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:

A repackaged CRISPR platform increases homology-directed repair for yeast engineering

Abstract

Inefficient homology-directed repair (HDR) constrains CRISPR–Cas9 genome editing in organisms that preferentially employ nonhomologous end joining (NHEJ) to fix DNA double-strand breaks (DSBs). Current strategies used to alleviate NHEJ proficiency involve NHEJ disruption. To confer precision editing without NHEJ disruption, we identified the shortcomings of the conventional CRISPR platforms and developed a CRISPR platform—lowered indel nuclease system enabling accurate repair (LINEAR)—which enhanced HDR rates (to 67–100%) compared to those in previous reports using conventional platforms in four NHEJ-proficient yeasts. With NHEJ preserved, we demonstrate its ability to survey genomic landscapes, identifying loci whose spatiotemporal genomic architectures yield favorable expression dynamics for heterologous pathways. We present a case study that deploys LINEAR precision editing and NHEJ-mediated random integration to rapidly engineer and optimize a microbial factory to produce (S)-norcoclaurine. Taken together, this work demonstrates how to leverage an antagonizing pair of DNA DSB repair pathways to expand the current collection of microbial factories.

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: Surveying NHEJ preference in industrially relevant yeast species.
Fig. 2: NHEJ disruption hinders growth and fermentation performance of S. stipitis.
Fig. 3: Leveraging geminin dynamics to establish a temporal expression control.
Fig. 4: LINEAR CRISPR platform.
Fig. 5: Comparison of editing efficiency between the LINEAR platform and the conventional CRISPR platforms in various nonconventional species.
Fig. 6: Case study demonstrating the utility of LINEAR and NHEJ synergistically in strain engineering.

Similar content being viewed by others

Data availability

Source data are provided for the results presented in Main and Extended Data figures. Vectors containing LINEAR constructs for Y. lipolytica, K. marxianus, H. polymorpha and S. stipitis are available from Addgene (plasmid nos. 174837–174840). Raw reads and assembled genome sequences for the final engineered norcoclaurine-producing strains have been deposited in NCBI under BioProject ID PRJNA753835. Additional data are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

All code used for processing and assembly of genome sequencing reads (along with an accompanying method for how the code was implemented) is available on Zenodo: https://doi.org/10.5281/zenodo.5544230.

References

  1. Hittinger, C. T. et al. Genomics and the making of yeast biodiversity. Curr. Opin. Genet. Dev. 35, 100–109 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Thorwall, S., Schwartz, C., Chartron, J. W. & Wheeldon, I. Stress-tolerant non-conventional microbes enable next-generation chemical biosynthesis. Nat. Chem. Biol. 16, 113–121 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Goncalves, F. A., Colen, G. & Takahashi, J. A. Yarrowia lipolytica and its multiple applications in the biotechnological industry. ScientificWorldJournal 2014, 476207 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Fonseca, G. G., Heinzle, E., Wittmann, C. & Gombert, A. K. The yeast Kluyveromyces marxianus and its biotechnological potential. Appl. Microbiol. Biotechnol. 79, 339–354 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Manfrao-Netto, J. H. C., Gomes, A. M. V. & Parachin, N. S. Advances in using Hansenula polymorpha as chassis for recombinant protein production. Front. Bioeng. Biotechnol. 7, 94 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gao, M. et al. Innovating a nonconventional yeast platform for producing shikimate as the building block of high-value aromatics. ACS Synth. Biol. 6, 29–38 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Lobs, A. K., Schwartz, C. & Wheeldon, I. Genome and metabolic engineering in non-conventional yeasts: current advances and applications. Synth. Syst. Biotechnol. 2, 198–207 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Kuck, U. & Hoff, B. New tools for the genetic manipulation of filamentous fungi. Appl. Microbiol. Biotechnol. 86, 51–62 (2010).

    Article  PubMed  Google Scholar 

  9. Mao, Z., Bozzella, M., Seluanov, A. & Gorbunova, V. Comparison of nonhomologous end joining and homologous recombination in human cells. DNA Repair (Amst.) 7, 1765–1771 (2008).

