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.

  • Article
  • Published:

YY1-controlled regulatory connectivity and transcription are influenced by the cell cycle

Abstract

Few transcription factors have been examined for their direct roles in physically connecting enhancers and promoters. Here acute degradation of Yin Yang 1 (YY1) in erythroid cells revealed its requirement for the maintenance of numerous enhancer–promoter loops, but not compartments or domains. Despite its reported ability to interact with cohesin, the formation of YY1-dependent enhancer–promoter loops does not involve stalling of cohesin-mediated loop extrusion. Integrating mitosis-to-G1-phase dynamics, we observed partial retention of YY1 on mitotic chromatin, predominantly at gene promoters, followed by rapid rebinding during mitotic exit, coinciding with enhancer–promoter loop establishment. YY1 degradation during the mitosis-to-G1-phase interval revealed a set of enhancer–promoter loops that require YY1 for establishment during G1-phase entry but not for maintenance in interphase, suggesting that cell cycle stage influences YY1’s architectural function. Thus, as revealed here for YY1, chromatin architectural functions of transcription factors can vary in their interplay with CTCF and cohesin as well as by cell cycle stage.

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: YY1 binding is necessary for the maintenance of CRE loops in asynchronous cells.
Fig. 2: YY1-anchored loops can form independently of CTCF and cohesin.
Fig. 3: Promoter-proximal YY1 binding drives YY1-dependent transcription in asynchronous cells.
Fig. 4: YY1 is partially retained on mitotic chromatin and rapidly recruited to CRE loop anchors during G1 entry.
Fig. 5: YY1 recruitment dynamics and requirements for CRE loop establishment during the mitosis-to-G1 transition.
Fig. 6: Transcriptional kinetics in G1 entry after depletion of YY1.
Fig. 7: Cell-cycle-dependent YY1 requirements for CRE loop formation.
Fig. 8: Model for CRE loop establishment and maintenance following mitosis.

Similar content being viewed by others

Data availability

The raw and processed Micro-C, ChIP–seq, RNA-seq and TT-seq data generated from this study are deposited in the Gene Expression Omnibus (GEO) database under accession GSE247254. External CTCF–AID Hi-C data from a previous study24 are available at GSE168251. External SMC3–AID Hi-C data from a previous study84 are available at GSE228402. External mitosis-to-G1 Hi-C and ChIP–seq data from a previous study67 are available at GSE129997. Source data are provided with this paper.

Code availability

The code used in this study is available at https://github.com/jclqrs/Lam_2024_Code and https://zenodo.org/doi/10.5281/zenodo.11992254.

References

  1. Furlong, E. E. M. & Levine, M. Developmental enhancers and chromosome topology. Science 361, 1341–1345 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zheng, H. & Xie, W. The role of 3D genome organization in development and cell differentiation. Nat. Rev. Mol. Cell Biol. 20, 535–550 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Zhang, H. & Blobel, G. A. Genome folding dynamics during the M-to-G1-phase transition. Curr. Opin. Genet Dev. 80, 102036 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Davidson, I. F. et al. DNA loop extrusion by human cohesin. Science 366, 1338–1345 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Kim, Y., Shi, Z., Zhang, H., Finkelstein, I. J. & Yu, H. Human cohesin compacts DNA by loop extrusion. Science 366, 1345–1349 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  8. 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 

  9. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sanborn, A. L. et al. Chromatin extrusion explains key features of loop and domain formation in wild-type and engineered genomes. Proc. Natl Acad. Sci. USA 112, E6456–E6465 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Davidson, I. F. & Peters, J.-M. Genome folding through loop extrusion by SMC complexes. Nat. Rev. Mol. Cell. Biol. 22, 445–464 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nasmyth, K. Disseminating the genome: joining, resolving, and separating sister chromatids during mitosis and meiosis. Annu. Rev. Genet. 35, 673–745 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Riggs, A. D. DNA methylation and late replication probably aid cell memory, and type I DNA reeling could aid chromosome folding and enhancer function. Philos. Trans. R. Soc. Lond. B Biol. Sci. 326, 285–297 (1990).

