The 3D genome and its impacts on human health and disease
Siqi Wang, Zhengyu Luo, Weiguang Liu, Tengfei Hu, Zhongying Zhao, Michael G. Rosenfeld, Xiaoyuan Song
The 3D genome and its impacts on human health and disease
Eukaryotic genomes are highly compacted in the cell nucleus. Two loci separated by a long linear distance can be brought into proximity in space through DNA-binding proteins and RNAs, which contributes profoundly to the regulation of gene expression. Recent technology advances have enabled the development and application of the chromosome conformation capture (3C) technique and a host of 3C-based methods that enable genome-scale investigations into changes in chromatin high-order structures during diverse physiological processes and diseases. In this review, we introduce 3C-based technologies and discuss how they can be utilized to glean insights into the impacts of three-dimensional (3D) genome organization in normal physiological and disease processes.
3D genome / 3C and 3C-based method / 3D genome changes in health and diseases
[1] |
Ou HD, Phan S, Deerinck TJ, et al. ChromEMT: visualizing 3D chromatin structure and compaction in interphase and mitotic cells. Science 2017;357:eaag0025.
CrossRef
Google scholar
|
[2] |
Flyamer IM, Gassler J, Imakaev M, et al. Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition. Nature 2017;544:110–114.
CrossRef
Google scholar
|
[3] |
Ke Y, Xu Y, Chen X, et al. 3D Chromatin structures of mature gametes and structural reprogramming during mammalian embryogenesis. Cell 2017;170:367–381.e20.
CrossRef
Google scholar
|
[4] |
Rosa-Garrido M, Chapski DJ, Schmitt AD, et al. High-resolution mapping of chromatin conformation in cardiac myocytes reveals structural remodeling of the epigenome in heart failure. Circulation 2017;136:1613–25.
CrossRef
Google scholar
|
[5] |
Nozaki T, Imai R, Tanbo M, et al. Dynamic organization of chromatin domains revealed by super-resolution live-cell imaging. Mol Cell 2017;67:282–293.e7.
CrossRef
Google scholar
|
[6] |
Du Z, Zheng H, Huang B, et al. Allelic reprogramming of 3D chromatin architecture during early mammalian development. Nature 2017;547:232–5.
CrossRef
Google scholar
|
[7] |
Zhang H, Emerson DJ, Gilgenast TG, et al. Chromatin structure dynamics during the mitosis-to-G1 phase transition. Nature 2019;576:158–62.
CrossRef
Google scholar
|
[8] |
Zheng H, Xie W. The role of 3D genome organization in development and cell differentiation. Nat Rev Mol Cell Biol 2019;20:535–50.
CrossRef
Google scholar
|
[9] |
Luo Z, Wang X, Jiang H, et al. Reorganized 3D genome structures support transcriptional regulation in mouse spermatogenesis. iScience 2020;23:101034.
CrossRef
Google scholar
|
[10] |
Luo Z, Hu T, Jiang H, et al. Rearrangement of macronucleus chromosomes correspond to TAD-like structures of micronucleus chromosomes in Tetrahymena thermophila. Genome Res 2020;30:406–14.
CrossRef
Google scholar
|
[11] |
Li D, He M, Tang Q, et al. Comparative 3D genome architecture in vertebrates. BMC Biol 2022;20:99.
CrossRef
Google scholar
|
[12] |
Di Stefano M, Nützmann HW. Modeling the 3D genome of plants. Nucleus 2021;12:65–81.
CrossRef
Google scholar
|
[13] |
Lieberman-Aiden E, van Berkum NL, Williams L, et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 2009;326:289–93.
CrossRef
Google scholar
|
[14] |
Cremer T, Cremer C. Chromosome territories, nuclear architecture and gene regulation in mammalian cells. Nat Rev Genet 2001;2:292–301.
CrossRef
Google scholar
|
[15] |
Dixon JR, Selvaraj S, Yue F, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 2012;485:376–80.
CrossRef
Google scholar
|
[16] |
Kempfer R, Pombo A. Methods for mapping 3D chromosome architecture. Nat Rev Genet 2020;21:207–26.
CrossRef
Google scholar
|
[17] |
Rao SS, Huntley MH, Durand NC, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 2014;159:1665–80.
CrossRef
Google scholar
|
[18] |
Schoenfelder S, Fraser P. Long-range enhancer–promoter contacts in gene expression control. Nat Rev Genet 2019;20:437–55.
CrossRef
Google scholar
|
[19] |
Talukder A, Hu H, Li X. An intriguing characteristic of enhancer-promoter interactions. BMC Genom 2021;22:163.
CrossRef
Google scholar
|
[20] |
Gacita AM, Fullenkamp DE, Ohiri J, et al. Genetic variation in enhancers modifies cardiomyopathy gene expression and progression. Circulation 2021;143:1302–16.
CrossRef
Google scholar
|
[21] |
Schwarzer W, Abdennur N, Goloborodko A, et al. Two independent modes of chromatin organization revealed by cohesin removal. Nature 2017;551:51–6.
CrossRef
Google scholar
|
[22] |
Fudenberg G, Imakaev M, Lu C, et al. Formation of chromosomal domains by loop extrusion. Cell Rep 2016;15:2038–49.
CrossRef
Google scholar
|
[23] |
Davidson IF, Bauer B, Goetz D, et al. DNA loop extrusion by human cohesin. Science 2019;366:1338–45.
CrossRef
Google scholar
|
[24] |
Davidson IF, Peters JM. Genome folding through loop extrusion by SMC complexes. Nat Rev Mol Cell Biol 2021;22:445–64.
