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:

Intrinsic targeting of host RNA by Cas13 constrains its utility

Abstract

Cas13 can be used for the knockdown, editing, imaging or detection of RNA and for RNA-based gene therapy. Here by using RNA immunoprecipitation sequencing, transcriptome profiling, biochemical analysis, high-throughput screening and machine learning, we show that Cas13 can intrinsically target host RNA in mammalian cells through previously unappreciated mechanisms. Different from its known cis/trans RNA-cleavage activity, Cas13 can also cleave host RNA via mechanisms that are transcript-specific, independent of the sequence of CRISPR RNA and dynamically dependent on the conformational state of Cas13, as we show for several Cas13-family effectors encoded in one-vector and two-vector lentiviral systems. Moreover, host genes involved in viral processes and whose transcripts are intrinsically targeted by Cas13 contribute to constraining the lentiviral delivery and expression of Cas13. Our findings offer guidance for the appropriate use of lentiviral Cas13 systems and highlight the need for caution regarding intrinsic RNA targeting in Cas13-based 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: Lentiviral defects of one-vector Cas13 systems.
Fig. 2: Lentiviral defects of two-vector Cas13 systems.
Fig. 3: Intrinsic RNA targets of different Cas13 effectors.
Fig. 4: Pre-crRNA cleavage activity underlies lentiviral defect of Cas13a.
Fig. 5: Endogenous RNA cleavage by Cas13a.
Fig. 6: Evaluation of Cas13 performance during high-throughput screening.

Similar content being viewed by others

Data availability

The data supporting the results in this study are available within the paper and its Supplementary information. The raw and analysed datasets generated in this study are available from the NCBI Gene Expression Omnibus database via the access code GSE202899. Source data are provided with this paper.

Code availability

The custom code for performing Lasso-based machine learning is available at https://github.com/cxlsky/Cas13-Lasso.

References

  1. Abudayyeh, O. O. et al. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science 353, aaf5573 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Terns, M. P. CRISPR-based technologies: impact of RNA-targeting systems. Mol. Cell 72, 404–412 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Abudayyeh, O. O. et al. A cytosine deaminase for programmable single-base RNA editing. Science 365, 382–386 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yang, L. Z. et al. Dynamic imaging of RNA in living cells by CRISPR-Cas13 systems. Mol. Cell 76, 981–997.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. East-Seletsky, A. et al. Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature 538, 270–273 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Powell, J. E. et al. Targeted gene silencing in the nervous system with CRISPR-Cas13. Sci. Adv. 8, eabk2485 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gootenberg, J. S. et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science 356, 438–442 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Perculija, V., Lin, J., Zhang, B. & Ouyang, S. Functional features and current applications of the RNA-targeting type VI CRISPR-Cas systems. Adv. Sci. 8, 2004685 (2021).

    Article  CAS  Google Scholar 

  9. Cox, D. B. T. et al. RNA editing with CRISPR-Cas13. Science 358, 1019–1027 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Abudayyeh, O. O. et al. RNA targeting with CRISPR-Cas13. Nature 550, 280–284 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  11. Konermann, S. et al. Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors. Cell 173, 665–676.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Smargon, A. A. et al. Cas13b is a type VI-B CRISPR-associated RNA-guided RNase differentially regulated by accessory proteins Csx27 and Csx28. Mol. Cell 65, 618–630.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yan, W. X. et al. Cas13d is a compact RNA-targeting type VI CRISPR effector positively modulated by a WYL-domain-containing accessory protein. Mol. Cell 70, 327–339.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ozcan, A. et al. Programmable RNA targeting with the single-protein CRISPR effector Cas7-11. Nature 597, 720–725 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Xu, C. et al. Programmable RNA editing with compact CRISPR-Cas13 systems from uncultivated microbes. Nat. Methods 18, 499–506 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Kannan, S. et al. Compact RNA editors with small Cas13 proteins. Nat. Biotechnol. 40, 194–197 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. O’Connell, M. R. Molecular mechanisms of RNA targeting by Cas13-containing type VI CRISPR-Cas systems. J. Mol. Biol. 431, 66–87 (2019).

    Article  PubMed  Google Scholar 

  18. Myhrvold, C. et al. Field-deployable viral diagnostics using CRISPR-Cas13. Science 360, 444–448 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, B. et al. Two HEPN domains dictate CRISPR RNA maturation and target cleavage in Cas13d. Nat. Commun. 10, 2544 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  20. Liu, L. et al. Two distant catalytic sites are responsible for C2c2 RNase activities. Cell 168, 121–134.e12 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Wessels, H. H. et al. Massively parallel Cas13 screens reveal principles for guide RNA design. Nat. Biotechnol. 38, 722–727 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, Y. et al. Optimized RNA-targeting CRISPR/Cas13d technology outperforms shRNA in identifying functional circRNAs. Genome Biol. 22, 41 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, S. et al. Screening for functional circular RNAs using the CRISPR-Cas13 system. Nat. Methods 18, 51–59 (2021).

    Article  PubMed  Google Scholar 

  24. Abbott, T. R. et al. Development of CRISPR as an antiviral strategy to combat SARS-CoV-2 and influenza. Cell 181, 865–876.e12 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kumar, A. et al. Genome-wide gene expression pattern underlying differential host response to high or low pathogenic H5N1 avian influenza virus in ducks. Acta Virol. 61, 66–76 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Rahim, M. N. et al. Global interactomics connect nuclear mitotic apparatus protein NUMA1 to influenza virus maturation. Viruses 10, 731 (2018).

  27. Yoh, S. M. et al. PQBP1 is a proximal sensor of the cGAS-dependent innate response to HIV-1. Cell 161, 1293–1305 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sammaibashi, S., Yamayoshi, S. & Kawaoka, Y. Strain-specific contribution of eukaryotic Elongation Factor 1 Gamma to the translation of influenza A virus proteins. Front. Microbiol. 9, 1446 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Chen, R. Y. et al. White spot syndrome virus entry is dependent on multiple endocytic routes and strongly facilitated by Cq-GABARAP in a CME-dependent manner. Sci. Rep. 6, 28694 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sadat, M. A. et al. Glycosylation, hypogammaglobulinemia, and resistance to viral infections. N. Engl. J. Med. 370, 1615–1625 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Santoni, G. et al. Involvement of the TRPML mucolipin channels in viral infections and anti-viral innate immune responses. Front. Immunol. 11, 739 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Besnard, E. et al. The mTOR complex controls HIV latency. Cell Host Microbe 20, 785–797 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Saha, A. et al. EBNA3C attenuates the function of p53 through interaction with inhibitor of growth family proteins 4 and 5. J. Virol. 85, 2079–2088 (2011).

  34. Xia, Z. et al. NAC1 potentiates cellular antiviral signaling by bridging MAVS and TBK1. J. Immunol. 203, 1001–1011 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. Tong, H. et al. High-fidelity Cas13 variants for targeted RNA degradation with minimal collateral effects. Nat. Biotechnol. 41, 108–119 (2023).

    Article  CAS  PubMed  Google Scholar 

  36. Su, Y. S., Hwang, L. H. & Chen, C. J. Heat shock protein A6, a novel HSP70, is induced during enterovirus A71 infection to facilitate internal ribosomal entry site-mediated translation. Front. Microbiol. 12, 664955 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Li, W. et al. Quality control, modeling, and visualization of CRISPR screens with MAGeCK-VISPR. Genome Biol. 16, 281 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cheng, X. et al. Modeling CRISPR-Cas13d on-target and off-target effects using machine learning approaches. Nat. Commun. 14, 752 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wang, Q. et al. The CRISPR-Cas13a gene-editing system induces collateral cleavage of RNA in glioma cells. Adv. Sci. 6, 1901299 (2019).

    Article  CAS  Google Scholar 

  40. Ai, Y., Liang, D. & Wilusz, J. E. CRISPR/Cas13 effectors have differing extents of off-target effects that limit their utility in eukaryotic cells. Nucleic Acids Res. 50, e65 (2022).

  41. Vialetto, E. et al. A target expression threshold dictates invader defense and prevents autoimmunity by CRISPR-Cas13. Cell Host Microbe 30, 1151–1162 (2022).

  42. Li, Y. et al. The collateral activity of RfxCas13d can induce lethality in a RfxCas13d knock-in mouse model. Genome Biol. 24, 20 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kelley, C. P., Haerle, M. C. & Wang, E. T. Negative autoregulation mitigates collateral RNase activity of repeat-targeting CRISPR-Cas13d in mammalian cells. Cell Rep. 40, 111226 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Shi, P. et al. Collateral activity of the CRISPR/RfxCas13d system in human cells. Commun. Biol. 6, 334 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, B. et al. Integrative analysis of pooled CRISPR genetic screens using MAGeCKFlute. Nat. Protoc. 14, 756–780 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hart, T., Brown, K. R., Sircoulomb, F., Rottapel, R. & Moffat, J. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol. Syst. Biol. 10, 733 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Du, Z. et al. Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat. Struct. Mol. Biol. 20, 908–913 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Xiao for technical advice and Y. Huang for sharing experimental material. This work was supported by the National Natural Science Foundation of China (31871344, 32071441), the Fundamental Research Funds for the Central Universities (N182005005, N2020001, N2220001), the 111 Project (B16009), the Key Laboratory of Bioresource Research and Development of Liaoning Province (2022JH13/10200026) and Liaoning Revitalization Talents Program (XLYC1807212) to T.F., and the research grant from the National Institutes of Health (R01HG010753) to W.L.

Author information

Authors and Affiliations

Authors

Contributions

T.F. and W.L. conceived the study and designed the research. Zexu Li and Zihan Li conducted most of the experiments. Zexu Li and X.C. conducted bioinformatics analysis. All the authors analysed the data. T.F. and W.L. wrote the paper with input from Zexu Li, Zihan Li and X.C., and help from all the other authors. T.F. and W.L. supervised the study.

Corresponding authors

Correspondence to Wei Li or Teng Fei.

Ethics declarations

Competing interests

W.L. is a paid consultant to Tavros Therapeutics, Inc. All other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Chunsheng Kang, Songying Ouyang and Li Yang 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 Assessment of lentiviral defect for one-vector Cas13 systems.

a, Western blot of protein extracts from HEK293FT cells transiently expressing different Cas13 effectors in one-vector format. b, Immunocytochemistry of Cas13 proteins showing localization and expression. Scale bar, 20 µm. c-d, Surviving cell number after puromycin selection for T47D (c) or LNCaP (d) cells infected by indicated one-vector Cas13 lentiviruses, mean ± SEM with n = 3, biological replicates. e, Cell viability analysis for HEK293FT cells with transient transfection of indicated one-vector lentiviral constructs, mean ± SEM with n = 3, biological replicates. Vector: lentiv2-w/o Cas9. f, Cell viability analysis for HEK293FT cells during lentivirus production by transient transfection of one-vector Cas13 along with packaging plasmids, mean ± SEM with n = 3, biological replicates. g-i, Assessment of surviving cell number (g), lentiviral RNA (h) and integrated lentiviral DNA (i) in A549 cells infected with indicated lentiviruses at low MOI. Data are mean ± SEM with n = 3, biological replicates (g), mean ± SD with n = 3, technical replicates (h-i).

Source data

Extended Data Fig. 2 Additional evaluation of lentiviral defect for one-vector Cas13 systems.

a-b, Time course examination of lentiviral RNA (a) or integrated DNA (b) post lentiviral infection in A549 cells, mean ± SD with n = 3, technical replicates. c-e, Assessment of surviving cell number (c), lentiviral RNA (d) and integrated lentiviral DNA (e) in A549 cells infected with indicated lentiviruses containing different crRNAs, mean ± SEM with n = 3. f, Evaluation of lentiviral defect for one-vector Cas13 systems under pHAGE-EF1α-puro plasmid backbone. Data are mean ± SEM with n = 3, biological replicates in cell survival assay, mean ± SD with n = 3, technical replicates in qPCR assays.

Source data

Extended Data Fig. 3 Evaluation of two-vector Cas13 systems for lentiviral defect.

a-c, Assessment of surviving cell number (a), lentiviral RNA (b) and integrated lentiviral DNA (c) in A549 cells infected with indicated lentiviruses at low MOI. d, Effect on cell survival after puromycin selection for A549 cells infected with high or low MOI of Cas13-only lentiviruses of two-vector system under pHAGE-EF1α-puro vector backbone, mean ± SEM with n = 3, biological replicates. e, Assessment of surviving cell number, lentiviral RNA and integrated lentiviral DNA levels using pHAGE-EF1α-puro vector backbone in A549 cells infected with indicated lentiviruses at high or low MOI. f, RNA levels in HEK293FT cells (left), HeLa cells (middle) and A549 cells (right) transiently transfected with pcDNA3.1-based Cas13 vectors by measuring NES element, mean ± SD with n = 3, technical replicates. g, Schematic of co-infection assay using indicated lentiviruses to evaluate lentiviral defect. Red arrowheads indicate detection region by qPCR at RNA and DNA levels. h-k, Assessment of surviving cell number (h) and lentiviral RNA levels using Cas13 (i), Cas13DR (j) or PuroR (k) elements in A549 cells co-infected with indicated lentiviruses. l, Integrated lentiviral DNA levels from genomic DNA of infected HeLa cells, U87 cells, SH-SY5Y cells and Neuro-2a cells by measuring NLS element, normalized to RPS28 or GAPDH (for SH-SY5Y), mean ± SD with n = 3, technical replicates. m-n, Cycloheximide (CHX) chase analysis for determining Cas13 protein stability in HEK293FT cells. Western blot (m) showing the protein levels at indicated time points and quantified band intensity was shown in (n).

Source data

Extended Data Fig. 4 Identification of intrinsic RNA targets of Cas13 by RIP-seq.

a, Schematic of RNA interactome and transcriptome analysis transiently transfected with indicated constructs in HEK293FT cells. Cas13b-crNT indicates a one-vector Cas13b system containing a non-targeting (NT) crRNA. Vector: lentiv2-w/o Cas9. b, The number of filtered strong RIP binding peaks (pileup > 15; fold enrichment > 4) in different samples. c, Loci and feature distribution of RIP-seq peaks for indicated samples. d, Top enriched motifs among RIP-seq peaks for indicated samples. e, Heatmap of genes associated with strong Cas13-bound peaks over the transcriptome. Vector: lentiv2-w/o Cas9. f, Top GO categories enriched among RIP-seq peak-associated genes across different samples.

Source data

Extended Data Fig. 5 Identification of intrinsic RNA targets of Cas13 by RNA-seq.

a, Heatmap of differentially expressed genes in Cas9- or Cas13-expressing HEK293FT cells. b, The number of differentially expressed genes that were up-regulated or down-regulated for indicated samples compared to vector control. c, Venn diagrams showing the overlaps of up- (left) or down-regulated (right) genes between Cas13a, Cas13b and Cas13d groups, versus vector. d-e, Top GO categories enriched among up- (d) or down-regulated (e) differentially expressed genes. f, Venn diagrams representing overlaps of up- (left) or down-regulated (right) genes in Cas13b or Cas13b-crNT groups, versus vector. g-h, RT-qPCR (g) and Western blot (h) validation for indicated gene expression in HEK293FT cells transiently transfected with lentiCRISPR v2-based Cas13b or Cas13b-crNT vectors, mean ± SD with n = 3, technical replicates for qPCR assay. i, Top 5 enriched GO categories of each Cas13-specific down- and up-regulated differentially expressed genes for Cas13a (top), Cas13b (middle) and Cas13d (bottom), versus vector.

Source data

Extended Data Fig. 6 Characterization of Cas13a variants.

a, Sequence alignment of catalytic domains for Cas13a derived from different bacteria strains. The conserved functional residues are shown in red star or by black arrow. b, Western blot of protein extracts from HEK293FT cells transiently transfected with indicated constructs. c, Coomassie blue staining showing purified Cas9 and Cas13a proteins. d, Cis RNA cleavage by indicated Cas13a variants during in vitro assay. Red arrow indicates the band position of on-target RNA or crRNA. e-f, Trans RNA cleavage by indicated Cas13a variants during in vitro assay. Results are shown by either fluorescence signal value (e) or direct visualization under blue light illuminator (f). a.u., arbitrary unit. mean ± SEM with n = 3, biological replicates. g, Pre-crRNA cleavage by indicated Cas13a variants during in vitro assay. Red arrows indicate band positions of intact and different cleavage patterns of RNA with structural schematic shown in right.

Source data

Extended Data Fig. 7 Profiling and validation of endogenous RNA targets by Cas13a variants.

