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:

Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells

Abstract

Human induced pluripotent stem cells (iPSCs) represent a unique opportunity for regenerative medicine because they offer the prospect of generating unlimited quantities of cells for autologous transplantation, with potential application in treatments for a broad range of disorders1,2,3,4. However, the use of human iPSCs in the context of genetically inherited human disease will require the correction of disease-causing mutations in a manner that is fully compatible with clinical applications3,5. The methods currently available, such as homologous recombination, lack the necessary efficiency and also leave residual sequences in the targeted genome6. Therefore, the development of new approaches to edit the mammalian genome is a prerequisite to delivering the clinical promise of human iPSCs. Here we show that a combination of zinc finger nucleases (ZFNs)7 and piggyBac8,9 technology in human iPSCs can achieve biallelic correction of a point mutation (Glu342Lys) in the α1-antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for α1-antitrypsin deficiency. Genetic correction of human iPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene-targeting technology that is currently available and crucially prevents contamination of the host genome with residual non-human sequences. Our results provide the first proof of principle, to our knowledge, for the potential of combining human iPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.

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: Correction of the G290T mutation in the Tyr gene in mouse iPSCs.
Figure 2: Correction of the Z mutation in human A1ATD-iPSCs.
Figure 3: Functional analysis of restored A1AT in corrected iPSC-derived hepatocyte-like cells.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

Gene Expression Omnibus

Data deposits

Exome sequence data have been deposited at the European Genome-Phenome Archive (http://www.ebi.ac.uk/ega/) hosted by the European Bioinformatics Institute under accession EGAS00001000055. CGH and SNP array data have been deposited with EBI ArrayExpress (http://www.ebi.ac.uk/arrayexpress/) under accession number E-MEXP-3316 and with Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) under accession number GSE31035, respectively.

References

  1. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007)

    Article  CAS  Google Scholar 

  2. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007)

    Article  ADS  CAS  Google Scholar 

  3. Stadtfeld, M. & Hochedlinger, K. Induced pluripotency: history, mechanisms, and applications. Genes Dev. 24, 2239–2263 (2010)

    Article  CAS  Google Scholar 

  4. Hanna, J. et al. Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science 318, 1920–1923 (2007)

    Article  ADS  CAS  Google Scholar 

  5. Fairchild, P. J. The challenge of immunogenicity in the quest for induced pluripotency. Nature Rev. Immunol. 10, 868–875 (2010)

    Article  CAS  Google Scholar 

  6. Tenzen, T., Zembowicz, F. & Cowan, C. A. Genome modification in human embryonic stem cells. J. Cell. Physiol. 222, 278–281 (2010)

    Article  CAS  Google Scholar 

  7. Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S. & Gregory, P. D. Genome editing with engineered zinc finger nucleases. Nature Rev. Genet. 11, 636–646 (2010)

    Article  CAS  Google Scholar 

  8. Yusa, K., Zhou, L., Li, M. A., Bradley, A. & Craig, N. L. A hyperactive piggyBac transposase for mammalian applications. Proc. Natl Acad. Sci. USA 108, 1531–1536 (2011)

    Article  ADS  CAS  Google Scholar 

  9. Wang, W. et al. Chromosomal transposition of PiggyBac in mouse embryonic stem cells. Proc. Natl Acad. Sci. USA 105, 9290–9295 (2008)

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. van der Weyden, L., Adams, D. J. & Bradley, A. Tools for targeted manipulation of the mouse genome. Physiol. Genomics 11, 133–164 (2002)

    Article  CAS  Google Scholar 

  12. Meier, I. D. et al. Short DNA sequences inserted for gene targeting can accidentally interfere with off-target gene expression. FASEB J. 24, 1714–1724 (2010)

    Article  CAS  Google Scholar 

  13. Lacoste, A., Berenshteyn, F. & Brivanlou, A. H. An efficient and reversible transposable system for gene delivery and lineage-specific differentiation in human embryonic stem cells. Cell Stem Cell 5, 332–342 (2009)

    Article  CAS  Google Scholar 

  14. Fraser, M. J., Ciszczon, T., Elick, T. & Bauser, C. Precise excision of TTAA-specific lepidopteran transposons piggyBac (IFP2) and tagalong (TFP3) from the baculovirus genome in cell lines from two species of Lepidoptera. Insect Mol. Biol. 5, 141–151 (1996)

    Article  CAS  Google Scholar 

  15. Yusa, K., Rad, R., Takeda, J. & Bradley, A. Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon. Nature Methods 6, 363–369 (2009)

    Article  CAS  Google Scholar 

  16. Rashid, S. T. et al. Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J. Clin. Invest. 120, 3127–3136 (2010)

    Article  CAS  Google Scholar 

  17. Perlmutter, D. H. Autophagic disposal of the aggregation-prone protein that causes liver inflammation and carcinogenesis in α-1-antitrypsin deficiency. Cell Death Differ. 16, 39–45 (2009)

    Article  CAS  Google Scholar 

  18. Gooptu, B. & Lomas, D. A. Conformational pathology of the serpins: themes, variations, and therapeutic strategies. Annu. Rev. Biochem. 78, 147–176 (2009)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. Mitalipova, M. M. et al. Preserving the genetic integrity of human embryonic stem cells. Nature Biotechnol. 23, 19–20 (2005)

    Article  CAS  Google Scholar 

  21. Baker, D. E. et al. Adaptation to culture of human embryonic stem cells and oncogenesis in vivo . Nature Biotechnol. 25, 207–215 (2007)

    Article  CAS  Google Scholar 

  22. Lefort, N. et al. Human embryonic stem cells reveal recurrent genomic instability at 20q11.21. Nature Biotechnol. 26, 1364–1366 (2008)

    Article  CAS  Google Scholar 

  23. Spits, C. et al. Recurrent chromosomal abnormalities in human embryonic stem cells. Nature Biotechnol. 26, 1361–1363 (2008)

    Article  CAS  Google Scholar 

  24. Touboul, T. et al. Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology 51, 1754–1765 (2010)

    Article  CAS  Google Scholar 

  25. Fusaki, N., Ban, H., Nishiyama, A., Saeki, K. & Hasegawa, M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad., Ser. B, Phys. Biol. Sci. 85, 348–362 (2009)

    Article  CAS  Google Scholar 

  26. Watanabe, K. et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nature Biotechnol. 25, 681–686 (2007)

    Article  CAS  Google Scholar 

  27. Cadinanos, J. & Bradley, A. Generation of an inducible and optimized piggyBac transposon system. Nucleic Acids Res. 35, e87 (2007)

    Article  Google Scholar 

  28. Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011)

    Article  CAS  Google Scholar 

  29. Liu, P., Jenkins, N. A. & Copeland, N. G. A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res. 13, 476–484 (2003)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  31. Beerli, R. R. & Barbas, C. F. III Engineering polydactyl zinc-finger transcription factors. Nature Biotechnol. 20, 135–141 (2002)

    Article  CAS  Google Scholar 

  32. Pavletich, N. P. & Pabo, C. O. Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 Å. Science 252, 809–817 (1991)

    Article  ADS  CAS  Google Scholar 

  33. Guschin, D. Y. et al. A rapid and general assay for monitoring endogenous gene modification. Methods Mol. Biol. 649, 247–256 (2010)

    Article  CAS  Google Scholar 

  34. Varela, I. et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469, 539–542 (2011)

    Article  ADS  CAS  Google Scholar 

  35. The 1000 Genomes Project Consortium . A map of human genome variation from population-scale sequencing. Nature 467, 1061–1073 (2010)

  36. Seki, T. et al. Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells. Cell Stem Cell 7, 11–14 (2010)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Klug and M. Minczuk for their advice, M. A. Li for comments on the manuscript, P. Ellis, N. Hammond and C. McGee for CGH analysis, the Sanger Institute sequencing facility for exome sequencing, N. Conte and S. Rice for assistance with bioinformatic analysis, M. Alexander for her help with cell culture reagents. We also thank L. Zhang, S. Hinkley and the production group for ZFN assembly and validation, K. Tong and X. Meng for technical assistance, J. C. Miller and E. Leung for ZFN off-target site analysis and S. Abrahamson and P. D. Gregory for careful reading of the manuscript. This work was supported by the Wellcome Trust (WT077187; A.B.), the MRC Senior non-clinical fellowship and the Cambridge Hospitals National Institute for Health Research Biomedical Research Center (L.V.), the Medical Research Council and Papworth NHS Trust (D.A.L.), the Bill and Melinda Gates Foundation, Inserm and Institut Pasteur (H.S.-M.) and Japan Science and Technology Agency (N.F.). K.Y. is supported by a postdoctoral fellowship of Japan Society for the Promotion of Science. S.T.R. and F.J.R. are Wellcome Trust Clinical Training Fellows. I.V. is supported by a fellowship from the International Human Frontiers Science Program Organization.

Author information

Authors and Affiliations

Authors

Contributions

K.Y. and S.T.R. are joint first authors. D.A.L., A.B. and L.V. contributed equally to this work. K.Y., S.T.R., D.A.L., A.B. and L.V. conceived the research and wrote the manuscript with comments from all authors. K.Y. performed gene correction in mouse and human iPSCs and conducted all experiments using piggyBac in Cambridge, UK. S.T.R., E.M., A.O., N.R.F.H., F.J.R., G.A. and S.J.M. performed in vitro phenotypic analysis of corrected human iPSCs. S.T.R., H.S.-M., S.D. and J.P.D.S. performed in vivo work. I.V. performed data analysis of exome sequencing. P.Q.-L., D.E.P. and M.C.H. generated and validated ZFNs. N.F. and M.H. generated Sendai virus vectors.

Corresponding authors

Correspondence to Allan Bradley or Ludovic Vallier.

Ethics declarations

Competing interests

[Competing financial interests: P.-Q.L., D.E.P. and M.C.H. are employees of Sangamo BioSciences. N.F. and M.H. are employees of DNAVEC. All other authors declare no competing financial interests.]

Supplementary information

Supplementary Information

The file contains Supplementary Figures 1-8 with legends, Supplementary Tables 1-5, Supplementary Text and additional references. (PDF 1907 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yusa, K., Rashid, S., Strick-Marchand, H. et al. Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394 (2011). https://doi.org/10.1038/nature10424

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10424

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