1887

Abstract

The discovery and adaptation of CRISPR/Cas systems within molecular biology has provided advances across biological research, agriculture and human health. Genomic manipulation through use of a CRISPR nuclease and programmed guide RNAs has become a common and widely accessible practice. The identification and introduction of new engineered variants and orthologues of Cas9 as well as alternative CRISPR systems such as the type V group have provided additional molecular options for editing. These include distinct PAM requirements, staggered DNA double-strand break formation, and the ability to multiplex guide RNAs from a single expression construct. Use of CRISPR/Cas has allowed for the construction and testing of a powerful genetic architecture known as a gene drive within eukaryotic model systems. Our previous work developed a drive within budding yeast using Cas9. Here, we installed the type V Cas12a (Cpf1) nuclease gene and its corresponding guide RNA to power a highly efficient artificial gene drive in diploid yeast. We examined the consequence of altering guide length or introduction of individual mutational substitutions to the crRNA sequence. Cas12a-dependent gene-drive function required a guide RNA of at least 18 bp and could not tolerate most changes within the 5′ end of the crRNA.

Keyword(s): Cas12a , CRISPR , gene drive and yeast
Funding
This study was supported by the:
  • National Institute of Food and Agriculture (Award Hatch Project 1013520)
    • Principle Award Recipient: GregoryC Finnigan
  • National Institutes of Health (Award P20 GM103418)
    • Principle Award Recipient: NotApplicable
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License.
Loading

Article metrics loading...

/content/journal/acmi/10.1099/acmi.0.000301
2021-12-17
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/acmi/3/12/acmi000301.html?itemId=/content/journal/acmi/10.1099/acmi.0.000301&mimeType=html&fmt=ahah

References

  1. Asmamaw M, Zawdie B. Mechanism and applications of CRISPR/Cas-9-mediated genome editing. Biologics 2021; 15:353–361 [View Article] [PubMed]
    [Google Scholar]
  2. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012; 337:816–821 [View Article] [PubMed]
    [Google Scholar]
  3. Jinek M, East A, Cheng A, Lin S, Ma E et al. RNA-programmed genome editing in human cells. Elife 2013; 2:e00471 [View Article] [PubMed]
    [Google Scholar]
  4. Sternberg SH, Doudna JA. Expanding the biologist’s toolkit with CRISPR-Cas9. Mol Cell 2015; 58:568–574 [View Article] [PubMed]
    [Google Scholar]
  5. Hou Z, Zhang Y, Propson NE, Howden SE, Chu L-F et al. Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc Natl Acad Sci U S A 2013; 110:15644–15649 [View Article] [PubMed]
    [Google Scholar]
  6. Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 2015; 523:481–485 [View Article] [PubMed]
    [Google Scholar]
  7. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 2015; 520:186–191 [View Article] [PubMed]
    [Google Scholar]
  8. Shmakov S, Abudayyeh OO, Makarova KS, Wolf YI, Gootenberg JS et al. Discovery and functional characterization of diverse class 2 CRISPR-Cas systems. Mol Cell 2015; 60:385–397 [View Article] [PubMed]
    [Google Scholar]
  9. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 2015; 163:759–771 [View Article] [PubMed]
    [Google Scholar]
  10. Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J et al. RNA targeting with CRISPR-Cas13. Nature 2017; 550:280–284 [View Article] [PubMed]
    [Google Scholar]
  11. Koonin EV, Makarova KS, Zhang F. Diversity, classification and evolution of CRISPR-Cas systems. Curr Opin Microbiol 2017; 37:67–78 [View Article] [PubMed]
    [Google Scholar]
  12. Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX et al. Rationally engineered Cas9 nucleases with improved specificity. Science 2016; 351:84–88 [View Article] [PubMed]
    [Google Scholar]
  13. Fonfara I, Richter H, Bratovič M, Le Rhun A, Charpentier E. The CRISPR-associated DNA-cleaving enzyme Cpf1 also processes precursor CRISPR RNA. Nature 2016; 532:517–521 [View Article] [PubMed]
    [Google Scholar]
  14. Paul B, Montoya G. CRISPR-Cas12a: Functional overview and applications. Biomed J 2020; 43:8–17 [View Article] [PubMed]
    [Google Scholar]
  15. Godfray HCJ, North A, Burt A. How driving endonuclease genes can be used to combat pests and disease vectors. BMC Biol 2017; 15:81 [View Article] [PubMed]
    [Google Scholar]
  16. Hammond AM, Galizi R. Gene drives to fight malaria: current state and future directions. Pathog Glob Health 2017; 111:412–423 [View Article] [PubMed]
    [Google Scholar]
  17. Bier E. Gene drives gaining speed. Nat Rev Genet 20211–18 [View Article] [PubMed]
    [Google Scholar]
  18. DiCarlo JE, Chavez A, Dietz SL, Esvelt KM, Church GM. Safeguarding CRISPR-Cas9 gene drives in yeast. Nat Biotechnol 2015; 33:1250–1255 [View Article] [PubMed]
    [Google Scholar]
  19. Gantz VM, Jasinskiene N, Tatarenkova O, Fazekas A, Macias VM et al. Highly efficient Cas9-mediated gene drive for population modification of the malaria vector mosquito Anopheles stephensi. Proc Natl Acad Sci U S A 2015; 112:E6736–43 [View Article] [PubMed]
    [Google Scholar]
  20. Hammond A, Galizi R, Kyrou K, Simoni A, Siniscalchi C et al. A CRISPR-Cas9 gene drive system targeting female reproduction in the malaria mosquito vector Anopheles gambiae. Nat Biotechnol 2016; 34:78–83 [View Article] [PubMed]
    [Google Scholar]
  21. Grunwald HA, Gantz VM, Poplawski G, Xu X-RS, Bier E et al. Super-Mendelian inheritance mediated by CRISPR-Cas9 in the female mouse germline. Nature 2019; 566:105–109 [View Article] [PubMed]
    [Google Scholar]
  22. Valderrama JA, Kulkarni SS, Nizet V, Bier E. A bacterial gene-drive system efficiently edits and inactivates a high copy number antibiotic resistance locus. Nat Commun 2019; 10:5726 [View Article] [PubMed]
    [Google Scholar]
  23. Walter M, Verdin E. Viral gene drive in herpesviruses. Nat Commun 2020; 11:4884 [View Article] [PubMed]
    [Google Scholar]
  24. Basgall EM, Goetting SC, Goeckel ME, Giersch RM, Roggenkamp E et al. Gene drive inhibition by the anti-CRISPR proteins AcrIIA2 and AcrIIA4 in Saccharomyces cerevisiae. Microbiology (Reading) 2018; 164:464–474 [View Article] [PubMed]
    [Google Scholar]
  25. Roggenkamp E, Giersch RM, Schrock MN, Turnquist E, Halloran M et al. Tuning CRISPR-Cas9 Gene Drives in Saccharomyces cerevisiae. G3 (Bethesda) 2018; 8:999–1018 [View Article] [PubMed]
    [Google Scholar]
  26. Goeckel ME, Basgall EM, Lewis IC, Goetting SC, Yan Y et al. Modulating CRISPR gene drive activity through nucleocytoplasmic localization of Cas9 in S. cerevisiae. Fungal Biol Biotechnol 2019; 6:2 [View Article] [PubMed]
    [Google Scholar]
  27. Finnigan GC, Thorner J. Complex in vivo ligation using homologous recombination and high-efficiency plasmid rescue from Saccharomyces cerevisiae. Bio Protoc 2015; 5:e1521 [View Article] [PubMed]
    [Google Scholar]
  28. Yan Y, Finnigan GC. Analysis of CRISPR gene drive design in budding yeast. Access Microbiol 2019; 1:e000059 [View Article] [PubMed]
    [Google Scholar]
  29. Finnigan GC, Thorner J. mCAL: a new approach for versatile multiplex action of Cas9 using one sgRNA and loci flanked by a programmed target sequence. G3 (Bethesda) 2016; 6:2147–2156 [View Article] [PubMed]
    [Google Scholar]
  30. Zheng L, Baumann U, Reymond JL. An efficient one-step site-directed and site-saturation mutagenesis protocol. Nucleic Acids Res 2004; 32:e115 [View Article] [PubMed]
    [Google Scholar]
  31. Pawluk A, Amrani N, Zhang Y, Garcia B, Hidalgo-Reyes Y et al. Naturally occurring off-switches for CRISPR-Cas9. Cell 2016; 167:1829–1838 [View Article] [PubMed]
    [Google Scholar]
  32. Rauch BJ, Silvis MR, Hultquist JF, Waters CS, McGregor MJ et al. Inhibition of CRISPR-Cas9 with bacteriophage proteins. Cell 2017; 168:150–158 [View Article] [PubMed]
    [Google Scholar]
  33. Marino ND, Zhang JY, Borges AL, Sousa AA, Leon LM et al. Discovery of widespread type I and type V CRISPR-Cas inhibitors. Science 2018; 362:240–242 [View Article] [PubMed]
    [Google Scholar]
  34. Watters KE, Fellmann C, Bai HB, Ren SM, Doudna JA. Systematic discovery of natural CRISPR-Cas12a inhibitors. Science 2018; 362:236–239 [View Article] [PubMed]
    [Google Scholar]
  35. Swiat MA, Dashko S, den Ridder M, Wijsman M, van der Oost J et al. FnCpf1: a novel and efficient genome editing tool for Saccharomyces cerevisiae. Nucleic Acids Res 2017; 45:12585–12598 [View Article] [PubMed]
    [Google Scholar]
  36. DiCarlo JE, Norville JE, Mali P, Rios X, Aach J et al. Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res 2013; 41:4336–4343 [View Article] [PubMed]
    [Google Scholar]
  37. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 2013; 31:827–832 [View Article] [PubMed]
    [Google Scholar]
  38. Lee K, Mackley VA, Rao A, Chong AT, Dewitt MA et al. Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering. Elife 2017; 6:e25312 [View Article] [PubMed]
    [Google Scholar]
  39. Zheng T, Hou Y, Zhang P, Zhang Z, Xu Y et al. Profiling single-guide RNA specificity reveals a mismatch sensitive core sequence. Sci Rep 2017; 7:40638 [View Article] [PubMed]
    [Google Scholar]
  40. Yan Y, Finnigan GC. Development of a multi-locus CRISPR gene drive system in budding yeast. Sci Rep 2018; 8:17277 [View Article] [PubMed]
    [Google Scholar]
  41. Vella MR, Gunning CE, Lloyd AL, Gould F. Evaluating strategies for reversing CRISPR-Cas9 gene drives. Sci Rep 2017; 7:11038 [View Article] [PubMed]
    [Google Scholar]
  42. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 2013; 154:442–451 [View Article] [PubMed]
    [Google Scholar]
  43. Kweon J, Jang A-H, Kim D-E, Yang JW, Yoon M et al. Fusion guide RNAs for orthogonal gene manipulation with Cas9 and Cpf1. Nat Commun 2017; 8:1723 [View Article] [PubMed]
    [Google Scholar]
  44. Kleinstiver BP, Tsai SQ, Prew MS, Nguyen NT, Welch MM et al. Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells. Nat Biotechnol 2016; 34:869–874 [View Article] [PubMed]
    [Google Scholar]
  45. Swarts DC, van der Oost J, Jinek M. Structural basis for guide RNA processing and seed-dependent DNA targeting by CRISPR-Cas12a. Mol Cell 2017; 66:221–233 [View Article] [PubMed]
    [Google Scholar]
  46. Murugan K, Seetharam AS, Severin AJ, Sashital DG. CRISPR-Cas12a has widespread off-target and dsDNA-nicking effects. J Biol Chem 2020; 295:5538–5553 [View Article] [PubMed]
    [Google Scholar]
  47. Wang Y, Wang M, Zheng T, Hou Y, Zhang P et al. Specificity profiling of CRISPR system reveals greatly enhanced off-target gene editing. Sci Rep 2020; 10:2269 [View Article] [PubMed]
    [Google Scholar]
  48. Creutzburg SCA, Wu WY, Mohanraju P, Swartjes T, Alkan F et al. Good guide, bad guide: spacer sequence-dependent cleavage efficiency of Cas12a. Nucleic Acids Res 2020; 48:3228–3243 [View Article] [PubMed]
    [Google Scholar]
  49. Thyme SB, Akhmetova L, Montague TG, Valen E, Schier AF. Internal guide RNA interactions interfere with Cas9-mediated cleavage. Nat Commun 2016; 7:11750 [View Article] [PubMed]
    [Google Scholar]
  50. Kim D, Kim J, Hur JK, Been KW, Yoon S-H et al. Genome-wide analysis reveals specificities of Cpf1 endonucleases in human cells. Nat Biotechnol 2016; 34:863–868 [View Article] [PubMed]
    [Google Scholar]
  51. Champer J, Reeves R, Oh SY, Liu C, Liu J et al. Novel CRISPR/Cas9 gene drive constructs reveal insights into mechanisms of resistance allele formation and drive efficiency in genetically diverse populations. PLoS Genet 2017; 13:e1006796 [View Article] [PubMed]
    [Google Scholar]
  52. Hammond AM, Kyrou K, Bruttini M, North A, Galizi R et al. The creation and selection of mutations resistant to a gene drive over multiple generations in the malaria mosquito. PLoS Genet 2017; 13:e1007039 [View Article] [PubMed]
    [Google Scholar]
  53. Noble C, Olejarz J, Esvelt KM, Church GM, Nowak MA. Evolutionary dynamics of CRISPR gene drives. Sci Adv 2017; 3:e1601964 [View Article] [PubMed]
    [Google Scholar]
  54. Unckless RL, Clark AG, Messer PW. Evolution of resistance against CRISPR/Cas9 Gene Drive. Genetics 2017; 205:827–841 [View Article] [PubMed]
    [Google Scholar]
  55. KaramiNejadRanjbar M, Eckermann KN, Ahmed HMM, Sánchez C HM, Dippel S et al. Consequences of resistance evolution in a Cas9-based sex conversion-suppression gene drive for insect pest management. Proc Natl Acad Sci U S A 2018; 115:6189–6194 [View Article] [PubMed]
    [Google Scholar]
  56. Champer J, Liu J, Oh SY, Reeves R, Luthra A et al. Reducing resistance allele formation in CRISPR gene drive. Proc Natl Acad Sci U S A 2018; 115:5522–5527 [View Article] [PubMed]
    [Google Scholar]
  57. Kyrou K, Hammond AM, Galizi R, Kranjc N, Burt A et al. A CRISPR-Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes. Nat Biotechnol 2018; 36:1062–1066 [View Article] [PubMed]
    [Google Scholar]
  58. Champer J, Lee E, Yang E, Liu C, Clark AG et al. A toxin-antidote CRISPR gene drive system for regional population modification. Nat Commun 2020; 11:1082 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/acmi/10.1099/acmi.0.000301
Loading
/content/journal/acmi/10.1099/acmi.0.000301
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error