a, The number of differentially expressed genes that were up-regulated or down-regulated among Cas13 variants compared to vector control. b-c, Top functional categories enriched among up- (b) or down-regulated (c) differentially expressed genes for indicated samples. d, RT-qPCR validation of indicated gene expression in HEK293FT cells transiently transfected with lentiCRISPR v2-based Cas13a and its variants, mean ± SD with n = 3, technical replicates. e, RNA expression change by RT-qPCR for indicated genes upon transient transfection with indicated constructs, mean ± SD with n = 3, technical replicates. f, Schematic for evaluating lentiviral capacity of HSPA6 knockdown by siRNA in HEK293FT cells. Lentiviruses (lentiCRISPR v2) produced in either control (siCtrl) or HSPA6 knockdown HEK293FT cells were used to infect A549 cells and lentiviral RNA or DNA level was determined. g, Assessment of HSPA6 knockdown efficiency in HEK293FT cells (left), lentiviral DNA (middle) and lentiviral RNA level (right) in A549 cells by measuring a region spanning NLS and FLAG tag in lentiCRISPR v2 vector, mean ± SD with n = 3, technical replicates. P values are determined via two-way ANOVA with Tukey’s multiple comparisons test or one-way ANOVA with Dunnett’s multiple comparisons test. h, RT-qPCR (left) and Western blot (right) analysis confirming the ectopic expression and knockdown of HSPA6 in HEK293FT cells. i, Integrated lentiviral DNA level of lentiCRISPR v2 vector in A549 cells by measuring NLS element after re-introduction of HSPA6 into HSPA6 knockdown HEK293FT cells by transient transfection, mean ± SD with n = 3, technical replicates.

Source data

Extended Data Fig. 8 Endogenous RNA targeting by Cas13.

a, RIP-qPCR validation of Cas13a binding sites. HEK293FT cells transiently expressing vector control (lenti-w/o Cas9) or Cas13a-Flag are analyzed. HA-IP serves as negative RIP control. GAPDH serves as negative site for Cas13a RIP enrichment. Values shown as mean ± SD with n = 3, technical replicates. *p < 0.001, two-way ANOVA with Sidak’s multiple comparisons test; ns means not significant. b-c, RT-qPCR (b) and Western blot (c) validation of indicated gene expression in HEK293FT cells transiently transfected with lentiCRISPR v2-based Cas13a and its variants. d, In vitro assay to assess the effect of Cas13a with differential conformation and complex constitution on endogenous RNA cleavage. e, Coomassie blue staining showing purified Cas9 and Cas13a/b/d proteins.

Source data

Extended Data Fig. 9 Cas13b/d-based cell fitness screens.

a, Schematic of computational pipeline to choose targeted RNA transcripts for Cas13b/d-based cell fitness screens. b, Statistics of sgRNA libraries for Cas13b/d-based screens. c, Scatter plot showing crRNA abundance change (log2 fold-change) in virus pool versus plasmid library for two biological replicates of either one-vector or two-vector Cas13d screens. R2, Pearson’s correlation coefficient. d, Scatter plot of β score (Day 33 versus Day 5) change for target transcripts between two biological replicates in Cas13d-null or Cas13d-expressing conditions of two-vector Cas13d screens. Red dots indicate core essential genes. e, The distribution of crRNA abundance across samples of different time points compared to plasmid pool for two-vector Cas13b screen. Boxes depict the upper and lower quartiles of the data, whiskers depict the maximum or minimum value within 1.5 times interquartile range above 75th or below 25th percentile, and black dots depict values that >1.5 times the interquartile range beyond either end of the box. f, The distribution of crRNA abundance (log2 fold-change) across samples of different time points compared to plasmid pool for two-vector Cas13b screens. g, Scatter plot of β score (Day 33 versus Day 5) change for target transcripts between two biological replicates in Cas13b-null or Cas13b-expressing conditions of two-vector Cas13b screens. Red dots indicate core essential genes.

Source data

Extended Data Fig. 10 Lasso model construction strategy and comparative summary of different types of Cas13 RNA targeting.

a, Early loss of crRNAs targeting non-essential genes in one-vector or two-vector Cas13d screens. Log2 fold-change (Day 5 vs. plasmid pool) < −0.5. b, Schematic of lasso-based machine learning model to determine crRNA-specific features underlying lentiviral defect. c, Comparison of features between different types of Cas13 activities.

Source data

Supplementary information

Supplementary Information

Supplementary discussion and references.

Reporting Summary

Supplementary dataset 1

Oligonucleotides, primers and crRNA sequences.

Supplementary Dataset 2

RIP-seq data.

Supplementary Dataset 3

RNA-seq data.

Supplementary Dataset 4

Cas13 libraries for high-throughput screening.

Supplementary Dataset 5

Cas13 screening data.

Supplementary Dataset 6

Unprocessed western blots and unprocessed gels.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Z., Li, Z., Cheng, X. et al. Intrinsic targeting of host RNA by Cas13 constrains its utility. Nat. Biomed. Eng 8, 177–192 (2024). https://doi.org/10.1038/s41551-023-01109-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-023-01109-y

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