CrossRef
Google scholar
|
[25] |
Mifsud B, Tavares-Cadete F, Young AN, et al. Mapping long-range promoter contacts in human cells with high-resolution capture Hi-C. Nat Genet 2015;47:598–606.
CrossRef
Google scholar
|
[26] |
Mora A, Sandve GK, Gabrielsen OS, et al. In the loop: promoter-enhancer interactions and bioinformatics. Brief Bioinform 2016;17:980–95.
CrossRef
Google scholar
|
[27] |
Weintraub AS, Li CH, Zamudio AV, et al. YY1 Is a Structural regulator of enhancer-promoter loops. Cell 2017;171:1573–1588.e28.
CrossRef
Google scholar
|
[28] |
Sun F, Sun T, Kronenberg M, et al. The Pol II preinitiation complex (PIC) influences Mediator binding but not promoter-enhancer looping. Genes Dev 2021;35:1175–89.
CrossRef
Google scholar
|
[29] |
Wang R, Chen F, Chen Q, et al. MyoD is a 3D genome structure organizer for muscle cell identity. Nat Commun 2022;13:205.
CrossRef
Google scholar
|
[30] |
Bonev B, Mendelson Cohen N, Szabo Q, et al. Multiscale 3D genome rewiring during mouse neural development. Cell 2017;171:557–572.e24.
CrossRef
Google scholar
|
[31] |
Yin Y, Lu JY, Zhang X, et al. U1 snRNP regulates chromatin retention of noncoding RNAs. Nature 2020;580:147–50.
CrossRef
Google scholar
|
[32] |
Hacisuleyman E, Goff LA, Trapnell C, et al. Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre. Nat Struct Mol Biol 2014;21:198–206.
CrossRef
Google scholar
|
[33] |
Korostowski L, Sedlak N, Engel N. The Kcnq1ot1 long non-coding RNA affects chromatin conformation and expression of Kcnq1, but does not regulate its imprinting in the developing heart. PLoS Genet 2012;8:e1002956.
CrossRef
Google scholar
|
[34] |
Fanucchi S, Fok ET, Dalla E, et al. Immune genes are primed for robust transcription by proximal long noncoding RNAs located in nuclear compartments. Nat Genet 2019;51:138–50.
CrossRef
Google scholar
|
[35] |
Tachiwana H, Yamamoto T, Saitoh N. Gene regulation by non-coding RNAs in the 3D genome architecture. Curr Opin Genet Dev 2020;61:69–74.
CrossRef
Google scholar
|
[36] |
Wang XQ, Dostie J. Reciprocal regulation of chromatin state and architecture by HOTAIRM1 contributes to temporal collinear HOXA gene activation. Nucleic Acids Res 2017;45:1091–104.
CrossRef
Google scholar
|
[37] |
Luo H, Zhu G, Eshelman MA, et al. HOTTIP-dependent R-loop formation regulates CTCF boundary activity and TAD integrity in leukemia. Mol Cell 2022;82:833–51.e11.
CrossRef
Google scholar
|
[38] |
Cai Z, Cao C, Ji L, et al. RIC-seq for global in situ profiling of RNA-RNA spatial interactions. Nature 2020;582:432–7.
CrossRef
Google scholar
|
[39] |
Cao J, Luo Z, Cheng Q, et al. Three-dimensional regulation of transcription. Protein Cell 2015;6:241–53.
CrossRef
Google scholar
|
[40] |
Quinodoz S, Guttman M. Long noncoding RNAs: an emerging link between gene regulation and nuclear organization. Trends Cell Biol 2014;24:651–63.
CrossRef
Google scholar
|
[41] |
Bauer M, Vidal E, Zorita E, et al. Chromosome compartments on the inactive X guide TAD formation independently of transcription during X-reactivation. Nat Commun 2021;12:3499.
CrossRef
Google scholar
|
[42] |
Gibson BA, Doolittle LK, Schneider MWG, et al. Organization of chromatin by intrinsic and regulated phase separation. Cell 2019;179:470–84.e21.
CrossRef
Google scholar
|
[43] |
Wang J, Yu H, Ma Q, et al. Phase separation of OCT4 controls TAD reorganization to promote cell fate transitions. Cell Stem Cell 2021;28:1868–83.e11.
CrossRef
Google scholar
|
[44] |
Huo X, Ji L, Zhang Y, et al. The nuclear matrix protein SAFB cooperates with major satellite RNAs to stabilize heterochromatin architecture partially through phase separation. Mol Cell 2020;77:368–83.e7.
CrossRef
Google scholar
|
[45] |
Zhang H, Ji X, Li P, et al. Liquid-liquid phase separation in biology: mechanisms, physiological functions and human diseases. Sci China Life Sci 2020;63:953–85.
CrossRef
Google scholar
|
[46] |
Fang K, Wang J, Liu L, et al. Mapping nucleosome and chromatin architectures: a survey of computational methods. Comput Struct Biotechnol J 2022;20:3955–62.
CrossRef
Google scholar
|
[47] |
Knoch TA. Simulation of different three-dimensional polymer models of interphase chromosomes compared to experiments–an evaluation and review framework of the 3D genome organization. Semin Cell Dev Biol 2019;90:19–42.
CrossRef
Google scholar
|
[48] |
Dekker J, Rippe K, Dekker M, et al. Capturing chromosome conformation. Science 2002;295:1306–11.
