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:

In vivo simultaneous transcriptional activation of multiple genes in the brain using CRISPR–dCas9-activator transgenic mice

Abstract

Despite rapid progresses in the genome-editing field, in vivo simultaneous overexpression of multiple genes remains challenging. We generated a transgenic mouse using an improved dCas9 system that enables simultaneous and precise in vivo transcriptional activation of multiple genes and long noncoding RNAs in the nervous system. As proof of concept, we were able to use targeted activation of endogenous neurogenic genes in these transgenic mice to directly and efficiently convert astrocytes into functional neurons in vivo. This system provides a flexible and rapid screening platform for studying complex gene networks and gain-of-function phenotypes 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: Design of an improved activator SPH.
Fig. 2: Generation of a Cre-dependent SPH transgenic mouse and activation of genes and lncRNAs in primary cells.
Fig. 3: SPH-based targeted activations convert astrocytes into functional neurons in vivo.
Fig. 4: Complex transcriptional activation in the intact brain using a single sgRNA array.

Similar content being viewed by others

References

  1. Wu, Z., Yang, H. & Colosi, P. Effect of genome size on AAV vector packaging. Mol. Ther. 18, 80–86, https://doi.org/10.1038/mt.2009.255 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. Nat. Methods 12, 664–670, https://doi.org/10.1038/Nmeth.3433 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588, https://doi.org/10.1038/nature14136 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Zetsche, B., Volz, S. E. & Zhang, F. A split-Cas9 architecture for inducible genome editing and transcription modulation. Nat. Biotechnol. 33, 139–142 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Dahlman, J. E. et al. Orthogonal gene knockout and activation with a catalytically active Cas9 nuclease. Nat. Biotechnol. 34, 441–441 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183, https://doi.org/10.1016/j.cell.2013.02.022 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451, https://doi.org/10.1016/j.cell.2013.06.044 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646, https://doi.org/10.1016/j.cell.2014.09.039 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Perez-Pinera, P. et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat. Methods 10, 973–976, https://doi.org/10.1038/Nmeth.2600 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661, https://doi.org/10.1016/j.cell.2014.09.029 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zalatan, J. G. et al. Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds. Cell 160, 339–350, https://doi.org/10.1016/j.cell.2014.11.052 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Polstein, L. R. & Gersbach, C. A. A light-inducible CRISPR-Cas9 system for control of endogenous gene activation. Nat. Chem. Biol. 11, 198–200, https://doi.org/10.1038/nchembio.1753 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cheng, A. W. et al. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell. Res. 23, 1163–1171, https://doi.org/10.1038/cr.2013.122 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gao, Y. et al. Complex transcriptional modulation with orthogonal and inducible dCas9 regulators. Nat. Methods 13, 1043–1049, https://doi.org/10.1038/nmeth.4042 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Maeder, M. L. et al. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 10, 977–979, https://doi.org/10.1038/Nmeth.2598 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bikard, D. et al. Programmable repression and activation of bacterial gene expression using an engineered CRISPR-Cas system. Nucleic Acids Res. 41, 7429–7437, https://doi.org/10.1093/nar/gkt520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mali, P. et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31, 833–838, https://doi.org/10.1038/nbt.2675 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chavez, A. et al. Highly efficient Cas9-mediated transcriptional programming. Nat. Methods 12, 326–328, https://doi.org/10.1038/Nmeth.3312 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chavez, A. et al. Comparison of Cas9 activators in multiple species. Nat. Methods 13, 563–567, https://doi.org/10.1038/Nmeth.3871 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Black, J. B. et al. Targeted epigenetic remodeling of endogenous loci by CRISPR/Cas9-based transcriptional activators directly converts fibroblasts to neuronal cells. Cell. Stem Cell. 19, 406–414, https://doi.org/10.1016/j.stem.2016.07.001 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nihongaki, Y. et al. CRISPR-Cas9-based photoactivatable transcription systems to induce neuronal differentiation. Nat. Methods 14, 963–966, https://doi.org/10.1038/nmeth.4430 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Liu, Y. et al. Ascl1 converts dorsal midbrain astrocytes into functional neurons in vivo. J. Neurosci. 35, 9336–9355, doi:https://doi.org/10.1523/Jneurosci.3975-14.2015 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Heinrich, C. et al. Directing astroglia from the cerebral cortex into subtype specific functional neurons. PLoS. Biol. 8, e1000373, https://doi.org/10.1371/journal.pbio.1000373 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Cheng, L. et al. Direct conversion of astrocytes into neuronal cells by drug cocktail. Cell. Res. 25, 1269–1272, https://doi.org/10.1038/cr.2015.120 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Davis, R. L., Weintraub, H. & Lassar, A. B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 51, 987–1000, doi:https://doi.org/10.1016/0092-8674(87)90585-X (1987).

