CRISPR/Cas9-Mediated Genome Editing as a Therapeutic Approach for Leber Congenital Amaurosis 10 - PubMed (original) (raw)

CRISPR/Cas9-Mediated Genome Editing as a Therapeutic Approach for Leber Congenital Amaurosis 10

Guo-Xiang Ruan et al. Mol Ther. 2017.

Abstract

As the most common subtype of Leber congenital amaurosis (LCA), LCA10 is a severe retinal dystrophy caused by mutations in the CEP290 gene. The most frequent mutation found in patients with LCA10 is a deep intronic mutation in CEP290 that generates a cryptic splice donor site. The large size of the CEP290 gene prevents its use in adeno-associated virus (AAV)-mediated gene augmentation therapy. Here, we show that targeted genomic deletion using the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system represents a promising therapeutic approach for the treatment of patients with LCA10 bearing the CEP290 splice mutation. We generated a cellular model of LCA10 by introducing the CEP290 splice mutation into 293FT cells and we showed that guide RNA pairs coupled with SpCas9 were highly efficient at removing the intronic splice mutation and restoring the expression of wild-type CEP290. In addition, we demonstrated that a dual AAV system could effectively delete an intronic fragment of the Cep290 gene in the mouse retina. To minimize the immune response to prolonged expression of SpCas9, we developed a self-limiting CRISPR/Cas9 system that minimizes the duration of SpCas9 expression. These results support further studies to determine the therapeutic potential of CRISPR/Cas9-based strategies for the treatment of patients with LCA10.

Keywords: CEP290; CRISPR/Cas9; LCA10.

Copyright © 2017 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.

PubMed Disclaimer

Figures

Figure 1

Figure 1

Generation of an In Vitro Model of LCA10 Using CRISPR/Cas9 (A) A schematic diagram showing the IVS26 mutation (filled star) in the CEP290 gene and the locations of the sgRNA target (antisense strand) and ssODN (sense strand) used for introducing the IVS26 mutation. The DSB site (filled triangle) induced by SpCas9 was located 15 bp downstream of the IVS26 mutation. The splicing patterns for wild-type and mutant are represented by solid and dashed lines, respectively. (B and C) Basal levels of wild-type CEP290 mRNA (B) and mutant CEP290 mRNA (C) in the wild-type (white bar), heterozygous (gray bars), and mutant 293FT cells (black bars), as determined by qRT-PCR. CEP290 mRNA levels were normalized to the levels of Cyclophilin A (PPIA) mRNA. The data are presented as the means ± SD (n = 3). Comparisons were performed using one-way ANOVA followed by the Tukey honest significant difference (HSD) post hoc test. *p < 0.05; **p < 0.01; ***p < 0.001. (D) Immunoblot analysis of lysates prepared from the wild-type (WT), heterozygous (Het), and mutant 293FT cells (MT). The membrane was probed for CEP290 protein (top) and β-actin (bottom; as a loading control). The ratio of CEP290/β-actin is shown at the bottom, with the ratio for the wild-type cells set to 1.0.

Figure 2

Figure 2

Targeted Deletion of the IVS26 Mutation with Paired sgRNAs and SpCas9 (A) Schematic diagram depicting the strategy used to remove the IVS26 mutation of CEP290 (filled star). An upstream sgRNA directs the first Cas9 cleavage to a site located upstream of the IVS26 mutation, and a downstream sgRNA directs the second Cas9 cleavage to a site downstream of the mutation. The two cleavage ends are directly ligated through the NHEJ process, resulting in the excision of the intronic fragment flanking the IVS26 mutation. The truncated intron 26 is removed during mRNA processing by the RNA splicing machinery. The locations of the upstream (U1) and downstream (D1, D2, and D3) sgRNA guide sequences are indicated in the diagram. Note that D1 sgRNA targets the sense strand, whereas the other three sgRNAs target the antisense strand. (B) PCR analysis for targeted genomic deletion. 293FT cell lines were transfected with the indicated pairs of sgRNA and SpCas9. Primers were designed to bind outside of the region to be deleted. The upper bands represent PCR products amplified from intron 26 of wild-type CEP290, whereas the lower bands represent PCR products amplified from the CEP290 allele following genomic deletion. M, 1-kb DNA ladder. (C) Percentages of wild-type, truncated, and inverted DNA in the mutant 293FT cells transfected with paired sgRNAs and SpCas9, as determined by next-generation sequencing.

Figure 3

Figure 3

Rescue of the Expression of Wild-Type CEP290 with Paired sgRNAs and SpCas9 (A and B) The levels of wild-type (A) and mutant (B) CEP290 mRNAs in the wild-type (white bars), heterozygous (gray bars), and mutant 293FT (black bars) cells transfected with paired sgRNAs and SpCas9, as measured by RT-qPCR. The data are presented as the means ± SD (n = 2). Comparisons were performed using one-way ANOVA followed by the Tukey HSD post hoc test. *p < 0.05; **p < 0.01. Ctrl, control sgRNA pair. (C) Immunoblot analysis of lysates prepared from the mutant cells transfected with paired sgRNAs and SpCas9. The membrane was probed for CEP290 protein (top) and β-actin (bottom; as a loading control). The ratio of CEP290/β-actin is shown at the bottom, with the ratio for the control sgRNA pair set to 1.0.

Figure 4

Figure 4

Targeted Deletion of the IVS26 Mutation with Single SaCas9 Plasmids or Dual SpCas9 Plasmids (A) PCR analysis of mutant 293FT cells transfected with either the single pAAV-SaCas9-paired sgRNAs plasmids or the dual SpCas9 plasmids (pAAV-SpCas9 + pAAV-U1D3) for targeted genomic deletion. Dashes indicate that there is no sgRNA pair in this plasmid. (B and C) The levels of wild-type (B) and mutant (C) CEP290 mRNAs in the mutant cells transfected with either the single pAAV-SaCas9-paired sgRNAs plasmids (white bars) or the dual SpCas9 plasmids (gray bars), as measured by qRT-PCR. The data are presented as the means ± SD (n = 3). Comparisons were performed using one-way ANOVA followed by the Tukey HSD post hoc test. *p < 0.05; **p < 0.01; ***p < 0.001; #p < 0.05 compared with the mutant cells transfected with the pAAV-SaCas9 alone (control).

Figure 5

Figure 5

Targeted Deletion of a Fragment in Intron 25 of the Cep290 Gene in the Mouse Retina Using a Dual AAV System (A and B) Micron IV fluorescence images for treated retina 1 (A) and retina 4 (B) on day 28 after subretinal injection. (C) PCR analysis showing targeted genomic deletion in the mouse retinas that received (1) AAV5-RK-EGFP (control) or AAV5-U11D11-RK-EGFP (treated) and (2) AAV5-SpCas9. The upper bands correspond to PCR products amplified from intron 25 of wild-type Cep290 gene, whereas the lower bands correspond to PCR products amplified from the Cep290 allele following U11D11-guided genomic deletion. (D) Percentages of wild-type and truncated DNA in the four treated retinas, as determined by next-generation sequencing.

Figure 6

Figure 6

Self-Limiting CRISPR/SpCas9 System (A) Schematic diagram of the pAAV-SpCas9 plasmid. The sgRNA recognition sequences were incorporated into insertion site 1 (between the minCMV promoter and SpCas9) and/or insertion site 2 (between SpCas9-NLS and SV40 pA). ITR, inverted terminal repeat. (B) Immunoblot analysis of lysates prepared from the mutant 293FT cells transfected with the pAAV-U1D3 plasmid and the self-limiting pAAV-SpCas9 plasmids, which contained the U1 sgRNA recognition sequence (U1T) and/or the D3 sgRNA recognition sequence (D3T). The membrane was probed for SpCas9 protein (top) and β-actin (bottom; as a loading control). The ratio of SpCas9/β-actin is shown at the bottom, with the ratio for the control pAAV-SpCas9 plasmid without the sgRNA recognition sequence set to 1.0. Dashes indicate that there is no sgRNA recognition sequence at this site. (C) PCR analysis for targeted genomic deletion in the mutant cells transfected with the pAAV-U1D3 plasmid and the self-limiting pAAV-SpCas9 plasmids. Three selected samples were subjected to NGS analysis, and percentages of truncated DNA were shown below the gel image. (D and E) The levels of wild-type (D) and mutant (E) mRNAs in the mutant cells transfected with the pAAV-U1D3 plasmid and the self-limiting pAAV-SpCas9 plasmids, as measured by qRT-PCR. The data are presented as the means ± SD (n = 3). Comparisons were performed using one-way ANOVA followed by the Tukey HSD post hoc test. *p < 0.05; **p < 0.01; ***p < 0.001 (compared with the mutant cells transfected with the pAAV-U1D3 plasmid alone [control]).

Similar articles

Cited by

References

    1. Leber T. Ueber Retinitis pigmentosa und angeborene Amaurose. Graefes Arch. Clin. Exp. Ophthalmol. 1869;15:1–25.
    1. Cremers F.P., van den Hurk J.A., den Hollander A.I. Molecular genetics of Leber congenital amaurosis. Hum. Mol. Genet. 2002;11:1169–1176. - PubMed
    1. Koenekoop R.K. An overview of Leber congenital amaurosis: a model to understand human retinal development. Surv. Ophthalmol. 2004;49:379–398. - PubMed
    1. den Hollander A.I., Koenekoop R.K., Yzer S., Lopez I., Arends M.L., Voesenek K.E., Zonneveld M.N., Strom T.M., Meitinger T., Brunner H.G. Mutations in the CEP290 (NPHP6) gene are a frequent cause of Leber congenital amaurosis. Am. J. Hum. Genet. 2006;79:556–561. - PMC - PubMed
    1. Chang B., Khanna H., Hawes N., Jimeno D., He S., Lillo C., Parapuram S.K., Cheng H., Scott A., Hurd R.E. In-frame deletion in a novel centrosomal/ciliary protein CEP290/NPHP6 perturbs its interaction with RPGR and results in early-onset retinal degeneration in the rd16 mouse. Hum. Mol. Genet. 2006;15:1847–1857. - PMC - PubMed

MeSH terms

Substances

Supplementary concepts

LinkOut - more resources