Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2022 Feb 18;65(4):660–700. doi: 10.1007/s11427-021-2057-0

Gene editing and its applications in biomedicine

Guanglei Li 1,#, Xiangyang Li 1,#, Songkuan Zhuang 2,#, Liren Wang 3,#, Yifan Zhu 3,#, Yangcan Chen 4,5,6,#, Wen Sun 4,6,#, Zeguang Wu 7,#, Zhuo Zhou 7,#, Jia Chen 8,, Xingxu Huang 1,, Jin Wang 2,, Dali Li 3,, Wei Li 4,5,6,9,10,, Haoyi Wang 4,5,6,, Wensheng Wei 7,
PMCID: PMC8889061  PMID: 35235150

Abstract

The steady progress in genome editing, especially genome editing based on the use of clustered regularly interspaced short palindromic repeats (CRISPR) and programmable nucleases to make precise modifications to genetic material, has provided enormous opportunities to advance biomedical research and promote human health. The application of these technologies in basic biomedical research has yielded significant advances in identifying and studying key molecular targets relevant to human diseases and their treatment. The clinical translation of genome editing techniques offers unprecedented biomedical engineering capabilities in the diagnosis, prevention, and treatment of disease or disability. Here, we provide a general summary of emerging biomedical applications of genome editing, including open challenges. We also summarize the tools of genome editing and the insights derived from their applications, hoping to accelerate new discoveries and therapies in biomedicine.

Keywords: gene editing, CRISPR, high-throughput functional genomics, diagnostics, animal model, therapeutics

Acknowledgements

This work was supported by the National Natural Science Foundation of China (81830004, 31922046, 31770057, 31722036, 31930016, 31870893), the Sanming Project of Medicine in Shenzhen (SZSM202011017), the National Key Research and Development Program of China (2018YFA0801401, 2019YFA0110800, 2018YFA0107703, 2019YFA0110000, 2020YFA0707800, 2020YFA0707600), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA16030403, XDA16010503), Beijing Municipal Science & Technology Commission (Z181100001318009), Beijing Advanced Innovation Center for Genomics at Peking University and the Peking-Tsinghua Center for Life Sciences, and the National Major Science & Technology Project for Control and Prevention of Major Infectious Diseases in China (2018ZX10301401). We are grateful to Dr. Gaofeng Fan from ShanghaiTech University for manuscript editing.

Footnotes

Compliance and ethics

The author(s) declare that they have no conflict of interest.

Contributed equally to this work

Contributor Information

Jia Chen, Email: chenjia@shanghaitech.edu.cn.

Xingxu Huang, Email: huangxx@shanghaitech.edu.cn.

Jin Wang, Email: wangj01@hotmail.com.

Dali Li, Email: dlli@bio.ecnu.edu.cn.

Wei Li, Email: liwei@ioz.ac.cn.

Haoyi Wang, Email: wanghaoyi@ioz.ac.cn.

Wensheng Wei, Email: wswei@pku.edu.cn.

