Browse > Article
http://dx.doi.org/10.5808/GI.2017.15.4.114

CTCF, Cohesin, and Chromatin in Human Cancer  

Song, Sang-Hyun (Cancer Genomics Research Laboratory, Cancer Research Institute, Seoul National University)
Kim, Tae-You (Cancer Genomics Research Laboratory, Cancer Research Institute, Seoul National University)
Abstract
It is becoming increasingly clear that eukaryotic genomes are subjected to higher-order chromatin organization by the CCCTC-binding factor/cohesin complex. Their dynamic interactions in three dimensions within the nucleus regulate gene transcription by changing the chromatin architecture. Such spatial genomic organization is functionally important for the spatial disposition of chromosomes to control cell fate during development and differentiation. Thus, the dysregulation of proper long-range chromatin interactions may influence the development of tumorigenesis and cancer progression.
Keywords
cohesins; CTCF; gene expression regulation; higher-order chromatin structure; topologically associated domain;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Nagano T, Lubling Y, Stevens TJ, Schoenfelder S, Yaffe E, Dean W, et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 2013;502:59-64.   DOI
2 Osborne CS, Chakalova L, Mitchell JA, Horton A, Wood AL, Bolland DJ, et al. Myc dynamically and preferentially relocates to a transcription factory occupied by Igh. PLoS Biol 2007;5:e192.   DOI
3 Fudenberg G, Getz G, Meyerson M, Mirny LA. High order chromatin architecture shapes the landscape of chromosomal alterations in cancer. Nat Biotechnol 2011;29:1109-1113.   DOI
4 De S, Michor F. DNA replication timing and long-range DNA interactions predict mutational landscapes of cancer genomes. Nat Biotechnol 2011;29:1103-1108.   DOI
5 Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, et al. The landscape of somatic copy-number alteration across human cancers. Nature 2010;463:899-905.   DOI
6 Alabert C, Groth A. Chromatin replication and epigenome maintenance. Nat Rev Mol Cell Biol 2012;13:153-167.
7 Albertson DG. Gene amplification in cancer. Trends Genet 2006;22:447-455.   DOI
8 Hsu PY, Hsu HK, Lan X, Juan L, Yan PS, Labanowska J, et al. Amplification of distant estrogen response elements deregulates target genes associated with tamoxifen resistance in breast cancer. Cancer Cell 2013;24:197-212.   DOI
9 Donohoe ME, Zhang LF, Xu N, Shi Y, Lee JT. Identification of a Ctcf cofactor, Yy1, for the X chromosome binary switch. Mol Cell 2007;25:43-56.   DOI
10 Ishihara K, Oshimura M, Nakao M. CTCF-dependent chromatin insulator is linked to epigenetic remodeling. Mol Cell 2006;23:733-742.   DOI
11 de Wit E, Vos ES, Holwerda SJ, Valdes-Quezada C, Verstegen MJ, Teunissen H, et al. CTCF binding polarity determines chromatin looping. Mol Cell 2015;60:676-684.   DOI
12 de Wit E, Bouwman BA, Zhu Y, Klous P, Splinter E, Verstegen MJ, et al. The pluripotent genome in three dimensions is shaped around pluripotency factors. Nature 2013;501:227-231.   DOI
13 Schoenfelder S, Sexton T, Chakalova L, Cope NF, Horton A, Andrews S, et al. Preferential associations between co-regulated genes reveal a transcriptional interactome in erythroid cells. Nat Genet 2010;42:53-61.   DOI
14 Kagey MH, Newman JJ, Bilodeau S, Zhan Y, Orlando DA, van Berkum NL, et al. Mediator and cohesin connect gene expression and chromatin architecture. Nature 2010;467:430-435.   DOI
15 Phillips-Cremins JE, Sauria ME, Sanyal A, Gerasimova TI, Lajoie BR, Bell JS, et al. Architectural protein subclasses shape 3D organization of genomes during lineage commitment. Cell 2013;153:1281-1295.   DOI
16 Hnisz D, Day DS, Young RA. Insulated neighborhoods: structural and functional units of mammalian gene control. Cell 2016;167:1188-1200.   DOI
17 Merkenschlager M, Nora EP. CTCF and cohesin in genome folding and transcriptional gene regulation. Annu Rev Genomics Hum Genet 2016;17:17-43.   DOI
18 Dorsett D, Strom L. The ancient and evolving roles of cohesin in gene expression and DNA repair. Curr Biol 2012;22:R240-R250.   DOI
19 Zhang Y, McCord RP, Ho YJ, Lajoie BR, Hildebrand DG, Simon AC, et al. Spatial organization of the mouse genome and its role in recurrent chromosomal translocations. Cell 2012;148:908-921.   DOI
20 Misteli T, Soutoglou E. The emerging role of nuclear architecture in DNA repair and genome maintenance. Nat Rev Mol Cell Biol 2009;10:243-254.