    Article  CAS  PubMed  Google Scholar 

  26. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676, https://doi.org/10.1016/j.cell.2006.07.024 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Mertens, J., Marchetto, M. C., Bardy, C. & Gage, F. H. Evaluating cell reprogramming, differentiation and conversion technologies in neuroscience. Nat. Rev. Neurosci. 17, 424–437, https://doi.org/10.1038/nrn.2016.46 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Hilton, I. B. et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat. Biotechnol. 33, 510–517, https://doi.org/10.1038/nbt.3199 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247.e17, https://doi.org/10.1016/j.cell.2016.08.056 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Farzadfard, F., Perli, S. D. & Lu, T. K. Tunable and multifunctional eukaryotic transcription factors based on CRISPR/Cas. ACS Synth. Biol. 2, 604–613, https://doi.org/10.1021/sb400081r (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Han, K. et al. SHANK3 overexpression causes manic-like behavior with unique pharmacogenetic properties. Nature 503, 72–77, https://doi.org/10.1038/nature12630 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lombardi, L. M., Baker, S. A. & Zoghbi, H. Y. MECP2 disorders: from the clinic to mice and back. J. Clin. Invest. 125, 2914–2923, https://doi.org/10.1172/JCI78167 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Collins, A. L. et al. Mild overexpression of MeCP2 causes a progressive neurological disorder in mice. Hum. Mol. Genet. 13, 2679–2689, https://doi.org/10.1093/hmg/ddh282 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Joung, J. et al. Genome-scale activation screen identifies a lncRNA locus regulating a gene neighborhood. Nature 548, 343–346, https://doi.org/10.1038/nature23451 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Horlbeck, M. A. et al. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation. Elife 5, e19760, https://doi.org/10.7554/eLife.19760 (2016).

    PubMed  PubMed Central  Google Scholar 

  36. Simeonov, D. R. et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115, https://doi.org/10.1038/nature23875 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chow, R. D. et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat. Neurosci. 20, 1329–1341, https://doi.org/10.1038/nn.4620 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ding, S. et al. Efficient transposition of the piggyBac (PB) transposon in mammalian cells and mice. Cell 122, 473–483, https://doi.org/10.1016/j.cell.2005.07.013 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. McCarthy, K. D. & de Vellis, J. Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J. Cell. Biol. 85, 890–902, https://doi.org/10.1083/Jcb.85.3.890 (1980).

    Article  CAS  PubMed  Google Scholar 

  40. Deverman, B. E. et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 34, 204–209, https://doi.org/10.1038/nbt.3440 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, H. et al. Cerebellar modules operate at different frequencies. Elife 3, e02536, https://doi.org/10.7554/eLife.02536 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Liu, F., Song, Y. & Liu, D. Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA.Gene Ther. 6, 1258–1266, https://doi.org/10.1038/sj.gt.3300947 (1999).

