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:

In vivo genome editing using a high-efficiency TALEN system

Abstract

The zebrafish (Danio rerio) is increasingly being used to study basic vertebrate biology and human disease with a rich array of in vivo genetic and molecular tools. However, the inability to readily modify the genome in a targeted fashion has been a bottleneck in the field. Here we show that improvements in artificial transcription activator-like effector nucleases (TALENs) provide a powerful new approach for targeted zebrafish genome editing and functional genomic applications1,2,3,4,5. Using the GoldyTALEN modified scaffold and zebrafish delivery system, we show that this enhanced TALEN toolkit has a high efficiency in inducing locus-specific DNA breaks in somatic and germline tissues. At some loci, this efficacy approaches 100%, including biallelic conversion in somatic tissues that mimics phenotypes seen using morpholino-based targeted gene knockdowns6. With this updated TALEN system, we successfully used single-stranded DNA oligonucleotides to precisely modify sequences at predefined locations in the zebrafish genome through homology-directed repair, including the introduction of a custom-designed EcoRV site and a modified loxP (mloxP) sequence into somatic tissue in vivo. We further show successful germline transmission of both EcoRV and mloxP engineered chromosomes. This combined approach offers the potential to model genetic variation as well as to generate targeted conditional alleles.

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: Second-generation GoldyTALEN scaffold improves genome-editing efficacy.
Figure 2: Increased TALEN efficiency results in biallelic gene targeting.
Figure 3: Targeted genome editing using GoldyTALENs.
Figure 4: Germline mloxP integration into the crhr2 locus.
Figure 5: In vivo TALEN-induced genome editing outcomes.

Similar content being viewed by others

References

  1. Huang, P. et al. Heritable gene targeting in zebrafish using customized TALENs. Nature Biotechnol. 29, 699–700 (2011)

    Article  Google Scholar 

  2. Sander, J. D. et al. Targeted gene disruption in somatic zebrafish cells using engineered TALENs. Nature Biotechnol. 29, 697–698 (2011)

    Article  CAS  Google Scholar 

  3. Cade, L. et al. Highly efficient generation of heritable zebrafish gene mutations using homo- and heterodimeric TALENs. Nucleic Acids Res. 40, 8001–8010 (2012)

    Article  CAS  Google Scholar 

  4. Moore, F. E. et al. Improved somatic mutagenesis in zebrafish using transcription activator-like effector nucleases (TALENs). PLoS ONE 7, e37877 (2012)

    Article  CAS  ADS  Google Scholar 

  5. Dahlem, T. J. et al. Simple methods for generating and detecting locus-specific mutations induced with TALENs in the zebrafish genome. PLoS Genet. 8, e1002861 (2012)

    Article  CAS  Google Scholar 

  6. Nasevicius, A. & Ekker, S. C. Effective targeted gene ‘knockdown’ in zebrafish. Nature Genet. 26, 216–220 (2000)

    Article  CAS  Google Scholar 

  7. Doyon, Y. et al. Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases. Nature Biotechnol. 26, 702–708 (2008)

    Article  CAS  Google Scholar 

  8. Meng, X., Noyes, M. B., Zhu, L. J., Lawson, N. D. & Wolfe, S. A. Targeted gene inactivation in zebrafish using engineered zinc-finger nucleases. Nature Biotechnol. 26, 695–701 (2008)

    Article  CAS  Google Scholar 

  9. Foley, J. E. et al. Rapid mutation of endogenous zebrafish genes using zinc finger nucleases made by Oligomerized Pool ENgineering (OPEN). PLoS ONE 4, e4348 (2009)

    Article  ADS  Google Scholar 

  10. Lawson, N. D. & Wolfe, S. A. Forward and reverse genetic approaches for the analysis of vertebrate development in the zebrafish. Dev. Cell 21, 48–64 (2011)

    Article  CAS  Google Scholar 

  11. Moscou, M. J. & Bogdanove, A. J. A simple cipher governs DNA recognition by TAL effectors. Science 326, 1501 (2009)

    Article  CAS  ADS  Google Scholar 

  12. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009)

    Article  CAS  ADS  Google Scholar 

  13. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, e82 (2011)

    Article  CAS  Google Scholar 

  14. Bedell, V. M. et al. The lineage-specific gene ponzr1 is essential for zebrafish pronephric and pharyngeal arch development. Development 139, 793–804 (2012)

    Article  Google Scholar 

  15. Miller, J. C. et al. A TALE nuclease architecture for efficient genome editing. Nature Biotechnol. 29, 143–148 (2011)

    Article  CAS  Google Scholar 

  16. Mussolino, C. et al. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 39, 9283–9293 (2011)

    Article  CAS  Google Scholar 

  17. Hyatt, T. M. & Ekker, S. C. Vectors and techniques for ectopic gene expression in zebrafish. Methods Cell Biol. 59, 117–126 (1998)

    Article  Google Scholar 

  18. Wang, Y. et al. Moesin1 and Ve-cadherin are required in endothelial cells during in vivo tubulogenesis. Development 137, 3119–3128 (2010)

    Article  CAS  Google Scholar 

  19. Lawson, N. D. & Weinstein, B. M. In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev. Biol. 248, 307–318 (2002)

    Article  CAS  Google Scholar 

  20. Traver, D. et al. Transplantation and in vivo imaging of multilineage engraftment in zebrafish bloodless mutants. Nature Immunol. 4, 1238–1246 (2003)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Radecke, S., Radecke, F., Cathomen, T. & Schwarz, K. Zinc-finger nuclease-induced gene repair with oligodeoxynucleotides: wanted and unwanted target locus modifications. Mol. Ther. 18, 743–753 (2010)

