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.

  • Letter
  • Published:

Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells

Abstract

The activities and genome-wide specificities of CRISPR-Cas Cpf1 nucleases1 are not well defined. We show that two Cpf1 nucleases from Acidaminococcus sp. BV3L6 and Lachnospiraceae bacterium ND2006 (AsCpf1 and LbCpf1, respectively) have on-target efficiencies in human cells comparable with those of the widely used Streptococcus pyogenes Cas9 (SpCas9)2,3,4,5. We also report that four to six bases at the 3′ end of the short CRISPR RNA (crRNA) used to program Cpf1 nucleases are insensitive to single base mismatches, but that many of the other bases in this region of the crRNA are highly sensitive to single or double substitutions. Using GUIDE-seq and targeted deep sequencing analyses performed with both Cpf1 nucleases, we were unable to detect off-target cleavage for more than half of 20 different crRNAs. Our results suggest that AsCpf1 and LbCpf1 are highly specific in human cells.

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: On-target indel mutation percentages induced by Cpf1 nucleases in human cells.
Figure 2: Tolerance of AsCpf1 and LbCpf1 to mismatched or truncated crRNAs.
Figure 3: Genome-wide specificities of AsCpf1 and LbCpf1.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

References

  1. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163, 759–771 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    Article  PubMed  Google Scholar 

  5. Maeder, M.L. & Gersbach, C.A. Genome-editing technologies for gene and cell therapy. Mol. Ther. 24, 430–446 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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).

    Article  CAS  PubMed  Google Scholar 

  7. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Deltcheva, E. et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 471, 602–607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jinek, M. et al. RNA-programmed genome editing in human cells. eLife 2, e00471 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  12. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Slaymaker, I.M. et al. Rationally engineered Cas9 nucleases with improved specificity. Science 351, 84–88 (2016).

    Article  CAS  PubMed  Google Scholar 

  18. Schunder, E., Rydzewski, K., Grunow, R. & Heuner, K. First indication for a functional CRISPR/Cas system in Francisella tularensis. Int. J. Med. Microbiol. 303, 51–60 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Makarova, K.S. et al. An updated evolutionary classification of CRISPR-Cas systems. Nat. Rev. Microbiol. 13, 722–736 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fagerlund, R.D., Staals, R.H. & Fineran, P.C. The Cpf1 CRISPR-Cas protein expands genome-editing tools. Genome Biol. 16, 251 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Bae, S., Park, J. & Kim, J.S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 30, 1473–1475 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kleinstiver, B.P. et al. Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol. 33, 1293–1298 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kleinstiver, B.P. et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523, 481–485 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Yin, H. et al. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat. Biotechnol. 34, 328–333 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bolukbasi, M.F. et al. DNA-binding-domain fusions enhance the targeting range and precision of Cas9. Nat. Methods 12, 1150–1156 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Friedland, A.E. et al. Characterization of Staphylococcus aureus Cas9: a smaller Cas9 for all-in-one adeno-associated virus delivery and paired nickase applications. Genome Biol. 16, 257 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tsai, S.Q., Topkar, V.V., Joung, J.K. & Aryee, M.J. Open-source guideseq software for analysis of GUIDE-seq data. Nat. Biotechnol. 34, 483 (2016).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by a National Institutes of Health (NIH) Director's Pioneer Award (DP1 GM105378) and NIH R01 GM107427 to J.K.J., the Jim and Ann Orr Research Scholar Award (to J.K.J.), a Natural Sciences and Engineering Research Council of Canada Postdoctoral Fellowship (to B.P.K.), an award from the Massachusetts General Hospital Collaborative Center for X-Linked Dystonia-Parkinsonism, and an MGH Tosteson Award (to S.Q.T.). New reagents described in this work have been deposited with the non-profit plasmid distribution service Addgene (http://www.addgene.org/crispr-cas).

Author information

Authors and Affiliations

Authors

Contributions

B.P.K., S.Q.T., and J.K.J. conceived of and designed experiments. B.P.K., S.Q.T., M.S.P., N.T.N., and M.M.W. performed all human cell experiments and analyzed data. S.Q.T., J.M.L., Z.R.M., and M.J.A. analyzed the GUIDE-seq and targeted deep-sequencing data. B.P.K., S.Q.T., and J.K.J. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Benjamin P Kleinstiver or J Keith Joung.

Ethics declarations

Competing interests

J.K.J. is a consultant for Horizon Discovery. J.K.J. has financial interests in Beacon Genomics, Editas Medicine, Hera Testing Laboratories, Poseida Therapeutics, and Transposagen Biopharmaceuticals. JKJ's interests were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict of interest policies. S.Q.T., M.J.A., and J.K.J. are co-founders of Beacon Genomics, a company that is commercializing methods for determining nuclease specificity.

Integrated supplementary information

Supplementary Figure 1 Cpf1 target sites in endogenous genes known to be efficiently modified by SpCas9.