    Article  CAS  PubMed  Google Scholar 

  15. de Wit, E. & Nora, E. P. New insights into genome folding by loop extrusion from inducible degron technologies. Nat. Rev. Genet. 24, 73–85 (2023).

    Article  CAS  PubMed  Google Scholar 

  16. Rhodes, J. D. P. et al. Cohesin disrupts polycomb-dependent chromosome interactions in embryonic stem cells. Cell Rep. 30, 820–835 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kriz, A. J., Colognori, D., Sunwoo, H., Nabet, B. & Lee, J. T. Balancing cohesin eviction and retention prevents aberrant chromosomal interactions, polycomb-mediated repression, and X-inactivation. Mol. Cell 81, 1970–1987 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hsieh, T.-H. S. et al. Enhancer–promoter interactions and transcription are largely maintained upon acute loss of CTCF, cohesin, WAPL or YY1. Nat. Genet. 54, 1919–1932 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Thiecke, M. J. et al. Cohesin-dependent and -independent mechanisms mediate chromosomal contacts between promoters and enhancers. Cell Rep. 32, 107929 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Khattabi, L. E. et al. A pliable mediator acts as a functional rather than an architectural bridge between promoters and enhancers. Cell 178, 1145–1158 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Goel, V.Y., Huseyin, M.K. & Hansen, A. S. Region Capture Micro-C reveals coalescence of enhancers and promoters into nested microcompartments. Nat. Genet. 55, 1048–1056 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang, H. et al. CTCF and transcription influence chromatin structure re-configuration after mitosis. Nat. Commun. 12, 5157 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

  26. Li, X. et al. GAGA-associated factor fosters loop formation in the Drosophila genome. Mol. Cell 83, 1519–1526 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Linares-Saldana, R. et al. BRD4 orchestrates genome folding to promote neural crest differentiation. Nat. Genet. 53, 1480–1492 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Crump, N. T. et al. BET inhibition disrupts transcription but retains enhancer–promoter contact. Nat. Commun. 12, 223 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jaeger, M. G. et al. Selective mediator dependence of cell-type-specifying transcription. Nat. Genet. 52, 719–727 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sun, F. et al. The Pol II preinitiation complex (PIC) influences mediator binding but not promoter–enhancer looping. Gene Dev. 35, 1175–1189 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Ramasamy, S. et al. The mediator complex regulates enhancer–promoter interactions. Nat. Struct. Mol. Biol. 30, 991–1000 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang, S. et al. RNA polymerase II is required for spatial chromatin reorganization following exit from mitosis. Sci. Adv. 7, eabg8205 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang, S., Übelmesser, N., Barbieri, M. & Papantonis, A. Enhancer–promoter contact formation requires RNAPII and antagonizes loop extrusion. Nat. Genet. 55, 832–840 (2023).

    Article  CAS  PubMed  Google Scholar 

  35. Jiang, Y. et al. Genome-wide analyses of chromatin interactions after the loss of Pol I, Pol II, and Pol III. Genome Biol. 21, 158 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hsieh, T.-H. S. et al. Resolving the 3D landscape of transcription-linked mammalian chromatin folding. Mol. Cell. 78, 539–553 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Barshad, G. et al. RNA polymerase II dynamics shape enhancer–promoter interactions. Nat. Genet. 55, 1370–1380 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hwang, S. S. et al. Transcription factor YY1 is essential for regulation of the Th2 cytokine locus and for Th2 cell differentiation. Proc. Natl Acad. Sci. USA 110, 276–281 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Beagan, J. A. et al. YY1 and CTCF orchestrate a 3D chromatin looping switch during early neural lineage commitment. Genome Res. 27, 1139–1152 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Liu, H. et al. Yin Yang 1 is a critical regulator of B-cell development. Gene Dev. 21, 1179–1189 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Degner, S. C. 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. USA 108, 9566–9571 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Verma-Gaur, J. et al. Noncoding transcription within the Igh distal VH region at PAIR elements affects the 3D structure of the Igh locus in pro-B cells. Proc. Natl Acad. Sci. USA 109, 17004–17009 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Mehra, P. et al. YY1 controls Eμ-3′RR DNA loop formation and immunoglobulin heavy chain class switch recombination. Blood Adv. 1, 15–20 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Perreault, A. A., Brown, J. D. & Venters, B. J. Erythropoietin regulates transcription and YY1 dynamics in a pre-established chromatin architecture. iScience 23, 101583 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Fang, S. et al. Tet inactivation disrupts YY1 binding and long-range chromatin interactions during embryonic heart development. Nat. Commun. 10, 4297 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Kwon, D. Y. et al. Neuronal Yin Yang1 in the prefrontal cortex regulates transcriptional and behavioral responses to chronic stress in mice. Nat. Commun. 13, 55 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Atlasi, Y. et al. Epigenetic modulation of a hardwired 3D chromatin landscape in two naive states of pluripotency. Nat. Cell Biol. 21, 568–578 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. Li, L. et al. YY1 interacts with guanine quadruplexes to regulate DNA looping and gene expression. Nat. Chem. Biol. 17, 161–168 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Liu, T. et al. Matrin3 mediates differentiation through stabilizing chromatin loop-domain interactions and YY1 mediated enhancer–promoter interactions. Nat. Commun. 15, 1274 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Weintraub, A. S. et al. YY1 is a structural regulator of enhancer–promoter loops. Cell 171, 1573–1588 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cubeñas-Potts, C. & Corces, V. G. Architectural proteins, transcription, and the three-dimensional organization of the genome. FEBS Lett. 589, 2923–2930 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Kim, S. & Shendure, J. Mechanisms of interplay between transcription factors and the 3D genome. Mol. Cell 76, 306–319 (2019).

    Article  CAS  PubMed  Google Scholar 

  53. Lee, J., Krivega, I., Dale, R. K. & Dean, A. The LDB1 complex co-opts CTCF for erythroid lineage-specific long-range enhancer interactions. Cell Rep. 19, 2490–2502 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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 

  55. Krivega, I. & Dean, A. LDB1-mediated enhancer looping can be established independent of mediator and cohesin. Nucleic Acids Res. 45, 8255–8268 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Deng, W. et al. Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell 149, 1233–1244 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

  59. Breda, L. et al. Forced chromatin looping raises fetal hemoglobin in adult sickle cells to higher levels than pharmacologic inducers. Blood 128, 1139–1143 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. López-Perrote, A. et al. Structure of Yin Yang 1 oligomers that cooperate with RuvBL1–RuvBL2 ATPases. J. Biol. Chem. 289, 22614–22629 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Wu, S. et al. A YY1–INO80 complex regulates genomic stability through homologous recombination–based repair. Nat. Struct. Mol. Biol. 14, 1165–1172 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lu, Z., Assumpção, A. L. F. V., Viny, A. D., Levine, R. L. & Pan, X. YY1 controls hematopoietic stem cell quiescence by repressing cohesin expression. Blood 132, 3831 (2018).

    Article  Google Scholar 

  63. Pan, X. et al. YY1 controls Igκ repertoire and B‐cell development, and localizes with condensin on the Igκ locus. EMBO J. 32, 1168–1182 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Donohoe, M. E., Zhang, L.-F., Xu, N., Shi, Y. & Lee, J. T. Identification of a Ctcf cofactor, Yy1, for the X chromosome binary switch. Mol. Cell 25, 43–56 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Pentland, I. et al. Disruption of CTCF-YY1-dependent looping of the human papillomavirus genome activates differentiation-induced viral oncogene transcription. PLoS Biol. 16, e2005752 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Guo, C. et al. Two forms of loops generate the chromatin conformation of the immunoglobulin heavy-chain gene locus. Cell 147, 332–343 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Zhang, H. et al. Chromatin structure dynamics during the mitosis-to-G1 phase transition. Nature 576, 158–162 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Naumova, N. et al. Organization of the mitotic chromosome. Science 342, 948–953 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gibcus, J. H. et al. A pathway for mitotic chromosome formation. Science 359, 6376 (2018).

