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:

Identifying synergistic high-order 3D chromatin conformations from genome-scale nanopore concatemer sequencing

Abstract

High-order three-dimensional (3D) interactions between more than two genomic loci are common in human chromatin, but their role in gene regulation is unclear. Previous high-order 3D chromatin assays either measure distant interactions across the genome or proximal interactions at selected targets. To address this gap, we developed Pore-C, which combines chromatin conformation capture with nanopore sequencing of concatemers to profile proximal high-order chromatin contacts at the genome scale. We also developed the statistical method Chromunity to identify sets of genomic loci with frequencies of high-order contacts significantly higher than background (‘synergies’). Applying these methods to human cell lines, we found that synergies were enriched in enhancers and promoters in active chromatin and in highly transcribed and lineage-defining genes. In prostate cancer cells, these included binding sites of androgen-driven transcription factors and the promoters of androgen-regulated genes. Concatemers of high-order contacts in highly expressed genes were demethylated relative to pairwise contacts at the same loci. Synergies in breast cancer cells were associated with tyfonas, a class of complex DNA amplicons. These results rigorously link genome-wide high-order 3D interactions to lineage-defining transcriptional programs and establish Pore-C and Chromunity as scalable approaches to assess high-order genome structure.

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: Pore-C concatemers yield high-fidelity maps of proximal 3D contacts.
Fig. 2: Synergy algorithm uncovers cooperativity in high-order enhancer and promoter interactions.
Fig. 3: De novo discovery of reference-distal and interchromosomal cooperativity with Chromunity.
Fig. 4: High-order contacts in highly expressed genes are preferentially demethylated.
Fig. 5: Androgen stimulation induces enhancer and promoter cooperativity at androgen-dependent loci in prostate cells.
Fig. 6: Synergy associated with a complex cancer amplicon.

Similar content being viewed by others

Data availability

Sequence and pairwise and high-order contact data that support the findings of this study have been deposited in GEO with the accession code GSE149117.

Code availability

The source code of packages used in these analyses can be accessed at the following links along with dependencies: https://github.com/nanoporetech/pore-c, https://github.com/mskilab/chromunity and https://github.com/mskilab/gGnome. Python (3.7.*) and R (3.6.0) were used for developing these tools. https://github.com/mskilab/GxG

References

  1. Stadhouders, R., Filion, G. J. & Graf, T. Transcription factors and 3D genome conformation in cell-fate decisions. Nature 569, 345–354 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Finn, E. H. & Misteli, T. A genome disconnect. Nat. Genet. 51, 1205–1206 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Mirny, L. & Dekker, J. Mechanisms of chromosome folding and nuclear organization: their interplay and open questions. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a040147 (2021).

  4. Nollmann, M., Bennabi, I., Götz, M. & Gregor, T. The impact of space and time on the functional output of the genome. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a040378 (2021).

  5. McCord, R. P., Kaplan, N. & Giorgetti, L. Chromosome conformation capture and beyond: toward an integrative view of chromosome structure and function. Mol. Cell 77, 688–708 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Oudelaar, A. M. et al. Single-allele chromatin interactions identify regulatory hubs in dynamic compartmentalized domains. Nat. Genet. 50, 1744–1751 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ay, F. et al. Identifying multi-locus chromatin contacts in human cells using tethered multiple 3C. BMC Genomics 16, 121 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Darrow, E. M. et al. Deletion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture. Proc. Natl Acad. Sci. USA 113, 4504–4512 (2016).

