p53 inhibits CRISPR–Cas9 engineering in human pluripotent stem cells

CRISPR/Cas9 has revolutionized our ability to engineer genomes and conduct genome-wide screens in human cells1–3. Whereas some cell types are amenable to genome engineering, genomes of human pluripotent stem cells (hPSCs) have been difficult to engineer, with reduced efficiencies relative to tumour cell lines or mouse embryonic stem cells3–13. Here, using hPSC lines with stable integration of Cas9 or transient delivery of Cas9-ribonucleoproteins (RNPs), we achieved an average insertion or deletion (indel) efficiency greater than 80%. This high efficiency of indel generation revealed that double-strand breaks (DSBs) induced by Cas9 are toxic and kill most hPSCs. In previous studies, the toxicity of Cas9 in hPSCs was less apparent because of low transfection efficiency and subsequently low DSB induction3. The toxic response to DSBs was P53/TP53-dependent, such that the efficiency of precise genome engineering in hPSCs with a wild-type P53 gene was severely reduced. Our results indicate that Cas9 toxicity creates an obstacle to the high-throughput use of CRISPR/Cas9 for genome engineering and screening in hPSCs. Moreover, as hPSCs can acquire P53 mutations14, cell replacement therapies using CRISPR/Cas9-enginereed hPSCs should proceed with caution, and such engineered hPSCs should be monitored for P53 function.CRISPR–Cas9-induced DNA damage triggers p53 to limit the efficiency of gene editing in human pluripotent cells.

[1]  Steven Lin,et al.  Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery , 2014, eLife.

[2]  U. Moll,et al.  Links between mutant p53 and genomic instability , 2012, Journal of cellular biochemistry.

[3]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[4]  Y. Liu,et al.  Gene targeting in human pluripotent stem cells. , 2011, Methods in molecular biology.

[5]  R. Jaenisch,et al.  Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases , 2009, Nature Biotechnology.

[6]  T. Golub,et al.  Genomic Copy Number Dictates a Gene-Independent Cell Response to CRISPR/Cas9 Targeting. , 2016, Cancer discovery.

[7]  L. Banaszynski,et al.  A Rapid, Reversible, and Tunable Method to Regulate Protein Function in Living Cells Using Synthetic Small Molecules , 2006, Cell.

[8]  Luigi Naldini,et al.  Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery , 2007, Nature Biotechnology.

[9]  J. Trent,et al.  WAF1, a potential mediator of p53 tumor suppression , 1993, Cell.

[10]  Zengrong Zhu,et al.  An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. , 2014, Cell stem cell.

[11]  R. Jaenisch,et al.  Induced Pluripotent Stem Cells Meet Genome Editing. , 2016, Cell stem cell.

[12]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[13]  John A. Tallarico,et al.  Functional CRISPR screening identifies the ufmylation pathway as a regulator of SQSTM1/p62 , 2016, eLife.

[14]  R. Handsaker,et al.  Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations , 2017, Nature.

[15]  L. Vassilev,et al.  In Vivo Activation of the p53 Pathway by Small-Molecule Antagonists of MDM2 , 2004, Science.

[16]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[17]  Y Taya,et al.  Activation of the ATM kinase by ionizing radiation and phosphorylation of p53. , 1998, Science.

[18]  Jeffrey A. Porter,et al.  CLK2 inhibition ameliorates autistic features associated with SHANK3 deficiency , 2016, Science.

[19]  D. Lane,et al.  p53, guardian of the genome , 1992, Nature.

[20]  Yang Xu,et al.  Modeling disease in human ESCs using an efficient BAC-based homologous recombination system. , 2010, Cell stem cell.

[21]  Xin Li,et al.  A comparison of non-integrating reprogramming methods , 2014, Nature Biotechnology.

[22]  S. Jaeger,et al.  Causal Network Models for Predicting Compound Targets and Driving Pathways in Cancer , 2014, Journal of biomolecular screening.

[23]  Eli J. Fine,et al.  DNA targeting specificity of RNA-guided Cas9 nucleases , 2013, Nature Biotechnology.

[24]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[25]  Sam W. Lee,et al.  The guardian of the genome , 2015, Science.

[26]  Davide Heller,et al.  STRING v10: protein–protein interaction networks, integrated over the tree of life , 2014, Nucleic Acids Res..

[27]  D. Durocher,et al.  High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities , 2015, Cell.

[28]  T. Ichisaka,et al.  Suppression of induced pluripotent stem cell generation by the p53–p21 pathway , 2009, Nature.

[29]  L. Pevny,et al.  Human embryonic stem cells have constitutively active Bax at the Golgi and are primed to undergo rapid apoptosis. , 2012, Molecular cell.

[30]  A. Letai,et al.  High mitochondrial priming sensitizes hESCs to DNA-damage-induced apoptosis. , 2013, Cell stem cell.

[31]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[32]  James A. Thomson,et al.  Homologous recombination in human embryonic stem cells , 2003, Nature Biotechnology.

[33]  T. Nomura,et al.  Signaling through the antigen receptor of B lymphocytes activates a p53-independent pathway of c-Myc-induced apoptosis , 1999, Oncogene.

[34]  E. Lander,et al.  Identification and characterization of essential genes in the human genome , 2015, Science.

[35]  R. Dolmetsch,et al.  A deleterious Nav1.1 mutation selectively impairs telencephalic inhibitory neurons derived from Dravet Syndrome patients , 2016, eLife.

[36]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[37]  Ahmed Enayetallah,et al.  Causal reasoning on biological networks: interpreting transcriptional changes , 2012, Bioinform..

[38]  M. Savio,et al.  Multiple roles of the cell cycle inhibitor p21(CDKN1A) in the DNA damage response. , 2010, Mutation research.

[39]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[40]  David A. Scott,et al.  Double Nicking by RNA-Guided CRISPR Cas9 for Enhanced Genome Editing Specificity , 2013, Cell.

[41]  Hui Zhao,et al.  Knock-in of large reporter genes in human cells via CRISPR/Cas9-induced homology-dependent and independent DNA repair , 2016, Nucleic acids research.

[42]  K. Eggan,et al.  Genetic Ablation of AXL Does Not Protect Human Neural Progenitor Cells and Cerebral Organoids from Zika Virus Infection. , 2016, Cell stem cell.

[43]  Jiong Wu,et al.  Regulation of Apoptosis and Differentiation by p53 in Human Embryonic Stem Cells* , 2007, Journal of Biological Chemistry.

[44]  David A. Scott,et al.  Rationally engineered Cas9 nucleases with improved specificity , 2015, Science.

[45]  James A. Gagnon,et al.  Efficient CRISPR-Cas9-mediated generation of knockin human pluripotent stem cells lacking undesired mutations at the targeted locus. , 2015, Cell reports.

[46]  Namritha Ravinder,et al.  Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. , 2015, Journal of biotechnology.

[47]  Joshua M. Korn,et al.  CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions. , 2016, Cancer discovery.

[48]  M. Pfaffl,et al.  A new mathematical model for relative quantification in real-time RT-PCR. , 2001, Nucleic acids research.

[49]  N. Benvenisty,et al.  Pluripotent stem cells in disease modelling and drug discovery , 2016, Nature Reviews Molecular Cell Biology.