    Article  Google Scholar 

  70. Pelham-Webb, B. et al. H3K27ac bookmarking promotes rapid post-mitotic activation of the pluripotent stem cell program without impacting 3D chromatin reorganization. Mol. Cell 81, 1732–1748 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Abramo, K. et al. A chromosome folding intermediate at the condensin-to-cohesin transition during telophase. Nat. Cell Biol. 21, 1393–1402 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Chervova, A., Festuccia, N., Altamirano‐Pacheco, L., Dubois, A. & Navarro, P. A gene subset requires CTCF bookmarking during the fast post‐mitotic reactivation of mouse ES cells. EMBO Rep. 24, e56075 (2023).

    Article  CAS  PubMed  Google Scholar 

  73. Donohoe, M. E. et al. Targeted disruption of mouse Yin Yang 1 transcription factor results in peri-implantation lethality. Mol. Cell. Biol. 19, 7237–7244 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Coronado, D. et al. A short G1 phase is an intrinsic determinant of naïve embryonic stem cell pluripotency. Stem Cell Res. 10, 118–131 (2013).

    Article  PubMed  Google Scholar 

  75. Weiss, M. J., Yu, C. & Orkin, S. H. Erythroid-cell-specific properties of transcription factor GATA-1 revealed by phenotypic rescue of a gene-targeted cell line. Mol. Cell. Biol. 17, 1642–1651 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Krietenstein, N. et al. Ultrastructural details of mammalian chromosome architecture. Mol. Cell 78, 554–565 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Open2C, et al. Cooltools: enabling high-resolution Hi-C analysis in Python. PLoS Comput. Biol. 20, e1012067 (2024).

    Article  PubMed Central  Google Scholar 

  78. Zuin, J. et al. Nonlinear control of transcription through enhancer–promoter interactions. Nature 604, 571–577 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Rinzema, N. J. et al. Building regulatory landscapes reveals that an enhancer can recruit cohesin to create contact domains, engage CTCF sites and activate distant genes. Nat. Struct. Mol. Biol. 29, 563–574 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Xiao, J. Y., Hafner, A. & Boettiger, A. N. How subtle changes in 3D structure can create large changes in transcription. eLife 10, e64320 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Xi, W. & Beer, M. A. Loop competition and extrusion model predicts CTCF interaction specificity. Nat. Commun. 12, 1046 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Schwalie, P. C. et al. Co-binding by YY1 identifies the transcriptionally active, highly conserved set of CTCF-bound regions in primate genomes. Genome Biol. 14, R148 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Lu, Z. et al. Yin Yang 1 regulates cohesin complex protein SMC3 in mouse hematopoietic stem cells. Blood Adv. 25, 3076–3091 (2024).

    Article  Google Scholar 

  84. Zhao, H. et al. Genome folding principles uncovered in condensin-depleted mitotic chromosomes. Nat. Genet. 56, 1213–1224 (2024).

    Article  CAS  PubMed  Google Scholar 

  85. Calderon, L. et al. Cohesin-dependence of neuronal gene expression relates to chromatin loop length. eLife 11, e76539 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kane, L. et al. Cohesin is required for long-range enhancer action at the Shh locus. Nat. Struct. Mol. Biol. 29, 891–897 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Shi, Y., Seto, E., Chang, L.-S. & Shenk, T. Transcriptional repression by YY1, a human GLI-Krüippel-related protein, and relief of repression by adenovirus E1A protein. Cell 67, 377–388 (1991).

    Article  CAS  PubMed  Google Scholar 

  88. Seto, E., Shi, Y. & Shenk, T. YY1 is an initiator sequence-binding protein that directs and activates transcription in vitro. Nature 354, 241–245 (1991).

    Article  CAS  PubMed  Google Scholar 

  89. Park, K. & Atchison, M. L. Isolation of a candidate repressor/activator, NF-E1 (YY-1, δ), that binds to the immunoglobulin kappa 3′ enhancer and the immunoglobulin heavy-chain mu E1 site. Proc. Natl Acad. Sci. USA 88, 9804–9808 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Hariharan, N., Kelley, D. E. & Perry, R. P. Delta, a transcription factor that binds to downstream elements in several polymerase II promoters, is a functionally versatile zinc finger protein. Proc. Natl Acad. Sci. USA 88, 9799–9803 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Schwalb, B. et al. TT-seq maps the human transient transcriptome. Science 352, 1225–1228 (2016).

    Article  CAS  PubMed  Google Scholar 

  92. Gregersen, L. H., Mitter, R. & Svejstrup, J. Q. Using TTchem-seq for profiling nascent transcription and measuring transcript elongation. Nat. Protoc. 15, 604–627 (2020).

    Article  CAS  PubMed  Google Scholar 

  93. Wilkinson, F. H., Park, K. & Atchison, M. L. Polycomb recruitment to DNA in vivo by the YY1 REPO domain. Proc. Natl Acad. Sci. USA 103, 19296–19301 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Shah, M., Funnell, A. P. W., Quinlan, K. G. R. & Crossley, M. Hit and run transcriptional repressors are difficult to catch in the act. BioEssays 41, e1900041 (2019).

    Article  PubMed  Google Scholar 

  95. Banigan, E. J. et al. Transcription shapes 3D chromatin organization by interacting with loop extrusion. Proc. Natl Acad. Sci. USA 120, e2210480120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ito, K. & Zaret, K. S. Maintaining transcriptional specificity through mitosis. Annu. Rev. Genom. Hum. Genet. 23, 53–71 (2022).

    Article  CAS  Google Scholar 

  97. Kadauke, S. & Blobel, G. A. Mitotic bookmarking by transcription factors. Epigenetics Chromatin 6, 6 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Rizkallah, R. & Hurt, M. M. Regulation of the transcription factor YY1 in mitosis through phosphorylation of its DNA-binding domain. Mol. Biol. Cell 20, 4766–4776 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Kassardjian, A. et al. The transcription factor YY1 is a novel substrate for aurora B kinase at G2/M transition of the cell cycle. PLoS ONE 7, e50645 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Alexander, K. E., Rizkallah, R. & Aurora, A. Phosphorylation of YY1 during mitosis inactivates its DNA binding activity. Sci. Rep. 7, 10084 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  101. Hsiung, C. C.-S. et al. Genome accessibility is widely preserved and locally modulated during mitosis. Genome Res. 25, 213–225 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kumaran, R. I. & Spector, D. L. A genetic locus targeted to the nuclear periphery in living cells maintains its transcriptional competence. J. Cell Biol. 180, 51–65 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Reddy, K. L., Zullo, J. M., Bertolino, E. & Singh, H. Transcriptional repression mediated by repositioning of genes to the nuclear lamina. Nature 452, 243–247 (2008).

    Article  CAS  PubMed  Google Scholar 

  104. Bridger, J. M., Boyle, S., Kill, I. R. & Bickmore, W. A. Re-modelling of nuclear architecture in quiescent and senescent human fibroblasts. Curr. Biol. 10, 149–152 (2000).

    Article  CAS  PubMed  Google Scholar 

  105. Mach, P. et al. Cohesin and CTCF control the dynamics of chromosome folding. Nat. Genet. 54, 1907–1918 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. 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 

  107. Alexander, J. M. et al. Live-cell imaging reveals enhancer-dependent Sox2 transcription in the absence of enhancer proximity. eLife 8, e41769 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Hsiung, C. C.-S. et al. A hyperactive transcriptional state marks genome reactivation at the mitosis–G1 transition. Gene Dev. 30, 1423–1439 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Ardakany, A. R., 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  Google Scholar 

  110. Matthey-Doret, C. et al. Computer vision for pattern detection in chromosome contact maps. Nat. Commun. 11, 5795 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Vian, L. et al. The energetics and physiological impact of cohesin extrusion. Cell 173, 1165–1178 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Liu, N. Q. et al. WAPL maintains a cohesin loading cycle to preserve cell-type-specific distal gene regulation. Nat. Genet. 53, 100–109 (2021).

