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:

Chromothripsis as an on-target consequence of CRISPR–Cas9 genome editing

Abstract

Genome editing has therapeutic potential for treating genetic diseases and cancer. However, the currently most practicable approaches rely on the generation of DNA double-strand breaks (DSBs), which can give rise to a poorly characterized spectrum of chromosome structural abnormalities. Here, using model cells and single-cell whole-genome sequencing, as well as by editing at a clinically relevant locus in clinically relevant cells, we show that CRISPR–Cas9 editing generates structural defects of the nucleus, micronuclei and chromosome bridges, which initiate a mutational process called chromothripsis. Chromothripsis is extensive chromosome rearrangement restricted to one or a few chromosomes that can cause human congenital disease and cancer. These results demonstrate that chromothripsis is a previously unappreciated on-target consequence of CRISPR–Cas9-generated DSBs. As genome editing is implemented in the clinic, the potential for extensive chromosomal rearrangements should be considered and monitored.

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: Micronucleation is an on-target consequence of CRISPR–Cas9 genome editing.
Fig. 2: Summary of genomic outcomes after the division of 18 micronucleated cells.
Fig. 3: CRISPR–Cas9 genome editing can cause chromothripsis.
Fig. 4: The impact of p53 status on the ability of micronucleated cells to undergo division.
Fig. 5: CRISPR–Cas9 genome editing induces chromosome bridge formation, adding to the genome complexity from micronuclei.
Fig. 6: Hallmark cytological features of chromothripsis after a genome-editing approach for the treatment of sickle cell disease.

Similar content being viewed by others

Data availability

Whole images of cells presented in Figs. 1f and 6e,i and Extended Data Fig. 2e and filtered SV calls are available at https://doi.org/10.5281/zenodo.4533299. Original images and videos that contribute to analyses in Figs. 1e, 4 and 6c,h, Extended Data Figs. 1d, 2 and 5c and Look-Seq experiments (Figs. 2, 3 and 5 and Extended Data Fig. 3) were not published due to constraints of file size but are available upon reasonable request. CD34+ HSPC-derived FISH and SKY images and analyses (Fig. 6d–g) were generated by the St. Jude Cytogenetic Shared Resource Laboratory and derived data supporting the findings in Fig. 6d–g are available from the corresponding authors upon request. Sequence read data are available in the Sequencing Read Archive under BioProject PRJNA676146. Source data are provided with this paper.

Code availability

Scripts used for sequencing data analysis (allelic copy number calculation and rearrangement detection) and for image analyses performed in Extended Data Fig. 2 are available at https://github.com/chengzhongzhangDFCI/CN_and_SV.

References

  1. Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR–Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Doudna, J. A. The promise and challenge of therapeutic genome editing. Nature 578, 229–236 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Xu, J. et al. Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing. Science 334, 993–996 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Orkin, S. H. & Bauer, D. E. Emerging genetic therapy for sickle cell disease. Annu. Rev. Med. 70, 257–271 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Wu, Y. et al. Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat. Med. 25, 776–783 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Frangoul, H. et al. CRISPR–Cas9 gene editing for sickle cell disease and β-thalassemia. N. Engl. J. Med. 384, 252–260 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Dever, D. P. et al. CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature 539, 384–389 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. DeWitt, M. A. et al. Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells. Sci. Transl. Med. 8, 360ra134 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Richardson, C. D. et al. CRISPR–Cas9 genome editing in human cells occurs via the Fanconi anemia pathway. Nat. Genet. 50, 1132–1139 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Romero, Z. et al. Editing the sickle cell disease mutation in human hematopoietic stem cells: comparison of endonucleases and homologous donor templates. Mol. Ther. 27, 1389–1406 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kim, D., Luk, K., Wolfe, S. A. & Kim, J. S. Evaluating and enhancing target specificity of gene-editing nucleases and deaminases. Annu. Rev. Biochem. 88, 191–220 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927–930 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Ihry, R. J. et al. p53 inhibits CRISPR–Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939–946 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. van den Berg, J. et al. A limited number of double-strand DNA breaks is sufficient to delay cell cycle progression. Nucleic Acids Res. 46, 10132–10144 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Enache, O. M. et al. Cas9 activates the p53 pathway and selects for p53-inactivating mutations. Nat. Genet. 52, 662–668 (2020).

  19. Whitworth, K. M. et al. Use of the CRISPR/Cas9 system to produce genetically engineered pigs from in vitro-derived oocytes and embryos. Biol. Reprod. 91, 78 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Shin, H. Y. et al. CRISPR/Cas9 targeting events cause complex deletions and insertions at 17 sites in the mouse genome. Nat. Commun. 8, 15464 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Adikusuma, F. et al. Large deletions induced by Cas9 cleavage. Nature 560, E8–E9 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765–771 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zuccaro, M. V. et al. Allele-specific chromosome removal after Cas9 cleavage in human embryos. Cell 183, 1650–1664 (2020).

  24. Weisheit, I. et al. Detection of deleterious on-target effects after HDR-mediated CRISPR editing. Cell Rep. 31, 107689 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Alanis-Lobato, G. et al. Frequent loss-of-heterozygosity in CRISPR–Cas9-edited early human embryos. Preprint at bioRxiv https://doi.org/10.1101/2020.06.05.135913 (2020).

  26. Cullot, G. et al. CRISPR–Cas9 genome editing induces megabase-scale chromosomal truncations. Nat. Commun. 10, 1136 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Stadtmauer, E. A. et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Zhang, C. Z. et al. Chromothripsis from DNA damage in micronuclei. Nature 522, 179–184 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Umbreit, N. T. et al. Mechanisms generating cancer genome complexity from a single cell division error. Science 368, eaba0712 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu, P. et al. Chromosome catastrophes involve replication mechanisms generating complex genomic rearrangements. Cell 146, 889–903 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kloosterman, W. P. & Cuppen, E. Chromothripsis in congenital disorders and cancer: similarities and differences. Curr. Opin. Cell Biol. 25, 341–348 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ly, P. et al. Chromosome segregation errors generate a diverse spectrum of simple and complex genomic rearrangements. Nat. Genet. 51, 705–715 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Consortium, I. T. P.-C. Ao. W. G. Pan-cancer analysis of whole genomes. Nature 578, 82–93 (2020).

    Article  CAS  Google Scholar 

  36. Cortes-Ciriano, I. et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat. Genet. 52, 331–341 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Leibowitz, M. L., Zhang, C. Z. & Pellman, D. Chromothripsis: a new mechanism for rapid karyotype evolution. Annu. Rev. Genet. 49, 183–211 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Ly, P. & Cleveland, D. W. Rebuilding chromosomes after catastrophe: emerging mechanisms of chromothripsis. Trends Cell Biol. 27, 917–930 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Soto, M., Garcia-Santisteban, I., Krenning, L., Medema, R. H. & Raaijmakers, J. A. Chromosomes trapped in micronuclei are liable to segregation errors. J. Cell Sci. 131, jcs214742 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. 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 

  41. McKinley, K. L. & Cheeseman, I. M. Large-scale analysis of CRISPR/Cas9 cell-cycle knockouts reveals the diversity of p53-dependent responses to cell-cycle defects. Dev. Cell 40, 405–420 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Brinkman, E. K. et al. Kinetics and fidelity of the repair of Cas9-induced double-strand DNA breaks. Mol. Cell 70, 801–813 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wu, J., Tang, B. & Tang, Y. Allele-specific genome targeting in the development of precision medicine. Theranostics 10, 3118–3137 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Stark, J. M. & Jasin, M. Extensive loss of heterozygosity is suppressed during homologous repair of chromosomal breaks. Mol. Cell Biol. 23, 733–743 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rao, P. N., Johnson, R. T. & Sperling, K. Premature Chromosome Condensation: Application in Basic, Clinical, and Mutation Research xvi (Academic Press, 1982).

    Book  Google Scholar 

  46. Hoffelder, D. R. et al. Resolution of anaphase bridges in cancer cells. Chromosoma 112, 389–397 (2004).

    Article  PubMed  Google Scholar 

  47. Terradas, M., Martin, M., Tusell, L. & Genesca, A. DNA lesions sequestered in micronuclei induce a local defective-damage response. DNA Repair 8, 1225–1234 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Crasta, K. et al. DNA breaks and chromosome pulverization from errors in mitosis. Nature 482, 53–58 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hatch, E. M., Fischer, A. H., Deerinck, T. J. & Hetzer, M. W. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 154, 47–60 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ly, P. et al. Selective Y centromere inactivation triggers chromosome shattering in micronuclei and repair by non-homologous end joining. Nat. Cell Biol. 19, 68–75 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Liu, S. et al. Nuclear envelope assembly defects link mitotic errors to chromothripsis. Nature 561, 551–555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kneissig, M. et al. Micronuclei-based model system reveals functional consequences of chromothripsis in human cells. eLife 8, e50292 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 575, 210–216 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ikeda, K. et al. Efficient scarless genome editing in human pluripotent stem cells. Nat. Methods 15, 1045–1047 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. Liang, D. et al. Frequent gene conversion in human embryos induced by double strand breaks. Preprint at bioRxiv https://doi.org/10.1101/2020.06.19.162214 (2020).

  56. Korbel, J. O. & Campbell, P. J. Criteria for inference of chromothripsis in cancer genomes. Cell 152, 1226–1236 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Vazquez-Diez, C., Yamagata, K., Trivedi, S., Haverfield, J. & FitzHarris, G. Micronucleus formation causes perpetual unilateral chromosome inheritance in mouse embryos. Proc. Natl Acad. Sci. USA 113, 626–631 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Minocherhomji, S. et al. Replication stress activates DNA repair synthesis in mitosis. Nature 528, 286–290 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Cleal, K., Jones, R. E., Grimstead, J. W., Hendrickson, E. A. & Baird, D. M. Chromothripsis during telomere crisis is independent of NHEJ, and consistent with a replicative origin. Genome Res. 29, 737–749 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Maciejowski, J., Li, Y., Bosco, N., Campbell, P. J. & de Lange, T. Chromothripsis and kataegis induced by telomere crisis. Cell 163, 1641–1654 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Maciejowski, J. et al. APOBEC3-dependent kataegis and TREX1-driven chromothripsis during telomere crisis. Nat. Genet. 52, 884–890 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ribeyre, C. & Shore, D. Regulation of telomere addition at DNA double-strand breaks. Chromosoma 122, 159–173 (2013).

    Article  CAS  PubMed  Google Scholar 

  63. Maciejowski, J. & de Lange, T. Telomeres in cancer: tumour suppression and genome instability. Nat. Rev. Mol. Cell Biol. 18, 175–186 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Canela, A. et al. DNA breaks and end resection measured genome-wide by end sequencing. Mol. Cell 63, 898–911 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. McClintock, B. The stability of broken ends of chromosomes in Zea mays. Genetics 26, 234–282 (1941).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Campbell, P. J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Li, Y. et al. Constitutional and somatic rearrangement of chromosome 21 in acute lymphoblastic leukaemia. Nature 508, 98–102 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ma, H. et al. Correction of a pathogenic gene mutation in human embryos. Nature 548, 413–419 (2017).

    Article  CAS  PubMed  Google Scholar 

  69. Egli, D. et al. Inter-homologue repair in fertilized human eggs? Nature 560, E5–E7 (2018).

    Article  CAS  PubMed  Google Scholar 

  70. Finn, J. D. et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 22, 2227–2235 (2018).

    Article  CAS  PubMed  Google Scholar 

  71. Humbert, O., Peterson, C. W., Norgaard, Z. K., Radtke, S. & Kiem, H. P. A nonhuman primate transplantation model to evaluate hematopoietic stem cell gene editing strategies for β-hemoglobinopathies. Mol. Ther. Methods Clin. Dev. 8, 75–86 (2018).

    Article  CAS  PubMed  Google Scholar 

  72. Humbert, O. et al. Therapeutically relevant engraftment of a CRISPR–Cas9-edited HSC-enriched population with HbF reactivation in nonhuman primates. Sci. Transl. Med. 11, eaaw3768 (2019).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  73. Demirci, S. et al. BCL11A enhancer-edited hematopoietic stem cells persist in rhesus monkeys without toxicity. J. Clin. Invest. 130, 6677–6687 (2020).

  74. Lu, Y. et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat. Med. 26, 732–740 (2020).

    Article  CAS  PubMed  Google Scholar 

  75. Luc, S. et al. Bcl11a deficiency leads to hematopoietic stem cell defects with an aging-like phenotype. Cell Rep. 16, 3181–3194 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Sanders, A. D. et al. Single-cell analysis of structural variations and complex rearrangements with tri-channel processing. Nat. Biotechnol. 38, 343–354 (2020).

    Article  CAS  PubMed  Google Scholar 

  77. McDermott, D. H. et al. Chromothriptic cure of WHIM syndrome. Cell 160, 686–699 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Maeder, M. L. et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat. Med. 25, 229–233 (2019).

    Article  CAS  PubMed  Google Scholar 

  79. Lomova, A. et al. Improving gene editing outcomes in human hematopoietic stem and progenitor cells by temporal control of DNA repair. Stem Cells 37, 284–294 (2019).

    Article  CAS  PubMed  Google Scholar 

  80. Metais, J. Y. et al. Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. Blood Adv. 3, 3379–3392 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Weber, L. et al. Editing a γ-globin repressor binding site restores fetal hemoglobin synthesis and corrects the sickle cell disease phenotype. Sci. Adv. 6, eaay9392 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Howden, S. E. et al. A Cas9 variant for efficient generation of indel-free knockin or gene-corrected human pluripotent stem cells. Stem Cell Rep. 7, 508–517 (2016).

    Article  CAS  Google Scholar 

  83. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Connelly, J. P. & Pruett-Miller, S. M. CRIS.py: a versatile and high-throughput analysis program for CRISPR-based genome editing. Sci. Rep. 9, 4194 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to R. Jaenisch, S. Markoulaki, A. Spektor and members of the Pellman and Weiss laboratories for discussions, I. Cheeseman for the doxycycline-inducible Cas9 RPE-1 cell line, D. Cullins for assistance with single-cell sorting of HSPCs and N. Mynhier for help with data visualization. This work was supported by the National Science Foundation Graduate Research Fellowship under grant no. DGE1144152 (M.L.L.), the National Cancer Institute career transition award K22CA216319 (C.-Z.Z.), the Howard Hughes Medical Institute (D.P.), NIH grants R01 CA213404 (D.P.), F32 DK118822 (P.A.D.) and P01 HL053749 (M.J.W.), the Assisi Foundation (M.J.W.), the Doris Duke Charitable Foundation (M.J.W.) and St. Jude/ALSAC. The St. Jude Cytogenetic and Center for Advanced Genome Engineering Shared Resource Laboratories are supported by NIH grant P30 CA21765 and by St. Jude/ALSAC. We thank the members of the St. Jude Children’s Research Hospital Center for Advanced Genome Engineering and Cytogenetics core facilities.

Author information

Authors and Affiliations

Authors

Contributions

M.L.L., S.P. and D.P. conceived the project; M.L.L., S.P., D.P. and P.A.D. designed the experiments; M.L.L. and S.P. performed the experiments, except for the human CD34+ HSPC experiments that were carried out by P.A.D. and Y.Y.; L.S. performed library preparation and sequencing for RPE-1 cells. M.L.L., S.P., L.J.B. and C.-Z.Z. analyzed data; C.-Z.Z. and L.J.B. developed and performed the computational analysis; M.L.L., S.P. and D.P. wrote the manuscript; all authors discussed the results and commented on the manuscript; M.J.W. supervised the human blood cell experiments; P.A.D. and L.J.B. made equal contributions to this work. D.P. supervised the study.

Corresponding authors

Correspondence to Mitchell J. Weiss or David Pellman.

Ethics declarations

Competing interests

M.J.W. is a consultant for Rubius Inc., Cellarity Inc., Beam Therapeutics and Esperion; none of the consulting work is relevant to the current project. C.-Z.Z. is a scientific adviser for Pillar BioSciences. D.P. is a member of the Volastra Therapeutics scientific advisory board. All other authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Fyodor Urnov and the other, anonymous, reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Micronucleus formation after CRISPR-Cas9 genome editing in several cell lines.

a, Experimental schemes. Top, RNP transfection. Bottom, inducible Cas9 expression with constitutive expression of gRNAs (RPE-1 cells). G0 cell cycle block was by serum starvation. Dividing cell cartoon represents approximate time of cell division. b, Micronucleation frequency after CRISPR-Cas9 RNP transfection in asynchronous cells. Left, editing efficiency. Right, frequency of micronucleation for these RNP transfections. (n = 3 experiments with 1339, 1231, 1220, 1236, and 1237 cells scored, left to right). Error bars: mean +/- SEM, two-tailed Fisher’s exact test. c, Representative Western blot of Cas9 levels at the indicated times after induction with doxycycline. 1st division is 24 hours after serum starve release, and 2nd division is 48 hours after release. Dox is doxycycline. n = 3 experiments. d, Number of cleaved chromosome arms contained within micronuclei for the indicated gRNAs and Cas9 expression strategies (RPE-1 cells) determined by FISH to detect the centromere (RNP Cas9) and/or subtelomere of the targeted chromosome (RNP Cas9 and Dox-inducible Cas9). RNP Cas9: for 2p: n = 2 experiments with 64 micronuclei counted, 4q: n = 2 experiments with 58 micronuclei counted, 5q: n= 3 experiments with 116 micronuclei counted, Xq: n = 2 experiments with 96 micronuclei counted; (Dox) Doxycycline-inducible Cas9; n = 3 experiments; 168 micronuclei counted per condition. e, Frequency of micronucleation in synchronized BJ fibroblasts after RNP transfection; (n = 3 experiments with 2378, 2487, 2423, 2714 cells, left to right). Error bars: mean +/- SEM, two-tailed Fisher’s exact test. f, Left, percentage of MN containing the targeted chromosome arm for the chr5q-targeting gRNA in BJ cells, as counted using subtelomeric FISH probes. Right, the number of chr5q chromosome arms per micronucleus in BJ cells, determined from centromere-specific and subtelomere-specific FISH probes. (n = 2 experiments counting 109 micronuclei). g, Cut site and FISH probe locations for allele-specific gRNA experiments. PAM sequence is in bold, with the polymorphic site in red. Orange star is the centromere FISH probe and green circle the subtelomere FISH probe. gRNAs target the reference allele. h, Editing efficiency after Cas9/gRNA RNP transfection with allele-specific gRNAs. (n = 3 experiments). Error bars: mean +/- SEM. i, Micronucleation frequency from samples in (h). (n = 3 experiments with 7066, 7041, 7253, cells scored for micronucleation, left to right). Error bars: mean +/- SEM, two-tailed Fisher’s exact test. (j) Left, percentage of MN containing the targeted chromosome arm for the allele-specific gRNAs, as scored using subtelomeric FISH probes. Right, pie chart of the number of targeted arms per micronucleus in RPE-1 cells, as determined from subtelomere-specific FISH probes. (n = 3 experiments counting 123 and 184 micronuclei, left to right) Error bars: mean +/- SEM.

Source data

Extended Data Fig. 2 DNA damage, nuclear envelope rupture and reduced DNA replication in CRISPR-MN.

a, Nuclear envelope rupture frequency for CRISPR-MN as compared to spindle checkpoint inhibitor-induced micronuclei. Rupture was defined as an MN:PN ratio of lamin B receptor (LBR)49 intensity > 3 (n = 3 experiments with 201 and 167 micronuclei analyzed for chr5q, p = 0.2216 and 165 and 152 micronuclei counted for chr6q, p = 0.2034). Error bars: mean +/- SEM, two-tailed Fisher’s exact test. b, DNA replication defect of CRISPR-MN. EdU fluorescence intensity was measured after a 5-hour pulse. Only cells that had entered S-phase were scored (>150 a.u. EdU signal in primary nucleus). Dotted red line is normal levels of DNA replication in the micronucleus relative to the primary nucleus (n = 3 experiments with 109 and 97 micronucleated cells analyzed for chr5q, p = 0.1698 and 65 and 73 micronucleated cells analyzed for chr6q, p = 0.6948). Error bars: mean +/- SEM; two-tailed Mann-Whitney U-test. c, CRISPR-MN acquire DNA damage. Shown is the frequency of γH2AX positive micronuclei (> 3 standard deviations above mean signal in primary nuclei) for the indicated gRNAs using the inducible Cas9 system (n = 3 experiments with 203 and 184 micronucleated cells analyzed for chr5q, p = 0.6870 and 175 and 169 cells analyzed for chr6q, p = 0.8053). Error bars: mean +/- SEM, two-tailed Fisher’s exact test. d, CRISPR-MN acquire DNA damage (RNP Cas9 system). Shown is the frequency of γH2AX positive micronuclei for the indicated gRNAs (n = 2 experiments with 56, 46, 82, and 50 micronucleated cells analyzed, left to right). e, Example images of data from panel d, showing γH2AX labeling. White arrows: micronuclei. Scale bars, 5 μm. The γH2AX focus in the primary nucleus likely decorates the centric portion of the broken chromosome. Alternatively, or additionally, it may label a DNA break on the homolog.

Source data

Extended Data Fig. 3 Haplotype copy number and SVs for the targeted chromosome for each sample in the paper.

Haplotype-resolved copy number and structural variant analysis for the targeted chromosome for each granddaughter pair. Red and blue dots represent 1 Mb copy number bins for each homolog, and curved lines represent structural variants of ≥ 1 Mb that could be on either homolog. Top, ‘granddaughter a’; middle, ‘granddaughter b’; bottom, sum copy number for each homolog for the pair of cells. Note that in most cases there should be a total of two red and two blue copies per granddaughter pair, and deviation from this represents certain missegregation or events, such as first-generation bridge formation. Copy number alterations occurring only in one daughter without a corresponding or reciprocal change in the other daughter were attributed to random noise due to variability in genome amplification quality. Text: inferred most likely explanation for each copy number and rearrangement profile. Note that alternative explanations exist for many samples, such as a G1 cut followed by replication of the cut chromosome.

Extended Data Fig. 4 Clustering of DNA breakpoints, indicative of chromothripsis, on the telomeric side of the CRISPR-Cas9-targeted cut site.

Breakpoint density for each daughter pair telomeric of the cut-site (red), relative to the rest of the genome (black), normalized by read depth. Data include both inter- and intra-chromosomal rearrangements. Significance is derived from a one-sided Poisson test28. p – values are rounded to the nearest exponent, except for those <10−30. Bolded p - values denote significance after Bonferroni correction. Bonferroni-corrected a = 0.0028.

Source data

Extended Data Fig. 5 Chromosome bridge formation after CRISPR-Cas9 genome editing.

a, A bridge formed during the first cell division after Cas9 addition yields shared losses (left granddaughter pair) or gains (right granddaughter pair) depending upon how the bridge breaks. This copy number alteration will be on the centromeric side of the CRISPR-Cas9 break. Cells and chromosomes are depicted as in Fig. 3. The non-micronucleated daughter cell is faded and not followed. In this example, the micronuclear chromosome from the first division is not reincorporated and becomes a micronucleus in one granddaughter. b, A bridge formed in the second cell division yields reciprocal copy number gains and losses centromeric of the break (comparing the granddaughters). The non-micronucleated daughter cell is faded and not followed. c, The frequency of detectable chromosome bridges by live-cell imaging after CRISPR-Cas9 genome editing in RPE-1 cells expressing a fluorescence reporter that marks chromosome bridges efficiently (GFP-BAF). DNA breaks were induced with the Chr5q-targeting inducible Cas9 system after treatment with siRNA against TP53 or non-targeting siRNA. Chromosome bridges frequently arise when a micronucleus forms in at least one daughter cell in the first division (MN+), whereas when a micronucleus is not formed, bridge formation is uncommon (MN-). In the second division, micronucleated cells are more prone to bridge formation (MN+) as compared to non-micronucleated cells (MN-). Bridge formation is more frequent in the second division, which may be explained by isolation of the acentric arm from the centric fragment of the chromosome (p53 siRNA: n = 6 experiments with 175 and 172 cell divisions imaged [division 1] and 136 and 132 divisions imaged [division 2]; non-targeting siRNA: n = 3 experiments with 89 and 90 cell divisions imaged [division 1] and 43 and 58 divisions imaged [division 2]). Error bars: mean +/- SEM, two-tailed Fisher’s exact test.

Source data

Extended Data Fig. 6 Allele ratios of heterozygous SNPs from CD34+ HSPC colonies after editing.

a, Map of SNP locations, cut site, and the centromere (CEN) on chromosome 2 (not to scale). b, The distribution of A-allele frequencies for samples where A-allele and B-allele frequencies comprise greater than 90 % of the sequence reads. The p-values for SNPs 1–8 are p = 0.1089, 0.3140, 0.9967, 0.7792. 0.2751, 0.4659, 0.3178, and 0.2239 respectively (two-tailed Mann-Whitney U test). SNP5 exhibited a strong deviation from a 50:50 allelic ratio even in unedited controls, which may reflect a PCR amplification artifact. Because of this, SNP5 was excluded from subsequent analysis. c, Heatmap of allele frequency data for all samples (Cas9, left; Cas9 + Chr2p gRNA, right). The heatmap is divided into sections based on the minimum sequencing read depth. Minimum sequencing read depth was defined by the SNP with the lowest number of reads in the sample. Samples with low read depth exhibited high variability in allelic ratios, likely reflecting low input DNA from small colonies. Because we lack phasing information, any deviation from a 50:50 allele ratio for multiple adjacent SNPs suggests segmental copy number alterations. See Supplementary Note for methods and additional discussion. For this experiment, only several hundred clones could feasibly be grown and analyzed, whereas patients will receive tens to hundreds of millions of edited cells. From the several hundred clones in our experiment, we only expect ~20 cells containing micronuclei based on micronucleation rates measured in Fig. 6. Extrapolating from these data, patients will receive millions of micronucleated cells, each one with the potential to undergo chromothripsis and grow into a clone. We note that this assay will not detect copy-number neutral chromothripsis nor chromothripsis that maintains copy number and heterozygosity at the assayed SNPs, with rearrangements located on other segments of the edited chromosome. Moreover, this approach has a limited ability to detect copy number gains or subclonal events that result from ongoing genomic instability triggered by micronucleation or bridging derived from the initial editing.

Source data

Supplementary information

Supplementary Information

Supplementary Note

Reporting Summary

Supplementary Video 1

Representative Look-Seq video of a reincorporated micronucleus, as in Fig. 3a,b (GFP–H2B). Pair 5.6. Timestamp shows relative time in minutes. Widefield imaging under a ×20 objective.

Supplementary Video 2

Representative Look-Seq video of a persistent micronucleus, as in Fig. 3c,d (GFP–H2B). Pair 5.10. Timestamp shows relative time in minutes. Widefield imaging under a ×20 objective.

Supplementary Tables

Supplementary Table 1. Summary of gRNA species. Table of gRNA sequences and coordinates used in this study. Supplementary Table 2. One-sided Poisson test for enrichment of rearrangements across all chromosome arms. One-sided Poisson tests were performed to test for enrichment of rearrangement intrachromosomal breakpoints on each chromosome arm in all granddaughter pairs. The table includes breakpoints present in each genome and on the targeted arm. Also included is the fraction of total genomic reads aligning to each arm. Supplementary Table 3. Primer sequences for editing efficiency analysis. Table of forward and reverse primers used to amplify DNA for analyses of editing efficiency.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1c

Unprocessed western blot.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leibowitz, M.L., Papathanasiou, S., Doerfler, P.A. et al. Chromothripsis as an on-target consequence of CRISPR–Cas9 genome editing. Nat Genet 53, 895–905 (2021). https://doi.org/10.1038/s41588-021-00838-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00838-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research