DOI QR코드

DOI QR Code

Generation of knockout mouse models of cyclin-dependent kinase inhibitors by engineered nuclease-mediated genome editing

  • Park, Bo Min (Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University) ;
  • Roh, Jae-il (Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University) ;
  • Lee, Jaehoon (Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University) ;
  • Lee, Han-Woong (Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University)
  • Received : 2018.10.20
  • Accepted : 2018.12.08
  • Published : 2018.12.31

Abstract

Cell cycle dysfunction can cause severe diseases, including neurodegenerative disease and cancer. Mutations in cyclin-dependent kinase inhibitors controlling the G1 phase of the cell cycle are prevalent in various cancers. Mice lacking the tumor suppressors $p16^{Ink4a}$ (Cdkn2a, cyclin-dependent kinase inhibitor 2a), $p19^{Arf}$ (an alternative reading frame product of Cdkn2a,), and $p27^{Kip1}$ (Cdkn1b, cyclin-dependent kinase inhibitor 1b) result in malignant progression of epithelial cancers, sarcomas, and melanomas, respectively. Here, we generated knockout mouse models for each of these three cyclin-dependent kinase inhibitors using engineered nucleases. The $p16^{Ink4a}$ and $p19^{Arf}$ knockout mice were generated via transcription activator-like effector nucleases (TALENs), and $p27^{Kip1}$ knockout mice via clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9). These gene editing technologies were targeted to the first exon of each gene, to induce frameshifts producing premature termination codons. Unlike preexisting embryonic stem cell-based knockout mice, our mouse models are free from selectable markers or other external gene insertions, permitting more precise study of cell cycle-related diseases without confounding influences of foreign DNA.

Keywords

Acknowledgement

Supported by : National Research Foundation of the Republic of Korea, Korea's Ministry of Food and Drug Safety (MFDS)

References

  1. Artelt P, Grannemann R, Stocking C, Friel J, Bartsch J, Hauser H. The prokaryotic neomycin-resistance-encoding gene acts as a transcriptional silencer in eukaryotic cells. Gene 1991; 99(2): 249-254. https://doi.org/10.1016/0378-1119(91)90134-W
  2. Baker DJ, Perez-Terzic C, Jin F, Pitel KS, Niederlander NJ, Jeganathan K, Yamada S, Reyes S, Rowe L, Hiddinga HJ, Eberhardt NL, Terzic A, van Deursen JM. Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat Cell Biol 2008; 10(7): 825-836. https://doi.org/10.1038/ncb1744
  3. Besson A, Dowdy SF, Roberts JM. CDK inhibitors: cell cycle regulators and beyond. Dev Cell 2008; 14(2): 159-169. https://doi.org/10.1016/j.devcel.2008.01.013
  4. Capparelli C, Chiavarina B, Whitaker-Menezes D, Pestell TG, Pestell RG, Hulit J, Ando S, Howell A, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. CDK inhibitors (p16/p19/p21) induce senescence and autophagy in cancer-associated fibroblasts, “fueling” tumor growth via paracrine interactions, without an increase in neo-angiogenesis. Cell Cycle 2012; 11(19): 3599-3610. https://doi.org/10.4161/cc.21884
  5. Choi JY, Yun WB, Kim JE, Lee MR, Park JJ, Song BR, Kim HR, Park JW, Kang MJ, Kang BC, Lee HW, Hwang DY. Successful development of squamous cell carcinoma and hyperplasia in RGEN-mediated p27 KO mice after the treatment of DMBA and TPA. Lab Anim Res 2018; 34(3): 118-125. https://doi.org/10.5625/lar.2018.34.3.118
  6. Deng D, Yan C, Pan X, Mahfouz M, Wang J, Zhu JK, Shi Y, Yan N. Structural basis for sequence-specific recognition of DNA by TAL effectors. Science 2012; 335(6069): 720-723. https://doi.org/10.1126/science.1215670
  7. Doench JG, Fusi N, Sullender M, Hegde M, Vaimberg EW, Donovan KF, Smith I, Tothova Z, Wilen C, Orchard R, Virgin HW, Listgarten J, Root DE. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPRCas9. Nat Biotechnol 2016; 34(2): 184-191. https://doi.org/10.1038/nbt.3437
  8. Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, Li Y, Fine EJ, Wu X, Shalem O, Cradick TJ, Marraffini LA, Bao G, Zhang F. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 2013; 31(9): 827-832. https://doi.org/10.1038/nbt.2647
  9. Joung JK, Sander JD. TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol 2013; 14(1): 49-55. https://doi.org/10.1038/nrm3486
  10. Kurokawa K, Tanaka T, Kato J. p19ARF prevents G1 cyclindependent kinase activation by interacting with MDM2 and activating p53 in mouse fibroblasts. Oncogene 1999; 18(17): 2718-2727. https://doi.org/10.1038/sj.onc.1202628
  11. Lee JH, Rho JI, Devkota S, Sung HY, Lee HW. Developing genetically engineered mouse models using engineered nucleases: Current status, challenges, and the way forward. Drug Discov Today 2016; 20: 13-20. https://doi.org/10.1016/j.ddtec.2016.07.006
  12. Li J, Poi MJ, Tsai MD. Regulatory mechanisms of tumor suppressor P16(INK4A) and their relevance to cancer. Biochemistry 2011; 50(25): 5566-5582. https://doi.org/10.1021/bi200642e
  13. Liggett WH Jr, Sidransky D. Role of the p16 tumor suppressor gene in cancer. J Clin Oncol 1998; 16(3): 1197-1206. https://doi.org/10.1200/JCO.1998.16.3.1197
  14. Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013; 140(15): 3079-3093. https://doi.org/10.1242/dev.091744
  15. McKeller RN, Fowler JL, Cunningham JJ, Warner N, Smeyne RJ, Zindy F, Skapek SX. The Arf tumor suppressor gene promotes hyaloid vascular regression during mouse eye development. Proc Natl Acad Sci U S A 2002; 99(6): 3848-3853. https://doi.org/10.1073/pnas.052484199
  16. Nagy Z. The dysregulation of the cell cycle and the diagnosis of Alzheimer's disease. Biochim Biophys Acta 2007; 1772(4): 402-408. https://doi.org/10.1016/j.bbadis.2006.11.001
  17. Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, Shishido N, Horii I, Loh DY, Nakayama K. Mice lacking p27(Kip1) display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors. Cell 1996; 85(5): 707-720. https://doi.org/10.1016/S0092-8674(00)81237-4
  18. Popp MW, Maquat LE. Leveraging Rules of Nonsense-Mediated mRNA Decay for Genome Engineering and Personalized Medicine. Cell 2016; 165(6): 1319-1322. https://doi.org/10.1016/j.cell.2016.05.053
  19. Roh JI, Cheong C, Sung YH, Lee J, Oh J, Lee BS, Lee JE, Gho YS, Kim DK, Park CB, Lee JH, Lee JW, Kang SM, Lee HW. Perturbation of NCOA6 leads to dilated cardiomyopathy. Cell Rep 2014; 8(4): 991-998. https://doi.org/10.1016/j.celrep.2014.07.027
  20. Roh JI, Kim Y, Oh J, Kim Y, Lee J, Lee J, Chun KH, Lee HW. Hexokinase 2 is a molecular bridge linking telomerase and autophagy. PLoS One 2018; 13(2): e0193182. https://doi.org/10.1371/journal.pone.0193182
  21. Roh JI, Lee J, Park SU, Kang YS, Lee J, Oh AR, Choi DJ, Cha JY, Lee HW. CRISPR-Cas9-mediated generation of obese and diabetic mouse models. Exp Anim 2018; 67(2): 229-237. https://doi.org/10.1538/expanim.17-0123
  22. Romagosa C, Simonetti S, Lopez-Vicente L, Mazo A, Lleonart ME, Castellvi J, Ramon y Cajal S. p16(Ink4a) overexpression in cancer: a tumor suppressor gene associated with senescence and high-grade tumors. Oncogene 2011; 30(18): 2087-2097. https://doi.org/10.1038/onc.2010.614
  23. Sa G, Guo Y, Stacey DW. The regulation of S phase initiation by p27Kip1 in NIH3T3 cells. Cell Cycle 2005; 4(4): 618-627.
  24. Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol 2014; 32(4): 347-355. https://doi.org/10.1038/nbt.2842
  25. Shankland SJ. Cell-cycle control and renal disease. Kidney Int 1997; 52(2): 294-308. https://doi.org/10.1038/ki.1997.335
  26. Sharpless NE, Ramsey MR, Balasubramanian P, Castrillon DH, DePinho RA. The differential impact of p16(INK4a) or p19(ARF) deficiency on cell growth and tumorigenesis. Oncogene 2004; 23(2): 379-385. https://doi.org/10.1038/sj.onc.1207074
  27. Sung YH, Baek IJ, Kim DH, Jeon J, Lee J, Lee K, Jeong D, Kim JS, Lee HW. Knockout mice created by TALEN-mediated gene targeting. Nat Biotechnol 2013; 31(1): 23-24. https://doi.org/10.1038/nbt.2477
  28. Sung YH, Jin Y, Kim S, Lee HW. Generation of knockout mice using engineered nucleases. Methods 2014; 69(1): 85-93. https://doi.org/10.1016/j.ymeth.2014.02.009
  29. Sung YH, Kim JM, Kim HT, Lee J, Jeon J, Jin Y, Choi JH, Ban YH, Ha SJ, Kim CH, Lee HW, Kim JS. Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Res 2014; 24(1): 125-131. https://doi.org/10.1101/gr.163394.113
  30. Ulanet DB, Hanahan D. Loss of p19(Arf) facilitates the angiogenic switch and tumor initiation in a multi-stage cancer model via p53-dependent and independent mechanisms. PLoS One 2010; 5(8): e12454. https://doi.org/10.1371/journal.pone.0012454
  31. Valera A, Perales JC, Hatzoglou M, Bosch F. Expression of the neomycin-resistance (neo) gene induces alterations in gene expression and metabolism. Hum Gene Ther 1994; 5(4): 449-456. https://doi.org/10.1089/hum.1994.5.4-449
  32. Weber JD, Jeffers JR, Rehg JE, Randle DH, Lozano G, Roussel MF, Sherr CJ, Zambetti GP. p53-independent functions of the p19(ARF) tumor suppressor. Genes Dev 2000; 14(18): 2358-2365. https://doi.org/10.1101/gad.827300
  33. Zhivotovsky B, Orrenius S. Cell cycle and cell death in disease: past, present and future. J Intern Med 2010; 268(5): 395-409. https://doi.org/10.1111/j.1365-2796.2010.02282.x

Cited by

  1. Divergence of the PIERCE1 expression between mice and humans as a p53 target gene vol.15, pp.8, 2020, https://doi.org/10.1371/journal.pone.0236881
  2. Effect of PIERCE1 on colorectal cancer vol.69, pp.4, 2020, https://doi.org/10.1538/expanim.19-0155