    Article  CAS  PubMed  Google Scholar 

  113. Rylski, M. et al. GATA-1-mediated proliferation arrest during erythroid maturation. Mol. Cell. Biol. 23, 5031–5042 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Lam, J. jclqrs/Lam_2024_Code: v1.0.0 (v1.0.0). Zenodo 10.5281/zenodo.11992255 (2024).

  115. Langmead, B., Wilks, C., Antonescu, V. & Charles, R. Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics 35, 421–432 (2018).

    Article  PubMed Central  Google Scholar 

  116. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Danecek, P. et al. Twelve years of SAMtools and BCFtools. GigaScience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Feng, J., Liu, T., Qin, B., Zhang, Y. & Liu, X. S. Identifying ChIP–seq enrichment using MACS. Nat. Protoc. 7, 1728–1740 (2012).

    Article  CAS  PubMed  Google Scholar 

  119. Ross-Innes, C. S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Dale, R. K., Pedersen, B. S. & Quinlan, A. R. Pybedtools: a flexible Python library for manipulating genomic datasets and annotations. Bioinformatics 27, 3423–3424 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  125. Zhu, A., Ibrahim, J. G. & Love, M. I. Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences. Bioinformatics 35, 2084–2092 (2018).

    Article  PubMed Central  Google Scholar 

  126. Xu, W. et al. CoolBox: a flexible toolkit for visual analysis of genomics data. BMC Bioinformatics 22, 489 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank E. Nora, E. Joyce and members of the Blobel Laboratory for helpful discussions. The authors would also like to thank the Children’s Hospital of Philadelphia Flow Cytometry Core for technical assistance with cell sorting. This work was supported by grants T32GM007170, T32HG000046 and F30DK132824 (to J.C.L.), T32GM008216 and the Blavatnik Family Fellowship Award (to N.G.A.), R24DK106766 (to R.C.H. and G.A.B.), National Science Foundation of China (grant 32100422 to H.Z.) and R01DK054937, R01DK058044 and U01DK127405 (to G.A.B.).

Author information

Authors and Affiliations

Authors

Contributions

J.C.L. and G.A.B. conceived the study. J.C.L., N.G.A. and S.C.M. performed the Micro-C experiments. J.C.L. and S.C.M. performed cell synchronization and ChIP experiments. S.W. performed the TT-seq experiments and sequencing alignment. J.C.L. and A.H. generated the YY1–AID cell line. K.A.H. performed RT–qPCR validation. C.A.K., B.G. and R.C.H. prepared ChIP–seq and RNA-seq libraries and performed sequencing and preprocessed sequencing data. J.C.L. analyzed all datasets and interpreted the results. N.G.A. and H.Z. helped with the interpretation of results. J.C.L. and G.A.B. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Gerd A. Blobel.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Guillaume Andrey, Bas van Steensel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Compartments and domains are maintained in the absence of YY1.

a, YY1-AID cell counts for −auxin and +auxin conditions. Results are shown as mean ± s.d. (n = 3 biological replicates, two-sided independent t-test). b, Western blot for YY1 in parental cells and YY1-AID cells after an auxin time-course. c, Histogram of YY1 ChIP–seq signal at YY1 peaks for −auxin and +auxin conditions. d, ChIP–seq tracks for YY1 (Active Motif antibody), YY1 (Bethyl antibody) and H3K27ac from −auxin and +auxin conditions. e, Heatmaps of YY1 ChIP–seq signal (Bethyl antibody) and mean log2-fold change (+auxin/−auxin) centered on all YY1 peaks (n = 2 biological replicates). f, Micro-C contact versus distance curves for each biological replicate. g, Heatmap of Pearson correlations of compartment eigenvector 1 values (EV1) between biological replicates. h, Micro-C contact maps from −auxin (top) and +auxin (bottom) conditions along with tracks of compartment EV1, with positive values corresponding to A compartment and negative values corresponding to B compartment. i, Histogram of EV1 values from −auxin and +auxin contact maps (n = 25,257 bins). j, Aggregate domain plot for all domains called in the −auxin contact map, centered on upstream and downstream boundaries. k, Histogram of log2 insulation score values from −auxin and +auxin contact maps for all boundaries called across both conditions. l, Histogram of log2 insulation score values from −auxin and +auxin contact maps for the subset of boundaries that have YY1 binding within ±50 kb. m, Micro-C contact maps from −auxin (top) and +auxin (bottom) conditions, annotated with example TAD calls. Corresponding tracks show log2 insulation scores (IS).

