1. Introduction
The CRISPR/Cas9 gene editing tool allows for precise and efficient fragment insertion into the genome [
1]. In this system, the RNA-guided Cas9 nuclease causes double-strand breaks (DSBs), which can be repaired by non-homologous end joining (NHEJ), resulting in random small deletions or insertions (indels) or homology-directed repair (HDR), enabling the introduction of foreign genes carried by donor DNA [
2]. CRISPR/Cas9, followed by HDR, provides a versatile method for generating transgenic animals when combined with somatic cell nuclear transfer (SCNT) and embryo transfer [
3,
4,
5,
6,
7,
8,
9,
10]. However, the limited efficiency of HDR, which is restricted to the S and G2 phases of the cell cycle, remains a significant obstacle [
11]. The higher cost and technological complexity of SCNT have also increased the difficulty of the production of transgenic animals. Therefore, we expect to find a method of using microinjection to insert large fragments of exogenous genes into
goat embryos.
Prime editing (PE), a novel approach derived from the CRISPR/Cas9 system, involves a nickase Cas9 (H840A), a Moloney murine leukemia virus reverse transcriptase (M-MLV RT), and a pegRNA containing a spacer sequence, a scaffold, a reverse transcriptase template (RTT), and a prime binding site (PBS) sequence [
12]. PE enables base substitutions, small insertions, and deletions without the need for DSBs or donor DNA [
13]. Compared to CRISPR/Cas9-HDR and base editing (BE), PE has lower off-target effects, increased safety, and enhanced flexibility while being unaffected by the cell cycle stage [
14]. Large fragments of foreign genes can be integrated into the attB sequence inserted into the embryo genome by the twinPE system by site-specific recombination and can meet the needs of the production of transgenic animals. However, the efficiency of PE is still low. Consequently, provided that its efficiency can be improved, PE has the potential to achieve greater efficacy for precise gene editing by embryo microinjection.
Previously, multiple approaches have aimed at improving PE efficiency [
15,
16,
17,
18,
19,
20,
21]. In this study, we improved the prime editor using the
HOXB13-5′ UTR RNA motif and the integrated twinPE plasmid and demonstrated the incorporation of base substitutions, small insertions, and deletions, as well as the attB sequence at various target loci in HEK293T cells. Additionally, the attB sequence was introduced into the
Rosa26 and
CCR5 gene loci in
goat cells and embryos with the aim of enabling the insertion of foreign genes into this attB site via site-specific recombination (SSR).
3. Discussion
Prime editing enables high-efficiency deletion, substitution, or insertion of larger DNA sequences at targeted endogenous genomic sites in human cells. It provides more editing types than base editing (BE) and has similar editing precision and flexibility compared to CRISPR/Cas9 but with very low levels of off-target activities. However, considering the wide potential of the PE system for livestock gene breeding, it is necessary to improve its editing efficiency.
To address this issue, we developed two strategies to optimize the prime editor and analyzed its efficiency by inserting an attB sequence in the goat genome at target sites previously screened by the SSA reporter system. Of note, any exogenous gene can be integrated into this sequence by site-specific recombination.
In the first strategy, we improved the PE3 and twinPE system by incorporating RNA motif to the 3′ terminal of pegRNA and, as such, developed an integrated twinPE system. In fact, the RNA motif derived from the
HOXB13-5′ UTR forms a stem-ring structure, which was previously shown to protect the RNA’s degradation [
23]. Meanwhile, the spacer sequence and PBS sequence of pegRNA have complementary sequences. The pairing of the complementary stretches destroys the pegRNA single-strand structure and, as such, prevents complementary pairing with DNA. Importantly, the RNA motif maintains the structural independence of these two fragments.