CrossRef
Google scholar
|
[49] |
Zhao Z, Tavoosidana G, Sjölinder M, et al. Circular chromosome conformation capture (4C) uncovers extensive networks of epigenetically regulated intra- and interchromosomal interactions. Nat Genet 2006;38:1341–7.
CrossRef
Google scholar
|
[50] |
Simonis M, Klous P, Splinter E, et al. Nuclear organization of active and inactive chromatin domains uncovered by chromosome conformation capture-on-chip (4C). Nat Genet 2006;38:1348–54.
CrossRef
Google scholar
|
[51] |
Dostie J, Richmond TA, Arnaout RA, et al. Chromosome Conformation Capture Carbon Copy (5C): a massively parallel solution for mapping interactions between genomic elements. Genome Res 2006;16:1299–309.
CrossRef
Google scholar
|
[52] |
Harismendy O, Notani D, Song X, et al. 9p21 DNA variants associated with coronary artery disease impair interferon-γ signalling response. Nature 2011;470:264–8.
CrossRef
Google scholar
|
[53] |
Fullwood MJ, Liu MH, Pan YF, et al. An oestrogen-receptor-alpha-bound human chromatin interactome. Nature 2009;462:58–64.
CrossRef
Google scholar
|
[54] |
Fang R, Yu M, Li G, et al. Mapping of long-range chromatin interactions by proximity ligation-assisted ChIP-seq. Cell Res 2016;26:1345–8.
CrossRef
Google scholar
|
[55] |
Mumbach MR, Rubin AJ, Flynn RA, et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat Methods 2016;13:919–22.
CrossRef
Google scholar
|
[56] |
Hsieh TH, Weiner A, Lajoie B, et al. Mapping nucleosome resolution chromosome folding in yeast by micro-C. Cell 2015;162:108–19.
CrossRef
Google scholar
|
[57] |
Lai B, Tang Q, Jin W, et al. Trac-looping measures genome structure and chromatin accessibility. Nat Methods 2018;15:741–7.
CrossRef
Google scholar
|
[58] |
Li T, Jia L, Cao Y, et al. OCEAN-C: mapping hubs of open chromatin interactions across the genome reveals gene regulatory networks. Genome Biol 2018;19:54.
CrossRef
Google scholar
|
[59] |
Wei X, Xiang Y, Peters DT, et al. HiCAR is a robust and sensitive method to analyze open-chromatin-associated genome organization. Mol Cell 2022;82:1225–38.e6.
CrossRef
Google scholar
|
[60] |
Luo Z, Zhang R, Hu T, et al. NicE-C efficiently reveals open chromatin- associated chromosome interactions at high resolution. Genome Res 2022;32:534–44.
CrossRef
Google scholar
|
[61] |
Liang Z, Li G, Wang Z, et al. BL-Hi-C is an efficient and sensitive approach for capturing structural and regulatory chromatin interactions. Nat Commun 2017;8:1622.
CrossRef
Google scholar
|
[62] |
Lin D, Hong P, Zhang S, et al. Digestion-ligation-only Hi-C is an efficient and cost-effective method for chromosome conformation capture. Nat Genet 2018;50:754–63.
CrossRef
Google scholar
|
[63] |
Niu L, Shen W, Huang Y, et al. Amplification-free library preparation with SAFE Hi-C uses ligation products for deep sequencing to improve traditional Hi-C analysis. Commun Biol 2019;2:267.
CrossRef
Google scholar
|
[64] |
Ma W, Ay F, Lee C, et al. Fine-scale chromatin interaction maps reveal the cis-regulatory landscape of human lincRNA genes. Nat Methods 2015;12:71–8.
CrossRef
Google scholar
|
[65] |
Quinodoz SA, Ollikainen N, Tabak B, et al. Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 2018;174:744–57.e24.
CrossRef
Google scholar
|
[66] |
Beagrie RA, Scialdone A, Schueler M, et al. Complex multi-enhancer contacts captured by genome architecture mapping. Nature 2017;543:519–24.
CrossRef
Google scholar
|
[67] |
Zheng M, Tian SZ, Capurso D, et al. Multiplex chromatin interactions with single-molecule precision. Nature 2019;566:558–62.
CrossRef
Google scholar
|
[68] |
Hell SW, Wichmann J. Breaking the diffraction resolution limit by stimulated emission: stimulated-emission-depletion fluorescence microscopy. Opt Lett 1994;19:780–2.
CrossRef
Google scholar
|
[69] |
Feng H, Wang X, Xu Z, et al. Super-resolution fluorescence microscopy for single cell imaging. Adv Exp Med Biol 2018;1068:59–71.
CrossRef
Google scholar
|
[70] |
Bates M, Jones SA, Zhuang X. Stochastic optical reconstruction microscopy (STORM): a method for superresolution fluorescence imaging. Cold Spring Harb Protoc 2013;2013:498–520.
CrossRef
Google scholar
|
[71] |
Gwosch KC, Pape JK, Balzarotti F, et al. MINFLUX nanoscopy delivers 3D multicolor nanometer resolution in cells. Nat Methods 2020;17:217–24.
CrossRef
Google scholar
|
[72] |
Nguyen HQ, Chattoraj S, Castillo D, et al. 3D mapping and accelerated super-resolution imaging of the human genome using in situ sequencing. Nat Methods 2020;17:822–32.
CrossRef
Google scholar
|
[73] |
Wang S, Su J-H, Beliveau BJ, et al. Spatial organization of chromatin domains and compartments in single chromosomes. Science 2016;353:598–602.