Source data

Extended Data Fig. 2 Chromatin loop changes after YY1 depletion in asynchronous cells.

a, Venn diagram of loop calls from −auxin and +auxin contact maps. b, Bar plot of loop change counts, stratified by the condition in which the loop was called. c, Box plot of loop strengths in the −auxin contact map, stratified by loop change (two-sided Mann–Whitney U test). d, Box plot of loop strengths in the +auxin contact map, stratified by loop change (two-sided Mann–Whitney U test). e, Counts of uncategorized loops across categories of looping change and YY1 occupancy. f, Pileup plots of all H3K27ac-H3K27ac and YY1–YY1 loops detected in the −auxin condition, weakened YY1–YY1 loops and strengthened YY1–YY1 loops (1 kb resolution, ±30 kb window). g, Pileup plots of weakened YY1–YY1 loops for individual biological replicates. Loops are centered on YY1 ChIP–seq peaks (n = 555 loops, 1 kb resolution, ±30 kb window). h, Enrichment of different factor occupancy patterns in strengthened CRE loops (*padj < 0.05, two-sided Fisher’s exact test, Benjamini–Hochberg multiple testing correction).

Extended Data Fig. 3 CTCF, cohesin and LDB1 peaks remain stable upon YY1 depletion.

a, Heatmaps showing CTCF ChIP–seq signal at all CTCF peaks before and after YY1 depletion in asynchronous cells. b, Heatmaps showing RAD21 ChIP–seq signal at all RAD21 peaks before and after asynchronous YY1 depletion. c, Heatmaps showing LDB1 ChIP–seq signal at all LDB1 peaks before and after YY1 depletion in asynchronous cells. d, Heatmaps showing H3K27ac ChIP–seq signal at all H3K27ac peaks before and after YY1 depletion in asynchronous cells. e, Heatmaps showing CTCF, RAD21 and YY1 ChIP–seq signal at their respective peaks before and after CTCF depletion in asynchronous cells. f, Box plot of loop strength fold changes of YY1-independent structural loops after YY1, CTCF or SMC3 depletion. g, Box plot of loop strengths of YY1-independent CRE loops after YY1, CTCF or SMC3 degradation. h, Pileup plots of YY1-independent CRE loops, based on observed/expected signal from 10k resolution YY1-AID, CTCF-AID and SMC3-AID contact maps. i, Box plot of loop strength fold changes of YY1-independent CRE loops after YY1, CTCF or SMC3 depletion. j, Box plot of loop strength fold changes of YY1-dependent CRE loops after YY1, CTCF or SMC3 depletion.

Extended Data Fig. 4 Examples of YY1-dependent loops after cohesin depletion.

a, Contact maps showing an example of a YY1-dependent loop (blue arrows) that persists after SMC3 depletion. Upper heatmap shows interactions before/after YY1 depletion, and lower heatmap shows interactions before/after SMC3 depletion. Tracks show YY1, CTCF, RAD21 and H3K27ac ChIP–seq in untreated YY1-AID cells. b, A different example of a YY1-dependent loop that persists after SMC3 depletion. c, Histogram plot displaying CRE loop length versus the log2-fold change in loop strength after cohesin depletion in asynchronous cells. d, Box plot displaying CRE loop lengths, stratified for cohesin-dependence and YY1 dependence (two-sided Mann–Whitney U test).

