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.

  • Review Article
  • Published:

Genome editing with engineered zinc finger nucleases

Key Points

  • Targeted genetic engineering in many important model systems and in human tissue culture cells has historically been challenging. This has changed dramatically over the past 5 years with the development of zinc finger nuclease (ZFN) technology.

  • A ZFN is an artificial endonuclease that consists of a designed zinc finger protein (ZFP) fused to the cleavage domain of the FokI restriction enzyme. A ZFN may be redesigned to cleave new targets by developing ZFPs with new sequence specificities.

  • For genome engineering, a ZFN is targeted to cleave a chosen genomic sequence. The cleavage event induced by the ZFN provokes cellular repair processes that in turn mediate efficient modification of the targeted locus.

  • If the ZFN-induced cleavage event is resolved via non-homologous end joining, this can result in small deletions or insertions, effectively leading to gene knockout. This approach has now been used to establish facile and efficient reverse genetics (that is, reverse genetics that does not require selection) in Drosophila melanogaster, zebrafish, rats, Arabidopsis thaliana and mammalian somatic cells.

  • If the break is resolved via a homology-based process in the presence of an investigator-provided donor, small changes or entire transgenes can be transferred, often without selection, into the chromosome; this is referred to as 'gene correction' and 'gene addition', respectively. This approach has been used to make novel alleles in D. melanogaster, mammalian cells and tobacco, and has been used to drive targeted integration in maize, tobacco and human embryonic stem and induced pluripotent stem cells.

  • Therapeutic application of ZFN technology requires the engineering of ZFNs that are highly specific in their action. Three clinical trials with ZFNs are underway, including one in which T cells are isolated from a patient infected with HIV, treated with ZFNs that disrupt the chemokine (C-C motif) receptor type 5 (CCR5) gene to make them resistant to virus infection, and transferred back to the patient.

Abstract

Reverse genetics in model organisms such as Drosophila melanogaster, Arabidopsis thaliana, zebrafish and rats, efficient genome engineering in human embryonic stem and induced pluripotent stem cells, targeted integration in crop plants, and HIV resistance in immune cells — this broad range of outcomes has resulted from the application of the same core technology: targeted genome cleavage by engineered, sequence-specific zinc finger nucleases followed by gene modification during subsequent repair. Such 'genome editing' is now established in human cells and a number of model organisms, thus opening the door to a range of new experimental and therapeutic possibilities.

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: Structure and design of zinc finger nucleases.
Figure 2: Eliminating potential homodimerization.
Figure 3: Types of genome editing made possible using zinc finger nucleases.

Similar content being viewed by others

References

  1. Carroll, D. Progress and prospects: zinc-finger nucleases as gene therapy agents. Gene Ther. 15, 1463–1468 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Bibikova, M., Golic, M., Golic, K. G. & Carroll, D. Targeted chromosomal cleavage and mutagenesis in Drosophila using zinc-finger nucleases. Genetics 161, 1169–1175 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Bibikova, M., Beumer, K., Trautman, J. K. & Carroll, D. Enhancing gene targeting with designed zinc finger nucleases. Science 300, 764 (2003). References 2 and 3 are early studies in the field of genome editing that applied ZFNs for the disruption and correction of endogenous genes in D. melanogaster.

    CAS  PubMed  Google Scholar 

  4. Kim, Y. G., Cha, J. & Chandrasegaran, S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc. Natl Acad. Sci. USA 93, 1156–1160 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Bibikova, M. et al. Stimulation of homologous recombination through targeted cleavage by chimeric nucleases. Mol. Cell Biol. 21, 289–297 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Miller, J., McLachlan, A. D. & Klug, A. Repetitive zinc-binding domains in the protein transcription factor IIIA from Xenopus oocytes. EMBO J. 4, 1609–1614 (1985). This paper describes the discovery of the Cys2-His2 zinc finger — the most common DNA recognition motif in metazoa.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Wolfe, S. A., Nekludova, L. & Pabo, C. O. DNA recognition by Cys2His2 zinc finger proteins. Annu. Rev. Biophys. Biomol. Struct. 29, 183–212 (2000).

