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:

Multiplexed genome regulation in vivo with hyper-efficient Cas12a

Abstract

Multiplexed modulation of endogenous genes is crucial for sophisticated gene therapy and cell engineering. CRISPR–Cas12a systems enable versatile multiple-genomic-loci targeting by processing numerous CRISPR RNAs (crRNAs) from a single transcript; however, their low efficiency has hindered in vivo applications. Through structure-guided protein engineering, we developed a hyper-efficient Lachnospiraceae bacterium Cas12a variant, termed hyperCas12a, with its catalytically dead version hyperdCas12a showing significantly enhanced efficacy for gene activation, particularly at low concentrations of crRNA. We demonstrate that hyperdCas12a has comparable off-target effects compared with the wild-type system and exhibits enhanced activity for gene editing and repression. Delivery of the hyperdCas12a activator and a single crRNA array simultaneously activating the endogenous Oct4, Sox2 and Klf4 genes in the retina of post-natal mice alters the differentiation of retinal progenitor cells. The hyperCas12a system offers a versatile in vivo tool for a broad range of gene-modulation and gene-therapy applications.

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: Development of combinatorial dCas12a mutants with superior activity under conditions of low crRNA levels.
Fig. 2: HyperCas12a outperforms WT dCas12 in CRISPR repression and in vivo gene editing.
Fig. 3: HyperdCas12a enables multiplex activation of endogenous genes.
Fig. 4: In vivo gene activation by hyperdCas12a with single crRNA and poly-crRNA arrays.
Fig. 5: In vivo multiplex gene activation by hyperdCas12a compared with dCas12a alternatives.
Fig. 6: Multiplex gene activation by hyperdCas12a induces the migration of retina progenitor cells and altered differentiation in vivo.

Similar content being viewed by others

Data availability

Whole-transcriptome sequencing data can be accessed in Gene Expression Omnibus under the accession code GSE166817. Key constructs and plasmids will be available on Addgene (https://www.addgene.org/Stanley_Qi/). All other data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

The gene-level FPKM values were calculated using a custom Python script available at https://github.com/QilabGitHub/FPKMcalculation. The semi-fluorescence intensities of individual cells (microscopy) were quantitated with a semi-automated image analysis pipeline based on MATLAB (version R2019a) available at https://github.com/QilabGitHub/dCas12a-microscopy.

References

  1. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Doudna, J. A. & Charpentier, E. The new frontier of genome engineering with CRISPR–Cas9. Science 346, 1258096 (2014).

    Article  PubMed  CAS  Google Scholar 

  3. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR–Cas system. Cell 163, 759–771 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zetsche, B. et al. Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nat. Biotechnol. 35, 31–34 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Kleinstiver, B. P. et al. Engineered CRISPR–Cas12a variants with increased activities and improved targeting ranges for gene, epigenetic and base editing. Nat. Biotechnol. 37, 276–282 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Campa, C. C., Weisbach, N. R., Santinha, A. J., Incarnato, D. & Platt, R. J. Multiplexed genome engineering by Cas12a and CRISPR arrays encoded on single transcripts. Nat. Methods 16, 887–893 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Fonfara, I., Richter, H., Bratovi, M., Le Rhun, Ä. & Charpentier, E. The CRISPR-associated DNA-cleaving enzyme Cpf1 also processes precursor CRISPR RNA. Nature 532, 517–521 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Gier, R. A. et al. High-performance CRISPR–Cas12a genome editing for combinatorial genetic screening. Nat. Commun. 11, 3455 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kim, D. et al. Genome-wide analysis reveals specificities of Cpf1 endonucleases in human cells. Nat. Biotechnol. 34, 863–868 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Bin Moon, S. et al. Highly efficient genome editing by CRISPR–Cpf1 using CRISPR RNA with a uridinylate-rich 3′-overhang. Nat. Commun. 9, 3651 (2018).

