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.

  • Technical Report
  • Published:

CRISPR interference-based specific and efficient gene inactivation in the brain

An Author Correction to this article was published on 15 March 2018

This article has been updated

Abstract

CRISPR–Cas9 has been demonstrated to delete genes in postmitotic neurons. Compared to the establishment of proliferative cell lines or animal strains, it is more challenging to acquire a highly homogeneous consequence of gene editing in a stable neural network. Here we show that dCas9-based CRISPR interference (CRISPRi) can efficiently silence genes in neurons. Using a pseudotarget fishing strategy, we demonstrate that CRISPRi shows superior targeting specificity without detectable off-target activity. Furthermore, CRISPRi can achieve multiplex inactivation of genes fundamental for neurotransmitter release with high efficiency. By developing conditional CRISPRi tools targeting synaptotagmin I (Syt1), we modified the excitatory to inhibitory balance in the dentate gyrus of the mouse hippocampus and found that the dentate gyrus has distinct regulatory roles in learning and affective processes in mice. We therefore recommend CRISPRi as a useful tool for more rapid investigation of gene function in the mammalian brain.

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: CRISPRi efficiently inactivates gene expression in primary neurons.
Fig. 2: CRISPRi shows prominent targeting specificity in neurons.
Fig. 3: CRISPRi-based conditional inactivation of Syt1 shifts the E-I balance of the DG.
Fig. 4: Tuning of the E-I balance has distinct learning and mental regulation in the DG.
Fig. 5: Flexible multiplex gene silencing using CRISPRi in the mouse brain.

Similar content being viewed by others

Change history

  • 15 March 2018

    In the version of this article initially published, the affiliation for Jian Zhang and Shuangli Mi was incomplete. In addition to the Key Laboratory of Genomics and Precision Medicine, they are also affiliated with the University of Chinese Academy of Sciences, Beijing, China. In Supplementary Fig. 1h,l, the molecular mass marker accompanying Snap25 was labeled 58 kDa; the correct value is 25 kDa. In Supplementary Fig. 9b,c, the top panel was labeled Syt1, with molecular mass markers ranging from 46 to 100 kDa; it is actually Snap25, with molecular mass markers ranging from 17 to 46 kDa. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Kheirbek, M. A. et al. Differential control of learning and anxiety along the dorsoventral axis of the dentate gyrus. Neuron 77, 955–968 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Ramirez, S. et al. Activating positive memory engrams suppresses depression-like behaviour. Nature 522, 335–339 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Chen, Y. et al. Engineering human stem cell lines with inducible gene knockout using CRISPR/Cas9. Cell. Stem Cell. 17, 233–244 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Bauer, D. E., Canver, M. C. & Orkin, S. H. Generation of genomic deletions in mammalian cell lines via CRISPR/Cas9. J. Vis. Exp. 95, e52118 (2015).

    Google Scholar 

  5. Merkle, F. T. et al. Efficient CRISPR-Cas9-mediated generation of knockin human pluripotent stem cells lacking undesired mutations at the targeted locus. Cell Rep. 11, 875–883 (2015).

  6. Yang, H. et al. One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell 154, 1370–1379 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  10. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Incontro, S., Asensio, C. S., Edwards, R. H. & Nicoll, R. A. Efficient, complete deletion of synaptic proteins using CRISPR. Neuron 83, 1051–1057 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Straub, C., Granger, A. J., Saulnier, J. L. & Sabatini, B. L. CRISPR/Cas9-mediated gene knock-down in post-mitotic neurons. PLoS One 9, e105584 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Swiech, L. et al. In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9. Nat. Biotechnol. 33, 102–106 (2015).

    Article  PubMed  CAS  Google Scholar 

  15. Dominguez, A. A., Lim, W. A. & Qi, L. S. Beyond editing: repurposing CRISPR-Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell. Biol. 17, 5–15 (2016).