We screened RNAmotif1 and RNAmotif2 from HOXB13-5′ UTR and added these sequences to the 3′ terminal of pegRNA. Next, we evidenced that RNAmotif2-pegRNA increased editing efficiency in HEK293T by 1.35-fold compared to WT pegRNA. Additionally, we evidenced that RNAmotif2-pegRNA provides notable enhancements over WT pegRNA at most target sites. For example, RNAmotif2-pegRNA-mediated FANCF 261 (6 bp del) achieved a 3.58-fold increased efficiency compared to WT pegRNA.
In the second strategy, we constructed all twinPE components—PE protein, pegRNA1, and pegRNA2—on the same plasmid. This plasmid provided an integrated twinPE system and included an EGFP detection marker for transfected cells. As such, this construct improved the transfection probability of the twinPE system into the same cell, with only one plasmid bearing all required components, and improved the detection efficiency of prime editing without the need for positive cell enrichment. The results indicated that the integrated twinPE editing efficiency was significantly improved and achieved 2.19-fold at FANCF 456–591 and 2.11-fold at CCR5 3027–3126. Based on these results, we speculated that integrated twinPE might have higher efficiency in hard-to-transfect cell lines.
To verify the effectiveness of the optimized PE system in integrating attB sequences into the goat genome, we used the SSA reporter system to select six pairs of target sites with higher cleavage efficiency of pegRNA-Cas9 and applied the enhanced RNAmotif2-integrated-twinPE system to insert an attB sequence into the Rosa26 and CCR5 gene loci of goat fibroblasts. In total, three out of six target sites could be edited by twinPE, while another three sites were not modified. In fact, an integration efficiency of 14.48% at Rosa26–80–183 demonstrated that the PE system works in goat cells with editing efficiencies that are directly related to the target site. Based on these results obtained in goat fibroblasts, we then inserted an attB sequence into goat parthenogenetic activation embryos genome following microinjection of the twinPE mRNA. With a concentration of 200 ng/µL PE2 mRNA, 50 ng/µL pegRNA1, and 50 ng/µL pegRNA2, editing was detected in 5 out of 42 embryos (11.9%) whereas with a concentration of 400 ng/µL PE2 mRNA, 100 ng/µL pegRNA1, and 100 ng/µL pegRNA2, editing could be detected in 4 out of 49 embryos, (6.8%). These results demonstrated that RNAmotif2-twinPE can mediate insertion in goat embryos. We chose parthenogenetically-activated goat embryos, as they are easily obtained and can indicate the impact of editing on embryonic development efficiency, similar to in vitro fertilization embryos and in vivo embryos. However, the limitation of parthenogenetically-activated goat embryos is that they cannot develop into a complete individual. Hence, in our future studies, we will insert attB sequences into the genomes of goats in vivo embryos by PE system and evaluate the developmental ability of transgenic embryos through embryo transfer. Of note, this must be based on the premise that the efficiency of prime editing can be significantly improved.
Finally, we verified whether the binding of twinPE and SSR can mediate the insertion of large exogenous genes into designated genomic loci. We decided to insert EGFP and Puro into FANCF and CCR5 as this choice would enable direct observation and drug screening, while the length of this exogenous gene can meet the needs of transgenic animal production. We have proven that integrated twinPE with the SSR system can effectively mediate the insertion of exogenous genes in HEK293T cells. To evaluate the existence of the off-target integration at pseudo-attB sites, we compared sequences in the human genome that are similar to our attB sequence. The comparison results indicated that there are no pseudo-attB sites in the human genome similar to our sequence, indicating that our attB sequence is, in fact, highly specific. We subsequently conducted the same experiment in goat cells, but we could not select positive clones after screening. This may be due to the low transfection efficiency of primary cells and the low editing efficiency of prime editing in goat cells.
Overall, this study established a significantly improved prime editing system and evidenced its capacity to insert an attB sequence in the goat genome. This represents an important addition to the prime editing toolbox for future development and applications.
