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:

Histone acetylation dynamics modulates chromatin conformation and allele-specific interactions at oncogenic loci

Abstract

In cancer cells, enhancer hijacking mediated by chromosomal alterations and/or increased deposition of acetylated histone H3 lysine 27 (H3K27ac) can support oncogene expression. However, how the chromatin conformation of enhancer–promoter interactions is affected by these events is unclear. In the present study, by comparing chromatin structure and H3K27ac levels in normal and lymphoma B cells, we show that enhancer–promoter-interacting regions assume different conformations according to the local abundance of H3K27ac. Genetic or pharmacological depletion of H3K27ac decreases the frequency and the spreading of these interactions, altering oncogene expression. Moreover, enhancer hijacking mediated by chromosomal translocations influences the epigenetic status of the regions flanking the breakpoint, prompting the formation of distinct intrachromosomal interactions in the two homologous chromosomes. These interactions are accompanied by allele-specific gene expression changes. Overall, our work indicates that H3K27ac dynamics modulates interaction frequency between regulatory regions and can lead to allele-specific chromatin configurations to sustain oncogene expression.

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: Repositioning of enhancer regions in chromatin subcompartment domains.
Fig. 2: Changes in H3K27ac modulate the frequency and size of EPIs.
Fig. 3: Reduction of H3K27ac modulates the frequency and the size of EPIs.
Fig. 4: Depletion of H3K27ac modulates the conformation of the BCL11A enhancer–promoter loop.
Fig. 5: Enhancer region on chr.3 regulates the expression of BCL6 and MYC.
Fig. 6: Allele-specific epigenetic marks and chromatin interactions at the MYC locus.
Fig. 7: Allele-specific epigenetic marks and chromatin interactions at the BCL2 locus.

Similar content being viewed by others

Data availability

RNA-seq and H3K27ac ChIP–seq data were obtained as follows: for lymphoblastoid cells from ref. 68 and the ArrayExpress Archive (http://www.ebi.ac.uk/arrayexpress), accession nos. E-MTAB-3656 and E-MTAB-3657, for GM12878 from ENCODE accession nos. GSE78551 and GSM733771, for SU-DHL-4, accession nos. GSM1227199 and GSM1703927, for OCI-LY7, accession nos. GSM1227199 and GSE86708, for DoHH2 ENCODE accession nos. GSM2366283 and GSE86743, for Karpas-422 accession no. GSE86733 (H3K27ac), for primary centrocytes and centroblasts, accession nos. GSE62246 and GSE89688, for five primary follicular lymphoma and two GC diffuse large B-cell lymphoma BLUEPRINT, accession no. EGAD00001001502. WGS data and RNA-seq data were obtained for lymphoma cell lines and for 30 primary DLBCLs and their matched controls from dbGaP (phs000235.v13.p2). WGS data and RNA-seq data for Burkitt’s lymphoma or DLBCL with t(8;14) were downloaded from the Malignant Lymphoma MMML ICGC portal. RNA-seq data were generated for Karpas-422 and WSU-DLCL2 that were nontreated and treated by A-485 and for three independent clones WSU-DLCL2 with BCL11A knockout (data are deposited in the Gene Expression Omnibus database at accession no. GSE168471). CTCF ChIP–seq data were obtained from ENCODE for GM12878 and we generated the data for Karpas-422 and WSU-DLCL2 that were nontreated and treated by A-485 (accession no. GSE168470). H3K36me3 ChIP–seq data were generated for WSU-DLCL2 (accession no. GSE168472). Hi-C data for nontreated WSU-DLCL2 and Karpas-422 were obtained from Donaldson-Collier et al.4 and generated for the treated replicates for the present study (accession no. GSE168470). Source data are provided with this paper.

Code availability

Code availability for Calder method: https://github.com/CSOgroup/CALDER.

References

  1. Feinberg, A. P., Koldobskiy, M. A. & Göndör, A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat. Rev. Genet. 17, 284–299 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Donaldson-Collier, M. C. et al. EZH2 oncogenic mutations drive epigenetic, transcriptional, and structural changes within chromatin domains. Nat. Genet. https://doi.org/10.1038/s41588-018-0338-y (2019).

