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 utero CRISPR-mediated therapeutic editing of metabolic genes

Abstract

In utero gene editing has the potential to prenatally treat genetic diseases that result in significant morbidity and mortality before or shortly after birth. We assessed the viral vector–mediated delivery of CRISPR–Cas9 or base editor 3 in utero, seeking therapeutic modification of Pcsk9 or Hpd in wild-type mice or the murine model of hereditary tyrosinemia type 1, respectively. We observed long-term postnatal persistence of edited cells in both models, with reduction of plasma PCSK9 and cholesterol levels following in utero Pcsk9 targeting and rescue of the lethal phenotype of hereditary tyrosinemia type 1 following in utero Hpd targeting. The results of this proof-of-concept work demonstrate the possibility of efficiently performing gene editing before birth, pointing to a potential new therapeutic approach for selected congenital genetic disorders.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: In utero base editing of the Pcsk9 gene.
Fig. 2: Functional effects of in utero Pcsk9 base editing and comparison to postnatal editing.
Fig. 3: In utero base editing of Hpd in the Fah–/– mouse model.
Fig. 4: In utero Hpd base editing improves liver function and rescues the lethal phenotype of Fah–/– mice.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available within the paper and its supplementary information files. DNA sequencing data have been deposited on the NCBI Sequence Read Archive under accession SRP155635.

References

  1. Davey, M. G. et al. Induction of immune tolerance to foreign protein via adeno-associated viral vector gene transfer in mid-gestation fetal sheep. PLoS ONE. 12, e0171132 (2017).

    Article  Google Scholar 

  2. Sabatino, D. E. et al. Persistent expression of hF.IX After tolerance induction by in utero or neonatal administration of AAV-1-F.IX in hemophilia B mice. Mol. Ther. 15, 1677–1685 (2007).

    Article  CAS  Google Scholar 

  3. Mingozzi, F. et al. CD8+ T-cell responses to adeno-associated virus capsid in humans. Nat. Med. 13, 419–422 (2007).

    Article  CAS  Google Scholar 

  4. Moss, R. B. et al. Repeated aerosolized AAV-CFTR for treatment of cystic fibrosis: a randomized placebo-controlled phase 2B trial. Hum. Gene Ther. 18, 726–732 (2007).

    Article  CAS  Google Scholar 

  5. Endo, M. et al. The developmental stage determines the distribution and duration of gene expression after early intra-amniotic gene transfer using lentiviral vectors. Gene Ther. 17, 61–71 (2010).

    Article  CAS  Google Scholar 

  6. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).

    Article  CAS  Google Scholar 

  7. Kim, D. et al. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat. Biotechnol. 35, 475–480 (2017).

    Article  CAS  Google Scholar 

  8. Ahi, Y. S., Bangari, D. S. & Mittal, S. K. Adenoviral vector immunity: its implications and circumvention strategies. Curr. Gene Ther. 11, 307–320 (2011).

    Article  CAS  Google Scholar 

  9. Cohen, J. C., Boerwinkle, E., Mosley, T. H. Jr. & Hobbs, H. H. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N. Engl. J. Med. 354, 1264–1272 (2006).

    Article  CAS  Google Scholar 

  10. Ding, Q. et al. Permanent alteration of PCSK9 with in vivo CRISPR–Cas9 genome editing. Circ. Res. 115, 488–492 (2014).

    Article  CAS  Google Scholar 

  11. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186–191 (2015).

    Article  CAS  Google Scholar 

  12. Chadwick, A. C., Wang, X. & Musunuru, K. In vivo base editing of PCSK9 (proprotein convertase subtilisin/kexin type 9) as a therapeutic alternative to genome editing. Arterioscler. Thromb. Vasc. Biol. 37, 1741–1747 (2017).

    Article  CAS  Google Scholar 

  13. Yin, H. et al. Structure-guided chemical modification of guide RNA enables potent non-viral in vivo genome editing. Nat. Biotechnol. 35, 1179–1187 (2017).

    Article  CAS  Google Scholar 

  14. Wang, X. et al. CRISPR–Cas9 targeting of PCSK9 in human hepatocytes in vivo: brief report. Arterioscler. Thromb. Vasc. Biol. 36, 783–786 (2016).

  15. Lipshutz, G. S., Flebbe-Rehwaldt, L. & Gaensler, K. M. Reexpression following readministration of an adenoviral vector in adult mice after initial in utero adenoviral administration. Mol. Ther. 2, 374–380 (2000).

    Article  CAS  Google Scholar 

  16. Waddington, S. N. et al. In utero gene transfer of human factor IX to fetal mice can induce postnatal tolerance of the exogenous clotting factor. Blood 101, 1359–1366 (2003).

    Article  CAS  Google Scholar 

  17. Morrow, G. & Tanguay, R. M. Biochemical and clinical aspects of hereditary tyrosinemia type 1. Adv. Exp. Med. Biol. 959, 9–21 (2017).

    Article  Google Scholar 

  18. Yin, H. et al. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat. Biotechnol. 32, 551–553 (2014).

    Article  CAS  Google Scholar 

  19. Yin, H. et al. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat. Biotechnol. 34, 328–333 (2016).

  20. Pankowicz, F. P. et al. Reprogramming metabolic pathways in vivo with CRISPR/Cas9 genome editing to treat hereditary tyrosinaemia. Nat. Commun. 7, 12642 (2016).

    Article  CAS  Google Scholar 

  21. Aponte, J. L. et al. Point mutations in the murine fumarylacetoacetate hydrolase gene: animal models for the human genetic disorder hereditary tyrosinemia type 1. Proc. Natl Acad. Sci. USA 98, 641–645 (2001).

    Article  CAS  Google Scholar 

  22. Azuma, H. et al. Robust expansion of human hepatocytes in Fah /–/Rag2 –/–/Il2rg –/– mice. Nat. Biotechnol. 25, 903–910 (2007).

  23. Paulk, N. K. et al. Adeno-associated virus gene repair corrects a mouse model of hereditary tyrosinemia in vivo. Hepatology 51, 1200–1208 (2010).

    Article  CAS  Google Scholar 

  24. Daer, R. M., Cutts, J. P., Brafman, D. A. & Haynes, K. A. The impact of chromatin dynamics on Cas9-mediated genome editing in human cells. ACS Synth. Biol. 6, 428–438 (2017).

    Article  CAS  Google Scholar 

  25. Finn, J. D. et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Rep. 22, 2227–2235 (2018).

    Article  CAS  Google Scholar 

  26. Rafati, M., Mohamadhashem, F., Hoseini, A., Ramandi, S. D. & Ghaffari, S. R. Prenatal diagnosis of tyrosinemia type 1 using next generation sequencing. Fetal. Pediatr. Pathol. 35, 282–285 (2016).

    Article  CAS  Google Scholar 

  27. Haeussler, M. et al. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome. Biol. 17, 148 (2016).

    Article  Google Scholar 

  28. Boelig, M. M. et al. The intravenous route of injection optimizes engraftment and survival in the murine model of in utero hematopoietic cell transplantation. Biol. Blood Marrow Transplant. 22, 991–999 (2016).

    Article  Google Scholar 

  29. Wang, D. et al. Adenovirus-mediated somatic genome editing of Pten by CRISPR/Cas9 in mouse liver in spite of Cas9-specific immune responses. Hum. Gene Ther. 26, 432–442 (2015).

    Article  CAS  Google Scholar 

  30. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371–376 (2017).

    Article  CAS  Google Scholar 

  31. Yang, Y. et al. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice. Nat. Biotechnol. 34, 334–338 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants T32-HL007843 (A.C.W.), R01-HL118744 and R01-HL126875 (K.M.) from the National Institutes of Health; grant UL1-TR001878 from the National Center for Advancing Translational Sciences of the National Institutes of Health and the Institute for Translational Medicine and Therapeutics at the University of Pennsylvania (W.H.P. and K.M.); grant GE-16-001-IU from the University of Pennsylvania Orphan Disease Center (W.H.P.); the Winkelman Family Fund in Cardiovascular Innovation (K.M.); the Burroughs Wellcome Fund Career Award for Medical Scientists (R.J.); and generous family gifts to the Children’s Hospital of Philadelphia (CHOP) (W.H.P.). We thank A. Weilerstein and L. Ma for their help with animal care, the Translational Core Laboratory at CHOP for assistance with liver function tests, and A. Radu, the Pathology Core at CHOP and the Histology Core at the Cardiovascular Institute at the University of Pennsylvania for their assistance with histology.

Author information

Authors and Affiliations

Authors

Contributions

A.C.R., J.D.S., A.C.C., and H.A.H. performed the experiments and acquired and analyzed the data. A.C.R. was the lead individual on the tyrosinemia experiments, J.D.S. was the lead individual on the Pcsk9 experiments, and A.C.C. did all initial in vitro work and continued as an instrumental contributor to both the Pcsk9 and tyrosinemia projects. N.J.A. and B.E.C. provided technical help, performed the prenatal injections, and assisted in acquiring the data. H.L. provided technical help, performed the in vitro ELISA assays, and acquired/analyzed the data. P.Z. provided technical help and designed plasmids. K.S. and W.L. provided technical help, performed the qRT-PCR experiments, and assisted in acquiring the data. L.L. performed critical technical help. D.A. and W.Z. designed the initial R26mTmG/+ screening studies. R.J. provided critical technical help and the histologic analyses. E.M.M. designed the original R26mTmG/+ screening studies and provided critical experimental guidance. K.M. and W.H.P. designed all the experiments, oversaw the performance of all the experiments, analyzed the data, and wrote the paper with support from A.C.R., J.D.S., and A.C.C.

Corresponding authors

Correspondence to Kiran Musunuru or William H. Peranteau.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Tables 1–5

Reporting Summary

Supplementary Video 1

In utero vitelline vein injection of an E16 fetus with dilute trypan blue

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rossidis, A.C., Stratigis, J.D., Chadwick, A.C. et al. In utero CRISPR-mediated therapeutic editing of metabolic genes. Nat Med 24, 1513–1518 (2018). https://doi.org/10.1038/s41591-018-0184-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0184-6

This article is cited by

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