Sites targeted by Cpf1 and SpCas9 in four genes are highlighted in blue and grey, respectively. Target site sequences and primers used to amplify these loci are provided in Supplementary Tables 1 and 5, respectively.

Supplementary Figure 2 On-target time-course activity assays of AsCpf1, LbCpf1, and SpCas9.

Time-course assays were performed using guide RNAs targeted to matched sites in human cells. Percent modification activity determined by T7E1 assay; error bars, s.e.m.; n = 3.

Supplementary Figure 3 Cpf1 activity and specificity with alternate length crRNAs.

(a) Indel mutation percentages induced by AsCpf1 and LbCpf1 at three different endogenous sites using crRNAs with variable length 3’ end truncations, determined by T7E1 assay. Error bars, s.e.m.; n = 3; nt, nucleotide. (b) Endogenous gene modification by AsCpf1 and LbCpf1 using crRNAs with 20 nt spacers that contain singly mismatched bases. Activity determined by T7E1 assay; error bars, s.e.m.; n = 3.

Supplementary Figure 4 Tag integration and overall mutagenesis percentages observed in GUIDE-seq experiments in U2OS cells.

(a,b) On-target GUIDE-seq tag integration (a) and overall mutagenesis (b) determined by RFLP and T7E1 assays, respectively, for the 11 matched target sites examined by GUIDE-seq in Figure 3b. Error bars, s.e.m.; n = 3. (c) Ratios of tag integration to overall mutagenesis (from data in a and b) for the 11 matched target sites. (d) Summary of RFLP:T7E1 ratios from c, with means and 95% confidence intervals shown. (e,f) On-target GUIDE-seq tag integration (e) and overall mutagenesis percentages (f) determined by RFLP and T7E1 assays, respectively, for the 9 different endogenous human gene target sites examined by GUIDE-seq in Figure 3b. Error bars, s.e.m.; n = 3. (g) Summary of RFLP:T7E1 ratios calculated from the data in e and f, with means and 95% confidence intervals shown.

Supplementary Figure 5 Genome-wide specificities of AsCpf1 and LbCpf1 using the crRNA targeted to matched site 6.

Off-target sites were determined using GUIDE-seq. Mismatched positions in the target sites of off-targets are highlighted in color, and GUIDE-seq read counts shown to the right of the on- and off-target sequences represent a measure of cleavage efficiency at a given site.

Supplementary Figure 6 GUIDE-seq with AsCpf1 and LbCpf1 in HEK293 cells.

(a,b) On-target GUIDE-seq tag integration (a) and overall mutagenesis (b) determined by RFLP and T7E1 assays, respectively, for 6 crRNAs. Error bars, s.e.m.; n = 3. (c) Ratios of tag integration to overall mutagenesis (from data in a and b), with means and 95% confidence intervals shown. (d) Off-target sites for AsCpf1 and LbCpf1 with three different crRNAs in HEK293 and U2OS cells determined using GUIDE-seq (U2OS cell data is re-represented from Figure 3b for comparative purposes). Mismatched positions in the target sites of off-targets are highlighted in color, and GUIDE-seq read counts shown to the right of the on- and off-target sequences represent a measure of cleavage efficiency at a given site.

Supplementary Figure 7 Deep sequencing of on-target and closely mismatched potential Cpf1 off-target sites.

Triplicate data points representing percent modification are plotted for control, AsCpf1, and LbCpf1 experiments; if less than 3 data points are shown for any given condition, the value is either zero or not determined (see Supplementary Table 4). Mismatched nucleotides in off-target sites are colored; off-target sites are numbered as shown in Supplementary Tables 4 or 5; on, on-target site; Gs, off-target site detected in GUIDE-seq experiments shown in Fig. 3b and Supplementary Fig. 5; Ss, statistically significant indel formation above background control levels determined using the Bonferroni correction (Supplementary Table 4).

Supplementary Figure 8 Venn diagrams of Cpf1 and SpCas9 targeting ranges based on availability of sites in the reference human genome.

(a, b) Targetable genetic elements are defined as those that contain the predicted cleavage site of at least one Cpf1 or SpCas9 target site, without or with a polIII promoter required 5’-guanine for SpCas9 (panels a and b, respectively; within N21GG versus TTTN24 motifs or GN20GG versus TTTN24 motifs, respectively). Numbers in the bottom right of each plot represent genetic elements not targetable by either SpCas9 or Cpf1. TSS, transcription start site.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Note 1 (PDF 1741 kb)

Supplementary Table 1

gRNA target sites. (XLSX 24 kb)

Supplementary Table 2

In Silico off-targets. (XLSX 63 kb)

Supplementary Table 3

Deep sequencing data and p-values. (XLSX 268 kb)

Supplementary Table 4

GUIDE-seq data. (XLSX 79 kb)

Supplementary Table 5

Oligonucleotides. (XLSX 19 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kleinstiver, B., Tsai, S., Prew, M. et al. Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells. Nat Biotechnol 34, 869–874 (2016). https://doi.org/10.1038/nbt.3620

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3620

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