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:

Photoactivatable CRISPR-Cas9 for optogenetic genome editing

Abstract

We describe an engineered photoactivatable Cas9 (paCas9) that enables optogenetic control of CRISPR-Cas9 genome editing in human cells. paCas9 consists of split Cas9 fragments and photoinducible dimerization domains named Magnets. In response to blue light irradiation, paCas9 expressed in human embryonic kidney 293T cells induces targeted genome sequence modifications through both nonhomologous end joining and homology-directed repair pathways. Genome editing activity can be switched off simply by extinguishing the light. We also demonstrate activation of paCas9 in spatial patterns determined by the sites of irradiation. Optogenetic control of targeted genome editing should facilitate improved understanding of complex gene networks and could prove useful in biomedical applications.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Design and characterization of photoactivatable Cas9.
Figure 2: Optogenetic genome editing of mammalian endogenous genes by the photoactivatable Cas9.
Figure 3: Spatiotemporal control of Cas9 nuclease activity with optimized paCas9-2.
Figure 4: Optogenetic control of RNA-guided transcription interference with padCas9.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  4. Dow, L.E. et al. Inducible in vivo genome editing with CRISPR-Cas9. Nat. Biotechnol. 33, 390–394 (2015).

    Article  CAS  Google Scholar 

  5. González, F. et al. An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. Cell Stem Cell 15, 215–226 (2014).

    Article  Google Scholar 

  6. Zetsche, B., Volz, S.E. & Zhang, F. A split-Cas9 architecture for inducible genome editing and transcription modulation. Nat. Biotechnol. 33, 139–142 (2015).

    Article  CAS  Google Scholar 

  7. 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).

    Article  CAS  Google Scholar 

  8. 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).

    Article  CAS  Google Scholar 

  9. Ramakrishna, S. et al. Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA. Genome Res. 24, 1020–1027 (2014).

    Article  CAS  Google Scholar 

  10. Laplante, M. & Sabatini, D.M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    Article  CAS  Google Scholar 

  11. Brieke, C., Rohrbach, F., Gottschalk, A., Mayer, G. & Heckel, A. Light-controlled tools. Angew. Chem. Int. Edn Engl. 51, 8446–8476 (2012).

    Article  CAS  Google Scholar 

  12. Lee, H., Larson, D. & Lawrence, D. Illuminating the chemistry of life: design, synthesis, and applications of “caged” and related photoresponsive compounds. ACS Chem. Biol. 4, 409–427 (2009).

    Article  CAS  Google Scholar 

  13. DeRose, R., Miyamoto, T. & Inoue, T. Manipulating signaling at will: chemically-inducible dimerization (CID) techniques resolve problems in cell biology. Pflugers Arch. 465, 409–417 (2013).

    Article  CAS  Google Scholar 

  14. Nishimasu, H. et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell 156, 935–949 (2014).

    Article  CAS  Google Scholar 

  15. Anders, C., Niewoehner, O., Duerst, A. & Jinek, M. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature 513, 569–573 (2014).

    Article  CAS  Google Scholar 

  16. Kennedy, M.J. et al. Rapid blue-light–mediated induction of protein interactions in living cells. Nat. Methods 7, 973–975 (2010).

    Article  CAS  Google Scholar 

  17. Bugaj, L.J., Choksi, A.T., Mesuda, C.K., Kane, R.S. & Schaffer, D.V. Optogenetic protein clustering and signaling activation in mammalian cells. Nat. Methods 10, 249–252 (2013).

    Article  CAS  Google Scholar 

  18. Kawano, F., Suzuki, H., Furuya, A. & Sato, M. Engineered pairs of distinct photoswitches for optogenetic control of cellular proteins. Nat. Commun. 6, 6256 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Fu, Y. et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 31, 822–826 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  25. Qi, L.S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  Google Scholar 

  26. Voon, D.C. et al. Use of mRNA- and protein-destabilizing elements to develop a highly responsive reporter system. Nucleic Acids Res. 33, e27 (2005).

    Article  Google Scholar 

  27. Lin, Y. et al. Rapidly reversible manipulation of molecular activity with dual chemical dimerizers. Angew. Chem. Int. Edn Engl. 52, 6450–6454 (2013).

    Article  CAS  Google Scholar 

  28. Shen, Z. et al. Conditional knockouts generated by engineered CRISPR-Cas9 endonuclease reveal the roles of coronin in C. elegans neural development. Dev. Cell 30, 625–636 (2014).

    Article  CAS  Google Scholar 

  29. Nihongaki, Y., Yamamoto, S., Kawano, F., Suzuki, H. & Sato, M. CRISPR-Cas9-based photoactivatable transcription system. Chem. Biol. 22, 169–174 (2015).

    Article  CAS  Google Scholar 

  30. Polstein, L.R. & Gersbach, C.A. A light-inducible CRISPR-Cas9 system for control of endogenous gene activation. Nat. Chem. Biol. 11, 198–200 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank J. Galipon for technical assistance. This work was supported by Platform for Dynamic Approaches to Living System from the Ministry of Education, Culture, Sports, Science and Technology, Japan (MEXT) and by grants from Japan Society for the Promotion of Science (JSPS).