21 Awad TA, Bigler J, Ulmer JE, Hu YJ, Moore JM, Lutz M, et al. Negative transcriptional regulation mediated by thyroid hormone response element 144 requires binding of the multivalent factor CTCF to a novel target DNA sequence. J Biol Chem 1999;274:27092-27098.   DOI
22 Rubio ED, Reiss DJ, Welcsh PL, Disteche CM, Filippova GN, Baliga NS, et al. CTCF physically links cohesin to chromatin. Proc Natl Acad Sci U S A 2008;105:8309-8314.   DOI
23 Defossez PA, Kelly KF, Filion GJ, Perez-Torrado R, Magdinier F, Menoni H, et al. The human enhancer blocker CTC-binding factor interacts with the transcription factor Kaiso. J Biol Chem 2005;280:43017-43023.   DOI
24 Xiao T, Wallace J, Felsenfeld G. Specific sites in the C terminus of CTCF interact with the SA2 subunit of the cohesin complex and are required for cohesin-dependent insulation activity. Mol Cell Biol 2011;31:2174-2183.   DOI
25 Parelho V, Hadjur S, Spivakov M, Leleu M, Sauer S, Gregson HC, et al. Cohesins functionally associate with CTCF on mammalian chromosome arms. Cell 2008;132:422-433.   DOI
26 Stedman W, Kang H, Lin S, Kissil JL, Bartolomei MS, Lieberman PM. Cohesins localize with CTCF at the KSHV latency control region and at cellular c-myc and H19/Igf2 insulators. EMBO J 2008;27:654-666.   DOI
27 Nasmyth K, Haering CH. Cohesin: its roles and mechanisms. Annu Rev Genet 2009;43:525-558.   DOI
28 Merkenschlager M, Odom DT. CTCF and cohesin: linking gene regulatory elements with their targets. Cell 2013;152:1285-1297.   DOI
29 Tonkin ET, Wang TJ, Lisgo S, Bamshad MJ, Strachan T. NIPBL, encoding a homolog of fungal Scc2-type sister chromatid cohesion proteins and fly Nipped-B, is mutated in Cornelia de Lange syndrome. Nat Genet 2004;36:636-641.   DOI
30 Bell AC, West AG, Felsenfeld G. The protein CTCF is required for the enhancer blocking activity of vertebrate insulators. Cell 1999;98:387-396.   DOI
31 Kanduri C, Pant V, Loukinov D, Pugacheva E, Qi CF, Wolffe A, et al. Functional association of CTCF with the insulator upstream of the H19 gene is parent of origin-specific and methylation-sensitive. Curr Biol 2000;10:853-856.   DOI
32 Bell AC, Felsenfeld G. Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature 2000;405:482-485.   DOI
33 Chen X, Xu H, Yuan P, Fang F, Huss M, Vega VB, et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008;133: 1106-1117.   DOI
34 Bulger M, Schubeler D, Bender MA, Hamilton J, Farrell CM, Hardison RC, et al. A complex chromatin landscape revealed by patterns of nuclease sensitivity and histone modification within the mouse beta-globin locus. Mol Cell Biol 2003;23:5234-5244.   DOI
35 Barber TD, McManus K, Yuen KW, Reis M, Parmigiani G, Shen D, et al. Chromatid cohesion defects may underlie chromosome instability in human colorectal cancers. Proc Natl Acad Sci U S A 2008;105:3443-3448.   DOI
36 Marson A, Levine SS, Cole MF, Frampton GM, Brambrink T, Johnstone S, et al. Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 2008;134:521-533.   DOI
37 Farrell CM, West AG, Felsenfeld G. Conserved CTCF insulator elements flank the mouse and human beta-globin loci. Mol Cell Biol 2002;22:3820-3831.   DOI
38 Hou C, Zhao H, Tanimoto K, Dean A. CTCF-dependent enhancer-blocking by alternative chromatin loop formation. Proc Natl Acad Sci U S A 2008;105:20398-20403.   DOI
