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.

  • Review Article
  • Published:

Gene regulation on extrachromosomal DNA

Abstract

Oncogene amplification on extrachromosomal DNA (ecDNA) is prevalent in human cancer and is associated with poor outcomes. Clonal, megabase-sized circular ecDNAs in cancer are distinct from nonclonal, small sub-kilobase-sized DNAs that may arise during normal tissue homeostasis. ecDNAs enable profound changes in gene regulation beyond copy-number gains. An emerging principle of ecDNA regulation is the formation of ecDNA hubs: micrometer-sized nuclear structures of numerous copies of ecDNAs tethered by proteins in spatial proximity. ecDNA hubs enable cooperative and intermolecular sharing of DNA regulatory elements for potent and combinatorial gene activation. The 3D context of ecDNA shapes its gene expression potential, selection for clonal heterogeneity among ecDNAs, distribution through cell division, and reintegration into chromosomes. Technologies for studying gene regulation and structure of ecDNA are starting to answer long-held questions on the distinct rules that govern cancer genes beyond chromosomes.

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: Unique characteristics of ecDNA.
Fig. 2: ecDNA hubs drive oncogene expression and may shape cancer-cell evolution.
Fig. 3: Genetic and structural basis of the regulatory circuitry on ecDNA.
Fig. 4: Technologies used to reveal ecDNA gene regulation and structure.

Similar content being viewed by others

References

  1. Cox, D., Yuncken, C. & Spriggs, A. I. Minute chromatin bodies in malignant tumours of childhood. Lancet 286, 55–58 (1965). The original description of ecDNA in tumor cells manifesting as small chromatin bodies on chromosome spreads.

    Article  Google Scholar 

  2. Spriggs, A. I., Boddington, M. M. & Clarke, C. M. Chromosomes of human cancer cells. Br. Med J. 2, 1431–1435 (1962).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hoff, D. D. V., Needham-VanDevanter, D. R., Yucel, J., Windle, B. E. & Wahl, G. M. Amplified human MYC oncogenes localized to replicating submicroscopic circular DNA molecules. Proc. Natl Acad. Sci. USA 85, 4804–4808 (1988).

    Article  Google Scholar 

  4. Turner, K. M. et al. Extrachromosomal oncogene amplification drives tumour evolution and genetic heterogeneity. Nature 543, 122–125 (2017). Systematic analysis of human cancer models using sequencing and cytogenetics identified ecDNAs in nearly half of human cancers and not in normal cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kim, H. et al. Extrachromosomal DNA is associated with oncogene amplification and poor outcome across multiple cancers. Nat. Genet. 52, 891–897 (2020). Comprehensive analysis of primary tumors found increased oncogene transcription and worsened outcomes linked to ecDNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kohl, N. E. et al. Transposition and amplification of oncogene-related sequences in human neuroblastomas. Cell 35, 359–367 (1983).

    Article  CAS  PubMed  Google Scholar 

  7. Benner, S., Wahl, G. & Hoff, D. V. Double minute chromosomes and homogeneously staining regions in tumors taken directly from patients versus in human tumor cell lines. Anti-cancer Drugs 2, 11–26 (1991).

    Article  CAS  PubMed  Google Scholar 

  8. Bigner, S. H., Mark, J. & Bigner, D. D. Cytogenetics of human brain tumors. Cancer Genet. Cytogenetics 47, 141–154 (1990).

    Article  CAS  Google Scholar 

  9. Storlazzi, C. T. et al. Gene amplification as double minutes or homogeneously staining regions in solid tumors: origin and structure. Genome Res. 20, 1198–1206 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yoshimoto, M. et al. MYCN gene amplification: identification of cell populations containing double minutes and homogeneously staining regions in neuroblastoma tumors. Am. J. Pathol. 155, 1439–1443 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vicario, R. et al. Patterns of HER2 gene amplification and response to anti-HER2 therapies. PLoS ONE 10, e0129876 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. McGill, J. R. et al. Double minutes are frequently found in ovarian carcinomas. Cancer Genet. Cytogenetics 71, 125–131 (1993).

    Article  CAS  Google Scholar 

  13. Lin, C. C. et al. Evolution of karyotypic abnormalities and C-MYC oncogene amplification in human colonic carcinoma cell lines. Chromosoma 92, 11–15 (1985).

    Article  CAS  PubMed  Google Scholar 

  14. Wahl, G. M. The importance of circular DNA in mammalian gene amplification. Cancer Res. 49, 1333–1340 (1989).

    CAS  PubMed  Google Scholar 

  15. Quinn, L. A., Moore, G. E., Morgan, R. T. & Woods, L. K. Cell lines from human colon carcinoma with unusual cell products, double minutes, and homogeneously staining regions. Cancer Res. 39, 4914–4924 (1979).