    CAS  PubMed  Google Scholar 

  8. Pavletich, N. P. & Pabo, C. O. Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 A. Science 252, 809–817 (1991). This study describes the first X-ray structure of a Cys2-His2 ZFP–DNA complex.

    CAS  PubMed  Google Scholar 

  9. Segal, D. J. et al. Evaluation of a modular strategy for the construction of novel polydactyl zinc finger DNA-binding proteins. Biochemistry 42, 2137–2148 (2003).

    CAS  PubMed  Google Scholar 

  10. Rebar, E. J. & Pabo, C. O. Zinc finger phage: affinity selection of fingers with new DNA-binding specificities. Science 263, 671–673 (1994).

    CAS  PubMed  Google Scholar 

  11. Jamieson, A. C., Kim, S. H. & Wells, J. A. In vitro selection of zinc fingers with altered DNA-binding specificity. Biochemistry 33, 5689–5695 (1994).

    CAS  PubMed  Google Scholar 

  12. Dreier, B., Beerli, R. R., Segal, D. J., Flippin, J. D. & Barbas, C. F. Development of zinc finger domains for recognition of the 5′-ANN-3′ family of DNA sequences and their use in the construction of artificial transcription factors. J. Biol. Chem. 276, 29466–29478 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Dreier, B. et al. Development of zinc finger domains for recognition of the 5′-CNN-3′ family DNA sequences and their use in the construction of artificial transcription factors. J. Biol. Chem. 280, 35588–35597 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Segal, D. J., Dreier, B., Beerli, R. R. & Barbas, C. F. Toward controlling gene expression at will: selection and design of zinc finger domains recognizing each of the 5′-GNN-3′ DNA target sequences. Proc. Natl Acad. Sci. USA 96, 2758–2763 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Choo, Y. & Klug, A. Toward a code for the interactions of zinc fingers with DNA: selection of randomized fingers displayed on phage. Proc. Natl Acad. Sci. USA 91, 11163–11167 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Bae, K. H. et al. Human zinc fingers as building blocks in the construction of artificial transcription factors. Nature Biotech. 21, 275–280 (2003).

    CAS  Google Scholar 

  17. 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 

  18. Segal, D. J., Crotty, J. W., Bhakta, M. S., Barbas, C. F. & Horton, N. C. Structure of Aart, a designed six-finger zinc finger peptide, bound to DNA. J. Mol. Biol. 363, 405–421 (2006).

    CAS  PubMed  Google Scholar 

  19. Wolfe, S. A., Grant, R. A., Elrod-Erickson, M. & Pabo, C. O. Beyond the 'recognition code' structures of two Cys2His2 zinc finger/TATA box complexes. Structure 9, 717–723 (2001).

    CAS  PubMed  Google Scholar 

  20. Pavletich, N. P. & Pabo, C. O. Crystal structure of a five-finger GLI–DNA complex: new perspectives on zinc fingers. Science 261, 1701–1707 (1993).

    CAS  PubMed  Google Scholar 

  21. Fairall, L., Schwabe, J. W., Chapman, L., Finch, J. T. & Rhodes, D. The crystal structure of a two zinc-finger peptide reveals an extension to the rules for zinc-finger/DNA recognition. Nature 366, 483–487 (1993).

    CAS  PubMed  Google Scholar 

  22. Houbaviy, H. B., Usheva, A., Shenk, T. & Burley, S. K. Cocrystal structure of YY1 bound to the adeno-associated virus P5 initiator. Proc. Natl Acad. Sci. USA 93, 13577–13582 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Nolte, R. T., Conlin, R. M., Harrison, S. C. & Brown, R. S. Differing roles for zinc fingers in DNA recognition: structure of a six-finger transcription factor IIIA complex. Proc. Natl Acad. Sci. USA 95, 2938–2943 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Ramirez, C. L. et al. Unexpected failure rates for modular assembly of engineered zinc fingers. Nature Methods 5, 374–375 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Vanamee, E. S., Santagata, S. & Aggarwal, A. K. FokI requires two specific DNA sites for cleavage. J. Mol. Biol. 309, 69–78 (2001).