39 Hou C, Dale R, Dean A. Cell type specificity of chromatin organization mediated by CTCF and cohesin. Proc Natl Acad Sci U S A 2010;107:3651-3656.   DOI
40 Murrell A, Heeson S, Reik W. Interaction between differentially methylated regions partitions the imprinted genes Igf2 and H19 into parent-specific chromatin loops. Nat Genet 2004;36:889-893.   DOI
41 Cancer Genome Atlas Research Network, Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013;368:2059-2074.   DOI
42 Kon A, Shih LY, Minamino M, Sanada M, Shiraishi Y, Nagata Y, et al. Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms. Nat Genet 2013;45:1232-1237.   DOI
43 Solomon DA, Kim T, Diaz-Martinez LA, Fair J, Elkahloun AG, Harris BT, et al. Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science 2011;333:1039-1043.   DOI
44 Taatjes DJ. The human Mediator complex: a versatile, genome-wide regulator of transcription. Trends Biochem Sci 2010;35:315-322.   DOI
45 Wendt KS, Yoshida K, Itoh T, Bando M, Koch B, Schirghuber E, et al. Cohesin mediates transcriptional insulation by CCCTCbinding factor. Nature 2008;451:796-801.   DOI
46 Nativio R, Wendt KS, Ito Y, Huddleston JE, Uribe-Lewis S, Woodfine K, et al. Cohesin is required for higher-order chromatin conformation at the imprinted IGF2-H19 locus. PLoS Genet 2009;5:e1000739.   DOI
47 Hadjur S, Williams LM, Ryan NK, Cobb BS, Sexton T, Fraser P, et al. Cohesins form chromosomal cis-interactions at the developmentally regulated IFNG locus. Nature 2009;460:410-413.   DOI
48 Yan J, Enge M, Whitington T, Dave K, Liu J, Sur I, et al. Transcription factor binding in human cells occurs in dense clusters formed around cohesin anchor sites. Cell 2013;154:801-813.   DOI
49 Phillips JE, Corces VG. CTCF: master weaver of the genome. Cell 2009;137:1194-1211.   DOI
50 Mehta GD, Kumar R, Srivastava S, Ghosh SK. Cohesin: functions beyond sister chromatid cohesion. FEBS Lett 2013;587:2299-2312.   DOI
51 Seitan VC, Faure AJ, Zhan Y, McCord RP, Lajoie BR, Ing-Simmons E, et al. Cohesin-based chromatin interactions enable regulated gene expression within preexisting architectural compartments. Genome Res 2013;23:2066-2077.   DOI
52 Schaaf CA, Kwak H, Koenig A, Misulovin Z, Gohara DW, Watson A, et al. Genome-wide control of RNA polymerase II activity by cohesin. PLoS Genet 2013;9:e1003382.   DOI
53 Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 2012;485:376-380.   DOI
54 Nora EP, Lajoie BR, Schulz EG, Giorgetti L, Okamoto I, Servant N, et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 2012;485:381-385.   DOI
55 Ong CT, Corces VG. CTCF: an architectural protein bridging genome topology and function. Nat Rev Genet 2014;15:234-246.   DOI
56 Wijchers PJ, de Laat W. Genome organization influences partner selection for chromosomal rearrangements. Trends Genet 2011;27:63-71.   DOI
57 Hark AT, Schoenherr CJ, Katz DJ, Ingram RS, Levorse JM, Tilghman SM. CTCF mediates methylation-sensitive enhancer-blocking activity at the H19/Igf2 locus. Nature 2000;405:486-489.   DOI
58 Kurukuti S, Tiwari VK, Tavoosidana G, Pugacheva E, Murrell A, Zhao Z, et al. CTCF binding at the H19 imprinting control region mediates maternally inherited higher-order chromatin conformation to restrict enhancer access to Igf2. Proc Natl Acad Sci U S A 2006;103:10684-10689.   DOI
59 Valadez-Graham V, Razin SV, Recillas-Targa F. CTCF-dependent enhancer blockers at the upstream region of the chicken alpha-globin gene domain. Nucleic Acids Res 2004;32:1354-1362.   DOI
60 Degner SC, Verma-Gaur J, Wong TP, Bossen C, Iverson GM, Torkamani A, et al. CCCTC-binding factor (CTCF) and cohesin influence the genomic architecture of the Igh locus and antisense transcription in pro-B cells. Proc Natl Acad Sci U S A 2011;108:9566-9571.   DOI
61 Pant V, Kurukuti S, Pugacheva E, Shamsuddin S, Mariano P, Renkawitz R, et al. Mutation of a single CTCF target site within the H19 imprinting control region leads to loss of Igf2 imprinting and complex patterns of de novo methylation upon maternal inheritance. Mol Cell Biol 2004;24:3497-3504.   DOI
62 Dorsett D, Merkenschlager M. Cohesin at active genes: a unifying theme for cohesin and gene expression from model organisms to humans. Curr Opin Cell Biol 2013;25:327-333.   DOI
63 Roix JJ, McQueen PG, Munson PJ, Parada LA, Misteli T. Spatial proximity of translocation-prone gene loci in human lymphomas. Nat Genet 2003;34:287-291.   DOI
64 Wang H, Maurano MT, Qu H, Varley KE, Gertz J, Pauli F, et al. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res 2012;22:1680-1688.   DOI
65 Wang XQ, Dostie J. Chromosome folding and its regulation in health and disease. Curr Opin Genet Dev 2017;43:23-30.   DOI
66 Rodriguez C, Borgel J, Court F, Cathala G, Forne T, Piette J. CTCF is a DNA methylation-sensitive positive regulator of the INK/ARF locus. Biochem Biophys Res Commun 2010;392:129-134.   DOI
67 Witcher M, Emerson BM. Epigenetic silencing of the p16(INK4a) tumor suppressor is associated with loss of CTCF binding and a chromatin boundary. Mol Cell 2009;34:271-284.   DOI
68 Lai AY, Fatemi M, Dhasarathy A, Malone C, Sobol SE, Geigerman C, et al. DNA methylation prevents CTCF-mediated silencing of the oncogene BCL6 in B cell lymphomas. J Exp Med 2010;207:1939-1950.   DOI
69 Ribeiro de Almeida C, Stadhouders R, de Bruijn MJ, Bergen IM, Thongjuea S, Lenhard B, et al. The DNA-binding protein CTCF limits proximal Vkappa recombination and restricts kappa enhancer interactions to the immunoglobulin kappa light chain locus. Immunity 2011;35:501-513.   DOI
70 Seitan VC, Hao B, Tachibana-Konwalski K, Lavagnolli T, Mira-Bontenbal H, Brown KE, et al. A role for cohesin in T-cell-receptor rearrangement and thymocyte differentiation. Nature 2011;476:467-471.   DOI
71 Ling JQ, Li T, Hu JF, Vu TH, Chen HL, Qiu XW, et al. CTCF mediates interchromosomal colocalization between Igf2/H19 and Wsb1/Nf1. Science 2006;312:269-272.   DOI
72 Haering CH, Farcas AM, Arumugam P, Metson J, Nasmyth K. The cohesin ring concatenates sister DNA molecules. Nature 2008;454:297-301.   DOI
73 Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 2014;159:1665-1680.   DOI
74 Guo Y, Xu Q, Canzio D, Shou J, Li J, Gorkin DU, et al. CRISPR inversion of CTCF sites alters genome topology and enhancer/promoter function. Cell 2015;162:900-910.   DOI
75 Canela A, Maman Y, Jung S, Wong N, Callen E, Day A, et al. Genome organization drives chromosome fragility. Cell 2017; 170:507-521.e518.   DOI
76 Gregor A, Oti M, Kouwenhoven EN, Hoyer J, Sticht H, Ekici AB, et al. De novo mutations in the genome organizer CTCF cause intellectual disability. Am J Hum Genet 2013;93:124-131.   DOI