  5. Achinger-Kawecka, J. et al. Epigenetic reprogramming at estrogen-receptor binding sites alters 3D chromatin landscape in endocrine-resistant breast cancer. Nat. Commun. 11, 320 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Viny, A. D. et al. Cohesin members Stag1 and Stag2 display distinct roles in chromatin accessibility and topological control of HSC self-renewal and differentiation. Cell Stem Cell 25, 682–696.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Beagan, J. A. & Phillips-Cremins, J. E. On the existence and functionality of topologically associating domains. Nat. Genet. 52, 8–16 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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 

  9. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320.e24 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schoenfelder, S. & Fraser, P. Long-range enhancer–promoter contacts in gene expression control. Nat. Rev. Genet. 20, 437–455 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Weischenfeldt, J. et al. Pan-cancer analysis of somatic copy-number alterations implicates IRS4 and IGF2 in enhancer hijacking. Nat. Genet. 49, 65–74 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Koche, R. P. et al. Extrachromosomal circular DNA drives oncogenic genome remodeling in neuroblastoma. Nat. Genet. 52, 29–34 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Morton, A. R. et al. Functional enhancers shape extrachromosomal oncogene amplifications. Cell 179, 1330–1341.e13 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hnisz, D. et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science https://doi.org/10.1126/science.aad9024 (2016).

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ernst, J. et al. Systematic analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Calo, E. & Wysocka, J. Modification of enhancer chromatin: what, how and why? Mol. Cell 49, 825–837 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Andersson, R. & Sandelin, A. Determinants of enhancer and promoter activities of regulatory elements. Nat. Rev. Genet. 21, 71–87 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Bartman, C. R., Hsu, S. C., Hsiung, C. C.-S., Raj, A. & Blobel, G. A. Enhancer regulation of transcriptional bursting parameters revealed by forced chromatin looping. Mol. Cell 62, 237–247 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Larsson, A. J. M. et al. Genomic encoding of transcriptional burst kinetics. Nature 565, 251–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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 

  25. 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 

  26. Rosencrance, C. D. et al. Chromatin hyperacetylation impacts chromosome folding by forming a nuclear subcompartment. Mol. Cell 78, 112–126.e12 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  28. Liu, Y. et al. Systematic inference and comparison of multi-scale chromatin sub-compartments connects spatial organization to cell phenotypes. Nat. Commun. https://doi.org/10.1038/s41467-021-22666-3 (2021).

  29. Amoli, M. M., Carthy, D., Platt, H. & Ollier, W. E. R. EBV Immortalization of human B lymphocytes separated from small volumes of cryo-preserved whole blood. Int. J. Epidemiol. 37, i41–i45 (2008).

    Article  PubMed  Google Scholar 

  30. Koues, O. I. et al. Enhancer sequence variants and transcription-factor deregulation synergize to construct pathogenic regulatory circuits in B-cell lymphoma. Immunity 42, 186–198 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Zhang, J. et al. The CREBBP acetyltransferase is a haploinsufficient tumor suppressor in B-cell lymphoma. Cancer Discov. 7, 322–337 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Basso, K. & Dalla-Favera, R. Roles of BCL6 in normal and transformed germinal center B cells. Immunol. Rev. 247, 172–183 (2012).

    Article  PubMed  Google Scholar 

  33. Hsu, J. et al. CHD7 and Runx1 interaction provides a braking mechanism for hematopoietic differentiation. Proc. Natl Acad. Sci. USA 117, 23626–23635 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Li, J. et al. The EMT transcription factor Zeb2 controls adult murine hematopoietic differentiation by regulating cytokine signaling. Blood 129, 460–472 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Robles, E. F. et al. Homeobox NKX2-3 promotes marginal-zone lymphomagenesis by activating B-cell receptor signalling and shaping lymphocyte dynamics. Nat. Commun. 7, 11889 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nagel, S. et al. NKL homeobox gene NKX2-2 is aberrantly expressed in Hodgkin lymphoma. Oncotarget 9, 37480–37496 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Battistello, E. et al. Pan-SRC kinase inhibition blocks B-cell receptor oncogenic signaling in non-Hodgkin lymphoma. Blood https://doi.org/10.1182/blood-2017-10-809210 (2018).

  38. Carty, M. et al. An integrated model for detecting significant chromatin interactions from high-resolution Hi-C data. Nat. Commun. 8, 15454 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lasko, L. M. et al. Discovery of a potent catalytic p300/CBP inhibitor that targets lineage-specific tumors. Nature 550, 128–132 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bauer, D. E. et al. An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level. Science 342, 253–257 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Canver, M. C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature 527, 192–197 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Okosun, J. et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat. Genet. 46, 176–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Rao, P. H. et al. Chromosomal and gene amplification in diffuse large B-cell lymphoma. Blood 92, 234–240 (1998).

