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:

Scalable biological signal recording in mammalian cells using Cas12a base editors

Abstract

Biological signal recording enables the study of molecular inputs experienced throughout cellular history. However, current methods are limited in their ability to scale up beyond a single signal in mammalian contexts. Here, we develop an approach using a hyper-efficient dCas12a base editor for multi-signal parallel recording in human cells. We link signals of interest to expression of guide RNAs to catalyze specific nucleotide conversions as a permanent record, enabled by Cas12’s guide-processing abilities. We show this approach is plug-and-play with diverse biologically relevant inputs and extend it for more sophisticated applications, including recording of time-delimited events and history of chimeric antigen receptor T cells’ antigen exposure. We also demonstrate efficient recording of up to four signals in parallel on an endogenous safe-harbor locus. This work provides a versatile platform for scalable recording of signals of interest for a variety of biological applications.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Base editing with hyperCas12a as a platform for signal recording.
Fig. 2: Modular recording of diverse biological signals.
Fig. 3: Recording of analog signal characteristics.
Fig. 4: Temporal resolution of cellular history within recording window.
Fig. 5: Dual-function effector and recorder cells.
Fig. 6: Simultaneous recording of multiple signals in parallel.

Similar content being viewed by others

Data availability

All data supporting main figures and extended data figures have been included in source data files. Plasmids are available from Addgene with the following accession codes: 183098, 183203, 183204, 183205, 183206, 183207, 183623, 183624, 183625, 183626, 183627, 183628 and 183629. Raw deep sequencing data are available at National Center for Biotechnology Information Bioproject PRJNA818698. No custom code was used in this study. Source data are provided with this paper.

References

  1. Burrill, D. R. & Silver, P. A. Making cellular memories. Cell 140, 13–18 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sheth, R. U. & Wang, H. H. DNA-based memory devices for recording cellular events. Nat. Rev. Genet. 19, 718–732 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Farzadfard, F. & Lu, T. K. Emerging applications for DNA writers and molecular recorders. Science 361, 870–875 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kotula, J. W. et al. Programmable bacteria detect and record an environmental signal in the mammalian gut. Proc. Natl Acad. Sci. USA 111, 4838–4843 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burrill, D. R., Inniss, M. C., Boyle, P. M. & Silver, P. A. Synthetic memory circuits for tracking human cell fate. Genes Dev. 26, 1486–1497 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ajo-Franklin, C. M. et al. Rational design of memory in eukaryotic cells. Genes Dev. 21, 2271–2276 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bonnet, J., Subsoontorn, P. & Endy, D. Rewritable digital data storage in live cells via engineered control of recombination directionality. Proc. Natl Acad. Sci. USA 109, 8884–8889 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Farzadfard, F. & Lu, T. K. Synthetic biology. Genomically encoded analog memory with precise in vivo DNA writing in living cell populations. Science 346, 1256272 (2014).

  9. Yang, L. et al. Permanent genetic memory with >1-byte capacity. Nat. Methods 11, 1261–1266 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Siuti, P., Yazbek, J. & Lu, T. Synthetic circuits integrating logic and memory in living cells. Nat. Biotechnol. 31, 448–452 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Shipman, S. L., Nivala, J., Macklis, J. D. & Church, G. M. Molecular recordings by directed CRISPR spacer acquisition. Science 353, aaf1175 (2016).

  12. Sheth, R. U., Yim, S. S., Wu, F. L. & Wang, H. H. Multiplex recording of cellular events over time on CRISPR biological tape. Science 358, 1457–1461 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Schmidt, F., Cherepkova, M. Y. & Platt, R. J. Transcriptional recording by CRISPR spacer acquisition from RNA. Nature 562, 380–385 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Perli, S. D., Cui, C. H. & Lu, T. K. Continuous genetic recording with self-targeting CRISPR-Cas in human cells. Science 353, aag0511 (2016).