    Article  CAS  Google Scholar 

  11. Li, F. et al. Comparison of CRISPR/Cas endonucleases for in vivo retinal gene editing. Front. Cell. Neurosci. 14, 570917 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tak, Y. E. et al. Inducible and multiplex gene regulation using CRISPR-Cpf1-based transcription factors. Nat. Methods 14, 1163–1166 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ling, X. et al. Improving the efficiency of CRISPR-Cas12a-based genome editing with site-specific covalent Cas12a-crRNA conjugates. Mol Cell 81, 4747–4756 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Zhang, L. et al. AsCas12a ultra nuclease facilitates the rapid generation of therapeutic cell medicines. Nat. Commun. 12, 3908 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jones, S. K. et al. Massively parallel kinetic profiling of natural and engineered CRISPR nucleases. Nat. Biotechnol. 39, 84–93 (2021).

    Article  PubMed  CAS  Google Scholar 

  16. Liu, P. et al. Enhanced Cas12a editing in mammalian cells and zebrafish. Nucleic Acids Res. 47, 4169–4180 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kocak, D. D. et al. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nat. Biotechnol. 37, 657–666 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nguyen, L. T., Smith, B. M. & Jain, P. K. Enhancement of trans-cleavage activity of Cas12a with engineered crRNA enables amplified nucleic acid detection. Nat. Commun. 11, 4906 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wang, D., Zhang, F. & Gao, G. CRISPR-based therapeutic genome editing: strategies and in vivo delivery by AAV vectors. Cell 181, 136–150 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhang, Y. et al. Enhanced CRISPR–Cas9 correction of Duchenne muscular dystrophy in mice by a self-complementary AAV delivery system. Sci. Adv. 6, eaay6812 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yamano, T. et al. Structural basis for the canonical and non-canonical PAM recognition by CRISPR–Cpf1. Mol. Cell 67, 633–645 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gao, L. et al. Engineered Cpf1 variants with altered PAM specificities. Nat. Biotechnol. 35, 789–792 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kempton, H. R. et al. Multiple input sensing and signal integration using a split Cas12a system. Mol. Cell 78, 184–191 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Vora, S. et al. Rational design of a compact CRISPR–Cas9 activator for AAV-mediated delivery. Preprint at bioRxiv https://doi.org/10.1101/298620 (2018).

  25. Wang, Q. et al. Mouse γ-synuclein promoter-mediated gene expression and editing in mammalian retinal ganglion cells. J. Neurosci. 40, 3896–3914 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Levy, J. M. et al. Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nat. Biomed. Eng. 4, 97–110 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Feng, G. et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron 28, 41–51 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Kleinstiver, B. P. et al. Genome-wide specificities of CRISPR–Cas Cpf1 nucleases in human cells. Nat. Biotechnol. 34, 869–874 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liu, Y. et al. CRISPR activation screens systematically identify factors that drive neuronal fate and reprogramming. Cell Stem Cell 23, 758–771 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Liu, P., Chen, M., Liu, Y., Qi, L. S. & Ding, S. CRISPR-based chromatin remodeling of the endogenous Oct4 or Sox2 locus enables reprogramming to pluripotency. Cell Stem Cell 22, 252–261 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gonatopoulos-Pournatzis, T. et al. Genetic interaction mapping and exon-resolution functional genomics with a hybrid Cas9–Cas12a platform. Nat. Biotechnol. 38, 638–648 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Breinig, M. et al. Multiplexed orthogonal genome editing and transcriptional activation by Cas12a. Nat. Methods 16, 51–54 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. Matsuda, T. & Cepko, C. L. Controlled expression of transgenes introduced by in vivo electroporation. Proc. Natl Acad. Sci. USA 104, 1027–1032 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang, S., Sengel, C., Emerson, M. M. & Cepko, C. L. A gene regulatory network controls the binary fate decision of rod and bipolar cells in the vertebrate retina. Dev. Cell 30, 513–527 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Chan, C. S. Y. et al. Cell type- and stage-specific expression of Otx2 is regulated by multiple transcription factors and cis-regulatory modules in the retina. Development 147, dev187922 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rocha-Martins, M. et al. De novo genesis of retinal ganglion cells by targeted expression of Klf4 in vivo. Development 146, dev176586 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. Sharma, P. et al. Oct4 mediates Müller glia reprogramming and cell cycle exit during retina regeneration in zebrafish. Life Sci. Alliance 2, e201900548 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Lin, Y. P., Ouchi, Y., Satoh, S. & Watanabe, S. Sox2 plays a role in the induction of amacrine and Müller glial cells in mouse retinal progenitor cells. Investig. Ophthalmol. Vis. Sci. 50, 68–74 (2009).