    Article  Google Scholar 

  9. Olivares-Chauvet, P. et al. Capturing pairwise and multi-way chromosomal conformations using chromosomal walks. Nature 540, 296–300 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Allahyar, A. et al. Enhancer hubs and loop collisions identified from single-allele topologies. Nat. Genet. 50, 1151–1160 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Beagrie, R. A. et al. Complex multi-enhancer contacts captured by genome architecture mapping. Nature 543, 519–524 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Bhat, P., Honson, D. & Guttman, M. Nuclear compartmentalization as a mechanism of quantitative control of gene expression. Nat. Rev. Mol. Cell Biol. 22, 653–670 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Hnisz, D., Shrinivas, K., Young, R. A., Chakraborty, A. K. & Sharp, P. A. A phase separation model for transcriptional control. Cell 169, 13–23 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Spielmann, M., Lupiáñez, D. G. & Mundlos, S. Structural variation in the 3D genome. Nat. Rev. Genet. 19, 453–467 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. 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 

  19. Salameh, T. J. et al. A supervised learning framework for chromatin loop detection in genome-wide contact maps. Nat. Commun. 11, 3428 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Weinreb, C. & Raphael, B. J. Identification of hierarchical chromatin domains. Bioinformatics 32, 1601–1609 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Yang, T. et al. HiCRep: assessing the reproducibility of Hi-C data using a stratum-adjusted correlation coefficient. Genome Res. 27, 1939–1949 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kaplan, N. & Dekker, J. High-throughput genome scaffolding from in vivo DNA interaction frequency. Nat. Biotechnol. 31, 1143–1147 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Burton, J. N. et al. Chromosome-scale scaffolding of de novo genome assemblies based on chromatin interactions. Nat. Biotechnol. 31, 1119–1125 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Dudchenko, O. et al. De novo assembly of the Aedes aegypti genome using Hi-C yields chromosome-length scaffolds. Science 356, 92–95 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ghurye, J. et al. Integrating Hi-C links with assembly graphs for chromosome-scale assembly. PLoS Comput. Biol. 15, e1007273 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bickhart, D. M. et al. Single-molecule sequencing and chromatin conformation capture enable de novo reference assembly of the domestic goat genome. Nat. Genet. 49, 643–650 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zook, J. M. et al. An open resource for accurately benchmarking small variant and reference calls. Nat. Biotechnol. 37, 561–566 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shafin, K. et al. Nanopore sequencing and the Shasta toolkit enable efficient de novo assembly of eleven human genomes. Nat. Biotechnol. 38, 1044–1053 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jiang, S. et al. The Epstein–Barr virus regulome in lymphoblastoid cells. Cell Host Microbe 22, 561–573 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wood, C. D. et al. MYC activation and BCL2L11 silencing by a tumour virus through the large-scale reconfiguration of enhancer-promoter hubs. eLife 5, e18270 (2016).

  31. Zhao, B. et al. Epstein–Barr virus exploits intrinsic B-lymphocyte transcription programs to achieve immortal cell growth. Proc. Natl Acad. Sci. USA 108, 14902–14907 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Palmer, C., Diehn, M., Alizadeh, A. A. & Brown, P. O. Cell-type specific gene expression profiles of leukocytes in human peripheral blood. BMC Genomics 7, 115 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  33. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  35. Disteche, C. M. & Berletch, J. B. X-chromosome inactivation and escape. J. Genet. 94, 591–599 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lee, J. T. Gracefully ageing at 50, X-chromosome inactivation becomes a paradigm for RNA and chromatin control. Nat. Rev. Mol. Cell Biol. 12, 815–826 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  38. Froberg, J. E., Pinter, S. F., Kriz, A. J., Jégu, T. & Lee, J. T. Megadomains and superloops form dynamically but are dispensable for X-chromosome inactivation and gene escape. Nat. Commun. 9, 5004 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hankey, W., Chen, Z. & Wang, Q. Shaping chromatin states in prostate cancer by pioneer transcription factors. Cancer Res. 80, 2427–2436 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Augello, M. A. et al. CHD1 loss alters AR binding at lineage-specific enhancers and modulates distinct transcriptional programs to drive prostate tumorigenesis. Cancer Cell 35, 603–617 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Maciejowski, J. & Imielinski, M. Modeling cancer rearrangement landscapes. Curr. Opin. Syst. Biol. 1, 54–61 (2017).