4. Materials and Methods
4.1. Plasmid Construction
The plasmids pCMV-PE2 (#132775), pGL3-U6-sgRNA-EGFP (#107721), and pSSA-1-3 (#35091) were obtained from Addgene. For expression of the integrated twinPE, each part of the plasmid backbone was amplified from pCMV-PE2 and pGL3-U6-sgRNA-EGFP using PrimeSTAR DNA Polymerase (Takara, Dalian, China) and linked by Gibson Assembly. For expression of the WT-PE2, the plasmid backbone was cut by NotІ-HF®v2 (NEB, Beijing, China) and KpnІ-HF®v2 (NEB, Beijing, China), after which each fragment was amplified from 2X_pX458_pSpCas9(BB)-2A-GFP (Plasmid #172221) and pCMV-PE2 and linked by Gibson Assembly. The spacer oligos, scaffold oligos, 3′ extension oligos of pegRNA, and RNA motif-3′ extension oligos of RNA motif-pegRNA were synthesized, annealed, and assembled by DNA ligase (Takara, Dalian, China).
4.2. Cell Culture and Transfection
HEK293T and goat fibroblast cells isolated from the ear margin of female goats aged 1 to 3 months were grown in Dulbecco’s modified Eagle’s medium (DMEM, Gibco, New York, NY, USA) or DMEM/F-12 (Gibco, New York, NY, USA), respectively, supplemented with 10% fetal bovine serum (FBS, Gibco, Queensland, Australia) at 37 °C in humidified incubator with 5% CO2. After seeding cells onto a 12-well plate 1 day before transfection, each well was transfected with 2 µg of plasmids and 10 µL PEI (Polysciences, Shanghai, China) at approximately 60% cell confluency. For the PE3 system, the proportion of plasmids was PE2/pegRNA/sgRNA = 9/3/1. For the twinPE system, the proportion of plasmids was PE2/pegRNA1/pegRNA2 = 6/1/1. For the SSA system, the proportion of plasmids was pSSA-1-3/PE2(WT)/sgRNA/pRL-SV40 = 18/15/4/2. One hour before transfection, the medium was changed to an FBS-free medium and to DMEM supplemented with 10% FBS twelve hours after transfection.
4.3. SSA Reporter System
HEK293T cells were seeded onto a 24-well plate and, the next day, transfected with pSSA-1-3 plasmids of each target site with pCMV-WT-PE2, sgRNA, and pRL-SV40 in triplicate. Forty-eight hours after transfection, cells were washed with PBS and lysed with 1×cell lysis buffer of TransDetect® Double-Luciferase Reporter Assay Kit (Transgene, Beijing, China). Following centrifugation at 12,000× g for 20 min at 4 °C, 20 µL supernatant was added into 3 wells of a 96-well plate. Then, 50 µL Luciferase Reaction Reagent was added into each well, the plate was mixed by vibration, and the chemiluminescence of firefly luciferase was detected with a luminometer. Next, 50 µL Luciferase Reaction Reagent ІІ was added into each well, the plate was mixed by vibration, and the chemiluminescence of renilla luciferase was detected. The relative cutting efficiency of pegRNA was reflected by the ratio of firefly and renilla luciferase.
4.4. Genome DNA Preparation and Detected with PCR
Three days after transfection, cells were washed with PBS and lysed with trypsin (Gibco, NY, USA), after which genomic DNA was extracted with a Genomic DNA kit (Tiangen, Beijing, China). Genomic DNA was amplified with PCR primers and revealed with agarose gel electrophoresis. For junction PCR, edited cells which were revealed objective stripe while WT cells were not. The objective stripe was purified and cloned in pMD19T for subsequent Sanger sequencing.
4.5. Targeted Deep Sequencing and Data Analysis
Sequences ranging from 250 to 300 bp, including the editing, were amplified by PCR and sequenced on an Illumina MiSeq. Alignment of amplicon sequences to a reference sequence was performed using CRISPResso2 (Cambridge, MA, USA). CRISPResso2 was run in BE mode for single base mutation and in HDR mode for insertion or deletion edits.