Author information

Authors and Affiliations

Authors

Contributions

Y.N. and M.S. conceived the project. Y.N., F.K., T.N. and M.S. designed the experiments. Y.N. performed experiments and analyzed data. Y.N. and M.S. wrote the manuscript.

Corresponding author

Correspondence to Moritoshi Sato.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Construction of rapamycin-inducible split-Cas9.

(a) Candidate 18 split sites of Streptococcus pyogenes Cas9 protein. (b) Constructs of rapamycin-inducible Cas9. N- and C- fragments of Cas9 were fused with FRB and FKBP, respectively.

Supplementary Figure 2 Screening of split-Cas9 fragments.

(a) Ligand-induced Cas9 activity measured by luciferase reporter plasmid HDR assay. HEK293T cells were transfected with N-fragment of Cas9 fused with FRB, C-fragment of Cas9 fused with FKBP, stop codon-inserted luciferase reporter and promoter-less luciferase donor vector. (b) Second screening of Cas9 split sites in the vicinity of residues 713 and 714.

Supplementary Figure 3 Schematic of paCas9.

The schematic is based on the crystal structures of Cas9 (PDB ID: 4UN3) (ref. 17) and the light-state dimer of Vivid (PDB ID: 3RH8) (supplementary ref. 1), a precursor protein of Magnets. N713 and C714 fragments are shown in red and blue, respectively. Positive Magnet and negative Magnet are shown in gold and cyan, respectively. sgRNA and targeted DNA are shown in green and yellow, respectively. The positions connected between Cas9 fragments and Magnets are circled in black.

Supplementary Figure 4 Time-course analysis of paCas9-mediated indel mutation in EMX1 locus.

HEK293T cells were transfected with paCas9-1 targeting EMX1. After 20 h incubation, the cells were illuminated by 1.2 W/m2 blue light and genomic DNA was extracted at indicated time points. Indel mutation frequencies were evaluated by mismatch-sensitive T7E1 assay. Data are shown as the mean ± s.e.m. (n=4 from two individual experiments with biological duplicates). Indel frequencies at 0, 1, 3 and 6 h were below the detection limit (1%).

Supplementary Figure 5 paCas9-mediated indel mutation of human EMX1 locus in HeLa cells.

Indel mutation frequencies were evaluated by mismatch-sensitive T7E1 assay.

Supplementary Figure 6 paCas9 nickase can facilitate efficient genome editing using a pair of sgRNAs.

(a) Schematic of light-induced DNA double-strand breaks using a pair of sgRNAs with paCas9 D10A nickase. By introducing D10A mutation in N-terminal fragment of Cas9, paCas9 nuclease can be converted into paCas9 nickase. Using a pair of sgRNAs targeting opposite strands of targeted gene, paCas9 nickase can offer optical control of DNA double-stranded break in targeted locus. (b) Schematic of the human EMX1 locus presenting the target region of a paired sgRNAs indicated by blue lines. PAMs are also shown in red. Red allows indicate putative cleavage site. (c) Representative T7E1 assay result for calculating indel mutation rates induced by paCas9 nickase (mean, n=2 from independent experiments).

Supplementary Figure 7 Spatial surrogate reporter activation by paCas9.

(a) Schematic of surrogate EGFP reporter system (ref. 24, 25). The surrogate reporter consists of mCherry, target sequence of paCas9 (here EMX1 target site) and EGFP. In the dark, while mCherry is constitutively expressed from CMV promoter, EGFP fluorescence is not observed because EGFP gene is out of frame in the absence of Cas9 activity. Upon blue light irradiation, paCas9 is activated and a double-strand break is introduced into the EMX1 target site of the reporter. By NHEJ pathway, this break is repaired with frameshift mutations. This frameshift mutation makes EGFP gene in frame, causing the expression of mCherry-EGFP fusion protein. (b) Slit-patterned activation of paCas9. HEK293T cells were transfected with paCas9-2, surrogate EGFP reporter and sgRNA targeting the surrogate reporter. Twenty hours post transfection, samples were incubated under dark state, global blue light irradiation, and slit-patterned blue light using a photomask for 24 h.

Supplementary Figure 8 Full-length gel images.

Full-length gel images corresponding to Figs 2a, c, d, f, 3a, f and Supplementary Figs 5. and 6c. Unrelated lanes are marked with cross.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 2 and 3, and Supplementary Notes 1 and 2 (PDF 1694 kb)

Supplementary Table 1

Target sequences of sgRNAs and oligonucleotide sequences for constructing sgRNAs. (XLSX 24 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nihongaki, Y., Kawano, F., Nakajima, T. et al. Photoactivatable CRISPR-Cas9 for optogenetic genome editing. Nat Biotechnol 33, 755–760 (2015). https://doi.org/10.1038/nbt.3245

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3245

This article is cited by

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