    Article  PubMed  Google Scholar 

  42. Hadi, K. et al. Distinct classes of complex structural variation uncovered across thousands of cancer genome graphs. Cell 183, 197–210 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hung, K. L. et al. ecDNA hubs drive cooperative intermolecular oncogene expression. Nature 600, 731–736 (2021).

  44. Zhu, Y. et al. Oncogenic extrachromosomal DNA functions as mobile enhancers to globally amplify chromosomal transcription. Cancer Cell 39, 694–707 (2021).

  45. Koche, R. P. et al. Extrachromosomal circular DNA drives oncogenic genome remodeling in neuroblastoma. Nat. Genet. 52, 29–34 (2019).

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

  47. Wu, S. et al. Circular ecDNA promotes accessible chromatin and high oncogene expression. Nature 575, 699–703 (2019).

  48. Ghandi, M. et al. Next-generation characterization of the cancer cell line encyclopedia. Nature 569, 503–508 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science 361, eaar3958 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Guo, Y. E. et al. Pol II phosphorylation regulates a switch between transcriptional and splicing condensates. Nature 572, 543–548 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Basu, S. et al. Unblending of transcriptional condensates in human repeat expansion disease. Cell 181, 1062–1079 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Li, J. et al. Single-gene imaging links genome topology, promoter–enhancer communication and transcription control. Nat. Struct. Mol. Biol. 27, 1032–1040 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Boija, A. et al. Transcription factors activate genes through the phase-separation capacity of their activation domains. Cell 175, 1842–1855 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Stortz, M., Presman, D. M., Pecci, A. & Levi, V. Phasing the intranuclear organization of steroid hormone receptors. Biochem. J 478, 443–461 (2021).

    Article  CAS  PubMed  Google Scholar 

  55. Dixon, J. R. et al. Integrative detection and analysis of structural variation in cancer genomes. Nat. Genet. 50, 1388–1398 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang, X. et al. Genome-wide detection of enhancer-hijacking events from chromatin interaction data in rearranged genomes. Nat. Methods 18, 661–668 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Rubin, M. A., Bristow, R. G., Thienger, P. D., Dive, C. & Imielinski, M. Impact of lineage plasticity to and from a neuroendocrine phenotype on progression and response in prostate and lung cancers. Mol. Cell 80, 562–577 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Koster, J. & Rahmann, S. Snakemake—a scalable bioinformatics workflow engine. Bioinformatics 28, 2520–2522 (2012).

    Article  PubMed  Google Scholar 

  59. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Ramírez, F. et al. High-resolution TADs reveal DNA sequences underlying genome organization in flies. Nat. Commun. 9, 189 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Abdennur, N. & Mirny, L. Cooler: scalable storage for Hi-C data and other genomically-labeled arrays. Bioinformatics 36, 311–316 (2020).

    Article  CAS  PubMed  Google Scholar 

  62. Knight, P. A. & Ruiz, D. A fast algorithm for matrix balancing. IMA J. Numer. Anal. 33, 1029–1047 (2013).

    Article  Google Scholar 

  63. Agrawal, R., Imielinski, T. & Swami, A. Mining association rules between sets of items in large databases. ACM SIGMOD Record 22, 207–216 (1993).

    Article  Google Scholar 

  64. Clauset, A., Newman, M. E. J. & Moore, C. Finding community structure in very large networks. Phys. Rev. 70, 066111 (2004).

    Google Scholar 

  65. Martin, M. et al. Whatshap: fast and accurate read-based phasing. Preprint at bioRxiv https://doi.org/10.1101/085050 (2016).

  66. Gamaarachchi, H., Lam, C. W., Jayatilaka, G., Samarakoon, H. & Smith, M. A. GPU accelerated adaptive banded event alignment for rapid comparative nanopore signal analysis. BMC Bioinform. 21, 343 (2020).

    Article  CAS  Google Scholar 

  67. Simpson, J. T. et al. Detecting DNA cytosine methylation using nanopore sequencing. Nat. Methods 14, 407–410 (2017).

    Article  CAS  PubMed  Google Scholar 

  68. Cotton, A. M. et al. Landscape of DNA methylation on the X chromosome reflects CpG density, functional chromatin state and X-chromosome inactivation. Hum. Mol. Genet. 24, 1528–1539 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Haeussler, M. et al. The UCSC Genome Browser database: 2019 update. Nucleic Acids Res. 47, D853–D858 (2019).

  70. Raudsepp, T. & Chowdhary, B. P. The eutherian pseudoautosomal region. Cytogenet. Genome Res. 147, 81–94 (2015).

    Article  PubMed  Google Scholar 

  71. Wala, J. A. et al. SvABA: genome-wide detection of structural variants and indels by local assembly. Genome Res. 28, 581–591 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Tate, J. G. et al. COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res. 47, 941–947 (2019).

    Article  Google Scholar 

Download references

Acknowledgements

We thank J. Skok for helpful comments on the manuscript. M.I. is supported by a Burroughs Wellcome Fund Career Award for Medical Scientists, Doris Duke Clinical Foundation Clinical Scientist Development Award and The Pershing Square Sohn Prize for Young Investigators in Cancer Research.

Author information

Authors and Affiliations

Authors

Contributions

A.S.D., N.U. and M.I. conceptualized the project. A.S.D., N.U., M.P., E.H. and M.I. curated the data. A.S.D., N.U., M.P., L.L., J.M.B., W.L., P.R., H.G.O., E.H. and M.I. performed the formal analysis. S.J., D.J.T. and M.I. acquired funding. A.S.D., N.U., M.P., S.S., X.D., M.A.A., C.E.B., A.M., E.H., S.J. and M.I. performed the investigations. A.S.D., N.U., M.P., L.L., P.R., S.S., D.S., C.T., S.J., E.H. and M.I. developed the methodology. E.H. and M.I. administered the project. S.J. and M.I. provided resources. A.S.D., M.P., E.H. and M.I. developed software. S.J. and M.I. supervised the project. A.S.D., N.U., M.P., X.D., S.K., H.T., E.A., D.W., J.M.M., E.H., S.J. and M.I. performed validations. A.S.D., M.P., E.H., and M.I. visualized the data. A.S.D., N.U., M.P., E.H. and M.I. wrote the original draft. All authors reviewed and edited the final draft.

Corresponding author

Correspondence to Marcin Imieliński.

Ethics declarations

Competing interests

M.P., S.S., X.D., L.L., C.T., P.R., D.S., D.T., S.J. and E.H. are employees of, and are stock option holders in, ONT. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers 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 Pore-C assay development.

(a) An overview of Pore-C alignment. A directed acyclic graph (DAG) is built by partially ordering reference alignments on each concatemer query sequence. The final concatemer alignment comprises a greedy traversal of the DAG (see Methods for more details). (b) Comparison of cumulative fragment length distributions (left) and densities (right) associated with three different restriction enzyme based protocols (NlaIII, DpnIII and HindIII). (c) Comparison of virtual pairwise contacts generated per Gb sequenced for Pore-C for the three restriction enzyme based protocols (NlaIII: n = 13, DpnIII: n = 6, and HindIII: n = 2). P-values obtained using two-sided Wilcoxon rank sum test. For all boxplots, the centerline corresponds to the mean, box limits to the interquartile range (IQR) and the whiskers to the last datum within 1.5 * IQR. (d) Stratum adjusted correlation coefficients (SCC) comparing single MinION runs (M1 and M2), single PromethION runs (P1 and P2) and combination of P1 and P2 with the full GM12878 Hi-C data set (n = 23 chromosomes for each run). The bars represent mean SCC score for all chromosomes for each run with standard error of mean. (e) Comparison of 500 kb compartment scores (CS) between Pore-C (1.89 billion virtual pairwise contacts) and Hi-C ( ~ 4 billion pairwise contacts) for GM12878. (f) Comparison of 50 kb topologically associated domain insulation scores (IS) between Pore-C and Hi-C. (g) Jaccard similarity between TADtree TAD boundaries in gold standard Hi-C data and one of the four data sets on the X axis. (h) Aggregate peak analysis (APA) comparing Pore-C virtual pairwise and Hi-C pairwise contact density within 100 kb of Hi-C loop anchors. Each 10 kb by 10 kb pixel represents the total number of contacts detected across the entire loop set in a standard coordinate system centered around each loop anchor. (i) Number of structural loops called by Peakachu19 in two Hi-C replicate, SPRITE and Pore-C. (j) Precision and recall of structural loops called in Pore-C and Hi-C replicates relative to gold standard data Hi-C calls. (k) APA peaks for structural loops called in Pore-C separated by structural loop calling scores, showing stronger signal in higher confidence structural loops. (l) Stratum adjusted correlation coefficients (SCCs) between the full Hi-C dataset for GM12878 with GM12878 Pore-C, GM12878 SPRITE, after separating high order (3-way or higher) and pairwise contacts (n = 23 chromosomes for each run). For all boxplots, the centerline corresponds to the mean, box limits to the interquartile range (IQR) and the whiskers to the last datum within 1.5 * IQR.

Extended Data Fig. 2 Pore-C concatemers guide denovo human assembly correction and scaffolding.

(a) The Shasta assembler was used to generate a draft assembly using a PromethION flow cell of nanopore WGS of the HG002 GIAB sample; the resulting contigs are plotted in length order (topmost track). Next, virtual pairwise contacts from a PromethION flow cell of Pore-C of HG002 and WGS-derived HG002 contigs were provided to the 3D-DNA tool (second track from the top) to generate scaffolds. A second round of scaffolding was carried out using SALSA2 followed by the ‘Purge Haplotigs’ tool to remove regional duplications caused by un-collapsed heterozygosity (third track from the top). The resulting assembly shows high congruity with the reference genome (bottom track). (b) Illustration of the scaffolding process for chromosomes 3, 4 and 5. The top contact map shows the contact density derived from Pore-C reads mapped against length ordered contigs from the draft assembly. The bottom contact map is derived from the same Pore-C reads mapped against the final scaffolds. The center track shows how the contigs have been ordered, re-oriented and merged into the final scaffolds, which are at or near chromosome length. (c) Performance of different assembly approaches with respect to contig/scaffold size. The combination of all three methods gives the best result with a scaffold NG50 of 125 Mb. The human reference genome with scaffold gaps removed is shown for comparison. (d) Top 5 scaffold alignments for each chromosome are plotted as a proportion of non-gap bases in each chromosome, showing that most chromosomes are assembled into a single large scaffold.

Extended Data Fig. 3 Chromunity and Synergy.

(a) Example showing relationship between concatemer communities, contact frequency, and the resulting bin-set in the Chromunity analysis. The contact frequency is computed as the number of concatemers in the community within 1 kb of the given reference location. The bin-set is defined as the union of reference locations at or above 85th percentile contact frequency. (b) Volcano plots of Synergy model results for collections of random bin-sets chosen to match enhancers and promoters bin-sets in covariate space. (c) Contact map of an exemplar window with synergistic bin-sets before (top) and after (bottom) concatemer shuffling (see Methods). (d) Example distribution of concatemer order before and after shuffling in the window shown in the previous panel. Shuffled concatemers retain the original order distribution after shuffling. (e) HiCRep SCC of shuffled vs. original contact maps showing close correspondence of pairwise interaction frequencies after concatemer shuffling. The bar represents mean SCC score for chromosome 1 (n = 70 bin-sets) with standard error of mean. Each data point represents SCC score for single bin-set. (f) Volcano plots of Synergy results from sliding window Chromunity analysis of GM12878. Additional volcano plots represent identical analyses using (1) shuffled concatemers for identical bin-sets and (2) random bin-sets chosen to match candidate synergies in covariate space. (g) Fraction of synergies relative to total candidate bin-sets nominated by the concatemer community detection step of Chromunity (n = 3120 bin-sets). Shuffled (n = 2890) and random (n = 1852) analyses defined as in panel F. Fisher’s exact test was used to determine enrichment of synergy between various groups. Two-sided P values were obtained for each comparison. Error bars on bar plots represent 95% confidence intervals on the Bernoulli trial parameter. (h) Volcano plots of Synergy model results for de novo detected high order interactions for inter-chromosomal (red) and intra-chromosomal (blue) RE Chromunity derived bin-sets. Shuffled and random bin-sets defined as in F. (i) Similar to G, fraction of candidate bin-sets from sliding window Chromunity analysis of GM12878 showing synergy across inter- and intra-chromosomal interactions (n = 661) relative to shuffled concatemers (n = 661) and random bin-sets (n = 425). Fisher’s exact test was used to determine enrichment of synergy between various groups. Two-sided P values were obtained for each comparison. Error bars on bar plots represent 95% confidence intervals on the Bernoulli trial parameter. (j) Circos plots showing all inter- and intra-chromosomal synergies from Chromunity analysis of E-P targets in GM12878. The blue highlighted regions are approximate genomic locations shown to be interacting by SPRITE.

