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.

  • Article
  • Published:

Rapid reverse genetic screening using CRISPR in zebrafish

Subjects

Abstract

Identifying genes involved in biological processes is critical for understanding the molecular building blocks of life. We used engineered CRISPR (clustered regularly interspaced short palindromic repeats) to efficiently mutate specific loci in zebrafish (Danio rerio) and screen for genes involved in vertebrate biological processes. We found that increasing CRISPR efficiency by injecting optimized amounts of Cas9-encoding mRNA and multiplexing single guide RNAs (sgRNAs) allowed for phenocopy of known mutants across many phenotypes in embryos. We performed a proof-of-concept screen in which we used intersecting, multiplexed pool injections to examine 48 loci and identified two new genes involved in electrical-synapse formation. By deep sequencing target loci, we found that 90% of the genes were effectively screened. We conclude that CRISPR can be used as a powerful reverse genetic screening strategy in vivo in a vertebrate system.

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

Figure 1: Optimizing multiplexed CRISPR screening for electrical synapses.
Figure 2: CRISPR screening identified new genes required for electrical synaptogenesis.

Similar content being viewed by others

Accession codes

Primary accessions

BioProject

Sequence Read Archive

References

  1. Chang, N. et al. Genome editing with RNA-guided Cas9 nuclease in zebrafish embryos. Cell Res. 23, 465–472 (2013).

    Article  CAS  Google Scholar 

  2. Hwang, W.Y. et al. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat. Biotechnol. 31, 227–229 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Peng, Y. et al. Making designer mutants in model organisms. Development 141, 4042–4054 (2014).

    Article  CAS  Google Scholar 

  5. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  Google Scholar 

  6. Jao, L.-E., Wente, S.R. & Chen, W. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proc. Natl. Acad. Sci. USA 110, 13904–13909 (2013).

    Article  CAS  Google Scholar 

  7. Sung, Y.H. et al. Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Res. 24, 125–131 (2014).

    Article  CAS  Google Scholar 

  8. Rohrschneider, M.R., Elsen, G.E. & Prince, V.E. Zebrafish Hoxb1a regulates multiple downstream genes including prickle1b. Dev. Biol. 309, 358–372 (2007).

    Article  CAS  Google Scholar 

  9. Jessen, J.R. et al. Zebrafish trilobite identifies new roles for Strabismus in gastrulation and neuronal movements. Nat. Cell Biol. 4, 610–615 (2002).

    Article  CAS  Google Scholar 

  10. Satou, C. et al. Functional role of a specialized class of spinal commissural inhibitory neurons during fast escapes in zebrafish. J. Neurosci. 29, 6780–6793 (2009).

    Article  CAS  Google Scholar 

  11. Pereda, A.E. Electrical synapses and their functional interactions with chemical synapses. Nat. Rev. Neurosci. 15, 250–263 (2014).

    Article  CAS  Google Scholar 

  12. Rash, J.E. et al. Molecular and functional asymmetry at a vertebrate electrical synapse. Neuron 79, 957–969 (2013).

    Article  CAS  Google Scholar 

  13. Wang, T., Wei, J.J., Sabatini, D.M. & Lander, E.S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  Google Scholar 

  14. Craig, A.M. & Kang, Y. Neurexin-neuroligin signaling in synapse development. Curr. Opin. Neurobiol. 17, 43–52 (2007).

    Article  CAS  Google Scholar 

  15. Xiao, A. et al. Chromosomal deletions and inversions mediated by TALENs and CRISPR/Cas in zebrafish. Nucleic Acids Res. 41, e141 (2013).

    Article  CAS  Google Scholar 

  16. Hsu, P.D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    Article  CAS  Google Scholar 

  17. Gagnon, J.A. et al. Efficient mutagenesis by Cas9 protein-mediated oligonucleotide insertion and large-scale assessment of single-guide RNAs. PLoS One 9, e98186 (2014).