    Article  CAS  PubMed  Google Scholar 

  44. Weniger, M. A. et al. Gains of the proto-oncogene BCL11A and nuclear accumulation of BCL11A XL protein are frequent in primary mediastinal B-cell lymphoma. Leukemia 20, 1880–1882 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Victora, G. D. et al. Identification of human germinal center light and dark zone cells and their relationship to human B-cell lymphomas. Blood 120, 2240 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bunting, K. L. et al. Multi-tiered reorganization of the genome during B cell affinity maturation anchored by a germinal center-specific locus control region. Immunity 45, 497–512 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chong, L. C. et al. High-resolution architecture and partner genes of MYC rearrangements in lymphoma with DLBCL morphology. Blood Adv. 2, 2755–2765 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ohno, H., Nakagawa, M., Kishimori, C., Fukutsuka, K. & Honjo, G. Cryptic t(3;8)(q27;q24) and/or MYC-BCL6 linkage associated with MYC expression by immunohistochemistry is frequent in multiple-hit B-cell lymphomas. Blood Cancer J. 7, e578 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ryan, R. J. H. et al. Detection of enhancer-associated rearrangements reveals mechanisms of oncogene dysregulation in B-cell lymphoma. Cancer Discov. 5, 1058–1071 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cho, S. W. et al. Promoter of lncRNA gene PVT1 is a tumor-suppressor DNA boundary element. Cell 173, 1398–1412.e22 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Küppers, R. & Dalla-Favera, R. Mechanisms of chromosomal translocations in B cell lymphomas. Oncogene 20, 5580–5594 (2001).

    Article  PubMed  Google Scholar 

  52. Graninger, W. B., Seto, M., Boutain, B., Goldman, P. & Korsmeyer, S. J. Expression of Bcl-2 and Bcl-2-Ig fusion transcripts in normal and neoplastic cells. J. Clin. Invest. 80, 1512–1515 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Tsujimoto, Y., Cossman, J., Jaffe, E. & Croce, C. M. Involvement of the bcl-2 gene in human follicular lymphoma. Science 228, 1440–1443 (1985).

    Article  CAS  PubMed  Google Scholar 

  54. Boettiger, A. N. et al. Super-resolution imaging reveals distinct chromatin folding for different epigenetic states. Nature 529, 418–422 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Morgan, M. A. J. & Shilatifard, A. Reevaluating the roles of histone-modifying enzymes and their associated chromatin modifications in transcriptional regulation. Nat. Genet. 52, 1271–1281 (2020).

    Article  CAS  PubMed  Google Scholar 

  57. Douillet, D. et al. Uncoupling histone H3K4 trimethylation from developmental gene expression via an equilibrium of COMPASS, Polycomb and DNA methylation. Nat. Genet. 52, 615–625 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Pérez-Lluch, S. et al. Absence of canonical marks of active chromatin in developmentally regulated genes. Nat. Genet. 47, 1158–1167 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kilpinen, H. et al. Coordinated effects of sequence variation on DNA binding, chromatin structure, and transcription. Science 342, 744–747 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Gimelbrant, A., Hutchinson, J. N., Thompson, B. R. & Chess, A. Widespread monoallelic expression on human autosomes. Science 318, 1136–1140 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Schwartzman, O. et al. UMI-4C for quantitative and targeted chromosomal contact profiling. Nat. Methods 13, 685–691 (2016).

    Article  CAS  PubMed  Google Scholar 

  64. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Loh, P.-R., Palamara, P. F. & Price, A. L. Fast and accurate long-range phasing in a UK Biobank cohort. Nat. Genet. 48, 811–816 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Edge, P., Bafna, V. & Bansal, V. HapCUT2: robust and accurate haplotype assembly for diverse sequencing technologies. Genome Res. 27, 801–812 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ramos-Rodriguez, M. & Subirana-Granes, M. UMI4Cats: Processing, analysis and visualization of UMI-4C chromatin contact data. R package version 4.0 https://doi.org/10.18129/B9.bioc.UMI4Cats (2021).

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

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank EPFL facilities and, in particular, B. Mangeat for the sequencing facility and M. Pujol for cell sorting at CIML. This work was supported by the ISREC Foundation (E.O.), the Swiss National Science Foundation (grant number 31003A_182526) (E.O.) and Swiss Cancer Research foundation (grant number KFS-3982-08-2016-R) (E.O.), and the Gelu Foundation. G.C. is supported by the Giorgi–Cavaglieri Foundation. S.R. is supported by the French National Cancer Institute (INCa) Epigenetic and Cancer program. S.S. has been supported with a Marie Curie EPFL fellow.

Author information

Authors and Affiliations

Authors

Contributions

S.S. was involved in the experimental design. Y.L. developed a new method and analyzed Hi-C data. S.S. and Y.L. analyzed Hi-C, UMI-4C and ChIP–seq data, and performed the phasing analyses. M.D.C., R.L. and N.K. prepared Hi-C, UMI-4C, ChIP- and RNA-seq libraries, and performed all experimental validations. D.T. analyzed and normalized ChIP–seq data. S.R. obtained and sorted the germinal center cells and prepared cells for ChIP and Hi-C library preparation G.C. supervised the computational analyses. E.O. designed the study and wrote the manuscript, with comments from all authors.

Corresponding author

Correspondence to Elisa Oricchio.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Berkley Gryder, Rolf Ohlsson and the other, anonymous, reviewer for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 Genome-wide analyses of H3K27ac changes and chromatin sub-compartments.

a. t-SNE plot based on the levels of H3K27ac in 9250 MERs in lymphoblastoid samples (n = 76, blue, GM12878 in dark blue), GC, (n = 5 green), and GC-DLBCL (n = 5, red). b. Heatmap and clustering of 5 cell lines based on the Pearson’s correlation coefficients (values are indicated and color coded) between H3K27ac levels of MERs (n = 1644). c. Heatmap and clustering of 4 cell lines analyzed by Hi-C based on the Pearson’s correlation coefficients (values are indicated and color coded) between compartment domain ranks of MERs (n = 1644). d. For each pair of cell lines a scatterplot comparison shows the difference of compartment domain ranks computed by CALDER (x-axis) and of H3K27ac levels (y-axis) computed for each enhancer region (dots). Enhancer regions exhibiting concordant changes are color-coded (red positive differences, >5%, blue negative differences <−5%). For each comparison, the radius of the circle comprising 90% of the points (R90) is shown. e. For each comparison, Pearson’s correlation coefficient (x-axis) between differences of compartment domain ranks and of H3K27ac. Distribution of correlation values (colored) are compared to expected distributions (gray). f-g. Comparison of compartment domain rank differences computed with CALDER between (f) Karpas-422 and GC cells (x-axis) and Karpas-422 and GM12878 (y-axis) and between (g) WSU-DLCL2 and GC cells (x-axis) and WSU-DLCL2 and GM12878 (y-axis). Each dot is a merged enhancer region color coded by the difference of H3K27ac between Karpas-422 (left) or WSU-DLCL2 (right) and GC h. Overlap of repositioned enhancer regions towards a more active (red) or inactive (blue) sub-compartments in Karpas-422 and WSU-DLCL2 cells with respect to GM12878. P-values and odds-ratio (OR) were computed by two-sided Fisher’s exact test. i. Comparison of compartment domain rank differences computed with CALDER between (left) Patient 1 and GC cells (x-axis) and Patient 1 and GM12878 (y-axis) and between (right) Patient 7 and GC cells (x-axis) and Patient 7 and GM12878 (y-axis). j. Overlap of repositioned enhancer regions towards a more active (red) or inactive (blue) sub-compartments in Patient 1 and Patient 7 with respect to GM12878. P-values and odds-ratio (OR) were computed by two-sided Fisher’s exact test.

