Here, we present a modified method for cryopreservation of one-cell embryos as well as a protocol that couples the use of freeze-thawed embryos and electroporation for the efficient generation of genetically modified mice.
The use of genetically modified (GM) mice has become crucial for understanding gene function and deciphering the underlying mechanisms of human diseases. The CRISPR/Cas9 system allows researchers to modify the genome with unprecedented efficiency, fidelity, and simplicity. Harnessing this technology, researchers are seeking a rapid, efficient, and easy protocol for generating GM mice. Here we introduce an improved method for cryopreservation of one-cell embryos that leads to a higher developmental rate of the freeze-thawed embryos. By combining it with optimized electroporation conditions, this protocol allows for the generation of knockout and knock-in mice with high efficiency and low mosaic rates within a short time. Furthermore, we show a step-by-step explanation of our optimized protocol, covering CRISPR reagent preparation, in vitro fertilization, cryopreservation and thawing of one-cell embryos, electroporation of CRISPR reagents, mouse generation, and genotyping of the founders. Using this protocol, researchers should be able to prepare GM mice with unparalleled ease, speed, and efficiency.
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system is a scientific breakthrough that provides unprecedented targeted modification in the genome1. The CRISPR/Cas9 system is comprised of Cas9 protein and guide RNA (gRNA) with two molecular components: a target-specific CRISPR RNA (crRNA) and a trans-activating CRISPR RNA (tracrRNA)2 . A gRNA directs the Cas9 protein to the specific locus in the genome, 20 nucleotides complementary to crRNA, and adjacent to the protospacer adjacent motif (PAM). The Cas9 protein binds to the target sequence and induces double-strand breaks (DSBs) that are repaired by either error-prone nonhomologous end joining (NHEJ) or high fidelity homology-directed repair (HDR)3,4,5. The NHEJ leads to insertions or/and deletions (indels), and hence to gene loss of function when a coding sequence is targeted. The HDR leads to precise genome editing in the presence of a repair template containing homology sequences3,4,5. The NHEJ and HDR have been harnessed to generate knockout and knock-in mice, respectively.
While the CRISPR/Cas9 system has markedly accelerated the generation of GM mice with outstanding efficacy and fidelity, scientists who apply these methods often encounter technical challenges. First, conventional protocols require microinjection for introducing the CRISPR editing tools into the pronucleus of fertilized eggs6,7. This technique is time-consuming and usually requires extensive training. Thus, several groups replaced microinjection with electroporation8,9,10,11,12,13. However, in the early electroporation protocols fresh embryos were used for electroporation. This caused another problem, because preparing fresh embryos before each experiment is difficult14.
Recently we and others have combined the use of freeze-thawed embryos and electroporation for genome editing, which facilitates the generation of GM mice15,16. This protocol enables researchers without advanced embryo manipulation skills to rapidly generate animal models of human diseases with high efficiency. The protocol also significantly reduces practical challenges in generating GM mice, such as genetic heterogeneity in the founders16. To overcome mosaicism, we perform the electroporation of CRISPR reagents within 1 h after embryo thawing to ensure that editing occurs before the first replication of the genome. Another improvement includes the use of Cas9 protein instead of Cas9 mRNA to reduce undesirable mosaicism17. Furthermore, we developed an optimal method for one-cell embryo cryopreservation that increases the developmental rate to the two-cell stage16: use of fetal bovine serum (FBS) dramatically improves the survival of freeze-thawed oocytes after fertilization, perhaps by the same mechanism that makes freeze-thawed unfertilized oocytes more resilient18.
Here we present a comprehensive protocol for the generation of GM mice using freeze-thawed embryos, including the modified method for cryopreservation of one-cell C57BL/6J embryos. It includes 1) gRNA design, CRISPR reagent preparation and assembly; 2) IVF, cryopreservation, and thawing of one-cell embryos; 3) Electroporation of CRISPR reagents into freeze-thawed embryos; 4) Embryo transfer into the oviduct of pseudopregnant female mice; and 5) Genotyping and sequence analysis of the F0 founder animals.
All animal care and procedures performed in this study were undertaken according to the rules and regulations of the Guide for the Care and Use of Laboratory Animals. The experimental protocol was approved by the Animal Care Committee of Laboratory Animals of University of Toyama, University of Tokyo, Jichi University, and Max Planck Florida Institute for Neuroscience. Information about all reagents is showed in the Table of Materials.
1. CRISPR reagents design
2. In vitro fertilization, embryo cryopreservation, and freeze-thawing
3. Assembly and electroporation of CRISPR reagents
NOTE: For electroporation, we used a platinum plate electrode (height: 0.5 mm, length: 10 mm, width: 3 mm, gap: 1 mm) and a one-step type electroporator that were described previously8 (Table of Materials). A two-step type electroporator can be used too (Table of Materials).
4. Embryo transfer
5. Genotyping and sequence analysis
Our modified method for cryopreservation of one-cell embryos, including incubation in HTF containing 20% FBS for 10 min followed by cryopreservation in 1 M DMSO and DAP213 solution, improved the developmental rate of the freeze-thawed embryos into the two-cell stage (Figure 1, p = 0.009, Student’s t-test).The freeze-thawed embryos were used for the production of GM mice and electroporation conditions were optimized: five repeats of 25 V with 3 ms pulses and 97 ms intervals using an electroporator as described in the Protocol section. The applicability of the protocol was checked by generation of albino Tyrosinase gene (Tyr) knockout mice (Figure 2). To do so, gRNA targeting exon 2 was designed (Figure 2A) and CRISPR reagents were prepared as described in the Protocol section. The editing tools were electroporated within 1 h of embryo thawing to ensure genome editing occurred before the first replication of the genome and thus prevented mosaicism in the founders (Figure 2B). All the generated mice were albino, and only one mouse was mosaic, with a coat with white and black patches (Figure 2C). All the albino mice harboring two different mutant alleles (heterozygous mutation) except one mouse harboring only one allele (homozygous mutation) are shown in Figure 2E. It can be concluded that our method can provide an editing efficiency of near 100% with a low mosaic rate as confirmed by sequencing analysis as well as coat color (Figure 2C–E).
Next, the reproducibility of the protocol was checked by the generation of several lines of knockout and knock-in mice. As shown in Table 1, the protocol can generate knockout and knock-in mice with high efficiency and low mosaic rates. Most of the generated F0 mice were mated with wild type C57BL/6J mice to confirm the germline transmission of mutant alleles to the F1 generations. As anticipated, all the F1 offspring of the homozygous F0 mice were heterozygous, containing one mutant allele and one wild type allele. No disparate mutations were observed between genotyping of the founders and their generations. The described protocol generated GM mice within a short time (i.e., ~4 weeks) as shown in the workflow of the protocol in Figure 3.
Figure 1: Fetal bovine serum (FBS) improved the two-cell stage developmental rate of the freeze-thawed embryos. The number of FBS+ embryos was 286 (the experiment was repeated 3x), and the number of FBS- embryos was 272 (the experiment was repeated 3x). Data are presented as mean ± SEM. This figure is reproduced from Darwish M et al.16 with permission. Please click here to view a larger version of this figure.
Figure 2: Generation of Tyr knockout mice with high efficiency and low mosaic rate. This figure is modified from Darwish M et al.16 with permission. (A) Schematic illustration showing the design of gRNA. The PAM sequence is shown in red. (B) Illustration showing the timing of electroporation for the freeze-thawed embryos, the yellow symbol represents electroporation time. (C) Representative images of the generated Tyr knockout mice. (D) Sequence analysis of different alleles of the Tyr knockout mice. The target sequence is labeled in red and dashes indicate the deletion of nucleotides in the mutant alleles. WT = wild type M = mutant allele. (E) A representative part of the chromatogram of the albino mouse containing the M1 allele is shown in D. Please click here to view a larger version of this figure.
Figure 3: Workflow of the protocol for the generation of GM mice. The first step was the preparation of cryopreserved embryos. Female C57BL/6J mice were superovulated, first by PMSG injection, then 48 h later by hCG injection. COCs were collected 16 h later and subjected to IVF with spermatozoa collected from male C57BL/6J mice. Fertilized oocytes were cryopreserved and stored in liquid nitrogen until needed. The second step included thawing the embryos and electroporation. Cryopreserved embryos were thawed and CRISPR reagents (i.e., tracrRNA, crRNA, and Cas9 protein) were assembled and electroporated within 1 h after thawing the embryos. The third step included embryo transfer and the birth of mice. The day after electroporation, two-cell embryos were transferred to the oviduct of pseudopregnant female mice to generate genetically modified mice that were later genotyped using Sanger sequencing to confirm the editing efficiency. The time and effort required for preparing fertilized oocytes before every experiment (step 1) can be shortened if a large number of cryopreserved embryos is prepared in advance. This figure is adapted from Darwish M et al.16 Please click here to view a larger version of this figure.
Mutant mice | Electroporated embryos | 2-cell embryos | Transferred embryos | Pups (%) | Mutant mice(%) | No of mosaic mice |
Tyr KO | 124 | 84 | 48 | 12 (25) | 100 | 1 |
Glrb KO | 151 | 94 | 40 | 6 (15) | 100 | 0 |
Slc39a6 KO | 48 | 25 | 25 | 5 (20) | 100 | 0 |
Bag3 KI | 233 | 84 | 20 | 3 (15) | 50 | 1 |
Camk2a KI | 124 | 70 | 70 | 14 (20) | 64.3 | 0 |
Table 1: Production of several lines of GM mice using freeze-thawed embryos of C57BL/6J background.
Supplementary Table 1. Please click here to download this table.
The described protocol allows for the generation of GM mice with high efficiency and low mosaic rates (Table 1). It enables researchers without advanced embryo manipulation skills to create mutant mice easily because it takes advantage of the latest and most useful advances in both reproductive engineering and genome editing technologies: CRISPR/Cas9 ribonucleoprotein (RNP) and electroporation into freeze-thawed embryos. These advances facilitated and expedited the generation of the GM mice. As described in Figure 3, it takes ~4 weeks to generate the GM mice. Compared to other protocols using similar approaches26, our method is superior in terms of the efficiency, birth rate, and mosaicism.
The incubation of one-cell embryos in FBS before the cryopreservation is critical for improving the developmental rate of the embryos. The electroporation conditions of the cryopreserved embryos, described in the Protocol, allow a compromise between the editing efficiency of CRISPR reagents and the survival of the embryos. Indeed, harsher or milder conditions may affect the developmental rate of the embryos and the editing efficiency, respectively16. The timing of the electroporation (Figure 2B) is critical to overcome the mosaicism in founders and ensure that genome modification occurs in the presence of only two alleles. This is consistent with previous reports showing that early-stage electroporation could produce non-mosaic mutants17. In addition, the use of RNP instead of gRNA/mRNA decreases mosaic rate and improves editing efficiency17.
The described protocol features several advantages. First, it generates the mutant mice within a short time (~4 weeks). Second, it is a highly efficient and robust protocol; several lines of knockout (KO) and knock-in (KI) mice were generated with high mutation rates (KO = 100%, KI = 50 to 64.3%). Third, it is convenient and cost-effective. The use of complete synthetic crRNA, tracrRNA, ssODN, and Cas9 protein eliminates the need for tedious preparation of the Cas9 vectors and in vitro transcription. Instead, it allows researchers to simply use commercial reagents and standard equipment. Fourth, it does not use microinjection, which requires high technical skills. Fifth, it reduces the mosaicism in founders, and thus leads to more efficient germline transmission of the edited alleles, overcoming the complicated genotypic analysis of the mosaic mice. Finally, this protocol is efficient with the C57BL/6J inbred strain, which avoids the use of F1 hybrids, and thus the need to perform numerous backcrosses to get rid of genetic complexity. The genotyping of F1 generations confirmed accurate germline transmission. However, it is not recommended to study the phenotypes of the F0 mice due to allele complexity and the misleading prediction of genotyping of subsequent generation extrapolated from the genotyping of F0 founders14,27.
It is of prime importance to harness the use of the freeze-thawed embryos in the genome editing of nonhuman primates and large animals such as pigs and sheep, which are not always readily available. The methods of embryo freezing differ greatly depending on the animal species. Therefore, we think our cryopreservation protocol might be only applicable to mice. On the other hand, our protocol is potentially applicable to use engineering nucleases other than Cas9 for the generation of GM mice, because electroporation has been previously reported to introduce other nucleases such as Cas12a into the embryo efficiently28,29.
One limitation of the protocol is the precise integration of a long transgene into the genome, which is considered difficult in electroporation-based protocols. However, one potential solution was reported: the integration of transgenes up 4.9 Kb in the mouse genome was successfully performed by combining electroporation with an adeno-associated virus (AAV)-mediated HDR donor delivery system30, confirming an earlier publication31. Also, the potential occurrence of off-target side effects is a major concern with the use of CRISPR/Cas9 technology. We have not performed whole-genome sequencing of the edited mice to exclude the potential off-target effect. However, we have used CRISPR tools that have been reported to reduce the off-target effect, such as RNP32. Moreover, we used a high-fidelity Cas9 variant that significantly reduces off-target editing without sacrificing on-target performance33. Also, we designed gRNAs using CRISPRdirect software (https://crispr.dbcls.jp), which should result in target sequences with minimized off-target sites19. To confirm the genotype-phenotype causality and alleviate the concerns about off-target effects, researchers should generate several edited mice of the same genotype using different gRNAs or perform backcrossing of the generated mice for several generations.
The knockout mice generated using the NHEJ mechanism and frameshift mutations have been widely used and show relevant phenotypes. However, truncated residual protein may exist due to either translation reinitiation or skipping of the edited exon34,35. Therefore, to precisely interpret the phenotypes, characterization of residual protein function or expression is necessary. The generation of complete gene knockout mice using more than one gRNA would be a good alternative. However, particular attention should be paid to confirm that the gene does not contain intronic regions transcribed to noncoding RNAs, which might have regulatory functions and hence complicate the phenotypic analysis.
In this study, we show a simple protocol by which many researchers can generate genetically modified mice in 4 weeks or less. By combining the use of freeze-thawed embryos and electroporation, we render the preparation of mouse models of human diseases easy, quick, and efficient.
The authors have nothing to disclose.
We wish to thank Hitomi Sawada and Elizabeth Garcia for animal care. This work was supported by KAKENHI (15K20134, 17K11222, 16H06276 and 16K01946) and Hokugin Research Grant (to H.N.), and Jichi Medical University Young Investigator Award (to H.U.). The Otsuka Toshimi Scholarship Foundation supported M.D.
0.25 M Sucrose | ARK Resource Co., Ltd. (Kumamoto, Japan) | SUCROSE | |
1 M DMSO | ARK Resource Co., Ltd. (Kumamoto, Japan) | 1M DMSO | |
Butorphanol | Meiji Seika Pharma Co., Ltd. (Tokyo, Japan) | Vetorphale 5mg | |
Cas9 protein: Alt-R® S.p. HiFi Cas9 Nuclease 3NLS | Integrated DNA Technologies, Inc. (Coralville, IA) | 1081060 | |
C57BL/6J mice | Japan SLC (Hamamatsu, Japan) | N/A | |
DAP213 | ARK Resource Co., Ltd. (Kumamoto, Japan) | DAP213 | |
FBS | Sigma-Aldrich, Inc. (St. Louis, MO) | ES-009-C | |
hCG | MOCHIDA PHARMACEUTICAL CO., LTD (Tokyo, Japan) | HCG Mochida 3000 | |
HTF | ARK Resource Co., Ltd. (Kumamoto, Japan) | HTF | |
ICR mice | Japan SLC (Hamamatsu, Japan) | N/A | |
Isoflurane | Petterson Vet Supply, Inc. (Greeley, CO) | 07-893-1389 | |
KSOM | ARK Resource Co., Ltd. (Kumamoto, Japan) | KSOM | |
LN2 Tank | Chart Industries (Ball Ground, GA) | XC 34/18 | |
M2 | ARK Resource Co., Ltd. (Kumamoto, Japan) | M2 | |
Medetomidine | Nippon Zenyaku Kogyo Co.,Ltd. (Koriyama, Japan) | 1124401A1060 | |
Microscope | Nikon Co. (Tokyo, Japan) | SMZ745T | |
Midazolam | Sandoz K.K. (Tokyo, Japan) | 1124401A1060 | |
Nuclease free buffer | Integrated DNA Technologies, Inc. (Coralville, IA) | 1072570 | |
Nucleospin DNA extraction kit | Takara Bio Inc (Kusatsu, Japan) | 740952 .5 | |
One-hole slide glass | Matsunami Glass Ind., Ltd. (Kishiwada, Japan) | S339929 | |
One-step type Electroporator | BEX Co., Ltd. (Tokyo, Japan) | CUY21EDIT II | |
Paraffin Liquid | NACALAI TESQUE Inc. (Kyoto, Japan) | SP 26137-85 | |
Platinum plate electrode | BEX Co., Ltd. (Tokyo, Japan) | LF501PT1-10, GE-101 | |
PMSG | ASKA Animal Health Co., Ltd (Tokyo, Japan) | SEROTROPIN 1000 | |
Povidone iodide | Professional Disposables International, Inc. (Orangeburg, NY) | C12400 | |
Reduced-Serum Minimal Essential Medium: OptiMEM I | Sigma-Aldrich, Inc. (St. Louis, MO) | 22600134 | |
Two-step type Electroporator | Nepa Gene Co., Ltd. (Ichikawa, Japan) | NEPA21 |