Progress and Prospects of Gene Editing in Pluripotent Stem Cells
Abstract
:1. Introduction
2. DSB-Mediated Gene Editing by Programmable Nucleases
2.1. Zinc-Finger Nuclease (ZFN)
On- and Off-Targeting of ZFNs
2.2. Transcription Activator-like Effector Nucleases (TALENs)
2.3. CRISPR-Cas System
2.3.1. Evaluating Off-Target Effects of CRISPR-Cas9
3. Base Editors
3.1. Cytidine Base Editor (CBE)
3.2. Adenine Base Editor (ABE)
3.3. Glycosylase Base Editor (GBE) and C-to-G Base Editor (CGBE)
3.4. Adenine Transversion Editors (AYBE and AXBE)
3.5. Pros and Cons of Using Base Editors in PSCs
3.5.1. Dealing with Off-Target Effects of CBE and ABE
3.5.2. On- and Off-Targets of Other Base Editors in PSCs
4. Prime Editor (PE)
5. New Gene Editing Tools
5.1. CRISPR-Associated Transposon (CAST)
5.2. CRISPR-Associated Serine Recombinases (twinPE and PASTE)
5.3. Retron
5.4. SeLection by Essential-Gene Exon Knock-in (SLEEK)
6. Conclusions and Future Prospects
6.1. Improving Editing Efficiencies in Human PSCs
6.2. Conditional Gene Editing
6.3. CRISPR-Cas as DNA Recorders of Cell Fates
6.4. New Systems to Be Explored
6.5. Conclusions
Funding
Acknowledgments
Conflicts of Interest
References
- Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R.; et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Gulbranson, D.R.; Hou, Z.; Bolin, J.M.; Ruotti, V.; Probasco, M.D.; Smuga-Otto, K.; Howden, S.E.; Diol, N.R.; Propson, N.E.; et al. Chemically defined conditions for human iPSC derivation and culture. Nat. Methods 2011, 8, 424–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okita, K.; Matsumura, Y.; Sato, Y.; Okada, A.; Morizane, A.; Okamoto, S.; Hong, H.; Nakagawa, M.; Tanabe, K.; Tezuka, K.; et al. A more efficient method to generate integration-free human iPS cells. Nat. Methods 2011, 8, 409–412. [Google Scholar] [CrossRef] [Green Version]
- Fusaki, N.; Ban, H.; Nishiyama, A.; Saeki, K.; Hasegawa, M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2009, 85, 348–362. [Google Scholar] [CrossRef] [Green Version]
- Yu, J.; Hu, K.; Smuga-Otto, K.; Tian, S.; Stewart, R.; Slukvin, I.I.; Thomson, J.A. Human induced pluripotent stem cells free of vector and transgene sequences. Science 2009, 324, 797–801. [Google Scholar] [CrossRef] [Green Version]
- Guan, J.; Wang, G.; Wang, J.; Zhang, Z.; Fu, Y.; Cheng, L.; Meng, G.; Lyu, Y.; Zhu, J.; Li, Y.; et al. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature 2022, 605, 325–331. [Google Scholar] [CrossRef]
- Liuyang, S.; Wang, G.; Wang, Y.; He, H.; Lyu, Y.; Cheng, L.; Yang, Z.; Guan, J.; Fu, Y.; Zhu, J.; et al. Highly efficient and rapid generation of human pluripotent stem cells by chemical reprogramming. Cell Stem Cell 2023, 30, 450–459.e9. [Google Scholar] [CrossRef]
- Yamanaka, S. Pluripotent Stem Cell-Based Cell Therapy-Promise and Challenges. Cell Stem Cell 2020, 27, 523–531. [Google Scholar] [CrossRef]
- Kim, J.Y.; Nam, Y.; Rim, Y.A.; Ju, J.H. Review of the Current Trends in Clinical Trials Involving Induced Pluripotent Stem Cells. Stem Cell Rev. Rep. 2022, 18, 142–154. [Google Scholar] [CrossRef] [PubMed]
- Mazzini, L.; De Marchi, F. iPSC-based research in ALS precision medicine. Cell Stem Cell 2023, 30, 748–749. [Google Scholar] [CrossRef] [PubMed]
- Turan, S.; Farruggio, A.P.; Srifa, W.; Day, J.W.; Calos, M.P. Precise Correction of Disease Mutations in Induced Pluripotent Stem Cells Derived from Patients with Limb Girdle Muscular Dystrophy. Mol. Ther. 2016, 24, 685–696. [Google Scholar] [CrossRef] [Green Version]
- Sumer, S.A.; Hoffmann, S.; Laue, S.; Campbell, B.; Raedecke, K.; Frajs, V.; Clauss, S.; Kaab, S.; Janssen, J.W.G.; Jauch, A.; et al. Precise Correction of Heterozygous SHOX2 Mutations in hiPSCs Derived from Patients with Atrial Fibrillation via Genome Editing and Sib Selection. Stem Cell Rep. 2020, 15, 999–1013. [Google Scholar] [CrossRef]
- Li, Y.; Hermanson, D.L.; Moriarity, B.S.; Kaufman, D.S. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell 2018, 23, 181–192.e5. [Google Scholar] [CrossRef] [Green Version]
- Mattapally, S.; Pawlik, K.M.; Fast, V.G.; Zumaquero, E.; Lund, F.E.; Randall, T.D.; Townes, T.M.; Zhang, J. Human Leukocyte Antigen Class I and II Knockout Human Induced Pluripotent Stem Cell-Derived Cells: Universal Donor for Cell Therapy. J. Am. Heart Assoc. 2018, 7, e010239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef] [PubMed]
- Aurora, M.; Spence, J.R. hPSC-derived lung and intestinal organoids as models of human fetal tissue. Dev. Biol. 2016, 420, 230–238. [Google Scholar] [CrossRef] [Green Version]
- Frum, T.; Spence, J.R. hPSC-derived organoids: Models of human development and disease. J. Mol. Med. 2021, 99, 463–473. [Google Scholar] [CrossRef]
- Huch, M.; Dorrell, C.; Boj, S.F.; van Es, J.H.; Li, V.S.; van de Wetering, M.; Sato, T.; Hamer, K.; Sasaki, N.; Finegold, M.J.; et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 2013, 494, 247–250. [Google Scholar] [CrossRef] [Green Version]
- Lancaster, M.A.; Renner, M.; Martin, C.A.; Wenzel, D.; Bicknell, L.S.; Hurles, M.E.; Homfray, T.; Penninger, J.M.; Jackson, A.P.; Knoblich, J.A. Cerebral organoids model human brain development and microcephaly. Nature 2013, 501, 373–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Driehuis, E.; Kretzschmar, K.; Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 2020, 15, 3380–3409. [Google Scholar] [CrossRef] [PubMed]
- Bar-Ephraim, Y.E.; Kretzschmar, K.; Clevers, H. Organoids in immunological research. Nat. Rev. Immunol. 2020, 20, 279–293. [Google Scholar] [CrossRef] [PubMed]
- Li, C.; Huang, J.; Yu, Y.; Wan, Z.; Chiu, M.C.; Liu, X.; Zhang, S.; Cai, J.P.; Chu, H.; Li, G.; et al. Human airway and nasal organoids reveal escalating replicative fitness of SARS-CoV-2 emerging variants. Proc. Natl. Acad. Sci. USA 2023, 120, e2300376120. [Google Scholar] [CrossRef]
- Achberger, K.; Probst, C.; Haderspeck, J.; Bolz, S.; Rogal, J.; Chuchuy, J.; Nikolova, M.; Cora, V.; Antkowiak, L.; Haq, W.; et al. Merging organoid and organ-on-a-chip technology to generate complex multi-layer tissue models in a human retina-on-a-chip platform. eLife 2019, 8, e46188. [Google Scholar] [CrossRef]
- Chang, C.Y.; Ting, H.C.; Liu, C.A.; Su, H.L.; Chiou, T.W.; Lin, S.Z.; Harn, H.J.; Ho, T.J. Induced Pluripotent Stem Cell (iPSC)-Based Neurodegenerative Disease Models for Phenotype Recapitulation and Drug Screening. Molecules 2020, 25, 2000. [Google Scholar] [CrossRef]
- He, Z.; Maynard, A.; Jain, A.; Gerber, T.; Petri, R.; Lin, H.C.; Santel, M.; Ly, K.; Dupre, J.S.; Sidow, L.; et al. Lineage recording in human cerebral organoids. Nat. Methods 2022, 19, 90–99. [Google Scholar] [CrossRef]
- Daoud, A.; Munera, J.O. Insights into Human Development and Disease from Human Pluripotent Stem Cell Derived Intestinal Organoids. Front. Med. 2019, 6, 297. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.; Wang, Y.; Liu, L.P.; Li, Y.M.; Zheng, Y.W. Gene Editing in Pluripotent Stem Cells and Their Derived Organoids. Stem Cells Int. 2021, 2021, 8130828. [Google Scholar] [CrossRef]
- Hendriks, D.; Clevers, H.; Artegiani, B. CRISPR-Cas Tools and Their Application in Genetic Engineering of Human Stem Cells and Organoids. Cell Stem Cell 2020, 27, 705–731. [Google Scholar] [CrossRef] [PubMed]
- De Masi, C.; Spitalieri, P.; Murdocca, M.; Novelli, G.; Sangiuolo, F. Application of CRISPR/Cas9 to human-induced pluripotent stem cells: From gene editing to drug discovery. Hum. Genom. 2020, 14, 25. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ihry, R.J.; Worringer, K.A.; Salick, M.R.; 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. [Google Scholar] [CrossRef] [PubMed]
- Merkle, F.T.; Ghosh, S.; Kamitaki, N.; Mitchell, J.; Avior, Y.; Mello, C.; Kashin, S.; Mekhoubad, S.; Ilic, D.; Charlton, M.; et al. Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations. Nature 2017, 545, 229–233. [Google Scholar] [CrossRef] [Green Version]
- Capecchi, M.R. Altering the genome by homologous recombination. Science 1989, 244, 1288–1292. [Google Scholar] [CrossRef] [Green Version]
- Koller, B.H.; Smithies, O. Inactivating the beta 2-microglobulin locus in mouse embryonic stem cells by homologous recombination. Proc. Natl. Acad. Sci. USA 1989, 86, 8932–8935. [Google Scholar] [CrossRef]
- Rudin, N.; Sugarman, E.; Haber, J.E. Genetic and physical analysis of double-strand break repair and recombination in Saccharomyces cerevisiae. Genetics 1989, 122, 519–534. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Saretzki, G.; Armstrong, L.; Leake, A.; Lako, M.; von Zglinicki, T. Stress defense in murine embryonic stem cells is superior to that of various differentiated murine cells. Stem Cells 2004, 22, 962–971. [Google Scholar] [CrossRef]
- Maynard, S.; Swistowska, A.M.; Lee, J.W.; Liu, Y.; Liu, S.T.; Da Cruz, A.B.; Rao, M.; de Souza-Pinto, N.C.; Zeng, X.; Bohr, V.A. Human embryonic stem cells have enhanced repair of multiple forms of DNA damage. Stem Cells 2008, 26, 2266–2274. [Google Scholar] [CrossRef] [Green Version]
- Fu, X.; Cui, K.; Yi, Q.; Yu, L.; Xu, Y. DNA repair mechanisms in embryonic stem cells. Cell. Mol. Life Sci. 2017, 74, 487–493. [Google Scholar] [CrossRef] [PubMed]
- Tichy, E.D.; Pillai, R.; Deng, L.; Liang, L.; Tischfield, J.; Schwemberger, S.J.; Babcock, G.F.; Stambrook, P.J. Mouse embryonic stem cells, but not somatic cells, predominantly use homologous recombination to repair double-strand DNA breaks. Stem Cells Dev. 2010, 19, 1699–1711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Novo, N.; Romero-Tamayo, S.; Marcuello, C.; Boneta, S.; Blasco-Machin, I.; Velazquez-Campoy, A.; Villanueva, R.; Moreno-Loshuertos, R.; Lostao, A.; Medina, M.; et al. Beyond a platform protein for the degradosome assembly: The Apoptosis-Inducing Factor as an efficient nuclease involved in chromatinolysis. PNAS Nexus 2023, 2, pgac312. [Google Scholar] [CrossRef] [PubMed]
- Artus, C.; Boujrad, H.; Bouharrour, A.; Brunelle, M.N.; Hoos, S.; Yuste, V.J.; Lenormand, P.; Rousselle, J.C.; Namane, A.; England, P.; et al. AIF promotes chromatinolysis and caspase-independent programmed necrosis by interacting with histone H2AX. EMBO J. 2010, 29, 1585–1599. [Google Scholar] [CrossRef] [Green Version]
- Shivram, H.; Cress, B.F.; Knott, G.J.; Doudna, J.A. Controlling and enhancing CRISPR systems. Nat. Chem. Biol. 2021, 17, 10–19. [Google Scholar] [CrossRef] [PubMed]
- Liu, G.; Lin, Q.; Jin, S.; Gao, C. The CRISPR-Cas toolbox and gene editing technologies. Mol. Cell 2022, 82, 333–347. [Google Scholar] [CrossRef]
- Doudna, J.A. The promise and challenge of therapeutic genome editing. Nature 2020, 578, 229–236. [Google Scholar] [CrossRef]
- Wang, J.Y.; Doudna, J.A. CRISPR technology: A decade of genome editing is only the beginning. Science 2023, 379, eadd8643. [Google Scholar] [CrossRef]
- Katti, A.; Diaz, B.J.; Caragine, C.M.; Sanjana, N.E.; Dow, L.E. CRISPR in cancer biology and therapy. Nat. Rev. Cancer 2022, 22, 259–279. [Google Scholar] [CrossRef]
- Durai, S.; Mani, M.; Kandavelou, K.; Wu, J.; Porteus, M.H.; Chandrasegaran, S. Zinc finger nucleases: Custom-designed molecular scissors for genome engineering of plant and mammalian cells. Nucleic Acids Res. 2005, 33, 5978–5990. [Google Scholar] [CrossRef]
- Miller, J.; McLachlan, A.D.; Klug, A. Repetitive zinc-binding domains in the protein transcription factor IIIA from Xenopus oocytes. EMBO J. 1985, 4, 1609–1614. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.G.; Cha, J.; Chandrasegaran, S. Hybrid restriction enzymes: Zinc finger fusions to Fok I cleavage domain. Proc. Natl. Acad. Sci. USA 1996, 93, 1156–1160. [Google Scholar] [CrossRef] [PubMed]
- Paschon, D.E.; Lussier, S.; Wangzor, T.; Xia, D.F.; Li, P.W.; Hinkley, S.J.; Scarlott, N.A.; Lam, S.C.; Waite, A.J.; Truong, L.N.; et al. Diversifying the structure of zinc finger nucleases for high-precision genome editing. Nat. Commun. 2019, 10, 1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lombardo, A.; Genovese, P.; Beausejour, C.M.; Colleoni, S.; Lee, Y.L.; Kim, K.A.; Ando, D.; Urnov, F.D.; Galli, C.; Gregory, P.D.; et al. Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nat. Biotechnol. 2007, 25, 1298–1306. [Google Scholar] [CrossRef]
- Zou, J.; Maeder, M.L.; Mali, P.; Pruett-Miller, S.M.; Thibodeau-Beganny, S.; Chou, B.K.; Chen, G.; Ye, Z.; Park, I.H.; Daley, G.Q.; et al. Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell Stem Cell 2009, 5, 97–110. [Google Scholar] [CrossRef] [Green Version]
- Hockemeyer, D.; Soldner, F.; Beard, C.; Gao, Q.; Mitalipova, M.; DeKelver, R.C.; Katibah, G.E.; Amora, R.; Boydston, E.A.; Zeitler, B.; et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat. Biotechnol. 2009, 27, 851–857. [Google Scholar] [CrossRef] [Green Version]
- Miller, J.C.; Holmes, M.C.; Wang, J.; Guschin, D.Y.; Lee, Y.L.; Rupniewski, I.; Beausejour, C.M.; Waite, A.J.; Wang, N.S.; Kim, K.A.; et al. An improved zinc-finger nuclease architecture for highly specific genome editing. Nat. Biotechnol. 2007, 25, 778–785. [Google Scholar] [CrossRef]
- Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339, 819–823. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Gerstein, M.; Snyder, M. RNA-Seq: A revolutionary tool for transcriptomics. Nat. Rev. Genet. 2009, 10, 57–63. [Google Scholar] [CrossRef]
- Yusa, K.; Rashid, S.T.; Strick-Marchand, H.; Varela, I.; Liu, P.Q.; Paschon, D.E.; Miranda, E.; Ordonez, A.; Hannan, N.R.; Rouhani, F.J.; et al. Targeted gene correction of alpha1-antitrypsin deficiency in induced pluripotent stem cells. Nature 2011, 478, 391–394. [Google Scholar] [CrossRef] [Green Version]
- Pattanayak, V.; Ramirez, C.L.; Joung, J.K.; Liu, D.R. Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat. Methods 2011, 8, 765–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gabriel, R.; Lombardo, A.; Arens, A.; Miller, J.C.; Genovese, P.; Kaeppel, C.; Nowrouzi, A.; Bartholomae, C.C.; Wang, J.; Friedman, G.; et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat. Biotechnol. 2011, 29, 816–823. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.J.; Kim, E.; Kim, J.S. Targeted chromosomal deletions in human cells using zinc finger nucleases. Genome Res. 2010, 20, 81–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.J.; Kweon, J.; Kim, E.; Kim, S.; Kim, J.S. Targeted chromosomal duplications and inversions in the human genome using zinc finger nucleases. Genome Res. 2012, 22, 539–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boch, J.; Bonas, U. Xanthomonas AvrBs3 family-type III effectors: Discovery and function. Annu. Rev. Phytopathol. 2010, 48, 419–436. [Google Scholar] [CrossRef]
- Miller, J.C.; Tan, S.; Qiao, G.; Barlow, K.A.; Wang, J.; Xia, D.F.; Meng, X.; Paschon, D.E.; Leung, E.; Hinkley, S.J.; et al. A TALE nuclease architecture for efficient genome editing. Nat. Biotechnol. 2011, 29, 143–148. [Google Scholar] [CrossRef]
- Boch, J. TALEs of genome targeting. Nat. Biotechnol. 2011, 29, 135–136. [Google Scholar] [CrossRef]
- Qasim, W.; Zhan, H.; Samarasinghe, S.; Adams, S.; Amrolia, P.; Stafford, S.; Butler, K.; Rivat, C.; Wright, G.; Somana, K.; et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci. Transl. Med. 2017, 9, eaaj2013. [Google Scholar] [CrossRef]
- Shahryari, A.; Burtscher, I.; Nazari, Z.; Lickert, H. Engineering Gene Therapy: Advances and Barriers. Adv. Ther.-Ger. 2021, 4, 2100040. [Google Scholar] [CrossRef]
- Yahata, N.; Matsumoto, Y.; Omi, M.; Yamamoto, N.; Hata, R. TALEN-mediated shift of mitochondrial DNA heteroplasmy in MELAS-iPSCs with m.13513G>A mutation. Sci. Rep. 2017, 7, 15557. [Google Scholar] [CrossRef] [Green Version]
- Karakikes, I.; Termglinchan, V.; Cepeda, D.A.; Lee, J.; Diecke, S.; Hendel, A.; Itzhaki, I.; Ameen, M.; Shrestha, R.; Wu, H.; et al. A Comprehensive TALEN-Based Knockout Library for Generating Human-Induced Pluripotent Stem Cell-Based Models for Cardiovascular Diseases. Circ. Res. 2017, 120, 1561–1571. [Google Scholar] [CrossRef] [PubMed]
- Dreyer, A.K.; Hoffmann, D.; Lachmann, N.; Ackermann, M.; Steinemann, D.; Timm, B.; Siler, U.; Reichenbach, J.; Grez, M.; Moritz, T.; et al. TALEN-mediated functional correction of X-linked chronic granulomatous disease in patient-derived induced pluripotent stem cells. Biomaterials 2015, 69, 191–200. [Google Scholar] [CrossRef] [PubMed]
- Ramalingam, S.; Annaluru, N.; Kandavelou, K.; Chandrasegaran, S. TALEN-mediated generation and genetic correction of disease-specific human induced pluripotent stem cells. Curr. Gene Ther. 2014, 14, 461–472. [Google Scholar] [CrossRef]
- Mussolino, C.; Morbitzer, R.; Lutge, 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. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.; Kweon, J.; Kim, J.S. TALENs and ZFNs are associated with different mutation signatures. Nat. Methods 2013, 10, 185. [Google Scholar] [CrossRef]
- Kim, Y.; Kweon, J.; Kim, A.; Chon, J.K.; Yoo, J.Y.; Kim, H.J.; Kim, S.; Lee, C.; Jeong, E.; Chung, E.; et al. A library of TAL effector nucleases spanning the human genome. Nat. Biotechnol. 2013, 31, 251–258. [Google Scholar] [CrossRef]
- Veres, A.; Gosis, B.S.; Ding, Q.; Collins, R.; Ragavendran, A.; Brand, H.; Erdin, S.; Cowan, C.A.; Talkowski, M.E.; 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. [Google Scholar] [CrossRef] [Green Version]
- Smith, C.; Gore, A.; Yan, W.; Abalde-Atristain, L.; Li, Z.; He, C.; Wang, Y.; Brodsky, R.A.; Zhang, K.; Cheng, L.; et al. Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. Cell Stem Cell 2014, 15, 12–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barrangou, R.; Fremaux, C.; Deveau, H.; Richards, M.; Boyaval, P.; Moineau, S.; Romero, D.A.; Horvath, P. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315, 1709–1712. [Google Scholar] [CrossRef]
- Bolotin, A.; Quinquis, B.; Sorokin, A.; Ehrlich, S.D. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology 2005, 151, 2551–2561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef] [PubMed]
- Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; DiCarlo, J.E.; Norville, J.E.; Church, G.M. RNA-guided human genome engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakamura, M.; Gao, Y.; Dominguez, A.A.; Qi, L.S. CRISPR technologies for precise epigenome editing. Nat. Cell. Biol. 2021, 23, 11–22. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; La Russa, M.; Qi, L.S. CRISPR/Cas9 in Genome Editing and Beyond. Annu. Rev. Biochem. 2016, 85, 227–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yano, N.; Fedulov, A.V. Targeted DNA Demethylation: Vectors, Effectors and Perspectives. Biomedicines 2023, 11, 1334. [Google Scholar] [CrossRef]
- Brouns, S.J.; Jore, M.M.; Lundgren, M.; Westra, E.R.; Slijkhuis, R.J.; Snijders, A.P.; Dickman, M.J.; Makarova, K.S.; Koonin, E.V.; van der Oost, J. Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 2008, 321, 960–964. [Google Scholar] [CrossRef] [Green Version]
- Marraffini, L.A.; Sontheimer, E.J. CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA. Science 2008, 322, 1843–1845. [Google Scholar] [CrossRef] [Green Version]
- Zetsche, B.; Gootenberg, J.S.; Abudayyeh, O.O.; Slaymaker, I.M.; Makarova, K.S.; Essletzbichler, P.; Volz, S.E.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.T.; Kellner, M.J.; Regev, A.; et al. RNA targeting with CRISPR-Cas13. Nature 2017, 550, 280–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pickar-Oliver, A.; Gersbach, C.A. The next generation of CRISPR-Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 2019, 20, 490–507. [Google Scholar] [CrossRef]
- Shalem, O.; Sanjana, N.E.; Zhang, F. High-throughput functional genomics using CRISPR-Cas9. Nat. Rev. Genet. 2015, 16, 299–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Michels, B.E.; Mosa, M.H.; Streibl, B.I.; Zhan, T.; Menche, C.; Abou-El-Ardat, K.; Darvishi, T.; Czlonka, E.; Wagner, S.; Winter, J.; et al. Pooled In Vitro and In Vivo CRISPR-Cas9 Screening Identifies Tumor Suppressors in Human Colon Organoids. Cell Stem Cell 2020, 26, 782–792.e7. [Google Scholar] [CrossRef] [PubMed]
- Briganti, F.; Sun, H.; Wei, W.; Wu, J.; Zhu, C.; Liss, M.; Karakikes, I.; Rego, S.; Cipriano, A.; Snyder, M.; et al. iPSC Modeling of RBM20-Deficient DCM Identifies Upregulation of RBM20 as a Therapeutic Strategy. Cell Rep. 2020, 32, 108117. [Google Scholar] [CrossRef]
- Jeong, J.; Jager, A.; Domizi, P.; Pavel-Dinu, M.; Gojenola, L.; Iwasaki, M.; Wei, M.C.; Pan, F.; Zehnder, J.L.; Porteus, M.H.; et al. High-efficiency CRISPR induction of t(9;11) chromosomal translocations and acute leukemias in human blood stem cells. Blood Adv. 2019, 3, 2825–2835. [Google Scholar] [CrossRef] [Green Version]
- Maxwell, K.G.; Augsornworawat, P.; Velazco-Cruz, L.; Kim, M.H.; Asada, R.; Hogrebe, N.J.; Morikawa, S.; Urano, F.; Millman, J.R. Gene-edited human stem cell-derived beta cells from a patient with monogenic diabetes reverse preexisting diabetes in mice. Sci. Transl. Med. 2020, 12, eaax9106. [Google Scholar] [CrossRef]
- Takasato, M.; Er, P.X.; Chiu, H.S.; Little, M.H. Generation of kidney organoids from human pluripotent stem cells. Nat. Protoc. 2016, 11, 1681–1692. [Google Scholar] [CrossRef] [Green Version]
- Miller, A.J.; Dye, B.R.; Ferrer-Torres, D.; Hill, D.R.; Overeem, A.W.; Shea, L.D.; Spence, J.R. Generation of lung organoids from human pluripotent stem cells in vitro. Nat. Protoc. 2019, 14, 518–540. [Google Scholar] [CrossRef]
- Drakhlis, L.; Devadas, S.B.; Zweigerdt, R. Generation of heart-forming organoids from human pluripotent stem cells. Nat. Protoc. 2021, 16, 5652–5672. [Google Scholar] [CrossRef] [PubMed]
- Ouchi, R.; Togo, S.; Kimura, M.; Shinozawa, T.; Koido, M.; Koike, H.; Thompson, W.; Karns, R.A.; Mayhew, C.N.; McGrath, P.S.; et al. Modeling Steatohepatitis in Humans with Pluripotent Stem Cell-Derived Organoids. Cell Metab. 2019, 30, 374–384.e6. [Google Scholar] [CrossRef]
- Xiang, Y.; Tanaka, Y.; Patterson, B.; Kang, Y.J.; Govindaiah, G.; Roselaar, N.; Cakir, B.; Kim, K.Y.; Lombroso, A.P.; Hwang, S.M.; et al. Fusion of Regionally Specified hPSC-Derived Organoids Models Human Brain Development and Interneuron Migration. Cell Stem Cell 2017, 21, 383–398.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wimmer, R.A.; Leopoldi, A.; Aichinger, M.; Kerjaschki, D.; Penninger, J.M. Generation of blood vessel organoids from human pluripotent stem cells. Nat. Protoc. 2019, 14, 3082–3100. [Google Scholar] [CrossRef] [PubMed]
- Freedman, B.S.; Brooks, C.R.; Lam, A.Q.; Fu, H.; Morizane, R.; Agrawal, V.; Saad, A.F.; Li, M.K.; Hughes, M.R.; Werff, R.V.; et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat. Commun. 2015, 6, 8715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bian, S.; Repic, M.; Guo, Z.; Kavirayani, A.; Burkard, T.; Bagley, J.A.; Krauditsch, C.; Knoblich, J.A. Genetically engineered cerebral organoids model brain tumor formation. Nat. Methods 2018, 15, 631–639. [Google Scholar] [CrossRef] [PubMed]
- VanderWall, K.B.; Huang, K.C.; Pan, Y.; Lavekar, S.S.; Fligor, C.M.; Allsop, A.R.; Lentsch, K.A.; Dang, P.; Zhang, C.; Tseng, H.C.; et al. Retinal Ganglion Cells with a Glaucoma OPTN(E50K) Mutation Exhibit Neurodegenerative Phenotypes when Derived from Three-Dimensional Retinal Organoids. Stem Cell Rep. 2020, 15, 52–66. [Google Scholar] [CrossRef]
- Koehler, K.R.; Nie, J.; Longworth-Mills, E.; Liu, X.P.; Lee, J.; Holt, J.R.; Hashino, E. Generation of inner ear organoids containing functional hair cells from human pluripotent stem cells. Nat. Biotechnol. 2017, 35, 583–589. [Google Scholar] [CrossRef] [Green Version]
- Zhu, H.; Blum, R.H.; Bernareggi, D.; Ask, E.H.; Wu, Z.; Hoel, H.J.; Meng, Z.; Wu, C.; Guan, K.L.; Malmberg, K.J.; et al. Metabolic Reprograming via Deletion of CISH in Human iPSC-Derived NK Cells Promotes In Vivo Persistence and Enhances Anti-tumor Activity. Cell Stem Cell 2020, 27, 224–237.e6. [Google Scholar] [CrossRef]
- Woan, K.V.; Kim, H.; Bjordahl, R.; Davis, Z.B.; Gaidarova, S.; Goulding, J.; Hancock, B.; Mahmood, S.; Abujarour, R.; Wang, H.; et al. Harnessing features of adaptive NK cells to generate iPSC-derived NK cells for enhanced immunotherapy. Cell Stem Cell 2021, 28, 2062–2075.e5. [Google Scholar] [CrossRef]
- Koo, T.; Lee, J.; Kim, J.S. Measuring and Reducing Off-Target Activities of Programmable Nucleases Including CRISPR-Cas9. Mol. Cells 2015, 38, 475–481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cui, Z.; Liu, H.; Zhang, H.; Huang, Z.; Tian, R.; Li, L.; Fan, W.; Chen, Y.; Chen, L.; Zhang, S.; et al. The comparison of ZFNs, TALENs, and SpCas9 by GUIDE-seq in HPV-targeted gene therapy. Mol. Ther. Nucleic Acids 2021, 26, 1466–1478. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.; Bae, S.; Park, J.; Kim, E.; Kim, S.; Yu, H.R.; Hwang, J.; Kim, J.I.; Kim, J.S. Digenome-seq: Genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat. Methods 2015, 12, 237–243. [Google Scholar] [CrossRef]
- Tsai, S.Q.; Nguyen, N.T.; Malagon-Lopez, J.; Topkar, V.V.; Aryee, M.J.; Joung, J.K. CIRCLE-seq: A highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat. Methods 2017, 14, 607–614. [Google Scholar] [CrossRef] [Green Version]
- Tsai, S.Q.; Zheng, Z.; Nguyen, N.T.; Liebers, M.; Topkar, V.V.; Thapar, V.; Wyvekens, N.; Khayter, C.; Iafrate, A.J.; Le, L.P.; et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 2015, 33, 187–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ran, F.A.; Hsu, P.D.; Lin, C.Y.; Gootenberg, J.S.; Konermann, S.; Trevino, A.E.; Scott, D.A.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.S.; Dagdas, Y.S.; Kleinstiver, B.P.; Welch, M.M.; Sousa, A.A.; Harrington, L.B.; Sternberg, S.H.; Joung, J.K.; Yildiz, A.; Doudna, J.A. Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature 2017, 550, 407–410. [Google Scholar] [CrossRef] [Green Version]
- Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.; Joung, J.K. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.K.; Jeong, E.; Lee, J.; Jung, M.; Shin, E.; Kim, Y.H.; Lee, K.; Jung, I.; Kim, D.; Kim, S.; et al. Directed evolution of CRISPR-Cas9 to increase its specificity. Nat. Commun. 2018, 9, 3048. [Google Scholar] [CrossRef] [Green Version]
- Fu, Y.; Sander, J.D.; Reyon, D.; Cascio, V.M.; Joung, J.K. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat. Biotechnol. 2014, 32, 279–284. [Google Scholar] [CrossRef] [Green Version]
- Kocak, D.D.; Josephs, E.A.; Bhandarkar, V.; Adkar, S.S.; Kwon, J.B.; Gersbach, C.A. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nat. Biotechnol. 2019, 37, 657–666. [Google Scholar] [CrossRef] [PubMed]
- Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature 2017, 551, 464–471. [Google Scholar] [CrossRef] [Green Version]
- Kurt, I.C.; Zhou, R.; Iyer, S.; Garcia, S.P.; Miller, B.R.; Langner, L.M.; Grunewald, J.; Joung, J.K. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat. Biotechnol. 2021, 39, 41–46. [Google Scholar] [CrossRef]
- Zhao, D.; Li, J.; Li, S.; Xin, X.; Hu, M.; Price, M.A.; Rosser, S.J.; Bi, C.; Zhang, X. Glycosylase base editors enable C-to-A and C-to-G base changes. Nat. Biotechnol. 2021, 39, 35–40. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Hong, M.; Luan, C.; Gao, H.; Ru, G.; Guo, X.; Zhang, D.; Zhang, S.; Li, C.; Wu, J.; et al. Adenine transversion editors enable precise, efficient A*T-to-C*G base editing in mammalian cells and embryos. Nat. Biotechnol. 2023, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Tong, H.; Wang, X.; Liu, Y.; Liu, N.; Li, Y.; Luo, J.; Ma, Q.; Wu, D.; Li, J.; Xu, C.; et al. Programmable A-to-Y base editing by fusing an adenine base editor with an N-methylpurine DNA glycosylase. Nat. Biotechnol. 2023, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Grunewald, J.; Zhou, R.; Lareau, C.A.; Garcia, S.P.; Iyer, S.; Miller, B.R.; Langner, L.M.; Hsu, J.Y.; Aryee, M.J.; Joung, J.K. A dual-deaminase CRISPR base editor enables concurrent adenine and cytosine editing. Nat. Biotechnol. 2020, 38, 861–864. [Google Scholar] [CrossRef]
- Sakata, R.C.; Ishiguro, S.; Mori, H.; Tanaka, M.; Tatsuno, K.; Ueda, H.; Yamamoto, S.; Seki, M.; Masuyama, N.; Nishida, K.; et al. Base editors for simultaneous introduction of C-to-T and A-to-G mutations. Nat. Biotechnol. 2020, 38, 865–869. [Google Scholar] [CrossRef]
- Xie, J.; Huang, X.; Wang, X.; Gou, S.; Liang, Y.; Chen, F.; Li, N.; Ouyang, Z.; Zhang, Q.; Ge, W.; et al. ACBE, a new base editor for simultaneous C-to-T and A-to-G substitutions in mammalian systems. BMC Biol. 2020, 18, 131. [Google Scholar] [CrossRef]
- Zhang, X.; Zhu, B.; Chen, L.; Xie, L.; Yu, W.; Wang, Y.; Li, L.; Yin, S.; Yang, L.; Hu, H.; et al. Dual base editor catalyzes both cytosine and adenine base conversions in human cells. Nat. Biotechnol. 2020, 38, 856–860. [Google Scholar] [CrossRef]
- Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nishida, K.; Arazoe, T.; Yachie, N.; Banno, S.; Kakimoto, M.; Tabata, M.; Mochizuki, M.; Miyabe, A.; Araki, M.; Hara, K.Y.; et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 2016, 353, aaf8729. [Google Scholar] [CrossRef]
- Komor, A.C.; Zhao, K.T.; Packer, M.S.; Gaudelli, N.M.; Waterbury, A.L.; Koblan, L.W.; Kim, Y.B.; Badran, A.H.; Liu, D.R. 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. [Google Scholar] [CrossRef] [Green Version]
- Anzalone, A.V.; Koblan, L.W.; Liu, D.R. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 2020, 38, 824–844. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Park, J.E.; Paa, P.; Rajakumar, P.D.; Prekop, H.T.; Chew, Y.T.; Manivannan, S.N.; Chew, W.L. Programmable C:G to G:C genome editing with CRISPR-Cas9-directed base excision repair proteins. Nat. Commun. 2021, 12, 1384. [Google Scholar] [CrossRef]
- Zafra, M.P.; Schatoff, E.M.; Katti, A.; Foronda, M.; Breinig, M.; Schweitzer, A.Y.; Simon, A.; Han, T.; Goswami, S.; Montgomery, E.; et al. Optimized base editors enable efficient editing in cells, organoids and mice. Nat. Biotechnol. 2018, 36, 888–893. [Google Scholar] [CrossRef]
- Koblan, L.W.; Doman, J.L.; Wilson, C.; Levy, J.M.; Tay, T.; Newby, G.A.; Maianti, J.P.; Raguram, A.; Liu, D.R. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 2018, 36, 843–846. [Google Scholar] [CrossRef] [PubMed]
- Gaudelli, N.M.; Lam, D.K.; Rees, H.A.; Sola-Esteves, N.M.; Barrera, L.A.; Born, D.A.; Edwards, A.; Gehrke, J.M.; Lee, S.J.; Liquori, A.J.; et al. Directed evolution of adenine base editors with increased activity and therapeutic application. Nat. Biotechnol. 2020, 38, 892–900. [Google Scholar] [CrossRef]
- Liu, Y.; Zhou, C.; Huang, S.; Dang, L.; Wei, Y.; He, J.; Zhou, Y.; Mao, S.; Tao, W.; Zhang, Y.; et al. A Cas-embedding strategy for minimizing off-target effects of DNA base editors. Nat. Commun. 2020, 11, 6073. [Google Scholar] [CrossRef]
- Rees, H.A.; Wilson, C.; Doman, J.L.; Liu, D.R. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci. Adv. 2019, 5, eaax5717. [Google Scholar] [CrossRef] [Green Version]
- Grunewald, J.; Zhou, R.; Iyer, S.; Lareau, C.A.; Garcia, S.P.; Aryee, M.J.; Joung, J.K. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat. Biotechnol. 2019, 37, 1041–1048. [Google Scholar] [CrossRef] [PubMed]
- Jeong, Y.K.; Yu, J.; Bae, S. Construction of non-canonical PAM-targeting adenosine base editors by restriction enzyme-free DNA cloning using CRISPR-Cas9. Sci. Rep. 2019, 9, 4939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Z.; Tao, W.; Huang, S.; Sun, W.; Wang, Y.; Jiang, W.; Huang, X.; Lin, C.P. Engineering an adenine base editor in human embryonic stem cells with minimal DNA and RNA off-target activities. Mol. Ther. Nucleic Acids 2022, 29, 502–510. [Google Scholar] [CrossRef] [PubMed]
- Koblan, L.W.; Arbab, M.; Shen, M.W.; Hussmann, J.A.; Anzalone, A.V.; Doman, J.L.; Newby, G.A.; Yang, D.; Mok, B.; Replogle, J.M.; et al. Efficient C*G-to-G*C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nat. Biotechnol. 2021, 39, 1414–1425. [Google Scholar] [CrossRef]
- Chen, S.; Liu, Z.; Lai, L.; Li, Z. Efficient C-to-G Base Editing with Improved Target Compatibility Using Engineered Deaminase-nCas9 Fusions. CRISPR J. 2022, 5, 389–396. [Google Scholar] [CrossRef]
- Chen, L.; Zhu, B.; Ru, G.; Meng, H.; Yan, Y.; Hong, M.; Zhang, D.; Luan, C.; Zhang, S.; Wu, H.; et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. Nat. Biotechnol. 2023, 41, 663–672. [Google Scholar] [CrossRef]
- Zuo, E.; Sun, Y.; Wei, W.; Yuan, T.; Ying, W.; Sun, H.; Yuan, L.; Steinmetz, L.M.; Li, Y.; Yang, H. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 2019, 364, 289–292. [Google Scholar] [CrossRef]
- Yan, N.; Feng, H.; Sun, Y.; Xin, Y.; Zhang, H.; Lu, H.; Zheng, J.; He, C.; Zuo, Z.; Yuan, T.; et al. Cytosine base editors induce off-target mutations and adverse phenotypic effects in transgenic mice. Nat. Commun. 2023, 14, 1784. [Google Scholar] [CrossRef]
- Jin, S.; Zong, Y.; Gao, Q.; Zhu, Z.; Wang, Y.; Qin, P.; Liang, C.; Wang, D.; Qiu, J.L.; Zhang, F.; et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 2019, 364, 292–295. [Google Scholar] [CrossRef]
- Li, A.; Mitsunobu, H.; Yoshioka, S.; Suzuki, T.; Kondo, A.; Nishida, K. Cytosine base editing systems with minimized off-target effect and molecular size. Nat. Commun. 2022, 13, 4531. [Google Scholar] [CrossRef]
- McGrath, E.; Shin, H.; Zhang, L.; Phue, J.N.; Wu, W.W.; Shen, R.F.; Jang, Y.Y.; Revollo, J.; Ye, Z. Targeting specificity of APOBEC-based cytosine base editor in human iPSCs determined by whole genome sequencing. Nat. Commun. 2019, 10, 5353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grunewald, J.; Zhou, R.; Garcia, S.P.; Iyer, S.; Lareau, C.A.; Aryee, M.J.; Joung, J.K. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 2019, 569, 433–437. [Google Scholar] [CrossRef] [PubMed]
- Kumar, R.; DiMenna, L.J.; Chaudhuri, J.; Evans, T. Biological function of activation-induced cytidine deaminase (AID). Biomed. J. 2014, 37, 269–283. [Google Scholar] [CrossRef]
- Gehrke, J.M.; Cervantes, O.; Clement, M.K.; Wu, Y.; Zeng, J.; Bauer, D.E.; Pinello, L.; Joung, J.K. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat. Biotechnol. 2018, 36, 977–982. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Doman, J.L.; Raguram, A.; Newby, G.A.; Liu, D.R. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat. Biotechnol. 2020, 38, 620–628. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.B.; Komor, A.C.; Levy, J.M.; Packer, M.S.; Zhao, K.T.; 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Neugebauer, M.E.; Hsu, A.; Arbab, M.; Krasnow, N.A.; McElroy, A.N.; Pandey, S.; Doman, J.L.; Huang, T.P.; Raguram, A.; Banskota, S.; et al. Evolution of an adenine base editor into a small, efficient cytosine base editor with low off-target activity. Nat. Biotechnol. 2023, 41, 673–685. [Google Scholar] [CrossRef] [PubMed]
- Lam, D.K.; Feliciano, P.R.; Arif, A.; Bohnuud, T.; Fernandez, T.P.; Gehrke, J.M.; Grayson, P.; Lee, K.D.; Ortega, M.A.; Sawyer, C.; et al. Improved cytosine base editors generated from TadA variants. Nat. Biotechnol. 2023, 41, 686–697. [Google Scholar] [CrossRef]
- Richter, M.F.; Zhao, K.T.; Eton, E.; Lapinaite, A.; Newby, G.A.; Thuronyi, B.W.; Wilson, C.; Koblan, L.W.; Zeng, J.; Bauer, D.E.; et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat. Biotechnol. 2020, 38, 901. [Google Scholar] [CrossRef] [PubMed]
- Li, J.A.; Yu, W.X.; Huang, S.S.; Wu, S.S.; Li, L.P.; Zhou, J.K.; Cao, Y.; Huang, X.X.; Qiao, Y.B. Structure-guided engineering of adenine base editor with minimized RNA off-targeting activity. Nat. Commun. 2021, 12, 2287. [Google Scholar] [CrossRef]
- Chen, L.; Zhang, S.; Xue, N.; Hong, M.; Zhang, X.; Zhang, D.; Yang, J.; Bai, S.; Huang, Y.; Meng, H.; et al. Engineering a precise adenine base editor with minimal bystander editing. Nat. Chem. Biol. 2023, 19, 101–110. [Google Scholar] [CrossRef]
- Tu, T.X.; Song, Z.M.; Liu, X.Y.; Wang, S.X.; He, X.X.; Xi, H.T.; Wang, J.H.; Yan, T.; Chen, H.R.; Zhang, Z.W.; et al. A precise and efficient adenine base editor. Mol. Ther. 2022, 30, 2933–2941. [Google Scholar] [CrossRef] [PubMed]
- Park, J.C.; Jang, H.K.; Kim, J.; Han, J.H.; Jung, Y.; Kim, K.; Bae, S.; Cha, H.J. High expression of uracil DNA glycosylase determines C to T substitution in human pluripotent stem cells. Mol. Ther. Nucleic Acids 2022, 27, 175–183. [Google Scholar] [CrossRef]
- Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157. [Google Scholar] [CrossRef]
- Chen, P.J.; Liu, D.R. Prime editing for precise and highly versatile genome manipulation. Nat. Rev. Genet. 2023, 24, 161–177. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.J.; Hussmann, J.A.; Yan, J.; Knipping, F.; Ravisankar, P.; Chen, P.F.; Chen, C.; Nelson, J.W.; Newby, G.A.; Sahin, M.; et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 2021, 184, 5635–5652.e29. [Google Scholar] [CrossRef] [PubMed]
- Adikusuma, F.; Lushington, C.; Arudkumar, J.; Godahewa, G.I.; Chey, Y.C.J.; Gierus, L.; Piltz, S.; Geiger, A.; Jain, Y.; Reti, D.; et al. Optimized nickase- and nuclease-based prime editing in human and mouse cells. Nucleic Acids Res. 2021, 49, 10785–10795. [Google Scholar] [CrossRef] [PubMed]
- Jiang, T.T.; Zhang, X.O.; Weng, Z.P.; Xue, W. Deletion and replacement of long genomic sequences using prime editing. Nat. Biotechnol. 2022, 40, 227–234. [Google Scholar] [CrossRef]
- Tao, R.; Wang, Y.H.; Hu, Y.; Jiao, Y.G.; Zhou, L.F.; Jiang, L.R.; Li, L.; He, X.Y.; Li, M.; Yu, Y.M.; et al. WT-PE: Prime editing with nuclease wild-type Cas9 enables versatile large-scale genome editing. Signal Transduct. Tar. 2022, 7, 108. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Zhang, G.; Huang, S.; Liu, Y.; Tang, J.; Zhong, M.; Wang, X.; Sun, W.; Yao, Y.; Ji, Q.; et al. Development of a versatile nuclease prime editor with upgraded precision. Nat. Commun. 2023, 14, 305. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Lim, J.M.; Min, S.; Oh, J.S.; Kim, D.Y.; Woo, J.S.; Nishimasu, H.; Cho, S.R.; Yoon, S.; Kim, H.H. Generation of a more efficient prime editor 2 by addition of the Rad51 DNA-binding domain. Nat. Commun. 2021, 12, 5617. [Google Scholar] [CrossRef] [PubMed]
- Park, S.J.; Jeong, T.Y.; Shin, S.K.; Yoon, D.E.; Lim, S.Y.; Kim, S.P.; Choi, J.; Lee, H.; Hong, J.I.; Ahn, J.; et al. Targeted mutagenesis in mouse cells and embryos using an enhanced prime editor. Genome Biol. 2021, 22, 170. [Google Scholar] [CrossRef] [PubMed]
- Velimirovic, M.; Zanetti, L.C.; Shen, M.W.; Fife, J.D.; Lin, L.; Cha, M.; Akinci, E.; Barnum, D.; Yu, T.; Sherwood, R.I. Peptide fusion improves prime editing efficiency. Nat. Commun. 2022, 13, 3512. [Google Scholar] [CrossRef]
- Nelson, J.W.; Randolph, P.B.; Shen, S.P.; Everette, K.A.; Chen, P.J.; Anzalone, A.V.; An, M.; Newby, G.A.; Chen, J.C.; Hsu, A.; et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 2022, 40, 402–410. [Google Scholar] [CrossRef]
- Chow, R.D.; Chen, J.S.; Shen, J.; Chen, S.D. A web tool for the design of prime-editing guide RNAs. Nat. Biomed. Eng. 2021, 5, 190–194. [Google Scholar] [CrossRef]
- Hsu, J.Y.; Grunewald, J.; Szalay, R.; Shih, J.; Anzalone, A.V.; Lam, K.C.; Shen, M.W.; Petri, K.; Liu, D.R.; Joung, J.K.; et al. PrimeDesign software for rapid and simplified design of prime editing guide RNAs. Nat. Commun. 2021, 12, 1034. [Google Scholar] [CrossRef]
- Hwang, G.H.; Jeong, Y.K.; Habib, O.; Hong, S.A.; Lim, K.; Kim, J.S.; Bae, S. PE-Designer and PE-Analyzer: Web-based design and analysis tools for CRISPR prime editing. Nucleic Acids Res. 2021, 49, W499–W504. [Google Scholar] [CrossRef]
- Siegner, S.M.; Karasu, M.E.; Schroder, M.S.; Kontarakis, Z.; Corn, J.E. PnB Designer: A web application to design prime and base editor guide RNAs for animals and plants. BMC Bioinform. 2021, 22, 101. [Google Scholar] [CrossRef]
- Anderson, M.V.; Haldrup, J.; Thomsen, E.A.; Wolff, J.H.; Mikkelsen, J.G. pegIT-a web-based design tool for prime editing. Nucleic Acids Res. 2021, 49, W505–W509. [Google Scholar] [CrossRef]
- Yu, G.; Kim, H.K.; Park, J.; Kwak, H.; Cheong, Y.; Kim, D.; Kim, J.; Kim, J.; Kim, H.H. Prediction of efficiencies for diverse prime editing systems in multiple cell types. Cell 2023, 186, 2256–2272.e23. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Zhou, L.; Gao, B.Q.; Li, G.; Wang, X.; Wang, Y.; Wei, J.; Han, W.; Wang, Z.; Li, J.; et al. Highly efficient prime editing by introducing same-sense mutations in pegRNA or stabilizing its structure. Nat. Commun. 2022, 13, 1669. [Google Scholar] [CrossRef] [PubMed]
- Feng, Y.; Liu, S.; Mo, Q.; Liu, P.; Xiao, X.; Ma, H. Enhancing prime editing efficiency and flexibility with tethered and split pegRNAs. Protein Cell 2023, 14, 304–308. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Zhang, G.; Liu, Y.; Huang, S.; Qu, S.; Cheng, D.; Yao, Y.; Ji, Q.; Wang, X.; Huang, X.; Liu, J. Enhancement of prime editing via xrRNA motif-joined pegRNA. Nat. Commun. 2022, 13, 1856. [Google Scholar] [CrossRef]
- Huang, S.; Zhang, Z.; Tao, W.; Liu, Y.; Li, X.; Wang, X.; Harati, J.; Wang, P.Y.; Huang, X.; Lin, C.P. Broadening prime editing toolkits using RNA-Pol-II-driven engineered pegRNA. Mol. Ther. 2022, 30, 2923–2932. [Google Scholar] [CrossRef] [PubMed]
- Habib, O.; Habib, G.; Hwang, G.H.; Bae, S. Comprehensive analysis of prime editing outcomes in human embryonic stem cells. Nucleic Acids Res. 2022, 50, 1187–1197. [Google Scholar] [CrossRef]
- Zhou, M.; Tang, S.; Duan, N.; Xie, M.; Li, Z.; Feng, M.; Wu, L.; Hu, Z.; Liang, D. Targeted-Deletion of a Tiny Sequence via Prime Editing to Restore SMN Expression. Int. J. Mol. Sci. 2022, 23, 7941. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Busquets, O.; Verma, Y.; Syed, K.M.; Kutnowski, N.; Pangilinan, G.R.; Gilbert, L.A.; Bateup, H.S.; Rio, D.C.; Hockemeyer, D.; et al. Highly efficient generation of isogenic pluripotent stem cell models using prime editing. Elife 2022, 11, e79208. [Google Scholar] [CrossRef] [PubMed]
- Peters, J.E.; Makarova, K.S.; Shmakov, S.; Koonin, E.V. Recruitment of CRISPR-Cas systems by Tn7-like transposons. Proc. Natl. Acad. Sci. USA 2017, 114, E7358–E7366. [Google Scholar] [CrossRef] [PubMed]
- Chavez, M.; Qi, L.S. Site-Programmable Transposition: Shifting the Paradigm for CRISPR-Cas Systems. Mol. Cell 2019, 75, 206–208. [Google Scholar] [CrossRef]
- Strecker, J.; Ladha, A.; Gardner, Z.; Schmid-Burgk, J.L.; Makarova, K.S.; Koonin, E.V.; Zhang, F. RNA-guided DNA insertion with CRISPR-associated transposases. Science 2019, 365, 48–53. [Google Scholar] [CrossRef]
- Tou, C.J.; Orr, B.; Kleinstiver, B.P. Precise cut-and-paste DNA insertion using engineered type V-K CRISPR-associated transposases. Nat. Biotechnol. 2023, 41, 968–979. [Google Scholar] [CrossRef] [PubMed]
- Lampe, G.D.; King, R.T.; Halpin-Healy, T.S.; Klompe, S.E.; Hogan, M.I.; Vo, P.L.H.; Tang, S.; Chavez, A.; Sternberg, S.H. Targeted DNA integration in human cells without double-strand breaks using CRISPR-associated transposases. Nat. Biotechnol. 2023, 1–12. [Google Scholar] [CrossRef]
- Durrant, M.G.; Fanton, A.; Tycko, J.; Hinks, M.; Chandrasekaran, S.S.; Perry, N.T.; Schaepe, J.; Du, P.P.; Lotfy, P.; Bassik, M.C.; et al. Systematic discovery of recombinases for efficient integration of large DNA sequences into the human genome. Nat. Biotechnol. 2023, 41, 488–499. [Google Scholar] [CrossRef] [PubMed]
- Anzalone, A.V.; Gao, X.D.; Podracky, C.J.; Nelson, A.T.; Koblan, L.W.; Raguram, A.; Levy, J.M.; Mercer, J.A.M.; Liu, D.R. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat. Biotechnol. 2022, 40, 731–740. [Google Scholar] [CrossRef] [PubMed]
- Yarnall, M.T.N.; Ioannidi, E.I.; Schmitt-Ulms, C.; Krajeski, R.N.; Lim, J.; Villiger, L.; Zhou, W.; Jiang, K.; Garushyants, S.K.; Roberts, N.; et al. Drag-and-drop genome insertion of large sequences without double-strand DNA cleavage using CRISPR-directed integrases. Nat. Biotechnol. 2023, 41, 500–512. [Google Scholar] [CrossRef] [PubMed]
- Lampson, B.C.; Sun, J.; Hsu, M.Y.; Vallejo-Ramirez, J.; Inouye, S.; Inouye, M. Reverse transcriptase in a clinical strain of Escherichia coli: Production of branched RNA-linked msDNA. Science 1989, 243, 1033–1038. [Google Scholar] [CrossRef] [Green Version]
- Hsu, M.Y.; Inouye, M.; Inouye, S. Retron for the 67-base multicopy single-stranded DNA from Escherichia coli: A potential transposable element encoding both reverse transcriptase and Dam methylase functions. Proc. Natl. Acad. Sci. USA 1990, 87, 9454–9458. [Google Scholar] [CrossRef]
- Hsu, M.Y.; Eagle, S.G.; Inouye, M.; Inouye, S. Cell-free synthesis of the branched RNA-linked msDNA from retron-Ec67 of Escherichia coli. J. Biol. Chem. 1992, 267, 13823–13829. [Google Scholar] [CrossRef] [PubMed]
- Linquist, S.; Cottenie, K.; Elliott, T.A.; Saylor, B.; Kremer, S.C.; Gregory, T.R. Applying ecological models to communities of genetic elements: The case of neutral theory. Mol. Ecol. 2015, 24, 3232–3242. [Google Scholar] [CrossRef] [Green Version]
- Sharon, E.; Chen, S.A.; Khosla, N.M.; Smith, J.D.; Pritchard, J.K.; Fraser, H.B. Functional Genetic Variants Revealed by Massively Parallel Precise Genome Editing. Cell 2018, 175, 544–557.e16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lopez, S.C.; Crawford, K.D.; Lear, S.K.; Bhattarai-Kline, S.; Shipman, S.L. Precise genome editing across kingdoms of life using retron-derived DNA. Nat. Chem. Biol. 2022, 18, 199–206. [Google Scholar] [CrossRef] [PubMed]
- Kong, X.; Wang, Z.; Zhang, R.; Wang, X.; Zhou, Y.; Shi, L.; Yang, H. Precise genome editing without exogenous donor DNA via retron editing system in human cells. Protein Cell 2021, 12, 899–902. [Google Scholar] [CrossRef] [PubMed]
- Allen, A.G.; Khan, S.Q.; Margulies, C.M.; Viswanathan, R.; Lele, S.; Blaha, L.; Scott, S.N.; Izzo, K.M.; Gerew, A.; Pattali, R.; et al. A highly efficient transgene knock-in technology in clinically relevant cell types. Nat. Biotechnol. 2023, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.; Staahl, B.T.; Alla, R.K.; Doudna, J.A. Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife 2014, 3, e04766. [Google Scholar] [CrossRef]
- Li, M.; Zhong, A.; Wu, Y.; Sidharta, M.; Beaury, M.; Zhao, X.; Studer, L.; Zhou, T. Transient inhibition of p53 enhances prime editing and cytosine base-editing efficiencies in human pluripotent stem cells. Nat. Commun. 2022, 13, 6354. [Google Scholar] [CrossRef]
- Zhang, Z.; Baxter, A.E.; Ren, D.; Qin, K.; Chen, Z.; Collins, S.M.; Huang, H.; Komar, C.A.; Bailer, P.F.; Parker, J.B.; et al. Efficient engineering of human and mouse primary cells using peptide-assisted genome editing. Nat. Biotechnol. 2023, 1–11. [Google Scholar] [CrossRef]
- Foss, D.V.; Muldoon, J.J.; Nguyen, D.N.; Carr, D.; Sahu, S.U.; Hunsinger, J.M.; Wyman, S.K.; Krishnappa, N.; Mendonsa, R.; Schanzer, E.V.; et al. Peptide-mediated delivery of CRISPR enzymes for the efficient editing of primary human lymphocytes. Nat. Biomed. Eng. 2023, 7, 647–660. [Google Scholar] [CrossRef]
- Yu, C.; Liu, Y.; Ma, T.; Liu, K.; Xu, S.; Zhang, Y.; Liu, H.; La Russa, M.; Xie, M.; Ding, S.; et al. Small molecules enhance CRISPR genome editing in pluripotent stem cells. Cell Stem Cell 2015, 16, 142–147. [Google Scholar] [CrossRef] [Green Version]
- Chu, V.T.; Weber, T.; Wefers, B.; Wurst, W.; Sander, S.; Rajewsky, K.; Kuhn, R. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 2015, 33, 543–548. [Google Scholar] [CrossRef] [Green Version]
- Maruyama, T.; Dougan, S.K.; Truttmann, M.C.; Bilate, A.M.; Ingram, J.R.; Ploegh, H.L. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat. Biotechnol. 2015, 33, 538–542. [Google Scholar] [CrossRef]
- Robert, F.; Barbeau, M.; Ethier, S.; Dostie, J.; Pelletier, J. Pharmacological inhibition of DNA-PK stimulates Cas9-mediated genome editing. Genome Med. 2015, 7, 93. [Google Scholar] [CrossRef] [Green Version]
- Shin, H.R.; See, J.E.; Kweon, J.; Kim, H.S.; Sung, G.J.; Park, S.; Jang, A.H.; Jang, G.; Choi, K.C.; Kim, I.; et al. Small-molecule inhibitors of histone deacetylase improve CRISPR-based adenine base editing. Nucleic Acids Res. 2021, 49, 2390–2399. [Google Scholar] [CrossRef]
- Perli, S.D.; Cui, C.H.; Lu, T.K. Continuous genetic recording with self-targeting CRISPR-Cas in human cells. Science 2016, 353, aag0511. [Google Scholar] [CrossRef] [Green Version]
- Tang, W.; Liu, D.R. Rewritable multi-event analog recording in bacterial and mammalian cells. Science 2018, 360, aap8992. [Google Scholar] [CrossRef] [Green Version]
- McKenna, A.; Findlay, G.M.; Gagnon, J.A.; Horwitz, M.S.; Schier, A.F.; Shendure, J. Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science 2016, 353, aaf7907. [Google Scholar] [CrossRef] [Green Version]
- Choi, J.; Chen, W.; Minkina, A.; Chardon, F.M.; Suiter, C.C.; Regalado, S.G.; Domcke, S.; Hamazaki, N.; Lee, C.; Martin, B.; et al. A time-resolved, multi-symbol molecular recorder via sequential genome editing. Nature 2022, 608, 98–107. [Google Scholar] [CrossRef]
- Pausch, P.; Al-Shayeb, B.; Bisom-Rapp, E.; Tsuchida, C.A.; Li, Z.; Cress, B.F.; Knott, G.J.; Jacobsen, S.E.; Banfield, J.F.; Doudna, J.A. CRISPR-CasPhi from huge phages is a hypercompact genome editor. Science 2020, 369, 333–337. [Google Scholar] [CrossRef]
- Li, Z.; Zhong, Z.; Wu, Z.; Pausch, P.; Al-Shayeb, B.; Amerasekera, J.; Doudna, J.A.; Jacobsen, S.E. Genome editing in plants using the compact editor CasPhi. Proc. Natl. Acad. Sci. USA 2023, 120, e2216822120. [Google Scholar] [CrossRef] [PubMed]
- Al-Shayeb, B.; Skopintsev, P.; Soczek, K.M.; Stahl, E.C.; Li, Z.; Groover, E.; Smock, D.; Eggers, A.R.; Pausch, P.; Cress, B.F.; et al. Diverse virus-encoded CRISPR-Cas systems include streamlined genome editors. Cell 2022, 185, 4574–4586.e16. [Google Scholar] [CrossRef] [PubMed]
- Karvelis, T.; Druteika, G.; Bigelyte, G.; Budre, K.; Zedaveinyte, R.; Silanskas, A.; Kazlauskas, D.; Venclovas, C.; Siksnys, V. Transposon-associated TnpB is a programmable RNA-guided DNA endonuclease. Nature 2021, 599, 692–696. [Google Scholar] [CrossRef]
- Altae-Tran, H.; Kannan, S.; Demircioglu, F.E.; Oshiro, R.; Nety, S.P.; McKay, L.J.; Dlakic, M.; Inskeep, W.P.; Makarova, K.S.; Macrae, R.K.; et al. The widespread IS200/IS605 transposon family encodes diverse programmable RNA-guided endonucleases. Science 2021, 374, 57–65. [Google Scholar] [CrossRef] [PubMed]
- Sasnauskas, G.; Tamulaitiene, G.; Druteika, G.; Carabias, A.; Silanskas, A.; Kazlauskas, D.; Venclovas, C.; Montoya, G.; Karvelis, T.; Siksnys, V. TnpB structure reveals minimal functional core of Cas12 nuclease family. Nature 2023, 616, 384–389. [Google Scholar] [CrossRef] [PubMed]
- Han, D.; Xiao, Q.; Wang, Y.; Zhang, H.; Dong, X.; Li, G.; Kong, X.; Wang, S.; Song, J.; Zhang, W.; et al. Development of miniature base editors using engineered IscB nickase. Nat. Methods 2023, 20, 1029–1036. [Google Scholar] [CrossRef]
- Saito, M.; Xu, P.; Faure, G.; Maguire, S.; Kannan, S.; Altae-Tran, H.; Vo, S.; Desimone, A.; Macrae, R.K.; Zhang, F. Fanzor is a eukaryotic programmable RNA-guided endonuclease. Nature 2023, 1–3. [Google Scholar] [CrossRef] [PubMed]
- Jiang, K.; Lim, J.; Sgrizzi, S.; Trinh, M.; Kayabolen, A.; Yutin, N.; Koonin, E.V.; Abudayyeh, O.O.; Gootenberg, J.S. Programmable RNA-guided endonucleases are widespread in eukaryotes and their viruses. bioRxiv 2023. [Google Scholar] [CrossRef]
- Wilkinson, M.E.; Frangieh, C.J.; Macrae, R.K.; Zhang, F. Structure of the R2 non-LTR retrotransposon initiating target-primed reverse transcription. Science 2023, 380, 301–308. [Google Scholar] [CrossRef]
DSB-Dependent Editor | Base Editor | Prime Editor | ||||||
---|---|---|---|---|---|---|---|---|
ZFN | TALEN | SpCas9 | CBE | ABE | CGBE | AXBE/AYBE | ||
Type of DNA damage | DSB | DSB | DSB | SSB | SSB | SSB | SSB | SSB (PE2) or DSB (PE3) |
Type of editing | Indel; Knock-in; Base mutation /correction | Indel; Knock-in; Base mutation /correction | Indel; Knock-in; Translocation; Base mutation /correction | Base substitution | Base substitution | Base substitution | Base substitution; | Base substitution; Indel; Recombination |
p53 activation? | Yes | Yes | Yes | No | No | N/A | N/A | No |
On-target specificity | + | + | ++ | ++ | +++ | ++ | + (C/T mix) | +++ |
Off-target effects on DNA | ++ | ++ | ++ | ++ | Very low | ++ (based on CBE) | Very low (based on ABE) | Low |
Off-target effects on RNA | - | - | - | ++ | + | ++ (based on CBE) | + (based on ABE) | - |
Applied in human PSCs? | Yes | Yes | Yes | Yes | Yes | No | No | Yes |
Clinical trial? | Yes | Yes | Yes | Yes | Yes | No | No | No |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zhang, Z.; Bao, X.; Lin, C.-P. Progress and Prospects of Gene Editing in Pluripotent Stem Cells. Biomedicines 2023, 11, 2168. https://doi.org/10.3390/biomedicines11082168
Zhang Z, Bao X, Lin C-P. Progress and Prospects of Gene Editing in Pluripotent Stem Cells. Biomedicines. 2023; 11(8):2168. https://doi.org/10.3390/biomedicines11082168
Chicago/Turabian StyleZhang, Zhenwu, Xinyu Bao, and Chao-Po Lin. 2023. "Progress and Prospects of Gene Editing in Pluripotent Stem Cells" Biomedicines 11, no. 8: 2168. https://doi.org/10.3390/biomedicines11082168
APA StyleZhang, Z., Bao, X., & Lin, C. -P. (2023). Progress and Prospects of Gene Editing in Pluripotent Stem Cells. Biomedicines, 11(8), 2168. https://doi.org/10.3390/biomedicines11082168