    Article  CAS  Google Scholar 

  10. Cao, M., Gao, M., Ploessl, D., Song, C. & Shao, Z. CRISPR-mediated genome editing and gene repression in Scheffersomyces stipitis. Biotechnol. J. 13, e1700598 (2018).

    Article  PubMed  Google Scholar 

  11. Sishc, B. J. & Davis, A. J. The role of the core non-homologous end joining factors in carcinogenesis and cancer. Cancers (Basel) 9, 81 (2017).

  12. Ferguson, D. O. et al. The nonhomologous end-joining pathway of DNA repair is required for genomic stability and the suppression of translocations. Proc. Natl Acad. Sci. USA 97, 6630–6633 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lin, S., Staahl, B. T., Alla, R. K. & Doudna, J. A. Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife 3, e04766 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Janssen, J. M., Chen, X., Liu, J. & Goncalves, M. The chromatin structure of CRISPR-Cas9 target DNA controls the balance between mutagenic and homology-directed gene-editing events. Mol. Ther. Nucleic Acids 16, 141–154 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lieber, M. R. & Karanjawala, Z. E. Ageing, repetitive genomes and DNA damage. Nat. Rev. Mol. Cell Biol. 5, 69–75 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Mao, Z., Bozzella, M., Seluanov, A. & Gorbunova, V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell Cycle 7, 2902–2906 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Tang, X. F. et al. Two Geminin homologs regulate DNA replication in silkworm, Bombyx mori. Cell Cycle 16, 830–840 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kerns, S. L., Torke, S. J., Benjamin, J. M. & McGarry, T. J. Geminin prevents rereplication during xenopus development. J. Biol. Chem. 282, 5514–5521 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Quinn, L. M., Herr, A., McGarry, T. J. & Richardson, H. The Drosophila Geminin homolog: roles for Geminin in limiting DNA replication, in anaphase and in neurogenesis. Genes Dev. 15, 2741–2754 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. McGarry, T. J. & Kirschner, M. W. Geminin, an inhibitor of DNA replication, is degraded during mitosis. Cell 93, 1043–1053 (1998).

    Article  CAS  PubMed  Google Scholar 

  22. Gutschner, T., Haemmerle, M., Genovese, G., Draetta, G. F. & Chin, L. Post-translational regulation of Cas9 during G1 enhances homology-directed repair. Cell Rep. 14, 1555–1566 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Slater, M. L., Sharrow, S. O. & Gart, J. J. Cell cycle of Saccharomyces cerevisiae in populations growing at different rates. Proc. Natl Acad. Sci. USA 74, 3850–3854 (1977).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Donaldson, A. D. et al. CLB5-dependent activation of late replication origins in S. cerevisiae. Mol. Cell 2, 173–182 (1998).

    Article  CAS  PubMed  Google Scholar 

  25. Collado, M., Blasco, M. A. & Serrano, M. Cellular senescence in cancer and aging. Cell 130, 223–233 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. YMotoko Sasaki, Y. N. in Autophagy: Cancer, Other Pathologies, Inflammation, Immunity, Infection, and Aging (ed. Hayat, M. A.) pp. 293–303 (Academic Press, 2014).

  27. Akhmetov, A. et al. Single-step precision genome editing in yeast using CRISPR-Cas9. Bio. Protoc. 8, e2765 (2018).

  28. Lobs, A. K., Engel, R., Schwartz, C., Flores, A. & Wheeldon, I. CRISPR-Cas9-enabled genetic disruptions for understanding ethanol and ethyl acetate biosynthesis in Kluyveromyces marxianus. Biotechnol. Biofuels 10, 164 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Numamoto, M., Maekawa, H. & Kaneko, Y. Efficient genome editing by CRISPR/Cas9 with a tRNA-sgRNA fusion in the methylotrophic yeast Ogataea polymorpha. J. Biosci. Bioeng. 124, 487–492 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Juergens, H. et al. Contribution of Complex I NADH dehydrogenase to respiratory energy coupling in glucose-grown cultures of Ogataea parapolymorpha. Appl. Environ. Microbiol. 86, e00678–20 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gao, S. et al. Multiplex gene editing of the Yarrowia lipolytica genome using the CRISPR-Cas9 system. J. Ind. Microbiol. Biotechnol. 43, 1085–1093 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Schwartz, C. M., Hussain, M. S., Blenner, M. & Wheeldon, I. Synthetic RNA polymerase III promoters facilitate high-efficiency CRISPR-Cas9-mediated genome editing in Yarrowia lipolytica. ACS Synth. Biol. 5, 356–359 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Vogel, C. & Marcotte, E. M. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat. Rev. Genet. 13, 227–232 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Park, S. H. & Hahn, J. S. Development of an efficient cytosolic isobutanol production pathway in Saccharomyces cerevisiae by optimizing copy numbers and expression of the pathway genes based on the toxic effect of alpha-acetolactate. Sci. Rep. 9, 3996 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Li, N., Zeng, W., Xu, S. & Zhou, J. Toward fine-tuned metabolic networks in industrial microorganisms. Synth. Syst. Biotechnol. 5, 81–91 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Sato, H., Das, S., Singer, R. H. & Vera, M. Imaging of DNA and RNA in living eukaryotic cells to reveal spatiotemporal dynamics of gene expression. Annu. Rev. Biochem. 89, 159–187 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chen, R., Yang, S., Zhang, L. & Zhou, Y. J. Advanced strategies for production of natural products in yeast. iScience 23, 100879 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hagel, J. M. & Facchini, P. J. Benzylisoquinoline alkaloid metabolism: a century of discovery and a brave new world. Plant Cell Physiol. 54, 647–672 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. DeLoache, W. C. et al. An enzyme-coupled biosensor enables (S)-reticuline production in yeast from glucose. Nat. Chem. Biol. 11, 465–471 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Liu, Q. et al. Rewiring carbon metabolism in yeast for high level production of aromatic chemicals. Nat. Commun. 10, 4976 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhao, Y. et al. Leveraging the Hermes transposon to accelerate the development of nonconventional yeast-based microbial cell factories. ACS Synth. Biol. 9, 1736–1752 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Jung, S. T., Lauchli, R. & Arnold, F. H. Cytochrome P450: taming a wild type enzyme. Curr. Opin. Biotechnol. 22, 809–817 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Veith, A. & Moorthy, B. Role of cytochrome P450s in the generation and metabolism of reactive oxygen species. Curr. Opin. Toxicol. 7, 44–51 (2018).

    Article  PubMed  Google Scholar 

  44. Hrycay, E. G. & Bandiera, S. M. Involvement of cytochrome P450 in reactive oxygen species formation and cancer. Adv. Pharmacol. 74, 35–84 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Bendich, A. J. The end of the circle for yeast mitochondrial DNA. Mol. Cell 39, 831–832 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Cao, M. et al. Centromeric DNA facilitates nonconventional yeast genetic engineering. ACS Synth. Biol. 6, 1545–1553 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Cao, M., Seetharam, A. S., Severin, A. J. & Shao, Z. Rapid isolation of centromeres from Scheffersomyces stipitis. ACS Synth. Biol. 6, 2028–2034 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Stanford, W. L., Cohn, J. B. & Cordes, S. P. Gene-trap mutagenesis: past, present and beyond. Nat. Rev. Genet. 2, 756–768 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Nguyen, V. Q., Co, C. & Li, J. J. Cyclin-dependent kinases prevent DNA re-replication through multiple mechanisms. Nature 411, 1068–1073 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Sopko, R. et al. Mapping pathways and phenotypes by systematic gene overexpression. Mol. Cell 21, 319–330 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Boeke, J. D., Trueheart, J., Natsoulis, G. & Fink, G. R. 5-Fluoroorotic acid as a selective agent in yeast molecular genetics. Methods Enzymol. 154, 164–175 (1987).

    Article  CAS  PubMed  Google Scholar 

  52. Shao, Z., Zhao, H. & Zhao, H. DNA assembler, an in vivo genetic method for rapid construction of biochemical pathways. Nucleic Acids Res. 37, e16 (2009).

    Article  PubMed  Google Scholar 

  53. Herricks, T. et al. One-cell doubling evaluation by living arrays of yeast, ODELAY! G3 (Bethesda) 7, 279–288 (2017).

    Article  CAS  Google Scholar 

  54. Calvert, M. E., Lannigan, J. A. & Pemberton, L. F. Optimization of yeast cell cycle analysis and morphological characterization by multispectral imaging flow cytometry. Cytometry A 73, 825–833 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Labun, K. et al. CHOPCHOP v3: expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res. 47, W171–W174 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Laplaza, B. R. T. JoseM., Jin, Yong-Su & Jeffries, ThomasW. Sh ble and Cre adapted for functional genomics and metabolic engineering of Pichia stipitis. Enzyme Microb. Technol. 38, 741–747 (2006).

    Article  CAS  Google Scholar 

  57. Di Tommaso, P. et al. Nextflow enables reproducible computational workflows. Nat. Biotechnol. 35, 316–319 (2017).

    Article  PubMed  Google Scholar 

  58. De Coster, W., D’Hert, S., Schultz, D. T., Cruts, M. & Van Broeckhoven, C. NanoPack: visualizing and processing long-read sequencing data. Bioinformatics 34, 2666–2669 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Kolmogorov, M. et al. metaFlye: scalable long-read metagenome assembly using repeat graphs. Nat. Methods 17, 1103–1110 (2020).

    Article  CAS  PubMed  Google Scholar 

  60. Li, H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics 34, 3094–3100 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Science Foundation Grant (no. 1716837 to D.P., Y.Z., M.C., S.G., C.L., M.S., S.C., A.S., L.H. and Z.S.), the NSF Graduate Research Fellowship Program (to D.P.) and Griswold Internship (to M.G.). We thank T. W. Jeffries (professor emeritus, University of Wisconsin-Madison and the Founder of Xylome Corporation) for sharing S. stipitis FLP-UC7 (ura3-3, NRRL Y21448) and the plasmid pJML545. We thank I. Wheeldon, University of California at Riverside, for sharing K. marxianus YS402 (CBC6556 Δura3); J. Dueber, University of California at Berkeley for sharing the S. cerevisiae plasmid containing TyrH* and DOD; and S. Yang, Chinese Academy of Science, for sharing the Y. lipolytica plasmids pCas1-yl and pCAS2-yl. We also thank S. M. Rigby for flow cytometry and L. Showman and K. Narayanaswamy at the W. M. Keck Metabolomics Research Laboratory of ISU for the detection of (S)-norcoclaurine. Finally, we thank T. Murtha at the ISU DNA facility for preparing gDNA samples for Nanopore sequencing.

Author information

Authors and Affiliations

Authors

Contributions

D.P. and Z.S. conceptualized the initial idea, designed the experiments, performed troubleshooting and wrote the manuscript. Y.Z. contributed to aberrant recombination characterization and hGem tag characterization in S. stipitis. M.C. contributed to the initial design of the hGem tag. S.G. contributed to Y. lipolytica PEX10 deletion. C.L. contributed to the construction of H. polymorpha DL1 Δleu2. M.S., S.C., and A.S. contributed to genome sequence analysis. M.G. and L.H. aided in cloning efforts and LINEAR CRISPR editing efficiency determination.

Corresponding author

Correspondence to Zengyi Shao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Chemical Biology thanks Zihe Liu, Eric Young and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Literature survey to determine the most implemented CRISPR platform.

A compilation of 400 instances where studies deployed CRISPR for genome editing revealed that most researchers implement the conventional CRISPR plasmid and donor DNA Platform I. Studies were identified on PubMed using ‘Yeast CRISPR’ and ‘Mammal CRISPR’ in the search query.

