AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Full Length Article | Open Access

Nucleolus localization of SpyCas9 affects its stability and interferes with host protein translation in mammalian cells

Renke TanaWenhao DubYiyang LiuaXiaoji CongcMeirong BaidChenxiao JiangaZengxia LiaMinjia TancDengke K. MadQiang Huangb( )Wei Jianga( )Yongjun Danga( )
Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200043, PR China
Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

The CRISPR/Cas9 system, originally derived from the prokaryotic adaptive immune system, has been developed as efficient genome editing tools. It enables precise gene manipulation on chromosomal DNA through the specific binding of programmable sgRNA to target DNA, and the Cas9 protein, which has endonuclease activity, will cut a double strand break at specific locus. However, Cas9 is a foreign protein in mammalian cells, and the potential risks associated with its introduction into mammalian cells are not fully understood. In this study, we performed pull-down and mass spectrometry (MS) analysis of Streptococcus pyogenes Cas9 (SpyCas9) interacting proteins in HEK293T cells and showed that the majority of Cas9-associated proteins identified by MS were localized in the nucleolus. Interestingly, we further discovered that the Cas9 protein contains a sequence encoding a nucleolus detention signal (NoDS). Compared with wild-type (WT) Cas9, NoDS-mutated variants of Cas9 (mCas9) are less stable, although their gene editing activity is minimally affected. Overexpression of WT Cas9, but not mCas9, causes general effects on transcription and protein translation in the host cell. Overall, identification of NoDS in Cas9 will improve the understanding of Cas9's biological function in vivo, and the removal of NoDS in Cas9 may enhance its safety for future clinical use.

References

1

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(6096): 816-821.

2

Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121): 819-823.

3

Knott GJ, Doudna JA. CRISPR-Cas guides the future of genetic engineering. Science. 2018;361(6405): 866-869.

4

Kellner MJ, Koob JG, Gootenberg JS, Abudayyeh OO, Zhang F. SHERLOCK: nucleic acid detection with CRISPR nucleases. Nat Protoc. 2019;14(10): 2986-3012.

5

Replogle JM, Norman TM, Xu A, et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing. Nat Biotechnol. 2020;38(8): 954-961.

6

Jaitin DA, Weiner A, Yofe I, et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell. 2016;167(7): 1883-1896. e15.

7

Jost M, Weissman JS. CRISPR approaches to small molecule target identification. ACS Chem Biol. 2018;13(2): 366-375.

8

Kampmann M. Elucidating drug targets and mechanisms of action by genetic screens in mammalian cells. Commun (Camb). 2017;53(53): 7162-7167.

9

Swiech L, Heidenreich M, Banerjee A, et al. In vivo interrogation of gene function in the mammalian brain using CRISPR-Cas9. Nat Biotechnol. 2015;33(1): 102-106.

10

Lin A, Giuliano CJ, Sayles NM, Sheltzer JM. CRISPR/Cas9 mutagenesis invalidates a putative cancer dependency targeted in on-going clinical trials. Elife. 2017;6: e24179.

11

Dominguez AA, Lim WA, Qi LS. Beyond editing: repurposing CRISPR–Cas9 for precision genome regulation and interrogation. Nat Rev Mol Cell Biol. 2015;17(1): 5-15.

12

Yin H, Xue W, Chen S, et al. Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol. 2014;32(6): 551-553.

13

Kennedy EM, Bassit LC, Mueller H, et al. Suppression of hepatitis B virus DNA accumulation in chronically infected cells using a bacterial CRISPR/Cas RNA-guided DNA endonuclease. Virology. 2015;476: 196-205.

14

Eyquem J, Mansilla-Soto J, Giavridis T, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017;543(7643): 113-117.

15

Liu X, Zhang Y, Cheng C, et al. CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res. 2017;27(1): 154-157.

16

Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339(6121): 823-826.

17

Ran FA, Cong L, Yan WX, et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520(7546): 186-191.

18

Kang XJ, Caparas CIN, Soh BS, Fan Y. Addressing challenges in the clinical applications associated with CRISPR/Cas9 technology and ethical questions to prevent its misuse. Protein Cell. 2017;8(11): 791-795.

19

Charlesworth CT, Deshpande PS, Dever DP, et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat Med. 2019;25(2): 249-254.

20

Kim S, Koo T, Jee HG, et al. CRISPR RNAs trigger innate immune responses in human cells. Genome Res. 2018;28(3): 367-373.

21

Kleinstiver BP, Pattanayak V, Prew MS, et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature. 2016;529(7587): 490-495.

22

Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F. Rationally engineered Cas9 nucleases with improved specificity. Science. 2016;351(6268): 84-88.

23

Chen JS, Dagdas YS, Kleinstiver BP, et al. Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature. 2017;550(7676): 407-410.

24

Yu C, Liu Y, Ma T, et al. Small molecules enhance CRISPR genome editing in pluripotent stem cells. Cell Stem Cell. 2015;16(2): 142-147.

