Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jun 10.
Published in final edited form as: Nat Protoc. 2020 Aug 14;15(9):3009–3029. doi: 10.1038/s41596-020-0361-1

GOTI, a method to identify genome-wide off-target effects of genome editing in mouse embryos

Erwei Zuo 1,2,#, Yidi Sun 3,#, Wu Wei 4,5,6,#, Tanglong Yuan 2,#, Wenqin Ying 1, Hao Sun 7, Liyun Yuan 4, Lars M Steinmetz 6,8,9,*, Yixue Li 4,10,11,12,13,*, Hui Yang 1,*
PMCID: PMC8190672  NIHMSID: NIHMS1701453  PMID: 32796939

Abstract

Genome editing holds great potential for correcting pathogenic mutations. We developed a method named GOTI (genome-wide off-target analysis by two-cell embryo injection) to detect off-target mutations by editing one blastomere of two-cell mouse embryos using either CRISPR-Cas9 or base editors. GOTI directly compares edited and non-edited cells without the interference of genetic background and thus could detect potential off-target variants with high sensitivity. Importantly, the GOTI method was designed to detect potential off-target variants of any genome editing tools by the combination of experimental and computational approaches, which is critical for accurate evaluation of the safety of genome editing tools. Here we provide a detailed protocol for GOTI including mice mating, 2-cell embryo injection, E14.5 embryo digestion, FACS, whole genome sequencing and data analysis. To enhance the utility of GOTI, we also include a computational workflow named GOTI-seq (https://github.com/sydaileen/GOTI-seq) for the sequencing data analysis, which can generate the final genome-wide off-target variants from raw sequencing data directly. The protocol typically takes 20 days from the mice mating to sequencing and 7 days for sequencing data analysis.

Keywords: GOTI, off-target effects, genome editing, two-cell embryo injection

EDITORIAL SUMMARY

Here the Authors describe GOTI (genome-wide off-target analysis by two-cell embryo injection), a method to detect off-target mutations of either CRISPR-Cas9-based genome editing, or base editors. This is achieved by editing one blastomere of a two-cell mouse embryo so that edited and unedited cells from the same genetic background can be compared.

Introduction

The recent development of genome editing tools holds great promise in diverse fields such as animal disease modeling, gene therapy, drug development, genetically modified plants, and biofuel technology1. In addition, gene editing technology has accelerated the study of the functional organization of the genome and the causal links between genetic variations and biological phenotypes25. A booming genetic editing technology within the life sciences field, CRISPR holds great hope for the treatment of genetic diseases6,7. A majority of CRISPR-Cas9 edited products usually contain small indels at the target site due to the non-homologous end joining (NHEJ) in response to double strand breaks (DSBs)8,9. Base editors, on the other hand, induce base pair substitutions using deaminases at the target loci without generating DSBs1114. There are two classes of DNA base editors. Cytosine base editors (CBEs) convert C•G base pairs to A•G base pairs11 and adenine base editors (ABEs) conversely convert A•G base pairs to G•C base pairs13. However, the issue of off-target mutations, which may cause genetic instability and dysfunction, has been a major concern in the application of both these methods1519. Specifically, the potential for off-target effects remains a major barrier to the applications of genome editing for human gene therapy. Several techniques have been developed to detect genome-wide gene-editing off-target activity in cells, including selective enrichment and identification of adapter-tagged DNA ends by sequencing (SITE-Seq)20, high-throughput genome-wide translocation sequencing (HTGTS)21, genome-wide, unbiased identification of double-strand breaks (GUIDE-seq)22 and circularization for in vitro reporting of cleavage effects by sequencing (CIRCLE-seq)23. However, these approaches can only detect off-targets from double-strand breaks (DSBs) generated by genome editing, thus are not applicable to detect single-nucleotide variants (SNVs) in vivo. Given the rapidly increased application of base editors, it has become desirable to develop technologies for the comprehensive evaluation of off-target effects in an unbiased manner.

Recently, we have developed a genome-wide off-target effects detection method, GOTI, for the de novo identification of off-target mutations in mouse embryos24,25. This technique examines off-target effects in a cell population derived from a single gene-edited blastomere, whereas previous studies used large pools of cells within which gene editing outcomes are variable, resulting in loss of signal for random off-target effects due to population averaging. In addition, as the edited and non-edited cells are from one single-ancestor cell, GOTI can minimize any confounding effects of genetic background and somatic mutations. GOTI could thus be generalized to be used for genome editing tools that do not introduce DSBs, including triplex-forming oligonucleotides, base editors, and potentially any other strategy that produces indels or SNVs.

In this article, we systemically describe the experimental procedures of GOTI and bioinformatic pipeline GOTI-seq (https://github.com/sydaileen/GOTI-seq) in detail. We also emphasize the specific steps and small tricks that required great care to ensure the data quality generated. The GOTI method could be broadly applied to evaluate the specificity of genome editing tools in animal models.

Overview of GOTI

The overall workflow of GOTI is illustrated in Fig. 1. A mixture of Cre, gene editing mRNA (Cas9/BE3/ABE7.10) and sgRNA is injected into one blastomere of a 2-cell mouse embryo, derived from Ai9 male mice mating with wild-type female mice26. The action of Cre, injected into only one of the 2 cells of the embryo, is expected to generate a chimeric embryo with half of the cells labeled with tdTomato (colored red) with the Cre-LoxP system26,27 so that the cells that received editing reagents cells can be distinguished from those that did not. When the chimeric embryo reaches E14.5, it is minced into small pieces and digested into a single cell suspension (Fig. 2). The tdTomato+ cells and tdTomato cells are then separated by fluorescence-activated cell sorting (FACS). Next, the two populations of cells are independently processed for high-throughput whole genome sequencing (WGS). Typically, the process from the 2-cell injection to the completion of the final sequencing takes 3 weeks for an experienced operator (Fig. 1a).

Fig. 1. Schematics of GOTI method.

Fig. 1.

a, Overall workflow of GOTI. b, Flowchart of GOTI-seq data processing pipeline. File formats and software used for each step are denoted in parentheses.

Fig. 2. Isolation of embryonic cells.

Fig. 2.

Pictures of key steps 59–61. a, The embryos with the placenta. b, Dissect mouse embryos and wash the embryos with PBS twice briefly. c, Add 5 mL of pre-warmed 37°C Trypsin-EDTA (0.05%) and transfer to a 50mL centrifuge tube.

The WGS reads are subsequently processed by a customized pipeline illustrated in Fig. 1b. Raw sequencing reads are first quality checked and the adapters are further trimmed out. Qualified reads are then mapped to the reference genome. The mapped alignment (BAM format) files are sorted and duplicates marked before downstream analysis. The off-target SNVs and indels are identified by comparing the tdTomato+ cells with tdTomato cells using three variant calling algorithms as indicated (Fig. 1b). Several previous studies have reported the discordance among variant calling methods due to different mathematical models and algorithms2831. Mutect2 applies a Bayesian classifier to detect somatic mutations with even low allele fractions32. Strelka2 detects both SNVs and indels by a mixture-model-based estimation with high speed33. Lofreq is an ultra-sensitive variant caller to call somatic variants34. Meanwhile, Scalpel performs well for the discovery of insertions and deletions35. Different algorithms apply different models and have their own preferences to variants, so to reduce the false positives, we considered the overlap of three algorithms of SNVs or indels as the true variants and annotated these for functional analysis.

Applications of GOTI

We have demonstrated that GOTI is applicable to detect the off-target effects of various genome editing tools including CRISPR/Cas9, BE3 and ABE7.10. We have found numerous de novo SNVs induced by BE3, while CRISPR/Cas9 and ABE7.10 generated no unwanted mutations for the sgRNAs that were examined. Notably, the specificity of CRISPR/Cas9 depends heavily on the choice of sgRNA, so we cannot rule out the possibilities that some other sgRNAs may have significant off-target effects even though we have examined several. GOTI is a valuable method that is expected to help improve the current gene editors and facilitate the generation of new genome editing tools with higher specificity. This method could be applied in the preclinical evaluation of genome editing reagents before their approval for clinical trials in gene therapy. GOTI depends on Cre/LoxP recombination system to distinguish edited cells from unedited cells, so it could theoretically be used in other transgenic animal models carrying the reporting system like rat36,37 or pig38,39.

Alternative methods and advantages of GOTI

There have been several genome-wide methods previously developed for the detection of off-target effects. IDGV characterizes the genome-wide specificity by capturing DSBs in vivo40, HTGTS detects genome-wide translocations generated from DSBs in vitro21, GUIDE-seq relies on the capture of double-stranded oligodeoxynucleotides integrated into the DSBs in vivo22. EndoV-seq41 and Digenome-seq42,43 are both in vitro assays to investigate the sgRNA-dependent off-target effects of genome editing tools. CIRCLE-seq23 examines the in vitro DSBs of naked DNA for the prediction of off-target sites. In addition, DISCOVER-seq tracks the recruitment of MRE11 to DSBs in vivo44. These methods mostly rely on the generation of DSBs or nicks on the genome caused by the cleavage of Cas9, but base editors do not generate DSBs because they use catalytically deficient Cas9. Anderson K.R. et al.45 and Lyer V et al.46 also applied deep sequencing for the detection of sgRNA-dependent off-target effects in genomic specific regions based on sequence similarity. The sgRNA-dependent off-target effects could be solved by using different sgRNAs. However, none of these methods were applicable to detect random off-target effects which are independent of sgRNA or Cas9. Several previous studies also applied WGS to detect potential off-target effects of CRIPSR/Cas9 comparing edited and non-edited animals4450. However, the true off-target variants could not be distinguished from the single nucleotide polymorphisms (SNPs) by the genome comparison of two different individuals. One of the major advantages of GOTI is to directly compare edited and non-edited cells with identical genetic backgrounds. Therefore, potentially random off-target variants could be detected without bias even when they occur at low frequency.

The other main advantage of GOTI is its single-cell nature. The approach detects the off-target effects generated in a single ancestor cell and inherited by all descendant cells, which can be easily detected by sequencing, while previous work has been based on large pools of cells in which random off-target activity would be lost in the population average.

Limitations of GOTI

The embryo has to develop to E14.5 when the whole embryo could be readily digested to obtain enough single cells, so the duration of whole experiment takes up to one month. Also, GOTI relies on mice as a model system and requires regulatory approval with respect to animal welfare. However, GOTI can be theoretically conducted in other animal models with Cre/LoxP recombination system integrated. Another limitation is that GOTI is specific to the species in which it is performed. There are major ethical and legal considerations that prevent its use to assess off-target effects in the context of the human genome. In addition, the accomplishment of some procedures such as two-cell embryo injection requires microinjection apparatus, professional training and technical skill. GOTI is much more expensive compared to other methods based on the enrichment of different off-target sites, which could be used to detect sgRNA-dependent off-target effects.

Experimental design

The generation of GOTI reagents (Steps 1–13 and Box 1)

GOTI reagents consist of single-guide RNA (sgRNA), gene editor (Cas9 or base editor) mRNA and Cre mRNA. These components are generally transcribed in vitro from a T7 bacteriophage promoter (Supplementary Fig. 1). For sgRNA, a DNA template that contains T7 promotor, the designed sequence of sgRNA and the sgRNA scaffold is generated (as described in Steps 11–13) by performing a PCR reaction with the scaffold template (px330) and PCR primers (see Table 1). The forward primer must contain a T7 promoter sequence (20 bp), a sgRNA target sequence (20 bp) and a scaffold template-specific sequence (19 bp). The T7 promoter sequence and the scaffold template-specific sequence are fixed. The reverse primer is a short complementary sequence only targeting the end of the gRNA scaffold in the reverse direction. Then an sgRNA containing the sgRNA target sequence is created by in vitro transcription of the DNA template with the MEGA shortscript T7 kit. For gene editor mRNA, a DNA template that contains the T7 promotor, gene editor coding sequence and polyA sequence is generated (as described in Steps 1–8 for Cas9 or Box 1 for BE mRNA) by performing a PCR reaction with plasmid template and PCR primers (see Table 1). The forward primer includes the T7 promoter and 20–30 bp upstream sequence of start codon (ATG). The Reverse primer is located downstream of the polyA sequence. The mRNA encoding the gene editor is created by in vitro transcription of the DNA template with the mMESSAGE mMACHINE T7 ULTRA kit. For Cre mRNA, a DNA template that contains the T7 promotor, Cre coding sequence and polyA sequence is generated (as described in Steps 9–10) by performing a PCR reaction with pCAG-Cre plasmid template and PCR primers (see Table 1). The forward primer includes the T7 promoter and a sequence upstream of the start codon (ATG). Reverse primers are located downstream of the polyA sequence. The mRNA encoding Cre is created by in vitro transcription of the DNA template with the mMESSAGE mMACHINE T7 ULTRA kit.

Table 1.

Primers for in vitro transcription and genotyping

Primer Sequence (5′−3′) Purpose Step
Cas9 IVT F TAATACGACTCACTATAGGGAGATT
TCAGGTTGGACCGGTG
Generation of Cas9 mRNA 1
Cas9 IVT R GACGTCAGCGTTCGAATTGC Generation of Cas9 mRNA 1
BE3 IVT F TCCGCGGCCGCTAATACGACT Generation of BE3 mRNA 1
BE3 IVT R TGGTTCTTTCCGCCTCAGAAGCC Generation of BE3 mRNA 1
Cre IVT F TAATACGACTCACTATAGGGAGACA
GATCACCTTTCCTATCAACC
Generation of Cre mRNA 2
Cre IVT R TCGGTATTTCCAGCACACTGGA Generation of Cre mRNA 2
sgRNA IVT F TAATACGACTCACTATAGGGNNNNN
NNNNNNNNNNNNNNNGTTTTAGAG
CTAGAAATAG
Generation of sgRNA 9
sgRNA IVT R AAAAGCACCGACTCGGTGCC Generation of sgRNA 9
Tyr-OF GTTATCCTCACACTACTTCTG On-target genotyping 44
Tyr-OR GTAATCCTACCAAGAGTCTCA On-target genotyping 44
Tyr-IF TCCTCACACTACTTCTGATG On-target genotyping 44
Tyr-IR GTCTCAAGATGGAAGATCAC On-target genotyping 44

Validation of on-target editing efficiency in blastocyst (Steps 44–57)

To evaluate the efficiency of on-target editing in the GOTI system (to ensure that the injected embryos were edited by genome editing tools, as if not, downstream experiments would not be performed) in addition to transferring 80% of the injected embryos to the pseudopregnant mother, we culture the remaining 20% to the blastocyst stage in vitro (Step 34). The single blastocyst is then lysed and PCR amplification products of the targeting sites are ligated into T-Vectors. The ligation mixture is transformed into E. coli DH5α and cloned into E. coli (TA clone). Thirty TA clones are picked for Sanger sequencing to estimate the on-target editing efficiency. If at least 6 TA clones (20%) show on-target editing, further in vivo experiments (Steps 35–43) are performed.

FACS gating strategy for isolation of embryonic cells (Steps 58–67)

We describe a step-by-step protocol to digest embryos and isolate embryonic cells by flow cytometry. The procedure is simple and a cell suspension can be prepared from a single mouse embryo using enzymatic digestion and mechanical disaggregation in less than 35 minutes. To maximize cell viability, FACS sorting must be performed as soon as possible after preparation of the sample. We aim to finish sorting within 4 h after digesting embryos. We show the FACS gating strategy for isolation of embryo cells in Supplementary Fig. 2.

Sorting accuracy validation (Step 68)

After FACS, DNA of tdTomato+ and tdTomato cells are extracted separately and PCR amplified. These PCR products are ligated into T-Vectors, and 20 TA clones are picked for Sanger sequencing. Further experiments (Steps 69–76) will be conducted only if less than 10% (2/20) of clones show on-target editing in tdTomato cells and more than 20% (4/20) of clones show on-target editing in tdTomato+ cells. The sensitivity and specificity are used to validate the sorting accuracy of FACS. Some tdTomato+ cells may not be edited and tdTomato- cells might be edited as tdTomato+ and tdTomato- cells may be mis-sorted sometimes. So in order to maximize the sorting accuracy, we set sensitivity and specificity thresholds here to guarantee that tdTomato+ and tdTomato- cells for downstream analyses are well separated. Sensitivity is indicated by the percentage of tdTomato+ cells edited, and specificity is represented by the percentage of tdTomato cells edited (which did not receive editing reagents at the 1-cell stage).

Controls

Two kinds of controls are used in GOTI. The tdTomato cells are sequenced and analyzed together with tdTomato+ cells to eliminate the influence of genetic background. Plus, a control group with only the injection of Cre (Cre-only group) is necessary for the experiment to control the background noise in GOTI. In addition, the survival rate of embryos to blastocysts in the Cre-only group (usually more than 90%) acts as a control for evaluating the toxicity of genome editing tools. Genome editing tools with less than 80% blastocyst rate are potentially toxic to embryos as the normal blastocyst rates are above 80%16. Groups with different sgRNAs and no sgRNAs are optional to control for the sgRNA-dependent and independent off-target effects.

Materials

BIOLOGICAL MATERIALS

  • C57BL/6 female mice; Shanghai Laboratory Animal Center (SLAC) laboratory Animal Co., Ltd (Shanghai SLAC Laboratory)

  • Ai9 male mice26 (full name B6.Cg-Gt (ROSA) 26Sortm9 (CAG-td-Tomato) Hze/J), JAX strain 007909; The Jackson Laboratory.

  • ICR female mice; Shanghai SLAC Laboratory.

  • Vasectomized male mice (ICR strain mice); Shanghai SLAC Laboratory. !CAUTION Experimental procedures involving animals must be carried out according to all relevant institutional and governmental regulations.

  • DH5α competent cells (Tiangen, cat.no CB101)

Reagents

  • KOD-Plus-Neo (toyobo cat. no. KOD-401)

  • Premix Taq (Ex Taq Version 2.0 plus dye (Takara,cat. no.RR902A)

  • DL10,000 DNA Marker (Takara, cat.no 3584A)

  • DL15,000 DNA Marker (Takara, cat.no 3582B)

  • 6 × Loading Buffer (Takara, cat.no 9156)

  • EDTA (Sangon Biotech, cat. no. A500838-0500)

  • Tris (Sangon Biotech, cat.no A100826-0500)

  • Glacial acetic acid (Sangon Biotech, cat.no A501931-0500)

  • PCR DNA primers oligomers (Shanghai HuaGen Biotech Co., Ltd.)

  • mMESSAGE mMACHINE T7 ULTRA kit (Life Technologies, cat.no AM1345)

  • MEGA shortscript T7 kit (Life Technologies, cat.no AM1354)

  • MEGA clear kit (Life Technologies, cat.no AM1908)

  • HEPES-CZB medium (see REAGENT SETUP)

  • Pregnant mare serum gonadotropin (PMSG; Sigma, cat. no. G4527)

  • Human chorionic gonadotropin (hCG; Sigma, cat. no. C8554)

  • KSOM medium (Millipore, cat. no. MR-106-D) ! CAUTION Store the medium at −20°C.

  • After thawing, keep it at 4°C and use it within 2 weeks.

  • Hyaluronidase (Sigma, cat. no. H-3884)

  • M2 medium (Millipore, cat. no. MR-015-D)

  • Cytochalasin B (Sigma, cat. no. C6762)

  • DMSO (Sigma, cat. no. D2650) ! CAUTION It is flammable; harmful if swallowed; toxic when in contact with skin and eye; and use protective gloves and safety glasses when handling.

  • Mineral oil (Sigma, cat. no. M8410)

  • Nuclease-free water (Life Technologies, cat. no. AM9932)

  • Trypsin-EDTA (0.05%) (Gibco, cat. no. 25300062)

  • Dulbecco’s Modified Eagle Medium (DMEM) (GIBCO, cat. no.11965092)

  • Fetal Bovine Serum (FBS) (GIBCO, cat. no. 16000044)

  • Tween 20 (Sangon Biotech, cat.no A100777-0500)

  • Triton X-100 (Sangon Biotech, cat.no A110694-0500)

  • Proteinase K (Tiangen, cat.no RT403-02)

  • DNeasy blood and tissue kit (Qiagen; cat.no 69504)

  • Universal DNA Purification Kit (TIANGEN, cat.no DP214)

  • TIANprep Rapid Mini Plasmid Kit (TIANGEN, cat.no DP105)

  • px330 (Addgene plasmid #42230)

  • px260 (Addgene plasmid #42229)

  • pCMV-BE3 (Addgene plasmid #73021)

  • pCAG-Cre (Yang lab #ZP156)

  • FastDigest BbsI (Thermo Scientific, cat.no FD1014)

  • FastDigest PvuI (Thermo Scientific, cat.no FD0624)

  • FastDigest NotI (Thermo Scientific, cat.no FD0595)

  • Nucleic Acid Dye (TIANGEN, cat.no RT210)

  • NaCl (Sigma, cat.no S-5886)

  • KCl (Sigma, cat.no P-5405)

  • CaCl2 2H2O (Sigma, cat.no C-7902)

  • MgSO4 7H2O (Sigma, cat.no M-5921)

  • KH2PO4 (Sigma, cat.no P-5655)

  • EDTA·2Na·2H2O (Sigma, cat.no E-4884)

  • NaHCO3 (Sigma, cat.no S-5761)

  • L-glutamine (Sigma, cat.no G-8540)

  • Na-lactate (Sigma, cat.no L7900)

  • Sodium Pyouvate (Sigma, cat.no P-8574)

  • Sodium Penicillin (Sigma, cat.no P-3032)

  • Streptomycin (Sigma, cat.no S1277)

  • BSA (Sigma, cat.no A-3311)

  • HEPES (Sigma, cat.no H-7006)

  • H2O (Merck Millipore, cat.no TMS-006-B)

  • 2,2,2-tribromoethanol (Sigma, cat.no T48402)

  • 2-methyl-2-butanol (Sigma, cat.no 240486)

REAGENT SETUP

Ampicillin

Prepare a stock solution of 100 mg/ml in H2O and filter-sterilize. Store at −20°C for up to 12 months.

Cell lysis buffer

Component Volume Final Concentration
Tween (1% (wt/vol) Tween in ddH2O) 1ml 0.1% (wt/vol)
Triton X-100 (1% (wt/vol) Triton X-100 in ddH2O) 1ml 0.1% (wt/vol)
Proteinase K (20 mg/ml) 2ml 4 mg/ml)
ddH2O 6ml
Total 10ml

Store at 4°C for up to 3 months.

LB medium

Add 10g bacto-tryptone, 5g yeast extract and 5g NaCl to 1L ddH2O and sterilize by autoclaving. Store at 4°C for up to 1 month.

LB agar plates

Add 12 g agar to 1l LB medium before autoclaving. To prepare plates, allow medium to cool to 50°C and then add antibiotic stock to achieve a final concentration of 100mg/L, mix by gentle swirling and pour or pipette approximately 10 ml into each sterile Petri dish (60 mm diameter). Note that plates should be covered to prevent evaporation and stored agar side up at 4°C for up to 1 month until use.

50 × TAE buffer

242 g of Tris base, 18.61 g of EDTA and 57.1 ml of glacial acetic acid (100%) per liter of water (final pH 8.5). The stock can be stored at room temperature (25–30 °C) for up to 3 months.

HEPES-CZB medium

(1 liter) Store at 4°C for up to 2 month.

Component Final concentration (g/L) Final Concentration (mM)
NaCl 4.76 81.62
KCl 0.36 4.83
CaCl2 2H2O 0.25 1.70
MgSO4 7H2O 0.29 1.18
KH2PO4 0.16 1.18
EDTA·2Na2·H2O 0.04 0.11
NaHCO3 0.42 5.00
L-glutamine 0.15 1.00
Na-lactate 0.35 31.30
Sodium Pyouvate 0.03 0.27
Sodium Penicillin 3.56 100.00
Streptomycin 0.51 0.70
BSA 5 -
HEPES 0.125 0.48
H2O 1 -

Hyaluronidase (Hy), 10×stock, 10 mg/ml

Add 100 mg of Hy to 10 ml of M2 medium, and then divide it into 100 × 100μl tubes. Store at −20°C for up to 12 months.

M2 + Hy, 10 mg/ml

Add 100μl of Hy stock solution to 900μl M2 medium. Mix it just before use.

Cytochalasin B (CB), 50×stock, 500μg/ml

Add 2 mg of CB to 4 ml DMSO, and then divide it into 100 × 20μl tubes. Store at −20°C for up to 12 months.

HEPES-CZB + CB, 10μg/ml

Add 20μl of CB stock solution to 1ml HEPES-CZB. Mix it just before use.

Avertin A stock solution

Prepareby dissolving 10g of 2,2,2-tribromoethanol in 10 ml of 2-methyl-2-butanol in 50 °C water bath until it is fully dissolved. Prepare a working solution of 2.5% avertin by mixing 2.5ml stock solution with 97.5 ml PBS. Sterilize by passing solution through a 0.22 mm bottle top vacuum filter. Store the solution at 4 °C in the dark for up to 3 months.

DMEM medium

Add 50 ml of FBS to 450 ml of DMEM. Store at 4 °C for up to 2 months.

Equipment

  • Cell strainer (Falcon, 40 μm Cell Strainer, cat.no 352340)

  • Polystyrene round-bottom tube (5 ml, with 35-μm cell strainer cap, 12×75 mm2; BD

  • Biosciences, cat. no. 352235)Falcon 10-cm (100 × 20 mm) dishes; bottoms are suitable for oocyte/embryo collection and lids that are suitable for micromanipulation (Becton Dickinson, cat. no. 353003)

  • Falcon 6-cm (60 × 15 mm) dishes (Becton Dickinson, cat. no. 351007)

  • Microloader tips (Eppendorf, 5242 956.003)

  • Thin Wall Borosilicate glass with filament (Borosilicate, BF100-78-10)

  • Thin Wall Borosilicate glass without filament (Borosilicate, B100-75-10)

  • 0.2 mL PCR Tubes (Axygen cat. no.14-222-262)

  • FemtoJet microinjector (Eppendorf)

  • Inverted microscope with Hoffman optics (Olympus, IX73)

  • Micromanipulator set (Narishige, MMO-202ND)

  • CO2 incubator (Thermo, BB15)

  • Stereo microscope (Olympus, SZ61)

  • Cell Sorter (Beckman, MoFlo XDP)

  • Centrifuge (Eppendorf, 5424R)

  • Micropipette Puller (Sutter Instrumen, P97)

  • Micropipette Microforge (Narishige, MF-900)

  • Fluorescence microscopy (Olympus, BX51)

Procedure

Generation of Cas9 or BE3 mRNA ~10h

  • 1
    For vectors without T7 promoter, firstly provide a template for in vitro transcription of Cas9 or BE3 mRNA, add the T7 promoter sequence to the Cas9 or BE3 coding region by PCR amplification using the appropriate primer pair listed in Table 1 and the following reaction mix (see Box 1 instead for details of how to generate Cas9 or BE3 mRNA with T7 promoters):
    Component Volume Final Concentration
    10× PCR Buffer for KOD -Plus- Neo 5μl 1x
    2 mM dNTPs 5μl 0.2 mM each
    Cas9 IVT F (10μM) for Cas9 2μl 0.4μM
    Cas9 IVT R (10μM) for Cas9 2μl 0.4μM
    KOD -Plus- Neo (1U/μl) 1μl 1.0 U/50 μl
    px260 (100ng/pl) for Cas9 0.5 μl 1ng/μl
    25 mM MgSO4 3μl 1.5 mM
    ddH2O 31.5 μl
    Total 50μl
  • 2
    Perform PCR using the following cycling conditions:
    Cycle number Denature Extend
    1 95°C, 5min
    2–34 95°C, 30s 68°C, 5min
    35 72°C, 15min

Box 1. Linearized plasmid is used as a template for in vitro transcription

For vectors including T7 promoters ~20 bp upstream of coding sequence of gene editing protein, plasmid is linearized by restriction enzyme cutting. Restriction site is selected downstream of polyA. Here we provide a brief protocol for linearization of plasmid pCMV-BE3 and in vitro transcription of BE3 mRNA.

  1. Purify plasmid pcmv-be3, using the TIANprep Rapid Mini Plasmid Kit according to the manufacturer’s instructions.

  2. Prepare the following reaction mixture at room temperature:
    Component Volume Final Concentration
    10× FastDigest Green buffer 5 μl 1x
    0.1 μg/μl
    Plasmid pCMV-BE3 (0.5 μg/μl) 10 μl 5 μl / 50μl
    FastDigest BbsI 5 μl 5 μl / 50μl
    FastDigest PvuI 5 μl 5 μl / 50μl
    FastDigest NotI 5 μl -
    ddH2O 20 μl -
    Total 50μl
  3. Mix gently and spin down for a few seconds.

  4. Incubate at 37°C in water thermostat for 30 min.

  5. Prepare 1x TAE by diluting appropriate amount of 50 x TAE with deionized water.

  6. Mix 0.5g agarose powder with 50 mL 1xTAE in a 250ml conical flask.

  7. Microwave for 1–3min until the agarose is completely dissolved. If not, boil the solution again.

  8. Cool down the agarose solution to about 60 °C.

  9. Add 5 μL of Nucleic Acid Dye to the agarose solution and mix gently.

  10. Pour the agarose into a gel tray at room temperature and rest for 30 mins, until it has completely solidified.

  11. Remove gel dams and place agarose gel into the gel box. Add 1xTAE into the gel box until the gel is covered.

  12. Load 5 μL DNA Marker into the first lane of the gel.

  13. Load 50 μL digested plasmid DNA into the additional well of the gel.

  14. Run the gel at 110 V for 30 min. Three digested DNA fragments are supposed to have 5412, 1170 and 1410 bp in length, respectively, as shown in the Supplementary Fig. 3:.

    CAUTION! Examine the linearized template DNA on a gel to confirm that cleavage is complete.

  15. Excise the 5412 bp DNA fragment band under long wavelength UV light.

  16. Purify DNA fragment from agarose gels using the Universal DNA Purification Kit according to the manufacturer’s instructions.

  17. Elute the DNA fragment with 40 μl nuclease-free water.

  18. Use 1μg purified DNA fragment as the template for in vitro transcription of BE3 mRNA using the mMESSAGE mMACHINE T7 kit according to the protocol.

  19. After purification, dilute the BE3 mRNA to 500 ng/μL with nuclease-free water and check its quality on a 1% (wt/vol) agarose gel in TAE buffer. The in vitro transcribed BE mRNA should have a band at ~1000bp (Supplementary Fig. 3). Failed in vitro transcription products show no clear bands at these positions as also shown in Supplementary Fig. 3.

  20. Dispense 1 μl of the purified mRNA into 0.2 mL PCR Tubes.

    PAUSE POINT The samples could be stored at −80°C for up to 1 year.

  • 3

    Run 5 μL of PCR products on a 1% (wt/vol) agarose gel at 110V for 30 min with 6 × loading buffer in TAE buffer to validate that the DNA fragment is unique and of the expected size (~4.5 kb for Cas9; ~kb for BE3; Supplementary Fig. 3).

  • 4

    Purify PCR products, using the Universal DNA Purification Kit according to the manufacturer’s instructions.

  • 5

    Use 1μg purified PCR product as the template for in vitro transcription of Cas9 mRNA using the mMESSAGE mMACHINE T7 kit according to the kit protocol.

  • 6

    Purify the mRNA using the MEGAclear kit following the manufacturer’s instructions and elute the RNA in 100 μl of TB buffer. Determine the RNA concentration using the NanoDrop 2000 spectrophotometer following the manufacturer’s instructions.

  • 7

    Dilute the purified mRNA to 500 ng/μl in 0.1 mM RNase-free ddH2O and check its quality on a 1% (wt/vol) agarose gel in TAE buffer. The in vitro transcribed Cas9 mRNA should have a band at ~1000bp (Supplementary Fig. 3). Failed in vitro transcription products show no clear bands at these positions as also shown in Supplementary Fig. 3.

  • 8

    Dispense 1 μl of the purified mRNA into 0.2 mL PCR Tubes.

    PAUSE POINT The samples can be stored at −80°C for up to 1 year.

Generation of Cre mRNA~10h

  • 9
    To provide a template for in vitro transcription of Cre mRNA, add the T7 promoter sequence to the Cre coding region by PCR amplification using the appropriate primer pair listed in Table 1, and the following reaction mix:
    Component Volume Final Concentration
    10× PCR Buffer for KOD -Plus- Neo 5μl 1x
    2 mM dNTPs 5μl 0.2 mM each
    Cre IVT F (10μM) for Cre 2μl 0.4μM
    Cre IVT F (10μM) for Cre 2μl 0.4μM
    KOD -Plus- Neo (1U/pl) 1μl 1.0 U/50 μl
    pCAG-Cre (100ng/μl) for Cre 0.5μl 1ng/μl
    25 mM MgSO4 3μl 1.5 mM
    ddH2O 31.5μl
    Total 50μl
  • 10

    Repeat Steps 2–8, diluting the purified sgRNA to 500 ng/μl in 0.1 mM EDTA, the band in Step 3 should be of the expected size (~2kb; Supplementary Fig. 3).

    The in vitro transcribed Cre mRNA should have a band at ~400bp (Supplementary Fig. 3).

    PAUSE POINT The samples can be stored at −80°C for up to 1 year.

Generation of sgRNA ~8h

  • 11
    For sgRNA preparation, prepare the following PCR reaction mix to add the T7 promoter sequence to the sgRNA template by PCR amplification using the appropriate primer pair listed in Table 1:
    Component Volume Final Concentration
    10× PCR Buffer for KOD -Plus- Neo 5μl 1x
    2 mM dNTPs 5μl 0.2 mM each
    sgRNA IVT F (10μM) 2μl 0.4μM
    sgRNA IVT R (10μM) 2μl 0.4μM
    KOD -Plus- Neo (1.0 U/μl) 1μl 1.0 U/50 μl
    px330(100ng/μl) 0.5μl 1ng/μl
    25 mM MgSO4 3μl 1.5 mM
    ddH2O 31.5μl
    Total 50μl
  • 12
    Perform PCR using the following cycling conditions:
    Cycle number Denature Anneal Extend
    1 95°C, 5min
    2–34 95°C, 30s 60°C, 30s 72°C, 30s
    35 72°C, 5min
  • 13

    Repeat Step 10 (the band in Step 3 should be of the expected size (120 bp; Supplementary Fig. 3) and the in vitro transcribed sgRNA should have a clear band at ~100bp (Supplementary Fig. 3).

    PAUSE POINT The samples can be stored at −80°C for up to 1 year.

Superovulation and mating TIMING ~3 d; ~2 h hands-on

  • 14

    Inject 10 females C57BL/6 (3–4 weeks old) with 5 IU PMSG through intraperitoneal (i.p.) at 2:00 p.m. on day 1.

    ? TROUBLESHOOTING

  • 15

    Approximately 47–49 h after PMSG injection (1:00–3:00 p.m. on day 3), inject the females i.p. with 5 IU hCG to induce ovulation.

  • 16

    Put each hormone-stimulated female together with Ai9 males in a 1:1 ratio in a mating cage overnight.

    ? TROUBLESHOOTING

Zygote collection and processing ~3h

  • 17

    Prepare two 35-mm dishes each containing 14 drops (30 μl for each drop) of KSOM medium covered with mineral oil for embryo culture. Transfer dishes into a 37°C incubator for at least 20 min before use. Prepare one 100-mm dish with 8 drops (200 μl for each drop) M2 medium.

  • 18

    Euthanize females by CO2 asphyxiation at 22–24 h after hCG injection.

  • 19

    Isolate oviducts and place all the oviducts into one 200 μl drop of M2 medium in the 100 mm dishes (Supplementary Fig. 4).

  • 20

    Under the stereoscopic microscope, transfer one oviduct at a time into the second 200 μl drop of M2 medium (prepared in Step 17). Tear the oviduct where it is most swollen using a 1 ml syringe attached to a 26G needle, releasing the zygote-cumulus complexes (ZCCs). Repeat this step for every oviduct until all the zygotes are isolated.

  • 21

    Add 200 μl of hyaluronidase to the ZCCs in the droplet, and then place the dish into a 37°C incubator for 3 min.

    Critical Step: No longer than 3 min.

  • 22

    Pipette the droplet up and down several times with a yellow tip until the cumulus cells are completely removed from the zygotes. Successively transfer the zygote clockwise through five wash drops of M2 medium (prepared in Step 17).

  • 23

    Finally, transfer the zygotes to one drop of KSOM medium in the pre-equilibrated 35-mm dish from Step 17 using a hand pipette, pass the zygotes through six to ten additional KSOM droplets to wash and place the dish at 37°C in a 5% CO2 incubator up to 24h until ready for injection.

Microinjection Preparation ~20min

  • 24

    Approximately 48–50 h after hCG injection, the zygotes develop to the end of the two-cell stage and the cytoplasms of the two blastomeres should be completely separated.

  • 25

    Prepare the appropriate injection mix depending on the aim of the experiment, as outlined in the table below. Cas9 or BE3 mRNA and Cre mRNA stocks from Step 8 Box 1, and Step 10 and sgRNA stock from Step 13 are both at concentrations of 500 ng/μl. Cre mRNA stock should be diluted down to 20 ng/μl with Nuclease-free water.

    Combine the following reagents at room temperature in a sterile, RNase-free eight-well PCR strip.
    Component Volume Final concentration
    Cas9/BE3 mRNA (500 ng/μl) 1μl 50 ng/μl
    sgRNA (500 ng/μl) 1μl 50 ng/μl
    Cre mRNA (20 ng/μl) 1μl 2 ng/μl
    Nuclease-free water 7μl -
    Total 10μl -
    Mix all the injection components just before use.
  • 26

    Centrifuge components at 15,000 × g for 5 min at 4 °C and and store on ice ready for microinjection.

  • 27

    Load 3μl of injection mix into the injection needle using a microloader tip. Keep the injection needle vertically for 5 minutes by sticking it to a piece of plasticine to remove small air bubbles in the needle.

  • 28

    Place a droplet of HEPES-CZB + CB on top of a 10-cm dish, and then cover it with mineral oil. Place the dish under a stereoscopic microscope.

  • 29

    Attach the loaded injection needle to the instrument holder connected to the Femtojet, lower the capillary into the medium drop on the stage such that it is positioned in the center just below the embryos and switch on the Femtojet and allow it to reach pressure.

  • 30

    Attach the holding pipette to the other side of the micromanipulator.

Injection of embryos ~1h (Supplementary Video 1)

  • 31

    Transfer 100 2-cell stage embryos into a large drop of HEPES-CZB + CB (prepared in Step 28) using a hand pipette. The number of zygotes to be moved into the microinjection drop should be determined by the skills of the injector and quality of the setup.

    Critical Step: Do not attempt to work with more zygotes than can be injected within 30 minutes.

  • 32

    Determine whether the microinjection capillary is open and unclogged by placing the tip of the microinjection capillary close to a zygote in the same horizontal plane under a continuous flow stream. If the microinjection capillary is open, a stream of DNA will move the zygote away from the tip of the microinjection capillary. If the injection pipette is still not open, tip it carefully on the holding pipette until it breaks at a larger dimension.

  • 33

    Hold an embryo using a holding pipette. Insert the injection tip into one blastomere and pause briefly halfway inside the blastomere to see the formation of a small droplet around the injection tip.

  • 34

    When all the embryos in the chamber have been injected, they should immediately be moved back into KSOM medium from Step 17, wash 6–8 times and incubated at 37°C in a 5% CO2 for 30 min. At this point, 80% of the embryos will be transferred to a foster mother oviduct for development to E14.5 (Steps 35–43), while the other 20% will be cultured in vitro to blastocyst stage for genotyping (Steps 44–57).

Reimplantation of injected embryos ~2h

  • 35

    Prepare 4 pseudopregnant foster mothers by mating estrous ICR female mice with vasectomized male mice on the same day as zygote collection.

  • 36

    On the morning of embryo transfer, identify 0.5 days post coitum (dpc) foster mothers with visible copulatory plugs.

  • 37

    Anesthetize foster mother by peritoneal injection of Avertin using a 1-ml syringe attached to a 26-gauge needle, after having weighed foster mothers to calculate the injection dose of Avertin (2.5% Avertin solution at a dose of 0.01 ml/g of body weight).

  • 38

    After 1 min, check that the mouse is fully anesthetized by lightly pinching the most medial toe. Once the mouse is unresponsive, place the anesthetized mouse under the stereomicroscope and disinfect with 75% (vol/vol) ethanol.

  • 39

    Make a small longitudinal incision (≤1 cm) parallel to the midline at the level of the last rib and slide the skin to expose the body wall. Pick up the body wall with forceps over the site of the ovary and make a small incision through the body wall. Expose the ovary, oviduct, and part of the uterus through the incision.

  • 40

    Load the glass transfer pipette by drawing KSOM medium ~1 cm up the pipette, then one small air bubble, then 18–22 embryos from Step 34 within a minimal amount of medium, and finally another small air bubble.

  • 41

    Make a small hole in the upper ampulla using a 30G needle. Insert glass transfer pipette into the hole and transfer the embryos with air bubbles.

  • 42

    Remove the transfer pipette, gently return the ovary, oviduct, and uterus back inside the body and seal the incision with absorbable sutures.

  • 43

    Keep the mice warm on a 37°C warming plate until the mouse recovers from the effects of the anesthesia.

    ? TROUBLESHOOTING

Nested PCR detection of targeted embryos ~6h

  • 44

    Incubate the remaining 20% injected embryos from Step 34 at 37°C, 5% CO2 for 3 days until blastocyst stage.

  • 45

    Wash single blastocysts 3–6 times with KSOM.

  • 46

    Transfer blastocysts directly into PCR tubes (one blastocyst per tube), and add1.5 μl embryo lysis buffer.

    Critical Step: Using stereo microscope to confirm the blastocysts are indeed put into PCR tubes.

  • 47

    Incubate at 56°C for 30 min, heat inactivate at 95°C for 10 min.

  • 48
    Prepare nested PCR amplification reactions to amplify the crude DNA solutions, using the following reaction mix.
    Component Volume Final Concentration
    Premix Taq (Ex Taq Version 2.0 plus dye) 25μl 1x
    Tyr-OF/IF (10μM) 2μl 0.4μM
    Tyr-OR/IR (10μM) 2μl 0.4μM
    Crude DNA solution 1.5μl
    Nuclease-free water Up to 50μl
  • 49
    Perform PCR using the following cycling conditions:
    Cycle number Denature Anneal Extend
    1 95°C, 5min
    2–34 95°C, 30s 60°C, 30s 72C, 30s
    35 72°C, 5min
  • 50

    Purify PCR products from Step 49, using the Universal DNA Purification Kit according to the manufacturer’s instructions.

  • 51
    Clone purified PCR products into pMD-19T using the pMD-19T cloning vector kit, according to the manufacturer’s instructions.
    Component Volume Final Concentration
    pMD-19T (50 pg/μl) 0.5 μl 2.5 μg/μl
    PCR product (50 μg/μl) 1μl 50 μg/μl
    Solution I 5μl 1x
    Nuclease-free water Up to 10μl
    Mix all the components and incubate at 56°C for 30 min.
  • 52

    Transform the above reaction mix into E. coli DH5α and cloned into E. coli (TA clone).

  • 53

    Plate all of the transformation onto a 10 cm LB agar plate with 100ug/ml of ampicillin and incubate plates at 37°C overnight.

  • 54

    Pick a single colony from an agar plate using a pipet tip and drop the pipet tip in a tube containing 5 mL LB medium with 100ug/ml of ampicillin.

  • 55

    Incubate the bacteria in a shaking incubator for 15h at 37°C, 200 rpm.

  • 56

    Extract plasmid from E. coli using the TIANprep Rapid Mini Plasmid Kit according to the manufacturer’s instructions.

  • 57

    Determine the number of clones with indels or SNVs by Sanger Sequencing. 20 TA clones are picked for Sanger sequencing, further experiments will be conducted only if the on-target editing was more than 20% (4/20 clones).

Isolation of embryonic cells and fluorescent-activated cell sorting (FACS) ~10h

  • 58

    Euthanize 14.5-d pregnant mice from Step 43 by cervical dislocation or anesthesia and separate the embryos with the placenta, then aseptically dissect mouse embryos. Wash the embryos with PBS twice briefly.

  • 59

    Place each embryo into a 100-mm petri dish and mince them into the smallest possible pieces less than 1mm.

  • 60

    Add 5 mL of pre-warmed 37°C Trypsin-EDTA (0.05%) and transfer to a 50mL centrifuge tube and incubate at 37°C for 30 minutes.

  • 61

    Add 5 ml DMEM medium and Homogenize fetal tissues by passing 30–40 times through a 1ml pipette tip.

  • 62

    Centrifuge the cell suspension at 200g for 6 min at room tempetature, and resuspend the pellet in 5 mL of DMEM medium.

  • 63

    Filter the cell suspension through a 40-μm cell strainer.

  • 64

    Centrifuge the cell suspension at 200g for 6 min at room temperature, aspirate the medium gently without disturbing the pellet and resuspend the pellet in 5 mL of DMEM medium.

  • 65

    Pass cell suspension through a 35-μm cell strainer cap into a 5-ml polystyrene round-bottom tube.

  • 66

    Sort the tdTomato+ cells and tdTomato cells into two separate tubes (~4 million cells for each) using a Moflo XDP fluorescence-activated cell sorter (Beckman Coulter), respectively. The FSC/SSC gates of the starting cell population were set in order to include all cells. Then doublet cells were excluded by SSC-H vs SSC-A. Positive and negative boundaries were defined by control progeny cells of nonedited blastomeres (see Supplementary Figure 2.

  • 67

    Extract genomic DNA from sorted cells using the DNeasy blood and tissue kit according to the manufacturer’s instructions.

Sample Quality control (QC) ~36h

  • 68

    Perform Nested PCR amplification reactions on the extracted purified genomic DNA from tdTomato+ cells and tdTomato cells, respectively, as described in Steps 48–57. Ligate the PCR products into T-Vectors, and pick 20 TA clones for Sanger sequencing. Conduct further experiments (Steps 69–78) only if less than 10% (2/20) of clones show on-target editing in tdTomato cells and more than 20% (4/20) of clones show on-target editing in tdTomato+ cells.

WGS .Timing ~3 d

  • 69

    Perform whole genome sequencing on the extracted genomic DNA from Step 67 at mean coverage of 50x by Illumina HiSeq X Ten.

Processing of raw reads ~20 h

  • 70
    Quality control raw sequencing reads by fastQC, using the following commands:
    fastqc raw/cre_neg_R1.fastq.gz raw/cre_neg_R2.fastq.gz -o fastQC/pretrim
    fastqc raw/cre_pos_R1.fastq.gz raw/cre_pos_R2.fastq.gz -o fastQC/pretrim
    

    If the quality of sequencing reads is good (Quality score per base > 20; Supplementary Fig. 5), just skip to Step 73. Otherwise, apply Step 72 to trim low quality reads and remove adapter sequences (Supplementary Fig. 5).

  • 71
    (Optional) Use Trimmomatic to trim the low quality reads and adapter sequences in the FASTQ files, using the following commands:
    java -jar trimmomatic-0.36.jar PE -threads 8 cre_neg_R1.fastq.gz cre_neg_R2.fastq.gz
    fastQC/trim/cre_neg_R1_paired.fastq.gz fastQC/trim/cre_neg_R1_unpaired.fastq.gz
    fastQC/trim/cre_neg_R2_paired.fastq.gz fastQC/trim/cre_neg_R2_unpaired.fastq.gz
    ILLUMINACLIP:TruSeq3-PE.fa:2:30:10 LEADING:5 TRAILING:5 MINLEN:70
    java -jar trimmomatic-0.36.jar PE -threads 8 cre_neg_R1.fastq.gz cre_neg_R2.fastq.gz
    fastQC/trim/cre_pos_R1_paired.fastq.gz fastQC/trim/cre_pos_R1_unpaired.fastq.gz
    fastQC/trim/cre_pos_R2_paired.fastq.gz fastQC/trim/cre_pos_R2_unpaired.fastq.gz
    ILLUMINACLIP:TruSeq3-PE.fa:2:30:10 LEADING:5 TRAILING:5 MINLEN:70
    fastqc fastQC/trim/cre_neg_R1_paired.fastq.gz fastQC/trim/cre_neg_R2_paired.fastq.gz -o fastQC/posttrim
    fastqc fastQC/trim/cre_pos_R1_paired.fastq.gz fastQC/trim/cre_pos_R2_paired.fastq.gz -o fastQC/posttrim
    
  • 72
    Construct the BWA index file of the reference genome mm10.fa, using the following command:
    bwa index mm10.fa
    
  • 73
    Align clean reads to the reference genome using BWA mem and use Picard-tools to reorder, sort, add read groups and mark duplicates of the aligned BAM files as follows:
    bwa mem -t 8 -M mm10.fa fastQC/trim/cre_neg_R1_paired.fastq.gz fastQC/trim/cre_neg_R2_paired.fastq.gz |
    samtools view -bS -o cre_neg.bam
    bwa mem -t 8 -M mm10.fa fastQC/trim/cre_pos_R1_paired.fastq.gz fastQC/trim/cre_pos_R2_paired.fastq.gz |
    samtools view -bS -o cre_pos.bam
    java -Xmx20g -jar picard.jar ReorderSam INPUT=cre_neg.bam OUTPUT=cre_neg.reorder.bam
    REFERENCE=mm10.fa
    java -Xmx20g -jar picard.jar ReorderSam INPUT=cre_pos.bam OUTPUT=cre_pos.reorder.bam
    REFERENCE=mm10.fa
    java -Xmx20g -jar picard.jar SortSam INPUT=cre_neg.reorder.bam OUTPUT=cre_neg.reorder.sort.bam
    SORT_ORDER=coordinate
    java -Xmx20g -jar picard.jar SortSam INPUT=cre_pos.reorder.bam OUTPUT=cre_pos.reorder.sort.bam
    SORT_ORDER=coordinate
    java -Xmx20g -jar picard.jar AddOrReplaceReadGroups VALIDATION_STRINGENCY=SILENT
    INPUT=cre_neg.reorder.sort.bam OUTPUT=cre_neg.reorder.sort.add.bam RGLB=WES RGPL=Illumina
    RGPU=HiSeq RGSM=cre_neg
    java -Xmx20g -jar picard.jar AddOrReplaceReadGroups VALIDATION_STRINGENCY=SILENT
    INPUT=cre_pos.reorder.sort.bam OUTPUT=cre_pos.reorder.sort.add.bam RGLB=WES RGPL=Illumina
    RGPU=HiSeq RGSM=cre_neg
    java -Xmx20g -jar picard.jar MarkDuplicates VALIDATION_STRINGENCY=SILENT
    INPUT=cre_neg.reorder.sort.add.bam OUTPUT=cre_neg.reorder.sort.add.mkdup.bam
    METRICS_FILE=cre_neg.reorder.sort.add.mkdup.metrics
    java -Xmx20g -jar picard.jar MarkDuplicates VALIDATION_STRINGENCY=SILENT
    INPUT=cre_pos.reorder.sort.add.bam OUTPUT=cre_pos.reorder.sort.add.mkdup.bam
    METRICS_FILE=cre_pos.reorder.sort.add.mkdup.metrics
    java -Xmx20g -jar picard.jar BuildBamIndex VALIDATION_STRINGENCY=SILENT
    INPUT=cre_neg.reorder.sort.add.mkdup.bam
    java -Xmx20g -jar picard.jar BuildBamIndex VALIDATION_STRINGENCY=SILENT
    INPUT=cre_pos.reorder.sort.add.mkdup.bam
    

Detection of off-target effects ~30h

  • 74
    To detect whether off-target variants specifically existed in tdTomato+ cells, apply three variant calling tools to identify SNVs and indels by comparing tdTomato+ cell with tdTomato cells from the same embryo, using the following commands:
    java -Xmx20g -jar GenomeAnalysisTK.jar -R mm10.fa -T MuTect2 -I:tumor
    cre_pos.reorder.sort.add.mkdup.bam -I:normal cre_neg.reorder.sort.add.mkdup.bam -o
    mutect/cre_pos/output.vcf
    lofreq somatic -n cre_neg.reorder.sort.add.mkdup.bam -t cre_pos.reorder.sort.add.mkdup.bam -f mm10.fa –
    threads 8 -o lofreq/cre_pos
    strelka-2.7.1.centos5_x86_64/bin/configureStrelkaSomaticWorkflow.py --normalBam
    cre_neg.reorder.sort.add.mkdup.bam --tumorBam cre_pos.reorder.sort.add.mkdup.bam --referenceFasta
    mm10.fa --runDir strelka/cre_pos
    scalpel-0.5.3/scalpel-discovery --somatic –normal cre_neg.reorder.sort.add.mkdup.bam –tumor
    cre_pos.reorder.sort.add.mkdup.bam –bed mm10.bed --window 600 --numprocs 8 --ref mm10.fa --dir
    scalpel/cre_pos
    awk ‘$7==”PASS” {print $0}’ strelka/cre_pos/results/variants/somatic.snvs.vcf >
    strelka/cre_pos/results/variants/somatic.snvs.pass.vcf
    awk ‘$7==”PASS” {print $0}’ strelka/cre_pos/results/variants/somatic.indels.vcf >
    strelka/cre_pos/results/variants/somatic.indels.pass.vcf
    awk ‘$7==”PASS” {print $0}’ mutect/cre_pos/output.vcf > mutect/cre_pos/output.pass.vcf
    awk ‘length($4)==1 && length($5)==1 {print $0}’ mutect/cre_pos/output.pass.vcf >
    mutect/cre_pos/output.pass.snv.vcf
    awk ‘length($4)>1 ∥ length($5)>1 {print $0}’ mutect/cre_pos/output.pass.vcf >
    mutect/cre_pos/output.pass.indel.vcf
    
    The SNVs identified by all three algorithms Mutect2, Strelka2 and Lofreq are considered true SNVs, and the overlap of Mutect2, Strelka2 and Scalpel is identified as true indels. This step can be performed using the following commands:
    perl filter_overlap.pl mutect/cre_pos/output.pass.snv.vcf lofreq/cre_pos/somatic_final.snvs.vcf
    strelka/cre_pos/results/variants/somatic.snvs.pass.vcf cre_pos.snv.overlap.vcf
    perl filter_overlap.pl mutect/cre_pos/output.pass.indel.vcf scalpel/cre_pos/somatic_final.indels.vcf
    strelka/cre_pos/results/variants/somatic.indels.pass.vcf cre_pos.indel.overlap.vcf
    

Validation of the off-target variants using Sanger sequencing ~2d

  • 75

    Design primers for a dozen of the detected SNVs for Sanger sequencing. The primers should have ~50% GC content.

  • 76

    Perform PCR amplification as described in Steps 48–49 and Sanger sequence the purified PCR products.

Filtration and functional annotation ~10min

  • 77
    Use Annovar as follows to annotate the identified variants from Step 75 and only use those with allele frequency more than 10% for the final results.
    annotate_variation.pl -buildver mm10 -downdb -webfrom annovar refGene mousedb/
    table_annovar.pl cre_pos.snv.overlap.vcf mousedb -buildver mm10 -out cre_pos.snv.overlap.vcf.anno -remove
    -protocol refGene -operation g -nastring . -vcfinput
    perl awk_anno.pl cre_pos.snv.overlap.vcf.anno.mm10_multianno.txt cre_pos.anno.tsv
    awk ‘$10>0.1 {print $0}’ cre_pos.anno.tsv > cre_pos.anno.0.1.tsv
    

Sequence comparison between off-target and on-target variants ~20min

  • 78
    Retrieve the adjacent 23bp sequences of the off-target variants (5bp upstream and 17bp downstream as the target base of on-target site in this study is the 6th nucleotide of the sgRNA sequence) from the marked BAM file (output from Step 74) and then blasted with the on-target sequence sgRNA.fasta (23bp; 20 bp sgRNA target sequence + 3bp PAM). Meanwhile, blast the predicted off-target sites from Cas-OFFinder51 based on sequence similarity with the on-target sequences using National Center for Biotechnology Information (NCBI) Basic Local Alignment Search Tool (BLAST) as follows:
    cat cre_pos.anno.0.1.tsv | while read line
    do
     chr=$(echo $line | cut -d” “ -f 1)
     pos=$(echo $line | cut -d” “ -f 2)
     start=`expr $pos - 5`
    end=`expr $pos + 17`
     samtools faidx mm10.fa $chr:${start}-${end} >> cre_pos.anno.0.1.tsv.fasta
    done
    makeblastdb -in sgRNA.fasta -dbtype nucl -parse_seqids
    blastn -db sgRNA.fasta -query cre_pos.anno.0.1.tsv.fasta -dust no -outfmt 0 -word_size=7 -out
    cre_pos.sgRNA.blast.out
    

    This step reveals the sequence similarity between off-target variants and on-target edits. High sequence similarities indicate the off-target variants are associated with sgRNAs, while low sequence similarities suggest the off-target effects are sgRNA-independent.

Timing

  • Steps 1–8, generation of Cas9 mRNA: ~10h

  • Steps 9–11, generation of Cre mRNA: ~10h

  • Steps 12–13, generation of sgRNA: ~8h

  • Steps 14–16, superovulation and mating: ~3d

  • Steps 17–23, zygote collection and processing: ~3h

  • Steps 24–30, microinjection preparation: ~20min

  • Steps 31–34, injection of embryos: ~1h

  • Steps 35–43, reimplantation of injected embryos: ~2h

  • Steps 44–57, nested PCR detection of targeted embryos: ~6h

  • Steps 58–67, isolation of embryonic cells and fluorescent-activated cell sorting (FACS): ~10h

  • Steps 68, sample quality control (QC): ~36h

  • Steps 69, WGS: ~3d

  • Steps 70–73, processing of raw reads: ~20h

  • Step 74, detection of off-target effects: ~30h

  • Steps 75–76, validation of the off-target variations using Sanger sequencing: ~2d

  • Step 77, filtration and functional annotation: ~10 min

  • Step 78, sequence comparison between off-target and on-target sequences ~20 min

Troubleshooting

Troubleshooting advice can be found in Table 2.

Table 2.

Troubleshooting.

Step Problem Possible reason Solution
15 Too many or two few eggs Incorrect age of female mice Select female mice with 3–4 weeks old
16 Low fertilization ratio Frequent use of male mice Mate at most twice a week for the male mice, with at least two days between
44–46 Low development rate The injected mRNA was not pure and is toxic Re-generate the mRNAs from Steps 1–16
58–66 Too low a proportion of tdTomato+ cells after FACS
  1. Gene editing tools may be toxic for the development of tdTomato+ cells

  2. The targeting gene is essential or important to the development of cells

  3. The quality of mRNA is inadequate

  1. Choose high fidelity version of gene editing tools

  2. Select non essential genes as target genes

  3. Re-prepare mRNA for injection

74 Mutect2 takes too much time to complete Mutect2 takes a lot of memory when running WGS data Split the WGS into separate chromosomes to run each time

Anticipated results

A successful application of the experimental procedure will generate two equal populations of cells with tdTomato or not (Fig. 3a). Embryos with obviously unequal proportions of tdTomato+ cells should not be sequenced for further analysis (Supplementary Fig. 6). Examples of the on-target efficiency for CRISPR/Cas9 (Cas9-Tyr-C16) and BE3 (BE3-Tyr-C-#116) editing following injection of sgRNA are shown in Fig. 3b. The bioinformatic pipeline will reveal the number of off-target variants in each embryo (Supplementary Fig. 7). The off-target mutations induced by CRISPR/Cas9 or BE3 editing were compared and BE3 induced many more off-target edits than CRISPR/Cas9 (Fig. 3c). The off-target variants can be used for downstream analysis by group comparison, mutation bias and functional enrichment16. The adjacent sequences of identified variants are compared with the on-target sites (Fig. 3d), but poor sequence similarity is observed (mean Bit-score = 10.4 for Cas9-Tyr-C; mean Bit-score = 12.2 for BE3-Tyr-C-#1). By contrast, potential off-target sequences predicted by Cas-OFFinder show high similarity with the on-target sequence (mean Bit-score = 29.4; Fig. 3d). These results suggest that the off-target variants identified by GOTI in CRISPR/Cas9 or BE3 editing are sgRNA-independent.

Fig. 3. Anticipated results from GOTI.

Fig. 3.

a, The separation of tdTomato+ and tdTomato cells. FACS analysis for E14.5 embryo of Cas9-Tyr-C16. b, On-target efficiency for tdTomato+ and tdTomato cells based on WGS for Cas9-Tyr-C16 and BE3-Tyr-C-#116-treated embryos. The target site was edited with high efficiency in tdTomato+ cells, but not in tdTomato cells. The deletions induced by Cas9-Tyr-C and base substitutions in BE3-Tyr-C-#1 were highlighted in red rectangles. Dark lines represent deletions in the region and green rectangles represent substitutions. c, The number of off-target SNVs detected in Cas9-Tyr-C and BE3-Tyr-C-#1-treated embryos. BE3-Tyr-C-#1 induced much higher number of off-target variants than Cas9-Tyr-C-#1. d, The sequence similarity (Bit-score) between on- and off-target sequences identified by GOTI in Cas9-Tyr-C16 and BE3-Tyr-C-#116-treated embryos or predicted by Cas-OFFinder. The off-target SNVs are sgRNA independent. n = 1809 for Cas-OFFinder, n = 18 for Cas9-Tyr-C and n = 247 for BE3-Tyr-C-#1. Box-and-whisker plots: center line indicates median, the bottom and top lines of the box represents the first quartile and third quartile of the values, respectively. The bottom and top lines represent the minimum and maximum value. P values are calculated with two-sided Wilcoxon rank sum test.

Supplementary Material

Supplementary material
Supplementary video

Supplementary Video Gene editing system are introduced into one blastomere of two-cell mouse embryos by microinjection.

Download video file (1.9MB, mp4)

Acknowledgements

We thank FACS facility in ION. The work was supported by R&D Program of China (2018YFC2000100 and 2017YFC1001302 to HY, 2017YFC0908405 to WW), CAS Strategic Priority Research Program (XDB32060000), National Natural Science Foundation of China (31871502, 31522037), Shanghai Municipal Science and Technology Major Project (2018SHZDZX05), Shanghai City Committee of science and technology project (18411953700, 18JC1410100) and NIH P01 Center grant (P01HG00020527 to L.M.S.).

Footnotes

Competing interests

L.M.S. has consulted for companies on CRISPR editing. Other authors declare no competing financial interests.

Data availability

The sequencing data were deposited in NCBI Sequence Read Archive (SRA) under project accession SRP119022 and http://www.biosino.org/node/project/detail/OEP000195.

Code availability

The GOTI-seq pipeline is publicly available in GitHub at https://github.com/sydaileen/GOTI-seq.

Supplementary Information

Supplementary Video Gene editing system are introduced into one blastomere of two-cell mouse embryos by microinjection.

References

  • 1.Knott GJ & Doudna JA CRISPR-Cas guides the future of genetic engineering. Science 361, 866–869, doi: 10.1126/science.aat5011 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.de la Fuente-Nunez C & Lu TK CRISPR-Cas9 technology: applications in genome engineering, development of sequence-specific antimicrobials, and future prospects. Integr Biol (Camb) 9, 109–122, doi: 10.1039/c6ib00140h (2017). [DOI] [PubMed] [Google Scholar]
  • 3.Hsu PD, Lander ES & Zhang F Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278, doi: 10.1016/j.cell.2014.05.010 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Doudna JA & Charpentier E Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096, doi: 10.1126/science.1258096 (2014). [DOI] [PubMed] [Google Scholar]
  • 5.Cox DB, Platt RJ & Zhang F Therapeutic genome editing: prospects and challenges. Nat Med 21, 121–131, doi: 10.1038/nm.3793 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Musunuru K The Hope and Hype of CRISPR-Cas9 Genome Editing: A Review. JAMA Cardiol 2, 914–919, doi: 10.1001/jamacardio.2017.1713 (2017). [DOI] [PubMed] [Google Scholar]
  • 7.Jiang F & Doudna JA CRISPR-Cas9 Structures and Mechanisms. Annu Rev Biophys 46, 505–529, doi: 10.1146/annurev-biophys-062215-010822 (2017). [DOI] [PubMed] [Google Scholar]
  • 8.Wyman C & Kanaar R DNA double-strand break repair: all’s well that ends well. Annu Rev Genet 40, 363–383, doi: 10.1146/annurev.genet.40.110405.090451 (2006). [DOI] [PubMed] [Google Scholar]
  • 9.Paquet D et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533, 125–129, doi: 10.1038/nature17664 (2016). [DOI] [PubMed] [Google Scholar]
  • 10.Goodwin S, McPherson JD & McCombie WR Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet 17, 333–351, doi: 10.1038/nrg.2016.49 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Komor AC, Kim YB, Packer MS, Zuris JA & Liu DR Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424, doi: 10.1038/nature17946 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nishida K et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, doi: 10.1126/science.aaf8729 (2016). [DOI] [PubMed] [Google Scholar]
  • 13.Gaudelli NM et al. Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature 551, 464–471, doi: 10.1038/nature24644 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rees HA & Liu DR Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet, doi: 10.1038/s41576-018-0059-1 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pattanayak V et al. High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nat Biotechnol 31, 839–843, doi: 10.1038/nbt.2673 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zuo E et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289–292, doi: 10.1126/science.aav9973 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jin S et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 364, 292–295, doi: 10.1126/science.aaw7166 (2019). [DOI] [PubMed] [Google Scholar]
  • 18.Grunewald J et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433–437, doi: 10.1038/s41586-019-1161-z (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zhou C et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature, doi: 10.1038/s41586-019-1314-0 (2019). [DOI] [PubMed] [Google Scholar]
  • 20.Cameron P et al. Mapping the genomic landscape of CRISPR-Cas9 cleavage. Nat Methods 14, 600–606, doi: 10.1038/nmeth.4284 (2017). [DOI] [PubMed] [Google Scholar]
  • 21.Frock RL et al. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat Biotechnol 33, 179–186, doi: 10.1038/nbt.3101 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Tsai SQ et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat Biotechnol 33, 187–197, doi: 10.1038/nbt.3117 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tsai SQ et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat Methods 14, 607–614, doi: 10.1038/nmeth.4278 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jin S et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science , published online (2019). [DOI] [PubMed] [Google Scholar]
  • 25.Yang H et al. Base editing generates substantial off-target single nucleotide variants. doi: 10.1101/480145 %J bioRxiv (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Madisen L et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci 13, 133–140, doi: 10.1038/nn.2467 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wang L et al. CRISPR-Cas9-mediated genome editing in one blastomere of two-cell embryos reveals a novel Tet3 function in regulating neocortical development. Cell Res 27, 815–829, doi: 10.1038/cr.2017.58 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.O’Rawe J et al. Low concordance of multiple variant-calling pipelines: practical implications for exome and genome sequencing. Genome Med 5, 28, doi: 10.1186/gm432 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bao R et al. Review of current methods, applications, and data management for the bioinformatics analysis of whole exome sequencing. Cancer Inform 13, 67–82, doi: 10.4137/CIN.S13779 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Cameron DL, Di Stefano L & Papenfuss AT Comprehensive evaluation and characterisation of short read general-purpose structural variant calling software. Nat Commun 10, 3240, doi: 10.1038/s41467-019-11146-4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Field MA et al. Recurrent miscalling of missense variation from short-read genome sequence data. BMC Genomics 20, 546, doi: 10.1186/s12864-019-5863-2 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cibulskis K et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 31, 213–219, doi: 10.1038/nbt.2514 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kim S et al. Strelka2: fast and accurate calling of germline and somatic variants. Nat Methods 15, 591–594, doi: 10.1038/s41592-018-0051-x (2018). [DOI] [PubMed] [Google Scholar]
  • 34.Wilm A et al. LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets. Nucleic Acids Res 40, 11189–11201, doi: 10.1093/nar/gks918 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Narzisi G et al. Accurate de novo and transmitted indel detection in exome-capture data using microassembly. Nat Methods 11, 1033–1036, doi: 10.1038/nmeth.3069 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Schonig K et al. Conditional gene expression systems in the transgenic rat brain. BMC Biol 10, 77, doi: 10.1186/1741-7007-10-77 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bryda EC et al. A novel conditional ZsGreen-expressing transgenic reporter rat strain for validating Cre recombinase expression. Sci Rep 9, 13330, doi: 10.1038/s41598-019-49783-w (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang K et al. Cre-dependent Cas9-expressing pigs enable efficient in vivo genome editing. Genome Res 27, 2061–2071, doi: 10.1101/gr.222521.117 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Li L et al. Production of a reporter transgenic pig for monitoring Cre recombinase activity. Biochem Biophys Res Commun 382, 232–235, doi: 10.1016/j.bbrc.2009.02.146 (2009). [DOI] [PubMed] [Google Scholar]
  • 40.Gabriel R et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat Biotechnol 29, 816–823, doi: 10.1038/nbt.1948 (2011). [DOI] [PubMed] [Google Scholar]
  • 41.Liang P et al. Genome-wide profiling of adenine base editor specificity by EndoV-seq. Nat Commun 10, 67, doi: 10.1038/s41467-018-07988-z (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kim D et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat Methods 12, 237–243, 231 p following 243, doi: 10.1038/nmeth.3284 (2015). [DOI] [PubMed] [Google Scholar]
  • 43.Kim D et al. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat Biotechnol 35, 475–480, doi: 10.1038/nbt.3852 (2017). [DOI] [PubMed] [Google Scholar]
  • 44.Wienert B et al. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq. Science 364, 286–289, doi: 10.1126/science.aav9023 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Anderson KR et al. CRISPR off-target analysis in genetically engineered rats and mice. Nat Methods 15, 512–514, doi: 10.1038/s41592-018-0011-5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Iyer V et al. Off-target mutations are rare in Cas9-modified mice. Nat Methods 12, 479, doi: 10.1038/nmeth.3408 (2015). [DOI] [PubMed] [Google Scholar]
  • 47.Smith C et al. Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. Cell Stem Cell 15, 12–13, doi: 10.1016/j.stem.2014.06.011 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tang X et al. A large-scale whole-genome sequencing analysis reveals highly specific genome editing by both Cas9 and Cpf1 (Cas12a) nucleases in rice. Genome Biol 19, 84, doi: 10.1186/s13059-018-1458-5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Iyer V et al. No unexpected CRISPR-Cas9 off-target activity revealed by trio sequencing of gene-edited mice. PLoS Genet 14, e1007503, doi: 10.1371/journal.pgen.1007503 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Willi M, Smith HE, Wang C, Liu C & Hennighausen L Mutation frequency is not increased in CRISPR-Cas9-edited mice. Nat Methods 15, 756–758, doi: 10.1038/s41592-018-0148-2 (2018). [DOI] [PubMed] [Google Scholar]
  • 51.Bae S, Park J & Kim JS Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 30, 1473–1475, doi: 10.1093/bioinformatics/btu048 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary material
Supplementary video

Supplementary Video Gene editing system are introduced into one blastomere of two-cell mouse embryos by microinjection.

Download video file (1.9MB, mp4)

RESOURCES