Source data

Extended Data Fig. 2 The shortcomings associated with the conventional CRISPR platforms in S. stipitis.

a, The workflow for the conventional CRISPR plasmid and donor DNA platform (Platform I in Extended Data Fig. 1) deployed in S. stipitis, depicting various shortcomings our group has encountered that impeded obtaining correctly edited cells. b, The vector map of the consolidated ‘self-digesting’ plasmid, pCashGem-SD-Xyl2, highlighting the key design components (Platform IV in Extended Data Fig. 1). c, Transformation of UC7 with the pCashGem-SD-Xyl2 plasmid failed to address the shortcomings, with almost all the colonies displaying an episomal GFP expression profile.

Extended Data Fig. 3 Aberrant recombination characterization in S. stipitis.

a, Visual depiction of visibly intense GFP expression and subsequent loss of expression b, Vector map of the pGAOU plasmid, harboring the GAOU fragment flanked by SacI and XhoI restriction sites. The digested GAOU fragment was transformed into UC7. c, Ten strongly intense green colonies observed under the DR46B transilluminator. d, The GFP expression intensity of the ten variants was evaluated by flow cytometry after 36 h of cultivation. Numbers correspond to the mean GFP expression intensity. Among these ten variants, only #5, #7, and #9 exhibited genomic expression of GFP (sharp peaks). The gating methodology was summarized in Supplementary Information.

Extended Data Fig. 4 Verification of ‘pseudo’ plasmids formed by aberrant recombination events in S. stipitis.

a, Transformation of E. coli BW25141 with the five plasmids isolated from S. stipitis variants. b, Enzyme digestion verification of pGAOU-X vectors isolated from E. coli DH5α. Lane 1: M, GeneRuler 1 kb DNA Ladder. Lanes 2 and 3: pGAOU-1 isolates. Lanes 4 and 5: pGAOU-2 isolates. Lanes 6 and 7: pGAOU-3 isolates. Individual restriction digestions for each biological sample were performed once. c, Vector maps of the isolated pGAOU-X plasmids. d, Transformation of UC7 with pGAOU-2 and pGAOU-3. Plates were observed under a DR46B transilluminator.

Extended Data Fig. 5 Design protocol for the LINEAR platform.

a, Cloning of individual LINEAR components. b, Assembly of LINEAR components into a cloning vector. Digestion and gel electrophoresis of the assembled vector yields the LINEAR fragment used in genome editing.

Extended Data Fig. 6 Comparison of the conventional CRISPR platforms versus LINEAR highlights growth deficiency associated with constitutive Cas9 expression.

All platforms utilized hGem tagged-Cas9. Plate pictures were taken at various timepoints post-transformation.

Extended Data Fig. 7 Assessing the contribution of the hGem tag on Xyl2:URA3 LINEAR editing efficiency with 500-bp homology arms.

a, Equimolar quantities of each LINEAR fragment were transformed to S. stipitis UC7. Plate pictures were taken at 48 h post-transformation. Ten colonies were selected at random (circled) for downstream analysis. b, The expected edit outcome on Xyl2 locus. c, PCR products obtained from the primer pair in b using the genomic DNA isolated from the colonies circled in a as templates. Individual PCR reactions for each biological sample were performed once. d, Sequencing of the amplicon associated with Cas9 #4 revealed an erroneous insertion of the donor template at the 5’ end of Xyl2.

Source data

Extended Data Fig. 8 Assessing the contribution of the hGem tag on Xyl2:URA3 LINEAR editing efficiency with 50-bp homology arms.

a, Equimolar quantities of each LINEAR fragment were transformed to S. stipitis UC7. Plate pictures were taken at 48 h post-transformation. Ten colonies selected at random (circled) for downstream analysis. Note that GFP cassette was placed in between sgRNA cassette and the donor DNA as a negative reporter to assess aberrant recombination. The colonies with strong GFP expression levels were false positives. b, The expected edit outcome on Xyl2 locus and the intact locus. c, PCR products obtained from the primer pair in b using the genomic DNA isolated from the colonies circled in a as templates. Individual PCR reactions for each biological sample were performed once. d, Sequencing of the amplicons associated with Cas9-hGem #10 and Cas9 #6 confirmed the presence of the intact wild type Xyl2. e, Sequencing of the amplicon associated with Cas9 #2 revealed an erroneous insertion of the donor template along with a portion of the upstream region of the LINEAR fragment.