    Article  Google Scholar 

  41. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Venkatesh, A., Ma, S., Langellotto, F., Gao, G. & Punzo, C. Retinal gene delivery by rAAV and DNA electroporation. Curr. Protoc. Microbiol. https://doi.org/10.1002/9780471729259.mc14d04s28 (2013).

  43. Matharu, N. et al. CRISPR-mediated activation of a promoter or enhancer rescues obesity caused by haploinsufficiency. Science 363, eaau0629 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Liao, C. et al. Modular one-pot assembly of CRISPR arrays enables library generation and reveals factors influencing crRNA biogenesis. Nat. Commun. 10, 2948 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Magnusson, J. P. et al. Enhanced Cas12a multi-gene regulation using a CRISPR array separator. eLife 10:e66406 (2021).

  46. 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 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nuñez, J. K. et al. Genome-wide programmable transcriptional memory by CRISPR-based epigenome editing. Cell 184, 2503–2519 (2021).

    Article  PubMed  CAS  Google Scholar 

  48. Nakamura, M., Ivec, A. E., Gao, Y. & Qi, L. S. Durable CRISPR-based epigenetic silencing. BioDesign Res. 2021, 981582 (2021).

  49. Kemaladewi, D. U. et al. A mutation-independent approach for muscular dystrophy via upregulation of a modifier gene. Nature 572, 125–130 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Xu, X. et al. Engineered miniature CRISPR–Cas system for mammalian genome regulation and editing. Mol. Cell. 81, 4333–4345 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Guo, L. Y. et al. Multiplex CRISPR genome regulation in mouse retina with hyper-efficient Cas12a. Protoc. Exch. https://doi.org/10.21203/rs.3.pex-1811/v1 (2022).

  52. Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 37, 224–226 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  54. Anders, S., Pyl, P. T. & Huber, W. HTSeq-A Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Kwong, J. M. K., Caprioli, J. & Piri, N. RNA binding protein with multiple splicing: a new marker for retinal ganglion cells. Investig. Ophthalmol. Vis. Sci. 51, 1052–1058 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

L.Y.G. acknowledges support from a Stanford’s National Eye Institute (NEI) T32 Vision Training Grant (grant no. 5T32EY020485), the Knights Templar Eye Foundation (KTEF) and the VitreoRetinal Surgery Foundation (VRSF). L.S.Q. acknowledges support from the Li Ka Shing Foundation, National Science Foundation CAREER award (award no. 2046650), National Institutes of Health (grant no. 1U01DK127405), Chan Zuckerberg Initiative Neurodegeneration Challenge Network, Stanford Maternal and Child Health Research Institute (MCHRI) through the Uytengsu–Hamilton 22q11 Neuropsychiatry Research Award Program, Stanford–Coulter Translational Research Grant, and California Institute for Regenerative Medicine (CIRM; grant no. DISC2-12669). S.W. is supported by funding from the American Diabetes Association (grant no. 1-16-INI-16), NIH grant no. 1R01EY03258501, NIH grant no. R01NS109990, NIH-NEI grant no. P30-EY026877 and Research to Prevent Blindness, Inc. BioRender.com was used to generate illustrations in Fig. 1b, 1g, 2a, 2c, 2f, 3a, 4a and 6e. The members of the Qi, Wang and Hu laboratories provided valuable discussion and support for this project.

Author information

Authors and Affiliations

Authors

Contributions

L.Y.G. and L.S.Q. conceived the idea for this study. L.Y.G., J.B., S.W. and L.S.Q. designed the experiments. L.Y.G., J.B., X.Z., H.R.K., B.G., D.A.R. and R.M.J. performed the ex vivo experiments. L.Y.G., J.B., A.E.D. and P.L. performed the in vivo experiments with guidance from S.W. and Y.H. L.Y.G., J.B., S.W. and L.S.Q. analysed the experimental data. A.C. and X.L. performed the computational analyses of the sequencing data, and X.Z. and R.M.J. analysed the imaging data. X.X. provided reagents. L.Y.G. and L.S.Q. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Lucie Y. Guo, Sui Wang or Lei S. Qi.

Ethics declarations

Competing interests

The authors have filed a provisional patent via Stanford University related to the work (US patent no. 63/148,652). L.S.Q. is a founder and scientific advisory board member of Epicrispr Biotechnologies.

Peer review

Peer review information

Nature Cell Biology thanks Stephen H. Tsang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Gating strategies for flow cytometry.

a, Standard strategy for gating cells based on forward scatter (FSC) and side scatter (SSA) (left), then further gating for singlets based on FSC-height (FSC-H) and FSC-area (FSC-A) (right). To analyse transfected cells, further gating is applied to the singlet population based on fluorescence intensity. Please note that for Fig. 1c-e, and Extended Data Fig. 2a–j, that gating strategy is included within the figure. b, Gating strategy for experiments with Cas12a-mCherry plasmid and a CAG-crRNA plasmid (without fluorophore), thus mCherry+ cells are used for analysis. c, Gating strategy for Fig. 1g, in which 3 plasmids are co-transfected. d, Gating strategy for some experiments with Cas12a-mCherry plasmid and U6-crRNA (with BFP). e, Mean BFP fluorescence across the mutants tested in Fig. 1c. f, Mean mCherry fluorescence among mutants tested in Fig. 1c. In e-f, each data point represents the mean GFP intensity of an independent experiment, with each bar representing the average of 2 or more independent experiments. g, Schematic of the LbCas12a protein domains and location of four of the most potent point mutants, with alignment across various Cas12 species. The relevant Asp (D) or Glu(E) residues are highlighted in red.

Source data

Extended Data Fig. 2 Optimizing the nuclear localizing signal and comparing to enAsdCas12a.

a. Schematic to test two different nuclear localization signals. Constructs containing either 2×SV40 or 2×Myc NLS fused with WT dCas12a are co-transfected with Tet crRNA in TRE3-GFP HEK293T reporter cells. b, Representative flow cytometry histogram of BFP intensity, showing threshold for BFP + cells, and subset of ‘low BFP + ’ cells (similar to Fig. 1d). c-d, GFP fluorescence in BFP + (c) or ‘low BFP + ’ cells (d). e-g, GFP fluorescence in BFP + (f) or low BFP + cells (g) to compare WT versus hyperdCas12a (hyp) with 2×Myc NLS, as well as BFP and mCherry average fluorescence in each gated BFP group. h-i, Alignment of the structure of LbCas12a versus AsCas12a proteins (h) and alignment of peptide sequences (i) encompassing mutations harboured by enAsCas12a, a reported enhanced variant of Cas12a from Acidaminococcus with the E174R/S542R/K548R mutations5 corresponding to homologous residues (D156R/G532R/K538R) mutations in LbCas12a. j, Comparison of variants containing mutations of homologous residues in LbCas12a in ‘low BFP + ’ cells. Interestingly, D156R combined with G532R and/or K538R did not achieve activation higher than the single D156R mutant, in contrast to results with homologous residues in AsCas12a5. k, Comparison of hyperdCas12a versus enAsdCas12a with a single crRNA driven by U6 promoter in 1:1 versus 1:0.2 ratio of dCas12:crRNA, in TRE3G-GFP HEK293T cells. l, Comparison of hyperdCas12a versus enAsdCas12a with single crRNA driven by CAG promoter flanked by direct repeats (DR) specific to LbCas12a versus AsCas12a. m, Comparison of hyperdCas12a versus enAsdCas12a with dual crRNAs containing crTet on the second position and non-targeting crLacZ on the first position flanked by As or Lb direct repeats (DR). All transfections in this figure were carried out in TRE3G-GFP HEK293T reporter cells. Bar graph in f, g and k-m shows the mean of n ≥ 3 independent experiments; bar graph in j shows the mean of n ≥ 2 independent experiments; each data point represents value of an independent experiment.