References

  1. Abudayyeh OO, Gootenberg JS, Konermann S, Joung J, Slaymaker IM, Cox DBT, Shmakov S, Makarova KS, Semenova E, Minakhin L, et al. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science. 2016;353:aaf5573. doi: 10.1126/science.aaf5573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Abudayyeh OO, Gootenberg JS, Essletzbichler P, Han S, Joung J, Belanto JJ, Verdine V, Cox DBT, Kellner MJ, Regev A, et al. RNA targeting with CRISPR-Cas13. Nature. 2017;550:280–284. doi: 10.1038/nature24049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Abudayyeh OO, Gootenberg JS, Franklin B, Koob J, Kellner MJ, Ladha A, Joung J, Kirchgatterer P, Cox DBT, Zhang F. A cytosine deaminase for programmable single-base RNA editing. Science. 2019;365:382–386. doi: 10.1126/science.aax7063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ackerman CM, Myhrvold C, Thakku SG, Freije CA, Metsky HC, Yang DK, Ye SH, Boehm CK, Kosoko-Thoroddsen TSF, Kehe J, et al. Massively multiplexed nucleic acid detection with Cas13. Nature. 2020;582:277–282. doi: 10.1038/s41586-020-2279-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Adamson B, Norman TM, Jost M, Cho MY, Nuñez JK, Chen Y, Villalta JE, Gilbert LA, Horlbeck MA, Hein MY, et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell. 2016;167:1867–1882. doi: 10.1016/j.cell.2016.11.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Adikusuma F, Piltz S, Corbett MA, Turvey M, McColl SR, Helbig KJ, Beard MR, Hughes J, Pomerantz RT, Thomas PQ. Large deletions induced by Cas9 cleavage. Nature. 2018;560:E8–E9. doi: 10.1038/s41586-018-0380-z. [DOI] [PubMed] [Google Scholar]
  7. Aiuti A, Slavin S, Aker M, Ficara F, Deola S, Mortellaro A, Morecki S, Andolfi G, Tabucchi A, Carlucci F, et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science. 2002;296:2410–2413. doi: 10.1126/science.1070104. [DOI] [PubMed] [Google Scholar]
  8. Aiuti A, Cattaneo F, Galimberti S, Benninghoff U, Cassani B, Callegaro L, Scaramuzza S, Andolfi G, Mirolo M, Brigida I, et al. Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med. 2009;360:447–458. doi: 10.1056/NEJMoa0805817. [DOI] [PubMed] [Google Scholar]
  9. Akinc A, Querbes W, De S, Qin J, Frank-Kamenetsky M, Jayaprakash KN, Jayaraman M, Rajeev KG, Cantley WL, Dorkin JR, et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol Ther. 2010;18:1357–1364. doi: 10.1038/mt.2010.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Al-Shayeb B, Sachdeva R, Chen LX, Ward F, Munk P, Devoto A, Castelle CJ, Olm MR, Bouma-Gregson K, Amano Y, et al. Clades of huge phages from across Earth’s ecosystems. Nature. 2020;578:425–431. doi: 10.1038/s41586-020-2007-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Allen AG, Chung CH, Atkins A, Dampier W, Khalili K, Nonnemacher MR, Wigdahl B. Gene editing of HIV-1 co-receptors to prevent and/or cure virus infection. Front Microbiol. 2018;9:2940. doi: 10.3389/fmicb.2018.02940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Aman R, Mahas A, Marsic T, Hassan N, Mahfouz MM. Efficient, rapid, and sensitive detection of plant RNA viruses with one-pot RT-RPA-CRISPR/Cas12a assay. Front Microbiol. 2020;11:610872. doi: 10.3389/fmicb.2020.610872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Anders C, Niewoehner O, Duerst A, Jinek M. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature. 2014;513:569–573. doi: 10.1038/nature13579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 2019;576:149–157. doi: 10.1038/s41586-019-1711-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Anzalone AV, Koblan LW, Liu DR. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol. 2020;38:824–844. doi: 10.1038/s41587-020-0561-9. [DOI] [PubMed] [Google Scholar]
  16. Auer TO, Duroure K, De Cian A, Concordet JP, Del Bene F. Highly efficient CRISPR/Cas9-mediated knock-in in zebrafish by homology-independent DNA repair. Genome Res. 2014;24:142–153. doi: 10.1101/gr.161638.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bai J, Lin H, Li H, Zhou Y, Liu J, Zhong G, Wu L, Jiang W, Du H, Yang J, et al. Cas12a-based on-site and rapid nucleic acid detection of African swine fever. Front Microbiol. 2019;10:2830. doi: 10.3389/fmicb.2019.02830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bainbridge JWB, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, Viswanathan A, Holder GE, Stockman A, Tyler N, et al. Effect of gene therapy on visual function in Leber’s congenital amaurosis. N Engl J Med. 2008;358:2231–2239. doi: 10.1056/NEJMoa0802268. [DOI] [PubMed] [Google Scholar]
  19. Bainbridge JWB, Mehat MS, Sundaram V, Robbie SJ, Barker SE, Ripamonti C, Georgiadis A, Mowat FM, Beattie SG, Gardner P J, et al. Long-term effect of gene therapy on Leber’s congenital amaurosis. N Engl J Med. 2015;372:1887–1897. doi: 10.1056/NEJMoa1414221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bakondi B, Lv W, Lu B, Jones MK, Tsai Y, Kim KJ, Levy R, Akhtar AA, Breunig JJ, Svendsen CN, et al. In vivo CRISPR/Cas9 gene editing corrects retinal dystrophy in the S334ter-3 rat model of autosomal dominant retinitis pigmentosa. Mol Ther. 2016;24:556–563. doi: 10.1038/mt.2015.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Balderston S, Taulbee JJ, Celaya E, Fung K, Jiao A, Smith K, Hajian R, Gasiunas G, Kutanovas S, Kim D, et al. Discrimination of single-point mutations in unamplified genomic DNA via Cas9 immobilized on a graphene field-effect transistor. Nat Biomed Eng. 2021;5:713–725. doi: 10.1038/s41551-021-00706-z. [DOI] [PubMed] [Google Scholar]
  22. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, Romero DA, Horvath P. CRISPR provides acquired resistance against viruses in prokaryotes. Science. 2007;315:1709–1712. doi: 10.1126/science.1138140. [DOI] [PubMed] [Google Scholar]
  23. Barzel A, Paulk NK, Shi Y, Huang Y, Chu K, Zhang F, Valdmanis PN, Spector LP, Porteus MH, Gaensler KM, et al. Promoterless gene targeting without nucleases ameliorates haemophilia B in mice. Nature. 2015;517:360–364. doi: 10.1038/nature13864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Batra R, Nelles DA, Pirie E, Blue SM, Marina RJ, Wang H, Chaim IA, Thomas JD, Zhang N, Nguyen V, et al. Elimination of toxic microsatellite repeat expansion RNA by RNA-targeting Cas9. Cell. 2017;170:899–912. doi: 10.1016/j.cell.2017.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Beerli RR, Barbas CF. Engineering polydactyl zinc-finger transcription factors. Nat Biotechnol. 2002;20:135–141. doi: 10.1038/nbt0202-135. [DOI] [PubMed] [Google Scholar]
  26. Benjamin R, Graham C, Yallop D, Jozwik A, Mirci-Danicar OC, Lucchini G, Pinner D, Jain N, Kantarjian H, Boissel N, et al. Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B-cell acute lymphoblastic leukaemia: results of two phase 1 studies. Lancet. 2020;396:1885–1894. doi: 10.1016/S0140-6736(20)32334-5. [DOI] [PubMed] [Google Scholar]
  27. Bennett J, Ashtari M, Wellman J, Marshall KA, Cyckowski LL, Chung DC, McCague S, Pierce EA, Chen Y, Bennicelli JL, et al. AAV2 gene therapy readministration in three adults with congenital blindness. Sci Transl Med. 2012;4:120ra115. doi: 10.1126/scitranslmed.3002865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Bennett J, Wellman J, Marshall KA, McCague S, Ashtari M, DiStefano-Pappas J, Elci OU, Chung DC, Sun J, Wright JF, et al. Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial. Lancet. 2016;388:661–672. doi: 10.1016/S0140-6736(16)30371-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bibikova M, Golic M, Golic KG, Carroll D. Targeted chromosomal cleavage and mutagenesis in Drosophila using zinc-finger nucleases. Genetics. 2002;161:1169–1175. doi: 10.1093/genetics/161.3.1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Billon P, Bryant EE, Joseph SA, Nambiar TS, Hayward SB, Rothstein R, Ciccia A. CRISPR-mediated base editing enables efficient disruption of eukaryotic genes through induction of STOP codons. Mol Cell. 2017;67:1068–1079. doi: 10.1016/j.molcel.2017.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Bjurström CF, Mojadidi M, Phillips J, Kuo C, Lai S, Lill GR, Cooper A, Kaufman M, Urbinati F, Wang X, et al. Reactivating fetal hemoglobin expression in human adult erythroblasts through BCL11A Knockdown Using Targeted Endonucleases. Mol Ther Nucleic Acids. 2016;5:e351. doi: 10.1038/mtna.2016.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Bloom K, Ely A, Mussolino C, Cathomen T, Arbuthnot P. Inactivation of hepatitis B virus replication in cultured cells and in vivo with engineered transcription activator-like effector nucleases. Mol Ther. 2013;21:1889–1897. doi: 10.1038/mt.2013.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Boch J, Bonas U. Xanthomonas AvrBs3 family-type III effectors: discovery and function. Annu Rev Phytopathol. 2010;48:419–436. doi: 10.1146/annurev-phyto-080508-081936. [DOI] [PubMed] [Google Scholar]
  34. Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, Lahaye T, Nickstadt A, Bonas U. Breaking the code of DNA binding specificity of TAL-type III effectors. Science. 2009;326:1509–1512. doi: 10.1126/science.1178811. [DOI] [PubMed] [Google Scholar]
  35. Boettcher M, Tian R, Blau JA, Markegard E, Wagner RT, Wu D, Mo X, Biton A, Zaitlen N, Fu H, et al. Dual gene activation and knockout screen reveals directional dependencies in genetic networks. Nat Biotechnol. 2018;36:170–178. doi: 10.1038/nbt.4062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology. 2005;151:2551–2561. doi: 10.1099/mic.0.28048-0. [DOI] [PubMed] [Google Scholar]
  37. Boye SE, Huang WC, Roman AJ, Sumaroka A, Boye SL, Ryals R C, Olivares MB, Ruan Q, Tucker BA, Stone EM, et al. Natural history of cone disease in the murine model of Leber congenital amaurosis due to CEP 290 mutation: determining the timing and expectation of therapy. PLoS ONE. 2014;9:e92928. doi: 10.1371/journal.pone.0092928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Broughton JP, Deng X, Yu G, Fasching CL, Servellita V, Singh J, Miao X, Streithorst JA, Granados A, Sotomayor-Gonzalez A, et al. CRISPR-Cas12-based detection of SARS-CoV-2. Nat Biotechnol. 2020;38:870–874. doi: 10.1038/s41587-020-0513-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Burr ML, Sparbier CE, Chan YC, Williamson JC, Woods K, Beavis PA, Lam EYN, Henderson MA, Bell CC, Stolzenburg S, et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature. 2017;549:101–105. doi: 10.1038/nature23643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Butler JR, Martens GR, Estrada JL, Reyes LM, Ladowski JM, Galli C, Perota A, Cunningham CM, Tector M, Joseph Tector A. Silencing porcine genes significantly reduces human-anti-pig cytotoxicity profiles: an alternative to direct complement regulation. Transgenic Res. 2016;25:751–759. doi: 10.1007/s11248-016-9958-0. [DOI] [PubMed] [Google Scholar]
  41. Cai P, Otten ABC, Cheng B, Ishii MA, Zhang W, Huang B, Qu K, Sun BK. A genome-wide long noncoding RNA CRISPRi screen identifies PRANCR as a novel regulator of epidermal homeostasis. Genome Res. 2020;30:22–34. doi: 10.1101/gr.251561.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Cai Y, Cheng T, Yao Y, Li X, Ma Y, Li L, Zhao H, Bao J, Zhang M, Qiu Z, et al. In vivo genome editing rescues photoreceptor degeneration via a Cas9/RecA-mediated homology-directed repair pathway. Sci Adv. 2019;5:eaav3335. doi: 10.1126/sciadv.aav3335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE, Shalem O, Chen DD, Schupp PG, Vinjamur DS, Garcia SP, et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature. 2015;527:192–197. doi: 10.1038/nature15521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Carreras A, Pane LS, Nitsch R, Madeyski-Bengtson K, Porritt M, Akcakaya P, Taheri-Ghahfarokhi A, Ericson E, Bjursell M, Perez-Alcazar M, et al. In vivo genome and base editing of a human PCSK9 knock-in hypercholesterolemic mouse model. BMC Biol. 2019;17:4. doi: 10.1186/s12915-018-0624-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Carroll KJ, Makarewich CA, McAnally J, Anderson DM, Zentilin L, Liu N, Giacca M, Bassel-Duby R, Olson EN. A mouse model for adult cardiac-specific gene deletion with CRISPR/Cas9. Proc Natl Acad Sci USA. 2016;113:338–343. doi: 10.1073/pnas.1523918113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Ceccaldi R, Rondinelli B, D’Andrea AD. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 2016;26:52–64. doi: 10.1016/j.tcb.2015.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Cermak T, Doyle EL, Christian M, Wang L, Zhang Y, Schmidt C, Baller JA, Somia NV, Bogdanove AJ, Voytas DF. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 2011;39:e82. doi: 10.1093/nar/gkr218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Chadwick AC, Wang X, Musunuru K. In vivo base editing of PCSK9 (proprotein convertase subtilisin/kexin type 9) as a therapeutic alternative to genome editing. Arterioscler Thromb Vasc Biol. 2017;37:1741–1747. doi: 10.1161/ATVBAHA.117.309881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Chang N, Sun C, Gao L, Zhu D, Xu X, Zhu X, Xiong JW, Xi JJ. Genome editing with RNA-guided Cas9 nuclease in zebrafish embryos. Cell Res. 2013;23:465–472. doi: 10.1038/cr.2013.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Charlesworth CT, Deshpande PS, Dever DP, Camarena J, Lemgart VT, Cromer MK, Vakulskas CA, Collingwood MA, Zhang L, Bode NM, et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat Med. 2019;25:249–254. doi: 10.1038/s41591-018-0326-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Chen H, Shi M, Gilam A, Zheng Q, Zhang Y, Afrikanova I, Li J, Gluzman Z, Jiang R, Kong LJ, et al. Hemophilia A ameliorated in mice by CRISPR-based in vivo genome editing of human factor VIII. Sci Rep. 2019;9:16838. doi: 10.1038/s41598-019-53198-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Chen J, Yang B, Yang L. To BE or not to BE, that is the question. Nat Biotechnol. 2019;37:520–522. doi: 10.1038/s41587-019-0119-x. [DOI] [PubMed] [Google Scholar]
  53. Chen J, Zhang W, Lin J, Wang F, Wu M, Chen C, Zheng Y, Peng X, Li J, Yuan Z. An efficient antiviral strategy for targeting hepatitis B virus genome using transcription activator-like effector nucleases. Mol Ther. 2014;22:303–311. doi: 10.1038/mt.2013.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Chen JS, Dagdas YS, Kleinstiver BP, Welch MM, Sousa AA, Harrington LB, Sternberg SH, Joung JK, Yildiz A, Doudna JA. Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature. 2017;550:407–410. doi: 10.1038/nature24268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Chen JS, Ma E, Harrington LB, Da Costa M, Tian X, Palefsky JM, Doudna JA. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science. 2018;360:436–439. doi: 10.1126/science.aar6245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Chen S, Sanjana NE, Zheng K, Shalem O, Lee K, Shi X, Scott D A, Song J, Pan JQ, Weissleder R, et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell. 2015;160:1246–1260. doi: 10.1016/j.cell.2015.02.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Chen Y, Zheng Y, Kang Y, Yang W, Niu Y, Guo X, Tu Z, Si C, Wang H, Xing R, et al. Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. Hum Mol Genet. 2015;24:3764–3774. doi: 10.1093/hmg/ddv120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Chen Y, Niu Y, Ji W. Genome editing in nonhuman primates: approach to generating human disease models. J Intern Med. 2016;280:246–251. doi: 10.1111/joim.12469. [DOI] [PubMed] [Google Scholar]
  59. Chen Y, Yu J, Niu Y, Qin D, Liu H, Li G, Hu Y, Wang J, Lu Y, Kang Y, et al. Modeling rett syndrome using TALEN-edited MECP2 mutant cynomolgus monkeys. Cell. 2017;169:945–955. doi: 10.1016/j.cell.2017.04.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Chen Z, Arai E, Khan O, Zhang Z, Ngiow SF, He Y, Huang H, Manne S, Cao Z, Baxter AE, et al. In vivo CD8+ T cell CRISPR screening reveals control by Fli1 in infection and cancer. Cell. 2021;184:1262–1280. doi: 10.1016/j.cell.2021.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Cheng L, Li Y, Qi Q, Xu P, Feng R, Palmer L, Chen J, Wu R, Yee T, Zhang J, et al. Single-nucleotide-level mapping of DNA regulatory elements that control fetal hemoglobin expression. Nat Genet. 2021;53:869–880. doi: 10.1038/s41588-021-00861-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Chertow DS. Next-generation diagnostics with CRISPR. Science. 2018;360:381–382. doi: 10.1126/science.aat4982. [DOI] [PubMed] [Google Scholar]
  63. Chesselet MF, Carmichael ST. Animal models of neurological disorders. Neurotherapeutics. 2012;9:241–244. doi: 10.1007/s13311-012-0118-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Chiu W, Lin TY, Chang YC, Isahwan-Ahmad Mulyadi Lai H, Lin S C, Ma C, Yarmishyn AA, Lin SC, Chang KJ, Chou YB, et al. An update on gene therapy for inherited retinal dystrophy: experience in Leber congenital amaurosis clinical trials. Int J Mol Sci. 2021;22:4534. doi: 10.3390/ijms22094534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Choi, J., Chen, W., Suiter, C.C., Lee, C., Chardon, F.M., Yang, W., Leith, A., Daza, R.M., Martin, B., and Shendure, J. (2021). Precise genomic deletions using paired prime editing. Nat Biotechnol doi: 10.1038/s41587-021-01025-z. [DOI] [PMC free article] [PubMed]
  66. Choo Y, Sánchez-García I, Klug A. In vivo repression by a site-specific DNA-binding protein designed against an oncogenic sequence. Nature. 1994;372:642–645. doi: 10.1038/372642a0. [DOI] [PubMed] [Google Scholar]
  67. Choong CJ, Baba K, Mochizuki H. Gene therapy for neurological disorders. Expert Opin Biol Ther. 2016;16:143–159. doi: 10.1517/14712598.2016.1114096. [DOI] [PubMed] [Google Scholar]
  68. Chow RD, Chen S. Cancer CRISPR screens in vivo. Trends Cancer. 2018;4:349–358. doi: 10.1016/j.trecan.2018.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Chow RD, Guzman CD, Wang G, Schmidt F, Youngblood MW, Ye L, Errami Y, Dong MB, Martinez MA, Zhang S, et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat Neurosci. 2017;20:1329–1341. doi: 10.1038/nn.4620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hummel A, Bogdanove AJ, Voytas DF. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics. 2010;186:757–761. doi: 10.1534/genetics.110.120717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Cibelli JB, Gurdon JB. Custom-made oocytes to clone non-human primates. Cell. 2018;172:647–649. doi: 10.1016/j.cell.2018.01.030. [DOI] [PubMed] [Google Scholar]
  72. Cideciyan AV, Hauswirth WW, Aleman TS, Kaushal S, Schwartz S B, Boye SL, Windsor EAM, Conlon TJ, Sumaroka A, Pang JJ, et al. Human RPE65 gene therapy for Leber congenital amaurosis: persistence of early visual improvements and safety at 1 year. Hum Gene Ther. 2009;20:999–1004. doi: 10.1089/hum.2009.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Cimprich KA, Cortez D. ATR: an essential regulator of genome integrity. Nat Rev Mol Cell Biol. 2008;9:616–627. doi: 10.1038/nrm2450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339:819–823. doi: 10.1126/science.1231143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Conway A, Mendel M, Kim K, McGovern K, Boyko A, Zhang L, Miller JC, DeKelver RC, Paschon DE, Mui BL, et al. Non-viral delivery of zinc finger nuclease mRNA enables highly efficient in vivo genome editing of multiple therapeutic gene targets. Mol Ther. 2019;27:866–877. doi: 10.1016/j.ymthe.2019.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Cooper ML, Choi J, Staser K, Ritchey JK, Devenport JM, Eckardt K, Rettig MP, Wang B, Eissenberg LG, Ghobadi A, et al. An “off-the-shelf” fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia. 2018;32:1970–1983. doi: 10.1038/s41375-018-0065-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Corman, V.M., Landt, O., Kaiser, M., Molenkamp, R., Meijer, A., Chu, D. K., Bleicker, T., Brünink, S., Schneider, J., Schmidt, M.L., et al. (2020). Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Eurosurveillance 25. [DOI] [PMC free article] [PubMed]
  78. Cortez JT, Montauti E, Shifrut E, Gatchalian J, Zhang Y, Shaked O, Xu Y, Roth TL, Simeonov DR, Zhang Y, et al. CRISPR screen in regulatory T cells reveals modulators of Foxp3. Nature. 2020;582:416–420. doi: 10.1038/s41586-020-2246-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Cox DBT, Platt RJ, Zhang F. Therapeutic genome editing: prospects and challenges. Nat Med. 2015;21:121–131. doi: 10.1038/nm.3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Cox DBT, Gootenberg JS, Abudayyeh OO, Franklin B, Kellner M J, Joung J, Zhang F. RNA editing with CRISPR-Cas13. Science. 2017;358:1019–1027. doi: 10.1126/science.aaq0180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Cradick TJ, Keck K, Bradshaw S, Jamieson AC, McCaffrey AP. Zinc-finger nucleases as a novel therapeutic strategy for targeting hepatitis B virus DNAs. Mol Ther. 2010;18:947–954. doi: 10.1038/mt.2010.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, Lu Y, Tang Z, Zhang H, Hindy G, et al. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med. 2014;371:22–31. doi: 10.1056/NEJMoa1307095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Cuella-Martin R, Hayward SB, Fan X, Chen X, Huang JW, Taglialatela A, Leuzzi G, Zhao J, Rabadan R, Lu C, et al. Functional interrogation of DNA damage response variants with base editing screens. Cell. 2021;184:1081–1097. doi: 10.1016/j.cell.2021.01.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Cui C, Liu Y, Gerloff D, Rohde C, Pauli C, Köhn M, Misiak D, Oellerich T, Schwartz S, Schmidt LH, et al. NOP10 predicts lung cancer prognosis and its associated small nucleolar RNAs drive proliferation and migration. Oncogene. 2021;40:909–921. doi: 10.1038/s41388-020-01570-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Cyranoski D. First monkeys cloned with technique that made Dolly the sheep. Nature. 2018;553:387–388. doi: 10.1038/d41586-018-01027-z. [DOI] [PubMed] [Google Scholar]
  86. Czernik M, Anzalone DA, Palazzese L, Oikawa M, Loi P. Somatic cell nuclear transfer: failures, successes and the challenges ahead. Int J Dev Biol. 2019;63:123–130. doi: 10.1387/ijdb.180324mc. [DOI] [PubMed] [Google Scholar]
  87. Dai Y, Somoza RA, Wang L, Welter JF, Li Y, Caplan AI, Liu CC. Exploring the trans-cleavage activity of CRISPR-Cas12a (cpf1) for the development of a universal electrochemical biosensor. Angew Chem Int Ed. 2019;58:17399–17405. doi: 10.1002/anie.201910772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Daponte V, Tylzanowski P, Forlino A. Appendage regeneration in vertebrates: what makes this possible? Cells. 2021;10:242. doi: 10.3390/cells10020242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. den Hollander AI, Koenekoop RK, Yzer S, Lopez I, Arends ML, Voesenek KEJ, Zonneveld MN, Strom TM, Meitinger T, Brunner HG, et al. Mutations in the CEP290 (NPHP6) gene are a frequent cause of Leber congenital amaurosis. Am J Hum Genet. 2006;79:556–561. doi: 10.1086/507318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Deng W, Shi X, Tjian R, Lionnet T, Singer RH. CASFISH: CRISPR/Cas9-mediated in situ labeling of genomic loci in fixed cells. Proc Natl Acad Sci USA. 2015;112:11870–11875. doi: 10.1073/pnas.1515692112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Deriano L, Roth DB. Modernizing the nonhomologous end-joining repertoire: alternative and classical NHEJ share the stage. Annu Rev Genet. 2013;47:433–455. doi: 10.1146/annurev-genet-110711-155540. [DOI] [PubMed] [Google Scholar]
  92. Dersh D, Phelan JD, Gumina ME, Wang B, Arbuckle JH, Holly J, Kishton RJ, Markowitz TE, Seedhom MO, Fridlyand N, et al. Genome-wide screens identify lineage- and tumor-specific genes modulating MHC-I- and MHC-II-restricted immunosurveillance of human lymphomas. Immunity. 2021;54:116–131. doi: 10.1016/j.immuni.2020.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, Pavel-Dinu M, Saxena N, Wilkens AB, Mantri S, et al. CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature. 2016;539:384–389. doi: 10.1038/nature20134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Diao Y, Li B, Meng Z, Jung I, Lee AY, Dixon J, Maliskova L, Guan KL, Shen Y, Ren B. A new class of temporarily phenotypic enhancers identified by CRISPR/Cas9-mediated genetic screening. Genome Res. 2016;26:397–405. doi: 10.1101/gr.197152.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Diao Y, Fang R, Li B, Meng Z, Yu J, Qiu Y, Lin KC, Huang H, Liu T, Marina RJ, et al. A tiling-deletion-based genetic screen for cis-regulatory element identification in mammalian cells. Nat Methods. 2017;14:629–635. doi: 10.1038/nmeth.4264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Didigu CA, Wilen CB, Wang J, Duong J, Secreto AJ, Danet-Desnoyers GA, Riley JL, Gregory PD, June CH, Holmes MC, et al. Simultaneous zinc-finger nuclease editing of the HIV coreceptors ccr5 and cxcr4 protects CD4+ T cells from HIV-1 infection. Blood. 2014;123:61–69. doi: 10.1182/blood-2013-08-521229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Ding Q, Strong A, Patel KM, Ng SL, Gosis BS, Regan SN, Cowan CA, Rader DJ, Musunuru K. Permanent alteration of PCSK9 with in vivo CRISPR-Cas9 genome editing. Circ Res. 2014;115:488–492. doi: 10.1161/CIRCRESAHA.115.304351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Ding W, Hu Z, Zhu D, Jiang X, Yu L, Wang X, Zhang C, Wang L, Ji T, Li K, et al. Zinc finger nucleases targeting the human papillomavirus E7 oncogene induce E7 disruption and a transformed phenotype in HPV16/18-positive cervical cancer cells. Clin Cancer Res. 2014;20:6495–6503. doi: 10.1158/1078-0432.CCR-14-0250. [DOI] [PubMed] [Google Scholar]
  99. Ding X, Yin K, Li Z, Lalla RV, Ballesteros E, Sfeir MM, Liu C. Ultrasensitive and visual detection of SARS-CoV-2 using all-in-one dual CRISPR-Cas12a assay. Nat Commun. 2020;11:4711. doi: 10.1038/s41467-020-18575-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Dixit A, Parnas O, Li B, Chen J, Fulco CP, Jerby-Arnon L, Marjanovic ND, Dionne D, Burks T, Raychowdhury R, et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell. 2016;167:1853–1866. doi: 10.1016/j.cell.2016.11.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Doman JL, Raguram A, Newby GA, Liu DR. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat Biotechnol. 2020;38:620–628. doi: 10.1038/s41587-020-0414-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Dong C, Qu L, Wang H, Wei L, Dong Y, Xiong S. Targeting hepatitis B virus cccDNA by CRISPR/Cas9 nuclease efficiently inhibits viral replication. Antiviral Res. 2015;118:110–117. doi: 10.1016/j.antiviral.2015.03.015. [DOI] [PubMed] [Google Scholar]
  103. Dong MB, Wang G, Chow RD, Ye L, Zhu L, Dai X, Park JJ, Kim HR, Errami Y, Guzman CD, et al. Systematic immunotherapy target discovery using genome-scale in vivo CRISPR screens in CD8 T cells. Cell. 2019;178:1189–1204. doi: 10.1016/j.cell.2019.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Dow LE, Lowe SW. Life in the fast lane: mammalian disease models in the genomics era. Cell. 2012;148:1099–1109. doi: 10.1016/j.cell.2012.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Dow LE, Fisher J, O’Rourke KP, Muley A, Kastenhuber ER, Livshits G, Tschaharganeh DF, Socci ND, Lowe SW. Inducible in vivo genome editing with CRISPR-Cas9. Nat Biotechnol. 2015;33:390–394. doi: 10.1038/nbt.3155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Dunbar, C.E., High, K.A., Joung, J.K., Kohn, D.B., Ozawa, K., and Sadelain, M. (2018). Gene therapy comes of age. Science 359. [DOI] [PubMed]
  107. East-Seletsky A, O’Connell MR, Knight SC, Burstein D, Cate JHD, Tjian R, Doudna JA. Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature. 2016;538:270–273. doi: 10.1038/nature19802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Ebbesen KK, Kjems J, Hansen TB. Circular RNAs: identification, biogenesis and function. Biochim Biophys Acta. 2016;1859:163–168. doi: 10.1016/j.bbagrm.2015.07.007. [DOI] [PubMed] [Google Scholar]
  109. Esrick EB, Lehmann LE, Biffi A, Achebe M, Brendel C, Ciuculescu MF, Daley H, MacKinnon B, Morris E, Federico A, et al. Post-transcriptional genetic silencing of BCL11A to treat sickle cell disease. N Engl J Med. 2021;384:205–215. doi: 10.1056/NEJMoa2029392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Feng X, Bednarz AL, Colloms SD. Precise targeted integration by a chimaeric transposase zinc-finger fusion protein. Nucleic Acids Res. 2010;38:1204–1216. doi: 10.1093/nar/gkp1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Fischer, K., Rieblinger, B., Hein, R., Sfriso, R., Zuber, J., Fischer, A., Klinger, B., Liang, W., Flisikowski, K., Kurome, M., et al. (2020). Viable pigs after simultaneous inactivation of porcine MHC class I and three xenoreactive antigen genes GGTA1, CMAH and B4GALNT2. Xenotransplantation 27. [DOI] [PubMed]
  112. Fozouni P, Son S, Díaz de León Derby M, Knott GJ, Gray CN, D’Ambrosio MV, Zhao C, Switz NA, Kumar GR, Stephens SI, et al. Amplification-free detection of SARS-CoV-2 with CRISPR-Cas13a and mobile phone microscopy. Cell. 2021;184:323–333. doi: 10.1016/j.cell.2020.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Frangoul H, Altshuler D, Cappellini MD, Chen YS, Domm J, Eustace BK, Foell J, de la Fuente J, Grupp S, Handgretinger R, et al. CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N Engl J Med. 2021;384:252–260. doi: 10.1056/NEJMoa2031054. [DOI] [PubMed] [Google Scholar]
  114. Frock RL, Hu J, Meyers RM, Ho YJ, Kii E, Alt FW. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat Biotechnol. 2015;33:179–186. doi: 10.1038/nbt.3101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Fu R, Fang M, Xu K, Ren J, Zou J, Su L, Chen X, An PP, Yu D, Ka M, et al. Generation of GGTA1−/−β2MT−/−CIITA−/− pigs using CRISPR/Cas9 technology to alleviate xenogeneic immune reactions. Transplantation. 2020;104:1566–1573. doi: 10.1097/TP.0000000000003205. [DOI] [PubMed] [Google Scholar]
  116. Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol. 2014;32:279–284. doi: 10.1038/nbt.2808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Fulco CP, Munschauer M, Anyoha R, Munson G, Grossman SR, Perez EM, Kane M, Cleary B, Lander ES, Engreitz JM. Systematic mapping of functional enhancer-promoter connections with CRISPR interference. Science. 2016;354:769–773. doi: 10.1126/science.aag2445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Ganbaatar U, Liu C. CRISPR-based COVID-19 testing: toward next-generation point-of-care diagnostics. Front Cell Infect Microbiol. 2021;11:663949. doi: 10.3389/fcimb.2021.663949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Gao X, Tao Y, Lamas V, Huang M, Yeh WH, Pan B, Hu YJ, Hu J H, Thompson DB, Shu Y, et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature. 2018;553:217–221. doi: 10.1038/nature25164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Garrelfs SF, Frishberg Y, Hulton SA, Koren MJ, O’Riordan WD, Cochat P, Deschênes G, Shasha-Lavsky H, Saland JM, Van’t Hoff WG, et al. Lumasiran, an RNAi therapeutic for primary hyperoxaluria type 1. N Engl J Med. 2021;384:1216–1226. doi: 10.1056/NEJMoa2021712. [DOI] [PubMed] [Google Scholar]
  121. Gasiunas G, Barrangou R, Horvath P, Siksnys V. Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci USA. 2012;109:E2579–E2586. doi: 10.1073/pnas.1208507109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Gasperini M, Findlay GM, McKenna A, Milbank JH, Lee C, Zhang MD, Cusanovich DA, Shendure J. CRISPR/Cas9-mediated scanning for regulatory elements required for HPRT1 expression via thousands of large, programmed genomic deletions. Am J Hum Genet. 2017;101:192–205. doi: 10.1016/j.ajhg.2017.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, Liu DR. Programmable base editing of A·T to G·C in genomic DNA without DNA cleavage. Nature. 2017;551:464–471. doi: 10.1038/nature24644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Giannelli SG, Luoni M, Castoldi V, Massimino L, Cabassi T, Angeloni D, Demontis GC, Leocani L, Andreazzoli M, Broccoli V. Cas9/sgRNA selective targeting of the P23H Rhodopsin mutant allele for treating retinitis pigmentosa by intravitreal AAV9.PHP.B-based delivery. Hum Mol Genet. 2018;27:761–779. doi: 10.1093/hmg/ddx438. [DOI] [PubMed] [Google Scholar]
  125. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres SE, Stern-Ginossar N, Brandman O, Whitehead EH, Doudna JA, et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell. 2013;154:442–451. doi: 10.1016/j.cell.2013.06.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Gillmore JD, Gane E, Taubel J, Kao J, Fontana M, Maitland ML, Seitzer J, O’Connell D, Walsh KR, Wood K, et al. CRISPR-Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med. 2021;385:493–502. doi: 10.1056/NEJMoa2107454. [DOI] [PubMed] [Google Scholar]
  127. Goh, C.J.H., Wong, J.H., El Farran, C., Tan, B.X., Coffill, C.R., Loh, Y.H., Lane, D., and Arumugam, P. (2021). Identification of pathways modulating vemurafenib resistance in melanoma cells via a genome-wide CRISPR/Cas9 screen. G3 11. [DOI] [PMC free article] [PubMed]
  128. Gootenberg JS, Abudayyeh OO, Kellner MJ, Joung J, Collins JJ, Zhang F. Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science. 2018;360:439–444. doi: 10.1126/science.aaq0179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Gootenberg JS, Abudayyeh OO, Lee JW, Essletzbichler P, Dy AJ, Joung J, Verdine V, Donghia N, Daringer NM, Freije CA, et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. Science. 2017;356:438–442. doi: 10.1126/science.aam9321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Grünewald J, Zhou R, Garcia SP, Iyer S, Lareau CA, Aryee MJ, Joung JK. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature. 2019;569:433–437. doi: 10.1038/s41586-019-1161-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Grünewald J, Zhou R, Iyer S, Lareau CA, Garcia SP, Aryee MJ, Joung JK. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat Biotechnol. 2019;37:1041–1048. doi: 10.1038/s41587-019-0236-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Guan Y, Ma Y, Li Q, Sun Z, Ma L, Wu L, Wang L, Zeng L, Shao Y, Chen Y, et al. CRISPR/Cas9-mediated somatic correction of a novel coagulator factorIX gene mutation ameliorates hemophilia in mouse. EMBO Mol Med. 2016;8:477–488. doi: 10.15252/emmm.201506039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Guilinger JP, Thompson DB, Liu DR. Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nat Biotechnol. 2014;32:577–582. doi: 10.1038/nbt.2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Guo M, Xu Y, Dong Z, Zhou Z, Cong N, Gao M, Huang W, Wang Y, Liu G, Xian X. Inactivation of ApoC3 by CRISPR/Cas9 protects against atherosclerosis in hamsters. Circ Res. 2020;127:1456–1458. doi: 10.1161/CIRCRESAHA.120.317686. [DOI] [PubMed] [Google Scholar]
  135. Guo Q, Feng X, Zhou Y. PCSK9 variants in familial hypercholesterolemia: a comprehensive synopsis. Front Genet. 2020;11:1020. doi: 10.3389/fgene.2020.01020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Guo S, Chen Y, Liu J, Zhang X, Liu Z, Zhou Z, Wei W. Low-density lipoprotein receptor-related protein 1 is a CROPs-associated receptor for Clostridioides infection toxin B. Sci China Life Sci. 2022;65:107–118. doi: 10.1007/s11427-021-1943-9. [DOI] [PubMed] [Google Scholar]
  137. Gurumurthy CB, O’Brien AR, Quadros RM, Adams J, Jr, Alcaide P, Ayabe S, Ballard J, Batra SK, Beauchamp MC, Becker KA, et al. Reproducibility of CRISPR-Cas9 methods for generation of conditional mouse alleles: a multi-center evaluation. Genome Biol. 2019;20:171. doi: 10.1186/s13059-019-1776-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. György B, Nist-Lund C, Pan B, Asai Y, Karavitaki KD, Kleinstiver BP, Garcia SP, Zaborowski MP, Solanes P, Spataro S, et al. Allele-specific gene editing prevents deafness in a model of dominant progressive hearing loss. Nat Med. 2019;25:1123–1130. doi: 10.1038/s41591-019-0500-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Haapaniemi E, Botla S, Persson J, Schmierer B, Taipale J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat Med. 2018;24:927–930. doi: 10.1038/s41591-018-0049-z. [DOI] [PubMed] [Google Scholar]
  140. Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, et al. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118:3132–3142. doi: 10.1172/JCI35700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Hajian R, Balderston S, Tran T, deBoer T, Etienne J, Sandhu M, Wauford NA, Chung JY, Nokes J, Athaiya M, et al. Detection of unamplified target genes via CRISPR-Cas9 immobilized on a graphene field-effect transistor. Nat Biomed Eng. 2019;3:427–437. doi: 10.1038/s41551-019-0371-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Han K, Jeng EE, Hess GT, Morgens DW, Li A, Bassik MC. Synergistic drug combinations for cancer identified in a CRISPR screen for pairwise genetic interactions. Nat Biotechnol. 2017;35:463–474. doi: 10.1038/nbt.3834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Hanna RE, Hegde M, Fagre CR, DeWeirdt PC, Sangree AK, Szegletes Z, Griffith A, Feeley MN, Sanson KR, Baidi Y, et al. Massively parallel assessment of human variants with base editor screens. Cell. 2021;184:1064–1080. doi: 10.1016/j.cell.2021.01.012. [DOI] [PubMed] [Google Scholar]
  144. Harel I, Benayoun BA, Machado B, Singh PP, Hu CK, Pech MF, Valenzano DR, Zhang E, Sharp SC, Artandi SE, et al. A platform for rapid exploration of aging and diseases in a naturally shortlived vertebrate. Cell. 2015;160:1013–1026. doi: 10.1016/j.cell.2015.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Harrington LB, Burstein D, Chen JS, Paez-Espino D, Ma E, Witte IP, Cofsky JC, Kyrpides NC, Banfield JF, Doudna JA. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science. 2018;362:839–842. doi: 10.1126/science.aav4294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Harris RS, Liddament MT. Retroviral restriction by APOBEC proteins. Nat Rev Immunol. 2004;4:868–877. doi: 10.1038/nri1489. [DOI] [PubMed] [Google Scholar]
  147. He J, Hu P, Gao Y, Zheng S, Xu C, Liu R, Fang L, Li R, Han C, An J, et al. Comparison and application of different immunoassay methods for the detection of SARS-CoV-2. J Med Virol. 2020;92:2777–2784. doi: 10.1002/jmv.26187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Heianza Y, Qi L. Impact of genes and environment on obesity and cardiovascular disease. Endocrinology. 2019;160:81–100. doi: 10.1210/en.2018-00591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Hendel A, Bak RO, Clark JT, Kennedy AB, Ryan DE, Roy S, Steinfeld I, Lunstad BD, Kaiser RJ, Wilkens AB, et al. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nat Biotechnol. 2015;33:985–989. doi: 10.1038/nbt.3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Hess GT, Frésard L, Han K, Lee CH, Li A, Cimprich KA, Montgomery SB, Bassik MC. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat Methods. 2016;13:1036–1042. doi: 10.1038/nmeth.4038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Hickman AB, Dyda F. DNA transposition at work. Chem Rev. 2016;116:12758–12784. doi: 10.1021/acs.chemrev.6b00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Holme E, Lindstedt S. Tyrosinaemia type I and NTBC (2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione) J Inherit Metab Dis. 1998;21:507–517. doi: 10.1023/A:1005410820201. [DOI] [PubMed] [Google Scholar]
  153. Horlbeck MA, Gilbert LA, Villalta JE, Adamson B, Pak RA, Chen Y, Fields AP, Park CY, Corn JE, Kampmann M, et al. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation. eLife. 2016;5:e19760. doi: 10.7554/eLife.19760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Horlbeck MA, Liu SJ, Chang HY, Lim DA, Weissman JS. Fitness effects of CRISPR/Cas9-targeting of long noncoding RNA genes. Nat Biotechnol. 2020;38:573–576. doi: 10.1038/s41587-020-0428-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, Li Y, Fine EJ, Wu X, Shalem O, et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol. 2013;31:827–832. doi: 10.1038/nbt.2647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Hu JH, Miller SM, Geurts MH, Tang W, Chen L, Sun N, Zeina C M, Gao X, Rees HA, Lin Z, et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature. 2018;556:57–63. doi: 10.1038/nature26155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Hu Z, Yu L, Zhu D, Ding W, Wang X, Zhang C, Wang L, Jiang X, Shen H, He D, et al. Disruption of HPV16-E7 by CRISPR/Cas system induces apoptosis and growth inhibition in HPV16 positive human cervical cancer cells. Biomed Res Int. 2014;2014:1–9. doi: 10.1155/2014/612823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Hu Z, Ding W, Zhu D, Yu L, Jiang X, Wang X, Zhang C, Wang L, Ji T, Liu D, et al. TALEN-mediated targeting of HPV oncogenes ameliorates HPV-related cervical malignancy. J Clin Invest. 2015;125:425–436. doi: 10.1172/JCI78206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Huang D, Qian J, Shi Z, Zhao J, Fang M, Xu Z. CRISPR-Cas12a-assisted multicolor biosensor for semiquantitative point-of-use testing of the nopaline synthase terminator in genetically modified crops by unaided eyes. ACS Synth Biol. 2020;9:3114–3123. doi: 10.1021/acssynbio.0c00365. [DOI] [PubMed] [Google Scholar]
  160. Huang M, Zhou X, Wang H, Xing D. Clustered regularly interspaced short palindromic repeats/Cas9 triggered isothermal amplification for site-specific nucleic acid detection. Anal Chem. 2018;90:2193–2200. doi: 10.1021/acs.analchem.7b04542. [DOI] [PubMed] [Google Scholar]
  161. Huang W, Yu L, Wen D, Wei D, Sun Y, Zhao H, Ye Y, Chen W, Zhu Y, Wang L, et al. A CRISPR-Cas12a-based specific enhancer for more sensitive detection of SARS-CoV-2 infection. Ebiomedicine. 2020;61:103036. doi: 10.1016/j.ebiom.2020.103036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Huang Y, Ding C, Liang P, Li D, Tang Y, Meng W, Sun H, Lu H, Chen Y, Chen X, et al. HBB-deficient Macaca fascicularis monkey presents with human β-thalassemia. Protein Cell. 2019;10:538–542. doi: 10.1007/s13238-019-0627-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Huang Y, Paxton WA, Wolinsky SM, Neumann AU, Zhang L, He T, Kang S, Ceradini D, Jin Z, Yazdanbakhsh K, et al. The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat Med. 1996;2:1240–1243. doi: 10.1038/nm1196-1240. [DOI] [PubMed] [Google Scholar]
  164. Humbert O, Kiem HP. Long-term increase in fetal hemoglobin expression in nonhuman primates following transplantation of autologous Bcl11a nuclease-edited HSCs. Blood. 2015;126:2035. doi: 10.1182/blood.V126.23.2035.2035. [DOI] [Google Scholar]
  165. Humbert O, Peterson CW, Norgaard ZK, Radtke S, Kiem HP. A nonhuman primate transplantation model to evaluate hematopoietic stem cell gene editing strategies for β-hemoglobinopathies. Mol Ther Methods Clin Dev. 2018;8:75–86. doi: 10.1016/j.omtm.2017.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Ihry RJ, Worringer KA, Salick MR, Frias E, Ho D, Theriault K, Kommineni S, Chen J, Sondey M, Ye C, et al. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nat Med. 2018;24:939–946. doi: 10.1038/s41591-018-0050-6. [DOI] [PubMed] [Google Scholar]
  167. Ivics Z, Li MA, Mátés L, Boeke JD, Nagy A, Bradley A, Izsvák Z. Transposon-mediated genome manipulation in vertebrates. Nat Methods. 2009;6:415–422. doi: 10.1038/nmeth.1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Izpisua Belmonte JC, Callaway EM, Caddick SJ, Churchland P, Feng G, Homanics GE, Lee KF, Leopold DA, Miller CT, Mitchell JF, et al. Brains, genes, and primates. Neuron. 2015;86:617–631. doi: 10.1016/j.neuron.2015.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Jacobson SG, Cideciyan AV, Roman AJ, Sumaroka A, Schwartz S B, Heon E, Hauswirth WW. Improvement and decline in vision with gene therapy in childhood blindness. N Engl J Med. 2015;372:1920–1926. doi: 10.1056/NEJMoa1412965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Jaitin DA, Weiner A, Yofe I, Lara-Astiaso D, Keren-Shaul H, David E, Salame TM, Tanay A, van Oudenaarden A, Amit I. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell. 2016;167:1883–1896. doi: 10.1016/j.cell.2016.11.039. [DOI] [PubMed] [Google Scholar]
  171. Jasin M, Rothstein R. Repair of strand breaks by homologous recombination. Cold Spring Harb Perspect Biol. 2013;5:a012740. doi: 10.1101/cshperspect.a012740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Jeong YK, Song B, Bae S. Current status and challenges of DNA base editing tools. Mol Ther. 2020;28:1938–1952. doi: 10.1016/j.ymthe.2020.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Ji H, Jiang Z, Lu P, Ma L, Li C, Pan H, Fu Z, Qu X, Wang P, Deng J, et al. Specific reactivation of latent HIV-1 by dCas9-SunTag-VP64-mediated guide RNA targeting the HIV-1 promoter. Mol Ther. 2016;24:508–521. doi: 10.1038/mt.2016.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Jiang C, Trudeau SJ, Cheong TC, Guo R, Teng M, Wang LW, Wang Z, Pighi C, Gautier-Courteille C, Ma Y, et al. CRISPR/Cas9 screens reveal multiple layers of B cell CD40 regulation. Cell Rep. 2019;28:1307–1322. doi: 10.1016/j.celrep.2019.06.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Jiang, T., Zhang, X.O., Weng, Z., and Xue, W. (2021). Programming large target genomic deletion and concurrent insertion via a prime editing-based method: PEDAR. bioRxiv 2021.2005.2012.443800.
  176. Jiao C, Sharma S, Dugar G, Peeck NL, Bischler T, Wimmer F, Yu Y, Barquist L, Schoen C, Kurzai O, et al. Noncanonical crRNAs derived from host transcripts enable multiplexable RNA detection by Cas9. Science. 2021;372:941–948. doi: 10.1126/science.abe7106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Jin S, Fei H, Zhu Z, Luo Y, Liu J, Gao S, Zhang F, Chen YH, Wang Y, Gao C. Rationally designed APOBEC3B cytosine base editors with improved specificity. Mol Cell. 2020;79:728–740. doi: 10.1016/j.molcel.2020.07.005. [DOI] [PubMed] [Google Scholar]
  178. Jin S, Lin Q, Luo Y, Zhu Z, Liu G, Li Y, Chen K, Qiu JL, Gao C. Genome-wide specificity of prime editors in plants. Nat Biotechnol. 2021;39:1292–1299. doi: 10.1038/s41587-021-00891-x. [DOI] [PubMed] [Google Scholar]
  179. Jin S, Zong Y, Gao Q, Zhu Z, Wang Y, Qin P, Liang C, Wang D, Qiu JL, Zhang F, et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science. 2019;364:292–295. doi: 10.1126/science.aaw7166. [DOI] [PubMed] [Google Scholar]
  180. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337:816–821. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. RNA-programmed genome editing in human cells. eLife. 2013;2:e00471. doi: 10.7554/eLife.00471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Joung J, Engreitz JM, Konermann S, Abudayyeh OO, Verdine VK, Aguet F, Gootenberg JS, Sanjana NE, Wright JB, Fulco CP, et al. Genome-scale activation screen identifies a lncRNA locus regulating a gene neighbourhood. Nature. 2017;548:343–346. doi: 10.1038/nature23451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Joung J, Ladha A, Saito M, Kim NG, Woolley AE, Segel M, Barretto RPJ, Ranu A, Macrae RK, Faure G, et al. Detection of SARS-CoV-2 with SHERLOCK one-pot testing. N Engl J Med. 2020;383:1492–1494. doi: 10.1056/NEJMc2026172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Jung IY, Kim YY, Yu HS, Lee M, Kim S, Lee J. CRISPR/Cas9-mediated knockout of DGK improves antitumor activities of human T cells. Cancer Res. 2018;78:4692–4703. doi: 10.1158/0008-5472.CAN-18-0030. [DOI] [PubMed] [Google Scholar]
  185. Kaczmarek JC, Kowalski PS, Anderson DG. Advances in the delivery of RNA therapeutics: from concept to clinical reality. Genome Med. 2017;9:60. doi: 10.1186/s13073-017-0450-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Kang, Y., Chu, C., Wang, F., and Niu, Y. (2019). CRISPR/Cas9-mediated genome editing in nonhuman primates. Dis Model Mech 12. [DOI] [PMC free article] [PubMed]
  187. Kennedy EM, Cullen BR. Gene editing: A new tool for viral disease. Annu Rev Med. 2017;68:401–411. doi: 10.1146/annurev-med-051215-031129. [DOI] [PubMed] [Google Scholar]
  188. Khanna KK, Keating KE, Kozlov S, Scott S, Gatei M, Hobson K, Taya Y, Gabrielli B, Chan D, Lees-Miller SP, et al. ATM associates with and phosphorylates p53: mapping the region of interaction. Nat Genet. 1998;20:398–400. doi: 10.1038/3882. [DOI] [PubMed] [Google Scholar]
  189. Kim D, Bae S, Park J, Kim E, Kim S, Yu HR, Hwang J, Kim JI, Kim JS. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat Methods. 2015;12:237–243. doi: 10.1038/nmeth.3284. [DOI] [PubMed] [Google Scholar]
  190. Kim D, Luk K, Wolfe SA, Kim JS. Evaluating and enhancing target specificity of gene-editing nucleases and deaminases. Annu Rev Biochem. 2019;88:191–220. doi: 10.1146/annurev-biochem-013118-111730. [DOI] [PubMed] [Google Scholar]
  191. Kim DY, Moon SB, Ko JH, Kim YS, Kim D. Unbiased investigation of specificities of prime editing systems in human cells. Nucleic Acids Res. 2020;48:10576–10589. doi: 10.1093/nar/gkaa764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Kim S, Kim D, Cho SW, Kim J, Kim JS. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 2014;24:1012–1019. doi: 10.1101/gr.171322.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Kim S, Koo T, Jee HG, Cho HY, Lee G, Lim DG, Shin HS, Kim JS. CRISPR RNAs trigger innate immune responses in human cells. Genome Res. 2018;28:367–373. doi: 10.1101/gr.231936.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Kim YB, Komor AC, Levy JM, Packer MS, Zhao KT, Liu D R. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat Biotechnol. 2017;35:371–376. doi: 10.1038/nbt.3803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Kim YG, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci USA. 1996;93:1156–1160. doi: 10.1073/pnas.93.3.1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Klann TS, Black JB, Chellappan M, Safi A, Song L, Hilton IB, Crawford GE, Reddy TE, Gersbach CA. CRISPR-Cas9 epigenome editing enables high-throughput screening for functional regulatory elements in the human genome. Nat Biotechnol. 2017;35:561–568. doi: 10.1038/nbt.3853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z, Joung JK. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature. 2016;529:490–495. doi: 10.1038/nature16526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT, Zheng Z, Gonzales APW, Li Z, Peterson RT, Yeh JRJ, et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. 2015;523:481–485. doi: 10.1038/nature14592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Kleinstiver BP, Tsai SQ, Prew MS, Nguyen NT, Welch MM, Lopez JM, McCaw ZR, Aryee MJ, Joung JK. Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells. Nat Biotechnol. 2016;34:869–874. doi: 10.1038/nbt.3620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Klompe SE, Vo PLH, Halpin-Healy TS, Sternberg SH. Transposon-encoded CRISPR-Cas systems direct RNA-guided DNA integration. Nature. 2019;571:219–225. doi: 10.1038/s41586-019-1323-z. [DOI] [PubMed] [Google Scholar]
  201. Klug A. The discovery of zinc fingers and their applications in gene regulation and genome manipulation. Annu Rev Biochem. 2010;79:213–231. doi: 10.1146/annurev-biochem-010909-095056. [DOI] [PubMed] [Google Scholar]
  202. Kocak DD, Josephs EA, Bhandarkar V, Adkar SS, Kwon JB, Gersbach CA. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nat Biotechnol. 2019;37:657–666. doi: 10.1038/s41587-019-0095-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. 2016;533:420–424. doi: 10.1038/nature17946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Komor AC, Zhao KT, Packer MS, Gaudelli NM, Waterbury AL, Koblan LW, Kim YB, Badran AH, Liu DR. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci Adv. 2017;3:eaao4774. doi: 10.1126/sciadv.aao4774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Konermann S, Brigham MD, Trevino AE, Hsu PD, Heidenreich M, Le Cong M, Platt RJ, Scott DA, Church GM, Zhang F. Optical control of mammalian endogenous transcription and epigenetic states. Nature. 2013;500:472–476. doi: 10.1038/nature12466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Konermann S, Lotfy P, Brideau NJ, Oki J, Shokhirev MN, Hsu PD. Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors. Cell. 2018;173:665–676. doi: 10.1016/j.cell.2018.02.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Korkmaz G, Lopes R, Ugalde AP, Nevedomskaya E, Han R, Myacheva K, Zwart W, Elkon R, Agami R. Functional genetic screens for enhancer elements in the human genome using CRISPR-Cas9. Nat Biotechnol. 2016;34:192–198. doi: 10.1038/nbt.3450. [DOI] [PubMed] [Google Scholar]
  208. Korkmaz G, Manber Z, Lopes R, Prekovic S, Schuurman K, Kim Y, Teunissen H, Flach K, Wit E, Galli GG, et al. A CRISPR-Cas9 screen identifies essential CTCF anchor sites for estrogen receptor-driven breast cancer cell proliferation. Nucleic Acids Res. 2019;47:9557–9572. doi: 10.1093/nar/gkz675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Kosicki M, Tomberg K, Bradley A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat Biotechnol. 2018;36:765–771. doi: 10.1038/nbt.4192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Kovač A, Miskey C, Menzel M, Grueso E, Gogol-Döring A, Ivics Z. RNA-guided retargeting of Sleeping Beauty transposition in human cells. eLife. 2020;9:e53868. doi: 10.7554/eLife.53868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Kunkel TA, Erie DA. Eukaryotic mismatch repair in relation to DNA replication. Annu Rev Genet. 2015;49:291–313. doi: 10.1146/annurev-genet-112414-054722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Kurata JS, Lin RJ. MicroRNA-focused CRISPR-Cas9 library screen reveals fitness-associated miRNAs. RNA. 2018;24:966–981. doi: 10.1261/rna.066282.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Ladowski J, Martens G, Estrada J, Tector M, Tector J. The desirable donor pig to eliminate all xenoreactive antigens. Xenotransplantation. 2019;26:e12504. doi: 10.1111/xen.12504. [DOI] [PubMed] [Google Scholar]
  214. Landrum MJ, Lee JM, Riley GR, Jang W, Rubinstein WS, Church DM, Maglott DR. ClinVar: public archive of relationships among sequence variation and human phenotype. Nucl Acids Res. 2014;42:D980–D985. doi: 10.1093/nar/gkt1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Lansing, F., Mukhametzyanova, L., Rojo-Romanos, T., Iwasawa, K., Kimura, M., Paszkowski-Rogacz, M., Karpinski, J., Grass, T., Sonntag, J., Schneider, P.M., et al. (2020a). Correction of a Factor VIII genomic inversion with designer-recombinases. bioRxiv2020.2011.2002.328013. [DOI] [PMC free article] [PubMed]
  216. Lansing F, Paszkowski-Rogacz M, Schmitt LT, Schneider PM, Rojo Romanos T, Sonntag J, Buchholz F. A heterodimer of evolved designer-recombinases precisely excises a human genomic DNA locus. Nucleic Acids Res. 2020;48:472–485. doi: 10.1093/nar/gkz1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Latella MC, Di Salvo MT, Cocchiarella F, Benati D, Grisendi G, Comitato A, Marigo V, Recchia A. In vivo editing of the human mutant rhodopsin gene by electroporation of plasmid-based CRISPR/Cas9 in the mouse retina. Mol Ther Nucleic Acids. 2016;5:e389. doi: 10.1038/mtna.2016.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Lee JK, Jeong E, Lee J, Jung M, Shin E, Kim YH, Lee K, Jung I, Kim D, Kim S, et al. Directed evolution of CRISPR-Cas9 to increase its specificity. Nat Commun. 2018;9:3048. doi: 10.1038/s41467-018-05477-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Lee RA, Puig HD, Nguyen PQ, Angenent-Mari NM, Donghia N M, McGee JP, Dvorin JD, Klapperich CM, Pollock NR, Collins JJ. Ultrasensitive CRISPR-based diagnostic for field-applicable detection of Plasmodium species in symptomatic and asymptomatic malaria. Proc Natl Acad Sci USA. 2020;117:25722–25731. doi: 10.1073/pnas.2010196117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Lei L, Chen H, Xue W, Yang B, Hu B, Wei J, Wang L, Cui Y, Li W, Wang J, et al. APOBEC3 induces mutations during repair of CRISPR-Cas9-generated DNA breaks. Nat Struct Mol Biol. 2018;25:45–52. doi: 10.1038/s41594-017-0004-6. [DOI] [PubMed] [Google Scholar]
  221. Lei Z, Meng H, Lv Z, Liu M, Zhao H, Wu H, Zhang X, Liu L, Zhuang Y, Yin K, et al. Detect-seq reveals out-of-protospacer editing and target-strand editing by cytosine base editors. Nat Methods. 2021;18:643–651. doi: 10.1038/s41592-021-01172-w. [DOI] [PubMed] [Google Scholar]
  222. Leung, R.K.K., Cheng, Q.X., Wu, Z.L., Khan, G., Liu, Y., Xia, H.Y., and Wang, J. (2021). CRISPR-Cas12-based nucleic acids detection systems. Methods doi: 10.1016/j.ymeth.2021.02.018. [DOI] [PubMed]
  223. Li D, Qiu Z, Shao Y, Chen Y, Guan Y, Liu M, Li Y, Gao N, Wang L, Lu X, et al. Heritable gene targeting in the mouse and rat using a CRISPR-Cas system. Nat Biotechnol. 2013;31:681–683. doi: 10.1038/nbt.2661. [DOI] [PubMed] [Google Scholar]
  224. Li H, Li M, Yang Y, Wang F, Wang F, Li C. Aptamerlinked CRISPR/Cas12a-based immunoassay. Anal Chem. 2021;93:3209–3216. doi: 10.1021/acs.analchem.0c04687. [DOI] [PubMed] [Google Scholar]
  225. Li H, Xing S, Xu J, He Y, Lai Y, Wang Y, Zhang G, Guo S, Deng M, Zeng M, et al. Aptamer-based CRISPR/Cas12a assay for the ultrasensitive detection of extracellular vesicle proteins. Talanta. 2021;221:121670. doi: 10.1016/j.talanta.2020.121670. [DOI] [PubMed] [Google Scholar]
  226. Li J, Yang S, Zuo C, Dai L, Guo Y, Xie G. Applying CRISPR-Cas12a as a signal amplifier to construct biosensors for Non-DNA targets in ultralow concentrations. ACS Sens. 2020;5:970–977. doi: 10.1021/acssensors.9b02305. [DOI] [PubMed] [Google Scholar]
  227. Li L, Li S, Wu N, Wu J, Wang G, Zhao G, Wang J. HOLMESv2: a CRISPR-Cas12b-assisted platform for nucleic acid detection and DNA methylation quantitation. ACS Synth Biol. 2019;8:2228–2237. doi: 10.1021/acssynbio.9b00209. [DOI] [PubMed] [Google Scholar]
  228. Li N, Tang N, Cheng C, Hu T, Wei X, Han W, Wang H. Improving the anti-solid tumor efficacy of CAR-T cells by inhibiting adenosine signaling pathway. Oncoimmunology. 2020;9:1824643. doi: 10.1080/2162402X.2020.1824643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Li P, Kleinstiver BP, Leon MY, Prew MS, Navarro-Gomez D, Greenwald SH, Pierce EA, Joung JK, Liu Q. Allele-specific CRISPR-Cas9 genome editing of the single-base P23H mutation for rhodopsin-associated dominant retinitis pigmentosa. CRISPR J. 2018;1:55–64. doi: 10.1089/crispr.2017.0009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Li S, Li X, Xue W, Zhang L, Yang LZ, Cao SM, Lei YN, Liu C X, Guo SK, Shan L, et al. Screening for functional circular RNAs using the CRISPR-Cas13 system. Nat Methods. 2021;18:51–59. doi: 10.1038/s41592-020-01011-4. [DOI] [PubMed] [Google Scholar]
  231. Li SY, Cheng QX, Liu JK, Nie XQ, Zhao GP, Wang J. CRISPR-Cas12a has both cis- and trans-cleavage activities on single-stranded DNA. Cell Res. 2018;28:491–493. doi: 10.1038/s41422-018-0022-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Li SY, Cheng QX, Wang JM, Li XY, Zhang ZL, Gao S, Cao R B, Zhao GP, Wang J. CRISPR-Cas12a-assisted nucleic acid detection. Cell Discov. 2018;4:20. doi: 10.1038/s41421-018-0028-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Li X, Wang Y, Liu Y, Yang B, Wang X, Wei J, Lu Z, Zhang Y, Wu J, Huang X, et al. Base editing with a Cpf1-cytidine deaminase fusion. Nat Biotechnol. 2018;36:324–327. doi: 10.1038/nbt.4102. [DOI] [PubMed] [Google Scholar]
  234. Li Y, Li S, Wang J, Liu G. CRISPR/Cas systems towards next-generation biosensing. Trends Biotechnol. 2019;37:730–743. doi: 10.1016/j.tibtech.2018.12.005. [DOI] [PubMed] [Google Scholar]
  235. Li Y, Yao L, Li J, Chen L, Song Y, Cai Z, Yang C. Stability issues of RT-PCR testing of SARS-CoV-2 for hospitalized patients clinically diagnosed with COVID-19. J Med Virol. 2020;92:903–908. doi: 10.1002/jmv.25786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Li Z, Zheng W, Wang H, Cheng Y, Fang Y, Wu F, Sun G, Sun G, Lv C, Hui B. Application of animal models in cancer research: recent progress and future prospects. Cancer Manag Res Volume. 2021;13:2455–2475. doi: 10.2147/CMAR.S302565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Liang M, Li Z, Wang W, Liu J, Liu L, Zhu G, Karthik L, Wang M, Wang KF, Wang Z, et al. A CRISPR-Cas12a-derived biosensing platform for the highly sensitive detection of diverse small molecules. Nat Commun. 2019;10:3672. doi: 10.1038/s41467-019-11648-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Liang Y, Zhang G, Li Q, Han L, Hu X, Guo Y, Tao W, Zhao X, Guo M, Gan T, et al. TRIM26 is a critical host factor for HCV replication and contributes to host tropism. Sci Adv. 2021;7:eabd9732. doi: 10.1126/sciadv.abd9732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Lin Q, Jin S, Zong Y, Yu H, Zhu Z, Liu G, Kou L, Wang Y, Qiu JL, Li J, et al. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat Biotechnol. 2021;39:923–927. doi: 10.1038/s41587-021-00868-w. [DOI] [PubMed] [Google Scholar]
  240. Lin Q, Lv JN, Wu KC, Zhang CJ, Liu Q, Jin ZB. Generation of nonhuman primate model of cone dysfunction through in situ AAV-mediated CNGB3 ablation. Mol Ther Methods Clin Dev. 2020;18:869–879. doi: 10.1016/j.omtm.2020.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Ling S, Yang S, Hu X, Yin D, Dai Y, Qian X, Wang D, Pan X, Hong J, Sun X, et al. Lentiviral delivery of co-packaged Cas9 mRNA and a Vegfa-targeting guide RNA prevents wet age-related macular degeneration in mice. Nat Biomed Eng. 2021;5:144–156. doi: 10.1038/s41551-020-00656-y. [DOI] [PubMed] [Google Scholar]
  242. Liu PF, Zhao KR, Liu ZJ, Wang L, Ye SY, Liang GX. Cas12a-based electrochemiluminescence biosensor for target amplification-free DNA detection. Biosens Bioelectron. 2021;176:112954. doi: 10.1016/j.bios.2020.112954. [DOI] [PubMed] [Google Scholar]
  243. Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, Horuk R, MacDonald ME, Stuhlmann H, Koup RA, Landau NR. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell. 1996;86:367–377. doi: 10.1016/S0092-8674(00)80110-5. [DOI] [PubMed] [Google Scholar]
  244. Liu, S.J., Horlbeck, M.A., Cho, S.W., Birk, H.S., Malatesta, M., He, D., Attenello, F.J., Villalta, J.E., Cho, M.Y., Chen, Y., et al. (2017a). CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science 355. [DOI] [PMC free article] [PubMed]
  245. Liu X, Zhang Y, Cheng C, Cheng AW, Zhang X, Li N, Xia C, Wei X, Liu X, Wang H. CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res. 2017;27:154–157. doi: 10.1038/cr.2016.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Liu XS, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, Shu J, Dadon D, Young RA, Jaenisch R. Editing DNA methylation in the mammalian genome. Cell. 2016;167:233–247. doi: 10.1016/j.cell.2016.08.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Liu Y, Cao Z, Wang Y, Guo Y, Xu P, Yuan P, Liu Z, He Y, Wei W. Genome-wide screening for functional long noncoding RNAs in human cells by Cas9 targeting of splice sites. Nat Biotechnol. 2018;36:1203–1210. doi: 10.1038/nbt.4283. [DOI] [PubMed] [Google Scholar]
  248. Liu Y, Liu Z, Cao Z, Wei W. Reply to: fitness effects of CRISPR/Cas9-targeting of long noncoding RNA genes. Nat Biotechnol. 2020;38:577–578. doi: 10.1038/s41587-020-0431-5. [DOI] [PubMed] [Google Scholar]
  249. Liu Y, Yang G, Huang S, Li X, Wang X, Li G, Chi T, Chen Y, Huang X, Wang X. Enhancing prime editing by Csy4-mediated processing of pegRNA. Cell Res. 2021;31:1134–1136. doi: 10.1038/s41422-021-00520-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Liu Z, Cai Y, Wang Y, Nie Y, Zhang C, Xu Y, Zhang X, Lu Y, Wang Z, Poo M, et al. Cloning of macaque monkeys by somatic cell nuclear transfer. Cell. 2018;172:881–887. doi: 10.1016/j.cell.2018.01.020. [DOI] [PubMed] [Google Scholar]
  251. Lu S, Li F, Chen Q, Wu J, Duan J, Lei X, Zhang Y, Zhao D, Bu Z, Yin H. Rapid detection of African swine fever virus using Cas12a-based portable paper diagnostics. Cell Discov. 2020;6:18. doi: 10.1038/s41421-020-0151-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Luo W, Galvan DL, Woodard LE, Dorset D, Levy S, Wilson M H. Comparative analysis of chimeric ZFP-, TALE- and Cas9-piggyBac transposases for integration into a single locus in human cells. Nucleic Acids Res. 2017;45:8411–8422. doi: 10.1093/nar/gkx572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Ma Y, Zhang J, Yin W, Zhang Z, Song Y, Chang X. Targeted AID-mediated mutagenesis (TAM) enables efficient genomic diversification in mammalian cells. Nat Methods. 2016;13:1029–1035. doi: 10.1038/nmeth.4027. [DOI] [PubMed] [Google Scholar]
  254. Ma Y, Zhang X, Shen B, Lu Y, Chen W, Ma J, Bai L, Huang X, Zhang L. Generating rats with conditional alleles using CRISPR/Cas9. Cell Res. 2014;24:122–125. doi: 10.1038/cr.2013.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Maeder ML, Stefanidakis M, Wilson CJ, Baral R, Barrera LA, Bounoutas GS, Bumcrot D, Chao H, Ciulla DM, DaSilva JA, et al. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med. 2019;25:229–233. doi: 10.1038/s41591-018-0327-9. [DOI] [PubMed] [Google Scholar]
  256. Maguire AM, High KA, Auricchio A, Wright JF, Pierce EA, Testa F, Mingozzi F, Bennicelli JL, Ying G, Rossi S, et al. Age-dependent effects of RPE65 gene therapy for Leber’s congenital amaurosis: a phase 1 dose-escalation trial. Lancet. 2009;374:1597–1605. doi: 10.1016/S0140-6736(09)61836-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Maguire AM, Simonelli F, Pierce EA, Pugh EN, Jr, Mingozzi F, Bennicelli J, Banfi S, Marshall KA, Testa F, Surace EM, et al. Safety and efficacy of gene transfer for Leber’s congenital amaurosis. N Engl J Med. 2008;358:2240–2248. doi: 10.1056/NEJMoa0802315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Makarova KS, Wolf YI, Iranzo J, Shmakov SA, Alkhnbashi OS, Brouns SJJ, Charpentier E, Cheng D, Haft DH, Horvath P, et al. Evolutionary classification of CRISPR-Cas systems: a burst of class 2 and derived variants. Nat Rev Microbiol. 2020;18:67–83. doi: 10.1038/s41579-019-0299-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM. RNA-guided human genome engineering via Cas9. Science. 2013;339:823–826. doi: 10.1126/science.1232033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Mandal PK, Ferreira LMR, Collins R, Meissner TB, Boutwell CL, Friesen M, Vrbanac V, Garrison BS, Stortchevoi A, Bryder D, et al. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell. 2014;15:643–652. doi: 10.1016/j.stem.2014.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Mao XY, Dai JX, Zhou HH, Liu ZQ, Jin WL. Brain tumor modeling using the CRISPR/Cas9 system: state of the art and view to the future. Oncotarget. 2016;7:33461–33471. doi: 10.18632/oncotarget.8075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Maresch R, Mueller S, Veltkamp C, Öllinger R, Friedrich M, Heid I, Steiger K, Weber J, Engleitner T, Barenboim M, et al. Multiplexed pancreatic genome engineering and cancer induction by transfection-based CRISPR/Cas9 delivery in mice. Nat Commun. 2016;7:10770. doi: 10.1038/ncomms10770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Martin RM, Ikeda K, Cromer MK, Uchida N, Nishimura T, Romano R, Tong AJ, Lemgart VT, Camarena J, Pavel-Dinu M, et al. Highly efficient and marker-free genome editing of human pluripotent stem cells by CRISPR-Cas9 RNP and AAV6 donor-mediated homologous recombination. Cell Stem Cell. 2019;24:821–828. doi: 10.1016/j.stem.2019.04.001. [DOI] [PubMed] [Google Scholar]
  264. Martinez-Lage M, Torres-Ruiz R, Rodriguez-Perales S. CRISPR/Cas9 technology: applications and human disease modeling. In: Progress in Molecular Biology and Translational Science. New York: Academic Press; 2017. pp. 23–48. [DOI] [PubMed] [Google Scholar]
  265. Meinke G, Bohm A, Hauber J, Pisabarro MT, Buchholz F. Cre recombinase and other tyrosine recombinases. Chem Rev. 2016;116:12785–12820. doi: 10.1021/acs.chemrev.6b00077. [DOI] [PubMed] [Google Scholar]
  266. Menchaca A, Dos Santos-Neto PC, Souza-Neves M, Cuadro F, Mulet AP, Tesson L, Chenouard V, Guiffès A, Heslan JM, Gantier M, et al. Otoferlin gene editing in sheep via CRISPR-assisted ssODN-mediated Homology Directed Repair. Sci Rep. 2020;10:5995. doi: 10.1038/s41598-020-62879-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Merkle T, Merz S, Reautschnig P, Blaha A, Li Q, Vogel P, Wettengel J, Li JB, Stafforst T. Precise RNA editing by recruiting endogenous ADARs with antisense oligonucleotides. Nat Biotechnol. 2019;37:133–138. doi: 10.1038/s41587-019-0013-6. [DOI] [PubMed] [Google Scholar]
  268. Miller J, McLachlan AD, Klug A. Repetitive zinc-binding domains in the protein transcription factor IIIA from Xenopus oocytes. EMBO J. 1985;4:1609–1614. doi: 10.1002/j.1460-2075.1985.tb03825.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Miller JC, Tan S, Qiao G, Barlow KA, Wang J, Xia DF, Meng X, Paschon DE, Leung E, Hinkley SJ, et al. A TALE nuclease architecture for efficient genome editing. Nat Biotechnol. 2011;29:143–148. doi: 10.1038/nbt.1755. [DOI] [PubMed] [Google Scholar]
  270. Miller SM, Wang T, Randolph PB, Arbab M, Shen MW, Huang T P, Matuszek Z, Newby GA, Rees HA, Liu DR. Continuous evolution of SpCas9 variants compatible with non-G PAMs. Nat Biotechnol. 2020;38:471–481. doi: 10.1038/s41587-020-0412-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  271. Mimitou EP, Cheng A, Montalbano A, Hao S, Stoeckius M, Legut M, Roush T, Herrera A, Papalexi E, Ouyang Z, et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat Methods. 2019;16:409–412. doi: 10.1038/s41592-019-0392-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Mojica FJM, Díez-Villaseñor C, García-Martínez J, Soria E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J Mol Evol. 2005;60:174–182. doi: 10.1007/s00239-004-0046-3. [DOI] [PubMed] [Google Scholar]
  273. Moreno AM, Palmer N, Alemán F, Chen G, Pla A, Jiang N, Leong Chew W, Law M, Mali P. Immune-orthogonal orthologues of AAV capsids and of Cas9 circumvent the immune response to the administration of gene therapy. Nat Biomed Eng. 2019;3:806–816. doi: 10.1038/s41551-019-0431-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Moscou MJ, Bogdanove AJ. A simple cipher governs DNA recognition by TAL effectors. Science. 2009;326:1501. doi: 10.1126/science.1178817. [DOI] [PubMed] [Google Scholar]
  275. Mu W, Tang N, Cheng C, Sun W, Wei X, Wang H. In vitro transcribed sgRNA causes cell death by inducing interferon release. Protein Cell. 2019;10:461–465. doi: 10.1007/s13238-018-0605-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Mukama O, Wu J, Li Z, Liang Q, Yi Z, Lu X, Liu Y, Liu Y, Hussain M, Makafe GG, et al. An ultrasensitive and specific point-of-care CRISPR/Cas12 based lateral flow biosensor for the rapid detection of nucleic acids. Biosens Bioelectron. 2020;159:112143. doi: 10.1016/j.bios.2020.112143. [DOI] [PubMed] [Google Scholar]
  277. Müller U, Barr-Gillespie PG. New treatment options for hearing loss. Nat Rev Drug Discov. 2015;14:346–365. doi: 10.1038/nrd4533. [DOI] [PubMed] [Google Scholar]
  278. Mussolino C, Morbitzer R, Lütge F, Dannemann N, Lahaye T, Cathomen T. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 2011;39:9283–9293. doi: 10.1093/nar/gkr597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Mussolino C, Alzubi J, Fine EJ, Morbitzer R, Cradick TJ, Lahaye T, Bao G, Cathomen T. TALENs facilitate targeted genome editing in human cells with high specificity and low cytotoxicity. Nucleic Acids Res. 2014;42:6762–6773. doi: 10.1093/nar/gku305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Musunuru K, Chadwick AC, Mizoguchi T, Garcia SP, DeNizio JE, Reiss CW, Wang K, Iyer S, Dutta C, Clendaniel V, et al. In vivo CRISPR base editing of PCSK9 durably lowers cholesterol in primates. Nature. 2021;593:429–434. doi: 10.1038/s41586-021-03534-y. [DOI] [PubMed] [Google Scholar]
  281. Myhrvold C, Freije CA, Gootenberg JS, Abudayyeh OO, Metsky H C, Durbin AF, Kellner MJ, Tan AL, Paul LM, Parham LA, et al. Field-deployable viral diagnostics using CRISPR-Cas13. Science. 2018;360:444–448. doi: 10.1126/science.aas8836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Najm FJ, Strand C, Donovan KF, Hegde M, Sanson KR, Vaimberg EW, Sullender ME, Hartenian E, Kalani Z, Fusi N, et al. Orthologous CRISPR-Cas9 enzymes for combinatorial genetic screens. Nat Biotechnol. 2018;36:179–189. doi: 10.1038/nbt.4048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  283. Nakamura M, Gao Y, Dominguez AA, Qi LS. CRISPR technologies for precise epigenome editing. Nat Cell Biol. 2021;23:11–22. doi: 10.1038/s41556-020-00620-7. [DOI] [PubMed] [Google Scholar]
  284. Naldini L. Ex vivo gene transfer and correction for cell-based therapies. Nat Rev Genet. 2011;12:301–315. doi: 10.1038/nrg2985. [DOI] [PubMed] [Google Scholar]
  285. Newby GA, Yen JS, Woodard KJ, Mayuranathan T, Lazzarotto C R, Li Y, Sheppard-Tillman H, Porter SN, Yao Y, Mayberry K, et al. Base editing of haematopoietic stem cells rescues sickle cell disease in mice. Nature. 2021;595:295–302. doi: 10.1038/s41586-021-03609-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Nguyen GN, Everett JK, Kafle S, Roche AM, Raymond HE, Leiby J, Wood C, Assenmacher CA, Merricks EP, Long CT, et al. A long-term study of AAV gene therapy in dogs with hemophilia A identifies clonal expansions of transduced liver cells. Nat Biotechnol. 2021;39:47–55. doi: 10.1038/s41587-020-0741-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Nguyen LT, Smith BM, Jain PK. Enhancement of transcleavage activity of Cas12a with engineered crRNA enables amplified nucleic acid detection. Nat Commun. 2020;11:4906. doi: 10.1038/s41467-020-18615-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Ning B, Yu T, Zhang S, Huang Z, Tian D, Lin Z, Niu A, Golden N, Hensley K, Threeton B, et al. A smartphone-read ultrasensitive and quantitative saliva test for COVID-19. Sci Adv. 2021;7:eabe3703. doi: 10.1126/sciadv.abe3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Nishida, K., Arazoe, T., Yachie, N., Banno, S., Kakimoto, M., Tabata, M., Mochizuki, M., Miyabe, A., Araki, M., Hara, K.Y., et al. (2016). Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353. [DOI] [PubMed]
  290. Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N, Ishitani R, Zhang F, Nureki O. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. 2014;156:935–949. doi: 10.1016/j.cell.2014.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Nishimasu H, Shi X, Ishiguro S, Gao L, Hirano S, Okazaki S, Noda T, Abudayyeh OO, Gootenberg JS, Mori H, et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science. 2018;361:1259–1262. doi: 10.1126/science.aas9129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Niu D, Wei HJ, Lin L, George H, Wang T, Lee IH, Zhao HY, Wang Y, Kan Y, Shrock E, et al. Inactivation of porcine endogenous retrovirus in pigs using CRISPR-Cas9. Science. 2017;357:1303–1307. doi: 10.1126/science.aan4187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Niu Y, Shen B, Cui Y, Chen Y, Wang J, Wang L, Kang Y, Zhao X, Si W, Li W, et al. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene targeting in one-cell embryos. Cell. 2014;156:836–843. doi: 10.1016/j.cell.2014.01.027. [DOI] [PubMed] [Google Scholar]
  294. Norman TM, Horlbeck MA, Replogle JM, Ge AY, Xu A, Jost M, Gilbert LA, Weissman JS. Exploring genetic interaction manifolds constructed from rich single-cell phenotypes. Science. 2019;365:786–793. doi: 10.1126/science.aax4438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Nouri R, Jiang Y, Lian XL, Guan W. Sequence-specific recognition of HIV-1 DNA with solid-state CRISPR-Cas12a-assisted nanopores (SCAN) ACS Sens. 2020;5:1273–1280. doi: 10.1021/acssensors.0c00497. [DOI] [PubMed] [Google Scholar]
  296. Novarino, G. (2017). Rett syndrome modeling goes simian. Sci Transl Med 9. [DOI] [PubMed]
  297. Nuñez JK, Chen J, Pommier GC, Cogan JZ, Replogle JM, Adriaens C, Ramadoss GN, Shi Q, Hung KL, Samelson AJ, et al. Genome-wide programmable transcriptional memory by CRISPR-based epigenome editing. Cell. 2021;184:2503–2519. doi: 10.1016/j.cell.2021.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Owens JB, Urschitz J, Stoytchev I, Dang NC, Stoytcheva Z, Belcaid M, Maragathavally KJ, Coates CJ, Segal DJ, Moisyadi S. Chimeric piggyBac transposases for genomic targeting in human cells. Nucleic Acids Res. 2012;40:6978–6991. doi: 10.1093/nar/gks309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Owens JB, Mauro D, Stoytchev I, Bhakta MS, Kim MS, Segal D J, Moisyadi S. Transcription activator like effector (TALE)-directed piggyBac transposition in human cells. Nucleic Acids Res. 2013;41:9197–9207. doi: 10.1093/nar/gkt677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Pan B, Géléoc GS, Asai Y, Horwitz GC, Kurima K, Ishikawa K, Kawashima Y, Griffith AJ, Holt JR. TMC1 and TMC2 are components of the mechanotransduction channel in hair cells of the mammalian inner ear. Neuron. 2013;79:504–515. doi: 10.1016/j.neuron.2013.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Pankowicz FP, Barzi M, Legras X, Hubert L, Mi T, Tomolonis JA, Ravishankar M, Sun Q, Yang D, Borowiak M, et al. Reprogramming metabolic pathways in vivo with CRISPR/Cas9 genome editing to treat hereditary tyrosinaemia. Nat Commun. 2016;7:12642. doi: 10.1038/ncomms12642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Pardee K, Green AA, Takahashi MK, Braff D, Lambert G, Lee J W, Ferrante T, Ma D, Donghia N, Fan M, et al. Rapid, low-cost detection of Zika virus using programmable biomolecular components. Cell. 2016;165:1255–1266. doi: 10.1016/j.cell.2016.04.059. [DOI] [PubMed] [Google Scholar]
  303. Patchsung M, Jantarug K, Pattama A, Aphicho K, Suraritdechachai S, Meesawat P, Sappakhaw K, Leelahakorn N, Ruenkam T, Wongsatit T, et al. Clinical validation of a Cas13-based assay for the detection of SARS-CoV-2 RNA. Nat Biomed Eng. 2020;4:1140–1149. doi: 10.1038/s41551-020-00603-x. [DOI] [PubMed] [Google Scholar]
  304. Pattanayak V, Ramirez CL, Joung JK, Liu DR. Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat Methods. 2011;8:765–770. doi: 10.1038/nmeth.1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  305. Peng J, Zhou Y, Zhu S, Wei W. High-throughput screens in mammalian cells using the CRISPR-Cas9 system. FEBS J. 2015;282:2089–2096. doi: 10.1111/febs.13251. [DOI] [PubMed] [Google Scholar]
  306. Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O, Wang N, Lee G, Bartsevich VV, Lee YL, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 2008;26:808–816. doi: 10.1038/nbt1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Pickar-Oliver A, Gersbach CA. The next generation of CRISPR-Cas technologies and applications. Nat Rev Mol Cell Biol. 2019;20:490–507. doi: 10.1038/s41580-019-0131-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  308. Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L, Kempton HR, Dahlman JE, Parnas O, Eisenhaure TM, Jovanovic M, et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell. 2014;159:440–455. doi: 10.1016/j.cell.2014.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Pourcel C, Salvignol G, Vergnaud G. CRISPR elements in Yersinia pestis acquire new repeats by preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies. Microbiology. 2005;151:653–663. doi: 10.1099/mic.0.27437-0. [DOI] [PubMed] [Google Scholar]
  310. Provasi E, Genovese P, Lombardo A, Magnani Z, Liu PQ, Reik A, Chu V, Paschon DE, Zhang L, Kuball J, et al. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat Med. 2012;18:807–815. doi: 10.1038/nm.2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, Lim WA. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013;152:1173–1183. doi: 10.1016/j.cell.2013.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  312. Qiu XY, Zhu LY, Zhu CS, Ma JX, Hou T, Wu XM, Xie SS, Min L, Tan DA, Zhang DY, et al. Highly effective and low-cost microRNA detection with CRISPR-Cas9. ACS Synth Biol. 2018;7:807–813. doi: 10.1021/acssynbio.7b00446. [DOI] [PubMed] [Google Scholar]
  313. Qu L, Yi Z, Zhu S, Wang C, Cao Z, Zhou Z, Yuan P, Yu Y, Tian F, Liu Z, et al. Programmable RNA editing by recruiting endogenous ADAR using engineered RNAs. Nat Biotechnol. 2019;37:1059–1069. doi: 10.1038/s41587-019-0178-z. [DOI] [PubMed] [Google Scholar]
  314. Rajagopal N, Srinivasan S, Kooshesh K, Guo Y, Edwards MD, Banerjee B, Syed T, Emons BJM, Gifford DK, Sherwood R I. High-throughput mapping of regulatory DNA. Nat Biotechnol. 2016;34:167–174. doi: 10.1038/nbt.3468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Ramakrishna S, Kwaku Dad AB, Beloor J, Gopalappa R, Lee SK, Kim H. Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA. Genome Res. 2014;24:1020–1027. doi: 10.1101/gr.171264.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Ramanan V, Shlomai A, Cox DBT, Schwartz RE, Michailidis E, Bhatta A, Scott DA, Zhang F, Rice CM, Bhatia SN. CRISPR/Cas9 cleavage of viral DNA efficiently suppresses hepatitis B virus. Sci Rep. 2015;5:10833. doi: 10.1038/srep10833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, Zetsche B, Shalem O, Wu X, Makarova KS, et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520:186–191. doi: 10.1038/nature14299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, Trevino AE, Scott DA, Inoue A, Matoba S, Zhang Y, et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell. 2013;154:1380–1389. doi: 10.1016/j.cell.2013.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Raper SE, Chirmule N, Lee FS, Wivel NA, Bagg A, Gao G, Wilson JM, Batshaw ML. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Mol Genet Metab. 2003;80:148–158. doi: 10.1016/j.ymgme.2003.08.016. [DOI] [PubMed] [Google Scholar]
  320. Rasys AM, Park S, Ball RE, Alcala AJ, Lauderdale JD, Menke DB. CRISPR-Cas9 gene editing in lizards through microinjection of unfertilized oocytes. Cell Rep. 2019;28:2288–2292. doi: 10.1016/j.celrep.2019.07.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Rees HA, Komor AC, Yeh WH, Caetano-Lopes J, Warman M, Edge ASB, Liu DR. Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery. Nat Commun. 2017;8:15790. doi: 10.1038/ncomms15790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Reik, A., Zhou, Y., Wagner, J., Hamlett, A., Mendel, M., Liu, P.Q., Lee, G., Paschon, D., Rebar, E., and Ando, D. (2008). Zinc finger nucleases targeting the glucocorticoid receptor allow IL-13 zetakine transgenic CTLs to kill glioblastoma cells in vivo in the presence of immunosuppressing glucocorticoids. In: AACR Annual Meeting. San Diego, CA.
  323. Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res. 2017;23:2255–2266. doi: 10.1158/1078-0432.CCR-16-1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Ren Q, Li C, Yuan P, Cai C, Zhang L, Luo GG, Wei W. A Dual-reporter system for real-time monitoring and high-throughput CRISPR/Cas9 library screening of the hepatitis C virus. Sci Rep. 2015;5:8865. doi: 10.1038/srep08865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Replogle JM, Norman TM, Xu A, Hussmann JA, Chen J, Cogan J Z, Meer EJ, Terry JM, Riordan DP, Srinivas N, et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing. Nat Biotechnol. 2020;38:954–961. doi: 10.1038/s41587-020-0470-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Richter MF, Zhao KT, Eton E, Lapinaite A, Newby GA, Thuronyi BW, Wilson C, Koblan LW, Zeng J, Bauer DE, et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat Biotechnol. 2020;38:883–891. doi: 10.1038/s41587-020-0453-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Rodriguez CM, Todd PK. New pathologic mechanisms in nucleotide repeat expansion disorders. Neurobiol Dis. 2019;130:104515. doi: 10.1016/j.nbd.2019.104515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  328. Rothgangl T, Dennis MK, Lin PJC, Oka R, Witzigmann D, Villiger L, Qi W, Hruzova M, Kissling L, Lenggenhager D, et al. In vivo adenine base editing of PCSK9 in macaques reduces LDL cholesterol levels. Nat Biotechnol. 2021;39:949–957. doi: 10.1038/s41587-021-00933-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Rouet P, Smih F, Jasin M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol Cell Biol. 1994;14:8096–8106. doi: 10.1128/mcb.14.12.8096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  330. Ruan GX, Barry E, Yu D, Lukason M, Cheng SH, Scaria A. CRISPR/Cas9-mediated genome editing as a therapeutic approach for leber congenital amaurosis 10. Mol Ther. 2017;25:331–341. doi: 10.1016/j.ymthe.2016.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  331. Rubin AJ, Parker KR, Satpathy AT, Qi Y, Wu B, Ong AJ, Mumbach MR, Ji AL, Kim DS, Cho SW, et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell. 2019;176:361–376. doi: 10.1016/j.cell.2018.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Rudich P, Lamitina T. Models and mechanisms of repeat expansion disorders: a worm’s eye view. J Genet. 2018;97:665–677. doi: 10.1007/s12041-018-0950-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, Lim WA, Marson A. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep. 2017;7:737. doi: 10.1038/s41598-017-00462-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  334. Russell S, Bennett J, Wellman JA, Chung DC, Yu ZF, Tillman A, Wittes J, Pappas J, Elci O, McCague S, et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65 -mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet. 2017;390:849–860. doi: 10.1016/S0140-6736(17)31868-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  335. Ryczek N, Hryhorowicz M, Zeyland J, Lipiński D, Słomski R. CRISPR/Cas technology in pig-to-human xenotransplantation research. Int J Mol Sci. 2021;22:3196. doi: 10.3390/ijms22063196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  336. Ryu SM, Koo T, Kim K, Lim K, Baek G, Kim ST, Kim HS, Kim DE, Lee H, Chung E, et al. Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy. Nat Biotechnol. 2018;36:536–539. doi: 10.1038/nbt.4148. [DOI] [PubMed] [Google Scholar]
  337. Saayman SM, Lazar DC, Scott TA, Hart JR, Takahashi M, Burnett JC, Planelles V, Morris KV, Weinberg MS. Potent and targeted activation of latent HIV-1 using the CRISPR/dCas9 activator complex. Mol Ther. 2016;24:488–498. doi: 10.1038/mt.2015.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Salter JD, Bennett RP, Smith HC. The APOBEC Protein family: united by structure, divergent in function. Trends Biochem Sci. 2016;41:578–594. doi: 10.1016/j.tibs.2016.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: current perspectives. Adv Drug Deliv Rev. 2020;154–155:37–63. doi: 10.1016/j.addr.2020.06.002. [DOI] [PubMed] [Google Scholar]
  340. Sánchez-Rivera FJ, Papagiannakopoulos T, Romero R, Tammela T, Bauer MR, Bhutkar A, Joshi NS, Subbaraj L, Bronson RT, Xue W, et al. Rapid modelling of cooperating genetic events in cancer through somatic genome editing. Nature. 2014;516:428–431. doi: 10.1038/nature13906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  341. Sato K, Oiwa R, Kumita W, Henry R, Sakuma T, Ito R, Nozu R, Inoue T, Katano I, Sato K, et al. Generation of a nonhuman primate model of severe combined immunodeficiency using highly efficient genome editing. Cell Stem Cell. 2016;19:127–138. doi: 10.1016/j.stem.2016.06.003. [DOI] [PubMed] [Google Scholar]
  342. Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, Radisic M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials. 2019;198:3–26. doi: 10.1016/j.biomaterials.2018.09.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Schmid-Burgk JL, Gao L, Li D, Gardner Z, Strecker J, Lash B, Zhang F. Highly parallel profiling of Cas9 variant specificity. Mol Cell. 2020;78:794–800. doi: 10.1016/j.molcel.2020.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  344. Seidah NG, Awan Z, Chrétien M, Mbikay M. PCSK9: a key modulator of cardiovascular health. Circ Res. 2014;114:1022–1036. doi: 10.1161/CIRCRESAHA.114.301621. [DOI] [PubMed] [Google Scholar]
  345. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelsen TS, Heckl D, Ebert BL, Root DE, Doench JG, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014;343:84–87. doi: 10.1126/science.1247005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Shalem O, Sanjana NE, Zhang F. High-throughput functional genomics using CRISPR-Cas9. Nat Rev Genet. 2015;16:299–311. doi: 10.1038/nrg3899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  347. Shams A, Higgins SA, Fellmann C, Laughlin TG, Oakes BL, Lew R, Kim S, Lukarska M, Arnold M, Staahl BT, et al. Comprehensive deletion landscape of CRISPR-Cas9 identifies minimal RNA-guided DNA-binding modules. Nat Commun. 2021;12:5664. doi: 10.1038/s41467-021-25992-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  348. Shao N, Han X, Song Y, Zhang P, Qin L. CRISPR-Cas12a coupled with platinum nanoreporter for visual quantification of SNVs on a volumetric bar-chart chip. Anal Chem. 2019;91:12384–12391. doi: 10.1021/acs.analchem.9b02925. [DOI] [PubMed] [Google Scholar]
  349. Shao Y, Guan Y, Wang L, Qiu Z, Liu M, Chen Y, Wu L, Li Y, Ma X, Liu M, et al. CRISPR/Cas-mediated genome editing in the rat via direct injection of one-cell embryos. Nat Protoc. 2014;9:2493–2512. doi: 10.1038/nprot.2014.171. [DOI] [PubMed] [Google Scholar]
  350. Sharon E, Chen SAA, Khosla NM, Smith JD, Pritchard JK, Fraser HB. Functional genetic variants revealed by massively parallel precise genome editing. Cell. 2018;175:544–557. doi: 10.1016/j.cell.2018.08.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  351. Shen B, Zhang W, Zhang J, Zhou J, Wang J, Chen L, Wang L, Hodgkins A, Iyer V, Huang X, et al. Efficient genome modification by CRISPR-Cas9 nickase with minimal off-target effects. Nat Methods. 2014;11:399–402. doi: 10.1038/nmeth.2857. [DOI] [PubMed] [Google Scholar]
  352. Shen JP, Zhao D, Sasik R, Luebeck J, Birmingham A, Bojorquez-Gomez A, Licon K, Klepper K, Pekin D, Beckett AN, et al. Combinatorial CRISPR-Cas9 screens for de novo mapping of genetic interactions. Nat Methods. 2017;14:573–576. doi: 10.1038/nmeth.4225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Shi K, Xie S, Tian R, Wang S, Lu Q, Gao D, Lei C, Zhu H, Nie Z. A CRISPR-Cas autocatalysis-driven feedback amplification network for supersensitive DNA diagnostics. Sci Adv. 2021;7:eabc7802. doi: 10.1126/sciadv.abc7802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Shiloh Y, Ziv Y. The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. Nat Rev Mol Cell Biol. 2013;14:197–210. doi: 10.1038/nrm3546. [DOI] [PubMed] [Google Scholar]
  355. Shinoda H, Taguchi Y, Nakagawa R, Makino A, Okazaki S, Nakano M, Muramoto Y, Takahashi C, Takahashi I, Ando J, et al. Amplification-free RNA detection with CRISPR-Cas13. Commun Biol. 2021;4:476. doi: 10.1038/s42003-021-02001-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Shirley JL, de Jong YP, Terhorst C, Herzog RW. Immune responses to viral gene therapy vectors. Mol Ther. 2020;28:709–722. doi: 10.1016/j.ymthe.2020.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Simeonov DR, Gowen BG, Boontanrart M, Roth TL, Gagnon JD, Mumbach MR, Satpathy AT, Lee Y, Bray NL, Chan AY, et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature. 2017;549:111–115. doi: 10.1038/nature23875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  358. Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F. Rationally engineered Cas9 nucleases with improved specificity. Science. 2016;351:84–88. doi: 10.1126/science.aad5227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Song CQ, Jiang T, Richter M, Rhym LH, Koblan LW, Zafra MP, Schatoff EM, Doman JL, Cao Y, Dow LE, et al. Adenine base editing in an adult mouse model of tyrosinaemia. Nat Biomed Eng. 2020;4:125–130. doi: 10.1038/s41551-019-0357-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Sternberg SH, Redding S, Jinek M, Greene EC, Doudna JA. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature. 2014;507:62–67. doi: 10.1038/nature13011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Strecker J, Jones S, Koopal B, Schmid-Burgk J, Zetsche B, Gao L, Makarova KS, Koonin EV, Zhang F. Engineering of CRISPR-Cas12b for human genome editing. Nat Commun. 2019;10:212. doi: 10.1038/s41467-018-08224-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  362. Strecker J, Ladha A, Gardner Z, Schmid-Burgk JL, Makarova KS, Koonin EV, Zhang F. RNA-guided DNA insertion with CRISPR-associated transposases. Science. 2019;365:48–53. doi: 10.1126/science.aax9181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  363. Swiech L, Heidenreich M, Banerjee A, Habib N, Li Y, Trombetta J, Sur M, Zhang F. In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9. Nat Biotechnol. 2015;33:102–106. doi: 10.1038/nbt.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Tanenbaum ME, Gilbert LA, Qi LS, Weissman JS, Vale RD. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell. 2014;159:635–646. doi: 10.1016/j.cell.2014.09.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Tao D, Liu J, Nie X, Xu B, Tran-Thi TN, Niu L, Liu X, Ruan J, Lan X, Peng G, et al. Application of CRISPR-Cas12a enhanced fluorescence assay coupled with nucleic acid amplification for the sensitive detection of african swine fever virus. ACS Synth Biol. 2020;9:2339–2350. doi: 10.1021/acssynbio.0c00057. [DOI] [PubMed] [Google Scholar]
  366. Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt S K, Surosky RT, Giedlin MA, Nichol G, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014;370:901–910. doi: 10.1056/NEJMoa1300662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  367. Thompson AA, Walters MC, Kwiatkowski J, Rasko JEJ, Ribeil J A, Hongeng S, Magrin E, Schiller GJ, Payen E, Semeraro M, et al. Gene therapy in patients with transfusion-dependent β-thalassemia. N Engl J Med. 2018;378:1479–1493. doi: 10.1056/NEJMoa1705342. [DOI] [PubMed] [Google Scholar]
  368. Thuronyi BW, Koblan LW, Levy JM, Yeh WH, Zheng C, Newby GA, Wilson C, Bhaumik M, Shubina-Oleinik O, Holt JR, et al. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat Biotechnol. 2019;37:1070–1079. doi: 10.1038/s41587-019-0193-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. Tian T, Shu B, Jiang Y, Ye M, Liu L, Guo Z, Han Z, Wang Z, Zhou X. An ultralocalized Cas13a assay enables universal and nucleic acid amplification-free single-molecule RNA diagnostics. ACS Nano. 2021;15:1167–1178. doi: 10.1021/acsnano.0c08165. [DOI] [PubMed] [Google Scholar]
  370. Tibbetts RS, Brumbaugh KM, Williams JM, Sarkaria JN, Cliby W A, Shieh SY, Taya Y, Prives C, Abraham RT. A role for ATR in the DNA damage-induced phosphorylation of p53. Genes Dev. 1999;13:152–157. doi: 10.1101/gad.13.2.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  371. Torikai H, Reik A, Liu PQ, Zhou Y, Zhang L, Maiti S, Huls H, Miller JC, Kebriaei P, Rabinovitch B, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119:5697–5705. doi: 10.1182/blood-2012-01-405365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Torikai H, Reik A, Soldner F, Warren EH, Yuen C, Zhou Y, Crossland DL, Huls H, Littman N, Zhang Z, et al. Toward eliminating HLA class I expression to generate universal cells from allogeneic donors. Blood. 2013;122:1341–1349. doi: 10.1182/blood-2013-03-478255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Torres-Ruiz R, Rodriguez-Perales S. CRISPR-Cas9: a revolutionary tool for cancer modelling. Int J Mol Sci. 2015;16:22151–22168. doi: 10.3390/ijms160922151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  374. Traxler EA, Yao Y, Wang YD, Woodard KJ, Kurita R, Nakamura Y, Hughes JR, Hardison RC, Blobel GA, Li C, et al. A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat Med. 2016;22:987–990. doi: 10.1038/nm.4170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Tsai SQ, Wyvekens N, Khayter C, Foden JA, Thapar V, Reyon D, Goodwin MJ, Aryee MJ, Joung JK. Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing. Nat Biotechnol. 2014;32:569–576. doi: 10.1038/nbt.2908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV, Thapar V, Wyvekens N, Khayter C, Iafrate AJ, Le LP, et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat Biotechnol. 2015;33:187–197. doi: 10.1038/nbt.3117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Tsai SQ, Nguyen NT, Malagon-Lopez J, Topkar VV, Aryee MJ, Joung JK. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat Methods. 2017;14:607–614. doi: 10.1038/nmeth.4278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Tsuchiya Y, Minami Y, Umemura Y, Watanabe H, Ono D, Nakamura W, Takahashi T, Honma S, Kondoh G, Matsuishi T, et al. Disruption of MeCP2 attenuates circadian rhythm in CRISPR/Cas9-based Rett syndrome model mouse. Genes Cells. 2015;20:992–1005. doi: 10.1111/gtc.12305. [DOI] [PubMed] [Google Scholar]
  379. Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, Augustus S, Jamieson AC, Porteus MH, Gregory PD, Holmes MC. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature. 2005;435:646–651. doi: 10.1038/nature03556. [DOI] [PubMed] [Google Scholar]
  380. Van Duyne GD. Cre recombinase. Microbiol Spectr. 2015;3:MDNA3. doi: 10.1128/microbiolspec.MDNA3-0014-2014. [DOI] [PubMed] [Google Scholar]
  381. Veres A, Gosis BS, Ding Q, Collins R, Ragavendran A, Brand H, Erdin S, Cowan CA, Talkowski ME, Musunuru K. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell. 2014;15:27–30. doi: 10.1016/j.stem.2014.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Villiger L, Grisch-Chan HM, Lindsay H, Ringnalda F, Pogliano CB, Allegri G, Fingerhut R, Häberle J, Matos J, Robinson MD, et al. Treatment of a metabolic liver disease by in vivo genome base editing in adult mice. Nat Med. 2018;24:1519–1525. doi: 10.1038/s41591-018-0209-1. [DOI] [PubMed] [Google Scholar]
  383. Villiger L, Rothgangl T, Witzigmann D, Oka R, Lin PJC, Qi W, Janjuha S, Berk C, Ringnalda F, Beattie MB, et al. In vivo cytidine base editing of hepatocytes without detectable off-target mutations in RNA and DNA. Nat Biomed Eng. 2021;5:179–189. doi: 10.1038/s41551-020-00671-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Vo PLH, Ronda C, Klompe SE, Chen EE, Acree C, Wang HH, Sternberg SH. CRISPR RNA-guided integrases for high-efficiency, multiplexed bacterial genome engineering. Nat Biotechnol. 2021;39:480–489. doi: 10.1038/s41587-020-00745-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Wagner DL, Amini L, Wendering DJ, Burkhardt LM, Akyüz L, Reinke P, Volk HD, Schmueck-Henneresse M. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nat Med. 2019;25:242–248. doi: 10.1038/s41591-018-0204-6. [DOI] [PubMed] [Google Scholar]
  386. Wallace J, Hu R, Mosbruger TL, Dahlem TJ, Stephens WZ, Rao D S, Round JL, O’Connell RM. Genome-wide CRISPR-Cas9 screen identifies microRNAs that regulate myeloid leukemia cell growth. PLoS ONE. 2016;11:e0153689. doi: 10.1371/journal.pone.0153689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  387. Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358–378. doi: 10.1038/s41573-019-0012-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Wang D, Zhang F, Gao G. CRISPR-based therapeutic genome editing: strategies and in vivo delivery by AAV vectors. Cell. 2020;181:136–150. doi: 10.1016/j.cell.2020.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  389. Wang G, Chow RD, Ye L, Guzman CD, Dai X, Dong MB, Zhang F, Sharp PA, Platt RJ, Chen S. Mapping a functional cancer genome atlas of tumor suppressors in mouse liver using AAV-CRISPR-mediated direct in vivo screening. Sci Adv. 2018;4:eaao5508. doi: 10.1126/sciadv.aao5508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  390. Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, Jaenisch R. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013;153:910–918. doi: 10.1016/j.cell.2013.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Wang, J., Chen, Q., Li, S., and Li, X. (2017a). An application of a Cas protein, and a method and kit for detecting a target nucleic acid molecule. CN111094588A.
  392. Wang J, Quake SR. RNA-guided endonuclease provides a therapeutic strategy to cure latent herpesviridae infection. Proc Natl Acad Sci USA. 2014;111:13157–13162. doi: 10.1073/pnas.1410785111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  393. Wang K, Jin Q, Ruan D, Yang Y, Liu Q, Wu H, Zhou Z, Ouyang Z, Liu Z, Zhao Y, et al. Cre-dependent Cas9-expressing pigs enable efficient in vivo genome editing. Genome Res. 2017;27:2061–2071. doi: 10.1101/gr.222521.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  394. Wang L, Li L, Ma Y, Hu H, Li Q, Yang Y, Liu W, Yin S, Li W, Fu B, et al. Reactivation of γ-globin expression through Cas9 or base editor to treat β-hemoglobinopathies. Cell Res. 2020;30:276–278. doi: 10.1038/s41422-019-0267-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  395. Wang L, Smith J, Breton C, Clark P, Zhang J, Ying L, Che Y, Lape J, Bell P, Calcedo R, et al. Meganuclease targeting of PCSK9 in macaque liver leads to stable reduction in serum cholesterol. Nat Biotechnol. 2018;36:717–725. doi: 10.1038/nbt.4182. [DOI] [PubMed] [Google Scholar]
  396. Wang L, Xue W, Yan L, Li X, Wei J, Chen M, Wu J, Yang B, Yang L, Chen J. Enhanced base editing by co-expression of free uracil DNA glycosylase inhibitor. Cell Res. 2017;27:1289–1292. doi: 10.1038/cr.2017.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  397. Wang L, Xue W, Zhang H, Gao R, Qiu H, Wei J, Zhou L, Lei Y N, Wu X, Li X, et al. Eliminating base-editor-induced genome-wide and transcriptome-wide off-target mutations. Nat Cell Biol. 2021;23:552–563. doi: 10.1038/s41556-021-00671-4. [DOI] [PubMed] [Google Scholar]
  398. Wang L, Yang Y, Breton CA, White J, Zhang J, Che Y, Saveliev A, McMenamin D, He Z, Latshaw C, et al. CRISPR/Cas9-mediated in vivo gene targeting corrects hemostasis in newborn and adult factor IX-knockout mice. Blood. 2019;133:2745–2752. doi: 10.1182/blood.2019000790. [DOI] [PubMed] [Google Scholar]
  399. Wang M, Chen K, Wu Q, Peng R, Zhang R, Li J. RCasFISH: CRISPR/dCas9-mediated in situ imaging of mRNA transcripts in fixed cells and tissues. Anal Chem. 2020;92:2468–2475. doi: 10.1021/acs.analchem.9b03797. [DOI] [PubMed] [Google Scholar]
  400. Wang Q, Zhang B, Xu X, Long F, Wang J. CRISPR-typing PCR (ctPCR), a new Cas9-based DNA detection method. Sci Rep. 2018;8:14126. doi: 10.1038/s41598-018-32329-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  401. Wang R, Zhao X, Chen X, Qiu X, Qing G, Zhang H, Zhang L, Hu X, He Z, Zhong D, et al. Rolling circular amplification (RCA)-assisted CRISPR/Cas9 cleavage (RACE) for Highly specific detection of multiple extracellular vesicle microRNAs. Anal Chem. 2020;92:2176–2185. doi: 10.1021/acs.analchem.9b04814. [DOI] [PubMed] [Google Scholar]
  402. Wang T, Liu Y, Sun HH, Yin BC, Ye BC. An RNA-guided Cas9 nickase-based method for universal isothermal DNA amplification. Angew Chem Int Ed. 2019;58:5382–5386. doi: 10.1002/anie.201901292. [DOI] [PubMed] [Google Scholar]
  403. Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014;343:80–84. doi: 10.1126/science.1246981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Wang, X., Huang, R., Zhang, L., Li, S., Luo, J., Gu, Y., Chen, Z., Zheng, Q., Chao, T., Zheng, W., et al. (2018d). A severe atherosclerosis mouse model on the resistant NOD background. Dis Model Mech 11. [DOI] [PMC free article] [PubMed]
  405. Wang X, Li J, Wang Y, Yang B, Wei J, Wu J, Wang R, Huang X, Chen J, Yang L. Efficient base editing in methylated regions with a human APOBEC3A-Cas9 fusion. Nat Biotechnol. 2018;36:946–949. doi: 10.1038/nbt.4198. [DOI] [PubMed] [Google Scholar]
  406. Wang X, Xiong E, Tian T, Cheng M, Lin W, Wang H, Zhang G, Sun J, Zhou X. Clustered regularly interspaced short palindromic repeats/Cas9-mediated lateral flow nucleic acid assay. ACS Nano. 2020;14:2497–2508. doi: 10.1021/acsnano.0c00022. [DOI] [PubMed] [Google Scholar]
  407. Wang X, Ding C, Yu W, Wang Y, He S, Yang B, Xiong YC, Wei J, Li J, Liang J, et al. Cas12a base editors induce efficient and specific editing with low DNA damage response. Cell Rep. 2020;31:107723. doi: 10.1016/j.celrep.2020.107723. [DOI] [PubMed] [Google Scholar]
  408. Wang X, Yu H, Lei A, Zhou J, Zeng W, Zhu H, Dong Z, Niu Y, Shi B, Cai B, et al. Generation of gene-modified goats targeting MSTN and FGF5 via zygote injection of CRISPR/Cas9 system. Sci Rep. 2015;5:13878. doi: 10.1038/srep13878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  409. Wang Y, Du Y, Shen B, Zhou X, Li J, Liu Y, Wang J, Zhou J, Hu B, Kang N, et al. Efficient generation of gene-modified pigs via injection of zygote with Cas9/sgRNA. Sci Rep. 2015;5:8256. doi: 10.1038/srep08256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  410. Wang Y, Du Y, Zhou X, Wang L, Li J, Wang F, Huang Z, Huang X, Wei H. Efficient generation of B2m-null pigs via injection of zygote with TALENs. Sci Rep. 2016;6:38854. doi: 10.1038/srep38854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  411. Weber J, Öllinger R, Friedrich M, Ehmer U, Barenboim M, Steiger K, Heid I, Mueller S, Maresch R, Engleitner T, et al. CRISPR/Cas9 somatic multiplex-mutagenesis for high-throughput functional cancer genomics in mice. Proc Natl Acad Sci USA. 2015;112:13982–13987. doi: 10.1073/pnas.1512392112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Weber ND, Stone D, Sedlak RH, De Silva Feelixge HS, Roychoudhury P, Schiffer JT, Aubert M, Jerome KR. AAV-mediated delivery of zinc finger nucleases targeting hepatitis B virus inhibits active replication. PLoS ONE. 2014;9:e97579. doi: 10.1371/journal.pone.0097579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  413. Wei JJ. Accurate and sensitive analysis of Staphylococcus aureus through CRISPR-Cas12a based recycling signal amplification cascades for early diagnosis of skin and soft tissue infections. J Microbiol Methods. 2021;183:106167. doi: 10.1016/j.mimet.2021.106167. [DOI] [PubMed] [Google Scholar]
  414. Wei Y, Zhou Y, Liu Y, Ying W, Lv R, Zhao Q, Zhou H, Zuo E, Sun Y, Yang H, et al. Indiscriminate ssDNA cleavage activity of CRISPR-Cas12a induces no detectable off-target effects in mouse embryos. Protein Cell. 2021;12:741–745. doi: 10.1007/s13238-021-00824-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  415. Wernike K, Keller M, Conraths FJ, Mettenleiter TC, Groschup MH, Beer M. Pitfalls in SARS-CoV-2 PCR diagnostics. Transbound Emerg Dis. 2021;68:253–257. doi: 10.1111/tbed.13684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  416. Wilde JJ, Aida T, Del Rosario RCH, Kaiser T, Qi P, Wienisch M, Zhang Q, Colvin S, Feng G. Efficient embryonic homozygous gene conversion via RAD51-enhanced interhomolog repair. Cell. 2021;184:3267–3280. doi: 10.1016/j.cell.2021.04.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. Wilkinson AC, Dever DP, Baik R, Camarena J, Hsu I, Charlesworth CT, Morita C, Nakauchi H, Porteus MH. Cas9-AAV6 gene correction of beta-globin in autologous HSCs improves sickle cell disease erythropoiesis in mice. Nat Commun. 2021;12:686. doi: 10.1038/s41467-021-20909-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. Witzigmann D, Kulkarni JA, Leung J, Chen S, Cullis PR, van der Meel R. Lipid nanoparticle technology for therapeutic gene regulation in the liver. Adv Drug Deliv Rev. 2020;159:344–363. doi: 10.1016/j.addr.2020.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  419. Wong ASL, Choi GCG, Cui CH, Pregernig G, Milani P, Adam M, Perli SD, Kazer SW, Gaillard A, Hermann M, et al. Multiplexed barcoded CRISPR-Cas9 screening enabled by CombiGEM. Proc Natl Acad Sci USA. 2016;113:2544–2549. doi: 10.1073/pnas.1517883113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  420. Wu WH, Tsai YT, Justus S, Lee TT, Zhang L, Lin CS, Bassuk A G, Mahajan VB, Tsang SH. CRISPR repair reveals causative mutation in a preclinical model of retinitis pigmentosa. Mol Ther. 2016;24:1388–1394. doi: 10.1038/mt.2016.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Wu Y, Zeng J, Roscoe BP, Liu P, Yao Q, Lazzarotto CR, Clement K, Cole MA, Luk K, Baricordi C, et al. Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat Med. 2019;25:776–783. doi: 10.1038/s41591-019-0401-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. Xiao AT, Tong YX, Zhang S. False negative of RT-PCR and prolonged nucleic acid conversion in COVID-19: Rather than recurrence. J Med Virol. 2020;92:1755–1756. doi: 10.1002/jmv.25855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. Xing S, Lu Z, Huang Q, Li H, Wang Y, Lai Y, He Y, Deng M, Liu W. An ultrasensitive hybridization chain reaction-amplified CRISPR-Cas12a aptasensor for extracellular vesicle surface protein quantification. Theranostics. 2020;10:10262–10273. doi: 10.7150/thno.49047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Xiong Y, Zhang J, Yang Z, Mou Q, Ma Y, Xiong Y, Lu Y. Functional DNA regulated CRISPR-Cas12a sensors for point-of-care diagnostics of non-nucleic-acid targets. J Am Chem Soc. 2020;142:207–213. doi: 10.1021/jacs.9b09211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. Xu C, Zhou Y, Xiao Q, He B, Geng G, Wang Z, Cao B, Dong X, Bai W, Wang Y, et al. Programmable RNA editing with compact CRISPR-Cas13 systems from uncultivated microbes. Nat Methods. 2021;18:499–506. doi: 10.1038/s41592-021-01124-4. [DOI] [PubMed] [Google Scholar]
  426. Xu CF, Chen GJ, Luo YL, Zhang Y, Zhao G, Lu ZD, Czarna A, Gu Z, Wang J. Rational designs of in vivo CRISPR-Cas delivery systems. Adv Drug Deliv Rev. 2021;168:3–29. doi: 10.1016/j.addr.2019.11.005. [DOI] [PubMed] [Google Scholar]
  427. Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, Wang L, Liu T, Wang X, Zhang B, et al. CRISPR-edited stem cells in a patient with HIV and acute lymphocytic leukemia. N Engl J Med. 2019;381:1240–1247. doi: 10.1056/NEJMoa1817426. [DOI] [PubMed] [Google Scholar]
  428. Xu P, Liu Z, Liu Y, Ma H, Xu Y, Bao Y, Zhu S, Cao Z, Wu Z, Zhou Z, et al. Genome-wide interrogation of gene functions through base editor screens empowered by barcoded sgRNAs. Nat Biotechnol. 2021;39:1403–1413. doi: 10.1038/s41587-021-00944-1. [DOI] [PubMed] [Google Scholar]
  429. Xu P, Tong Y, Liu XZ, Wang TT, Cheng L, Wang BY, Lv X, Huang Y, Liu DP. Both TALENs and CRISPR/Cas9 directly target the HBB IVS2–654 (C>T) mutation in β-thalassemia-derived iPSCs. Sci Rep. 2015;5:12065. doi: 10.1038/srep12065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  430. Xu W. Microinjection and micromanipulation: a historical perspective. In: Liu C, Du Y, editors. Microinjection. Methods in Molecular Biology. New York: Humana Press; 2019. pp. 1–16. [DOI] [PubMed] [Google Scholar]
  431. Xue W, Chen S, Yin H, Tammela T, Papagiannakopoulos T, Joshi N S, Cai W, Yang G, Bronson R, Crowley DG, et al. CRISPR-mediated direct mutation of cancer genes in the mouse liver. Nature. 2014;514:380–384. doi: 10.1038/nature13589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  432. Yan H, Niimi M, Matsuhisa F, Zhou H, Kitajima S, Chen Y, Wang C, Yang X, Yao J, Yang D, et al. Apolipoprotein CIII deficiency protects against atherosclerosis in knockout rabbits. Arterioscler Thromb Vasc Biol. 2020;40:2095–2107. doi: 10.1161/ATVBAHA.120.314368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Yan S, Tu Z, Liu Z, Fan N, Yang H, Yang S, Yang W, Zhao Y, Ouyang Z, Lai C, et al. A Huntingtin knockin pig model recapitulates features of selective neurodegeneration in Huntington’s disease. Cell. 2018;173:989–1002. doi: 10.1016/j.cell.2018.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Yang B, Li X, Lei L, Chen J. APOBEC: from mutator to editor. J Genet Genomics. 2017;44:423–437. doi: 10.1016/j.jgg.2017.04.009. [DOI] [PubMed] [Google Scholar]
  435. Yang, D., Zhang, J., Xu, J., Zhu, T., Fan, Y., Fan, J., and Chen, Y.E. (2013a). Production of apolipoprotein C-III knockout rabbits using zinc finger nucleases. J Vis Exp doi: 10.3791/50957. [DOI] [PMC free article] [PubMed]
  436. Yang H, Wang H, Shivalila CS, Cheng AW, Shi L, Jaenisch R. One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell. 2013;154:1370–1379. doi: 10.1016/j.cell.2013.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  437. Yang H, Gao P, Rajashankar KR, Patel DJ. PAMdependent target DNA recognition and cleavage by C2c1 CRISPR-Cas endonuclease. Cell. 2016;167:1814–1828. doi: 10.1016/j.cell.2016.11.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Yang L, Yang B, Chen J. One prime for all editing. Cell. 2019;179:1448–1450. doi: 10.1016/j.cell.2019.11.030. [DOI] [PubMed] [Google Scholar]
  439. Yang L, Wang L, Huo Y, Chen X, Yin S, Hu Y, Zhang X, Zheng R, Geng H, Han H, et al. Amelioration of an inherited metabolic liver disease through creation of a de novo start codon by cytidine base editing. Mol Ther. 2020;28:1673–1683. doi: 10.1016/j.ymthe.2020.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  440. Ye L, Park JJ, Dong MB, Yang Q, Chow RD, Peng L, Du Y, Guo J, Dai X, Wang G, et al. In vivo CRISPR screening in CD8 T cells with AAV-Sleeping Beauty hybrid vectors identifies membrane targets for improving immunotherapy for glioblastoma. Nat Biotechnol. 2019;37:1302–1313. doi: 10.1038/s41587-019-0246-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Yeh, W.H., Shubina-Oleinik, O., Levy, J.M., Pan, B., Newby, G.A., Wornow, M., Burt, R., Chen, J.C., Holt, J.R., and Liu, D.R. (2020). In vivo base editing restores sensory transduction and transiently improves auditory function in a mouse model of recessive deafness. Sci Transl Med 12. [DOI] [PMC free article] [PubMed]
  442. Yin D, Ling S, Wang D, Dai Y, Jiang H, Zhou X, Paludan SR, Hong J, Cai Y. Targeting herpes simplex virus with CRISPR-Cas9 cures herpetic stromal keratitis in mice. Nat Biotechnol. 2021;39:567–577. doi: 10.1038/s41587-020-00781-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  443. Yin H, Xue W, Chen S, Bogorad RL, Benedetti E, Grompe M, Koteliansky V, Sharp PA, Jacks T, Anderson DG. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol. 2014;32:551–553. doi: 10.1038/nbt.2884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  444. Yin H, Song CQ, Dorkin JR, Zhu LJ, Li Y, Wu Q, Park A, Yang J, Suresh S, Bizhanova A, et al. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat Biotechnol. 2016;34:328–333. doi: 10.1038/nbt.3471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  445. Yin H, Kauffman KJ, Anderson DG. Delivery technologies for genome editing. Nat Rev Drug Discov. 2017;16:387–399. doi: 10.1038/nrd.2016.280. [DOI] [PubMed] [Google Scholar]
  446. Yin H, Song CQ, Suresh S, Wu Q, Walsh S, Rhym LH, Mintzer E, Bolukbasi MF, Zhu LJ, Kauffman K, et al. Structure-guided chemical modification of guide RNA enables potent non-viral in vivo genome editing. Nat Biotechnol. 2017;35:1179–1187. doi: 10.1038/nbt.4005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  447. Yin, S., Ma, L., Shao, T., Zhang, M., Guan, Y., Wang, L., Hu, Y., Chen, X., Han, H., Shen, N., et al. (2020). Enhanced genome editing to ameliorate a genetic metabolic liver disease through co-delivery of adeno-associated virus receptor. Sci China Life Sci doi: 10.1007/s11427-020-1744-6. [DOI] [PubMed]
  448. Yuan, F., Guo, L., Park, K.H., Woollard, J.R., Taek-Geun, K., Jiang, K., Melkamu, T., Zang, B., Smith, S.L., Fahrenkrug, S.C., et al. (2018). Ossabaw pigs with a PCSK9 gain-of-function mutation develop accelerated coronary atherosclerotic lesions: a novel model for preclinical studies. J Am Heart Assoc 7. [DOI] [PMC free article] [PubMed]
  449. Yue H, Shu B, Tian T, Xiong E, Huang M, Zhu D, Sun J, Liu Q, Wang S, Li Y, et al. Droplet Cas12a assay enables DNA quantification from unamplified samples at the single-molecule level. Nano Lett. 2021;21:4643–4653. doi: 10.1021/acs.nanolett.1c00715. [DOI] [PubMed] [Google Scholar]
  450. Zabaleta N, Barberia M, Martin-Higueras C, Zapata-Linares N, Betancor I, Rodriguez S, Martinez-Turrillas R, Torella L, Vales A, Olagüe C, et al. CRISPR/Cas9-mediated glycolate oxidase disruption is an efficacious and safe treatment for primary hyperoxaluria type I. Nat Commun. 2018;9:5454. doi: 10.1038/s41467-018-07827-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  451. Zeng J, Wu Y, Ren C, Bonanno J, Shen AH, Shea D, Gehrke JM, Clement K, Luk K, Yao Q, et al. Therapeutic base editing of human hematopoietic stem cells. Nat Med. 2020;26:535–541. doi: 10.1038/s41591-020-0790-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  452. Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, Essletzbichler P, Volz SE, Joung J, van der Oost J, Regev A, et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. 2015;163:759–771. doi: 10.1016/j.cell.2015.09.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. Zhang B, Wang Q, Xu X, Xia Q, Long F, Li W, Shui Y, Xia X, Wang J. Detection of target DNA with a novel Cas9/sgRNAs-associated reverse PCR (CARP) technique. Anal Bioanal Chem. 2018;410:2889–2900. doi: 10.1007/s00216-018-0873-5. [DOI] [PubMed] [Google Scholar]
  454. Zhang D, Yan Y, Que H, Yang T, Cheng X, Ding S, Zhang X, Cheng W. CRISPR/Cas12a-mediated interfacial cleaving of hairpin DNA reporter for electrochemical nucleic acid sensing. ACS Sens. 2020;5:557–562. doi: 10.1021/acssensors.9b02461. [DOI] [PubMed] [Google Scholar]
  455. Zhang JP, Cheng XX, Zhao M, Li GH, Xu J, Zhang F, Yin MD, Meng FY, Dai XY, Fu YW, et al. Curing hemophilia A by NHEJ-mediated ectopic F8 insertion in the mouse. Genome Biol. 2019;20:276. doi: 10.1186/s13059-019-1907-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  456. Zhang K, Deng R, Teng X, Li Y, Sun Y, Ren X, Li J. Direct visualization of single-nucleotide variation in mtDNA using a CRISPR/Cas9-mediated proximity ligation assay. J Am Chem Soc. 2018;140:11293–11301. doi: 10.1021/jacs.8b05309. [DOI] [PubMed] [Google Scholar]
  457. Zhang W, Shi L, Zhao Z, Du P, Ye X, Li D, Cai Z, Han J, Cai J. Disruption of CTLA-4 expression on peripheral blood CD8+ T cell enhances anti-tumor efficacy in bladder cancer. Cancer Chemother Pharmacol. 2019;83:911–920. doi: 10.1007/s00280-019-03800-x. [DOI] [PubMed] [Google Scholar]
  458. Zhang W, Wan H, Feng G, Qu J, Wang J, Jing Y, Ren R, Liu Z, Zhang L, Chen Z, et al. SIRT6 deficiency results in developmental retardation in cynomolgus monkeys. Nature. 2018;560:661–665. doi: 10.1038/s41586-018-0437-z. [DOI] [PubMed] [Google Scholar]
  459. Zhang, X., Li, T., Ou, J., Huang, J., and Liang, P. (2021). Homology-based repair induced by CRISPR-Cas nucleases in mammalian embryo genome editing. Protein Cell doi: 10.1007/s13238-021-00838-7. [DOI] [PMC free article] [PubMed]
  460. Zhang X, Yue D, Wang Y, Zhou Y, Liu Y, Qiu Y, Tian F, Yu Y, Zhou Z, Wei W. PASTMUS: mapping functional elements at single amino acid resolution in human cells. Genome Biol. 2019;20:279. doi: 10.1186/s13059-019-1897-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  461. Zhang Y, Qian L, Wei W, Wang Y, Wang B, Lin P, Liu W, Xu L, Li X, Liu D, et al. Paired design of dCas9 as a systematic platform for the detection of featured nucleic acid sequences in pathogenic strains. ACS Synth Biol. 2017;6:211–216. doi: 10.1021/acssynbio.6b00215. [DOI] [PubMed] [Google Scholar]
  462. Zhang Y, Zhang X, Cheng C, Mu W, Liu X, Li N, Wei X, Liu X, Xia C, Wang H. CRISPR-Cas9 mediated LAG-3 disruption in CAR-T cells. Front Med. 2017;11:554–562. doi: 10.1007/s11684-017-0543-6. [DOI] [PubMed] [Google Scholar]
  463. Zhao X, Zeng L, Mei Q, Luo Y. Allosteric probe-initiated wash-free method for sensitive extracellular vesicle detection through dual cycle-assisted CRISPR-Cas12a. ACS Sens. 2020;5:2239–2246. doi: 10.1021/acssensors.0c00944. [DOI] [PubMed] [Google Scholar]
  464. Zhao X, Zhang G, Liu S, Chen X, Peng R, Dai L, Qu X, Li S, Song H, Gao Z, et al. Human neonatal fc receptor is the cellular uncoating receptor for enterovirus B. Cell. 2019;177:1553–1565. doi: 10.1016/j.cell.2019.04.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  465. Zhao X, Zhang W, Qiu X, Mei Q, Luo Y, Fu W. Rapid and sensitive exosome detection with CRISPR/Cas12a. Anal Bioanal Chem. 2020;412:601–609. doi: 10.1007/s00216-019-02211-4. [DOI] [PubMed] [Google Scholar]
  466. Zheng Q, Cai X, Tan MH, Schaffert S, Arnold CP, Gong X, Chen CZ, Huang S. Precise gene deletion and replacement using the CRISPR/Cas9 system in human cells. BioTechniques. 2014;57:115–124. doi: 10.2144/000114196. [DOI] [PubMed] [Google Scholar]
  467. Zheng R, Li Y, Wang L, Fang X, Zhang J, He L, Yang L, Li D, Geng H. CRISPR/Cas9-mediated metabolic pathway reprogramming in a novel humanized rat model ameliorates primary hyperoxaluria type 1. Kidney Int. 2020;98:947–957. doi: 10.1016/j.kint.2020.04.049. [DOI] [PubMed] [Google Scholar]
  468. Zhong H, Chen Y, Li Y, Chen R, Mardon G. CRISPR-engineered mosaicism rapidly reveals that loss of Kcnj13 function in mice mimics human disease phenotypes. Sci Rep. 2015;5:8366. doi: 10.1038/srep08366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  469. Zhou C, Sun Y, Yan R, Liu Y, Zuo E, Gu C, Han L, Wei Y, Hu X, Zeng R, et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature. 2019;571:275–278. doi: 10.1038/s41586-019-1314-0. [DOI] [PubMed] [Google Scholar]
  470. Zhou T, Huang M, Lin J, Huang R, Xing D. High-fidelity CRISPR/Cas13a trans-cleavage-triggered rolling circle amplified DNAzyme for visual profiling of microRNA. Anal Chem. 2021;93:2038–2044. doi: 10.1021/acs.analchem.0c03708. [DOI] [PubMed] [Google Scholar]
  471. Zhou W, Hu L, Ying L, Zhao Z, Chu PK, Yu XF. A CRISPR-Cas9-triggered strand displacement amplification method for ultrasensitive DNA detection. Nat Commun. 2018;9:5012. doi: 10.1038/s41467-018-07324-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  472. Zhou X, Wang L, Du Y, Xie F, Li L, Liu Y, Liu C, Wang S, Zhang S, Huang X, et al. Efficient generation of gene-modified pigs harboring precise orthologous human mutation via CRISPR/Cas9-induced homology-directed repair in zygotes. Hum Mutat. 2016;37:110–118. doi: 10.1002/humu.22913. [DOI] [PubMed] [Google Scholar]
  473. Zhou X, Xin J, Fan N, Zou Q, Huang J, Ouyang Z, Zhao Y, Zhao B, Liu Z, Lai S, et al. Generation of CRISPR/Cas9-mediated gene-targeted pigs via somatic cell nuclear transfer. Cell Mol Life Sci. 2015;72:1175–1184. doi: 10.1007/s00018-014-1744-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  474. Zhou Y, Sharma J, Ke Q, Landman R, Yuan J, Chen H, Hayden D S, Fisher Iii JW, Jiang M, Menegas W, et al. Atypical behaviour and connectivity in SHANK3-mutant macaques. Nature. 2019;570:326–331. doi: 10.1038/s41586-019-1278-0. [DOI] [PubMed] [Google Scholar]
  475. Zhou Y, Zhu S, Cai C, Yuan P, Li C, Huang Y, Wei W. High-throughput screening of a CRISPR/Cas9 library for functional genomics in human cells. Nature. 2014;509:487–491. doi: 10.1038/nature13166. [DOI] [PubMed] [Google Scholar]
  476. Zhou Z, Wei W. PrePAIRing Cas9s for screening success. Nat Biotechnol. 2018;36:147–148. doi: 10.1038/nbt.4075. [DOI] [PubMed] [Google Scholar]
  477. Zhu S, Li W, Liu J, Chen CH, Liao Q, Xu P, Xu H, Xiao T, Cao Z, Peng J, et al. Genome-scale deletion screening of human long non-coding RNAs using a paired-guide RNA CRISPR-Cas9 library. Nat Biotechnol. 2016;34:1279–1286. doi: 10.1038/nbt.3715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  478. Zhu S, Cao Z, Liu Z, He Y, Wang Y, Yuan P, Li W, Tian F, Bao Y, Wei W. Guide RNAs with embedded barcodes boost CRISPR-pooled screens. Genome Biol. 2019;20:20. doi: 10.1186/s13059-019-1628-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  479. Zhu, S., Liu, Y., Zhou, Z., Zhang, Z., Xiao, X., Liu, Z., Chen, A., Dong, X., Tian, F., Chen, S., et al. (2021). Genome-wide CRISPR activation screen identifies candidate receptors for SARS-CoV-2 entry. Sci China Life Sci doi: 10.1007/s11427-021-1990-5. [DOI] [PMC free article] [PubMed]
  480. Zimmerman PA, Buckler-White A, Alkhatib G, Spalding T, Kubofcik J, Combadiere C, Weissman D, Cohen O, Rubbert A, Lam G, et al. Inherited resistance to HIV-1 conferred by an inactivating mutation in CC chemokine receptor 5: studies in populations with contrasting clinical phenotypes, defined racial background, and quantified risk. Mol Med. 1997;3:23–36. doi: 10.1007/BF03401665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  481. Zuccaro MV, Xu J, Mitchell C, Marin D, Zimmerman R, Rana B, Weinstein E, King RT, Palmerola KL, Smith ME, et al. Allele-specific chromosome removal after Cas9 cleavage in human embryos. Cell. 2020;183:1650–1664. doi: 10.1016/j.cell.2020.10.025. [DOI] [PubMed] [Google Scholar]
  482. Zuckermann M, Hovestadt V, Knobbe-Thomsen CB, Zapatka M, Northcott PA, Schramm K, Belic J, Jones DTW, Tschida B, Moriarity B, et al. Somatic CRISPR/Cas9-mediated tumour suppressor disruption enables versatile brain tumour modelling. Nat Commun. 2015;6:7391. doi: 10.1038/ncomms8391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  483. Zuo E, Sun Y, Wei W, Yuan T, Ying W, Sun H, Yuan L, Steinmetz LM, Li Y, Yang H. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science. 2019;364:289–292. doi: 10.1126/science.aav9973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Zuo E, Sun Y, Yuan T, He B, Zhou C, Ying W, Liu J, Wei W, Zeng R, Li Y, et al. A rationally engineered cytosine base editor retains high on-target activity while reducing both DNA and RNA off-target effects. Nat Methods. 2020;17:600–604. doi: 10.1038/s41592-020-0832-x. [DOI] [PubMed] [Google Scholar]
  485. Zuris JA, Thompson DB, Shu Y, Guilinger JP, Bessen JL, Hu JH, Maeder ML, Joung JK, Chen ZY, Liu DR. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol. 2015;33:73–80. doi: 10.1038/nbt.3081. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Science China. Life Sciences are provided here courtesy of Nature Publishing Group

RESOURCES