    Article  PubMed  Google Scholar 

  15. Tang, W. & Liu, D. R. Rewritable multi-event analog recording in bacterial and mammalian cells. Science 360, eaap8992 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Farzadfard, F. et al. Single-nucleotide-resolution computing and memory in living cells. Mol. Cell 75, 769–780.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Loveless, T. B. et al. Lineage tracing and analog recording in mammalian cells by single-site DNA writing. Nat. Chem. Biol. https://doi.org/10.1038/s41589-021-00769-8 (2021).

  18. Park, J. et al. ll Recording of elapsed time and temporal information about biological events using Cas9. Cell 184, 1047–1063 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. Zetsche, B. et al. Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nat. Biotechnol. 35, 31–34 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Zhong, G., Wang, H., Li, Y., Tran, M. H. & Farzan, M. Cpf1 proteins excise CRISPR RNAs from mRNA transcripts in mammalian cells. Nat. Chem. Biol. 13, 839–841 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kempton, H., Goudy, L., Love, K. & Qi, L. Multiple input sensing and signal integration using a split Cas12a system. Mol. Cell 78, 184–191.e3 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. 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 https://doi.org/10.1038/s41592-019-0508-6 (2019).

  25. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, X. et al. Cas12a base editors induce efficient and specific editing with low DNA damage response. Cell Rep. 31, 107723 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Stadtmauer, E. A. et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020).

  29. Li, X. et al. Base editing with a Cpf1–cytidine deaminase fusion. Nat. Biotechnol. 36, 324–327 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Richter, M. F. et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat. Biotechnol. https://doi.org/10.1038/s41587-020-0453-z (2020).

  32. 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 

  33. Guo, L. et al. Multiplexed genome regulation in vivo with hyper-efficient Cas12a. Nat. Cell Biol. 24, 590–600 (2022).

  34. Huang, T. P., Newby, G. A. & Liu, D. R. Precision genome editing using cytosine and adenine base editors in mammalian cells. Nat. Protoc. https://doi.org/10.1038/s41596-020-00450-9 (2021).

  35. 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 

  36. Nissim, L., Perli, S. D., Fridkin, A., Perez-Pinera, P. & Lu, T. K. Multiplexed and programmable regulation of gene networks with an integrated RNA and CRISPR/Cas toolkit in human cells. Mol. Cell 54, 698–710 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Taniguchi, K. & Karin, M. NF-κB, inflammation, immunity and cancer: coming of age. Nat. Rev. Immunol. 18, 309–324 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Ede, C., Chen, X., Lin, M.-Y. & Chen, Y. Y. Quantitative analyses of core promoters enable precise engineering of regulated gene expression in mammalian cells. ACS Synth. Biol. 5, 395–404 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Carlezon, W., Duman, R. & Nestler, E. The many faces of CREB. Trends Neurosci. 28, 436–445 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Crabtree, G. R. & Olson, E. N. NFAT signaling: choreographing the social lives of cells. Cell 109, S67–S79 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. Shaulian, E. & Karin, M. AP-1 in cell proliferation and survival. Oncogene 20, 2390–2400 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Nguyen, T., Sherratt, P. J. & Pickett, C. B. Regulatory mechanisms controlling gene expression mediated by the antioxidant response element. Annu. Rev. Pharmacol. Toxicol. 43, 233–260 (2003).

  43. Morsut, L. et al. Engineering customized cell sensing and response behaviors using synthetic notch receptors. Cell https://doi.org/10.1016/j.cell.2016.01.012 (2016).

  44. Lim, W. A. & June, C. H. The principles of engineering immune cells to treat cancer. Cell 168, 724–740 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Schukur, L., Geering, B., Charpin-El Hamri, G. & Fussenegger, M. Implantable synthetic cytokine converter cells with AND-gate logic treat experimental psoriasis. Sci. Transl. Med. 7, 318ra201 (2015).

    Article  PubMed  Google Scholar 

  46. Smole, A., Lainšček, D., Bezeljak, U., Horvat, S. & Jerala, R. A synthetic mammalian therapeutic gene circuit for sensing and suppressing inflammation. Mol. Ther. 25, 102–119 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Monaco, C., Nanchahal, J., Taylor, P. & Feldmann, M. Anti-TNF therapy: past, present and future. Int. Immunol. 27, 55–62 (2015).