CrossRef
Google scholar
|
[74] |
Nguyen HQ, Chattoraj S, Castillo D, et al. 3D mapping and accelerated super-resolution imaging of the human genome using in situ sequencing. Nat Methods 2020;17:822–32.
CrossRef
Google scholar
|
[75] |
Xu J, Ma H, Jin J, et al. Super-resolution imaging of higher-order chromatin structures at different epigenomic states in single mammalian cells. Cell Rep 2018;24:873–82.
CrossRef
Google scholar
|
[76] |
Su JH, Zheng P, Kinrot SS, et al. Genome-scale imaging of the 3D organization and transcriptional activity of chromatin. Cell 2020;182:1641–59.e26.
CrossRef
Google scholar
|
[77] |
Zhuang X. Spatially resolved single-cell genomics and transcriptomics by imaging. Nat Methods 2021;18:18–22.
CrossRef
Google scholar
|
[78] |
Tan L, Xing D, Chang C-H, et al. Three-dimensional genome structures of single diploid human cells. Science 2018;361:924–8.
CrossRef
Google scholar
|
[79] |
Nagano T, Lubling Y, Stevens TJ, et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 2013;502:59–64.
CrossRef
Google scholar
|
[80] |
Lin D, Xu W, Hong P, et al. Decoding the spatial chromatin organization and dynamic epigenetic landscapes of macrophage cells during differentiation and immune activation. Nat Commun 2022;13:5857.
CrossRef
Google scholar
|
[81] |
Arrastia MV, Jachowicz JW, Ollikainen N, et al. Single-cell measurement of higher-order 3D genome organization with scSPRITE. Nat Biotechnol 2022;40:64–73.
CrossRef
Google scholar
|
[82] |
Tan L, Ma W, Wu H, et al. Changes in genome architecture and transcriptional dynamics progress independently of sensory experience during post-natal brain development. Cell 2021;184:741–58.e17.
CrossRef
Google scholar
|
[83] |
Lee DS, Luo C, Zhou J, et al. Simultaneous profiling of 3D genome structure and DNA methylation in single human cells. Nat Methods 2019;16:999–1006.
CrossRef
Google scholar
|
[84] |
Li G, Liu Y, Zhang Y, et al. Joint profiling of DNA methylation and chromatin architecture in single cells. Nat Methods 2019;16:991–3.
CrossRef
Google scholar
|
[85] |
Xia C, Babcock HP, Moffitt JR, et al. Multiplexed detection of RNA using MERFISH and branched DNA amplification. Sci Rep 2019;9:7721.
CrossRef
Google scholar
|
[86] |
Lincoln R, Bossi ML, Remmel M, et al. A general design of caging-group-free photoactivatable fluorophores for live-cell nanoscopy. Nat Chem 2022;14:1013–20.
CrossRef
Google scholar
|
[87] |
Hu M, Deng K, Selvaraj S, et al. HiCNorm: removing biases in Hi-C data via Poisson regression. Bioinformatics 2012;28:3131–3.
CrossRef
Google scholar
|
[88] |
Imakaev M, Fudenberg G, McCord RP, et al. Iterative correction of Hi-C data reveals hallmarks of chromosome organization. Nat Methods 2012;9:999–1003.
CrossRef
Google scholar
|
[89] |
Wu HJ, Michor F. A computational strategy to adjust for copy number in tumor Hi-C data. Bioinformatics 2016;32:3695–701.
CrossRef
Google scholar
|
[90] |
Stansfield JC, Cresswell KG, Vladimirov VI, et al. HiCcompare: an R-package for joint normalization and comparison of HI-C datasets. BMC Bioinf 2018;19:279.
CrossRef
Google scholar
|
[91] |
Spill YG, Castillo D, Vidal E, et al. Binless normalization of Hi-C data provides significant interaction and difference detection independent of resolution. Nat Commun 2019;10:1938.
CrossRef
Google scholar
|
[92] |
Norton HK, Emerson DJ, Huang H, et al. Detecting hierarchical genome folding with network modularity. Nat Methods 2018;15:119–22.
CrossRef
Google scholar
|
[93] |
Chen F, Li G, Zhang MQ, et al. HiCDB: a sensitive and robust method for detecting contact domain boundaries. Nucleic Acids Res 2018;46:11239–50.
CrossRef
Google scholar
|
[94] |
Li A, Yin X, Xu B, et al. Decoding topologically associating domains with ultra-low resolution Hi-C data by graph structural entropy. Nat Commun 2018;9:3265.
CrossRef
Google scholar
|
[95] |
Li X, Zeng G, Li A, et al. DeTOKI identifies and characterizes the dynamics of chromatin TAD-like domains in a single cell. Genome Biol 2021;22:217.
CrossRef
Google scholar
|
[96] |
Durand NC, Shamim MS, Machol I, et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C Experiments. Cell Syst 2016;3:95–8.
CrossRef
Google scholar
|
[97] |
Weinreb C, Raphael BJ. Identification of hierarchical chromatin domains. Bioinformatics 2016;32:1601–9.
CrossRef
Google scholar
|
[98] |
Oluwadare O, Cheng J. ClusterTAD: an unsupervised machine learning approach to detecting topologically associated domains of chromosomes from Hi-C data. BMC Bioinf 2017;18:480.
CrossRef
Google scholar
|
[99] |
Zhang YW, Chen L, Li SC. Detecting TAD-like domains from RNA-associated interactions. Nucleic Acids Res 2022;50:e88–8.