77 Le May N, Fradin D, Iltis I, Bougneres P, Egly JM. XPG and XPF endonucleases trigger chromatin looping and DNA demethylation for accurate expression of activated genes. Mol Cell 2012;47:622-632.   DOI
78 Kang JY, Song SH, Yun J, Jeon MS, Kim HP, Han SW, et al. Disruption of CTCF/cohesin-mediated high-order chromatin structures by DNA methylation downregulates PTGS2 expression. Oncogene 2015;34:5677-5684.   DOI
79 Mullighan CG, Zhang J, Kasper LH, Lerach S, Payne-Turner D, Phillips LA, et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 2011;471:235-239.   DOI
80 Dolnik A, Engelmann JC, Scharfenberger-Schmeer M, Mauch J, Kelkenberg-Schade S, Haldemann B, et al. Commonly altered genomic regions in acute myeloid leukemia are enriched for somatic mutations involved in chromatin remodeling and splicing. Blood 2012;120:e83-e92.   DOI
81 Katainen R, Dave K, Pitkanen E, Palin K, Kivioja T, Valimaki N, et al. CTCF/cohesin-binding sites are frequently mutated in cancer. Nat Genet 2015;47:818-821.   DOI
82 Kong A, Steinthorsdottir V, Masson G, Thorleifsson G, Sulem P, Besenbacher S, et al. Parental origin of sequence variants associated with complex diseases. Nature 2009;462:868-874.   DOI
83 Ji X, Dadon DB, Powell BE, Fan ZP, Borges-Rivera D, Shachar S, et al. 3D chromosome regulatory landscape of human pluripotent cells. Cell Stem Cell 2016;18:262-275.   DOI
84 Tang Z, Luo OJ, Li X, Zheng M, Zhu JJ, Szalaj P, et al. CTCF-mediated human 3D genome architecture reveals chromatin topology for transcription. Cell 2015;163:1611-1627.   DOI
85 Flavahan WA, Drier Y, Liau BB, Gillespie SM, Venteicher AS, Stemmer-Rachamimov AO, et al. Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 2016;529:110-114.   DOI
86 Yun J, Song SH, Kang JY, Park J, Kim HP, Han SW, et al. Reduced cohesin destabilizes high-level gene amplification by disrupting pre-replication complex bindings in human cancers with chromosomal instability. Nucleic Acids Res 2016;44:558-572.   DOI
87 Deardorff MA, Wilde JJ, Albrecht M, Dickinson E, Tennstedt S, Braunholz D, et al. RAD21 mutations cause a human cohesinopathy. Am J Hum Genet 2012;90:1014-1027.   DOI
88 Yun J, Song SH, Kim HP, Han SW, Yi EC, Kim TY. Dynamic cohesin-mediated chromatin architecture controls epithelialmesenchymal plasticity in cancer. EMBO Rep 2016;17:1343-1359.   DOI
89 Hnisz D, Weintraub AS, Day DS, Valton AL, Bak RO, Li CH, et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science 2016;351:1454-1458.   DOI
90 Lupianez DG, Kraft K, Heinrich V, Krawitz P, Brancati F, Klopocki E, et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 2015;161:1012-1025.   DOI
91 Pott S, Lieb JD. What are super-enhancers? Nat Genet 2015;47:8-12.   DOI
92 Amabile A, Migliara A, Capasso P, Biffi M, Cittaro D, Naldini L, et al. Inheritable silencing of endogenous genes by hit-andrun targeted epigenetic editing. Cell 2016;167:219-232.e214.   DOI
93 Liu XS, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, et al. Editing DNA methylation in the mammalian genome. Cell 2016;167:233-247.e217.   DOI
94 Simeonov DR, Gowen BG, Boontanrart M, Roth TL, Gagnon JD, Mumbach MR, et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 2017;549: 111-115.   DOI