  48. Beirnaert, E. et al. Bivalent llama single-domain antibody fragments against tumor necrosis factor have picomolar potencies due to intramolecular interactions. Front. Immunol. 8, 867 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Roybal, K. T. et al. Engineering T cells with customized therapeutic response programs using synthetic notch receptors. Cell 167, 419–432.e16 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Papapetrou, E. P. & Schambach, A. Gene insertion into genomic safe harbors for human gene therapy. Mol. Ther. 24, 678–684 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lawrence, T. The nuclear factor NF-κB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 1, a001651 (2009).

  52. Tamura, T., Yanai, H., Savitsky, D. & Taniguchi, T. The IRF family transcription factors in immunity and oncogenesis. Annu. Rev. Immunol. 26, 535–584 (2008).

  53. Frieda, K. L. et al. Synthetic recording and in situ readout of lineage information in single cells. Nature 541, 107–111 (2017).

    Article  CAS  PubMed  Google Scholar 

  54. Frei, T. et al. Characterization and mitigation of gene expression burden in mammalian cells. Nat. Commun. 11, 4641 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chang, A. L., Wolf, J. J. & Smolke, C. D. Synthetic RNA switches as a tool for temporal and spatial control over gene expression. Curr. Opin. Biotechnol. 23, 679–688 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Banaszynski, L. A., Chen, L., Maynard-Smith, L. A., Ooi, A. G. L. & Wandless, T. J. A rapid, reversible, and tunable method to regulate protein function in living cells using synthetic small molecules. Cell 126, 995 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Watters, K. E., Fellmann, C., Bai, H. B., Ren, S. M. & Doudna, J. A. Systematic discovery of natural CRISPR-Cas12a inhibitors. Science 362, 236–239 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Jillette, N., Du, M., Zhu, J. J., Cardoz, P. & Cheng, A. W. Split selectable markers. Nat. Commun. 10, 4968 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Qi laboratory, including M. Chavez, V. Tieu, P. Finn, T. Abbott, A. Chemparathy, X. Xu, J. Bian and M. Nakamura, as well as E. Gonzalez Diaz and F. Yang for advice and helpful discussions. The authors also thank the Stanford Shared Protein and Nucleic Acid Facility for technical support. H.R.K. acknowledges support from the National Science Foundation Graduate Research Fellowship Program, Stanford Bio-X Fellowship Program and Siebel Scholars Foundation. L.S.Q. acknowledges support from the Li Ka Shing Foundation, the Stanford Maternal and Child Health Research Institute through the Uytengsu-Hamilton 22q11 Neuropsychiatry Research Award Program, National Science Foundation CAREER award (award no. 2046650) and California Institute for Regenerative Medicine (CIRM, DISC2-12669). L.S.Q. is a Chan Zuckerberg Biohub investigator. The work is supported by National Science Foundation CAREER award (award no. 2046650) and the Li Ka Shing Foundation.

Author information

Authors and Affiliations

Authors

Contributions

H.R.K. and L.S.Q. conceived the idea. H.R.K. designed and performed experiments for all figures. K.S.L. designed constructs and performed experiments for initial testing of Cas12 base editor constructs. L.Y.G. designed constructs and contributed for testing of hyperCas12 mutants. H.R.K. analyzed the data. H.R.K. and L.S.Q. wrote the manuscript. All authors read and commented on the manuscript.

Corresponding author

Correspondence to Lei S. Qi.

Ethics declarations

Competing interests

The authors have filed a provisional patent via Stanford University partly related to the work (US patent no. 63/148,652; international application patent no. PCT/US2022/016223). L.S.Q. is a founder of Epicrispr Biotechnologies.