    CAS  PubMed  Google Scholar 

  16. Carroll, S. M. et al. Double minute chromosomes can be produced from precursors derived from a chromosomal deletion. Mol. Cell. Biol. 8, 1525–1533 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Maurer, B. J., Lai, E., Hamkalo, B. A., Hood, L. & Attardi, G. Novel submicroscopic extrachromosomal elements containing amplified genes in human cells. Nature 327, 434–437 (1987).

    Article  CAS  PubMed  Google Scholar 

  18. Pauletti, G., Lai, E. & Attardi, G. Early appearance and long-term persistence of the submicroscopic extrachromosomal elements (amplisomes) containing the amplified DHFR genes in human cell lines. Proc. Natl Acad. Sci. USA 87, 2955–2959 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang, Y. et al. eccDNAs are apoptotic products with high innate immunostimulatory activity. Nature 599, 308–314 (2021).

  20. Møller, H. D. et al. Circular DNA elements of chromosomal origin are common in healthy human somatic tissue. Nat. Commun. 9, 1069 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Møller, H. D., Parsons, L., Jørgensen, T. S., Botstein, D. & Regenberg, B. Extrachromosomal circular DNA is common in yeast. Proc. Natl Acad. Sci. USA 112, E3114–E3122 (2015).

  22. Paulsen, T., Kumar, P., Koseoglu, M. M. & Dutta, A. Discoveries of extrachromosomal circles of DNA in normal and tumor cells. Trends Genet. 34, 270–278 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  24. Hung, K. L. et al. ecDNA hubs drive cooperative intermolecular oncogene expression. Nature 600, 731–736 (2021). Discovery of ecDNA hubs that enable intermolecular activation of oncogene expression through enhancer–promoter interactions.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Levan, A. & Levan, G. Have double minutes functioning centromeres? Hereditas 88, 81–92 (1978). Conclusive evidence that ecDNAs lack centromeres, which explains their distinct mode of random segregation that results in copy-number heterogeneity.

    Article  CAS  PubMed  Google Scholar 

  26. Lundberg, G. et al. Binomial mitotic segregation of MYCN-carrying double minutes in neuroblastoma illustrates the role of randomness in oncogene amplification. PLoS ONE 3, e3099 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Lange, J. T. et al. Principles of ecDNA random inheritance drive rapid genome change and therapy resistance in human cancers. Preprint at bioRxiv https://doi.org/10.1101/2021.06.11.447968 (2021).

  28. Ståhl, F., Wettergren, Y. & Levan, G. Amplicon structure in multidrug-resistant murine cells: a nonrearranged region of genomic DNA corresponding to large circular DNA. Mol. Cell. Biol. 12, 1179–1187 (1992).

    PubMed  PubMed Central  Google Scholar 

  29. Nathanson, D. A. et al. Targeted therapy resistance mediated by dynamic regulation of extrachromosomal mutant EGFR DNA. Science 343, 72–76 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Yu, M. & Ren, B. The three-dimensional organization of mammalian genomes. Annu. Rev. Dev. Cell Biol. 33, 265–289 (2017).

  31. Cremer, T. & Cremer, M. Chromosome territories. Cold Spring Harb. Perspect. Biol. 2, a003889 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Spilianakis, C. G., Lalioti, M. D., Town, T., Lee, G. R. & Flavell, R. A. Interchromosomal associations between alternatively expressed loci. Nature 435, 637–645 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Apostolou, E. & Thanos, D. Virus infection induces NF-κB-dependent interchromosomal associations mediating monoallelic IFN-β gene expression. Cell 134, 85–96 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Maass, P. G., Barutcu, A. R. & Rinn, J. L. Interchromosomal interactions: a genomic love story of kissing chromosomes. J. Cell Biol. 218, 27–38 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yi, E. et al. Live-cell imaging shows uneven segregation of extrachromosomal DNA elements and transcriptionally active extrachromosomal DNA hubs in cancer. Cancer Discov. 12, 468–483 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Itoh, N. & Shimizu, N. DNA replication-dependent intranuclear relocation of double minute chromatin. J. Cell Sci. 111, 3275–3285 (1998).

    Article  CAS  PubMed  Google Scholar 

  37. Misteli, T. Beyond the sequence: cellular organization of genome function. Cell 128, 787–800 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Kanda, T., Sullivan, K. F. & Wahl, G. M. Histone-GFP fusion protein enables sensitive analysis of chromosome dynamics in living mammalian cells. Curr. Biol. 8, 377–385 (1998).

    Article  CAS  PubMed  Google Scholar 

  39. Oobatake, Y. & Shimizu, N. Double-strand breakage in the extrachromosomal double minutes triggers their aggregation in the nucleus, micronucleation, and morphological transformation. Genes Chromosomes Cancer 59, 133–143 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Chong, S. et al. Imaging dynamic and selective low-complexity domain interactions that control gene transcription. Science 361, eaar2555 (2018).