Extended Data Fig. 5 Maintenance of transcription requires continuous presence of YY1.

a, Principal component analysis (PCA) of individual biological replicates of Pol II ChIP–seq. b, Representative tracks of RPM-normalized YY1 ChIP–seq from untreated YY1-AID cells and Pol II ChIP–seq before/after YY1 depletion from biological replicates of asynchronous cells. c, Histogram plot showing transcription change for genes versus YY1 peak strength at gene promoters (Spearman correlation coefficient = −0.41). d, Histogram of the multiplicity of active genes associated with CRE loops. e, Histogram of the multiplicity of CRE loops associated with active genes. f, Box plot showing transcription changes for different looping configurations at genes that do not have YY1 binding at the promoter (two-sided Mann–Whitney U test; from top to bottom: p = 1e−7, p = 1e−4, p = 0.28). g, Scatter plot showing loop strength change versus transcription change in asynchronous YY1-AID at loops that have YY1 at the distal enhancer but not at the promoter (Spearman correlation coefficient = 0.003). h, Box plot showing log2-fold change in traveling ratio for genes after YY1 depletion in asynchronous cells (two-sided Mann–Whitney U test).

Extended Data Fig. 6 YY1 chromatin binding dynamics during the mitosis-to-G1 transition.

a, Representative plots of the gating strategy for purification of mitosis-to-G1 populations. b, Heatmap showing the Pearson correlation in YY1 occupancy between biological replicates of YY1 ChIP–seq for mitosis-to-G1 stages. c, YY1 ChIP–qPCR in asynchronous (async) cells and synchronized/sorted prometaphase cells. Primers, labeled with the nearest or overlapping gene, include YY1 binding sites that display mitotic retention as well as sites that have no mitotic retention on ChIP–seq (n = 3 biological replicates). d, Prometaphase YY1 ChIP–seq track plotted alongside in silico generated tracks simulating prometaphase background and various levels of interphase contamination. e, Box plot showing YY1 binding at retained peaks genome-wide for different levels of simulated interphase contamination (two-sided Mann–Whitney U test).

Extended Data Fig. 7 Compartmentalization and TAD establishment following YY1 depletion.

a, YY1 ChIP–qPCR of synchronized, sorted prometaphase cells after mitosis-to-G1 depletion of YY1 (n = 3 biological replicates). b, Bar plot of the percent of cells in G1 after 2-hour release from nocodazole arrest after mitosis-to-G1 depletion of YY1, as assessed by DAPI signal from flow cytometry (two-sided Mann–Whitney U test). c, Micro-C contact probability curves for all biological replicates. d, Representative Micro-C contact maps with tracks of compartmentalization, with positive eigenvector 1 values (EV1) corresponding to A compartment and negative values corresponding to B compartment. e, Heatmap of Pearson correlations of subcompartment eigenvector 1 (EV1) values between biological replicates. f, Histogram plot of EV1 values after mitosis-to-G1 depletion. g, Histogram of log2 insulation score values for all boundaries detected in the −auxin_m contact map. h, Representative Micro-C contact map annotated with example TAD calls. Tracks show log2 insulation score values. i, Aggregate domain plot for all TADs called in control contact map, centered on upstream and downstream boundaries. j, Box plot showing loop strengths of all mid G1-detected YY1−YY1 loops weakened by mitosis-to-G1 depletion of YY1 (two-sided Mann–Whitney U test). k, Pileup plots corresponding to loops included in j.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Supplementary Methods.

Reporting Summary

Supplementary Tables

Supplementary Table 1: Oligo sequences. Supplementary Table 2: TAD list for asynchronous cells. Supplementary Table 3: TAD list for mid G1 cells. Supplementary Table 4: Loop calls for asynchronous cells. Supplementary Table 5: Loop calls for mid G1 cells. Supplementary Table 6. Pol II ChIP-seq DESeq2 results for asynchronous cells. Supplementary Table 7: Pol II ChIP-seq DESeq2 results for G1 stages. Supplementary Table 8: RNA-seq DESeq2 results. Supplementary Table 9: TT-seq DESeq2 results. Supplementary Table 10: Micro-C sequencing statistics.

Source data

Source Data Extended Data Fig. 1

Unprocessed western blot for Extended Data Fig. 1b.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lam, J.C., Aboreden, N.G., Midla, S.C. et al. YY1-controlled regulatory connectivity and transcription are influenced by the cell cycle. Nat Genet (2024). https://doi.org/10.1038/s41588-024-01871-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41588-024-01871-y

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