Peer review

Peer review information

Nature Chemical Biology thanks the anonymous reviewers 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 Base editing with hyperCas12a for signal recording.

a, Left, representative flow cytometry histograms showing GFP fluorescence in HEK cells 3 days post-transfection with GFP* reporter, U6-driven guide and wild type (WT) or mutant (hyper) dCas12a-ABE. crLacZ, non-targeting control. Right, quantification of percentage GFP + cells for 3 independent replicates. To go with Fig. 1c. b, Representative flow cytometry histograms for GFP* HEK cells transfected with the dox-recording constructs, 3 days post-transfection and stimulation with dox. To go with Fig. 1f. c, Representative flow cytometry histograms for GFP* HEK cells transfected with hyperdCas12a base editor and NFKB-driven crGFP*, 3 days post-transfection and stimulation with TNFα. To go with Fig. 1g.

Source data

Extended Data Fig. 2 Optimizing inducible crRNAs for lentiviral transduction.

a, Left, designs tested for transferring the inducible guide cassette into a lentiviral backbone. The inducible construct was placed in reverse orientation, to prevent the polyA from disrupting lentiviral packaging. Right, quantification of reporter GFP fluorescence in HEK cells transiently transfected with inducible guide, mutant base editor, and reporter, as well as additional rtTA for design 2. Data shown for 2 independent replicates, 3 days post-transfection and stimulation with dox. Design 1 was selected moving forward, as despite slightly worse performance than design 2 it had benefit of including constitutive open reading frame for selection. b, Left, schematic showing incorporation of additional repeat-spacer cassette to increase amount of guide available per mRNA transcript. Right, GFP fluorescence in GFP* HEK cells stably transduced with dox-inducible guide, 3 days post-transfection with base editor and stimulation with dox. Data shown for 3 independent replicates. c, Left, representative histogram of GFP* HEK cells stably transduced with NFKB-crRNA, 3 days post-transfection with base editor and stimulation with TNFα. Right, quantification of mean fluorescence and percent GFP + for 3 independent replicates. d, Quantification of A→G mutations in stably transduced NFKB-crRNA cells at the targeted stop codon in the GFP* locus as measured by MiSeq. Data are shown for 3 independent replicates.

Source data

Extended Data Fig. 3 Dual signal recording using the GFP** reporter.

a, Schematic showing the GFP** dual recording reporter, which expresses GFP upon the activity of two distinct GFP* guides removing two stop codons. b, Left, representative histograms of HEK cells 3 days post-transfection with GFP**, base editor, and combinations of U6-crGFP*. Right, quantification of mean GFP for 3 independent replicates. c, Modified lentiviral constructs used to transduce two inducible guides into cells and select with a single antibiotic, through use of split hygromycin. d, Representative histograms of GFP** HEK cells stably transduced with NFKB and TRE3G inducible guides shown in c and transfected with base editor, 3 days posttransfection and stimulation with TNFα/dox. e, Quantification of mean fluorescence (left) and percentage GFP + (right) of GFP** HEK cells stably transduced with NFKB and TRE3G inducible guides and transfected with base editor, 3 days post transfection and stimulation with TNFα/dox. All data shown for 3 independent replicates. f, Quantification of A→G mutations at each targeted stop codon in the GFP** locus as measured by MiSeq. Data are shown for 3 independent replicates.

Source data

Extended Data Fig. 4 Modular recording of diverse biological signals.

a, Schematic showing modular recording by incorporating different inducible promoters into the guide construct. b, Cellular memory of NFAT signaling, stimulated by PMA. c, Cellular memory of oxidative stress, stimulated by tBHQ. ROS = reactive oxygen species. All experiments in b-c shown in GFP* HEK cells stably transduced with respective inducible guide, for 3 independent replicates 3 days post transfection with base editor.