Source data

Extended Data Fig. 9 Assessing the contribution of the hGem tag on Xyl2:GSAU LINEAR editing efficiency.

a, Equimolar quantities of each LINEAR fragment were transformed to S. stipitis UC7. Plate pictures were taken at 48 h post-transformation. Ten colonies that displayed genomic GFP expression as visualized on transilluminator selected (circled) for downstream analysis. Slightly different from the design used to achieve Xyl2:URA3 LINEAR editing with 50-bp homology arms, GFP cassette was included in the donor DNA as a positive reporter. b, PCR analysis of the 5’ end of the Xyl2 locus. Due to the large size of the inserted pathway, two reactions were designed to assess the nature of integration at each 500-bp homology arm. PCR products were obtained from the primer pair using the genomic DNA isolated from the colonies circled in a as templates. Individual PCR reactions for each biological sample were performed once. c, PCR analysis of the 3’ end of the Xyl2 locus. Individual PCR reactions for each biological sample were performed once. d, Sequencing of the amplicon associated with Cas9 #9 revealed an erroneous insertion of the downstream portion of the donor template at the 3’ end of Xyl2. Wild type stands for a ~100-bp sequence originating from the wild type Xyl2 locus between the upstream and the downstream homology arms.

Source data

Extended Data Fig. 10 Pathway map depicting the aromatic amino acid pathway and the de novo synthesis of (S)-norcoclaurine.

a, Glycolysis and the pentose phosphate pathway (PPP) channel carbon fluxes into the aromatic amino acid (AAA) pathway. Shikimate can serve as a reporter for assessing AAA pathway flux. Tkt1, Aro4*, Aro1, and Aro7* comprise the four-enzyme, upstream L-tyrosine precursor module. The red lines depict L-tyrosine feedback inhibition on the native Aro4 and Aro7 enzymes. b, (S)-betalamic acid, a product of the biosensing module of TyrH* and DOD, spontaneously condenses with endogenous amino acids to produce yellow pigmented betaxanthins. c, TyrH*, DODC, and NCS comprise the three-enzyme, downstream (S)-norcoclaurine module. Metabolite abbreviations: G6P, glucose-6-phosphate; PEP, phosphoenolpyruvate; E4P, erythrose 4-phosphate; DAHP, 3-deoxy-D-arabino-heptulosonate 7-phosphate; DHQ, 3-dehydroquinic acid; DHS, 3-dehydroshikimate; S3P, shikmate-3-phosphate; EPSP, 5-enolpyruvylshikimate-3-phosphate; L-DOPA, L-3,4-dihydroxyphenylalanine; 4-HPAA, 4-hydroxyphenylacetaldehyde. Enzyme abbreviations: Tkt1, transketolase; Aro4*, DAHP synthase feedback inhibition-resistant mutant (Aro4K220L); Aro1, pentafunctional Aro1 polypeptide; Aro1*, Aro1 mutant (Aro1D900A) with the shikimate kinase domain inactivated, enabling shikimate accumulation; Aro7*, chorismate mutase feedback inhibition-resistant mutant (Aro7G139S); TyrH*, tyrosine hydroxylase mutant (TyrHW13L, F309L) from Berberis vulgaris with low L-DOPA oxidase activity; DOD, L-DOPA dioxygenase from Mirabilis jalapa; DODC, L-DOPA decarboxylase from Pseudomonas putida; NCS, norcoclaurine synthase from Papaver somniferum.

Supplementary information

Supplementary Information

Supplementary Discussions 1–4, Figs. 1–22 and Tables 1–5.

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. 6

Statistical source data.

Source Data Extended Data Fig. 1

Compilation of references.

Source Data Extended Data Fig. 7

Unprocessed gel.

Source Data Extended Data Fig. 8

Unprocessed gel.

Source Data Extended Data Fig. 9

Unprocessed gel.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ploessl, D., Zhao, Y., Cao, M. et al. A repackaged CRISPR platform increases homology-directed repair for yeast engineering. Nat Chem Biol 18, 38–46 (2022). https://doi.org/10.1038/s41589-021-00893-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00893-5

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