholar 

  119. Klar, T. A., Jakobs, S., Dyba, M., Egner, A. & Hell, S. W. Fluorescence microscopy with diffraction resolution barrier broken by stimulated emission. Proc. Natl Acad. Sci. USA 97, 8206–8210 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Hofmann, M., Eggeling, C., Jakobs, S. & Hell, S. W. Breaking the diffraction barrier in fluorescence microscopy at low light intensities by using reversibly photoswitchable proteins. Proc. Natl Acad. Sci. USA 102, 17565–17569 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Gwosch, C. K. et al. MINFLUX nanoscopy delivers multicolor nanometer 3D-resolution in (living) cells. Nat. Methods 17, 217–224 (2020).

    Article  CAS  PubMed  Google Scholar 

  122. Beliveau, B. J. et al. Versatile design and synthesis platform for visualizing genomes with Oligopaint FISH probes. Proc. Natl Acad. Sci. USA 109, 21301–21306 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Wang, S. et al. Spatial organization of chromatin domains and compartments in single chromosomes. Science 353, 598–602 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ou, H. D. et al. ChromEMT: visualizing 3D chromatin structure and compaction in interphase and mitotic cells. Science 357, eaag0025 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Yaffe, E. & Tanay, A. Probabilistic modeling of Hi-C contact maps eliminates systematic biases to characterize global chromosomal architecture. Nat. Genet. 43, 1059–1065 (2011).

    Article  CAS  PubMed  Google Scholar 

  126. Imakaev, M. et al. Iterative correction of Hi-C data reveals hallmarks of chromosome organization. Nat. Methods 9, 999–1003 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ryba, T. et al. Evolutionarily conserved replication timing profiles predict long-range chromatin interactions and distinguish closely related cell types. Genome Res. 20, 761–770 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Di Pierro, M., Cheng, R. R., Aiden, E. L., Wolynes, P. G. & Onuchic, J. N. De novo prediction of human chromosome structures: epigenetic marking patterns encode genome architecture. Proc. Natl Acad. Sci. USA 114, 12126–12131 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Lin, D., Bonora, G., Yardimci, G. G. & Noble, W. S. Computational methods for analyzing and modeling genome structure and organization. Wiley Interdiscip. Rev. Syst. Biol. Med. 11, e1435 (2019).

    Article  PubMed  CAS  Google Scholar 

  130. Kalhor, R., Tjong, H., Jayathilaka, N., Alber, F. & Chen, L. Genome architectures revealed by tethered chromosome conformation capture and population-based modeling. Nat. Biotechnol. 30, 90–98 (2012).

    Article  CAS  Google Scholar 

  131. Crane, E. et al. Condensin-driven remodelling of X chromosome topology during dosage compensation. Nature 523, 240–244 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Shin, H. et al. TopDom: an efficient and deterministic method for identifying topological domains in genomes. Nucleic Acids Res. 44, e70 (2015). This article presents one of the most reliable and popular TAD callers.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Malik, L. & Patro, R. Rich chromatin structure prediction from Hi-C data. IEEE/ACM Trans. Comput. Biol. Bioinform. 16, 1448–1458 (2019).

    Article  CAS  PubMed  Google Scholar 

  134. Haddad, N., Vaillant, C. & Jost, D. IC-Finder: inferring robustly the hierarchical organization of chromatin folding. Nucleic Acids Res. 45, 81 (2017).

    CAS  Google Scholar 

  135. Soler-Vila, P., Cuscó, P., Farabella, I., Di Stefano, M. & Marti-Renom, M. A. Hierarchical chromatin organization detected by TADpole. Nucleic Acids Res. 48, e39 (2020). This paper presents one of the most recently developed TAD callers and is extremely valuable for its benchmarking quality and for multiple TAD caller comparisons.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Norton, H. K. et al. Detecting hierarchical genome folding with network modularity. Nat. Methods 15, 119–122 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Moller, J. & de Pablo, J. J. Bottom-up meets top-down: the crossroads of multiscale chromatin modeling. Biophys. J. 118, 2057–2065 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Bendandi, A., Dante, S., Zia, S. R., Diaspro, A. & Rocchia, W. Chromatin compaction multiscale modeling: a complex synergy between theory, simulation, and experiment. Front. Mol. Biosci. 7, 15 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Zufferey, M., Tavernari, D., Oricchio, E. & Ciriello, G. Comparison of computational methods for the identification of topologically associating domains. Genome Biol. 19, 217 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Forcato, M. et al. Comparison of computational methods for Hi-C data analysis. Nat. Methods 14, 679–685 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Ay, F., Bailey, T. L. & Noble, W. S. Statistical confidence estimation for Hi-C data reveals regulatory chromatin contacts. Genome Res. 24, 999–1011 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Carty, M. et al. An integrated model for detecting significant chromatin interactions from high-resolution Hi-C data. Nat. Commun. 8, 1–10 (2017).

    Article  CAS  Google Scholar 

  143. Pal, K., Forcato, M. & Ferrari, F. Hi-C analysis: from data generation to integration. Biophys. Rev. 11, 67–78 (2019).

    Article  PubMed  Google Scholar 

  144. Dozmorov, M., Sirusb & Benfeitas, R. Hi-C data analysis tools and papers. GitHub https://github.com/mdozmorov/HiC_tools/blob/master/README.md (2021).

  145. Roayaei Ardakany, A., Gezer, H. T., Lonardi, S. & Ay, F. Mustache: multi-scale detection of chromatin loops from Hi-C and Micro-C maps using scale-space representation. Genome Biol. 21, 256 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  146. Jordan Rowley, M. et al. Analysis of Hi-C data using SIP effectively identifies loops in organisms from C. elegans to mammals. Genome Res. 30, 447–458 (2020).

    Article  PubMed  CAS  Google Scholar 

  147. Ay, F. & Noble, W. S. Analysis methods for studying the 3D architecture of the genome. Genome Biol. 16, 183 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Nikumbh, S. & Pfeifer, N. Genetic sequence-based prediction of long-range chromatin interactions suggests a potential role of short tandem repeat sequences in genome organization. BMC Bioinformatics 18, 218 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Rhie, S. K. et al. Using 3D epigenomic maps of primary olfactory neuronal cells from living individuals to understand gene regulation. Sci. Adv. 4, eaav8550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Hafez, D. et al. McEnhancer: predicting gene expression via semi-supervised assignment of enhancers to target genes. Genome Biol. 18, 199 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Di Iulio, J. et al. The human noncoding genome defined by genetic diversity. Nat. Genet. 50, 333–337 (2018).

    Article  PubMed  CAS  Google Scholar 

  152. Javierre, B. M. et al. Lineage-specific genome architecture links enhancers and non-coding disease variants to target gene promoters. Cell 167, 1369–1384 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Mitchelmore, J., Grinberg, N. F., Wallace, C. & Spivakov, M. Functional effects of variation in transcription factor binding highlight long-range gene regulation by epromoters. Nucleic Acids Res. 48, 2866–2879 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Malysheva, V., Mendoza-Parra, M. A., Blum, M., Spivakov, M. & Gronemeyer, H. Gene regulatory network reconstruction incorporating 3D chromosomal architecture reveals key transcription factors and DNA elements driving neural lineage commitment. Preprint at bioRxiv https://doi.org/10.1101/303842 (2019).

    Article  Google Scholar 

  155. Madrid-Mencía, M., Raineri, E., Cao, T. B. N. & Pancaldi, V. Using GARDEN-NET and ChAseR to explore human haematopoietic 3D chromatin interaction networks. Nucleic Acids Res. 48, 4066–4080 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Zhang, Y. et al. Enhancing Hi-C data resolution with deep convolutional neural network HiCPlus. Nat. Commun. 9, 1–9 (2018).

    CAS  Google Scholar 

  157. Oluwadare, O., Highsmith, M. & Cheng, J. An overview of methods for reconstructing 3-D chromosome and genome structures from Hi-C data. Biol. Proced. Online 21, 1–20 (2019).

    Article  Google Scholar 

  158. Marti-Renom, M. A. et al. Challenges and guidelines toward 4D nucleome data and model standards. Nat. Genet. 50, 1352–1358 (2018).

    Article  CAS  PubMed  Google Scholar 

  159. MacPherson, Q., Beltran, B. & Spakowitz, A. J. Bottom-up modeling of chromatin segregation due to epigenetic modifications. Proc. Natl Acad. Sci. USA 115, 12739–12744 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Bianco, S., Chiariello, A. M., Annunziatella, C., Esposito, A. & Nicodemi, M. Predicting chromatin architecture from models of polymer physics. Chromosome Res. 25, 25–34 (2017).

    Article  CAS  PubMed  Google Scholar 

  161. Mirny, L. A. The fractal globule as a model of chromatin architecture in the cell. Chromosom. Res. 19, 37–51 (2011).

    Article  CAS  Google Scholar 

  162. Barbieri, M. et al. A model of the large-scale organization of chromatin. Biochem. Soc. Trans. 41, 508–512 (2013).

    Article  CAS  PubMed  Google Scholar 

  163. Jost, D., Carrivain, P., Cavalli, G. & Vaillant, C. Modeling epigenome folding: formation and dynamics of topologically associated chromatin domains. Nucleic Acids Res. 42, 9553–9561 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Jost, D. & Vaillant, C. Epigenomics in 3D: importance of long-range spreading and specific interactions in epigenomic maintenance. Nucleic Acids Res. 46, 2252–2264 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Barbieri, M. et al. Active and poised promoter states drive folding of the extended HoxB locus in mouse embryonic stem cells. Nat. Struct. Mol. Biol. 24, 515–524 (2017).

    Article  CAS  PubMed  Google Scholar 

  166. Falk, M. et al. Heterochromatin drives compartmentalization of inverted and conventional nuclei. Nature 570, 395–399 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Oliveira Junior, A. B., Contessoto, V. G., Mello, M. F. & Onuchic, J. N. A scalable computational approach for simulating complexes of multiple chromosomes. J. Mol. Biol. 433, 166700 (2020).

    Article  PubMed  CAS  Google Scholar 

  168. Ghosh, S. K. & Jost, D. How epigenome drives chromatin folding and dynamics, insights from efficient coarse-grained models of chromosomes. PLoS Comput. Biol. 14, e1006159 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Giorgetti, L. et al. Predictive polymer modeling reveals coupled fluctuations in chromosome conformation and transcription. Cell 157, 950–963 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Bianco, S. et al. Polymer physics predicts the effects of structural variants on chromatin architecture. Nat. Genet. 50, 662–667 (2018).

    Article  CAS  PubMed  Google Scholar 

  171. Contessoto, V. G. et al. The Nucleome Data Bank: web-based resources to simulate and analyze the three-dimensional genome. Nucleic Acids Res. 49, 172–182 (2020).

    Article  CAS  Google Scholar 

  172. Rousseau, M., Fraser, J., Ferraiuolo, M. A., Dostie, J. & Blanchette, M. Three-dimensional modeling of chromatin structure from interaction frequency data using Markov chain Monte Carlo sampling. BMC Bioinformatics 12, 414 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  173. Baú, D. et al. The three-dimensional folding of the α-globin gene domain reveals formation of chromatin globules. Nat. Struct. Mol. Biol. 18, 107–115 (2011).

    Article  PubMed  CAS  Google Scholar 

  174. Trussart, M. et al. Defined chromosome structure in the genome-reduced bacterium Mycoplasma pneumoniae. Nat. Commun. 8, 14665 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Zhu, G. et al. Reconstructing spatial organizations of chromosomes through manifold learning. Nucleic Acids Res. 46, e50 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Paulsen, J. et al. Long-range interactions between topologically associating domains shape the four-dimensional genome during differentiation. Nat. Genet. 51, 835–843 (2019).

    Article  CAS  PubMed  Google Scholar 

  177. Lesne, A., Riposo, J., Roger, P., Cournac, A. & Mozziconacci, J. 3D genome reconstruction from chromosomal contacts. Nat. Methods 11, 1141–1143 (2014).

    Article  CAS  PubMed  Google Scholar 

  178. Trieu, T., Oluwadare, O. & Cheng, J. Hierarchical reconstruction of high-resolution 3D models of large chromosomes. Sci. Rep. 9, 1–12 (2019).

    Article  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Di Stefano, M. et al. Transcriptional activation during cell reprogramming correlates with the formation of 3D open chromatin hubs. Nat. Commun. 11, 1–12 (2020).

    Article  CAS  Google Scholar 

  181. Li, Q. et al. The three-dimensional genome organization of Drosophila melanogaster through data integration. Genome Biol. 18, 145 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Hua, N. et al. Producing genome structure populations with the dynamic and automated PGS software. Nat. Protoc. 13, 915–926 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Whalen, S., Truty, R. M. & Pollard, K. S. Enhancer–promoter interactions are encoded by complex genomic signatures on looping chromatin. Nat. Genet. 48, 488–496 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Li, W., Wong, W. H. & Jiang, R. DeepTACT: predicting 3D chromatin contacts via bootstrapping deep learning. Nucleic Acids Res. 47, e60 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Fudenberg, G., Kelley, D. R. & Pollard, K. S. Predicting 3D genome folding from DNA sequence with Akita. Nat. Methods 17, 1111–1117 (2020). This is the first computational modelling paper describing modelling of chromatin conformation solely from the DNA.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Schwessinger, R. et al. DeepC: predicting 3D genome folding using megabase-scale transfer learning. Nat. Methods 17, 1118–1124 (2020). This article presents one of the first computational tools to model chromatin conformation solely from the DNA sequence. However, unlike Akita, it requires training on the epigenetics data.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Zhang, S., Chasman, D., Knaack, S. & Roy, S. In silico prediction of high-resolution Hi-C interaction matrices. Nat. Commun. 10, 1–18 (2019).

    Article  CAS  Google Scholar 

  188. Belokopytova, P. S., Nuriddinov, M. A., Mozheiko, E. A., Fishman, D. & Fishman, V. Quantitative prediction of enhancer–promoter interactions. Genome Res. 30, 72–84 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Qi, Y. et al. Data-driven polymer model for mechanistic exploration of diploid genome organization. Biophys. J. 119, 1905–1916 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. de Luca, K. L. & Kind, J. in Methods in Molecular Biology Vol. 2157 159–172 (Humana, 2021).

  191. Li, G. et al. Joint profiling of DNA methylation and chromatin architecture in single cells. Nat. Methods 16, 991–993 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Zheng, M. et al. Multiplex chromatin interactions with single-molecule precision. Nature 566, 558–562 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. You, Q. et al. Direct DNA crosslinking with CAP-C uncovers transcription-dependent chromatin organization at high resolution. Nat. Biotechnol. 39, 225–235 (2020).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  195. Girelli, G. et al. GPSeq reveals the radial organization of chromatin in the cell nucleus. Nat. Biotechnol. 38, 1184–1193 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Wang, Y. et al. SPIN reveals genome-wide landscape of nuclear compartmentalization. Genome Biol. 22, 36 (2021). This article presents the first tool that combines nuclear architecture (positioning) data from TSA-seq and DamID in order to compute spatial compartmentalization of chromatin in respect to the lamina or nuclear bodies such as nuclear speckles.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Kubalová, I. et al. Prospects and limitations of expansion microscopy in chromatin ultrastructure determination. Chromosom. Res. 28, 355–368 (2020).

    Article  Google Scholar 

  198. Shi, X. et al. Label-retention expansion microscopy. Preprint at bioRxiv https://doi.org/10.1101/687954 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  199. Wassie, A. T., Zhao, Y. & Boyden, E. S. Expansion microscopy: principles and uses in biological research. Nat. Methods 16, 33–41 (2019).

    Article  CAS  PubMed  Google Scholar 

  200. Alon, S. et al. Expansion sequencing: spatially precise in situ transcriptomics in intact biological systems. Science 371, eaax2656 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Robinett, C. C. et al. In vivo localization of DNA sequences and visualization of large-scale chromatin organization using lac operator/repressor recognition. J. Cell Biol. 135, 1685–1700 (1996).

    Article  CAS  PubMed  Google Scholar 

  202. Belmont, A. S. & Straight, A. F. In vivo visualization of chromosomes using lac operator-repressor binding. Trends Cell Biol. 8, 121–124 (1998).

    Article  CAS  PubMed  Google Scholar 

  203. Lucas, J. S., Zhang, Y., Dudko, O. K. & Murre, C. 3D trajectories adopted by coding and regulatory DNA elements: first-passage times for genomic interactions. Cell 158, 339–352 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Maass, P. G. et al. Spatiotemporal allele organization by allele-specific CRISPR live-cell imaging (SNP-CLING). Nat. Struct. Mol. Biol. 25, 176–184 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Wang, H. et al. CRISPR-mediated live imaging of genome editing and transcription. Science 365, 1301–1305 (2019).

    Article  CAS  PubMed  Google Scholar 

  207. Geng, Y. & Pertsinidis, A. CAS-LiveFISH: simple and versatile imaging of genomic loci in live mammalian cells and early pre-implantation embryos. Preprint at bioRxiv https://doi.org/10.1101/2020.08.25.265306 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  208. Neguembor, M. V. et al. (Po)STAC (polycistronic SunTAg modified CRISPR) enables live-cell and fixed-cell super-resolution imaging of multiple genes. Nucleic Acids Res. 46, e30 (2018).

    Article  PubMed  CAS  Google Scholar 

  209. Chu, L. et al. The 3D topography of mitotic chromosomes. Mol. Cell 79, 902–916.e6 (2020).

    Article  PubMed  CAS  Google Scholar 

  210. Liu, X. et al. In situ capture of chromatin interactions by biotinylated dCas9. Cell 170, 1028–1043 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Tsui, C. et al. dCas9-targeted locus-specific protein isolation method identifies histone gene regulators. Proc. Natl Acad. Sci. USA 115, 2734–2741 (2018).

    Article  CAS  Google Scholar 

  212. Myers, S. A. et al. Discovery of proteins associated with a predefined genomic locus via dCas9–APEX-mediated proximity labeling. Nat. Methods 15, 437–439 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  214. Gómez-Marín, 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  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  216. Monahan, K., Horta, A. & Lomvardas, S. LHX2- and LDB1-mediated trans interactions regulate olfactory receptor choice. Nature 565, 448–453 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. 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 

  218. Bhattacharyya, S., Chandra, V., Vijayanand, P. & Ay, F. Identification of significant chromatin contacts from HiChIP data by FitHiChIP. Nat. Commun. 10, 1–14 (2019).

    Article  CAS  Google Scholar 

  219. Takei, Y. et al. Global architecture of the nucleus in single cells by DNA seqFISH+ and multiplexed immunofluorescence. Preprint at bioRxiv https://doi.org/10.1101/2020.11.29.403055 (2020).

    Article  Google Scholar 

  220. 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 

  221. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Vakoc, C. R. et al. Proximity among distant regulatory elements at the β-globin locus requires GATA-1 and FOG-1. Mol. Cell 17, 453–462 (2005).

    Article  CAS  PubMed  Google Scholar 

  224. Splinter, E. & De Laat, W. The complex transcription regulatory landscape of our genome: control in three dimensions. EMBO J. 30, 4345–4355 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Zhao, Z. et al. Circular chromosome conformation capture (4C) uncovers extensive networks of epigenetically regulated intra- and interchromosomal interactions. Nat. Genet. 38, 1341–1347 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  228. Apostolou, E. et al. Genome-wide chromatin interactions of the nanog locus in pluripotency, differentiation, and reprogramming. Cell Stem Cell 12, 699–712 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Van De Werken, H. J. G. et al. in Methods in Enzymology Vol. 513 (eds Wu, C. & Allis, D) 89–112 (Academic, 2012).

  230. Bonev, B. & Cavalli, G. Organization and function of the 3D genome. Nat. Rev. Genet. 17, 661–678 (2016).

    Article  CAS  PubMed  Google Scholar 

  231. Y, G. H. et al. Highly rearranged chromosomes reveal uncoupling between genome topology and gene expression. Nat. Genet. 51, 1272–1282 (2019).

    Article  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  233. Ogiyama, Y., Schuettengruber, B., Papadopoulos, G. L., Chang, J.-M. & Cavalli, G. Polycomb-dependent chromatin looping contributes to gene silencing during Drosophila development. Mol. Cell 71, 73–88 (2018).

    Article  CAS  PubMed  Google Scholar 

  234. Flavahan, W. A. et al. Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs. Nature 575, 229–233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Tarjan, D. R., Flavahan, W. A. & Bernstein, B. E. Epigenome editing strategies for the functional annotation of CTCF insulators. Nat. Commun. 10, 4258 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Kim, J. H. et al. LADL: light-activated dynamic looping for endogenous gene expression control. Nat. Methods 16, 633–639 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Wang, H. et al. CRISPR-mediated programmable 3D genome positioning and nuclear organization. Cell 175, 1405–1417 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Di Stefano for critical reading of the Computational analysis and modelling section, B. Schuttengrueber and F. Bantignies for input on figures, and Q. Szabo for help with and input on Fig. 4. I.J. was supported by an European Molecular Biology Organization (EMBO) Long-Term Fellowship (ALTF 559-2018) and the Laboratory of Excellence EpiGenMed. Research in the G.C. laboratory is supported by grants from the European Research Council (Advanced Grant 3DEpi, under grant agreement No 788972), the European Union’s Horizon 2020 research and innovation programme (MuG, under grant agreement No 676556 and ChromDesign, under the Martie Sklodowska-Curie grant agreement No 813327), the Agence Nationale de la Recherche (ANR-15-CE12-0006 EpiDevoMath), the Fondation pour la Recherche Médicale (DEI20151234396), the MSDAVENIR foundation (Project GENE-IGH), the INSERM and the French National Cancer Institute (INCa).

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Giacomo Cavalli.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks Zhijun Duan, Bing Ren, Juan Vaquerizas, who co-reviewed with Elizabeth Ing-Simmons, and the other, anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

First principles

Basic building blocks of knowledge that cannot be deduced from any other preposition used for mathematical modelling of polymer behaviours.

Loop-extrusion model

A model suggesting that motor protein complexes such as cohesin or condensin form around chromatin and use the energy of ATP to slide through it while extruding the intervening region.

Tyramide signal amplification

(TSA). A method enabling sensitive detection of low-abundance molecules in fluorescent immunocytochemistry applications.

Multiway contacts

Chromatin contacts involving more than two chromatin fragments.

Nuclear speckles

Nuclear foci enriched in pre-mRNA splicing factors.

Polycomb

An evolutionarily conserved group of proteins involved in the regulation of a large group of developmental (and other) genes.

Cajal bodies

Nuclear bodies 0.3–1 μm in size that contain RNAs and proteins and are involved in RNA metabolism-related processes.

PML bodies

Nuclear bodies 0.1–1 μm in size that contain many components, including the promyelocytic leukaemia protein (PML), and are frequently localized near Cajal bodies.

Lamina-associated domains

Chromosome domains associated with the nuclear lamina in the 3D nuclear space.

Airy diffraction pattern

A diffused circle surrounded by rings of decreasing intensity generated when a laser passes through a circular opening.

Point spread function

The response of an imaging system to a point object. If the object is below the microscope resolution, it will appear larger than it really is.

Sub-diffractive point spread function

A point spread function of smaller size than that generated by diffraction-limited systems.

Dendrimer crosslinking

A procedure in which formaldehyde crosslinking can be followed or replaced by crosslinking with dendrimers, which are highly ordered, branched polymeric molecules of different sizes.

Diffraction limit

The points where two airy patterns are too close to be distinguishable.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jerkovic´, I., Cavalli, G. Understanding 3D genome organization by multidisciplinary methods. Nat Rev Mol Cell Biol 22, 511–528 (2021). https://doi.org/10.1038/s41580-021-00362-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-021-00362-w

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