4.6. Limited Dilution Method for Screening Monoclonal Cells
Three days following transfection, cells were washed with PBS and digested with trypsin. Following centrifugation at 1050 rpm for 5 min, it was resuspended in DMEM/F-12 medium at 1/10,000 dilution in a 10-cm culture dish. After 7 days of cultivation, the culture medium was discarded, washed with PBS, and digested with trypsin. The single-cell clones were gently aspirated with a 100 μL pipette. Then, a portion of the cells was extracted for identification, while the remaining part was used to continue cultivation.
4.7. Puromycin Method for Screening Polyclonal Cells
Three days following transfection, the cell culture medium was supplemented with 1 mg/mL of puromycin to remove cells not expressing puro protein. The cell culture medium was replaced every 2 d, and the puromycin concentration was maintained for 7 d. The positive cell clusters generated in the cell culture dishes could express green fluorescent protein.
4.8. Collection and In Vitro Culture of Goat Oocytes
Upon collection, goat ovaries were transported to the laboratory in physiological saline containing penicillin and streptomycin. The connective tissue around the ovaries was removed to fully expose the follicles. After rinsing the ovaries with 75% alcohol and physiological saline, they were placed in a 60-mm cell culture dish with PBS. Then, the follicles were punctured on the surface of the ovaries to allow the cumulus-oocyte complexes (COCs) to flow into the PBS fluid along with the follicular fluid. COCs with uniform cytoplasm, intact morphology, and complete cumulus cell coating were selected by using an oocyte probe under a stereomicroscope. The COCs were washed twice in PBS, transferred to an oocyte maturation medium (KSOM, Sigma, Saint Louis, MO, USA), and cultured in a cell culture incubator at 38.5 °C in a humidified incubator with 5% CO2.
4.9. Parthenogenetic Activation of Goat Oocytes
Following 22 h in vitro culture, the granulosa cells on the surface of the oocytes were removed, after which the oocytes were transferred to 0.1% hyaluronidase (HY, Sigma, Saint Louis, MO, USA) for 5 min to remove any remaining granulosa cells. Next, oocytes with the first polar body and intact form were selected and transferred to ION ionomycin (Sigma, Saint Louis, MO, USA) droplets and blown and aspirated in two ION ionomycin droplets for 3–5 min in sequence. After 5 min restoration in KSOM, the oocytes were sequentially washed in two 6-DMAP (Sigma, Saint Louis, MO, USA) microdroplets, collected in the third 6-DMAP droplet, and cultured in a cell culture incubator at 38.5 °C in a humidified incubator with 5% CO2 for 4 h.
4.10. Microinjection of Goat Embryos
Four microdroplets of microinjection solution were formed in the cover of a 35-mm culture dish, covered with mineral oil (Sigma, Saint Louis, MO, USA), and preheated to 37 °C. Then, the mRNA of twinPE system was mixed and centrifuged at 10,000 rpm/min for 20 min at 4 °C. Each embryo was injected with 7 pL RNA, washed in G1 embryo culture medium (Thermo Scientific, Boston, MA, USA) droplets, and transferred to G1 embryo culture medium in a four-well plate.
Then, 400 parthenogenetic activation embryos were divided into two groups. Group 1 was microinjected with 200 ng/µL PE2 mRNA, 50 ng/µL pegRNA1, and 50 ng/µL pegRNA2 and group 2 was microinjected with 400 ng/µL PE2 mRNA, 100 ng/µL pegRNA1, and 100 ng/µL pegRNA2. After 5 days of cultivation at 38.5 °C in a humidified incubator with 5% CO2, 42 morulas of group 1 and 59 morulas of group 2 were collected, each embryo was cracked individually, and twinPE efficiency was detected with junction PCR.
4.11. Statistical Analysis
All statistical analyses were performed using GraphPad Prism 6.0. Data were presented as standard error of the mean (SEM) and analyzed with a paired T-test from three independent experiments. Differences were considered statistically significant at p < 0.05.