Extended Data Fig. 4 Methylation signal in Pore-C.

(a) Correlation of methylation data obtained from Pore-C with whole genome bisulfite sequencing (WGBS, r = 0.92) and (b) Nanopore whole genome sequencing (r = 0.94). (c) Comparison of methylation signal from 2 Pore-C replicates with 2 ENCODE WGBS replicates with Pearson correlation coefficients shown in the upper right triangle. Very high correlation was observed between two replicates of Pore-C, exceeding that shown for the two ENCODE WGBS replicates. (d) Haplotype-specific CpG island methylation across genes present on the Xa and Xi chromosomes. Genes that escape X chromosome inactivation (XCI, n = 29) show no difference in methylation of promoter-proximal CpG islands (CGI) (left). Genes that undergo XCI (n = 208) show significantly elevated DNA methylation on Xi compared to Xa. For all boxplots, the centerline corresponds to the mean, box limits to the interquartile range (IQR) and the whiskers to the last datum within 1.5 * IQR. (e) Haplotype-specific methylation fraction at CpG islands relative to the transcription start site of X chromosome genes at three categories of loci on Xa and Xi. A dip in DNA methylation around the promoter in the pseudo-autosomal region (PAR), which escapes inactivation. The dip is present on Xa and in both haplotypes in the PAR, but not in other Xi regions. Random regions of genome with similar widths as the promoters are also shown for both Xa and Xi. (f) Haplotype-specific contact maps on chromosome X in GM12878. The inactive X (Xi) haplotype (top heat map) lacks the A/B compartmentalization present in the active X (Xa) haplotype (lowest heat map) but instead demonstrates two megadomains. The border of these two megadomains, called the ‘hinge region,’ shows a mega-loop of ~ 16 Mb between DXZ4 and FIRRE present in Xi but absent in Xa.

Extended Data Fig. 5 LNCaP Chromunity results and controls.

(a) Volcano plot of Synergy results for bin-sets nominated by sliding-window Chromunity for LNCaP cells with (DHT+, bottom) and without (DHT-, top) androgen stimulation. Additional analyses represent negative controls: ‘Shuffled’ analysis shows Synergy results for the same candidate bin-sets using shuffled concatemers (see Methods). ‘Random’ analysis shows Synergy results on original concatemers using random bin-sets chosen to match candidate bin-sets in covariate space (see Methods). (b) Same as (a) but using regulatory element (RE) Chromunity.

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Tables 1–5.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deshpande, A.S., Ulahannan, N., Pendleton, M. et al. Identifying synergistic high-order 3D chromatin conformations from genome-scale nanopore concatemer sequencing. Nat Biotechnol 40, 1488–1499 (2022). https://doi.org/10.1038/s41587-022-01289-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01289-z

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer