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.

  • Protocol
  • Published:

Genome editing using FACS enrichment of nuclease-expressing cells and indel detection by amplicon analysis

Abstract

This protocol describes methods for increasing and evaluating the efficiency of genome editing based on the CRISPR–Cas9 (clustered regularly interspaced short palindromic repeats–CRISPR-associated 9) system, transcription activator-like effector nucleases (TALENs) or zinc-finger nucleases (ZFNs). First, Indel Detection by Amplicon Analysis (IDAA) determines the size and frequency of insertions and deletions elicited by nucleases in cells, tissues or embryos through analysis of fluorophore-labeled PCR amplicons covering the nuclease target site by capillary electrophoresis in a sequenator. Second, FACS enrichment of cells expressing nucleases linked to fluorescent proteins can be used to maximize knockout or knock-in editing efficiencies or to balance editing efficiency and toxic/off-target effects. The two methods can be combined to form a pipeline for cell-line editing that facilitates the testing of new nuclease reagents and the generation of edited cell pools or clonal cell lines, reducing the number of clones that need to be generated and increasing the ease with which they are screened. The pipeline shortens the time line, but it most prominently reduces the workload of cell-line editing, which may be completed within 4 weeks.

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

Figure 1: Stages of IDAA.
Figure 2: IDAA reveals the indel signature of a given gRNA.
Figure 3: FACS-based genome editing.
Figure 4: Time line and overview of a pipeline for cell line editing based on IDAA and FACS.
Figure 5: Nuclease-2A-FP expression systems.
Figure 6: Setting of FACS parameters for nuclease-2A-FP genome editing.
Figure 7: FACS balancing of on-target and off-target mutagenesis.
Figure 8: Screening of gRNA designs using gRNA amplicon expression cassettes, FACS and IDAA.
Figure 9: Building up of genome modification levels in cell pools by repeated nuclease delivery and FACS.
Figure 10: Genotype screening of cell clones by IDAA.

Similar content being viewed by others

References

  1. Carroll, D. Genome engineering with targetable nucleases. Annu. Rev. Biochem. 83, 409–439 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Bae, S., Kweon, J., Kim, H.S. & Kim, J.-S. Microhomology-based choice of Cas9 nuclease target sites. Nat. Methods 11, 705–706 (2014).

    CAS  PubMed  Google Scholar 

  3. Urnov, F.D. et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature 435, 646–651 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Moehle, E.A. et al. Targeted gene addition into a specified location in the human genome using designed zinc finger nucleases. Proc. Natl. Acad. Sci. USA 104, 3055–3060 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Chen, F. et al. High-frequency genome editing using ssDNA oligonucleotides with zinc-finger nucleases. Nat. Methods 8, 753–755 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Yang, L. et al. Optimization of scarless human stem cell genome editing. Nucleic Acids Res. 41, 9049–9061 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Richardson, C.D., Ray, G.J., DeWitt, M.A., Curie, G.L. & Corn, J.E. Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, 339–344 (2016).

    CAS  PubMed  Google Scholar 

  8. Renaud, J.-B. et al. Improved genome editing efficiency and flexibility using modified oligonucleotides with TALEN and CRISPR-Cas9 nucleases. Cell Rep. 14, 2263–2272 (2016).

    CAS  PubMed  Google Scholar 

  9. Kim, H. & Kim, J.-S. A guide to genome engineering with programmable nucleases. Nat. Rev. Genet. 15, 321–334 (2014).

    CAS  PubMed  Google Scholar 

  10. Mussolino, C., Mlambo, T. & Cathomen, T. Proven and novel strategies for efficient editing of the human genome. Curr. Opin. Pharmacol. 24, 105–112 (2015).

    CAS  PubMed  Google Scholar 

  11. Maeder, M.L., Thibodeau-Beganny, S., Sander, J.D., Voytas, D.F. & Joung, J.K. Oligomerized pool engineering (OPEN): an 'open-source' protocol for making customized zinc-finger arrays. Nat. Protoc. 4, 1471–1501 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Kim, H.J., Lee, H.J., Kim, H., Cho, S.W. & Kim, J.-S. Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res. 19, 1279–1288 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Sander, J.D., Maeder, M.L. & Joung, J.K. Engineering designer nucleases with customized cleavage specificities. Curr. Protoc. Mol. Biol. 96, 12.13 (2011).

    Google Scholar 

  14. Urnov, F.D., Rebar, E.J., Holmes, M.C., Zhang, H.S. & Gregory, P.D. Genome editing with engineered zinc finger nucleases. Nat. Rev. Genet. 11, 636–646 (2010).

    CAS  PubMed  Google Scholar 

  15. Sanjana, N.E. et al. A transcription activator-like effector toolbox for genome engineering. Nat. Protoc. 7, 171–192 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Reyon, D. et al. FLASH assembly of TALENs for high-throughput genome editing. Nat. Biotechnol. 30, 460–465 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sander, J.D. & Joung, J.K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347–355 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ran, F.A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. 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 

  20. Wright, A.V., Nuñez, J.K. & Doudna, J.A. Biology and applications of CRISPR systems: harnessing nature's toolbox for genome engineering. Cell 164, 29–44 (2016).

    CAS  PubMed  Google Scholar 

  21. Tsai, S.Q. & Joung, J.K. Defining and improving the genome-wide specificities of CRISPR-Cas9 nucleases. Nat. Rev. Genet. 17, 300–312 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, Z. et al. Fast and sensitive detection of indels induced by precise gene targeting. Nucleic Acids Res. 43, e59 (2015).

    PubMed  PubMed Central  Google Scholar 

  23. Duda, K. et al. High-efficiency genome editing via 2A-coupled co-expression of fluorescent proteins and zinc finger nucleases or CRISPR/Cas9 nickase pairs. Nucleic Acids Res. 42, e84 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, Z. et al. Engineered CHO cells for production of diverse, homogeneous glycoproteins. Nat. Biotechnol. 33, 842–844 (2015).

    CAS  PubMed  Google Scholar 

  25. Carrington, B., Varshney, G.K., Burgess, S.M. & Sood, R. CRISPR-STAT: an easy and reliable PCR-based method to evaluate target-specific sgRNA activity. Nucleic Acids Res. 43, e157 (2015).

    PubMed  PubMed Central  Google Scholar 

  26. Certo, M.T. et al. Tracking genome engineering outcome at individual DNA breakpoints. Nat. Methods 8, 671–676 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Mandal, P.K. et al. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell 15, 643–652 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Ding, Q. et al. A TALEN genome-editing system for generating human stem cell-based disease models. Cell Stem Cell 12, 238–251 (2013).

    CAS  PubMed  Google Scholar 

  29. Kim, H. et al. Surrogate reporters for enrichment of cells with nuclease-induced mutations. Nat. Methods 8, 941–943 (2011).

    CAS  PubMed  Google Scholar 

  30. Kim, H. et al. Magnetic separation and antibiotics selection enable enrichment of cells with ZFN/TALEN-induced mutations. PLoS One 8, e56476 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ramakrishna, S. et al. Surrogate reporter-based enrichment of cells containing RNA-guided Cas9 nuclease-induced mutations. Nat. Commun. 5, 3378 (2014).

    PubMed  Google Scholar 

  32. Tsai, S.Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    CAS  PubMed  Google Scholar 

  33. Miller, J.C. et al. An improved zinc-finger nuclease architecture for highly specific genome editing. Nat. Biotechnol. 25, 778–785 (2007).

    CAS  PubMed  Google Scholar 

  34. Yeung, A.T., Hattangadi, D., Blakesley, L. & Nicolas, E. Enzymatic mutation detection technologies. BioTechniques 38, 749–758 (2005).

    CAS  PubMed  Google Scholar 

  35. Vouillot, L., Thélie, A. & Pollet, N. Comparison of T7E1 and surveyor mismatch cleavage assays to detect mutations triggered by engineered nucleases. G3 (Bethesda) 5, 407–415 (2015).

    CAS  PubMed Central  Google Scholar 

  36. Oleykowski, C.A., Bronson Mullins, C.R., Godwin, A.K. & Yeung, A.T. Mutation detection using a novel plant endonuclease. Nucleic Acids Res. 26, 4597–4602 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Till, B.J., Burtner, C., Comai, L. & Henikoff, S. Mismatch cleavage by single-strand specific nucleases. Nucleic Acids Res. 32, 2632–2641 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Cross, M.J., Waters, D.L.E., Lee, L.S. & Henry, R.J. Endonucleolytic mutation analysis by internal labeling (EMAIL). Electrophoresis 29, 1291–1301 (2008).

    CAS  PubMed  Google Scholar 

  39. Tsuji, T. & Niida, Y. Development of a simple and highly sensitive mutation screening system by enzyme mismatch cleavage with optimized conditions for standard laboratories. Electrophoresis 29, 1473–1483 (2008).

    CAS  PubMed  Google Scholar 

  40. Qiu, P. et al. Mutation detection using Surveyor™ nuclease. BioTechniques 36, 702–707 (2004).

    CAS  PubMed  Google Scholar 

  41. Mashal, R.D., Koontz, J. & Sklar, J. Detection of mutations by cleavage of DNA heteroduplexes with bacteriophage resolvases. Nat. Genet. 9, 177–183 (1995).

    CAS  PubMed  Google Scholar 

  42. Güell, M., Yang, L. & Church, G.M. Genome editing assessment using CRISPR Genome Analyzer (CRISPR-GA). Bioinformatics 30, 2968–2970 (2014).

    PubMed  PubMed Central  Google Scholar 

  43. Brinkman, E.K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Mock, U., Hauber, I. & Fehse, B. Digital PCR to assess gene-editing frequencies (GEF-dPCR) mediated by designer nucleases. Nat. Protoc. 11, 598–615 (2016).

    CAS  PubMed  Google Scholar 

  45. Orlando, S.J. et al. Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res. 38, e152 (2010).

    PubMed  PubMed Central  Google Scholar 

  46. Maresca, M., Lin, V.G., Guo, N. & Yang, Y. Obligate Ligation-Gated Recombination (ObLiGaRe): custom-designed nuclease-mediated targeted integration through nonhomologous end joining. Genome Res. 23, 539–546 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Carlson, D.F. et al. Efficient TALEN-mediated gene knockout in livestock. Proc. Natl. Acad. Sci. USA 109, 17382–17387 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Frank, S., Skryabin, B.V. & Greber, B. A modified TALEN-based system for robust generation of knock-out human pluripotent stem cell lines and disease models. BMC Genomics 14, 1–9 (2013).

    Google Scholar 

  49. Ran, F.A. et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell 154, 1380–1389 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Mali, P. et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31, 833–838 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Shen, B. et al. Efficient genome modification by CRISPR-Cas9 nickase with minimal off-target effects. Nat. Methods 11, 399–402 (2014).

    CAS  PubMed  Google Scholar 

  52. Cho, S.W. et al. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res. 24, 132–141 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kim, S., Kim, D., Cho, S.W., Kim, J. & Kim, J.-S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012–1019 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Zuris, J.A. et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat. Biotechnol. 33, 73–80 (2015).

    CAS  PubMed  Google Scholar 

  55. Liang, X. et al. Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. J. Biotechnol. 208, 44–53 (2015).

    CAS  PubMed  Google Scholar 

  56. Yu, X. et al. Improved delivery of Cas9 protein/gRNA complexes using lipofectamine CRISPRMAX. Biotechnol. Lett. 38, 919–929 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Hendel, A. et al. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nat. Biotechnol. 33, 985–989 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Dieffenbach, C.W., Lowe, T.M. & Dveksler, G.S. General concepts for PCR primer design. Genome Res. 3, S30–S37 (1993).

    CAS  Google Scholar 

  59. Kibbe, W.A. OligoCalc: an online oligonucleotide properties calculator. Nucleic Acids Res. 35, W43–W46 (2007).

    PubMed  PubMed Central  Google Scholar 

  60. Korbie, D.J. & Mattick, J.S. Touchdown PCR for increased specificity and sensitivity in PCR amplification. Nat. Protoc. 3, 1452–1456 (2008).

    CAS  PubMed  Google Scholar 

  61. Hsu, P.D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Frock, R.L. et al. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat. Biotechnol. 33, 179–186 (2015).

    CAS  PubMed  Google Scholar 

  63. Wang, X. et al. Unbiased detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective lentiviral vectors. Nat. Biotechnol. 33, 175–178 (2015).

    CAS  PubMed  Google Scholar 

  64. Fu, Y., Sander, J.D., Reyon, D., Cascio, V.M. & Joung, J.K. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat. Biotechnol. 32, 279–284 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Kim, D. et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat. Methods 12, 237–243 (2015).

    CAS  PubMed  Google Scholar 

  66. Tsai, S.Q. et al. Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing. Nat. Biotechnol. 32, 569–576 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Guilinger, J.P., Thompson, D.B. & Liu, D.R. Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nat. Biotechnol. 32, 577–582 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Kleinstiver, B.P. et al. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490–495 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Slaymaker, I.M. et al. Rationally engineered Cas9 nucleases with improved specificity. Science 351, 84–88 (2015).

    PubMed  PubMed Central  Google Scholar 

  70. Muller, M. et al. Streptococcus thermophilus CRISPR-Cas9 systems enable specific editing of the human genome. Mol. Ther. 24, 636–644 (2016).

    PubMed  PubMed Central  Google Scholar 

  71. Bolukbasi, M.F., Gupta, A. & Wolfe, S.A. Creating and evaluating accurate CRISPR-Cas9 scalpels for genomic surgery. Nat. Methods 13, 41–50 (2016).

    CAS  PubMed  Google Scholar 

  72. Koo, T., Lee, J. & Kim, J.-S. Measuring and reducing off-target activities of programmable nucleases including CRISPR-Cas9. Mol. Cells 38, 475–481 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Doyon, Y. et al. Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures. Nat. Methods 8, 74–79 (2011).

    CAS  PubMed  Google Scholar 

  74. Nakade, S. et al. Microhomology-mediated end-joining-dependent integration of donor DNA in cells and animals using TALENs and CRISPR/Cas9. Nat. Commun. 5, 5560 (2014).

    CAS  PubMed  Google Scholar 

  75. Sakuma, T., Nakade, S., Sakane, Y., Suzuki, K.-I.T. & Yamamoto, T. MMEJ-assisted gene knock-in using TALENs and CRISPR-Cas9 with the PITCh systems. Nat. Protoc. 11, 118–133 (2016).

    CAS  PubMed  Google Scholar 

  76. Yoshimi, K. et al. ssODN-mediated knock-in with CRISPR-Cas for large genomic regions in zygotes. Nat. Commun. 7, 10431 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Chu, V.T. et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 33, 543–548 (2015).

    CAS  PubMed  Google Scholar 

  78. Park, J., Bae, S. & Kim, J.-S. Cas-Designer: a web-based tool for choice of CRISPR-Cas9 target sites. Bioinformatics 31, 4014–4016 (2015).

    CAS  PubMed  Google Scholar 

  79. Sander, J.D. et al. ZiFiT (Zinc Finger Targeter): an updated zinc finger engineering tool. Nucleic Acids Res. 38, W462–W468 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Heigwer, F., Kerr, G. & Boutros, M. E-CRISP: fast CRISPR target site identification. Nat Methods 11, 122–123 (2014).

    CAS  PubMed  Google Scholar 

  81. Hodgkins, A. et al. WGE: a CRISPR database for genome engineering. Bioinformatics 31, 3078–3080 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Moreno-Mateos, M.A. et al. CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat. Methods 12, 982–988 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Singh, R., Kuscu, C., Quinlan, A., Qi, Y. & Adli, M. Cas9-chromatin binding information enables more accurate CRISPR off-target prediction. Nucleic Acids Res. 43, e118 (2015).

    PubMed  PubMed Central  Google Scholar 

  84. Lee, C.M., Cradick, T.J., Fine, E.J. & Bao, G. Nuclease target site selection for maximizing on-target activity and minimizing off-target effects in genome editing. Mol. Ther. 24, 475–487 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Magnuson, V.L. et al. Substrate nucleotide-determined non-templated addition of adenine by Taq DNA polymerase: implications for PCR-based genotyping and cloning. BioTechniques 21, 700–709 (1996).

    CAS  PubMed  Google Scholar 

  86. Brownstein, M.J., Carpten, J.D. & Smith, J.R. Modulation of non-templated nucleotide addition by Taq DNA polymerase: primer modifications that facilitate genotyping. BioTechniques 20, 1004–1006 (1996).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Fossum (BRIC), C.K. Beier Holden, C. Andersen, and S. Narimatsu (Copenhagen Center for Glycomics), and A. Dürr (the Danish Stem Cell Center) for excellent technical assistance. We thank S. Miller, Washington University, for critical reading of the manuscript. This work was supported by the Danish Cancer Society (R124-A7632-15-S2 to M.F.), the Danish Council for Independent Research (0602-02368B to M.F.), the University of Copenhagen Excellence Programme for Interdisciplinary Research—CDO2016 (to M.F., E.P.B. and H.H.W.), the Novo Nordisk Foundation (to M.F., Y.N. and Z.Y., and NNF15CC0018344 to E.A.O.), the Lundbeck Foundation (R165-2013-15743 to F.N.), the National Institutes of Health (R21CA184656 to S.H.H.), the Medical Research Council UK (U117581329 to E.A.O.) and the Danish National Research Foundation (DNRF107 to E.P.B., H.H.W., Y.N., Z.Y. and H.C.).

Author information

Authors and Affiliations

Authors

Contributions

All authors were involved in designing and/or performing experiments underlying the manuscript or the development of the methods described. L.A.L., E.P.B. and M.F. wrote the manuscript with comments from the other authors.

Corresponding authors

Correspondence to Eric P Bennett or Morten Frödin.

Ethics declarations

Competing interests

E.P.B. declares competing financial interests (see the HTML version of this article for details). E.P.B. possesses the rights to a patent application describing the IDAA methodology.

Integrated supplementary information

Supplementary Figure 1 Sensitivity and quantitative performance of IDAA performed in an ABI 3500 instrument is comparable to Next-Generation Sequencing (MiSeq).

Indel analysis by (a) IDAA or (b) MiSeq of PCR amplicons from the Cosmc locus edited by CRISPR/Cas9 in a CHO cell pool. On the x-axis, indel sizes (bp) are indicated for some of the amplicons. In the zoom-in panels, a stippled line indicates the background signal level, defined based on the signals from 45-55 bp insertions that were considered background, since CRISPR/Cas9 rarely elicits insertions of such sizes. (c) Graphs of frequencies of indels a-m from the IDAA and MiSeq analyses. Note the high degree of agreement between the two analyses.

Supplementary Figure 2 IDAA can generate useful indel profiles from minute amounts of IDAA PCR amplicons.

The Trp53 locus was targeted with CRISPR/Cas9 in a mouse Neuro2A cell pool and the target site was amplified by IDAA PCR. The indicated amounts of the IDAA PCR were analyzed by (a) agarose gel electrophoresis (an arrow indicates the IDAA PCR amplicon) or (b) IDAA capillary electrophoresis. The size and frequency of selected indels are indicated. IDAA was performed in an ABI 3500 instrument.

Note that the indel profiles are almost identical across the range of IDAA amplicons analyzed and that a high-quality profile can be generated from amplicon amounts barely visible or undetectable by agarose gel analysis.

Supplementary Figure 3 Upper and lower detection levels for IDAA amplicons in ABI 3130 and 3500 instruments.

(a) Various amounts of a wt IDAA amplicon from a non-edited control sample were analyzed in ABI 3130 and 3500 instruments. When the loaded amplicons exceed the dynamic range, a smaller peak artifact often appears 20-30 bp ahead of the true signal (indicated by asterisk in the upper 3130 panel). (b) Zoom in on the peaks boxed in (a). The lower 3130 panel illustrates that also below the dynamic range, a specific signal can be discriminated from background signals.

Supplementary Figure 4 IDAA reveals the indel signature of a given gRNA, TALEN or ZFN.

IDAA profiles for two independent experiments using the same construct of FP-linked CRISPR/Cas9 (a,b), TALENs (c) or ZFNs (d), targeting various loci in K562 or HEK293 cells, as indicated. Prior to analysis, the nuclease-transfected cells were subjected to bulk FACS for the top 30% most fluorescent cells. The size and frequency of selected indels are indicated. IDAA profiles were generated in an ABI 3130 instrument.

Supplementary Figure 5 Using IDAA to estimate probability of germline transmission of indels from F0 mosaic fish and to identify F1 mutant fish in Danio rerio genome editing.

(a) In zebrafish genome editing, a major challenge is determination of the nature and degree of indel mosaicism and hence, probability of germline transmission of indels from F0 fish derived from one-cell embryos injected with Cas9 and gRNA. IDAA enables easy and rapid evaluation of these variables through analysis of F0 fin DNA, allowing optimal set-up of F0 breeding pairs. IDAA also enables easy identification of indels in subsequent generations through fin DNA analysis. (b) Somatic IDAA profiles of the bambi locus in fin DNA of F0 fish targeted at the one-cell embryo stage via injection of Cas9 and gRNA. F0 male #1 and F0 female #4 were chosen for mating due to the presence of predominant indels indicated by open or closed circles, suggesting high probability of germline transmission of these mutations. (c) IDAA profiles of 200 downstream F1 fish, showing that 4 out of 5 fish harbored the predominant indels identified in the F0 breeding pair, of which 3 were in the biallelic state (F1 #1, #3, #4) and one homozygous for an indel (F1 #4). In this example, IDAA was performed on embryos, but IDAA could also have been performed on fin DNA. The size and frequency of selected indels are indicated. IDAA profiles were generated in an ABI 3130 instrument.

Supplementary Figure 6 QCgRNA amplicon expression cassettes.

(a) Schematic showing the various elements of the QCgRNA amplicon primers QCgFwd, QCgX and QCgRev. Sequences annealing to U6 promotor template are shown in orange; gRNA design in red; tracr elements in blue; restriction enzyme sites for sub-cloning to pEPB104 in italics. Note that the gRNA design is incorporated into QCgX as the complementary sequence to the target, which is shown in green. The nucleotide (c) is only included in the QCgX primer, if the gRNA (=target) does not contain a G as the first (5´) nucleotide, which is the case in the present example. (b) Agarose gel (2%) electrophoresis check for the formation of full-length (f) products (arrow) by QCgRNA amplicon tri-primer PCRs, as compared to control (c) PCRs containing only QCgFwd and QCgX primers (Step 3 in Procedure). Various amounts of MassRuler Low Range DNA ladder (M) are run alongside to enable quantitation of the QCgRNA amplicons. (c) The indel profiles elicited by a gRNA design in a QCgRNA amplicon and a plasmid vector are identical, as illustrated by targeting GALNT10 in HEK293 cells. The GALNT10 QCgRNA amplicon was subcloned into pEPB104 plasmid (Addgene #68369; Supplementary Sequence 1) using EcoRI and KpnI restriction endonuclease sites present in both constructs. (d) A QCgRNA design found non-functional in one cell type typically remains non-functional when tested in other cell types, as illustrated with a POMT2 QCgRNA. IDAA profiles were generated in an (c) ABI 3500 or and (d) ABI 3130 instrument.

Supplementary information

Supplementary Figures and Text

Supplementary Figures 1–6, Supplementary Tables 1–3, the Supplementary Note, the Supplementary Data and the Supplementary Manual (PDF 5743 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lonowski, L., Narimatsu, Y., Riaz, A. et al. Genome editing using FACS enrichment of nuclease-expressing cells and indel detection by amplicon analysis. Nat Protoc 12, 581–603 (2017). https://doi.org/10.1038/nprot.2016.165

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2016.165

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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