25

Wienert B, Shin J, Zelin E, Pestal K, Corn JE. In vitro-transcribed guide RNAs trigger an innate immune response via the RIG-Ⅰ pathway. PLoS Biol. 2018;16(7): e2005840.

26

Chen B, Gilbert LA, Cimini BA, et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell. 2013;155(7): 1479-1491.

27

Hoellerich E, Dunagan C, Maring D, et al. Nucleolar localization of SmMAK16 protein from Schistosoma mansoni is regulated by three distinct signals that function independent of pH or phosphorylation status. Mol Biochem Parasitol. 2014;193(1): 9-16.

28

Audas TE, Jacob MD, Lee S. The nucleolar detention pathway. Cell Cycle. 2012;11(11): 2059-2062.

29

Audas TE, Jacob MD, Lee S. Immobilization of proteins in the nucleolus by ribosomal intergenic spacer noncoding RNA. Mol Cell. 2012;45(2): 147-157.

30

Sun H, Fu S, Cui S, et al. Development of a CRISPR-SaCas9 system for projection- and function-specific gene editing in the rat brain. Sci Adv. 2020;6(12): eaay6687.

31

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(3): 665-676. e614.

32

Fu Y, Foden JA, Khayter C, et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol. 2013;31(9): 822.

33

Kleinstiver BP, Pattanayak V, Prew MS, et al. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature. 2016;529(7587): 490.

34

Xie Q, Weng X, Lu L, Lin Z, Xu X, Fu C. A sensitive fluorescent sensor for quantification of alpha-fetoprotein based on immunosorbent assay and click chemistry. Biosens Bioelectron. 2016;77: 46-50.

35

Raska I, Shaw PJ, Cmarko D. Structure and function of the nucleolus in the spotlight. Curr Opin Cell Biol. 2006;18(3): 325-334.

36

Olson MOJ. Sensing cellular stress: another new function for the nucleolus. Sci STKE. 2004;2004(224): pe10.

37

Frottin F, Schueder F, Tiwary S, et al. The nucleolus functions as a phase-separated protein quality control compartment. Science. 2019;365(6451): 342-347.

38

Enache OM, Rendo V, Abdusamad M, et al. Cas9 activates the p53 pathway and selects for p53-inactivating mutations. Nat Genet. 2020;52(7): 662-668.

39

Bernardi R, Scaglioni PP, Bergmann S, Horn HF, Vousden KH, Pandolfi PP. PML regulates p53 stability by sequestering Mdm2 to the nucleolus. Nat Cell Biol. 2004;6(7): 665-672.

40

Rubbi CP, Milner J. Disruption of the nucleolus mediates stabilization of p53 in response to DNA damage and other stresses. EMBO J. 2003;22(22): 6068-6077.

41

Boyd MT, Vlatkovic N, Rubbi CP. The nucleolus directly regulates p53 export and degradation. J Cell Biol. 2011;194(5): 689-703.

42

Ye L, Wang J, Beyer AI, et al. Seamless modification of wild-type induced pluripotent stem cells to the natural CCR5Δ32 mutation confers resistance to HIV infection. Proc Natl Acad Sci USA. 2014;111(26): 9591-9596.

43

Schwank G, Koo BK, Sasselli V, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13(6): 653-658.

44

Geurts MH, de Poel E, Amatngalim GD, et al. CRISPR-based adenine editors correct nonsense mutations in a cystic fibrosis organoid biobank. Cell Stem Cell. 2020;26(4): 503-510.e7.

45

Maji B, Gangopadhyay SA, Lee M, et al. A high-throughput platform to identify small-molecule inhibitors of CRISPR-Cas9. Cell. 2019;177(4): 1067-1079.e19.

46

Gangopadhyay SA, Cox KJ, Manna D, et al. Precision control of CRISPR-Cas9 using small molecules and light. Biochemistry. 2019;58(4): 234-244.

47

Li Z, Cheng Z, Raghothama C, et al. USP9X controls translation efficiency via deubiquitination of eukaryotic translation initiation factor 4A1. Nucleic Acids Res. 2018;46(2): 823-839.

48

Huai C, Li G, Yao R, et al. Structural insights into DNA cleavage activation of CRISPR-Cas9 system. Nat Commun. 2017;8(1): 1375.

49

Lu T, Hu JC, Lu WC, et al. Identification of small molecule inhibitors targeting the SMARCA2 bromodomain from a high-throughput screening assay. Acta Pharmacol Sin. 2018;39(9): 1544-1552.

Genes & Diseases
Pages 731-740
Cite this article:
Tan R, Du W, Liu Y, et al. Nucleolus localization of SpyCas9 affects its stability and interferes with host protein translation in mammalian cells. Genes & Diseases, 2022, 9(3): 731-740. https://doi.org/10.1016/j.gendis.2020.09.003

410

Views

3

Downloads

11

Crossref

N/A

Web of Science

6

Scopus

0

CSCD

Altmetrics

Received: 14 July 2020
Revised: 14 September 2020
Accepted: 15 September 2020
Published: 25 September 2020
© 2020, Chongqing Medical University.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return