Extended Data Fig. 2 Genome wide analyses of enhancer promoter interactions (EPIs) and changes in H3K27ac.

a. Graphical representation of enhancer-promoter interactions (EPIs, top) analysis. The total number of tested and significant EPIs is reported for each cell line. b. Fraction of significant EPI that are expected (top) and that were observed (bottom) to occur between enhancers and promoters within the same compartment domain. c. Percentage of significantly different EPIs (y-axis) which are more frequent either in GC than in the indicated lymphoma samples (blue bars) or in lymphoma samples than in GC (red bars). Results are shown for enhancer regions that were repositioned from inactive to active compartment (left) or from active to inactive compartment (right) in lymphoma samples with respect to GC. d. Fold-change between the observed and expected number of significantly more frequent EPIs (y-axis) in Karpas-422 than in GM12878 (left) and in GC (right) with respect to the difference in H3K27ac in these regions. e. Number of significant interactions per EPI (that is, number of bins of the Hi-C map with significantly frequent interactions between the enhancer and promoter regions) (x-axis) with respect to the difference in H3K27ac in these regions. Results are shown for EPIs that were more frequent in Karpas-422 than in GM12878 (top) or in GC (bottom). f. Representative western blot image (n=2 independent experiments) detecting the H3K27ac and histone-3 treated with DMSO (vehicle) or 0.2 μM and 0.5 μM A-485 for 48h in the indicated lymphoma cell lines. g. Quantification of cell survival of Karpas-422 and Su-DHL-4 cells treated with 0.5 μM and 1μM A-485 or DMSO. h. Density plot of the p-values inferred by HiC-DC for each interaction in WSU-DLCL2 and Karpas-422 Hi-C maps connecting bins at most 2 Mb apart. P-values below 0.05 (left of black dashed line) indicate significant contacts.

Source data

Extended Data Fig. 3 Chromatin conformation analyses of BCL11A region upon pharmacological depletion of H3K27ac.

a. Representation of 20kb interacting regions on chr.2: 60.46–60–86 Mb color coded based on their q-value in Karpas-422 cells untreated and treated with A-485 inhibitor (top) and corresponding ChIP-sequencing track of H3K27ac (NRPM) (botton). b. Representative H3K27ac ChIP sequencing tracks for the indicated genomic locus of lymphoblastoid samples, GC, lymphoma cells and lymphoma patients (NRPM). Patient 1 and Patient 7 are reported as RRPM x 10−2. c. Spearman correlation plot of H3K27ac levels and BCL11A expression in 70 lymphoblastoid samples. two-tailed p value is calculated from the t-statistics of the corresponding correlation value. d. BCL11A expression levels in GC-DLBCL patients (n=30) and lymphoblastoid cells (n=70). The thick central line of each box plot represents the median expression value, the bounding box corresponds to the 25th–75th percentiles, and the whiskers extend up to 1.5 times the interquartile range. p-value was calculated with two tailed t-test. e. Quantification of expression changes of BCL11A upon treatment with A-485 0.5 μM for 48h in Karpas-422 (n= 6) and WSU-DLCL2 (n= 3) compared to the same cells treated with DMSO (vehicle Karpas-422 n= 5 and WSU-DLCL2 n=3). The black dots represent the number of independent experiments. Data are presented as mean value + SD. p-value were calculated using unpaired two-tailed t-test f. Hi-C contact maps of chr.2: 60.16–61.46 Mb region in Karpas-422 and WSU-DLCL2 cells untreated and treated with A-485. The lines in black delineate the compartment domains. The position of relevant genes is indicated. g. CTCF ChiP-seq tracks of chr.2: 60.16–61.46 Mb region in Karpas-422 and WSU-DLCL2 treated and untreated with A-485 (RPM). The detection of CTCF and their orientation is reported.

Extended Data Fig. 4 Chromatin conformation analyses of BCL11A region upon genetic modification of H3K27ac.

a. Quantification of BCL11A expression changes in Su-DHL-4 cells expressing dCas9-KRAB-sgRNA1, dCas9-KRAB-sgRNA2, dCas9-KRAB-sgRNA3 compared to cells expressing dCas9-KRAB. n=3 independent biological replicates. Data are presented as mean value + SD p-value were calculated by unpaired two-tailed t-test. b. H3K27ac ChIP sequencing tracks in Su-DHL-4 labelled with the position of the primers used for ChIP-qPCR and quantification of H3K27ac by ChIP-qPCR with the indicated primers. n=3 independent biological replicates. Data are presented as mean value ± SD. p-value were calculated by unpaired two-tailed t-test c. H3K27ac ChIP sequencing tracks in Su-DHL-4 on the regions on chr.2 (60.85–61.45 Mb) flanking the BCL11A region (RRPM x10−3). d. Hi-C contact map and ChIP sequencing track of H3K27ac (RPM) in K562 cells in the indicated genomic region. e. Representation of 20kb interacting regions on chr.2: 60.46–60–86 Mb color coded based on their q value in K562 cells. f. Representation of the number of reads spanning BCL11A exon2 detected by RNA-sequencing in K562 cells expressing dCas9-EP300 and dCas9-EP300-sgRNA2. g. Representative western-blot image (n=2) of BCL11A and tubulin in WSU-DLCL2 cells (control) and three independent BCL11A knock-out clones (KO#2, KO#12, KO#25). h. Bar plot of the representative gene set enriched categories that significantly scored.