    Article  CAS  Google Scholar 

  23. Thomson, J. G., Rucker, E. B., III & Piedrahita, J. A. Mutational analysis of loxP sites for efficient Cre-mediated insertion into genomic DNA. Genesis 36, 162–167 (2003)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  25. Meeker, N. D., Hutchinson, S. A., Ho, L. & Trede, N. S. Method for isolation of PCR-ready genomic DNA from zebrafish tissues. Biotechniques 43, 610–614 (2007)

    Article  CAS  Google Scholar 

  26. Blum, Y. et al. Complex cell rearrangements during intersegmental vessel sprouting and vessel fusion in the zebrafish embryo. Dev. Biol. 316, 312–322 (2008)

    Article  CAS  Google Scholar 

  27. Mahfouz, M. M. et al. De novo-engineered transcription activator-like effector (TALE) hybrid nuclease with novel DNA binding specificity creates double-strand breaks. Proc. Natl Acad. Sci. USA 108, 2623–2628 (2011)

    Article  CAS  ADS  Google Scholar 

Download references

Acknowledgements

State of Minnesota grant H001274506 to S.C.E. and D.F.V.; NIH GM63904 to S.C.E.; NIH grant P30DK084567 to S.C.E. and K.J.C.; NIH grant DK083219 to V.M.B.; Mayo Foundation; NIH DA032194 to K.J.C.; NIH grant R41HL108440 to D.F.C. and S.C.F.; NIH grant GM088424 to J.J.E.; NSF grant DBI0923827 to D.F.V.; General Research Fund (HKU771611, HKU771110, HKU769809M) from the Research Grant Council, The University of Hong Kong and the Tang King Yin Research Fund to A.C.M. and A.Y.H.L. We thank G. Davis for discussion on ssDNA use with custom restriction enzymes, H. J. Fadel for in vitro RNA synthesis, and S. Westcot for comments on this manuscript. We thank G. Moulder for help in DNA analyses and members of the Mayo Clinic Zebrafish Core Facility for excellent animal care.

Author information

Authors and Affiliations

Authors

Contributions

W.T. designed and constructed full-length pT3TS-Tal vectors targeting Danio rerio crhr1 and crhr2 against exon sequences provided by K.J.C.; D.F.C. and S.C.F. designed and produced the pT3TS-Tal and GoldyTALEN cloning vectors. D.F.C. synthesized crhr1 and crhr2 TALEN mRNA from the pT3TS-Tal vectors. C.G.S. designed and assembled ponzr1 TALENs and transferred TAL repeats from original crhr1 and crhr2 TALEN vectors into GoldyTALEN. T.L.P. and K.J.C. did initial crhr1 TALEN microinjections. T.L.P. performed initial characterization of crhr1 mutagenesis efficiency by PCR and restriction fragment length polymorphism analysis. T.L.P. and K.J.C. microinjected crhr2 TALEN and loxP oligonucleotides. T.L.P. performed initial characterization by PCR demonstrating loxP integration. R.G.K. fully characterized efficiency and sequence of somatic loxP insertions in the crhr2 locus. K.J.C. screened adult fin clips for mloxP integrations into crhr2. T.L.P. screened F1 offspring for mloxP integrations into crhr2 and together with J.M.C. cloned and sequenced integration events. K.J.C. designed experiments associated for crhr1 and crhr2 modification. K.J.C. selected loxP mutant JTZ17 for integration. A.C.M. developed initial zebrafish genetic testing, TALEN cell-free assays, and ssDNA HDR protocols. S.G.P. conducted the cell-free TALEN endonuclease assay. A.Y.H.L. contributed to the design of initial TALEN and ssDNA HDR experiments. Y.W. and J.J.E. conducted biallelic conversion TALEN experiments in somatic and germline tests. D.F.V. and S.C.E. initiated the strategy to use custom restriction enzymes for genome editing in zebrafish. S.C.E. developed the plan for HDR targeting using ssDNAs, conducted overall project design and data analysis, and wrote the initial manuscript text. All authors contributed to manuscript composition. V.M.B. and J.M.C. conducted ponzr1 and crhr1 TALEN scaffold comparison experiments. V.M.B. and K.J.C. designed ssDNA oligonucleotides for HDR experiments. V.M.B. and J.M.C. injected and screened the EcoRV and mloxP HDR experiments at the ponzr1 locus. J.M.C. ran quantitative data assessments and statistical analyses. V.M.B. and J.M.C. made first drafts and legends of Figs 1, 3 and 5. V.M.B. and J.M.C. conducted fin biopsy analyses of the ponzr1 locus. V.M.B. completed the analysis of ponzr1 germline transmission with assistance from J.M.C.

Corresponding author

Correspondence to Stephen C. Ekker.

Ethics declarations

Competing interests

S.C.F., J.J.E., K.J.C. and D.F.V. hold shares in Recombinetics, Inc., a company that uses TALENs for genome modification in large animals. D.F.V. is a listed inventor on a patent application titled “TAL effector-mediated DNA modification” that is co-owned by Iowa State University and the University of Minnesota, and has been licensed to Cellectis, a European biotechnology company.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-11, Supplementary Table 1 and legends for Supplementary Movies 1-3. (PDF 1937 kb)

Supplementary Movie 1

This movie shows red blood cell movement through the blood vessels in wild type embryos. (MOV 707 kb)

Supplementary Movie 2

This movie shows red blood cell movement through the blood vessels in cdh5 MO-injected embryos. (MOV 1831 kb)

Supplementary Movie 3

This movie shows red blood cell movement through the blood vessels in cdh5 GoldyTALEN-injected embryos. (MOV 89 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bedell, V., Wang, Y., Campbell, J. et al. In vivo genome editing using a high-efficiency TALEN system. Nature 491, 114–118 (2012). https://doi.org/10.1038/nature11537

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11537

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: 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