Tissue-specific genome editing in Ciona embryos by CRISPR/Cas9 - PubMed (original) (raw)

Tissue-specific genome editing in Ciona embryos by CRISPR/Cas9

Alberto Stolfi et al. Development. 2014 Nov.

Abstract

The CRISPR/Cas9 system has ushered in a new era of targeted genetic manipulations. Here, we report the use of CRISPR/Cas9 to induce double-stranded breaks in the genome of the sea squirt Ciona intestinalis. We use electroporation to deliver CRISPR/Cas9 components for tissue-specific disruption of the Ebf (Collier/Olf/EBF) gene in hundreds of synchronized Ciona embryos. Phenotyping of transfected embryos in the 'F0' generation revealed that endogenous Ebf function is required for specification of Islet-expressing motor ganglion neurons and atrial siphon muscles. We demonstrate that CRISPR/Cas9 is sufficiently effective and specific to generate large numbers of embryos carrying mutations in a targeted gene of interest, which should allow for rapid screening of gene function in Ciona.

Keywords: Ascidians; CRISPR/Cas9; Ciona; Ebf; Genome editing; Islet.

© 2014. Published by The Company of Biologists Ltd.

PubMed Disclaimer

Figures

Fig. 1.

Fig. 1.

CRISPR/Cas9 components in Ciona intestinalis. (A) Top: nls::dCas9::nls::eGFP protein used to assay nuclear localization in Ciona. dCas9 is Cas9 with two point mutations that render it catalytically dead. Bottom: nls::Cas9::nls used for targeted mutagenesis. (B) Tail bud-stage embryo electroporated with Mesp>nls::dCas9::nls::eGFP, confirming proper nuclear localization in B7.5 lineage cells. Scale bar: 25 μm. (C) Ebf.774 sgRNA (F+E version). The protospacer (red) is paired with its target in Ebf. ‘F’ modification is in orange, ‘E’ modification is in green. (D) In situ hybridization using eGFP probe in gastrula-stage embryo electroporated, with U6>sgRNA(F+E)::eGFP in right hemisphere, indicating successful transcription of the sgRNA in 100% of embryos (_n_=100).

Fig. 2.

Fig. 2.

CRISPR/Cas9-mediated mutagenesis of Ebf. (A) C. intestinalis Ebf gene, showing exons (solid boxes) and introns (scale bar: 1 kb). Exons are colored according to domains: green, N terminus+DNA-binding domain (DBD); orange, IPT; magenta, atypical HLH; brown, transactivation domain; gray, untranslated regions. Promoters (proximal and distal) are indicated by elbows. Alternative splicing is indicated by dotted line, giving rise to Ebf transcript variants shown below. Both contain the conserved zinc-coordinating motif of the DBD. (B) Alignment of wild-type and mutant Ebf alleles cloned from pooled embryos electroporated with 10 μg EF1α>nls::Cas9::nls, 10 μg U6>Ebf.774 and 10 μg U6>Ebf.813. Three out of seven clones had a mutation (two unique mutations). (C) Mutant Ebf alleles cloned from MACS-sorted hCD4+ cells dissociated from embryos electroporated with 10 μg EF1α>hCD4::mCherry, 25 μg EF1α>nls::Cas9::nls and 75 μg U6>Ebf.774 (see ‘Cleavage assay of genomic DNA from MACS-enriched transfected cells’ section for details). Six out of 13 clones had a mutation (all unique). Target sequences indicated in blue. Indels and substitutions are indicated in red.

Fig. 3.

Fig. 3.

Genomic cleavage assays. (A) Cleavage assay of Ebf exon 9 amplicon from pooled embryos electroporated with EF1α>nls::Cas9::nls+U6>Ebf.774 or EF1α>nls::Cas9::nls alone. Cleavage efficiency was calculated at 31.5%. Cleavage of Ebf exon 9 amplicon from control (_Cas9-_alone) embryos was not detected. Cleavage of kit control amplicon (1:1 mix of wild-type and mutant sequences) was 43.7%. (B) Assay of amplicon from MACS-sorted cells from embryos electroporated with 10 μg EF1α>nls::Cas9::nls, 10 μg U6>Ebf.774 and 10 μg EF1α>hCD4::mCherry (for MACS selection). Cleavage efficiency in sorted hCD4+ cells was 27.1%, versus 2.4% in hCD4− flow-through and 13.7% in unsorted cells from the same pool of dissociated embryos. (C) Assay of amplicon from MACS-sorted cells from embryos electroporated with 10 μg EF1α>hCD4::mCherry, 25 μg EF1α>nls::Cas9::nls and 75 μg U6>Ebf.774. Cleavage efficiency in sorted hCD4+ cells was 66.2% and 45.1% in unsorted cells from the same embryo pool (see

supplementary material Fig. S6

). (D) Assay of pooled embryos electroporated with EF1α>nls::Cas9::nls+U6>Ebf.774, collected at 4, 8 and 12 hpf. Cleavage bands are visible at 8 hpf, but not at 4 hpf. Efficiency does not increase substantially from 8 to 12 hpf.

Fig. 4.

Fig. 4.

Phenotypic assays for tissue-specific loss of Ebf in F0 embryos. (A) Larva (17.5 hpf, 22°C) electroporated with 35 μg Sox1/2/3>nls::Cas9::nls and 25 μg U6>Control sgRNA, showing normal expression of Isl>YFP reporter in A10.57 MN (observed in 68% of larvae, _n_=100). (B) CRISPR/Cas9-targeted mutagenesis of Ebf exon 9 (35 μg Sox1/2/3>nls::Cas9::nls, 25 μg U6>Ebf.774) results in only 23% of larvae showing MN-specific Isl reporter expression (_n_=100). (C) Lost Isl expression can be rescued by a CRISPR/Cas9-resistant form of Ebf (45 μg Ebf>Ebfm774), as Isl>YFP is now seen in 83% of larvae (_n_=100). (D) Larva (26 hpf, 18°C) electroporated with 50 μg Mesp>nls::Cas9::nls and 25 μg U6>Control, showing normal ASMP migration (seen in 96% of larvae, _n_=100) and _Isl_>mCherry reporter expression in ASMPs (filled arrowhead; seen in 53% of larvae, _n_=100). (E) Upon electroporation with 50 μg Mesp>nls::Cas9::nls and 25 μg U6>Ebf.774, ASMP migration is observed in only 22% of larvae (_n_=100) and _Isl_>mCherry expression in ASMPs is only seen in 3% of larvae (_n_=100; empty arrowhead). Asterisk indicates _Isl_>mCherry expression in A10.57 neuron, confirming that disruption of Ebf was B7.5 lineage specific. (F) Control larvae (22 hpf, 18°C) show normal expression of Mrf (assayed by in situ mRNA hybridization) in ASMPs (filled arrowheads; observed in 79% of larvae, _n_=100). (G) Mrf expression in ASMPs is downregulated upon CRISPR/Cas9 targeting of Ebf (50 μg Mesp>nls::Cas9::nls+25 μg U6>Ebf.774), as Mrf expression is seen in only 38% of larvae (_n_=100; empty arrowheads). (H,I) Histograms indicating the fraction of electroporated embryos displaying the phenotypes represented in A-G.

References

    1. Barrangou R., Fremaux C., Deveau H., Richards M., Boyaval P., Moineau S., Romero D. A. and Horvath P. (2007). CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709-1712 10.1126/science.1138140 - DOI - PubMed
    1. Beh J., Shi W., Levine M., Davidson B. and Christiaen L. (2007). FoxF is essential for FGF-induced migration of heart progenitor cells in the ascidian Ciona intestinalis. Development 134, 3297-3305 10.1242/dev.010140 - DOI - PubMed
    1. Brouns S. J. J., Jore M. M., Lundgren M., Westra E. R., Slijkhuis R. J. H., Snijders A. P. L., Dickman M. J., Makarova K. S., Koonin E. V. and van der Oost J. (2008). Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 321, 960-964 10.1126/science.1159689 - DOI - PMC - PubMed
    1. Chen B., Gilbert L. A., Cimini B. A., Schnitzbauer J., Zhang W., Li G.-W., Park J., Blackburn E. H., Weissman J. S., Qi L. S. et al. (2013). Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479-1491 10.1016/j.cell.2013.12.001 - DOI - PMC - PubMed
    1. Christiaen L., Wagner E., Shi W. and Levine M. (2009a). Isolation of sea squirt (Ciona) gametes, fertilization, dechorionation, and development. Cold Spring Harb. Protoc. 2009, ppdb.prot5344 10.1101/pdb.prot5344 - DOI - PubMed

Publication types

MeSH terms

LinkOut - more resources