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.

  • Article
  • Published:

Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors

Abstract

Genome editing with targeted nucleases and DNA donor templates homologous to the break site has proven challenging in human hematopoietic stem and progenitor cells (HSPCs), and particularly in the most primitive, long-term repopulating cell population. Here we report that combining electroporation of zinc finger nuclease (ZFN) mRNA with donor template delivery by adeno-associated virus (AAV) serotype 6 vectors directs efficient genome editing in HSPCs, achieving site-specific insertion of a GFP cassette at the CCR5 and AAVS1 loci in mobilized peripheral blood CD34+ HSPCs at mean frequencies of 17% and 26%, respectively, and in fetal liver HSPCs at 19% and 43%, respectively. Notably, this approach modified the CD34+CD133+CD90+ cell population, a minor component of CD34+ cells that contains long-term repopulating hematopoietic stem cells (HSCs). Genome-edited HSPCs also engrafted in immune-deficient mice long-term, confirming that HSCs are targeted by this approach. Our results provide a strategy for more robust application of genome-editing technologies in HSPCs.

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: HSPCs are efficiently transduced by AAV6.
Figure 2: Combination of ZFN mRNA and AAV6 vectors promotes high levels of site-specific genome editing at the CCR5 locus in HSPCs.
Figure 3: Site-specific genome editing by AAV6 vectors uses HDR.
Figure 4: Rates of bulk culture cell growth and genome modification in erythroid and myeloid lineages.
Figure 5: Genome editing in different HSPC subsets.
Figure 6: Engraftment of NSG mice with gene-edited HSPCs.

Similar content being viewed by others

References

  1. Aiuti, A. et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 296, 2410–2413 (2002).

    Article  CAS  Google Scholar 

  2. Cartier, N. et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326, 818–823 (2009).

    Article  CAS  Google Scholar 

  3. Biffi, A. et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 341, 1233158 (2013).

    Article  Google Scholar 

  4. Aiuti, A. et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science 341, 1233151 (2013).

    Article  Google Scholar 

  5. Cavazza, A., Moiani, A. & Mavilio, F. Mechanisms of retroviral integration and mutagenesis. Hum. Gene Ther. 24, 119–131 (2013).

    Article  CAS  Google Scholar 

  6. Braun, C.J. et al. Gene therapy for Wiskott-Aldrich syndrome--long-term efficacy and genotoxicity. Sci. Transl. Med. 6, 227ra33 (2014).

    Article  Google Scholar 

  7. HaceinBeyAbina, S. et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J. Clin. Invest. 118, 3132–3142 (2008).

    Article  CAS  Google Scholar 

  8. Gaj, T., Gersbach, C.A. & Barbas, C.F. III. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31, 397–405 (2013).

    Article  CAS  Google Scholar 

  9. Sung, P. & Klein, H. Mechanism of homologous recombination: mediators and helicases take on regulatory functions. Nat. Rev. Mol. Cell Biol. 7, 739–750 (2006).

    Article  CAS  Google Scholar 

  10. Wyman, C. & Kanaar, R. DNA double-strand break repair: all's well that ends well. Annu. Rev. Genet. 40, 363–383 (2006).

    Article  CAS  Google Scholar 

  11. Brugmans, L., Kanaar, R. & Essers, J. Analysis of DNA double-strand break repair pathways in mice. Mutat. Res. 614, 95–108 (2007).

    Article  CAS  Google Scholar 

  12. Kowalczykowski, S.C. Initiation of genetic recombination and recombination-dependent replication. Trends Biochem. Sci. 25, 156–165 (2000).

    Article  CAS  Google Scholar 

  13. 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).

    Article  CAS  Google Scholar 

  14. Tebas, P. et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med. 370, 901–910 (2014).

    Article  CAS  Google Scholar 

  15. Xu, J. et al. Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. Proc. Natl. Acad. Sci. USA 110, 6518–6523 (2013).