Source data

Extended Data Fig. 5 Recording memory of antigen encounter.

a, Constructs used for SynNotch-induced guide expression. b, Cellular memory of antigen encounter via SynNotch, in GFP* HEK cells transfected with guide, SynNotch, and base editor, then cocultured with -/+CD19 HEK cells. Data shown for 3 independent replicates c, Constructs used for recording CD19-CAR activation in Jurkat cells. The top two constructs were stably integrated, and the bottom two were electroporated. d, Representative flow cytometry histograms for signal recording of CD19 antigen encounter in Jurkat cells. Histograms show BFP (induced by NFAT promoter) vs GFP (from successful base editing event of GFP* reporter) in engineered Jurkat CAR-T cells, after 48 hours of co-culture with K562 cells -/+CD19. To go with Fig. 2d.

Source data

Extended Data Fig. 6 Characterization of recording parameters.

a, GFP* HEK cells stably transduced with lentiviral NFKBp-guide and transfected with base editor were stimulated with 100 ng/mL TNFα for 3 days, then analyzed via flow cytometry. Randomly selected sub-populations of cells of the indicated size were analyzed using downsampling. Statistical analysis was performed using a two-sided t test without adjustment for multiple comparisons. Data shown for 3 independent replicates. To go with Fig. 3a. b, The mean GFP fluorescence for each downsampled cell population for the NFKB-recording circuit, +TNFα condition is plotted against the number of cells analyzed, to go with Fig. 3a. Dotted lines with gray shading indicate standard deviation of 3 independent replicates. c, Dose response for GFP* HEK cells stably transduced with lentiviral NFKBp-guide and transfected with base editor. Mean BFP fluorescence is shown 3 days post-transfection and stimulation with TNFα. Error bars indicate standard deviation of 3 independent replicates. To go with Fig. 3b. d, Mean BFP (expressed under NFKB promoter) plotted against mean GFP (“memory” reporter) for TNFα dose-response experiment. Data were fitted using a linear regression. Error bars indicate standard deviation of 3 independent replicates. e, Experimental timeline used for testing different durations of TNFα stimulation.

Source data

Extended Data Fig. 7 Recording history of NFKB signaling at the AAVS1 locus.

a, Left, constructs used for signal recording using inducible AAVS1 targeting guides in HEK cells. Right, experimental timeline used for genomic DNA (gDNA) collection. b, Single-input recording at the AAVS1 locus in HEK cells transfected with base editor and NFKB-driven crAAVS1_4, and stimulated with TNFα for 3 days. The %A→G conversion as measured by MiSeq is shown for A11 (left) and A14 (right), with number indicating position of A relative to PAM. Data shown for 3 independent replicates. c, Left, constructs used for recording at the AAVS1 locus in Jurkat cells. Right, experimental timeline. d, Recording of NFKB-mediated inflammation in Jurkat cells at the AAVS1 locus, as measured by MiSeq. Data shown for 3 independent replicates.

Source data

Extended Data Fig. 8 Utilizing two parallel guides for more detailed record of input signal characteristics.

a, Schematic showing two input signals with distinctive patterns but the same area under the curve. b, Promoter engineering strategy used to reduce the sensitivity of NFKB-responsive promoter, to generate two inducible guide constructs that respond differently to input TNFα signal. c, Mean BFP fluorescence of HEK cells 3 days post-transfection with NFKB promoter variants and stimulation with TNFα. Left, raw mean BFP values are plotted. Right, mean BFP values normalized to the maximum of each respective promoter are plotted. Error bars indicate standard deviation of 3 independent replicates. d, Quantification of %A→G conversion as measured by MiSeq for HEK cells transfected with base editor, 5X-NFKB-crAAVS1_4, and 2X-NFKB-crAAVS1_8. One day after transfection, cells were stimulated with either 100 ng/mL TNFα for 0.25 hours, or 0.5 ng/mL TNFα for 50 hours. All data shown for 3 independent replicates. Statistical analysis was performed using a two-sided t test without adjustment for multiple comparisons.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–8 and Tables 1–6.

Reporting Summary

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. 4

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. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kempton, H.R., Love, K.S., Guo, L.Y. et al. Scalable biological signal recording in mammalian cells using Cas12a base editors. Nat Chem Biol 18, 742–750 (2022). https://doi.org/10.1038/s41589-022-01034-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01034-2

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