    CAS  PubMed  Google Scholar 

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

    CAS  Google Scholar 

  27. Szczepek, M. et al. Structure-based redesign of the dimerization interface reduces the toxicity of zinc-finger nucleases. Nature Biotech. 25, 786–793 (2007).

    CAS  Google Scholar 

  28. Shimizu, Y., Bhakta, M. S. & Segal, D. J. Restricted spacer tolerance of a zinc finger nuclease with a six amino acid linker. Bioorg. Med. Chem. Lett. 19, 3970–3972 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Handel, E. M., Alwin, S. & Cathomen, T. Expanding or restricting the target site repertoire of zinc-finger nucleases: the inter-domain linker as a major determinant of target site selectivity. Mol. Ther. 17, 104–111 (2009).

    PubMed  Google Scholar 

  30. Guo, J., Gaj, T. & Barbas, C. F. Directed evolution of an enhanced and highly efficient FokI cleavage domain for zinc finger nucleases. J. Mol. Biol. 400, 96–107 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Moynahan, M. E. & Jasin, M. Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nature Rev. Mol. Cell Biol. 11, 196–207 (2010).

    CAS  Google Scholar 

  32. Lieber, M. R. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu. Rev. Biochem. 79, 181–211 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Jackson, S. P. & Bartek, J. The DNA-damage response in human biology and disease. Nature 461, 1071–1078 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Beumer, K. J. et al. Efficient gene targeting in Drosophila by direct embryo injection with zinc-finger nucleases. Proc. Natl Acad. Sci. USA 105, 19821–19826 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  Google Scholar 

  36. 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 Biotech. 26, 695–701 (2008).

    CAS  Google Scholar 

  37. 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).

    PubMed  PubMed Central  Google Scholar 

  38. Geurts, A. M. et al. Knockout rats via embryo microinjection of zinc-finger nucleases. Science 325, 433 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Mashimo, T. et al. Generation of knockout rats with X-linked severe combined immunodeficiency (X-SCID) using zinc-finger nucleases. PLoS ONE 5, e8870 (2010).

    PubMed  PubMed Central  Google Scholar 

  40. Lloyd, A., Plaisier, C. L., Carroll, D. & Drews, G. N. Targeted mutagenesis using zinc-finger nucleases in Arabidopsis. Proc. Natl Acad. Sci. USA 102, 2232–2237 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, F. et al. High frequency targeted mutagenesis in Arabidopsis thaliana using zinc finger nucleases. Proc. Natl Acad. Sci. USA 107, 12023–12028 (2010).

    Google Scholar 

  42. Osakabe, K., Osakabe, Y. & Toki, S. Site-directed mutagenesis in Arabidopsis using custom-designed zinc finger nucleases. Proc. Natl Acad. Sci. USA, 107, 12034–12039 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Thomas, K. R., Folger, K. R. & Capecchi, M. R. High frequency targeting of genes to specific sites in the mammalian genome. Cell 44, 419–428 (1986).

    CAS  PubMed  Google Scholar 

  44. Kohli, M., Rago, C., Lengauer, C., Kinzler, K. W. & Vogelstein, B. Facile methods for generating human somatic cell gene knockouts using recombinant adeno-associated viruses. Nucleic Acids Res. 32, e3 (2004).

    PubMed  PubMed Central  Google Scholar 

  45. Ruis, B. L., Fattah, K. R. & Hendrickson, E. A. The catalytic subunit of DNA-dependent protein kinase regulates proliferation, telomere length, and genomic stability in human somatic cells. Mol. Cell Biol. 28, 6182–6195 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Khan, I. F. et al. Engineering of human pluripotent stem cells by AAV-mediated gene targeting. Mol. Ther. 18, 1192–1199 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Doyon, Y., Vo, T., Choi, V. M., Gregory, P. D. & Holmes, M. C. A transient cold shock enhances zinc-finger nuclease-mediated gene disruption. Nature Methods 7, 459–460 (2010).

    CAS  PubMed  Google Scholar 

  48. Santiago, Y. et al. Targeted gene knockout in mammalian cells using engineered zinc finger nucleases. Proc. Natl Acad. Sci. USA 105, 5809–5814 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Cost, G. J. et al. BAK and BAX deletion using zinc-finger nucleases yields apoptosis-resistant CHO cells. Biotechnol. Bioeng. 105, 330–340 (2009).

    Google Scholar 

  50. Liu, P. Q. et al. Generation of a triple-gene knockout mammalian cell line using engineered zinc-finger nucleases. Biotechnol. Bioeng. 106, 97–105 (2010).

    CAS  PubMed  Google Scholar 

  51. Lee, H. J., Kim, E. & Kim, J. S. Targeted chromosomal deletions in human cells using zinc finger nucleases. Genome Res. 20, 81–89 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Orlando, S. et al. Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res. 8 Jun 2010 (doi:10.1093/nar/gkq512).

    PubMed  PubMed Central  Google Scholar 

  53. Petolino, J. F. et al. Zinc finger nuclease-mediated transgene deletion. Plant Mol. Biol. 73, 617–628 (2010).

    CAS  PubMed  Google Scholar 

  54. Brunet, E. et al. Chromosomal translocations induced at specified loci in human stem cells. Proc. Natl Acad. Sci. USA 106, 10620–10625 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Brown, J. P., Wei, W. & Sedivy, J. M. Bypass of senescence after disruption of p21CIP1/WAF1 gene in normal diploid human fibroblasts. Science 277, 831–834 (1997).

    CAS  PubMed  Google Scholar 

  56. Rouet, P., Smih, F. & Jasin, M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell Biol. 14, 8096–8106 (1994). The conclusion drawn by this paper — that the DNA ends generated by a DSB in a mitotically dividing mammalian cell are recombinogenic — provided a starting point for the development of genome editing. See also reference 57.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Puchta, H., Dujon, B. & Hohn, B. Homologous recombination in plant cells is enhanced by in vivo induction of double strand breaks into DNA by a site-specific endonuclease. Nucleic Acids Res. 21, 5034–5040 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Porteus, M. H. & Baltimore, D. Chimeric nucleases stimulate gene targeting in human cells. Science 300, 763 (2003).

    PubMed  Google Scholar 

  59. Bozas, A., Beumer, K. J., Trautman, J. K. & Carroll, D. Genetic analysis of zinc-finger nuclease-induced gene targeting in Drosophila. Genetics 182, 641–651 (2009). This paper provides the first comprehensive study of the intranuclear requirements for resolving an endogenous DSB by NHEJ rather than HDR.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Beumer, K., Bhattacharyya, G., Bibikova, M., Trautman, J. K. & Carroll, D. Efficient gene targeting in Drosophila with zinc-finger nucleases. Genetics 172, 2391–2403 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  62. Maeder, M. L. et al. Rapid 'open-source' engineering of customized zinc-finger nucleases for highly efficient gene modification. Mol. Cell 31, 294–301 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Townsend, J. A. et al. High-frequency modification of plant genes using engineered zinc-finger nucleases. Nature 459, 442–445 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Formosa, T. & Alberts, B. M. DNA synthesis dependent on genetic recombination: characterization of a reaction catalyzed by purified bacteriophage T4 proteins. Cell 47, 793–806 (1986).

    CAS  PubMed  Google Scholar 

  65. Nassif, N., Penney, J., Pal, S., Engels, W. R. & Gloor, G. B. Efficient copying of nonhomologous sequences from ectopic sites via P-element-induced gap repair. Mol. Cell Biol. 14, 1613–1625 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. 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 

  67. Goldberg, A. D. et al. Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140, 678–691 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Lombardo, A. et al. Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nature Biotech. 25, 1298–1306 (2007).

    CAS  Google Scholar 

  69. Benabdallah, B. F. et al. Targeted gene addition to human mesenchymal stromal cells as a cell-based plasma-soluble protein delivery platform. Cytotherapy 12, 394–399 (2010).

    CAS  PubMed  Google Scholar 

  70. Zou, J. et al. Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Stem Cell 5, 97–110 (2009).

    CAS  Google Scholar 

  71. Hockemeyer, D. et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nature Biotech. 27, 851–857 (2009).

    CAS  Google Scholar 

  72. DeKelver, R. C. et al. Functional genomics, proteomics, and regulatory DNA analysis in isogenic settings using zinc finger nuclease-driven transgenesis into a safe harbor locus in the human. Genome Res. 20, 1133–1142 (2010). A useful feature of ZFN-driven genome editing is the ability it provides to perform mammalian somatic cell genetics experiments in isogenic settings. This paper provides several examples of such isogenic engineering.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Hanin, M. & Paszkowski, J. Plant genome modification by homologous recombination. Curr. Opin. Plant Biol. 6, 157–162 (2003).

    CAS  PubMed  Google Scholar 

  74. Cai, C. Q. et al. Targeted transgene integration in plant cells using designed zinc finger nucleases. Plant Mol. Biol. 69, 699–709 (2009).

    CAS  PubMed  Google Scholar 

  75. Shukla, V. K. et al. Precise genome modification in the crop species Zea mays using zinc-finger nucleases. Nature 459, 437–441 (2009).

    CAS  PubMed  Google Scholar 

  76. Perez, E. E. et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nature Biotech. 26, 808–816 (2008). This paper provides data underlying the first clinical trial on the genome editing of human cells. See also reference 81.

    CAS  Google Scholar 

  77. Tuerk, C. & Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249, 505–510 (1990).

    CAS  PubMed  Google Scholar 

  78. Blackwell, T. K. & Weintraub, H. Differences and similarities in DNA-binding preferences of MyoD and E2A protein complexes revealed by binding site selection. Science 250, 1104–1110 (1990).

    CAS  PubMed  Google Scholar 

  79. Phillips, C. M. et al. Identification of chromosome sequence motifs that mediate meiotic pairing and synapsis in, C. elegans. Nature Cell Biol. 11, 934–942 (2009).

    CAS  PubMed  Google Scholar 

  80. Reik, A. et al. Zinc finger nucleases targeting the glucocorticoid receptor allow IL-13 zetakine transgenic CTLs to kill glioblastoma cells in vivo in the presence of immunosuppressing glucocorticoids. Mol. Ther. 16, S13–S14 (2008).

    Google Scholar 

  81. Holt, N. et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nuclease targeted to CCR5 control HIV-1 in vivo. Nature Biotech. 20 Jul 2010 (doi:10.1038/nbt.1663).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bot., A. The landmark approval of Provenge, what it means to immunology and 'in this issue': the complex relation between vaccines and autoimmunity. Int. Rev. Immunol. 29, 235–238 (2010).

    CAS  PubMed  Google Scholar 

  83. Hutter, G. et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N. Engl. J. Med. 360, 692–698 (2009).

    PubMed  Google Scholar 

  84. Hurt, J. A., Thibodeau, S. A., Hirsh, A. S., Pabo, C. O. & Joung, J. K. Highly specific zinc finger proteins obtained by directed domain shuffling and cell-based selection. Proc. Natl Acad. Sci. USA 100, 12271–12276 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Mandell, J. G. & Barbas, C. F. Zinc Finger Tools: custom DNA-binding domains for transcription factors and nucleases. Nucleic Acids Res. 34, W516–W523 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Sander, J. D., Zaback, P., Joung, J. K., Voytas, D. F. & Dobbs, D. An affinity-based scoring scheme for predicting DNA-binding activities of modularly assembled zinc-finger proteins. Nucleic Acids Res. 37, 506–515 (2009).

    CAS  PubMed  Google Scholar 

  87. Moore, M., Klug, A. & Choo, Y. Improved DNA binding specificity from polyzinc finger peptides by using strings of two-finger units. Proc. Natl Acad. Sci. USA 98, 1437–1441 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Bartsevich, V. V. & Juliano, R. L. Regulation of the MDR1 gene by transcriptional repressors selected using peptide combinatorial libraries. Mol. Pharmacol. 58, 1–10 (2000).

    CAS  PubMed  Google Scholar 

  89. Isalan, M., Klug, A. & Choo, Y. A rapid, generally applicable method to engineer zinc fingers illustrated by targeting the HIV-1 promoter. Nature Biotech. 19, 656–660 (2001).

    CAS  Google Scholar 

  90. Greisman, H. A. & Pabo, C. O. A general strategy for selecting high-affinity zinc finger proteins for diverse DNA target sites. Science 275, 657–661 (1997).

    CAS  PubMed  Google Scholar 

  91. Rebar, E. J. & Miller, J. C. Design and applications of engineered zinc finger proteins. Biotech International 20–23 (April/May 2004).

  92. Malphettes, L. et al. Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies. Biotechnol. Bioeng. 106, 774–783 (2010).

    CAS  PubMed  Google Scholar 

  93. Siekmann, A. F., Standley, C., Fogarty, K. E., Wolfe, S. A. & Lawson, N. D. Chemokine signaling guides regional patterning of the first embryonic artery. Genes Dev. 23, 2272–2277 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Del Prete, G. Q. et al. Derivation and characterization of a simian immunodeficiency virus SIVmac239 variant with tropism for CXCR4. J. Virol. 83, 9911–9922 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Philip D. Gregory.

Ethics declarations

Competing interests

Fyodor D. Urnov, Edward J. Rebar, Michael C. Holmes, H. Steve Zhang and Philip D. Gregory are full time employees of Sangamo BioSciences Inc.

Related links

Related links

DATABASES

ClinicalTrials.gov 

NCT00842634

NCT01044654

NCT01082926

Glossary

Paralogues

Two or more genes within a given species that originated from a single parent gene via duplication events, usually with a subsequent, sometimes subtle, divergence of function.

Pseudogenes

Non-functional paralogues that often lack promoter or intron sequences.

Adeno-associated viruses

(AAVs). As defined by Flint and colleagues in Principles of Virology, AAV is a parvovirus (also known as a dependovirus) that can establish a latent infection during which its DNA is integrated into the host cell genome in an inactive state. AAV2 integrates into the PPP1R12C gene locus on chromosome 19.

Homing endonuclease

A 'meganuclease', such as I-SceI, that recognizes and cuts longer sequences (18 bp in the case of I-SceI) than those cut by commonly used restriction enzymes.

Episomal

The formal, technical definition of 'episome', and its distinction from the word 'plasmid' (as proposed by Jacob and Wollman in 1958), is complex. In vernacular use it means 'circular extrachromosomal DNA molecule present inside the nucleus'.

Protoplasts

Plant cells that lack a cell wall.

Calli

(Sing. callus.) Clusters of undifferentiated plant cells grown on solid medium that, in some species and under specific culture conditions, have the capacity to regenerate a whole plant.

Short hairpin RNA

Small RNAs that form hairpins that can induce sequence-specific silencing in mammalian cells through RNAi.

Systematic evolution of ligands by exponential enrichment

(SELEX). A method for identifying nucleic acid ligands for a chosen 'bait' molecule (typically a protein). In its most general form, the method comprises: incubation of a randomized nucleic acid library with the bait molecule; recovery of the bait, along with any bound nucleic acids; amplification of recovered nucleic acids via PCR; and sequencing to identify binding motifs.

Embryogenic suspension cells

Plant cells, derived from callus tissue or from an embryo, that can be maintained in liquid growth medium and that, under appropriate culture conditions, can be used to regenerate a whole plant.

Autologous

In transplantology, referring to cells or an organ transplanted from an individual to that same individual (often after some ex vivo procedure has been performed).

Intrabodies

Antibodies that are directed against intracellular target molecules and expressed within a specific subcellular compartment as directed by localization signals genetically fused to the amino or carboxyl terminus of a given antibody.

Aviraemic

Aviraemia refers to the lack of detectable virus in the circulation of an individual.

Ultradeep sequencing

An umbrella term that refers to several independent, proprietary, high-throughput DNA-sequencing technologies that use massively parallel sequencing-by-synthesis approaches. The new methods allow an increase in generated sequence per run of about two orders of magnitude compared with conventional Sanger sequencing technologies.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Urnov, F., Rebar, E., Holmes, M. et al. Genome editing with engineered zinc finger nucleases. Nat Rev Genet 11, 636–646 (2010). https://doi.org/10.1038/nrg2842

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg2842

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