    Article  CAS  PubMed  Google Scholar 

  43. Huang, P. et al. Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors. Nature 475, 386–389, https://doi.org/10.1038/nature10116 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Yang, S. et al. MANF regulates hypothalamic control of food intake and body weight. Nat. Commun. 8, 579, https://doi.org/10.1038/s41467-017-00750-x (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lundgaard, I. et al. Direct neuronal glucose uptake heralds activity-dependent increases in cerebral metabolism. Nat. Commun. 6, 6807, https://doi.org/10.1038/ncomms7807 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Greferath, U. et al. Inner retinal change in a novel rd1-FTL mouse model of retinal degeneration. Front. Cell. Neurosci. 9, 293, https://doi.org/10.3389/fncel.2015.00293 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Wang, P. et al. Dual role of Ski in pancreatic cancer cells: tumor-promoting versus metastasis-suppressive function. Carcinogenesis 30, 1497–1506, https://doi.org/10.1093/carcin/bgp154 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. McCracken, J. M. et al. C57BL/6 substrains exhibit different responses to acute carbon tetrachloride exposure: implications for work involving transgenic mice. Gene Expr. 17, 187–205, https://doi.org/10.3727/105221617X695050 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Yao, X. et al. Homology-mediated end joining-based targeted integration using CRISPR/Cas9. Cell. Res. 27, 801–814, https://doi.org/10.1038/cr.2017.76 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ivaniutsin, U., Chen, Y., Mason, J. O., Price, D. J. & Pratt, T. Adenomatous polyposis coli is required for early events in the normal growth and differentiation of the developing cerebral cortex. Neural Dev. 4, 3, https://doi.org/10.1186/1749-8104-4-3 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Ou, Z. et al. The combination of CRISPR/Cas9 and iPSC technologies in the gene therapy of human β-thalassemia in mice. Sci. Rep. 6, 32463, https://doi.org/10.1038/srep32463 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Huang, M. et al. Ptf1a, Lbx1 and Pax2 coordinate glycinergic and peptidergic transmitter phenotypes in dorsal spinal inhibitory neurons. Dev. Biol. 322, 394–405, https://doi.org/10.1016/j.ydbio.2008.06.031 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Schinko, J., Posnien, N., Kittelmann, S., Koniszewski, N. & Bucher, G. Single and double whole-mount in situ hybridization in red flour beetle (Tribolium) embryos. Cold Spring Harb. Protoc. 2009, t5258, https://doi.org/10.1101/pdb.prot5258 (2009).

    Article  Google Scholar 

  54. Ramer, M. S. Anatomical and functional characterization of neuropil in the gracile fasciculus. J. Comp. Neurol. 510, 283–296, https://doi.org/10.1002/cne.21785 (2008).

    Article  PubMed  Google Scholar 

  55. Ting, J. T., Daigle, T. L., Chen, Q. & Feng, G. Acute brain slice methods for adult and aging animals: application of targeted patch clamp analysis and optogenetics. Methods Mol. Biol. 1183, 221–242, https://doi.org/10.1007/978-1-4939-1096-0_14 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Li, C. L. et al. Somatosensory neuron types identified by high-coverage single-cell RNA-sequencing and functional heterogeneity. Cell. Res. 26, 83–102, https://doi.org/10.1038/cr.2015.149 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Li, E. Zuo, Y. Shi, Y. Liu, J. He, J. Pan, Y. Zhong, Y. Lu, Y. Zhang, J. Yang and X. Tang for technical assistance and valuable discussion. This work was supported by National Science and Technology Major Project (2017YFC1001302), CAS Strategic Priority Research Program (XDB02050007, XDA01010409), the MoST863 Program (2015AA020307), NSFC grants (31522037, 31500825, 31571509, 31522038), China Youth Thousand Talents Program (to H.Y.), Break through project of Chinese Academy of Sciences, Shanghai Sailing Plan for the Young Scientific Talents (15YF1414700), and The Ministry of Science and Technology of China (MOST; 2016YFA0100500).

Author information

Authors and Affiliations

Authors

Contributions

H.Z. designed and performed the experiments. J.L. designed and performed the in vivo gene activation in the liver and western blot. C.Z. designed the SPH activation system. N.G., H.L., X.H. and J.Z. cloned the vectors, and performed and analyzed the gene activation experiments in vitro, ex vivo and in vivo. X.S. and L.S. produced AAV8. Z.R., L.C. and F.L. designed and performed the in situ hybridization, ex vivo activation of Ascl1 and Neurog2 in astrocytes and assisted with the in vivo conversion experiments. C.L. and X.Z. performed single-cell RNA sequencing, Y.S. analyzed RNA sequencing and single-cell RNA-sequencing data. H.X. performed electrophysiology. Y.W., X.Y. and W.Y. generated the SPH transgenic mice. C.T. assisted with the immunofluorescence staining of brains and livers. P.H. designed and supervised the experiments of in vivo activation in SPH mice. H.Y. supervised the project and designed experiments. H.Z., P.H. and H.Y. wrote the manuscript.

