DAJIN enables multiplex genotyping to simultaneously validate intended and unintended target genome editing outcomes

Genome editing can introduce designed mutations into a target genomic site. Recent research has revealed that it can also induce various unintended events such as structural variations, small indels, and substitutions at, and in some cases, away from the target site. These rearrangements may result in confounding phenotypes in biomedical research samples and cause a concern in clinical or agricultural applications. However, current genotyping methods do not allow a comprehensive analysis of diverse mutations for phasing and mosaic variant detection. Here, we developed a genotyping method with an on-target site analysis software named Determine Allele mutations and Judge Intended genotype by Nanopore sequencer (DAJIN) that can automatically identify and classify both intended and unintended diverse mutations, including point mutations, deletions, inversions, and cis double knock-in at single-nucleotide resolution. Our approach with DAJIN can handle approximately 100 samples under different editing conditions in a single run. With its high versatility, scalability, and convenience, DAJIN-assisted multiplex genotyping may become a new standard for validating genome editing outcomes.

[1]  Satoru Takahashi,et al.  EXOC1 plays an integral role in spermatogonia pseudopod elongation and spermatocyte stable syncytium formation in mice , 2021, eLife.

[2]  R. Knight,et al.  High-accuracy long-read amplicon sequences using unique molecular identifiers with Nanopore or PacBio sequencing , 2021, Nature Methods.

[3]  L. Teboul,et al.  Anticipating and Identifying Collateral Damage in Genome Editing. , 2020, Trends in genetics : TIG.

[4]  S. Kaul,et al.  FREQUENT GENE CONVERSION IN HUMAN EMBRYOS INDUCED BY DOUBLE STRAND BREAKS , 2020, bioRxiv.

[5]  Takashi Yamamoto,et al.  A simple and practical workflow for genotyping of CRISPR–Cas9‐based knockout phenotypes using multiplexed amplicon sequencing , 2020, Genes to cells : devoted to molecular & cellular mechanisms.

[6]  A. Yoshiki,et al.  Efficient production of large deletion and gene fragment knock-in mice mediated by genome editing with Cas9-mouse Cdt1 in mouse zygotes. , 2020, Methods.

[7]  Waseem Qasim,et al.  Variability in Genome Editing Outcomes: Challenges for Research Reproducibility and Clinical Safety , 2020, Molecular therapy : the journal of the American Society of Gene Therapy.

[8]  Yanyi Huang,et al.  Long-read individual-molecule sequencing reveals CRISPR-induced genetic heterogeneity in human ESCs , 2020, bioRxiv.

[9]  Tobias Marschall,et al.  Haplotype threading: accurate polyploid phasing from long reads , 2020, Genome Biology.

[10]  A. Atala,et al.  CRISPR/Cas9 increases mitotic gene conversion in human cells , 2020, Gene Therapy.

[11]  A. Korobeynikov,et al.  Sequence Analysis , 2020, Encyclopedia of Bioinformatics and Computational Biology.

[12]  Yuri B Schwartz,et al.  Pervasive head-to-tail insertions of DNA templates mask desired CRISPR-Cas9–mediated genome editing events , 2020, Science Advances.

[13]  S. Sukumar,et al.  Targeted nanopore sequencing with Cas9-guided adaptor ligation , 2020, Nature Biotechnology.

[14]  Li Fang,et al.  NanoCaller for accurate detection of SNPs and indels in difficult-to-map regions from long-read sequencing by haplotype-aware deep neural networks , 2019, Genome Biology.

[15]  Ruibang Luo,et al.  Clair: Exploring the limit of using a deep neural network on pileup data for germline variant calling , 2019, bioRxiv.

[16]  Charles D. Yeh,et al.  Advances in genome editing through control of DNA repair pathways , 2019, Nature Cell Biology.

[17]  S. Wells,et al.  Application of long-read sequencing for robust identification of correct alleles in genome edited animals , 2019, bioRxiv.

[18]  Bo Huang,et al.  Deep profiling reveals substantial heterogeneity of integration outcomes in CRISPR knock-in experiments , 2019, bioRxiv.

[19]  Aidan R. O’Brien,et al.  Reproducibility of CRISPR-Cas9 methods for generation of conditional mouse alleles: a multi-center evaluation , 2019, Genome Biology.

[20]  Matthew C. Canver,et al.  CRISPResso2 provides accurate and rapid genome editing sequence analysis , 2019, Nature Biotechnology.

[21]  Mark S. Anderson,et al.  A large CRISPR-induced bystander mutation causes immune dysregulation , 2019, Communications Biology.

[22]  F. Matsuda,et al.  A mutation in transcription factor MAFB causes Focal Segmental Glomerulosclerosis with Duane Retraction Syndrome. , 2018, Kidney international.

[23]  A. Bradley,et al.  Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements , 2018, Nature Biotechnology.

[24]  Brent S. Pedersen,et al.  Bioconda: sustainable and comprehensive software distribution for the life sciences , 2018, Nature Methods.

[25]  Lydia Teboul,et al.  Application of long single-stranded DNA donors in genome editing: generation and validation of mouse mutants , 2018, BMC Biology.

[26]  Denise G. Lanza,et al.  Comparative analysis of single-stranded DNA donors to generate conditional null mouse alleles , 2018, BMC Biology.

[27]  Leland McInnes,et al.  UMAP: Uniform Manifold Approximation and Projection for Dimension Reduction , 2018, ArXiv.

[28]  Fengtang Yang,et al.  Revealing hidden complexities of genomic rearrangements generated with Cas9 , 2017, Scientific Reports.

[29]  Heng Li,et al.  Minimap2: pairwise alignment for nucleotide sequences , 2017, Bioinform..

[30]  Jianhui Gong,et al.  Correction of a pathogenic gene mutation in human embryos , 2017, Nature.

[31]  Michael C. Schatz,et al.  Accurate detection of complex structural variations using single molecule sequencing , 2017, Nature Methods.

[32]  Lothar Hennighausen,et al.  CRISPR/Cas9 targeting events cause complex deletions and insertions at 17 sites in the mouse genome , 2017, Nature Communications.

[33]  S. Wells,et al.  Analysing the outcome of CRISPR-aided genome editing in embryos: Screening, genotyping and quality control. , 2017, Methods.

[34]  Edwin Cuppen,et al.  Mapping and phasing of structural variation in patient genomes using nanopore sequencing , 2017, Nature Communications.

[35]  Leland McInnes,et al.  hdbscan: Hierarchical density based clustering , 2017, J. Open Source Softw..

[36]  Guillaume Pavlovic,et al.  Efficient and rapid generation of large genomic variants in rats and mice using CRISMERE , 2017, Scientific Reports.

[37]  Jin-Soo Kim,et al.  Cas-analyzer: an online tool for assessing genome editing results using NGS data , 2016, Bioinform..

[38]  David R. Liu,et al.  Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage , 2016, Nature.

[39]  Justin Chu,et al.  NanoSim: nanopore sequence read simulator based on statistical characterization , 2016, bioRxiv.

[40]  Steve D. M. Brown,et al.  Correction of the auditory phenotype in C57BL/6N mice via CRISPR/Cas9-mediated homology directed repair , 2016, Genome Medicine.

[41]  T. Mashimo,et al.  Simple Genome Editing of Rodent Intact Embryos by Electroporation , 2015, PloS one.

[42]  A. Regev,et al.  Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System , 2015, Cell.

[43]  Stefan Mundlos,et al.  Deletions, Inversions, Duplications: Engineering of Structural Variants using CRISPR/Cas in Mice. , 2015, Cell reports.

[44]  B. van Steensel,et al.  Easy quantitative assessment of genome editing by sequence trace decomposition , 2014, Nucleic acids research.

[45]  Kun Zhang,et al.  Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. , 2014, Cell stem cell.

[46]  Matthew C. Canver,et al.  Characterization of Genomic Deletion Efficiency Mediated by Clustered Regularly Interspaced Palindromic Repeats (CRISPR)/Cas9 Nuclease System in Mammalian Cells*♦ , 2014, The Journal of Biological Chemistry.

[47]  Satoru Takahashi,et al.  Simple generation of albino C57BL/6J mice with G291T mutation in the tyrosinase gene by the CRISPR/Cas9 system , 2014, Mammalian Genome.

[48]  Gang Bao,et al.  Quantifying genome-editing outcomes at endogenous loci with SMRT sequencing. , 2014, Cell reports.

[49]  Hao Yin,et al.  Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype , 2014, Nature Biotechnology.

[50]  Bo Zhang,et al.  Chromosomal deletions and inversions mediated by TALENs and CRISPR/Cas in zebrafish , 2013, Nucleic acids research.

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

[52]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[53]  Erin L. Doyle,et al.  Targeting DNA Double-Strand Breaks with TAL Effector Nucleases , 2010, Genetics.

[54]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[55]  K. Kurimoto,et al.  Critical function of Prdm14 for the establishment of the germ cell lineage in mice , 2008, Nature Genetics.

[56]  Hans-Peter Kriegel,et al.  LOF: identifying density-based local outliers , 2000, SIGMOD '00.

[57]  SallyAnn Harbison,et al.  A review of the potential of the MinION™ single‐molecule sequencing system for forensic applications , 2018, WIREs Forensic Science.

[58]  Tao Jiang,et al.  Sequence analysis H-PoP and H-PoPG: heuristic partitioning algorithms for single individual haplotyping of polyploids , 2016 .

[59]  Terrence S. Furey,et al.  The UCSC Table Browser data retrieval tool , 2004, Nucleic Acids Res..