    Article  Google Scholar 

  18. Howe, K. et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature 496, 498–503 (2013).

    Article  CAS  Google Scholar 

  19. Kelly, A. & Hurlstone, A.F. The use of RNAi technologies for gene knockdown in zebrafish. Brief Funct. Genomics 10, 189–196 (2011).

    Article  CAS  Google Scholar 

  20. Schulte-Merker, S. & Stainier, D.Y.R. Out with the old, in with the new: reassessing morpholino knockdowns in light of genome editing technology. Development 141, 3103–3104 (2014).

    Article  CAS  Google Scholar 

  21. Hruscha, A. et al. Efficient CRISPR/Cas9 genome editing with low off-target effects in zebrafish. Development 140, 4982–4987 (2013).

    Article  CAS  Google Scholar 

  22. Twigg, S.R.F. et al. Cellular interference in craniofrontonasal syndrome: males mosaic for mutations in the X-linked EFNB1 gene are more severely affected than true hemizygotes. Hum. Mol. Genet. 22, 1654–1662 (2013).

    Article  CAS  Google Scholar 

  23. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  Google Scholar 

  24. Bassett, A.R., Tibbit, C., Ponting, C.P. & Liu, J.-L. Highly efficient targeted mutagenesis of Drosophila with the CRISPR/Cas9 system. Cell Rep. 4, 220–228 (2013).

    Article  CAS  Google Scholar 

  25. Lo, T.-W. et al. Precise and heritable genome editing in evolutionarily diverse nematodes using TALENs and CRISPR/Cas9 to engineer insertions and deletions. Genetics 195, 331–348 (2013).

    Article  CAS  Google Scholar 

  26. Nakayama, T. et al. Cas9-based genome editing in Xenopus tropicalis. Methods Enzymol. 546, 355–375 (2014).

    Article  CAS  Google Scholar 

  27. Yen, S.-T. et al. Somatic mosaicism and allele complexity induced by CRISPR/Cas9 RNA injections in mouse zygotes. Dev. Biol. 393, 3–9 (2014).

    Article  CAS  Google Scholar 

  28. Yu, C., Zhang, Y., Yao, S. & Wei, Y. A PCR based protocol for detecting indel mutations induced by TALENs and CRISPR/Cas9 in zebrafish. PLoS One 9, e98282 (2014).

    Article  Google Scholar 

  29. Pan, L. et al. Rapid identification and recovery of ENU-induced mutations with next-generation sequencing and paired-end low-error analysis. BMC Genomics 16, 1263 (2015).

    Article  Google Scholar 

  30. Breese, M.R. & Liu, Y. NGSUtils: a software suite for analyzing and manipulating next-generation sequencing datasets. Bioinformatics 29, 494–496 (2013).

    Article  CAS  Google Scholar 

  31. Güell, M., Yang, L. & Church, G.M. Genome editing assessment using CRISPR Genome Analyzer (CRISPR-GA). Bioinformatics 30, 2968–2970 (2014).

    Article  Google Scholar 

  32. Kimmel, C.B., Ballard, W.W., Kimmel, S.R., Ullmann, B. & Schilling, T.F. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253–310 (1995).

    Article  CAS  Google Scholar 

  33. Higashijima, S., Hotta, Y. & Okamoto, H. Visualization of cranial motor neurons in live transgenic zebrafish expressing green fluorescent protein under the control of the islet-1 promoter/enhancer. J. Neurosci. 20, 206–218 (2000).

    Article  CAS  Google Scholar 

  34. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank R. Garcia for superb animal care, the Moens lab for discussion and editing, J. Spiewak and the Parichy lab at the University of Washington for slc24a5 mutant embryos, S. Peterson and the Postlethwait lab at the University of Oregon for advice on sgRNA cloning, and I. Phelps and D. Doherty for help and access to the MiSeq machine. Funding was provided by the National Institutes of Health (R01HD076585 and R21NS076950 to C.B.M. and K99NS085035 to A.C.M.).

Author information

Authors and Affiliations

Authors

Contributions

A.N.S., C.F.D., A.C.W. and A.C.M. performed experiments, acquired and quantified data, and generated images for publication. A.C.M., A.N.S. and C.B.M. wrote the manuscript. All authors edited the manuscript.

Corresponding authors

Correspondence to Adam C Miller or Cecilia B Moens.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Optimizing Cas9 and sgRNA concentrations for increasing mutation efficiency.

(a) Wild-type, 2-dpf slc24a5b1 embryos and a series of slc24a5 CRISPR-injected embryos; anterior is to the left. Wild-type pigmentation is lost in slc24a5 (‘golden’) mutants. Injection of Cas9-encoding mRNA and an sgRNA targeting slc24a5 resulted in embryos with mosaic loss of eye pigmentation (a ʹ). (b) Model of the slc24a5 CRISPR target site with arrows denoting PCR primers used for sequence analysis of the locus. (c) Sequencing from embryos injected with 100/1,200 pg of slc24a5 sgRNA/Cas9-encoding mRNA. Deletions are denoted by dashes (-), and insertions are in bold. The underlined sequence denotes the NGG motif used by Cas9. (d) Quantitation of eye pigmentation as assessed by mean pixel gray value of the eye with varying amounts of Cas9-encoding mRNA and a constant 100 pg of slc24a5 sgRNA. Each open triangle represents an individual animal. (e,f) Quantitation of toxicity seen in injected embryos for varying amounts of Cas9-encoding mRNA and a constant 100 pg of slc24a5 sgRNA (e) or a constant 1,200 pg of Cas9 with varying slc24a5 sgRNA (f). Toxicity encompassed embryo death, edema, localized cell death and general developmental defects. In d, N = 24 for each condition; error bars denote s.e.m. In e and f, N > 77 embryos for each point.

Source data

Supplementary Figure 2 Optimized CRISPR can recapitulate known mutant defects in injected embryos.

(a) Model of pk1b and vangl2 sgRNA target sites. Underlined sequence denotes the NGG motif used by Cas9. (b) Images are 20-µm dorsal-view projections of the hindbrain at 2 dpf. Anterior is up. Scale bar, 20 µm. Larvae are transgenic for Tg(isl1:GFP)rw0, which marks the branchiomotor neurons. A subset of these, the facial branchiomotor neurons (shown), undergo a stereotypical migration from rhombomere 4 (r4) to r6. Mutants in the planar cell polarity pathway (pk1b and vangl2) cause a failure in this migration. These phenotypes can be recapitulated in injected CRISPR F0 embryos. (c) Quantitation of migration defects. (d) Lateral views of 1-dpf embryos. Scale bar, 1 mm. vangl2 mutants have defects in anterior-posterior axis elongation due to defects in mesodermal convergence and extension. This can be recapitulated in injected CRISPR F0 embryos. (e) Quantitation of convergent extension defect. In c and e, N > 24 embryos for each bar.

Source data

Supplementary Figure 3 Optimizing Cas9 and sgRNA concentrations for electrical-synapse phenotypes.

(a) Quantitation of mosaic electrical-synapse loss in injected embryos for varying amounts of gjd1a sgRNA injected with 1,200 pg of Cas9-encoding mRNA. (b) Quantitation of toxicity seen in injected embryos for varying amounts of gjd1a sgRNA injected with 1,200 pg of Cas9-encoding mRNA. In a, N = 24 embryos for each bar. In b, N > 86 embryos for each point.

Source data

Supplementary Figure 4 Analysis of sgRNAs and genomic alterations in CRISPR injected embryos revealed by targeted deep sequencing of screen targets.

(a) Comparison of GC content versus indel frequency. Individual targets are denoted by open squares. The solid line is a linear trend line of the data (R2 = 0.0233). (b) Analysis of sgRNA indel efficiency based on binding the non-coding (–, N = 10) or coding (+, N = 32) strand. Box-and-whisker plot denotes median, quartiles, and extremes of the data. (c) Heat map for positions 2 to N of the sgRNA showing the relative increase in indel efficiency for each specific position. N is the first position of the PAM sequence. Color scale represents increased (orange) or decreased (cyan) indel efficiency relative to average. (d) Percentage of indels that caused out-of-frame changes to the target sequence. Individual targets are denoted by open squares. (e) Histogram of deletion positions relative to the NGG site bound by Cas9. Data are included from all targets aligned to the NGG. (f) Distribution of deletion sizes. Data are combined from all targets. The x-axes of e and f are log scales.

Source data

Supplementary Figure 5 Direct genotype-phenotype association for tjp1b and electrical-synapse loss.

(a) Each triangle represents an individual animal injected with 16.5/1,200 pg tjp1b_L sgRNA/Cas9-encoding mRNA. Spinal cords were analyzed for the loss of synapses and heads were assayed for mutational efficiency on the basis of qPCR analysis of the tjp1b locus. We found that heads and spinal cords had indistinguishable mutational efficiencies (heads, 0.59 ± 0.04; tail, 0.61 ± 0.05; P = 0.84; values are average ± s.e.m. compared by Student’s t-test). The square of the mutational efficiency is plotted on the x-axis on the basis of the expectation that phenotypes are expected to be due to biallelic loss of a gene. There is a strong correlation of genotype to phenotype (R2 = 0.98). The lines represent the expected genotype-phenotype association, assuming random mutagenesis and that deleterious alleles occur only with out-of-frame mutations (dotted line, y = (2/3)2x) or both in-frame and out-of-frame mutations (solid line, y = (3/3)2x). The fact that the phenotypic rate approaches 100% suggests either that in-frame deletions are deleterious or that heterozygous loss of tjp1b effects synapse formation. Note that we found that the qPCR genotypic analysis underestimated the mutational efficiency by 6%–15% (see Online Methods). This underestimation would result in an increased slope of genotype-phenotype association plotted here. (bd) Images are 15-µm dorsal-view projections of two spinal cord segments at 5 dpf. Anterior is to the left. Scale bar, 10 µm. Larvae were stained for Connexin36 (Cx36, white) to mark the electrical synapses and for neurofilaments (RMO44, red) to mark neuronal processes, including M and CoLo. Individual Cx36 channel is shown in neighboring panel. Heterozygous loss of tjp1b has no effect on M synapse formation, whether the germline-transmitted mutation is out-of-frame (b, tjp1bfh449/+) or in-frame (c, tjp1bfh451/+). However, trans-heterozygous tjp1b mutants carrying an out-of-frame and an in-frame mutation (tjp1bfh449/fh451) lose electrical synapses, confirming that in-frame mutations are deleterious at this locus (d). In a, N = 32 embryos.

Source data

Supplementary Figure 6 Retesting sgRNAs with low indel rates from the screen revealed that most were due to degradation.

(a) qPCR quantitation of mutational efficiency in injected embryos for each of the failed target sgRNAs from the screen and a newly synthesized version. Each was individually injected at 100/1,200 pg sgRNA/Cas9-encoding mRNA. Mutation efficiency was assessed by qPCR and was done in triplicate; error bars denote s.e.m. (b) nrxn1b_S contained two polymorphisms in the designed sgRNA as compared to the genomic sequence of the fish into which it was injected. These differences likely reflect its failure to induce indels. In a, each bar represents five embryos pooled in three replicates; error bars denote s.e.m.

Source data

Supplementary Figure 7 Visual method for design, synthesis, quality control and injection of sgRNAs and Cas9-encoding mRNA into zebrafish embryos.

See the Online Methods for details on each step.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Table 1 (PDF 5579 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shah, A., Davey, C., Whitebirch, A. et al. Rapid reverse genetic screening using CRISPR in zebrafish. Nat Methods 12, 535–540 (2015). https://doi.org/10.1038/nmeth.3360

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.3360

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing