Next Article in Journal
A New and Sensitive HPLC-UV Method for Rapid and Simultaneous Quantification of Curcumin and D-Panthenol: Application to In Vitro Release Studies of Wound Dressings
Next Article in Special Issue
Microfluidic Microalgae System: A Review
Previous Article in Journal
First Experimental Quantitative Charge Density Studies of Advanced Intermediate of Vitamin D Analogues
Previous Article in Special Issue
Occurrence of Mycosporine-like Amino Acids (MAAs) from the Bloom-Forming Cyanobacteria Aphanizomenon Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Development and Optimization of CRISPR Prime Editing System in Photoautotrophic Cells

1
State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan RD. Minhang District, Shanghai 200240, China
2
Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, 800 Dongchuan RD. Minhang District, Shanghai 200240, China
*
Author to whom correspondence should be addressed.
Molecules 2022, 27(6), 1758; https://doi.org/10.3390/molecules27061758
Submission received: 20 February 2022 / Revised: 2 March 2022 / Accepted: 7 March 2022 / Published: 8 March 2022

Abstract

:
Prime editor (PE), a versatile editor that allows the insertion and deletion of arbitrary sequences, and all 12-point mutations without double-strand breaks (DSB) and a donor template, dramatically enhances research capabilities. PE combines nickase Cas9(H840A) and reverse transcriptase (RT), along with prime editing guide RNA (pegRNA). It has been reported in several plant species, but a weak editing efficiency has led to a decrease in applications. This study reports an optimized-prime editor (O-PE) for endogenous gene editing in Arabidopsis thaliana cells, with an average 1.15% editing efficiency, which is 16.4-fold higher than previously reported. Meanwhile, we observed an increase in indels when testing alternative reverse transcriptase and found out that nCas9(H840A) fused to non-functional reverse transcriptase was responsible for the increase. This work develops an efficient prime editor for plant cells and provides a blueprint for applying PE in other photoautotrophic cells, such as microalgae, that have a high industrial value.

1. Introduction

The development of facile, precise, and efficient genetic editors is essential for engineering organisms. The Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR associated protein (CRISPR-Cas) system, which naturally evolved as a prokaryotic defense system has been extensively exploited to develop editing tools by combining it with cell’s repair mechanisms [1]. The early CRISPR-based editing tools relied heavily on the ability of Cas protein to introduce a double-strand break (DSB) at the target site. This was followed by the introduction of a donor template, along with a reliance on cell repair machinery, either homologues repair (HR) or non-homologues end-joining (NHEJ), for insertion or deletion of the sequence [2]. To counter the issues of DSB-based editors, base editing systems have been developed, which allow DSB-free and template-free editing [3,4]. However, base editors are restricted to base substitutions. There remains a gap for editors that allow DSB-free and template-free insertions, deletions, transition, and transversion; in short, an all-round editor.
Recently, a versatile editing tool, i.e., prime editor (PE), was reported, allowing insertion and deletion of an arbitrary sequence and all 12-point mutations, without DSB and a donor template [5]. PE consists of reverse transcriptase fused to nickase Cas9(H840A) via a flexible linker (nCas9-RT) (Figure 1). The nCas9-RT uses a uniquely designed prime editing guide RNA (pegRNA). The pegRNA contains a single guide RNA (sgRNA) along with primer binding site (PBS) and reverse transcriptase template (RT template), which carries the desired edits. Upon prime editing, nCas9 nicks the non-target strand, PBS binds to the 3′flap acting as a primer for RT, and transcribes the RT template containing the desired edit. The flap is resolved during DNA repair and replication, and the edit is permanently inserted into the target site. The first kind of PE, i.e., PE1, was optimized by developing an engineered M-MLV RT (D200N + L603W + T330P + T306K + W313F); thus, creating the PE2 system. PE2 led to a 1.6 to 5.1-fold increase in point mutation efficiency over PE1. PE2 was further engineered to develop PE3, which involved a second cleavage at an unedited strand 14 to 116 bases away from the original nick. This led to a further 4-fold increase in editing efficiency over PE2. A tool of such versatility holds great promise for plant genetic engineering. Prime editors have been successfully applied to higher plant cells such as rice [6,7,8,9,10,11,12,13], wheat [6], maize [14], tomato [15], tobacco [13], and Arabidopsis [13].
Here, we report the development of PE2 for Arabidopsis cells. The weak PE efficiency led us to optimize it to enhance editing efficiencies. We tested multiple combinations of promoters and reverse transcriptase. Our optimized-PE2 (O-PE2) system led to precise endogenous gene editing in Arabidopsis, with an average 1.15% editing efficiency, an 8.7-fold increase in editing efficiency compared to unoptimized-PE2 (pPE2-35Srat), which was 16.4-fold higher than a previous study [13]. Moreover, when testing alternative reverse transcriptase, we observed a rise in indels with PE2. After investigating the cause, we concluded that nCas9(H840A) fused to a non-functional RT was the culprit. Our strategy provides a blueprint for the development and evolution of PE into photoautotrophic microalgae, whose development of various value-added industrial and pharmaceutically active substances is hindered due to a limited molecular toolbox [16].

2. Results

2.1. Development of Prime Editing in E. coli

To verify the prime editing system, we initially developed a prime editor for E. coli, following the reported design in yeast and mammalian cells [5] (Figure 1). We first used a BglBrick Ps8K plasmid [17] to construct the PE2 system, consisting of arabinose-induced nCas9(H840A) fused to M-MLV RT at C-terminus via a flexible linker (Figure 1 and Figure 2A). Then, a second plasmid pTargetF-pegRNA-CmR was constructed based on pTargetF [3], consisting of a dysfunction chloramphenicol resistance gene CmR H139Y and constitutively expressed pegRNA targeting CmR H139Y (Figure 2A). The H139Y mutation renders CmR non-functional, while PE2 can revert the mutation, making CmR functional.
We carried out a two-plasmid transformation in electrocompetent E. coli MG1655 (Figure 2B), and the cells were then grown on an agar plate, with or without chloramphenicol. As mentioned in the methods, the editing efficiency was calculated and confirmed by Sanger sequencing. Editing via PE occurred and rendered the mutants chloramphenicol resistance, but the editing efficiency was extremely low up to 0.6 × 10−5 (Figure 2C). The conversion of A to G in CmR H139Y gene from the strains with chloramphenicol resistance was identified by sequencing (Figure 2C), confirming our PE is functional in E. coli.

2.2. Development of PE2 in Arabidopsis Cells

To develop PE2 for Arabidopsis cells, we constructed a pPE2-35SRat plasmid, based on pBbE1 [17] plasmid. The pPE2-35SRat plasmid consists of codon-optimized nCas9(H840A)-M-MLV RT fused to green fluorescence protein via T2A linker [18] and pegRNA expressed constitutively by Arabidopsis codon-optimized U6 promoter (Its sequence is shown in Supplementary Materials). The pegRNA targeted PDS3, which is important for dwarfism and mosaic albino phenotype [19], for +3 A to T base substitution (Figure 1).
To test the constructed PE2 in Arabidopsis cells, we transformed the pPE2-35SRat plasmid into Arabidopsis. We selected 3 to 4 weeks old Arabidopsis leaves and processed them into protoplasts and then transfected the pPE2-35SRat plasmid into them using a PEG transfection buffer. The positively transfected cells presented green fluoresce (Figure 3B), owing to the GFP gene in the plasmid. The transformed cells were selected and collected by flow cytometry, based on GFP fluoresces. The collected cells were then used for genome extraction, and PCR amplified the target sequences of PDS3-1. The amplicons were sequenced by next-generation sequencing and analyzed using CRISPResso2 [20]. The research scheme is shown in Figure 3A. The results showed a base substitution efficiency of 0.1329% (Figure 3C and Table 1), similar to the previous report [13]. However, the PE2 system was less efficient in Arabidopsis compared with other higher plants such as rice.

2.3. Optimization of PE2 in Arabidopsis Cells

Considering the weak PE2 efficiency, we tweaked the system using various promoters for the fusion protein and RTs. We tested 2x35S, RPS5A, and UBQ10 promoters, common promoters for protein expression in Arabidopsis cells. We also tested different RTs: Fusicatenibacter saccharivorans-RT [21] (Fs-RT), Marinomonas mediterranea-RT [22] (Mm-RT), Vibrio vulnificus-RT [23] (Vv-RT), and Arabidopsis codon-optimized-RT. All the sequences are shown in the Supplementary Materials. We constructed total of six plasmids based on these, named pPE2-35SAt, pPE2-RPS5AAt, pPE2-UBQ10At, pPE2-35SFs, pPE2-35SMm, and pPE2-35SVv (Figure 3D). These plasmids were individually introduced into the protoplasts for the PE editing test through the method mentioned earlier. Finally, we successfully obtained a highest efficiency of 1.44% (based on three independent experiments, the efficiencies were 1.44%, 1.38%, and 0.63%, respectively) by combining the 35S promoter and Arabidopsis codon-optimized reverse transcriptase (Figure 3C,D and Table 1), namely optimized-PE2 (O-PE2). The others did not work well, with their efficiencies ranging between 0.0187% and 0.0342% (Table 1). Compared to initially constructed pPE2-35SRat, pPE2-35SAt (O-PE2) boosted PE2 efficiency to 8.7-fold at PDS3-1 loci.
Based on the O-PE2 system, we developed a PE3/3b system (pPE3-35SAt/pPE3b-35SAt Figure 3D) in Arabidopsis cells. To our surprise, contrary to reports in other species and mammalian cells [5], PE3/3b did not significantly improve the modification efficiency, indicating that the addition of another sgRNA does not improve the efficiency in Arabidopsis cells in the current conditions.

2.4. nCas9(H840A) in PE2 Cause Indels

While testing alternate promoter/reverse-transcriptase combinations, we observed an increased variation in the DNA sequence at the target site with some combinations (Table 1 and Figure 4). The PE plasmids, pPE2-RPS5AAt, pPE2-UBQ10At, pPE2-35SFs, pPE2-35SMm, and pPE2-35SVv did not perform the desired editing. The sequencing results also showed a lower proportion of wild-type sequence reads between 40.36% to 84.56%, contrary to O-PE2, which had 94.23% wild-type reads. The altered reads included insertions, deletions, or substitutions near the target sequence. We hypothesized that in those cases, reverse transcriptase was non-functional, leaving the nCas9(H840A) cut to be repaired, which led to indels during the repair process. To confirm this hypothesis, we constructed pPE2-35SAt-NT, which lacks PBS and a RT template targeting the PDS3-1 locus. The NGS results showed only 50.73% of reads were the wild-type sequence, while the rest contained indels (Figure 4 and Table 1). These results supported the hypothesis that indels were observed when RT was non-functional. At PDS3-2, where PE2 worked, we observed negligible indels, indicating that indels are only caused when RT is non-functional (Table 1).

3. Discussion

CRISPR-Cas9-mediated gene-editing tools, including PE, have been widely applied in plant cells [24], to increase yield, regulate metabolites, improve stress resistance, etc. [25]. However, the editing efficiency of the PE system is still relatively lower than that of the traditional Cas9 editing tools. The editing efficiency of the PE system in different species is affected [26] by the selection of the spacer position, length of the PBS, size of the RT template, and their combination [5,6,27].
A previous report of the development of PE for Arabidopsis involved the insertion of GFP11 at the AT1G26660.1 locus, but reported an extremely low editing efficiency of 0.07 ± 0.12% [13]. In this paper, we improved the editing efficiency of PE in Arabidopsis, by optimizing combinations of different promoters and reverse-transcriptase. We used 35S promoter and Arabidopsis codon-optimized RT to achieve an average 1.15% single base substitution modification efficiency at the endogenous gene, for the first time. Further optimization and improvement of the editing efficiency of O-PE could involve adjusting pegRNA by following the guidelines from the research of the Gao group [26] and Liu group [28]. In addition, we tested PE3/3b in Arabidopsis and, consistent with previous studies [6], found no significant improvement in editing efficiency.
Furthermore, we found that nCas9(H840A) led to indels during the testing of alternate promoters and reverse-transcriptase. Previous work from the Liu group [29] reported the same observation in the base editor, in which nCas9(H840A), and not nCas9(D10A), caused a high proportion of indels in animal cells. However, this was random and altered with the target site. This study designed a plasmid (pPE2-35SAt-NT) as a control, which carried sgRNA. The analysis of NGS showed that at sites where PE2 was efficient, nCas9(H840A) caused a large number of indels when fused to dysfunctional reverse-transcriptase. Contrarily, when the reverse-transcriptase was functional, PE2 did not cause indels.
Adoption of PE is an important addition to the molecular toolbox and goes beyond Arabidopsis. Microalgae are considered a third-generation biofuel. It has incredibly high bioenergy, aquatic products, food, pharmaceutical, and medical value. The traditional method to improve the yield of microalgae is to control the accumulation of different metabolites by regulating the carbon flow, by changing the culture conditions, such as pH, temperature, and nitrogen stress. However, such methods have certain limitations that affect cell division and ultimately reduce the overall biological yield [30]. The development of versatile and efficient molecular tools can overcome such drawbacks, by overexpression or repression of desired pathways via genetic manipulation. CRISPR-Cas based tools have been deployed in Chlamydomonas reinhardtii [31]. However, the dependence of previous editors on DSB, homologous recombination, and the need for template DNA restricts their application. As a new generation of gene editors, PE has incomparable advantages over the previous generation and is an excellent candidate for gene editing of microalgae. To date, there have been no reports of prime editing in microalgae.
In summary, we developed an optimized-PE2 (O-PE) editor for Arabidopsis, achieving an average substitution efficiency of 1.15% of endogenous genes. We further investigated the indel formation in cases where RT was dysfunctional and proved nCas9(H840A) to be the main factor. Our strategy of constructing PE editors via a combination of various promoters, RT, and sequence codon-optimization provides a blueprint for the development of PE for genetically engineered high-yield microalgae [16,30,32].

4. Materials and Methods

4.1. Plasmid Construction

To construct vector pPE2-35SRat, NLS, U6 promoter, double 35S promoter, and nCas9(H840A) codon-optimized for Arabidopsis by Tsingke, Shanghai, China, the engineered M-MLV reverse transcriptase which was amplified from Addgene# 132775, Cambridge MA, USA, EGFP which was from Prof. Gong, and T2A linker were cloned into the vector pBbE1 [17] backbone, yielding the various nCas9(H840A)-RT fusion plasmids by Golden Gate. AT-RT were codon-optimized for Arabidopsis by Tsingke. The UBQ10 promoter was a gift from Prof. Gong. The natural reverse transcriptase-Cas1 fusion protein Mm-RT [22], Vv-RT [23] and Fs-RT [21] were synthesized by Genewiz, Shanghai, China. The pegRNAs were created by paired primers containing the target sgRNA, PBS, and RT template sequences using T4 ligase (NEB) and introduced into the vectors by Golden Gate. Another sgRNA of PE3/PE3b used the same method for introduction into vectors. All the plasmid constructions were confirmed by clone PCR, enzyme digest, and Sanger sequencing.
The E. coli PE system, i.e., pS8k-nCas9RT, consisted of a low copy Sc101 ori and kanamycin resistance gene cloned with arabinose-induced nickase Cas9(H840A) fused to engineered M-MLV reverse-transcriptase via XTEN linker [5]. The second plasmid pTargetF-pegRNA-CmR consisted of high copy colE1 ori, spectinomycin resistance gene, dysfunctioned chloramphenicol resistance gene CmR H193Y, and pegRNA expressed constitutively by J23119 promoter.

4.2. Efficiency Assay in E. coli

For PE assay in E. coli, we performed dual-plasmid transformation of pS8k-nCas9RT and pTargetF-pegRNA-CmR to electric component E. coli MG1655 cells. After transformation, cells were recovered for 1 h at 37 °C at 200 rpm. The recovered cells were transferred to 5 mL fresh LB and inoculated overnight at 37 °C at 220 rpm. The cells were then serially diluted and plated on kanamycin (Kan) + Spectinomycin (Spec) and kanamycin (Kan) + Spectinomycin (Spec) + Chloramphenicol (Cm) plates. The editing efficiency was calculated as follows:
No .   of   clones   on   Kan   +   Spec   +   Cm   plate No .   of   clones   on   Kan   +   Spec   plate

4.3. Protoplast Transfection

Plants were grown at 22 °C under 12 h light and 12 h dark cycles. Three to four-week-old Col-0 leaves were used for the protoplast transient assay, and the protocol was from Sheen Lab [33]. We isolated protoplasts from leaf strips using an enzyme solution (1.4% cellulose (Yakult, Nagoya, Japan), 0.32% macerozyme (Yakult), 0.4 M mannitol (Sigma, Burlington, VT, USA), 20 mM KCl (Sinopharm, Shanghai, China), 20 mM MES (4-Morpholineethanesulfonic acid hydrate, 2-(N-Morpholino) ethanesulfonic acid hydrate, Sigma), pH 5.7, 10 mM CaCl2 (Sinopharm), 0.1% BSA (bovine serum albumin, Sigma)), which was shaken (20 rpm) in the dark at room temperature for 2 h. The digest reaction was terminated by W5 solution (2 mM MES (pH 5.7) (Sigma), 154 mM NaCl (Sinopharm), 125 mM CaCl2 (Sinopharm), 5 mM KCl (Sinopharm)) and filtered through a 70-μm cell strainer (Falcon, New York, NY, USA). After removing undigested cellular debris, protoplasts were centrifuged (1400 rpm) and resuspended with pre-chilled W5 solution twice. The protoplasts were placed in 5 mL W5 solution in ice for at least 0.5 h, following washing steps. Then, after removing the supernatant, cells were quantified using a hemocytometer and resuspended in MMG solution (4 mM MES (pH 5.7) (Sigma), 0.4 M mannitol (Sigma), and 15 mM MgCl2 (Sinopharm)). The plasmids (10 μg) were introduced into 2 × 105 cells (100 μL) using 1.1-fold volume PEG transfection buffer (40% PEG4000 (poly ethylene glycol, Sigma), 0.4 M mannitol (Sigma), and 0.1 M CaCl2 (Sinopharm)). The transfections were terminated with 5-fold volume W5 solution and then centrifuged (100 g). The cells were resuspended with 250 μL WI solution (4 mM MES (pH 5.7) (Sigma), 0.5 M mannitol (Sigma), 20 mM KCl (Sinopharm)) into 24-well plates for 16 h in the dark, before microscopy assay or FACS (Fluorescence activated Cell Sorting) separation.

4.4. DNA Extraction and Amplicon Sequencing

Genomic DNA of protoplasts was extracted using a DNAsecure Plant Kit (Tiangen, Shanghai, China), and Target sequences were amplified using Phusion polymerase (Thermo Fisher Scientific, Waltham, MA, USA) with specific primers (Supplementary Materials) for Sanger sequencing (Tsingke, Shanghai, China) or next-generation sequencing (Sangon/Biozeron, Shanghai, China). NGS reads were analyzed using CRISPResso2 (version 2.0.38; Cambridge, MA, USA, 2019) [20].

Supplementary Materials

The supporting Sequence information can be downloaded at https://www.mdpi.com/article/10.3390/molecules27061758/s1.

Author Contributions

Conceptualization, Z.J., A., Y.X.; methodology, Z.J., A.; validation, Y.X.; formal analysis, Z.J.; investigation, Z.J., A., S.Z.; resources, Z.J., R.L., Y.J.; data curation, Z.J., A.; writing—original draft preparation, Z.J.; writing—review and editing, Z.J., A., Y.X.; supervision, Y.X.; project administration, Y.X.; funding acquisition, Y.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key R&D Program of China (2018YFA0901200), the National Natural Science Foundation of China (31870071), and the Oceanic Interdisciplinary Program of Shanghai Jiao Tong University (SL2020MS028).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available from the corresponding author: [email protected].

Acknowledgments

We thank Qingqiu Gong from Shanghai Jiao Tong University for valuable suggestions and the gift of plasmids in Arabidopsis and Haixia Jiang from Core Facility and Technical Service Center for SLSB, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University for technical support for FACS analysis.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples from the compounds are available from the corresponding author: [email protected].

References

  1. Hille, F.; Richter, H.; Wong, S.P.; Bratovic, M.; Ressel, S.; Charpentier, E. The Biology of CRISPR-Cas: Backward and Forward. Cell 2018, 172, 1239–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Paquet, D.; Kwart, D.; Chen, A.; Sproul, A.; Jacob, S.; Teo, S.; Olsen, K.M.; Gregg, A.; Noggle, S.; Tessier-Lavigne, M. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 2016, 533, 125–129. [Google Scholar] [CrossRef] [PubMed]
  3. Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature 2017, 551, 464–471. [Google Scholar] [CrossRef]
  4. Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016, 533, 420–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Raguram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149–157. [Google Scholar] [CrossRef]
  6. Lin, Q.; Zong, Y.; Xue, C.; Wang, S.; Jin, S.; Zhu, Z.; Wang, Y.; Anzalone, A.V.; Raguram, A.; Doman, J.L.; et al. Prime genome editing in rice and wheat. Nat. Biotechnol. 2020, 38, 582–585. [Google Scholar] [CrossRef]
  7. Tang, X.; Sretenovic, S.; Ren, Q.; Jia, X.; Li, M.; Fan, T.; Yin, D.; Xiang, S.; Guo, Y.; Liu, L.; et al. Plant Prime Editors Enable Precise Gene Editing in Rice Cells. Mol. Plant 2020, 13, 667–670. [Google Scholar] [CrossRef]
  8. Li, H.; Li, J.; Chen, J.; Yan, L.; Xia, L. Precise Modifications of Both Exogenous and Endogenous Genes in Rice by Prime Editing. Mol. Plant 2020, 13, 671–674. [Google Scholar] [CrossRef]
  9. Xu, R.; Li, J.; Liu, X.; Shan, T.; Qin, R.; Wei, P. Development of Plant Prime-Editing Systems for Precise Genome Editing. Plant Commun. 2020, 1, 100043. [Google Scholar] [CrossRef]
  10. Xu, W.; Zhang, C.; Yang, Y.; Zhao, S.; Kang, G.; He, X.; Song, J.; Yang, J. Versatile Nucleotides Substitution in Plant Using an Improved Prime Editing System. Mol. Plant 2020, 13, 675–678. [Google Scholar] [CrossRef]
  11. Butt, H.; Rao, G.S.; Sedeek, K.; Aman, R.; Kamel, R.; Mahfouz, M. Engineering herbicide resistance via prime editing in rice. Plant Biotechnol. J. 2020, 18, 2370–2372. [Google Scholar] [CrossRef] [PubMed]
  12. Hua, K.; Jiang, Y.; Tao, X.; Zhu, J.K. Precision genome engineering in rice using prime editing system. Plant Biotechnol. J. 2020, 18, 2167–2169. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, L.; Kaya, H.B.; Zhang, N.; Rai, R.; Willmann, M.R.; Carpenter, S.C.D.; Read, A.C.; Martin, F.; Fei, Z.; Leach, J.E.; et al. Spelling Changes and Fluorescent Tagging With Prime Editing Vectors for Plants. Front. Genome Ed. 2021, 3, 617553. [Google Scholar] [CrossRef] [PubMed]
  14. Jiang, Y.Y.; Chai, Y.P.; Lu, M.H.; Han, X.L.; Lin, Q.; Zhang, Y.; Zhang, Q.; Zhou, Y.; Wang, X.C.; Gao, C.; et al. Prime editing efficiently generates W542L and S621I double mutations in two ALS genes in maize. Genome Biol. 2020, 21, 257. [Google Scholar] [CrossRef] [PubMed]
  15. Lu, Y.; Tian, Y.; Shen, R.; Yao, Q.; Zhong, D.; Zhang, X.; Zhu, J.K. Precise genome modification in tomato using an improved prime editing system. Plant Biotechnol. J. 2021, 19, 415–417. [Google Scholar] [CrossRef] [PubMed]
  16. Patel, V.K.; Soni, N.; Prasad, V.; Sapre, A.; Dasgupta, S.; Bhadra, B. CRISPR-Cas9 System for Genome Engineering of Photosynthetic Microalgae. Mol. Biotechnol. 2019, 61, 541–561. [Google Scholar] [CrossRef]
  17. Lee, T.S.; Krupa, R.A.; Zhang, F.; Hajimorad, M.; Holtz, W.J.; Prasad, N.; Lee, S.K.; Keasling, J.D. BglBrick vectors and datasheets: A synthetic biology platform for gene expression. J. Biol. Eng. 2011, 5, 12. [Google Scholar] [CrossRef] [Green Version]
  18. Liu, Z.; Chen, O.; Wall, J.B.J.; Zheng, M.; Zhou, Y.; Wang, L.; Ruth Vaseghi, H.; Qian, L.; Liu, J. Systematic comparison of 2A peptides for cloning multi-genes in a polycistronic vector. Sci. Rep. 2017, 7, 2193. [Google Scholar] [CrossRef]
  19. Kang, B.C.; Yun, J.Y.; Kim, S.T.; Shin, Y.; Ryu, J.; Choi, M.; Woo, J.W.; Kim, J.S. Precision genome engineering through adenine base editing in plants. Nat. Plants 2018, 4, 427–431. [Google Scholar] [CrossRef]
  20. Clement, K.; Rees, H.; Canver, M.C.; Gehrke, J.M.; Farouni, R.; Hsu, J.Y.; Cole, M.A.; Liu, D.R.; Joung, J.K.; Bauer, D.E.; et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 2019, 37, 215–226. [Google Scholar] [CrossRef]
  21. Schmidt, F.; Cherepkova, M.Y.; Platt, R.J. Transcriptional recording by CRISPR spacer acquisition from RNA. Nature 2018, 562, 380–385. [Google Scholar] [CrossRef] [PubMed]
  22. Silas, S.; Mohr, G.; Sidote, D.J.; Markham, L.M.; Sanchez-Amat, A.; Bhaya, D.; Lambowitz, A.M.; Fire, A.Z. Direct CRISPR spacer acquisition from RNA by a natural reverse transcriptase-Cas1 fusion protein. Science 2016, 351, aad4234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gonzalez-Delgado, A.; Mestre, M.R.; Martinez-Abarca, F.; Toro, N. Spacer acquisition from RNA mediated by a natural reverse transcriptase-Cas1 fusion protein associated with a type III-D CRISPR-Cas system in Vibrio vulnificus. Nucleic Acids Res. 2019, 47, 10202–10211. [Google Scholar] [CrossRef] [PubMed]
  24. Zheng, Y.; Li, Q.; Ye, M.; Chen, A.; Wang, H. Applications of CRISPR/Cas9-based genome editing in the plant biology. Tur. J. Botany 2021, 45, 253–268. [Google Scholar] [CrossRef]
  25. Hassan, M.M.; Zhang, Y.; Yuan, G.; De, K.; Chen, J.G.; Muchero, W.; Tuskan, G.A.; Qi, Y.; Yang, X. Construct design for CRISPR/Cas-based genome editing in plants. Trends Plant Sci. 2021, 26, 1133–1152. [Google Scholar] [CrossRef]
  26. Lin, Q.; Jin, S.; Zong, Y.; Yu, H.; Zhu, Z.; Liu, G.; Kou, L.; Wang, Y.; Qiu, J.L.; Li, J.; et al. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat. Biotechnol. 2021, 39, 923–927. [Google Scholar] [CrossRef]
  27. Liu, Y.; Li, X.; He, S.; Huang, S.; Li, C.; Chen, Y.; Liu, Z.; Huang, X.; Wang, X. Efficient generation of mouse models with the prime editing system. Cell Discov. 2020, 6, 27. [Google Scholar] [CrossRef]
  28. Nelson, J.W.; Randolph, P.B.; Shen, S.P.; Everette, K.A.; Chen, P.J.; Anzalone, A.V.; An, M.; Newby, G.A.; Chen, J.C.; Hsu, A.; et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 2021. [Google Scholar] [CrossRef]
  29. Rees, H.A.; Yeh, W.H.; Liu, D.R. Development of hRad51-Cas9 nickase fusions that mediate HDR without double-stranded breaks. Nat. Commun. 2019, 10, 2212. [Google Scholar] [CrossRef]
  30. Banerjee, S.; Banerjee, S.; Ghosh, A.K.; Das, D. Maneuvering the genetic and metabolic pathway for improving biofuel production in algae: Present status and future prospective. Renew. Sustain. Energy Rev. 2020, 133, 110155. [Google Scholar] [CrossRef]
  31. Jiang, W.; Brueggeman, A.J.; Horken, K.M.; Plucinak, T.M.; Weeks, D.P. Successful transient expression of Cas9 and single guide RNA genes in Chlamydomonas reinhardtii. Eukaryot. Cell 2014, 13, 1465–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Lin, W.R.; Tan, S.I.; Hsiang, C.C.; Sung, P.K.; Ng, I.S. Challenges and opportunity of recent genome editing and multi-omics in cyanobacteria and microalgae for biorefinery. Bioresour. Technol. 2019, 291, 121932. [Google Scholar] [CrossRef] [PubMed]
  33. Yoo, S.D.; Cho, Y.H.; Sheen, J. Arabidopsis mesophyll protoplasts: A versatile cell system for transient gene expression analysis. Nat. Protoc. 2007, 2, 1565–1572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Illustration of PE system. Prime editor consists of nCas9(H840A) fused to reverse transcriptase (RT) via XTEN linker (a 16 amino acid flexible linker, black line). The fusion protein is guided by a pegRNA which consists of primer binding site (PBS), RT template and sgRNA. PBS (yellow), RT template (red). This figure shows how a mutation (+3 A to T) was introduced (green).
Figure 1. Illustration of PE system. Prime editor consists of nCas9(H840A) fused to reverse transcriptase (RT) via XTEN linker (a 16 amino acid flexible linker, black line). The fusion protein is guided by a pegRNA which consists of primer binding site (PBS), RT template and sgRNA. PBS (yellow), RT template (red). This figure shows how a mutation (+3 A to T) was introduced (green).
Molecules 27 01758 g001
Figure 2. Prime editing in E. coli. (A) Diagram of the PE2 system plasmids in E. coli. The pS8k-nCas9RT consists of nCas9(H840A) fused to M-MLV RT induced by arabinose inducible promoter and Kan resistance. The pTargetF-pegRNA-CmR consists of dysfunctional CmR H139Y, pegRNA targeting CmR H139Y expressed via constitutive J23119 promoter, and spectinomycin resistance gene. (B) Dual-plasmid transformed E. coli MG1655 was serially diluted and plated. Control diluted 10−4 on kan + spec plate (left), recovered CmR via PE2 on kan + spec + cm plate. (C) A sanger sequencing chromatogram and editing efficiency.
Figure 2. Prime editing in E. coli. (A) Diagram of the PE2 system plasmids in E. coli. The pS8k-nCas9RT consists of nCas9(H840A) fused to M-MLV RT induced by arabinose inducible promoter and Kan resistance. The pTargetF-pegRNA-CmR consists of dysfunctional CmR H139Y, pegRNA targeting CmR H139Y expressed via constitutive J23119 promoter, and spectinomycin resistance gene. (B) Dual-plasmid transformed E. coli MG1655 was serially diluted and plated. Control diluted 10−4 on kan + spec plate (left), recovered CmR via PE2 on kan + spec + cm plate. (C) A sanger sequencing chromatogram and editing efficiency.
Molecules 27 01758 g002
Figure 3. Prime editing in Arabidopsis cells. (A) The Arabidopsis cells were transfected with the PE system. The positively transfected cells were selected via flow cytometry. This was followed by genome extraction, amplification of target sequence, NGS, and data analysis. (B) Fluorescence of GFP+ protoplasts. Red signal in Cy5 channel indicates autofluorescence from chloroplast, green signal indicates GFP protein overexpressed in the cytoplasm from plasmids. Untreated protoplast served as a negative control. Scale bar, 20 µm. (C) Frequency reads with desired edits (%) of pPE2-35SRat and pPE2-35SAt at PDS3-1 site using next-generation sequencing (NGS). Black balls indicate independent results. Error bars represent mean ± s.d. of n = 3 independent biological replicates. (D) Features of each vector in Arabidopsis cells.
Figure 3. Prime editing in Arabidopsis cells. (A) The Arabidopsis cells were transfected with the PE system. The positively transfected cells were selected via flow cytometry. This was followed by genome extraction, amplification of target sequence, NGS, and data analysis. (B) Fluorescence of GFP+ protoplasts. Red signal in Cy5 channel indicates autofluorescence from chloroplast, green signal indicates GFP protein overexpressed in the cytoplasm from plasmids. Untreated protoplast served as a negative control. Scale bar, 20 µm. (C) Frequency reads with desired edits (%) of pPE2-35SRat and pPE2-35SAt at PDS3-1 site using next-generation sequencing (NGS). Black balls indicate independent results. Error bars represent mean ± s.d. of n = 3 independent biological replicates. (D) Features of each vector in Arabidopsis cells.
Molecules 27 01758 g003
Figure 4. NGS analyzed data. Alleles of PDS3-1 site isolated from protoplasts of Arabidopsis after editing with pPE2-35SAt, pPE2-35SAt-NT, and pPE2-35SMm. The amplicons sequenced by an Illumina MiSeq for NGS were analyzed using CRISPResso2. (A) represents the wild-type reference sequence. (B) represents the successful PE2 via pPE2-35SAt. (C) represents control consisting of sgRNA and not pegRNA. (D) represents indels when pPE2-35SMm was used.
Figure 4. NGS analyzed data. Alleles of PDS3-1 site isolated from protoplasts of Arabidopsis after editing with pPE2-35SAt, pPE2-35SAt-NT, and pPE2-35SMm. The amplicons sequenced by an Illumina MiSeq for NGS were analyzed using CRISPResso2. (A) represents the wild-type reference sequence. (B) represents the successful PE2 via pPE2-35SAt. (C) represents control consisting of sgRNA and not pegRNA. (D) represents indels when pPE2-35SMm was used.
Molecules 27 01758 g004
Table 1. Efficiencies of various prime editors in protoplasts of Arabidopsis thaliana.
Table 1. Efficiencies of various prime editors in protoplasts of Arabidopsis thaliana.
PlasmidTarget SiteAimAverage EfficiencyAverage Proportion of NGS Reads with No Changes (%)
Non 1PDS3-1/0.0203%90
pPE2-35SAtPDS3-1+3 A to T1.1506%94.23
pPE2-RPS5AAtPDS3-1+3 A to T0.0342%58.05
pPE2-UBQ10AtPDS3-1+3 A to T0.0194%77.89
pPE3b-35SAtPDS3-1+3 A to T0.0318%78.42
pPE2-35SRatPDS3-1+3 A to T0.1329%86.39
pPE2-35SFsPDS3-1+3 A to T0.0255%84.56
pPE2-35SMmPDS3-1+3 A to T0.0214%49.56
pPE2-35SVvPDS3-1+3 A to T0.0187%40.36
pPE2-35SAt-NTPDS3-1/0.0336%50.73
Non 1PDS3-2/0.0997%93.39
pPE2-35SAtPDS3-2+5 G to A0.1167%92.83
Non 1ALS-1/0.1369%93.47
pPE3-35SAtALS-1+2 A to G0.1547%91.71
1 No vectors.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jiang, Z.; Abdullah; Zhang, S.; Jiang, Y.; Liu, R.; Xiao, Y. Development and Optimization of CRISPR Prime Editing System in Photoautotrophic Cells. Molecules 2022, 27, 1758. https://doi.org/10.3390/molecules27061758

AMA Style

Jiang Z, Abdullah, Zhang S, Jiang Y, Liu R, Xiao Y. Development and Optimization of CRISPR Prime Editing System in Photoautotrophic Cells. Molecules. 2022; 27(6):1758. https://doi.org/10.3390/molecules27061758

Chicago/Turabian Style

Jiang, Zhengzheng, Abdullah, Shun Zhang, Yuli Jiang, Rui Liu, and Yi Xiao. 2022. "Development and Optimization of CRISPR Prime Editing System in Photoautotrophic Cells" Molecules 27, no. 6: 1758. https://doi.org/10.3390/molecules27061758

Article Metrics

Back to TopTop