    Article  CAS  Google Scholar 

  16. Lombardo, A. et al. Site-specific integration and tailoring of cassette design for sustainable gene transfer. Nat. Methods 8, 861–869 (2011).

    Article  CAS  Google Scholar 

  17. Coluccio, A. et al. Targeted gene addition in human epithelial stem cells by zinc-finger nuclease-mediated homologous recombination. Mol. Ther. 21, 1695–1704 (2013).

    Article  CAS  Google Scholar 

  18. Meyer, M., de Angelis, M.H., Wurst, W. & Kühn, R. Gene targeting by homologous recombination in mouse zygotes mediated by zinc-finger nucleases. Proc. Natl. Acad. Sci. USA 107, 15022–15026 (2010).

    Article  CAS  Google Scholar 

  19. Pei, Y. et al. A platform for rapid generation of single and multiplexed reporters in human iPSC lines. Sci. Rep. 5, 9205 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Genovese, P. et al. Targeted genome editing in human repopulating haematopoietic stem cells. Nature 510, 235–240 (2014).

    Article  CAS  Google Scholar 

  22. Baum, C.M., Weissman, I.L., Tsukamoto, A.S., Buckle, A.M. & Peault, B. Isolation of a candidate human hematopoietic stem-cell population. Proc. Natl. Acad. Sci. USA 89, 2804–2808 (1992).

    Article  CAS  Google Scholar 

  23. Doulatov, S., Notta, F., Laurenti, E. & Dick, J.E. Hematopoiesis: a human perspective. Cell Stem Cell 10, 120–136 (2012).

    Article  CAS  Google Scholar 

  24. Hoban, M.D. et al. Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. Blood 125, 2597–2604 (2015).

    Article  CAS  Google Scholar 

  25. Song, L. et al. Optimizing the transduction efficiency of capsid-modified AAV6 serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy 15, 986–998 (2013).

    Article  CAS  Google Scholar 

  26. Song, L. et al. High-efficiency transduction of primary human hematopoietic stem cells and erythroid lineage-restricted expression by optimized AAV6 serotype vectors in vitro and in a murine xenograft model in vivo. PLoS One 8, e58757 (2013).

    Article  CAS  Google Scholar 

  27. Veldwijk, M.R. et al. Pseudotyped recombinant adeno-associated viral vectors mediate efficient gene transfer into primary human CD34(+) peripheral blood progenitor cells. Cytotherapy 12, 107–112 (2010).

    Article  CAS  Google Scholar 

  28. Perez, E.E. et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat. Biotechnol. 26, 808–816 (2008).

    Article  CAS  Google Scholar 

  29. 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).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. Miller, J.C. et al. Improved specificity of TALE-based genome editing using an expanded RVD repertoire. Nat. Methods 12, 465–471 (2015).

    Article  CAS  Google Scholar 

  34. 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 

  35. Gabriel, R. et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat. Biotechnol. 29, 816–823 (2011).

    Article  CAS  Google Scholar 

  36. Li, L. et al. Genomic editing of the HIV-1 coreceptor CCR5 in adult hematopoietic stem and progenitor cells using zinc finger nucleases. Mol. Ther. 21, 1259–1269 (2013).

    Article  CAS  Google Scholar 

  37. Cheung, A.K., Hoggan, M.D., Hauswirth, W.W. & Berns, K.I. Integration of the adeno-associated virus genome into cellular DNA in latently infected human Detroit 6 cells. J. Virol. 33, 739–748 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Smith, J.R. et al. Robust, persistent transgene expression in human embryonic stem cells is achieved with AAVS1-targeted integration. Stem Cells 26, 496–504 (2008).

    Article  CAS  Google Scholar 

  39. Miller, D.G., Petek, L.M. & Russell, D.W. Adeno-associated virus vectors integrate at chromosome breakage sites. Nat. Genet. 36, 767–773 (2004).

    Article  CAS  Google Scholar 

  40. Lin, Y. & Waldman, A.S. Promiscuous patching of broken chromosomes in mammalian cells with extrachromosomal DNA. Nucleic Acids Res. 29, 3975–3981 (2001).

    Article  CAS  Google Scholar 

  41. Lin, Y. & Waldman, A.S. Capture of DNA sequences at double-strand breaks in mammalian chromosomes. Genetics 158, 1665–1674 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Murray, L. et al. Enrichment of human hematopoietic stem cell activity in the CD34+Thy-1+Lin- subpopulation from mobilized peripheral blood. Blood 85, 368–378 (1995).

    CAS  PubMed  Google Scholar 

  43. Majeti, R., Park, C.Y. & Weissman, I.L. Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood. Cell Stem Cell 1, 635–645 (2007).

    Article  CAS  Google Scholar 

  44. Negrin, R.S. et al. Transplantation of highly purified CD34+Thy-1+ hematopoietic stem cells in patients with metastatic breast cancer. Biol. Blood Marrow Transplant. 6, 262–271 (2000).

    Article  CAS  Google Scholar 

  45. Vose, J.M. et al. Transplantation of highly purified CD34+Thy-1+ hematopoietic stem cells in patients with recurrent indolent non-Hodgkin's lymphoma. Biol. Blood Marrow Transplant. 7, 680–687 (2001).

    Article  CAS  Google Scholar 

  46. Dick, J.E., Bhatia, M., Gan, O., Kapp, U. & Wang, J.C.Y. Assay of human stem cells by repopulation of NOD/SCID mice. Stem Cells 15(suppl. 1), 199–203, discussion 204–207 (1997).

    Article  Google Scholar 

  47. Holt, N. et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat. Biotechnol. 28, 839–847 (2010).

    Article  CAS  Google Scholar 

  48. Hogan, C.J., Shpall, E.J. & Keller, G. Differential long-term and multilineage engraftment potential from subfractions of human CD34+ cord blood cells transplanted into NOD/SCID mice. Proc. Natl. Acad. Sci. USA 99, 413–418 (2002).

    Article  CAS  Google Scholar 

  49. Dick, J.E., Guenechea, G., Gan, O.I. & Dorrell, C. In vivo dynamics of human stem cell repopulation in NOD/SCID mice. Ann. NY Acad. Sci. 938, 184–190 (2001).

    Article  CAS  Google Scholar 

  50. McKenzie, J.L., Gan, O.I., Doedens, M., Wang, J.C.Y. & Dick, J.E. Individual stem cells with highly variable proliferation and self-renewal properties comprise the human hematopoietic stem cell compartment. Nat. Immunol. 7, 1225–1233 (2006).

    Article  CAS  Google Scholar 

  51. Gu, A. et al. Engraftment and lineage potential of adult hematopoietic stem and progenitor cells is compromised following short-term culture in the presence of an aryl hydrocarbon receptor antagonist. Hum. Gene Ther. Methods 25, 221–231 (2014).

    Article  CAS  Google Scholar 

  52. Hofer, U. et al. Pre-clinical modeling of CCR5 knockout in human hematopoietic stem cells by zinc finger nucleases using humanized mice. J. Infect. Dis. 208 (suppl. 2), S160–S164 (2013).

    Article  CAS  Google Scholar 

  53. Branzei, D. & Foiani, M. Regulation of DNA repair throughout the cell cycle. Nat. Rev. Mol. Cell Biol. 9, 297–308 (2008).

    Article  CAS  Google Scholar 

  54. Notta, F. et al. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science 333, 218–221 (2011).

    Article  CAS  Google Scholar 

  55. Hirata, R., Chamberlain, J., Dong, R. & Russell, D.W. Targeted transgene insertion into human chromosomes by adeno-associated virus vectors. Nat. Biotechnol. 20, 735–738 (2002).

    Article  CAS  Google Scholar 

  56. Miller, D.G. et al. Gene targeting in vivo by adeno-associated virus vectors. Nat. Biotechnol. 24, 1022–1026 (2006).

    Article  CAS  Google Scholar 

  57. Russell, D.W. & Hirata, R.K. Human gene targeting by viral vectors. Nat. Genet. 18, 325–330 (1998).

    Article  CAS  Google Scholar 

  58. Rogers, C.S. et al. Production of CFTR-null and CFTR-DeltaF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer. J. Clin. Invest. 118, 1571–1577 (2008).

    Article  CAS  Google Scholar 

  59. Hirsch, M.L. Adeno-associated virus inverted terminal repeats stimulate gene editing. Gene Ther. 22, 190–195 (2015).

    Article  CAS  Google Scholar 

  60. Barzel, A. et al. Promoterless gene targeting without nucleases ameliorates haemophilia B in mice. Nature 517, 360–364 (2015).

    Article  CAS  Google Scholar 

  61. 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. Genome Res. 20, 1133–1142 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  63. Boczkowski, D., Nair, S.K., Nam, J.-H., Lyerly, H.K. & Gilboa, E. Induction of tumor immunity and cytotoxic T lymphocyte responses using dendritic cells transfected with messenger RNA amplified from tumor cells. Cancer Res. 60, 1028–1034 (2000).

    CAS  PubMed  Google Scholar 

  64. Wang, J. et al. Targeted gene addition to a predetermined site in the human genome using a ZFN-based nicking enzyme. Genome Res. 22, 1316–1326 (2012).

    Article  Google Scholar 

  65. Xiao, X., Li, J. & Samulski, R.J. Production of high-titer recombinant adeno-associated virus vectors in the absence of helper adenovirus. J. Virol. 72, 2224–2232 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  67. Guilinger, J.P. et al. Broad specificity profiling of TALENs results in engineered nucleases with improved DNA-cleavage specificity. Nat. Methods 11, 429–435 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by funding from the California HIV/AIDS Research Program ID12-USC-245 and F10-USC-207, US National Institutes of Health grants P01 HL73104, R01 DE025167, and U19 HL129902, the California Institute for Regenerative Medicine grant RT3-07848, and the James B. Pendleton Charitable Trust. We thank L. Truong, G. Lee and Y. Lee for CD34+ cells, E. Lopez, E. Seclen and J. Rathbun for assistance with humanized mice, E. Killingbeck and P. Cheung for MiSeq analysis, A. Goodwin, A. Kang, and H. Tran for AAV production, A. Reik and F. Urnov for AAVS1 ZFNs, and C. Wang for helpful discussions.

Author information

Authors and Affiliations

Authors

Contributions

J.W., C.M.E. and J.J.D. performed most of the experiments; S.B.H., P.W.-L.L., D.A.S., R.T.S. and G.N.L. developed assays and analyzed samples; J.W., C.M.E., P.D.G., M.C.H. and P.M.C. designed the experiments and analyzed data; J.W., C.M.E., M.C.H. and P.M.C. wrote the manuscript.

Corresponding authors

Correspondence to Michael C Holmes or Paula M Cannon.

Ethics declarations

Competing interests

The following authors are full-time employees of Sangamo BioSciences, Inc.; J.W., J.J.D., S.B.H., P.W.L., D.A.S., R.T.S., P.D.G. and M.C.H.

Integrated supplementary information

Supplementary Figure 1 Timing of AAV6 vector addition relative to ZFN mRNA electroporation.

Mobilized blood CD34+ HSPCs were transduced with AAV6 vectors containing a CCR5-XhoI donor (2,000 vg/cell) at the indicated times, pre- or post- electroporation with CCR5 ZFN mRNA (60 μg/ml). Cells were collected 5 days later and genomic DNA analyzed by Illumina deep sequencing to measure the extent of genome modification by (a) RFLP insertions and (b) indels. Data were combined from two independent experiments.

Supplementary Figure 2 AAV6 vectors and ZFN mRNA promote high levels of genome editing at the CCR5 locus in mobilized blood HSPC.

Mobilized blood CD34+ HSPCs were transduced with AAV6 vectors carrying the CCR5-RFLP donor at indicated doses (vg/cell) for 16 hours and/or electroporated with 40-60 μg/ml CCR5 ZFN mRNA. Cells were analyzed 4-5 days post-electroporation by Illumina deep sequencing to measure the efficiency of genome modification (% indels and RFLP). Results (mean ± SD) are combined from 19 separate experiments using 7 different HSPC donors. ns, no significant difference (p>0.05, unpaired t-test) in the % RFLP modification obtained at the 3,000 and 10,000 vg/cell doses of CCR5-RFLP, in combination with CCR5 ZFN mRNA.

Supplementary Figure 3 AAV6 vectors and ZFN mRNA promote high levels of genome editing at the CCR5 locus in fetal liver CD34+ HSPCs.

Fetal liver CD34+ HSPCs were transduced with AAV6-CCR5-GFP donor (1,000 vg/cell) for 24 hours then electroporated with CCR5 ZFN mRNA. Cells were analyzed at days 1 and 10 post electroporation by flow cytometry (a, c) and by In-Out PCR at day 10 (b). (a) is one representative experiment while (c) is the mean +/- SD for flow cytometry data from 4 independent experiments. **** p<0.0001, one-way ANOVA, Newman-Keuls post-test to compare all columns.

Supplementary Figure 4 AAV6 vectors and ZFN mRNA promote high levels of genome editing at the AAVS1 locus in mobilized blood HSPCs.

(a) Schematics of AAV vectors used to deliver AAVS1 homology donors. R and L refer to AAVS1 genomic sequences, comprising 801 and 840 bp respectively. (b) Mobilized blood CD34+ HSPCs were transduced with AAV6 vectors carrying the AAVS1-RFLP donor at indicated doses (vg/cell) for 16 hours and/or electroporated with AAVS1 ZFN mRNA. Cells were analyzed 3-5 days post-electroporation by Illumina deep sequencing to measure the efficiency of genome modification (% indels and RFLP). Results are combined from 6 experiments using 5 HSPC donors, and show mean +/- SD. No significant difference (p>0.05, unpaired t-test) on % RFLP was detected between cells treated with 1,000 and 3,000 vg/cell AAVS1-GFP in combination with ZFN mRNA treatment. (c) Confirmation of insertion of HindIII site at the AAVS1 locus by RFLP assay. One representative experiment is shown. (d) Mobilized blood HSPCs were treated as described in (b), but using the AAVS1-GFP donor. Cells were collected 3-6 days post-transduction and analyzed by flow cytometry for % GFP+, and by deep sequencing to measure % indels. Results were combined from 6 experiments using 4 HSPC donors and show mean +/- SD. * p<0.05, unpaired t-test between GFP+. (e) Flow cytometry plots from a representative experiment using 10,000 vg/cell AAVS1-GFP donor at 5 days post-electroporation.

Supplementary Figure 5 AAV6 vectors and ZFN mRNA promote high levels of genome editing at the AAVS1 locus in fetal liver HSPCs.

Fetal liver CD34+ HSPCs were transduced with AAVS1-GFP vectors (1,000 vg/cell) for 24 hours, then electroporated with/without AAVS1 ZFN mRNA. Cells were analyzed at day 10 post electroporation. Representative flow cytometry plots (a), or a specific In-Out PCR (b), are shown as measures of GFP addition, with the mean +/- SD for flow cytometry data from 3 independent experiments shown in (c). **** p<0.0001, unpaired t-test.

Supplementary Figure 6 Possible outcomes for AAV donor genomes.

(a) If an AAV genome containing homology arms flanking a transgene (eg promoter-GFP cassette) is introduced into a cell with matched ZFN, the possible outcomes include on-target insertion at the ZFN target site mediated by (ii) HDR, or (iii) NHEJ. In addition, off-target insertions by end-capture can occur at other DSB, which can arise from off-target activity of the nuclease (iv), or from random cellular events (v). (b) Expanded data set from Fig. 3f, showing frequency of different events. Data is mean ± SD from n=3 samples except the AAVS1 ZFN, no donor and CCR5 ZFN, no donor treatments, n=1. * p < 0.05, one-way ANOVA. (c) Relative contribution of different events to stable GFP expression at each locus, extrapolated from data in part (b), suggesting that 95.7% and 96.5% of events at the CCR5 and AAIVS1 loci, respectively, are HDR-mediated on-target GFP addition.

Supplementary Figure 7 Lineage analysis of human cells in NSG mice engrafted with genome edited HSPCs.

Neonatal NSG mice were engrafted with fetal liver HSPCs, either mock or treated with AAV6 donors (CCR5-GFP or CCR5-RFLP) and CCR5 ZFN mRNAs. Peripheral blood was harvested at 8, 12, and 16 weeks (a), and bone marrow and spleen harvested at 16 weeks (b), and analyzed for the indicated lineages of human cells. There were no significant differences in levels of each lineage between mice receiving mock and treated HSPCs in the blood (two-way ANOVA) or either tissue (one-way ANOVA).

Supplementary Figure 8 Genome edited cells are present in different lineages.

Four neonatal NSG mice were engrafted with fetal liver HSPCs, treated with CCR5-GFP vectors and CCR5 ZFN mRNA. Peripheral blood was analyzed by flow cytometry at 8, 12 and 16 weeks for GFP+ levels in each indicated human cell lineage. At the 16 week time point, bone marrow was similarly analyzed for GFP expression. No significant differences were noted in levels of GFP+ cells between lineages except in the bone marrow, where levels of GFP + cells in the CD4 + T cell fraction were significantly higher than in the other cell types (p <0.05, one-way ANOVA, Newman-Keuls post test to compare all cell types). (a) Combined data from 4 animals at each time point. (b) Representative plots showing GFP + cells in CD19 + B cells or CD4 + T cells, from blood of two different mice.

Supplementary Figure 9 Comparison of engraftment of NSG mice with fetal liver versus mobilized blood CD34+ HSPCs.

Neonatal NSG mice were transplanted with 1 million fetal liver (FL) HSPCs, and adult NSG mice were transplanted with 1 million mobilized blood (Mob) CD34+ HSPCs. The rates of engraftment with human cells achieved with mobilized blood CD34+ cells were significantly lower in blood and tissues (bone marrow and spleen) at later time points (**** = p<0.0001).

Supplementary Figure 10 Supporting data for Figure 2.

Uncropped images of gels shown in (a) Figure 2d and (b) Figure 2g. * indicates structures that can form during the last re-annealing step of the PCR, combining different types of CCR5 sequences (wild-type, indel-, or XhoI-containing sequences), and thereby resulting in heteroduplexes that are resistant to XhoI digestion. Alternatively, more than two single stranded amplicons can anneal to form complex structures.

Supplementary Figure 11 Supporting data for Figure 3.

Uncropped image of gel shown in Figure 3d.

Supplementary Figure 12 Supporting data for Figure 5.

(a) Gating strategy used to isolate primitive (P), early (E) and committed (C) progenitors from mobilized blood CD34+ HSPCs. (b) Uncropped image of gel shown in Figure 5d. (c) Uncropped image of gel shown in Figure 5g. (d) Uncropped image of gel shown in Figure 5h.

Supplementary Figure 13 Supporting data for Figure 6.

Uncropped image of gels shown in (a) Figure 6c (* = non-specific band; # = duplicate samples) and (b) Figure 6f.

Supplementary Figure 14 Amino acid sequences of AAVS1 ZFN pair used in this study.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14 and Supplementary Tables 1–4 (PDF 1824 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, J., Exline, C., DeClercq, J. et al. Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat Biotechnol 33, 1256–1263 (2015). https://doi.org/10.1038/nbt.3408

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3408

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