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Gibson, B. A. et al. Organization of chromatin by intrinsic and regulated phase separation. Cell 179, 470–484 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Shin, Y. et al. Liquid nuclear condensates mechanically sense and restructure the genome. Cell 175, 1481–1491 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lovén, J. et al. Selective inhibition of tumor oncogenes by disruption of super-enhancers. Cell 153, 320–334 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Henssen, A. et al. Targeting MYCN-driven transcription by BET-bromodomain inhibition. Clin. Cancer Res. 22, 2470–2481 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Morton, A. R. et al. Functional enhancers shape extrachromosomal oncogene amplifications. Cell 179, 1330–1341 (2019). Description of oncogene-enhancer co-amplification on ecDNAs, showing hijacking of both cognate and ectopic enhancers to drive oncogene expression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tanabe, H. et al. Evolutionary conservation of chromosome territory arrangements in cell nuclei from higher primates. Proc. Natl Acad. Sci. USA 99, 4424–4429 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Helmsauer, K. et al. Enhancer hijacking determines extrachromosomal circular MYCN amplicon architecture in neuroblastoma. Nat. Commun. 11, 5823 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Nikolaev, S. et al. Extrachromosomal driver mutations in glioblastoma and low-grade glioma. Nat. Commun. 5, 5690 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Risca, V. I., Denny, S. K., Straight, A. F. & Greenleaf, W. J. Variable chromatin structure revealed by in situ spatially correlated DNA cleavage mapping. Nature 541, 237–241 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Fragkos, M., Ganier, O., Coulombe, P. & Méchali, M. DNA replication origin activation in space and time. Nat. Rev. Mol. Cell Biol. 16, 360–374 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Carré-Simon, À. & Fabre, E. 3D genome organization: causes and consequences for DNA damage and repair. Genes 13, 7 (2022).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Deshpande, V. et al. Exploring the landscape of focal amplifications in cancer using AmpliconArchitect. Nat. Commun. 10, 392 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Luebeck, J. et al. AmpliconReconstructor integrates NGS and optical mapping to resolve the complex structures of focal amplifications. Nat. Commun. 11, 4374 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Xu, K. et al. Structure and evolution of double minutes in diagnosis and relapse brain tumors. Acta Neuropathol. 137, 123–137 (2019).

    Article  PubMed  Google Scholar 

  57. L’Abbate, A. et al. Genomic organization and evolution of double minutes/homogeneously staining regions with MYC amplification in human cancer. Nucleic Acids Res. 42, 9131–9145 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Shoshani, O. et al. Chromothripsis drives the evolution of gene amplification in cancer. Nature 591, 137–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Lupiáñez, D. G. et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 161, 1012–1025 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Uslu, V. V. et al. Long-range enhancers regulating Myc expression are required for normal facial morphogenesis. Nat. Genet. 46, 753–758 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Franke, M. et al. Formation of new chromatin domains determines pathogenicity of genomic duplications. Nature 538, 265–269 (2016).

    Article  CAS  PubMed  Google Scholar 

  62. 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 

  63. Pang, J. et al. Extrachromosomal DNA in HPV mediated oropharyngeal cancer drives diverse oncogene transcription. Clin. Cancer Res. 27, 6772–6786 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. 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 

  65. 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 

  66. 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 

  67. Schwab, M., Klempnauer, K. H., Alitalo, K., Varmus, H. & Bishop, M. Rearrangement at the 5′ end of amplified c-myc in human COLO 320 cells is associated with abnormal transcription. Mol. Cell. Biol. 6, 2752–2755 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. 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 

  69. Northcott, P. A. et al. Subgroup-specific structural variation across 1,000 medulloblastoma genomes. Nature 488, 49–56 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Tolomeo, D., Agostini, A., Visci, G., Traversa, D. & Storlazzi, C. T. PVT1: a long non-coding RNA recurrently involved in neoplasia-associated fusion transcripts. Gene 779, 145497 (2021).

    Article  CAS  PubMed  Google Scholar 

  71. Kalyana-Sundaram, S. et al. Gene fusions associated with recurrent amplicons represent a class of passenger aberrations in breast cancer. Neoplasia 14, 702–708 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Robinson, D. R. et al. Integrative clinical genomics of metastatic cancer. Nature 548, 297–303 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Chapman, O. S. et al. The landscape of extrachromosomal circular DNA in medulloblastoma. Preprint at bioRxiv https://doi.org/10.1101/2021.10.18.464907 (2021).

  74. 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 

  75. Sanyal, A., Lajoie, B. R., Jain, G. & Dekker, J. The long-range interaction landscape of gene promoters. Nature 489, 109–113 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Ganji, M. et al. Real-time imaging of DNA loop extrusion by condensin. Science 360, 102–105 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Terakawa, T. et al. The condensin complex is a mechanochemical motor that translocates along DNA. Science 358, 672–676 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Hamkalo, B. A., Farnham, P. J., Johnston, R. & Schimke, R. T. Ultrastructural features of minute chromosomes in a methotrexate-resistant mouse 3T3 cell line. Proc. Natl Acad. Sci. USA 82, 1126–1130 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. van der Bliek, A. M., Lincke, C. R. & Borst, P. Circular DNA of 3T6R50 double minute chromosomes. Nucleic Acids Res. 16, 4841–4851 (1988).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Rattner, J. B. & Lin, C. C. Ultrastructural organization of double minute chromosomes and HSR regions in human colon carcinoma cells. Cytogenetic Genome Res. 38, 176–181 (1984).

    Article  CAS  Google Scholar 

  82. VanDevanter, D. R., Piaskowski, V. D., Casper, J. T., Douglass, E. C. & Von Hoff, D. D. Ability of circular extrachromosomal DNA molecules to carry amplified MYCN protooncogenes in human neuroblastomas in vivo. J. Natl Cancer Inst. 82, 1815–1821 (1990).

    Article  CAS  PubMed  Google Scholar 

  83. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  84. Nagano, T. et al. Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature 547, 61–67 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Stevens, T. J. et al. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature 544, 59–64 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Hung, K. L. et al. Targeted profiling of human extrachromosomal DNA by CRISPR–CATCH. Preprint at bioRxiv https://doi.org/10.1101/2021.11.28.470285 (2022).

  88. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  89. Creyghton, M. P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl Acad. Sci. USA 107, 21931–21936 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Mumbach, M. R. et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Mumbach, M. R. et al. Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements. Nat. Genet. 49, 1602–1612 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Tang, Z. et al. CTCF-mediated human 3D genome architecture reveals chromatin topology for transcription. Cell 163, 1611–1627 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Lareau, C. A. et al. Droplet-based combinatorial indexing for massive-scale single-cell chromatin accessibility. Nat. Biotechnol. 37, 916–924 (2019).

    Article  CAS  PubMed  Google Scholar 

  95. Chen, S., Lake, B. B. & Zhang, K. High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell. Nat. Biotechnol. 37, 1452–1457 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ma, S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  98. Jiang, W. et al. Cas9-assisted targeting of chromosome segments CATCH enables one-step targeted cloning of large gene clusters. Nat. Commun. 6, 1–8 (2015).

    Google Scholar 

  99. Overhauser, J. in Pulsed-Field Gel Electrophoresis: Protocols, Methods, and Theories (eds. Burmeister, M. & Ulanovsky, L.) 129–134 (Humana Press, 1992).

  100. Cao, H. et al. Rapid detection of structural variation in a human genome using nanochannel-based genome mapping technology. GigaScience 3, 34 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  101. Baskin, F., Rosenberg, R. N. & Dev, V. Correlation of double-minute chromosomes with unstable multidrug cross-resistance in uptake mutants of neuroblastoma cells. Proc. Natl Acad. Sci. USA 78, 3654–3658 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Møller, H. D., Parsons, L., Jørgensen, T. S., Botstein, D. & Regenberg, B. Extrachromosomal circular DNA is common in yeast. Proc. Natl Acad. Sci. USA 112, E3114–E3122 (2015).

  103. Shoura, M. J. et al. Intricate and cell type-specific populations of endogenous circular DNA (eccDNA) in Caenorhabditis elegans and Homo sapiens. G3 7, 3295–3303 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Smith, C. A. & Vinograd, J. Small polydisperse circular DNA of HeLa cells. J. Mol. Biol. 69, 163–178 (1972).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

H.Y.C. is supported by US National Institutes of Health grant R35-CA209919 and is an Investigator of the Howard Hughes Medical Institute. P.S.M. is supported in part by grants U24CA264379 and R01 CA238249 from the US National Institutes of Health. H.Y.C. and P.S.M. are supported by Cancer Grand Challenges CGCSDF-2021\100007 with support from Cancer Research UK and the National Cancer Institute. K.L.H. is supported by a Stanford Graduate Fellowship.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Howard Y. Chang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Anton Henssen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Beth Moorefield, Tiago Faial and Carolina Perdigoto, in collaboration with the Nature Structural & Molecular Biology team.

Additional information

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

Rights and permissions

Springer Nature or its licensor 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

Hung, K.L., Mischel, P.S. & Chang, H.Y. Gene regulation on extrachromosomal DNA. Nat Struct Mol Biol 29, 736–744 (2022). https://doi.org/10.1038/s41594-022-00806-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00806-7

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