CrossRef
Google scholar
|
[100] |
Kaul A, Bhattacharyya S, Ay F. Identifying statistically significant chromatin contacts from Hi-C data with FitHiC2. Nat Protoc 2020;15:991–1012.
CrossRef
Google scholar
|
[101] |
Zhang Y, Blanchette M. Reference panel guided topological structure annotation of Hi-C data. Nat Commun 2022;13:7426.
CrossRef
Google scholar
|
[102] |
Luzhin AV, Golov AK, Gavrilov AA, et al. LASCA: loop and significant contact annotation pipeline. Sci Rep 2021;11:6361.
CrossRef
Google scholar
|
[103] |
Cao Y, Chen Z, Chen X, et al. Accurate loop calling for 3D genomic data with cLoops. bioRxiv 2020;36:666–75.
CrossRef
Google scholar
|
[104] |
Li D, Purushotham D, Harrison JK, et al. WashU Epigenome Browser update 2022. Nucleic Acids Res 2022;50:W774–81.
CrossRef
Google scholar
|
[105] |
Kim K, Jang I, Kim M, et al. 3DIV update for 2021: a comprehensive resource of 3D genome and 3D cancer genome. Nucleic Acids Res 2021;49:D38–46.
CrossRef
Google scholar
|
[106] |
Robinson JT, Turner D, Durand NC, et al. Juicebox.js provides a cloud-based visualization system for Hi-C data. Cell Syst 2018;6:256–8.e1.
CrossRef
Google scholar
|
[107] |
Martin JS, Xu Z, Reiner AP, et al. HUGIn: Hi-C unifying genomic interrogator. Bioinformatics 2017;33:3793–5.
CrossRef
Google scholar
|
[108] |
Li R, Liu Y, Li T, et al. 3Disease Browser: a web server for integrating 3D genome and disease-associated chromosome rearrangement data. Sci Rep 2016;6:34651.
CrossRef
Google scholar
|
[109] |
Wang X, Luan Y, Yue FE. A deep-learning framework for detecting a full range of structural variations from bulk and single-cell contact maps. Sci Adv 2022;8:eabn9215.
CrossRef
Google scholar
|
[110] |
Poszewiecka B, Pienkowski VM, Nowosad K, et al. TADeus2: a web server facilitating the clinical diagnosis by pathogenicity assessment of structural variations disarranging 3D chromatin structure. Nucleic Acids Res 2022;50:W744–52.
CrossRef
Google scholar
|
[111] |
Chakraborty A, Ay F. Identification of copy number variations and translocations in cancer cells from Hi-C data. Bioinformatics 2018;34:338–45.
CrossRef
Google scholar
|
[112] |
Requena F, Abdallah HH, García A, et al. CNVxplorer: a web tool to assist clinical interpretation of CNVs in rare disease patients. Nucleic Acids Res 2021;49:W93–W103.
CrossRef
Google scholar
|
[113] |
Wang S, Lee S, Chu C, et al. HiNT: a computational method for detecting copy number variations and translocations from Hi-C data. Genome Biol 2020;21:73.
CrossRef
Google scholar
|
[114] |
Zhang R, Zhou T, Ma J. Multiscale and integrative single-cell Hi-C analysis with Higashi. Nat Biotechnol 2022;40:254–61.
CrossRef
Google scholar
|
[115] |
Forcato M, Bicciato S. Computational analysis of Hi-C data. Methods Mol Biol 2021;2157:103–25.
CrossRef
Google scholar
|
[116] |
Nicoletti C. Methods for the differential analysis of Hi-C data. Methods Mol Biol 2022;2301:61–95.
CrossRef
Google scholar
|
[117] |
Naumova N, Imakaev M, Fudenberg G, et al. Organization of the mitotic chromosome. Science 2013;342:948–53.
CrossRef
Google scholar
|
[118] |
Oomen ME, Hansen AS, Liu Y, et al. CTCF sites display cell cycle-dependent dynamics in factor binding and nucleosome positioning. Genome Res 2019;29:236–49.
CrossRef
Google scholar
|
[119] |
Gibcus JH, Samejima K, Goloborodko A, et al. A pathway for mitotic chromosome formation. Science 2018;359:eaao6135.
CrossRef
Google scholar
|
[120] |
Wang Y, Wang H, Zhang Y, et al. Reprogramming of meiotic chromatin architecture during spermatogenesis. Mol Cell 2019;73:547–61. e6.
CrossRef
Google scholar
|
[121] |
Vara C, Paytuví-Gallart A, Cuartero Y, et al. Three-dimensional genomic structure and cohesin occupancy correlate with transcriptional activity during spermatogenesis. Cell Rep 2019;28:352–67. e9.
CrossRef
Google scholar
|
[122] |
Patel L, Kang R, Rosenberg SC, et al. Dynamic reorganization of the genome shapes the recombination landscape in meiotic prophase. Nat Struct Mol Biol 2019;26:164–74.
CrossRef
Google scholar
|
[123] |
Alavattam KG, Maezawa S, Sakashita A, et al. Attenuated chromatin compartmentalization in meiosis and its maturation in sperm development. Nat Struct Mol Biol 2019;26:175–84.
CrossRef
Google scholar
|
[124] |
Zuo W, Chen G, Gao Z, et al. Stage-resolved Hi-C analyses reveal meiotic chromosome organizational features influencing homolog alignment. Nat Commun 2021;12:5827.
CrossRef
Google scholar
|
[125] |
Du Z, Zheng H, Kawamura YK, et al. Polycomb group proteins regulate chromatin architecture in mouse oocytes and early embryos. Mol Cell 2020;77:825–39.e7.
CrossRef
Google scholar
|
[126] |
Chen X, Ke Y, Wu K, et al. Key role for CTCF in establishing chromatin structure in human embryos. Nature 2019;576:306–10.
CrossRef
Google scholar
|
[127] |
Hug CB, Vaquerizas JM. The birth of the 3D genome during early embryonic development. Trends Genet 2018;34:903–14.
CrossRef
Google scholar
|
[128] |
Ke Y, Xu Y, Chen X, et al. 3D Chromatin structures of mature gametes and structural reprogramming during mammalian embryogenesis. Cell 2017;170:367–81.e20.
CrossRef
Google scholar
|
[129] |
Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res 1965;37:614–36.
CrossRef
Google scholar
|
[130] |
Criscione SW, De Cecco M, Siranosian B, et al. Reorganization of chromosome architecture in replicative cellular senescence. Sci Adv 2016;2:e1500882.
CrossRef
Google scholar
|
[131] |
Suzuki M, Boothman DA. Stress-induced premature senescence (SIPS)—influence of SIPS on radiotherapy. J Radiat Res 2008;49:105–12.
CrossRef
Google scholar
|
[132] |
Chandra T, Ewels PA, Schoenfelder S, et al. Global reorganization of the nuclear landscape in senescent cells. Cell Rep 2015;10:471–83.
CrossRef
Google scholar
|
[133] |
Sati S, Bonev B, Szabo Q, et al. 4D genome rewiring during oncogene-induced and replicative senescence. Mol Cell 2020;78:522–38.e9.
CrossRef
Google scholar
|
[134] |
Zhang B, Long Q, Wu S, et al. KDM4 Orchestrates epigenomic remodeling of senescent cells and potentiates the senescence-associated secretory phenotype. Nat Aging 2021;1:454–72.
CrossRef
Google scholar
|
[135] |
Zhang X, Liu X, Du Z, et al. The loss of heterochromatin is associated with multiscale three-dimensional genome reorganization and aberrant transcription during cellular senescence. Genome Res 2021;31:1121–35.
CrossRef
Google scholar
|
[136] |
Liu Z, Ji Q, Ren J, et al. Large-scale chromatin reorganization reactivates placenta-specific genes that drive cellular aging. Dev Cell 2022;57:1347–68.e12.
CrossRef
Google scholar
|
[137] |
Deng L, Ren R, Liu Z, et al. Stabilizing heterochromatin by DGCR8 alleviates senescence and osteoarthritis. Nat Commun 2019;10:3329.
CrossRef
Google scholar
|
[138] |
Hu H, Ji Q, Song M, et al. ZKSCAN3 counteracts cellular senescence by stabilizing heterochromatin. Nucleic Acids Res 2020;48:6001–18.
CrossRef
Google scholar
|
[139] |
Aaltonen LA, Abascal F, Adams DJ, et al. Pan-cancer analysis of whole genomes. Nature 2020;578:82–93.
CrossRef
Google scholar
|
[140] |
Barutcu AR, Lajoie BR, McCord RP, et al. Chromatin interaction analysis reveals changes in small chromosome and telomere clustering between epithelial and breast cancer cells. Genome Biol 2015;16:214.
CrossRef
Google scholar
|
[141] |
Johnstone SE, Reyes A, Qi Y, et al. Large-scale topological changes restrain malignant progression in colorectal cancer. Cell 2020;182:1474–89.e23.
CrossRef
Google scholar
|
[142] |
Zhang, L, Song T, Yao M, et al. Integrative identification by Hi-C revealed distinct advanced structural variations in Lung Adenocarcinoma tissue. bioRxiv 2020.
CrossRef
Google scholar
|
[143] |
Martin RS, Das P, Marques RD, et al. Alterations in chromosome spatial compartmentalization classify prostate cancer progression. bioRxiv 2021.
CrossRef
Google scholar
|
[144] |
Hnisz D, Weintraub AS, Day DS, et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science (New York, N.Y.) 2016;351;1454–8.
CrossRef
Google scholar
|
[145] |
Flavahan WA, Drier Y, Liau BB, et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 2016;529:110–4.
CrossRef
Google scholar
|
[146] |
Northcott PA, Lee C, Zichner T, et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 2014;511:428–34.
CrossRef
Google scholar
|
[147] |
Ooi WF, Nargund AM, Lim KJ, et al. Integrated paired-end enhancer profiling and whole-genome sequencing reveals recurrent CCNE1 and IGF2 enhancer hijacking in primary gastric adenocarcinoma. Gut 2020;69:1039–52.
CrossRef
Google scholar
|
[148] |
Xu J, Song F, Lyu H, et al. Subtype-specific 3D genome alteration in acute myeloid leukaemia. Nature 2022;611:387–98.
CrossRef
Google scholar
|
[149] |
Weischenfeldt J, Dubash T, Drainas AP, et al. Pan-cancer analysis of somatic copy-number alterations implicates IRS4 and IGF2 in enhancer hijacking. Nat Genet 2017;49:65–74.
CrossRef
Google scholar
|
[150] |
Engreitz JM, Agarwala V, Mirny LA. Three-dimensional genome architecture influences partner selection for chromosomal translocations in human disease. PLoS One 2012;7:e44196.
CrossRef
Google scholar
|
[151] |
Mani RS, Tomlins SA, Callahan K, et al. Induced chromosomal proximity and gene fusions in prostate cancer. Science 2009;326:1230.
CrossRef
Google scholar
|
[152] |
Roix JJ, McQueen PG, Munson PJ, et al. Spatial proximity of translocation-prone gene loci in human lymphomas. Nat Genet 2003;34:287–91.
CrossRef
Google scholar
|
[153] |
Liu Y, Guo B, Aguilera-Jimenez E, et al. Chromatin looping shapes KLF5-dependent transcriptional programs in human epithelial cancers. Cancer Res 2020;80:5464–77.
CrossRef
Google scholar
|
[154] |
Luo Z, Rhie SK, Lay FD, et al. A prostate cancer risk element functions as a repressive loop that regulates HOXA13. Cell Rep 2017;21:1411–7.
CrossRef
Google scholar
|
[155] |
Iyyanki T, Zhang B, Wang Q, et al. Subtype-associated epigenomic landscape and 3D genome structure in bladder cancer. Genome Biol 2021;22:105.
CrossRef
Google scholar
|
[156] |
Chen B, Ma Y, Bi J, et al. Regulation network of colorectal-cancer-specific enhancers in the progression of colorectal cancer. Int J Mol Sci 2021;22:8337.
CrossRef
Google scholar
|
[157] |
Harewood L, Kishore K, Eldridge MD, et al. Hi-C as a tool for precise detection and characterisation of chromosomal rearrangements and copy number variation in human tumours. Genome Biol 2017;18:125.
CrossRef
Google scholar
|
[158] |
Dixon JR, Xu J, Dileep V, et al. Integrative detection and analysis of structural variation in cancer genomes. Nat Genet 2018;50:1388–98.
CrossRef
Google scholar
|
[159] |
Akdemir KC, Le VT, Chandran S, et al; PCAWG Structural Variation Working Group. Disruption of chromatin folding domains by somatic genomic rearrangements in human cancer. Nat Genet 2020;52:294–305.
CrossRef
Google scholar
|
[160] |
Stephens PJ, Greenman CD, Fu B, et al. Massive genomic rear-rangement acquired in a single catastrophic event during cancer development. Cell 2011;144:27–40.
CrossRef
Google scholar
|
[161] |
Korbel JO, Campbell PJ. Criteria for inference of chromothripsis in cancer genomes. Cell 2013;152:1226–36.
CrossRef
Google scholar
|
[162] |
Lupiáñez DG, Kraft K, Heinrich V, et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 2015;161:1012–25.
CrossRef
Google scholar
|
[163] |
Franke M, Ibrahim DM, Andrey G, et al. Formation of new chromatin domains determines pathogenicity of genomic duplications. Nature 2016;538:265–9.
CrossRef
Google scholar
|
[164] |
Lettice LA, Daniels S, Sweeney E, et al. Enhancer-adoption as a mechanism of human developmental disease. Hum Mutat 2011;32:1492–9.
CrossRef
Google scholar
|
[165] |
Ibn-Salem J, Köhler S, Love MI, et al. Deletions of chromosomal regulatory boundaries are associated with congenital disease. Genome Biol 2014;15:423.
CrossRef
Google scholar
|
[166] |
Takagi M, Sasaki G, Mitsui T, et al. A 2.0 Mb microdeletion in proximal chromosome 14q12, involving regulatory elements of FOXG1, with the coding region of FOXG1 being unaffected, results in severe developmental delay, microcephaly, and hypoplasia of the corpus callosum. Eur J Med Genet 2013;56:526–8.
CrossRef
Google scholar
|
[167] |
Milunsky JM, Maher TA, Zhao G, et al. TFAP2A mutations result in branchio-oculo-facial syndrome. Am J Hum Genet 2008;82:1171–7.
CrossRef
Google scholar
|
[168] |
Laugsch M, Bartusel M, Rehimi R, et al. Modeling the pathological long-range regulatory effects of human structural variation with patient-specific hiPSCs. Cell Stem Cell 2019;24:736–52.e12.
CrossRef
Google scholar
|
[169] |
Sun JH, Zhou L, Emerson DJ, et al. Disease-associated short tandem repeats co-localize with chromatin domain boundaries. Cell 2018;175:224–38.e15.
CrossRef
Google scholar
|
[170] |
Giorgio E, Robyr D, Spielmann M, et al. A large genomic deletion leads to enhancer adoption by the lamin B1 gene: a second path to autosomal dominant adult-onset demyelinating leukodystrophy (ADLD). Hum Mol Genet 2015;24:3143–54.
CrossRef
Google scholar
|
[171] |
Redin C, Brand H, Collins RL, et al. The genomic landscape of balanced cytogenetic abnormalities associated with human congenital anomalies. Nat Genet 2017;49:36–45.
CrossRef
Google scholar
|
[172] |
Razin SV, Gavrilov AA, Iarovaia OV. Modification of nuclear compartments and the 3D genome in the course of a viral infection. Acta Naturae 2020;12:34–46.
CrossRef
Google scholar
|
[173] |
Ho JSY, Mok BW, Campisi L, et al. TOP1 inhibition therapy protects against SARS-CoV-2-induced lethal inflammation. Cell 2021;184:2618–32.e17.
CrossRef
Google scholar
|
[174] |
Wang R, Lee J-H, Xiong F, et al. SARS-CoV-2 Restructures the host chromatin architecture. bioRxiv 2021.
CrossRef
Google scholar
|
[175] |
Moreau P, Cournac A, Palumbo GA, et al. Tridimensional infiltration of DNA viruses into the host genome shows preferential contact with active chromatin. Nat Commun 2018;9:4268.
CrossRef
Google scholar
|
[176] |
Yang B, Li B, Jia L, et al. 3D landscape of Hepatitis B virus interactions with human chromatins. Cell Discov 2020;6:95.
CrossRef
Google scholar
|
[177] |
Tang D, Zhao H, Wu Y, et al. Transcriptionally inactive hepatitis B virus episome DNA preferentially resides in the vicinity of chromosome 19 in 3D host genome upon infection. Cell Rep 2021;35:109288.
CrossRef
Google scholar
|
[178] |
Hensel KO, Cantner F, Bangert F, et al. Episomal HBV persistence within transcribed host nuclear chromatin compartments involves HBx. Epigenetics Chromatin 2018;11:34.
CrossRef
Google scholar
|
[179] |
Shen C, Feng X, Mao T, et al. Yin-Yang 1 and HBx protein activate HBV transcription by mediating the spatial interaction of cccDNA minichromosome with cellular chromosome 19p13.11. Emerg Microbes Infect 2020;9:2455–64.
CrossRef
Google scholar
|
[180] |
Péneau C, Imbeaud S, La Bella T, et al. Hepatitis B virus integrations promote local and distant oncogenic driver alterations in hepatocellular carcinoma. Gut 2022;71:616–26.
CrossRef
Google scholar
|
[181] |
Okabe A, Huang KK, Matsusaka K, et al. Cross-species chromatin interactions drive transcriptional rewiring in Epstein-Barr virus-positive gastric adenocarcinoma. Nat Genet 2020;52:919–30.
CrossRef
Google scholar
|
[182] |
Moquin SA, Thomas S, Whalen S, et al. The epstein-barr virus episome maneuvers between nuclear chromatin compartments during reactivation. J Virol 2018;92:e01413–17.
CrossRef
Google scholar
|
[183] |
Wang L, Laing J, Yan B, et al. Epstein-barr virus episome physically interacts with active regions of the host genome in lymphoblastoid cells. J Virol 2020;94:e01390–20.
CrossRef
Google scholar
|
[184] |
Kim KD, Tanizawa H, De Leo A, et al. Epigenetic specifications of host chromosome docking sites for latent Epstein-Barr virus. Nat Commun 2020;11:877.
CrossRef
Google scholar
|
[185] |
Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–49.
CrossRef
Google scholar
|
[186] |
Shen C, Liu Y, Shi S, et al. Long-distance interaction of the integrated HPV fragment with MYC gene and 8q24.22 region upregulating the allele-specific MYC expression in HeLa cells. Int J Cancer 2017;141:540–8.
CrossRef
Google scholar
|
[187] |
Peter M, Stransky N, Couturier J, et al. Frequent genomic structural alterations at HPV insertion sites in cervical carcinoma. J Pathol 2010;221:320–30.
CrossRef
Google scholar
|
[188] |
Groves IJ, Drane ELA, Michalski M, et al. Three-dimensional interactions between integrated HPV genomes and cellular chromatin dysregulate host gene expression in early cervical carcinogenesis. bioRxiv 2021.
CrossRef
Google scholar
|
[189] |
Cao C, Hong P, Huang X, et al. HPV-CCDC106 integration alters local chromosome architecture and hijacks an enhancer by three-dimensional genome structure remodeling in cervical cancer. J Genet Genomics 2020;47:437–50.
CrossRef
Google scholar
|
[190] |
Adeel MM, Jiang H, Arega Y, et al. Structural variations of the 3D genome architecture in cervical cancer development. Front Cell Dev Biol 2021;9:706375.
CrossRef
Google scholar
|
[191] |
Chu Z, Gu L, Hu Y, et al. STAG2 regulates interferon signaling in melanoma via enhancer loop reprogramming. Nat Commun 2022;13:1859.
CrossRef
Google scholar
|
[192] |
Dahiya R, Hu Q, Ly P. Mechanistic origins of diverse genome rear-rangements in cancer. Semin Cell Dev Biol 2022;123:100–9.
CrossRef
Google scholar
|
[193] |
Zakharova VV, Magnitov MD, Del Maestro L, et al. SETDB1 fuels the lung cancer phenotype by modulating epigenome, 3D genome organization and chromatin mechanical properties. Nucleic Acids Res 2022;50:4389–413.
CrossRef
Google scholar
|
[194] |
Bintu B, Mateo LJ, Su J-H, et al. Super-resolution chromatin tracing reveals domains and cooperative interactions in single cells. Science 2018;362:eaau1783.
CrossRef
Google scholar
|
[195] |
Bakir-Gungor B, Sezerman OU. A new methodology to associate SNPs with human diseases according to their pathway related context. PLoS One 2011;6:e26277.
CrossRef
Google scholar
|
[196] |
Maurano MT, Humbert R, Rynes E, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 2012;337:1190–5.
CrossRef
Google scholar
|
[197] |
Wang H, Yang B, Cai X, et al. Hepatocellular carcinoma risk variant modulates lncRNA HLA-DQB1-AS1 expression via a long-range enhancer-promoter interaction. Carcinogenesis 2021;42:1347–56.
CrossRef
Google scholar
|
[198] |
Baur B, Lee D-I, Haag J, et al. Deciphering the role of 3D genome organization in breast cancer susceptibility. Front Genet 2021;12:788318.
CrossRef
Google scholar
|
[199] |
Quinodoz SA, Guttman M. Essential roles for RNA in shaping nuclear organization. Cold Spring Harb Perspect Biol 2022;14:a039719.
CrossRef
Google scholar
|
/
〈 | 〉 |