Source data

Extended Data Fig. 3 Improved gene editing by hyperCas12a.

a. Nuclease-active WT Cas12a versus hyperCas12a were co-transfected with crGFP into HEK293T cells stably expressing GFP driven by SV40 promoter. b. Representative flow cytometry histogram showing threshold for mCherry+ cells. Analysis of mCherry+ cells are shown in Fig. 2d, while bulk cells (without sorting) were used for indel analysis (panels c-d). c. Indel activity at each nucleotide position, shown as percentage of total reads with a deletion at the position. The PAM is highlighted in pink. d, Indel patterns and corresponding ratios in total reads detected by deep sequencing as analysed by CRISPResso252.

Extended Data Fig. 4 Characterization of off-target effects of hyperdCas12a.

a. RNA-seq FPKMs (fragments per kilobase million fragments mapped) are plotted as transfected versus non-transfected cells. The transfected samples are TRE3G-GFP HEK293T reporter cells co-transfected with WT dCas12a or hyperdCas12a, and with non-targeting crRNA or crRNA targeting TRE3G. b. RNA-seq plots showing FKPM (Fragments Per Kilobase Million) between two biological duplicates for each condition. The calculated Pearson correlation coefficient for each condition is shown on the graph.

Extended Data Fig. 5 Dual antibiotic selection for co-transfected mouse P19 cells.

a, Mouse P19 cells were co-transfected with constructs expressing puromycin resistance (PuroR) and hygromycin resistance (HygroR), then selected with puromycin and hygromycin at 24 hr after transfection. Cells were collected for analysis 72 hr after transfection. b, Histograms showing percentage of BFP + (crRNA) and mCherry + (dCas12a) cells for non-transfected, non-selected, and Puro/Hygro-selected cells. c, Flow cytometry plots. Data in panels b-c are representative plots of n = 3 independent experiments.

Extended Data Fig. 6 Screening dCas12a crRNAs for activating endogenous Sox2.

a, Schematic of dCas12a crRNAs (red) targeting promoter of Sox2, and their relative positions to validated dCas9 sgRNAs31 that are functional (black) for activating Sox2. Arrows indicate sense or antisense binding of crRNAs/sgRNAs to target DNA. The genomic position of the first ‘T’ in PAM (relative to TSS, which is ‘0’) are shown for each crRNA targeting the Sox2 promoter. b, Immunostaining of Sox2 expression from activation by WT dCas12a–miniVPR with various Sox2 single crRNAs, compared to activation by dCas9-miniVPR (using a validated sgRNA, S84)31. Scale bar, 100 µm. c-d, Immunostaining of Sox2 expression and co-localization with BFP and mCherry for a pair of crRNAs (c) and a panel of ‘triplet’ crRNAs (d), demonstrating additive or synergistic effect when multiple crRNAs are used in tandem. Interestingly, addition of a third crRNA targeting a region between the paired crRNAs S1 and S2 decreases the level of activation. Inset (c) shows brightfield image to demonstrate nuclear localization of mCherry (hyperdCas12a) and target (Sox2), since BFP on crRNA plasmid precludes the use of an additional nuclear dye. White scale bar, 100 µm; yellow scale bar (within inset), 50 µm e-f. Automated quantitation of images in panels b-d. In panel e, 350-2000 cells for each condition were quantitated for one screening experiment with multiple fields of view. The exact number of cells for each condition is listed in the Source Data for Extended Data Fig. 6. NT, non-targeting crRNA. In panel f, 70-250 cells for each condition were quantitated for one screening experiment with multiple fields of view. For box-and-whisker plots, the box shows 25-75% (with bar at median, dot at mean), and whiskers encompass 10-90%, with individual data points shown for the lowest and highest 10% of each dataset. The exact number of cells for each condition are listed in the Source Data for Extended Data Fig. 6. NT, non-targeting crRNA.

Source data

Extended Data Fig. 7 Screening dCas12a crRNAs for activating endogenous Klf4.

a, Schematic of dCas12a crRNAs (red) targeting promoter of Klf4 and their relative positions to known dCas9 sgRNAs31 that are functional (black) or non-functional (grey) for activating Klf4. Arrows indicate sense or antisense binding of crRNAs/sgRNAs to the target DNA. The genomic position of the first ‘T’ in PAM (relative to TSS, which is ‘0’) are shown for each crRNA targeting to the Klf4 promoter. b, Immunostaining of Klf4. Inset shows brightfield image to demonstrate nuclear localization of mCherry (hyperdCas12a) and target (Klf4), since BFP on crRNA plasmid precludes the use of an additional nuclear dye. White scale bar, 100 µm; yellow scale bar (within inset), 50 µm c. Automated quantitation of images in panel b, where 200-600 cells for each condition were quantitated for one screening experiment with multiple fields of view. The exact number of cells for each condition is listed in the Source Data for Extended Data Fig. 7. NT, non-targeting crRNA.

Source data

Extended Data Fig. 8 Screening dCas12a crRNAs for activating endogenous Oct4.

a, Schematic of dCas12a crRNAs (red) targeting promoters of Oct4 and their relative positions to known dCas9 sgRNAs31 that are functional (black) or non-functional (grey) for activating Oct4. Arrows indicate sense or antisense binding of crRNAs/sgRNAs to the target DNA. The genomic position of the first ‘T’ in PAM (relative to TSS, which is ‘0’) are shown for each crRNA targeting to the Oct4 promoter. b, Immunostaining of Oct4. White scale bar, 100 µm. c, Inset shows merge with brightfield to demonstrate nuclear localization of mCherry (hyperdCas12a) and target (Oct4), since BFP on crRNA plasmid precludes the use of an additional nuclear dye. Yellow scale bar, 50 µm. d. Quantification of panel b, where 100-600 cells for each condition were quantitated over one screening experiment with multiple fields of view. The exact number of cells for each condition are listed in the Source Data for Extended Data Fig. 8. e. Immunostaining of Oct4 after activation by paired crRNA consisting of the two most potent crRNAs (O1 + O2), which shows lack of additive effect. White scale bar, 100 µm. f. Quantification in panels e, where 200-700 cells for each condition were quantitated over one screening experiment with multiple fields of view. For box-and-whisker plots, the box shows 25-75% (with bar at median, dot at mean), and whiskers encompass 10-90%, with individual data points shown for the lowest and highest 10% of each dataset. The exact number of cells for each condition is listed in the Source Data for Extended Data Fig. 8.

Source data

Extended Data Fig. 9 Enhanced multiplex activation by hyperdCas12a.

Multiplex endogenous gene activation by hyperdCas12a versus enAsdCas12a and 6-crRNA array targeting Oct4, Sox2 and Klf4 (OSK) in mouse P19 cells as measured by qPCR, in similar experiment as described in Fig. 3a. Each data point shows one independent measurement, and each bar shows the average of n = 3 independent experiments. DR, direct repeat; NT, non-targeting.

Source data

Extended Data Fig. 10 In vivo single crRNA activation by hyperdCas12a.

a-c, Constructs containing hyperdCas12a and single crRNA to Sox2 (a), Klf4 (b) or Oct4 (c) for in vivo electroporation in post-natal mouse retina and representative immunofluorescence images. CAG-GFP is used to mark the electroporated patch. Scale bar, 50 µm. Magnified views of the regions in the yellow boxes are shown in Fig. 6a. d, Quantification of percentages of Oct4+, Sox2+ or Klf4+ cells among HA + cells in INL. Bar graph shows the mean of 3 independent experiments, and each data point represents value of an independent experiment. e, Immunofluorescence images of in vivo electroporation in mouse retina with hyperdCas12a with non-targeting LacZ crRNA. Scale bar, 50 µm.

Source data

Supplementary information

Reporting Summary

Supplementary Tables 1–4

Supplementary Table 1. crRNA sequences. Supplementary Table 2. Plasmids used in each figure. Supplementary Table 3. Primer sequences. Supplementary Table 4. All genes from RNA-seq.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, L.Y., Bian, J., Davis, A.E. et al. Multiplexed genome regulation in vivo with hyper-efficient Cas12a. Nat Cell Biol 24, 590–600 (2022). https://doi.org/10.1038/s41556-022-00870-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00870-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research