Corresponding authors

Correspondence to Pengyu Huang or Hui Yang.

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 Design of a fluorescence reporter system.

(a) mCherry reporter is driven by a minimal CMV promoter and activated by an upstream sgRNA target site. Note that miniCMV is a weak promoter, thus the basal fluorescence level is very low. See also18. (b) BFP was co-transfected to control the transfection efficiency (n = 2 cell cultures). Scale bar: 100 μm.

Supplementary Figure 2 Flow cytometry analysis of SPH-based activation.

(a) Schematic of the vectors used to evaluate SPH-based activation. Cre recombinase was used to remove the LSL cassette in the SPH vector. (b) Representative images show that co-transfection of sgRNA-miniCMV, SPH and Cre led to enhanced expression of mCherry (n = 4 cell cultures, 4 images). mCherry was activated in 93 ± 1% of the cells expressing SPH activator system. All values are presented as mean ± s.e.m.

Supplementary Figure 3 SPH shows more potent activation efficiency compared to SAM and VPR in primary astrocytes.

(a) Schematic illustration of all plasmids. The expressions of all functional elements were probed by the fluorescence reporter mCherry or GFP. (b) 2 days after transfection, cells were prepared for FACS. The mCherry and GFP double-positive cells were collected for qPCR. (c) SPH achieved the highest level of activation, which supports the observations in cell lines (n = 2 cell cultures).

Supplementary Figure 4 Characterization of SPH transgenic mice.

(a) Schematic of the integration sites of SPH in a SPH transgenic mouse genome. (b) Genotyping of SPH transgenic mice. “+” indicates positive mice. Percentage of positive progenies was calculated on 101 mice. Gel image was cropped. (c) Systemic evaluation of the Alb-Cre effectiveness in cleaving LSL cassettes specifically in the liver. Note that Alb is a liver-specific promoter (n = 1 mouse). Gel image was cropped. (d) Fluorescence images of the primary SPH fibroblasts infected by lentivirus containing sgRNA-EF1a-Cre-mCherry, confirming expression of dCas9 and activators were induced by Cre recombinase (n = 3 mice). (e) Expressions of dCas9 in Cre-expressing fibroblasts. Gel image was cropped. (f) Characterization of Cre effectiveness in the liver after hydrodynamic injection of the plasmids expressing Cre and mCherry (n = 1 mouse). (g) Stereotactic Injection of AAV-hSyn-mCherry-Cre to induce neuron-specific expression of EGFP (n = 1 mouse). Red arrowheads indicate primers. Scale Bars: d, f, g, 20 μm.

Supplementary Figure 5 Targeted activation of individual genes and lncRNA in the liver through tail vein injection of plasmids.

(a) Experimental design. Plasmids with Cre, sgRNA and mCherry expression were delivered into the liver by hydrodynamic injection via tail vein. Note that mRNA was extracted from the liver without isolating mCherry-positive cells and used for qPCR analysis (n = 1 mouse). Scale Bar: 100 μm. (b) Targeted activation of individual genes in the liver after injection of plasmids expressing indicated sgRNAs (number of mice denoted within graph). Increased amount of ASCL1 protein was confirmed by western blot (n = 1 mouse). Gel images were cropped. (c) Induction of lncRNAs in the liver (n = 3 mice). (d) Activation of multiple genes in the liver through tail vein injection of a mixture of sgRNA-expressing vectors (n = 2 mice). The sgLacZ serves as a control sgRNA.

Supplementary Figure 6 Remodeling of metabolic liver zonation by activating Dkk1 in vivo.

(a) Schematic of the plasmid used for sgDkk1 expression. (b,c) SPH mice were hyperdynamically injected with sgRNA-mCherry plasmids. Immunofluorescence staining were performed 4–6 days after plasmids injections on liver sections. mCherry (red) was stained to indicate cells receiving plasmids. Both GS and CYP2E1 (cyan) were β-catenin target genes locating inpericentral area of the liver lobule. Arrows indicate mCherry-positive cells showing downregulation of GS and CYP2E1. Note that expression of GS and CYP2E1 in pericentral hepatocytes receiving sgDkk1-Cre-mCherry was inhibited. Scale bar = 50 μm. (d) Increased Dkk1 transcripts were quantified by qPCR (n = 5 mice). (e, f) Percentage of cells showing suppressed expression of GS (88/439 VS 0/165, sgLacZ: n = 2 mice, sgDkk1: n = 2 mice) or CYP2E1 (127/829 VS 0/66, sgLacZ: n = 2 mice, sgDkk1: n = 5 mice) in plasmid-expressing hepatocytes. (g) Schematic illustration of DKK1-mediated suppression of Wnt/β-catenin signaling. Activation of Dkk1 leads to reduced expressions of two β-catenin target genes; green circles indicate mCherry-positive cells showing downregulation of GS and CYP2E1. The sgLacZ serves as a control sgRNA.

Supplementary Figure 7 Prescreen of the effective sgRNAs.

(a) Prescreening in primary fibroblasts derived from SPH mice (n = 2 cell cultures) and (b) N2a cells (n = 1 cell culture). The sgRNAs used in the sgRNA arrays are denoted as red. The sgLacZ serves as a control sgRNA.

Supplementary Figure 8 Simultaneous activation of 8 genes and 2 lncRNAs in mCherry-positive cells via hydrodynamic injection of all-in-one plasmids.

(a) Schematic of the all-in-one vector used for sgRNAs and Cre expression, and experimental flow of gene activations in the liver of SPH transgenic mice. Note that mRNA was extracted from mCherry-positive cells. (b) Simultaneous activation of 8 genes and 2 lncRNAs in the liver (n = 3 mice). The sgLacZ serves as a control sgRNA.

Supplementary Figure 9 Simultaneous activation of 10 genetic elements including 8 genes and 2 lncRNAs in the liver via hydrodynamic injection of dual-AAV vectors.

(a) Experimental flow of gene activations in the liver of SPH transgenic mice. (b) Schematic of the AAV-10-sgRNA-array and AAV-Alb-Cre. (c) Simultaneous activation of 8 genes and 2 lncRNAs (n = 2 mice). SPH mouse injected with vehicle serves as a control.

Supplementary Figure 10 Simultaneous activation of multiple genes in single-cell level.

Primary astrocytes derived from the SPH mice were transfected with an all-in-one vector expressing Cre, mCherry and an sgRNA array targeting the promoters of 8 genes. Due to detection limit, 3 (Ascl1, Slc6a4 and Slc7a11) out of 8 genes (Neurog2, Slc6a4, Dkk1, Hbb, Slc7a11, IL10, Ascl1, Neurod1) were detected in single-cell RNA-seq data. The average expression levels (FPKM values) of Ascl1, Slc6a4 and Slc7a11 were normalized to control group (n = 3 cells). 6 cells receiving the all-in-one vector were analyzed. Control cells were introduced with sgLacZ constructs.

Supplementary Figure 11 Enhance transcription of Hbb and Dkk1 was confirmed by in situ hybridization.

Enhance transcription of Hbb and Dkk1 was detected in the cerebral cortex after injecting AAV-hSyn-Cre and AAV-sgDkk1-sgHbb (n = 1 mouse). Scale Bar: 20 μm. The contralateral cerebral cortex serves as control.

Supplementary Figure 12 Full scan of all gels.

a, for Supplementary Figure 4b. b, for Supplementary Figure 4c. c, for Supplementary Figure 4e. d, for Supplementary Figure 5b. e, for Figure 4e, GAPDH. f, for Figure 4e, DKK1. g, for Figure 4e, HBB.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, H., Liu, J., Zhou, C. et al. In vivo simultaneous transcriptional activation of multiple genes in the brain using CRISPR–dCas9-activator transgenic mice. Nat Neurosci 21, 440–446 (2018). https://doi.org/10.1038/s41593-017-0060-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-017-0060-6

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