    Article  PubMed  CAS  Google Scholar 

  16. Ma, H. et al. Multiplexed labeling of genomic loci with dCas9 and engineered sgRNAs using CRISPRainbow. Nat. Biotechnol. 34, 528–530 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Nelles, D. A. et al. Programmable RNA tracking in live cells with CRISPR/Cas9. Cell 165, 488–496 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Fulco, C. P. et al. Systematic mapping of functional enhancer-promoter connections with CRISPR interference. Science 354, 769–773 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882.e21 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Thakore, P. I. et al. Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nat. Methods 12, 1143–1149 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Liu, S. J. et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science 355, eaah7111 (2017).

    Article  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  25. Söllner, T. et al. SNAP receptors implicated in vesicle targeting and fusion. Nature 362, 318–324 (1993).

    Article  PubMed  Google Scholar 

  26. Südhof, T. C. & Rothman, J. E. Membrane fusion: grappling with SNARE and SM proteins. Science 323, 474–477 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Geppert, M. et al. Synaptotagmin I: a major Ca2+ sensor for transmitter release at a central synapse. Cell 79, 717–727 (1994).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Root, D. E., Hacohen, N., Hahn, W. C., Lander, E. S. & Sabatini, D. M. Genome-scale loss-of-function screening with a lentiviral RNAi library. Nat. Methods 3, 715–719 (2006).

    Article  PubMed  CAS  Google Scholar 

  30. Groffen, A. J. et al. Doc2b is a high-affinity Ca2+ sensor for spontaneous neurotransmitter release. Science 327, 1614–1618 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Yao, J., Gaffaney, J. D., Kwon, S. E. & Chapman, E. R. Doc2 is a Ca2+ sensor required for asynchronous neurotransmitter release. Cell 147, 666–677 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Dittgen, T. et al. Lentivirus-based genetic manipulations of cortical neurons and their optical and electrophysiological monitoring in vivo. Proc. Natl. Acad. Sci. USA 101, 18206–18211 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Oh, W. J., Noggle, S. A., Maddox, D. M. & Condie, B. G. The mouse vesicular inhibitory amino acid transporter gene: expression during embryogenesis, analysis of its core promoter in neural stem cells and a reconsideration of its alternate splicing. Gene 351, 39–49 (2005).

    Article  PubMed  CAS  Google Scholar 

  34. Kerr, A. M., Reisinger, E. & Jonas, P. Differential dependence of phasic transmitter release on synaptotagmin 1 at GABAergic and glutamatergic hippocampal synapses. Proc. Natl. Acad. Sci. USA 105, 15581–15586 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Jackman, S. L. & Regehr, W. G. The mechanisms and functions of synaptic facilitation. Neuron 94, 447–464 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Deneris, E. S. & Wyler, S. C. Serotonergic transcriptional networks and potential importance to mental health. Nat. Neurosci. 15, 519–527 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Pehrson, A. L. & Sanchez, C. Altered γ-aminobutyric acid neurotransmission in major depressive disorder: a critical review of the supporting evidence and the influence of serotonergic antidepressants. Drug. Des. Devel. Ther. 9, 603–624 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Perez, S. M. & Lodge, D. J. Hippocampal interneuron transplants reverse aberrant dopamine system function and behavior in a rodent model of schizophrenia. Mol. Psychiatry 18, 1193–1198 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Lewis, D. A., Hashimoto, T. & Volk, D. W. Cortical inhibitory neurons and schizophrenia. Nat. Rev. Neurosci. 6, 312–324 (2005).

    Article  PubMed  CAS  Google Scholar 

  40. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    Article  PubMed  CAS  Google Scholar 

  41. Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C. & Doudna, J. A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 507, 62–67 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Szczelkun, M. D. et al. Direct observation of R-loop formation by single RNA-guided Cas9 and Cascade effector complexes. Proc. Natl. Acad. Sci. USA 111, 9798–9803 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Lin, Y. et al. CRISPR/Cas9 systems have off-target activity with insertions or deletions between target DNA and guide RNA sequences. Nucleic Acids Res. 42, 7473–7485 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Wu, X. et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 32, 670–676 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Kuscu, C., Arslan, S., Singh, R., Thorpe, J. & Adli, M. Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease. Nat. Biotechnol. 32, 677–683 (2014).

    Article  PubMed  CAS  Google Scholar 

  46. O’Geen, H., Henry, I. M., Bhakta, M. S., Meckler, J. F. & Segal, D. J. A genome-wide analysis of Cas9 binding specificity using ChIP-seq and targeted sequence capture. Nucleic Acids Res. 43, 3389–3404 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Cencic, R. et al. Protospacer adjacent motif (PAM)-distal sequences engage CRISPR Cas9 DNA target cleavage. PLoS. One 9, e109213 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Duan, J. et al. Genome-wide identification of CRISPR/Cas9 off-targets in human genome. Cell. Res. 24, 1009–1012 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Knight, S. C. et al. Dynamics of CRISPR-Cas9 genome interrogation in living cells. Science 350, 823–826 (2015).

    Article  PubMed  CAS  Google Scholar 

  50. 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  PubMed  CAS  Google Scholar 

  51. Radzisheuskaya, A., Shlyueva, D., Müller, I. & Helin, K. Optimizing sgRNA position markedly improves the efficiency of CRISPR/dCas9-mediated transcriptional repression. Nucleic Acids Res. 44, e141 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Forrest, A. R. et al. A promoter-level mammalian expression atlas. Nature 507, 462–470 (2014).

    Article  PubMed  CAS  Google Scholar 

  53. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank P.D. Hsu (The Salk Institute for Biological Studies, USA) and S. Shi (Memorial Sloan Kettering Cancer Center, USA) for discussion and comments. We thank J. Guan (Tsinghua University, China) for material help. We also thank all members of the Yao laboratory for assistance. This work was supported by the National Key R&D Program of China (Grant No. 2016YFA0101900, 2016YFC0903301), National Natural Science Foundation of China (Grant No. 31771482, 31471020, 31161120358), National Basic Research Program of China (Grant No. 2015CB910603), Beijing Municipal Science & Technology Commission (Grant No.Z161100002616010), the Key Research Program of the CAS (Grant No. KJZD-EW-L14), the Open Project of Key Laboratory of Genomic and Precision Medicine of the CAS, the Open Project of State Key Laboratory of Membrane Biology of China, The JPB Foundation, The Leona M. and Harry B. Helmsley Charitable Trust, Annette C. Merle-Smith, NIH grants R01MH114030 (F.H.G) and NIH U19MH106434 (F.H.G), and The G. Harold & Leila Y. Mathers Foundation.

Author information

Authors and Affiliations

Authors

Contributions

J.Y. and F.H.G. conceived the project. J.Y. and S.M. designed the experiments. Y.Z., Y.-N.L., J.Z., Y.-Z.X. and S.M. conducted ChIP experiments. Y.Z., Y.-N.L. and X.Y. conducted western blotting, qPCR and molecular biology experiments. H.Q. and B.Y. performed electrophysiological experiments and analyzed data. W.S., Y.Z., Y.C., S.-Y.L. and Y.L. conducted stereotactic infusion and immunofluorescence experiments. W.S. conducted animal behavioral experiments and analyzed data. Y.Z. and J.Y. analyzed experimental results and wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Shuangli Mi or Jun Yao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 CRISPRi shows prominent targeting specificity in neurons.

(a, e and i) Design of Syt1- and Snap25-targeting sgRNA variant arrays to test the binding of dCas9-KRAB to these two ‘false’ targets. The colored nucleic acids indicate mismatches. (b, f and j) ChIP-qPCR analysis of dCas9-KRAB binding to the targeting sites with the navigation of sgRNA variants (n = 3 for all groups). (c, g and k) qRT-PCR analysis of mRNA expression in neurons expressing different sgRNA variants (n = 3 for all groups). (d, h and l) Immunoblot analysis and quantification of protein expression in neurons with co-expression of sgRNA variants and dCas9-KRAB (n = 3 for all groups). In d, h and l, blots were cropped; full-length blots are presented in Supplementary Figure 9. All data were obtained from neurons from three independent cultures with similar results and shown as mean ± s.e.m. Statistical significance was assessed by Unpaired Student’s t-test. *P < 0.05. For detailed numbers and statistical analysis, see Supplementary Table 3.

Supplementary Figure 2 Lentiviral expression of CRISPRi in the DG.

(a) Distance (in mm) from bregma along the rostrocaudal axis. Scale bar, 1 mm. (b) Quantification of dCas9-KRAB-expressing DAPI+/GFP+ cells (n = 3). Data were obtained from neurons from three mice with similar results and shown as mean ± s.e.m.

Supplementary Figure 3 Neuronal subtype-specific expression of dCas9-KRAB in primary neurons.

Representative immunofluorescence images of pCaMKIIα- and pVGAT-driving dCas9-KRAB for specific expression in glutamatergic and GABAergic neurons, respectively. Scale bar, 100 μm. Data were obtained from neurons from three independent cultures with similar results.

Supplementary Figure 4 Expression and function of Syt2 and Syt9 in GABAergic hippocampal neurons.

(a) The mRNA expression of Syt1/2/9 in the pVGAT::dCas9-KRAB+ GABAergic neurons and pVGAT::dCas9-KRAB- glutamatergic neurons (n = 3 for all groups). Data were obtained from neurons from three independent cultures with similar results and shown as mean ± s.e.m. Statistical significance was assessed by Unpaired Student’s t-test. **P = 0.001. (b, c) Representative traces (b) and average peak amplitudes (c) of AP-evoked IPSCs showing functions of Syt2 and Syt9 in the pVGAT::dCas9-KRAB+ GABAergic neurons (n = 35, 28, 13, 32). Data were obtained from neurons from three independent cultures with similar results, n represents the number cells analyzed. Quantification is represented as a box-and-whisker plot with upper and lower whiskers representing the maximum and minimum values, respectively; the boxes represent 2.5%, median and 97.5% quartiles. Statistical significance was assessed by Unpaired Student’s t-test; **P < 0.001. For detailed numbers and statistical analysis, see Supplementary Table 3.

Supplementary Figure 5 Locomotor activity of mice with conditional Syt1 KD in the DG.

(a,b) Average velocity (a) and total locomotor distance (b) of conditional Syt1 KD mice in a 10 min open field test (n = 10, 10, 10 mice). All data were shown as mean ± s.e.m. Statistical significance was assessed by Unpaired Student’s t-test. For detailed numbers and statistical analysis, see Supplementary Table 3.

Supplementary Figure 6 dCas9-based Syt1 enhancement in primary neurons.

(a) Schematic diagram of dCas9-based synergistic activation mediator (dCas9-SAM) system for Syt1 activation. Compass arrowheads indicate the sgRNA targeting sites. (b,c) Immunoblot analysis (b) and quantification (c) of Syt1 protein level in neurons expressing the dCas9-SAM system (n = 3 for all groups). In b, blots were cropped; full-length blots are presented in Supplementary Figure 9. (d) qRT-PCR analysis of Syt1 enhancement by the dCas9-SAM system (n = 3 for all groups). Data were obtained from neurons from three independent cultures with similar results and shown as mean ± s.e.m. Statistical significance was assessed by Unpaired Student’s t-test. *P < 0.05. For detailed numbers and statistical analysis, see Supplementary Table 3.

Supplementary Figure 7

Full-length blots presented in Figures 1 and 2.

Supplementary Figure 8

Full-length blots presented in Figures 3 and 5.

Supplementary Figure 9

Full-length blots presented in Supplementary Figures 1 and 6.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zheng, Y., Shen, W., Zhang, J. et al. CRISPR interference-based specific and efficient gene inactivation in the brain. Nat Neurosci 21, 447–454 (2018). https://doi.org/10.1038/s41593-018-0077-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0077-5

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