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:

A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice

Abstract

Many genetic liver diseases in newborns cause repeated, often lethal, metabolic crises. Gene therapy using nonintegrating viruses such as adeno-associated virus (AAV) is not optimal in this setting because the nonintegrating genome is lost as developing hepatocytes proliferate1,2. We reasoned that newborn liver may be an ideal setting for AAV-mediated gene correction using CRISPR-Cas9. Here we intravenously infuse two AAVs, one expressing Cas9 and the other expressing a guide RNA and the donor DNA, into newborn mice with a partial deficiency in the urea cycle disorder enzyme, ornithine transcarbamylase (OTC). This resulted in reversion of the mutation in 10% (6.7–20.1%) of hepatocytes and increased survival in mice challenged with a high-protein diet, which exacerbates disease. Gene correction in adult OTC-deficient mice was lower and accompanied by larger deletions that ablated residual expression from the endogenous OTC gene, leading to diminished protein tolerance and lethal hyperammonemia on a chow diet.

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: In vivo gene correction of the OTC locus in the spfash mouse liver by AAV.CRISPR-SaCas9.
Figure 2: Efficient restoration of OTC expression in the liver of spfash mice treated at neonatal stage by AAV8.CRISPR-SaCas9-mediated gene correction.
Figure 3: Time course of SaCas9 expression following neonatal vector administration and functional improvement following high-protein diet challenge.
Figure 4: Gene targeting/correction in the liver of spfash mice treated as adults by AAV8.CRISPR-SaCas9 vectors.

Similar content being viewed by others

References

  1. Cunningham, S.C., Dane, A.P., Spinoulas, A., Logan, G.J. & Alexander, I.E. Gene delivery to the juvenile mouse liver using AAV2/8 vectors. Mol. Ther. 16, 1081–1088 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Wang, L., Wang, H., Bell, P., McMenamin, D. & Wilson, J.M. Hepatic gene transfer in neonatal mice by adeno-associated virus serotype 8 vector. Hum. Gene Ther. 23, 533–539 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Batshaw, M.L., Tuchman, M., Summar, M., Seminara, J. & Members of the Urea Cycle Disorders Consortium. A longitudinal study of urea cycle disorders. Mol. Genet. Metab. 113, 127–130 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lichter-Konecki, U., Caldovic, L., Morizono, H. & Simpson, K. Ornithine transcarbamylase deficiency (1993–2013). in GeneReviews (eds. Pagon, R.A. et al.) http://www.ncbi.nlm.nih.gov/books/NBK154378/ (29 August 2013).

  5. Ah Mew, N., Krivitzky, L., McCarter, R., Batshaw, M., Tuchman, M. & Urea Cycle Disorders Consortium of the Rare Diseases Clinical Research Network. Clinical outcomes of neonatal onset proximal versus distal urea cycle disorders do not differ. J. Pediatr. 162, 324–9.e1 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Hodges, P.E. & Rosenberg, L.E. The spfash mouse: a missense mutation in the ornithine transcarbamylase gene also causes aberrant mRNA splicing. Proc. Natl. Acad. Sci. USA 86, 4142–4146 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sharma, R. et al. In vivo genome editing of the albumin locus as a platform for protein replacement therapy. Blood 126, 1777–1784 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. 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  PubMed  PubMed Central  Google Scholar 

  9. Anguela, X.M. et al. Robust ZFN-mediated genome editing in adult hemophilic mice. Blood 122, 3283–3287 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Friedland, A.E. et al. Characterization of Staphylococcus aureus Cas9: a smaller Cas9 for all-in-one adeno-associated virus delivery and paired nickase applications. Genome Biol. 16, 257 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Kleinstiver, B.P. et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523, 481–485 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Hsu, P.D., Lander, E.S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    PubMed  Google Scholar 

  16. 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  PubMed  PubMed Central  Google Scholar 

  17. Cheng, R. et al. Efficient gene editing in adult mouse livers via adenoviral delivery of CRISPR/Cas9. FEBS Lett. 588, 3954–3958 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Dingemanse, M.A. et al. Development of the ornithine cycle in rat liver: zonation of a metabolic pathway. Hepatology 24, 407–411 (1996).

    Article  CAS  PubMed  Google Scholar 

  19. Schmidt, F. & Grimm, D. CRISPR genome engineering and viral gene delivery: a case of mutual attraction. Biotechnol. J. 10, 258–272 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Vasileva, A. & Jessberger, R. Precise hit: adeno-associated virus in gene targeting. Nat. Rev. Microbiol. 3, 837–847 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Mahiny, A.J. et al. In vivo genome editing using nuclease-encoding mRNA corrects SP-B deficiency. Nat. Biotechnol. 33, 584–586 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Gao, G.P. et al. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc. Natl. Acad. Sci. USA 99, 11854–11859 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang, L. et al. AAV8-mediated hepatic gene transfer in infant rhesus monkeys (Macaca mulatta). Mol. Ther. 19, 2012–2020 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, L. et al. Targeted and genome-wide sequencing reveal single nucleotide variations impacting specificity of Cas9 in human stem cells. Nat. Commun. 5, 5507 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Tsai, S.Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Shi, Y., Falahati, R., Zhang, J., Flebbe-Rehwaldt, L. & Gaensler, K.M. Role of antigen-specific regulatory CD4+CD25+ T cells in tolerance induction after neonatal IP administration of AAV-hF.IX. Gene Ther. 20, 987–996 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Nivsarkar, M.S. et al. Evidence for contribution of CD4+ CD25+ regulatory T cells in maintaining immune tolerance to human factor IX following perinatal adenovirus vector delivery. J. Immunol. Res. 2015, 397879 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. LoDuca, P.A., Hoffman, B.E. & Herzog, R.W. Hepatic gene transfer as a means of tolerance induction to transgene products. Curr. Gene Ther. 9, 104–114 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hinderer, C. et al. Neonatal systemic AAV induces tolerance to CNS gene therapy in MPS I dogs and nonhuman primates. Mol. Ther. 23, 1298–1307 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lock, M. et al. Rapid, simple, and versatile manufacturing of recombinant adeno-associated viral vectors at scale. Hum. Gene Ther. 21, 1259–1271 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ran, F.A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Moscioni, D. et al. Long-term correction of ammonia metabolism and prolonged survival in ornithine transcarbamylase-deficient mice following liver-directed treatment with adeno-associated viral vectors. Mol. Ther. 14, 25–33 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Daly, T.M. AAV-mediated gene transfer to the liver. Methods Mol. Biol. 246, 195–199 (2004).

    CAS  PubMed  Google Scholar 

  34. Augustin, L., Mavinakere, M., Morizono, H. & Tuchman, M. Expression of wild-type and mutant human ornithine transcarbamylase genes in Chinese hamster ovary cells and lack of dominant negative effect of R141Q and R40H mutants. Pediatr. Res. 48, 842–846 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Morizono, H. et al. Expression, purification and kinetic characterization of wild-type human ornithine transcarbamylase and a recurrent mutant that produces 'late onset' hyperammonaemia. Biochem. J. 322, 625–631 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang, L. et al. Sustained correction of OTC deficiency in spf(ash) mice using optimized self-complementary AAV2/8 vectors. Gene Ther. 19, 404–410 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Penn Vector Core for supplying vectors, Penn Bioinformatics Core for assistance on deep sequencing data analysis, Y. Zhu and M. Nayalat for help on immunohistochemistry analysis, and L. Mays for assistance on manuscript preparation. This work was supported by National Institute of Child Health and Human Development P01-HD057247 (J.M.W.) and the Kettering Family Foundation (M.L.B.).

Author information

Authors and Affiliations

Authors

Contributions

L.W. and J.M.W. conceived this study. L.W., Y.Y. and J.M.W. designed the experiments. Y.Y., P.B., D.M., Z.H., J.W., H.Y. and C.X. performed the experiments. K.M. conducted the bioinformatics analysis of the deep sequencing data. J.M.W., L.W., Y.Y., P.B., H.M., K.M. and M.L.B. wrote and edited the manuscript.

Corresponding author

Correspondence to James M Wilson.

Ethics declarations

Competing interests

J.M. Wilson is an advisor to REGENXBIO, Dimension Therapeutics and Solid Gene Therapy, and is a founder of, holds equity in, and has a sponsored research agreement with REGENXBIO and Dimension Therapeutics; in addition, he is a consultant to several biopharmaceutical companies and is an inventor on patents licensed to various biopharmaceutical companies.

Integrated supplementary information

Supplementary Figure 1 In vitro validation of OTC sgRNAs and donor template.

(a) In vitro validation of sgRNAs targeted to OTC in the MC57G mouse cell line by transient transfection followed by 4-day puromycin enrichment and SURVEYOR nuclease assays. sgRNA1 showed the highest efficiency in inducing indels in the targeted loci and was therefore chosen for subsequent studies. Arrows denote SURVEYOR nuclease cleaved fragments of the OTC PCR products. Results were replicated in 2 independent experiments. (b) In vitro validation of OTC donor template. MC57G cells were transiently transfected with a plasmid co-expressing OTC sgRNA1, SaCas9, and an AgeI restriction site tagged OTC donor plasmid followed by 4-day puromycin enrichment. RFLP analysis was performed following AgeI digestion to detect HDR in vitro. Co-transfection of the AgeI-tagged OTC donor template with an SaCas9 plasmid without OTC sgRNA1 did not result in detectable HDR. Arrows denote AgeI-sensitive cleavage products resulting from HDR. Results were replicated in 2 independent experiments. Indel and HDR frequency were calculated based on band intensities31.

Supplementary Figure 2 Vector dose optimization to improve in vivo gene correction.

Postnatal day 2 spfash pups received temporal vein injection of 5x1010 GC AAV8.SaCas9 and either 5x1010 (n=5), 1x1011 (n=3), or 5x1011 (n=5) GC of AAV8.sgRNA1.donor vector. Liver samples were collected 3 weeks post vector treatment for analysis. (a) Quantification of gene correction based on the percentage of area on liver sections expressing OTC by immunostaining. (b) Quantification of OTC mRNA levels in the liver by RT-qPCR using primers spanning exons 4–5 to amplify wild-type OTC. Mean ± SEM are shown. ** P<0.01, Dunnett’s test.

Supplementary Figure 3 Time course of gene expression by Western analysis and HDR analysis by RFLP.

(a) HDR analysis by RFLP. OTC target region was PCR amplified from the liver genomic DNA isolated from untreated spfash mice or spfash mice treated with the dual AAV vectors. Untreated spfash control samples were collected at 8 weeks of age. Samples from the treated spfash mice were collected at 1, 3, and 8 weeks (n=3 animals per time point) following neonatal injection of the dual AAV8 vectors. Targeted animals received AAV8.SaCas9 (5x1010 GC/pup) and AAV8.sgRNA1.donor (5x1011 GC/pup). Untargeted animals received AAV8.SaCas9 (5x1010 GC/pup) and AAV8.control.donor (5x1011 GC/pup). AgeI digestion was performed and estimated HDR percentages are shown. (b) Western blot analysis. Liver lysates were prepared from untreated WT and spfash mice or spfash mice treated with the dual AAV vectors for detection of FLAG-SaCas9 and OTC protein.

Supplementary Figure 4 Examination of liver toxicity in animals treated with AAV8.CRISPR-SaCas9 dual vectors.

(a) Histological analysis on livers harvested 3 and 8 weeks following the dual vector treatment. Scale bar, 100 µm. (b) Liver transaminase levels in untreated spfash mice (n=9) or 8 weeks following dual vector treatment. Untargeted mice received 5x1010 GC AAV8.SaCas9 and 5x1011 of AAV8.control.donor vectors (n=8), while gene-targeted mice received 5x1010 GC AAV8.SaCas9 and 5x1011 GC of AAV8.sgRNA1.donor (n=7). Mean ± SEM are shown. There were no statistically significant differences between groups, Dunnett’s test.

Supplementary Figure 5 Examination of liver toxicity in adult animals treated with AAV8.CRISPR-SaCas9 dual vectors.

(a) Histological analysis on livers harvested 3 weeks (low-dose) or 2 weeks (high-dose) following dual vector treatment. Scale bar, 100 µm. (b) Liver transaminase levels in untreated spfash mice, or 3 weeks following low-dose dual vector treatment, or 2 weeks following high-dose dual vector treatment (n=3 for each group). Low-dose, untargeted mice received 1x1011 GC AAV8.SaCas9 and 1x1012 GC of AAV8.control.donor vectors, while low-dose, gene-targeted mice received 1x1011 GC AAV8.SaCas9 and 1x1012 GC of AAV8.sgRNA1.donor. High-dose, untargeted mice received 1x1012 GC AAV8.SaCas9 and 5x1012 GC of AAV8.control.donor vectors, while high-dose, gene-targeted mice received 1x1012 GC AAV8.SaCas9 and 5x1012 GC of AAV8.sgRNA1.donor. Mean ± SEM are shown. Adult animals received high-dose, gene-targeted vectors showed a trend of elevated ALT and AST levels, although not statistically different when compared with other groups (Dunnett’s test).

Supplementary Figure 6 Comparison of SaCas9 vector DNA and mRNA levels in the livers of neonatal treated and adult treated mice.

Neonatal spfash mice received 5x1010 GC AAV8.SaCas9 and 5x1011 GC of AAV8.sgRNA1.donor vectors and were sacrificed at 1 (n=5), 3 (n=6), and 8 weeks (n=7) after injection. Low-dose adult spfash mice received 1x1011 GC AAV8.SaCas9 and 1x1012 GC of AAV8.sgRNA1.donor vectors and were sacrificed at 3 weeks (n=3) after injection. High-dose adult spfash mice received 1x1012 GC AAV8.SaCas9 and 5x1012 GC of AAV8.sgRNA1.donor vectors and were sacrificed at 2 weeks (n=3) after injection. (a) Quantification of SaCas9 vector DNA in the liver by qPCR. (b) Quantification of SaCas9 mRNA in the liver by RT-qPCR. Mean ± SEM are shown.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1 and 3–6 (PDF 916 kb)

Supplementary Table 2 (XLSX 872 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, Y., Wang, L., Bell, P. 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). https://doi.org/10.1038/nbt.3469

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3469

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