Source data

Extended Data Fig. 5 Analyses of the chromatin structure in BCL6 and MYC loci.

a. Representative ChIP sequencing tracks of H3K27ac in lymphoblastoid cells, GC, DLBCL tumor cells, and in primarylymphoma samples (NRPM), Patient 1 and Patient 7 are reported as RRPM x10−2. b-c. Representation of 20kb interacting regions in chr.3 187.4–188.7 Mb color coded based on their q value (top) in Karpas-422 untreated (b) or treated with A-485 inhibitor (c) and corresponding H3K27ac ChIP-sequencing (NRPM). d. Quantification of expression changes of BCL6 upon treatment with A-485 0.5 μM for 48h in Karpas-422 (n=6) and WSU-DLCL2 (n=6) compared to the same cells treated with DMSO (vehicle, n=5). The black dots represent the number of independent experiments. Data are presented as mean value + SD. p-value were calculated using unpaired two-tailed t-test. e. Representation of copy number changes detected on chr. 8 and chr. 3 and graphical representation of the derivative chromosome t(3;8). f. Hi-C inter-chromosomal contact maps of chr.8 and chr.3 in the region spanning the breakpoint in WSU-DLCL2 cells. g. ChIP-sequencing track of H3K27ac (blue, NRPM) and UMI-4C domainogram with two different bait-primers representing the mean number of contacts (% of the maximum) on chr.8 spanning the breakpoint in WSU-DLCL2 cells treated with A-485 (0.5 μM for 48h) or DMSO as control.

Extended Data Fig. 6 Epigenetic marks distributed to each copy of chromosome 8 based on their haplotype.

a. Schematic representation of the phasing protocol used to define the haplotypes of the two copies of chr. 8. b.H3K27ac and H3K36me3 signal distribution in wild-type (WT) and translocated chromosome (TRA) in WSU-DLCL2 cells between chr.8:128,745–128,755 Mb region c. H3K27ac ChIP tracks of chr.8 indicated region in Karpas-422. In grey the total number of reads, in blue the number of reads with SNPs mapping on the haplotype 1 and red the number of reads with SNPs mapping on the haplotype 2 (NRPM). d. Representation of the reads distribution in whole genome sequencing (WGS), H3K27ac and H3K36me3 ChIP sequencing in the indicated genomic regions. In grey, genomic positions without detected SNPs and in two color bars genomic position with detected SNPs. e. H3K27ac and H3K36me3 ChIP tracks in the chr.8 indicated region (NRPM). In grey the total number of reads, in blue the reads with SNPs mapping on the haplotype 1 and red the number of reads with SNPs mapping on the haplotype 2. f. Quantification of the number of reads spanning the chr8:129,047,325 SNP harboring an adenine (A) or a guanine (G) detected by whole genome sequencing (WGS) and RNA- sequencing (RNA-seq) in Karpas-422 cells.

Extended Data Fig. 7 Chr.14–18 inter-chromosomal interactions in samples with and without translocation.

a-d. Hi-C inter-chromosomal contact maps of chr.14 and chr.18 in the region spanning the breakpoint in WSU-DLCL2 cells (a), Patient 1(b), GC (c) and Patient 7 (d).

Supplementary information

Supplementary Information

Supplementary Methods

Reporting Summary

Supplementary Tables

Supplementary Tables 1–7

Supplementary Data 1

Supplementary Data

Supplementary Data 2

Supplementary Data

Source data

Source Data Figure 2

Unprocessed western blots.

Source Data Extended Data Figure 2

Unprocessed western blots.

Source Data Extended Data Figure 4

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sungalee, S., Liu, Y., Lambuta, R.A. et al. Histone acetylation dynamics modulates chromatin conformation and allele-specific interactions at oncogenic loci. Nat Genet 53, 650–662 (2021). https://doi.org/10.1038/s41588-021-00842-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00842-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing