ABSTRACT
Genome-editing (GE) techniques like base editing are ideal for introducing novel gain-of-function mutations and in situ protein evolution. Features of base editors (BEs) such as higher efficacy, relaxed protospacer adjacent motif (PAM), and a broader editing window enables diversification of user-defined targeted locus. Cytosine (CBE) or adenine (ABE) BEs alone can only alter C-to-T or A-to-G in target sites. In contrast, dual BEs (ACBEs) can concurrently generate C-to-T and A-to-G modifications. Although BE tools have recently been applied in microbes, there is no report of ACBE for microbial GE. In this study, we engineered four improved ACBEs (iACBEs) tethering highly active CBE and ABE variants that can introduce synchronized C-to-T and A-to-G mutations in targeted loci. iACBE4 generated by evoCDA1-ABE9e fusion demonstrated a broader editing window (positions −6 to 15) and is also compatible with the multiplex editing approach in Escherichia coli. We further show that the iACBE4-NG containing PAM-relaxed nCas9-NG expands the targeting scope beyond NGG (N-A/G/C/T) PAM. As a proof-of-concept, iACBE was effectively utilized to identify previously unknown mutations in the rpoB gene, conferring gain-of-function, i.e., rifampicin resistance. The iACBE tool would expand the CRISPR-GE toolkit for microbial genome engineering and synthetic biology.
KEYWORDS: CRISPR, base editing, dual base editor, genome editing, microbial engineering
INTRODUCTION
Several genetic engineering tools based on CRISPR/Cas system have recently been developed and adopted to edit genetic information in different organisms (1–3). Base editors (BEs) are one of the emerging CRISPR technologies that offer unique features compared to traditional CRISPR/Cas9 uses. For example, the primary outcome of CRISPR/Cas9 application includes the insertion or deletion that typically introduces frameshift mutations leading to the generation of a premature stop codon (knockout) or nonfunctional peptides. In contrast, BE tools generate base substitutions using deaminases tethered with Cas9 variants and DNA repair components to create point mutations, allowing in vivo protein evolution (4). Moreover, double-strand breaks (DSBs)-mediated gene disruption by CRISPR/Cas9 is primarily the outcome of the error-prone nonhomologous end-joining (NHEJ) pathway. The active NHEJ pathway is absent in most bacteria, thus impeding the CRISPR/Cas9 use. Even when NHEJ is present in some bacteria, it is not as active as in mammalian cells. In this scenario, DSB-free BEs are promising tools for targeted base editing in bacteria.
Among the available BE arsenals, the main BE tools comprise cytosine base editor (CBE) (5, 6) and adenine base editor (ABE) (7) that enable C:G to T:A and A:T to G:C conversion, respectively. Individual CBE or ABE can generate mutations in only a single type of nucleotide base-pair. To broaden the range of DNA modifications, simultaneous C-to-T and A-to-G substitution was achieved by developing dual (adenine-cytosine) base editors (ACBEs) by combining the deaminases and related BE components of CBEs and ABEs (4, 8–14). ACBE-mediated simultaneous conversion of two types of DNA base-pairs on the same target site enables greater diversification of desired alleles in directed evolution studies. Saturated mutagenesis of target genes by ACBE is an efficient way for studying the genotype-phenotype relationships.
Although BE tools have recently been reported for microbial genome engineering (15), to the best of our knowledge, there is no report of ACBE tool establishment in microbial species, including bacteria. Also, most of the earlier ACBE types consisted of Streptococcus pyogenes Cas9 (SpCas9, hereafter Cas9) nickase (nCas9, D10A) that recognizes NGG (N- A/G/C/T) as a protospacer adjacent motif (PAM), offering a limited target range. One of the significant challenges for applying highly active BE-based tools in bacteria is the adverse effects of overexpressed BE components on cell growth and survival (15–17). In some cases, the use of inducible or relatively weak constitutive promoters, adding a protein degradation (LVA) tag, replacing nCas9 with catalytically dead Cas9 (dCas9, D10A+H840A), or a combination of all the above factors helped to reduce the cytotoxicity permitting the BE applications in different microbial species, including E. coli, Bacillus sp., Pseudomonas sp., Agrobacterium spp., Paenibacillus polymyxa, Streptomyces spp., etc. (15–20). Moreover, highly active BE variants such as evoCDA1, ABE8e, and ABE9e would pose further difficulty due to their broader editing window and faster base conversion rate than their predecessors (21–23). Recently, a platform named in vivo rapid investigation of BE components in E. coli (IRI-CCE) (24) was developed that comprises an optimized set of promoter-terminator combinations allowing an appropriate amount of BE expression with higher editing activities at desired target sites in plasmid DNA or genomic loci.
This study describes the establishment of improved ACBE (iACBE) systems for bacterial use by employing the IRI-CCE platform in E. coli. We engineered a series of iACBEs by fusing different CBE (PmCDA1, evoCDA1 [evolved version of PmCDA1], APOBEC3A) and ABE (ABE8e, ABE9e) variants to accomplish efficient A/C base editing in plasmid and chromosomal targets in E. coli. Furthermore, we comprehensively analyzed the applicability of iACBE4, the most active iACBE type comprising evoCDA1-ABE9e, by targeting 14 genomic target sites. To broaden the targeting range of iACBE4, nCas9-NG, a PAM-flexible Cas9 variant recognizing NGN as a PAM (25), was fused to generate an iACBE4-NG tool that enabled the editing of targets with non-NGG PAM. To avoid self-targeting frequency while using nCas9-NG, the modified single guide RNA (sgRNA) scaffold starting with “GCCCC” (esgRNA) (26) was evaluated. Using the optimized iACBE4 and iACBE4-NG, we targeted the RNA polymerase rpoB gene of E. coli implicated in rifampicin resistance (RifR). The screening of generated rpoB mutants identified previously unknown mutations in single or multiple amino acids bestowing RifR in E. coli cells. This study demonstrates the iACBE potential for mutant library construction of desired genes enabling the quick evaluation of the correlation between genotype and gain-of-function.
RESULTS
Design and construction of improved dual base editor (iACBE) system.
To characterize a dual base editing system for the bacterial application, we designed four ACBE architectures combining ABE8e or ABE9e with either of the three different CBE versions, i.e., PmCDA1 and evoCDA1 and APOBEC3A (Fig. 1A). In the first set of mammalian and plant ACBE reports, editing efficiencies from A-to-G were lower than those of C-to-T, thereby reducing the occurrence of synchronized base editing (4, 8, 9, 11, 14). The heterodimer of adenosine deaminase (TadA-TadA*) opted for the ABE7.10 variant or ACBEs (7–14). The TadA-TadA* convert A-to-G within the editing window of positions 4 to 8 in the protospacer region (considering the PAM at positions 21 to 23) with relatively low efficiency (7). We reasoned that using highly active TadA in ACBE would boost the prospects of attaining concurrent base editing. Therefore, we chose monomeric ABE8e (23) and ABE9e (22) for bacterial ACBE designs, the most active TadA versions so far. The fusion of TadA to the N-terminus of nCas9 was chosen since C-terminal fusion was reported to lack activity in ACBEs (14).
Meanwhile, PmCDA1, a sea lamprey cytidine deaminase, mediates C-to-T conversion within 1 to 5 bases as a canonical editing window in the protospacer region (5). Similar to the earlier mammalian ACBEs (8, 9, 11), PmCDA1 with one copy of uracil-DNA glycosylase inhibitor (UGI) was fused to the C-terminus and ABE8e at the N-terminus of nCas9, creating iACBE version 1 (iACBE1) (Fig. 1B). The use of UGI increases the purity of C-to-T conversion at target sites, a strategy used in previous reports of CBEs and ACBEs. Next, we chose evoCDA1 or APOBEC3A as ABE8e partners by fusing both the deaminases to the N-terminus of Cas9, producing iACBE2 and iACBE3, respectively. Two UGI (2xUGI) copies were connected at the C-terminus of nCas9, likewise used in previously published reports. Both evoCDA1 and APOBEC3A exhibited broader editing windows in E. coli and other organisms (21, 24, 27). The iACBE4 construct was generated by combining the most active individual BE deaminases, i.e., evoCDA1 and ABE9e (Fig. 1B). An optimized combination of promoter-terminator (pGlpT-TerL3S2P21) that allowed nonlethal expression of single-function BEs (ABEs and CBEs) in IRI-CCE (24), was adopted for the expression of iACBEs with a single-plasmid system in E. coli.
Establishment of iACBE system in E. coli.
To examine the synchronized base-editing activities of the four designed ACBE constructs, we performed editing tests at two sgRNA target regions (Target 1, T1 by Test sgRNA1; and Target 2, T2 by Test sgRNA2) (Fig. 1C). Individual constructs were transformed in E. coli, and plasmids isolated from four independent clones were investigated for mutagenesis in targeted regions by Sanger sequencing and the online EditR tool (28). The dual A/C base editing frequency of all the tested iACBE versions was found to be 100% showing concurrent A-to-G and C-to-T edits at one or more positions of As and Cs in the protospacer region (Fig. 1D).
Each ACBE type showed a variable range of editing efficiencies for specific A/C positions in the targeted region. Apparently, iACBE1, iACBE2, and iACBE4 commonly exhibited base-editing features similar to corresponding single-function BEs achieving 100% mutation at a minimum of one A/C position in the target region. While the on-target editing pattern of iACBE3 showed reduced concurrent A-to-G and C-to-T editing activities compared to their single BE counterparts (ABE8e or ABPOBE3A). Considering the combined data estimated in IRI-CCE platform of two tested targets with a minimum 5% base conversion rate, editing window length for ABE8e, ABE9e, PmCDA1, evoCDA1, and APOBEC3A spanned from 3 to 8, 3 to 8, 1 to 7, −6 to 10, −6 to 10, respectively (24). In the case of ACBEs, all four iACBE versions exhibited similar editing window lengths for A-to-G edits to that of ABEs, i.e., positions 3 to 8. C-to-T conversion window positioned from 1 to 7, -1 to 9, 3 to 9, and -1 to 9 for iACBE1 to 4, respectively (Fig. 1D). The lower amount of A/C base editing by iACBE3 was found in the editable window compared to ABE8e (range: 11% to 86% versus 10% to 99%) and APOBEC3A (0% to 36% versus 14% to 100%). Analysis of iACBE2 consisting of catalytically inactive dCas9 fusion also showed similar editing features compared to nCas9-based iACBE2 (Fig. S1). Overall, iACBE2 and iACBE4 showed a broader window and were the most effective iACBEs for installing concurrent A/C mutations among the tested versions. We chose iACBE4 (evoCDA1-ABE9e) for further characterization in subsequent experiments (Fig. 2A).
Evaluation of the editing efficiency by iACBE4 on E. coli genomic sites.
Next, we thoroughly assessed iACBE4 performance for mutating the endogenous target sites in E. coli. For targeting genomic loci, we chose 14 target sites having different A/C sequence contexts across the protospacer regions (Table S1). The pGlpT-driven iACBE4 effectively modified the available A/Cs in the target regions of all the 14 tested sites with variable efficiency (Fig. 2B). Notably, iACBE4 showed a broader editing window length of 21 nucleotides (positions −6 to 15) (Fig. 2C). Higher coediting of A/C was observed in assessed sites at positions 1 to 8. Among them, A-to-G and C-to-T conversions by iACBE4 were highest at positions 4 to 8 and 1 to 9, respectively, within the target region. The editing pattern also confirmed the 100% editing of at least one A or C in all the 14 targets, similarly observed for plasmid targets. Although editing efficacies of base editing systems may vary due to several factors like target site, sequence context, and experimental conditions (29), our results demonstrate the functionality of iACBE4 architecture for co-editing of A/C in the genomic context of E. coli.
iACBE4-mediated multiplex editing allows multisite editing in E. coli.
Multiplex editing includes simultaneous editing of two or more sites and offers a fast way of generating multisite mutants in a single generation. Multiplex editing with single-function BEs was achieved in several organisms, including bacteria and eukaryotes (16, 30, 31). We aimed to examine the potential of iACBE4 for concurrent base editing in multiple sites, which was not investigated in any of the previous ACBE studies. Five different multiplex ACBE systems (iACBE4-M1 to M5) were designed following a single-plasmid component system containing expression cassettes of BE fusions, sgRNAs, and target sites if needed (Fig. 3A). Dual base editing at two target DNA sites (2×) located on either the same gene (rpoB by iACBE4-M1, Fig. 3B); or two different genes (rpoB and rppH by iACBE4-M2) (Fig. 3C); or sites located on the plasmid and genomic site (T1 and rppH by iACBE4-M3, Fig. 3D); was successfully achieved in all the analyzed independent clones. In addition, simultaneous targeting of three genes (galK, rpoB, and rppH) with triple (3×) and quadruple (4×) sgRNAs assembled into iACBE4-M4 (Fig. 3E) and iACBE4-M5 (Fig. 3F), respectively, showed efficient co-editing of A/C positioned in targeted sites. Taken together, an optimized iACBE4 system could be utilized for efficient dual base multisite editing on the same or different genes in E. coli.
Evaluation of PAM-relaxed BE-NG and iACBE-NG tools in bacteria.
The iACBE constructs include nCas9, which requires NGG as a PAM motif. Also, the targeted A/C must be available within the editable window, restricting the choice of targetable loci in the genome for broader use. Considering the strict requirement of PAM as one of the constraints for using nCas9-based BE and iACBE, we replaced nCas9 with nCas9-NG, a nickase form of PAM-relaxed Cas9-NG, which recognizes NGN as a PAM motif in the target sites (25). Analysis of two targets with NGG PAM (T1 and T2) by nCas9-NG derived ABEs (NG-ABE8e, NG-ABE9e) and CBEs (NG-PmCDA1, NG-evoCD1, NG-APOBEC3A) revealed efficient editing in editable windows thereby confirming the compatibility of BE tools with nCas9-NG (Fig. S2). Editing efficacies and editable windows of nCas9-NG-BEs (Fig. S2) were similar to corresponding nCas9-BEs (Fig. 1D), reaching up to 100% in editing windows. Besides, Cas9-NG reported to self-target the sgRNA-expression cassette, possibly raising the off-target issue by producing secondary sgRNAs (26). Native sgRNA scaffold begins with GTT immediately after the protospacer sequence. We found that all the Cas9-NG-BEs recognize GTT as a PAM motif in the sgRNA cassette and induced base editing in the narrower window and a relatively lower percentage than on-target editing (Fig. S2). Surprisingly, PmCDA1-derived NG-CBE showed no self-editing for both the tested sites similar to a recent report in plants (32). These results indicated that regardless of self-editing, nCas9-NG-based BEs enable base editing in E. coli.
Inspired by the data of NG-BEs, we combined highly processive ABE8e-evoCDA1 and ABE9e-evoCDA1 with nCas9-NG to edit the NGN PAM sites generating iACBE2-NG and iACBE4-NG, respectively (Fig. 4; Table S2). We observed similar editing outcomes for iACBE2-NG (Fig. S3) and iACBE4-NG (Fig. 4) at T1 and T2 sites with the NGG motif, and hence, iACBE4-NG was pursued in further work. Target sites (T1 and T2) with NGN PAM (GGG, CGA, CGC, CGT) were cloned alongside desired ACBE components (Fig. 4A to D). To reduce self-editing in NG-based BEs, the modified sgRNA scaffold (esgRNA) was used (Fig. 4C) as described by Qin et al. (26) that preserved a high on-target indel generation rate while alleviating the self-editing in plants. At first, testing of iACBE4-NG with native sgRNA displayed higher co-editing of A/C at on-target and self-targeting regions in the T1 and T2 sites (Fig. 4E and F). For the T1 site, on-target editing against self-editing frequencies of positions 2 to 7 in the editable window with native sgRNA were 90.8% versus 30.8%, 83.8% versus 28.9%, 81.7% versus 26.9%, and 83% versus 23.1% at GGG, CGA, CGC, and CGT PAM motifs, respectively (Table S2). For T2 site, average on-target against self-editing frequencies of positions 1 to 8 with native sgRNA were 89.2% versus 35.6%, 77% versus 33.1%, 73.1% versus 30.4%, and 80.4% versus 25.1% at GGG, CGA, CGC, and CGT PAM motifs in the editable window, respectively (Table S2).
The iACBE4-NG with esgRNA (Fig. 4G) revealed efficient on-target editing at the T1 (Fig. 4H) and T2 (Fig. 4I) sites, while self-editing was significantly reduced. For instance, average on-target and self-editing frequencies by iACBE4-NG with esgRNA for the T1 site were 82.5% versus 0.9%, 68.6% versus 0.9%, 60.5% versus 0.9%, and 74.3% versus 0.8% at GGG, CGA, CGC, and CGT PAM motifs, respectively (Table S2). Also, on-target against self-editing frequencies with esgRNA for the T2 site at GGG, CGA, CGC, and CGT PAM motifs were 72.9% versus 0.9%, 58% versus 1.4%, 48.2% versus 1.1%, and 66.1% versus 1.7%, respectively (Table S2). Overall, using esgRNA with iACBE4-NG could alleviate the self-targeting effect with comparable or slightly lower on-target editing for plasmid target sites in E. coli with the same editing features as native sgRNA (Fig. S4).
Next, we applied the iACBE4-NG tool to edit the five genomic sites comprising native sgRNA and esgRNA with different NGN PAM motifs (Table S1). Among the analyzed genomic sites, there was no specific correlation between on-target and self-target editing frequencies by iACBE4-NG with native sgRNA scaffold (Fig. 5A). For instance, iACBE4-NG with native sgRNA displayed efficient editing at on-target (49.75% to 99% and 15.72% to 100%) and self-target (0% to 22% and 0% to 32%) regions for sites 1 and 3, respectively. At the same time, the iACBE4-NG construct showed high on-target editing but no self-targeting for sites 2, 4, and 5. Although we observed no self-editing in targeted genomic loci by iACBE4-NG with modified esgRNA, it also dramatically reduced the on-target A/C co-editing. All the five genomic sites by iACBE4-NG with esgRNA revealed a significant reduction in on-target editing activities (Fig. 5B).
Overall, iACBE-NG data indicate that the dual base editing tools comprising nCas9-NG and esgRNA are poorly compatible for modifying genomic loci, consistent with the recent ABE-NG reports in rice (33). Though the application of iACBE-NG greatly expands the choice of target sites to induce the dual base editing in E. coli, further research is needed to achieve high on-targeting with reduced or no self-editing with PAM-relaxed nucleases.
Evolution of rifampicin-resistant protein (rpoB) of E. coli by iACBE systems.
Engineered iACBE-based tools provide an excellent opportunity to mutate a gene of interest in native genetic background and simultaneously validate the gain-of-function. To demonstrate iACBE potential for in situ protein evolution studies, we chose the rpoB gene, which is implicated in rifampicin resistance (RifR) in E. coli and several pathogenic microbes (34–38), suggestive of its clinical significance. The rpoB gene is highly conserved among bacteria and encodes the beta-subunit of DNA-dependent RNA polymerase. Several point (single amino-acid) mutations in RpoB are well-documented, conferring RifR in E. coli (35–38) (Fig. 6A; Table S3). The mode of action involves the transcription inhibition via rifampicin binding to the catalytic core of the RpoB enzyme. Three zones in RpoB are considered RifR-determining region (RRDR) domains that include most of the known RifR-related point mutations (37).
The seven sites distributed across the three RRDR regions were randomly targeted (Fig. 6B). Sites 1 to 4 included protospacers with NGG PAM and sites 5 to 7 with NGN PAM. Seven independent iACBE4 constructs comprising desired nCas9 form (sites 1 to 4, nCas9; sites 5 to 7, nCas9-NG) were transformed into E. coli DH5-alpha strain and obtained clones were further screened for RifR (Fig. 6C). iACBE4-NG with native sgRNA or esgRNA were used for targeting sites 5 to 7. The multisite editing constructs comprising two (site 1 and 2) or three (site 1, 2, and 3) sgRNAs targeting the rpoB gene were also investigated, but clones mutated only in site 1 were recovered in the RifR screening, indicating mutants generated by multisite editing (at sites 2 and 3) were sensitive to rifampicin and did not survive. Single colony cultures were streaked or dotted on LB plates containing 50 μg/mL rifampicin to verify the RifR (Fig. 6D and E). The genotypes of independent RifR colonies that appeared on rifampicin-containing plates were determined by Sanger sequencing, and mutations with amino acid changes were mapped across the targeted sites. Among the tested regions, site 6 yielded no RifR clones. We identified a single or cluster of multiple amino acid substitutions responsible for RifR (Fig. 6F to G; Table S3). These results demonstrate the practical use of iACBE systems for saturated mutagenesis of the targeted genetic locus in E. coli cells, facilitating functional characterization of induced mutations thereof.
Genome-wide off-target evaluation of the iACBE systems in E. coli.
Precise editing without potential off-targets is critical for successfully applying CRISPR-based tools for genome engineering. To investigate potential off-targets induced by the iACBE systems, we carried out whole-genome sequencing (WGS) and unbiased profiling of single nucleotide variations (SNVs). One clone of each event of chromosome target (galK) editing from iACBE4 and iACBE4-NG with either native or esgRNA scaffold was analyzed using the WGS approach to assess the gRNA-independent SNVs (Fig. 7; Table S4). Additionally, WGS analysis of laboratory parental DH5α strain was performed to know the background noise of SNVs. We reasoned that aligning the WGS reads of each edited clone to the parental DH5α genome sequence allows a more precise evaluation of iACBE-specific off-targets, as performed previously (17, 30). SNV analysis revealed seven (laboratory DH5α strain), six (iACBE4 with galK gRNA1), 27 (iACBE4-NG-galK NG-gRNA1-native sgRNA), and four (iACBE4-NG-galK NG-gRNA1- esgRNA scaffold) off-target SNVs, resulting in two, four, 13, and two nonsynonymous mutations, respectively (Fig. 7; Table S4). These data are in line with previous reports on single BE-caused off-targets in other bacteria (17, 30). Therefore, genome-wide evaluation data indicate that our iACBE4 tool is a relatively safer GE system for E. coli.
DISCUSSION
We engineered the dual base editing systems and applied for the first time in bacteria, employing the fusion of highly active ABE and CBE variants. Optimized iACBE toolset can be used for programmable dual base editing with high efficiencies at single or multisite targets in E. coli. The iACBE4-NG generated by using engineered Cas9-NG nickase (nCas9-NG), which recognizes the NGN PAM motif, further expands the target range of the iACBE toolset. In addition, characterized iACBE system exhibits high editing efficiencies up to 100%, which considerably cuts the workload of mutant verification. Unlike CRISPR/Cas9, which needs DSB generation, the engineered iACBE system is self-sufficient in introducing base conversion independent of NHEJ.
The iACBE-based screening of rpoB mutants related to RifR demonstrates its use in exploring the role of induced mutations in protein studies. In earlier studies, single amino-acid modifications in RpoB conferring RifR in E. coli are well-documented (35–38). We identified single and clusters of multiple amino-acid substitutions in RpoB imparting RifR (Table S3). For example, our data reveal nine RifR mutants with single amino-acid substitutions. Among those, five mutations overlapped with the previous reports (S512F, D516N, H526Y, S531F, P564L), and four mutations were unique (S508F, K527R, H551Y, Q580W). Additionally, nine clusters of multiple substitutions were identified, expanding the rpoB mutant library conferring RifR phenotype (Table S3), which may help to understand the structural aspects of antibiotic resistance. We note that the rpoB is an essential gene; therefore, it is possible that some of the editing events are actually not possible to be effectively selected during competitive growth between rpoB mutants against wild-type cells in the editing pool.
Recently, the ABE8e variant in ACBE design showed higher synchronized A/C editing efficiency in mammalian cells (10, 12), which is consistent with the iACBE data. Combining PAM-relaxed Cas9 versions and highly active BE deaminases broadens the scope using ACBE tools. PAM-relaxed Cas9 variants could be prone to two types of unwanted off-target effects. First, PAM-relaxed Cas9 could produce off-target editing at sgRNA-dependent loci. Second, sites recognizable by secondary sgRNAs created through self-editing increase off-targeting chances. Self-targeting is problematic due to the secondary gRNA production, but it may also reduce the on-target effect. Although there is no direct report about using the CRISPR tool based on Cas9-NG with esgRNA in bacteria, several studies in plants have evaluated the Cas9-NG-mediated indel generation and BE activities with esgRNA to alleviate the self-editing (26, 33, 39). The esgRNA may be compatible with Cas9-NG for generating indels, but it is poorly compatible with BE-NG and ACBE-NG. We found that the iACBE4-NG with esgRNA displayed similar on-target editing activities and decreased self-targeting on plasmid target sites. However, we observed that the esgRNA is not compatible with the iACBE tool for targeting genomic sites and dramatically reduced on-target efficiency.
In the case of the PmCDA1-based CBE-NG tool, we did not observe self-editing in bacteria which is consistent with the previous reports of PmCDA1-based CBEs with nCas9 (40), Cas9-NG, and SpRY (32) in plant systems. Hence, when desirable, we propose PmCDA1 as a first-choice CBE with PAM-relaxed Cas9 variants. Moreover, in the present and earlier studies, BE-NG- or iACBE4-NG-mediated editing was not diminished at estimated target sites. Therefore, despite the self-editing, BE and ACBE tools based on Cas9-NG may edit the sites with non-NGG PAM expanding the targeting scope of bacterial genome editing.
SpRY is another PAM-relaxed Cas9 variant that recognizes NYN (Y- C or T) and NRN (R- A or G) as PAM motifs (41). The sgRNAs targeting sites with NYN PAM motifs generated higher self-editing frequencies than on-target editing by SpRY in plants (42, 43). Also, combining esgRNA with SpRY is of no help because it causes a substantial reduction in on-target activities and higher self-editing in rice (44), which is an obvious outcome with GCC as PAM in esgRNA sequence. Therefore, we believe that the SpRY-mediated iACBE tools may induce higher self-editing in sgRNA cassettes than corresponding Cas9-NG-based BEs. Thus, it would be worthy of engineering specialized sgRNAs or Cas9 variants when implementing the PAM-relaxed BE and iACBE toolbox in bacteria and other organisms.
Prime editor (PE), another CRISPR-based GE tool (45), can theoretically introduce all kinds of base substitutions. The efficiency of PEs is low in different organisms, including E. coli (46), and predefined mutation sets can only be introduced. Therefore, better ACBEs that can induce the randomized combination of multiple base substitutions would be excellent endowments for protein evolution studies. Also, glycosylase base editor (CGBE), a new BE tool, introduces C-to-G, C-to-A, and C-to-T mutations depending on the DNA repair responses in different organisms (47–50). Incorporating CGBE into dual base editing systems would further extend the diversity of saturated mutagenesis, as described in a recent report (51). In the future, essential genes in their natural genomic context will be attractive targets by iACBE-mediated GE because of their importance in evolution, metabolic engineering, development of antibiotic resistance, and newer strain designs.
Conclusions.
We developed iACBE, the first report of a dual base editing system for bacterial applications that provides a versatile tool for the induction of synchronized A-to-G and C-to-T mutations at the same target site. The successful application of iACBE4 for multiplex editing allowed concurrent diversification of multiple targets from plasmid or genomic sites. This study also reveals molecular insights into the compatibility of PAM-relaxed iACBE designs with native and esgRNA scaffolds for editing target sites with NGN PAM motifs. The iACBE toolset could be applied to introduce concurrent A/C base substitutions within the desired target genes in the genetic context facilitating functional analysis of constructed mutants in synthetic biology and basic microbiological research.
MATERIALS AND METHODS
E. coli strains and culture conditions.
The E. coli 10-beta strain was used for cloning of different biopart modules. The list of synthesized primers, generated plasmids, and biopart sequences used in the current study are provided in Table S5. Cloning and BE analyses were conducted in E. coli 10-beta and DH5α strains. The E. coli cells were grown in Luria-Bertani (LB) broth with suitable antibiotics.
E. coli strains were grown aerobically at 37°C in LB liquid medium (for 1 L, 5 g yeast extract, 10 g tryptone, and 10 g NaCl) or on LB-agar plates supplemented with desired antibiotics when required.
Plasmid constructs, sgRNA designing, and cloning.
The necessary bioparts such as different Cas9 forms, deaminases, UGI, promoters, and terminators were amplified by traditional PCR employing a high-fidelity version of Phusion DNA polymerase (Thermo Fisher Scientific, Waltham, MA). Golden Gate assembly (52) and MoClo (53) kits were utilized for cloning various plasmid vectors, following the instructions provided in the kit protocols. Type IIS enzymes, BpiI and BsaI, were mainly used in the digestion-ligation process. BsaI and BpiI recognition sites were removed from bioparts for cloning in the Golden Gate system.
The pGlpT promoter and TerL3S2P21 terminator (24) were used to express single and dual base editing systems. The DNA sequences are provided in Table S5. The synthesized ABE8e (8-point mutations in ABE7.10) from Bioneer Co. (Daejon, South Korea) was used as a template in PCR to obtain the ABE9e (additional V82S/Q154R) variant by site-directed mutagenesis. Three CBE deaminase modules along with linker sequences were prepared by PCR using plasmid templates ordered from the Addgene, namely, PmCDA1 (Addgene #79620) (5), evoCDA1 (Addgene #122608) (21), and APOBEC3A (Addgene #119770) (27), respectively. The C-terminal PmCDA1-1xUGI was cloned with nCas9. The ABE8e or ABE9e with evoCDA1 or A3A, including the XTEN linker, were tethered at the N-terminal of nCas9. The 2xUGI module was prepared using (Addgene #122608) as a PCR template to fuse with the desired CBEs and iACBEs containing evoCDA1 and APOBEC3A.
The plant AtU6 promoter was amplified from (Addgene #46968) to express sgRNAs with the native (sgRNA) or modified (esgRNA) scaffold. The nCas9(D10A) and nCas9-NG(D10A) were cloned by the PCR-cloning strategy using template plasmids (Addgene #49771) and (Addgene #125616), respectively. Required sgRNA sequences with native or modified scaffold (esgRNA) were PCR amplified using a plasmid template (Addgene #46966) for further cloning and combined with pAtU6 for expression in E. coli cells. Target regions (T1 and T2) with appropriate PAM motifs were cloned into a universal target-acceptor plasmid (24) using the Type IIS BsmBI enzyme by digestion-ligation method.
Bacterial transformation, plasmid isolation, and Sanger sequencing.
Cloning steps were performed by ligating the various biopart modules, followed by heat shock-mediated E. coli transformation. The 10-beta E. coli strains were used for cloning steps. Briefly, competent cells and digestion-ligation mix were exposed to heat shock at 45°C for 1 min, and 1 mL LB broth was added after incubating the tubes on ice for 3 min. Then, after shaking (180 rpm) the culture for 1 h at 37°C, it was spread on the LB agar (1.5%) plates containing appropriate antibiotics. Individual colonies appeared on LB-agar plates (with necessary antibiotics) after 18 h of incubation at 37°C were subjected to plasmid isolation and Sanger sequencing analysis at Cosmogentech Ltd. (Seoul, South Korea). For targeted mutagenesis assay by BE and iACBE systems, the E. coli cells were transformed with the desired plasmid vectors by heat shock method. Plasmid vectors consisting of BE or ACBE components without sgRNA-expression cassettes were used as control. As described earlier, independent clones from the LB-agar plate (with necessary antibiotics) were cultured in 3 mL antibiotic-containing LB media and allowed to grow at 37°C for a 24-h period. Plasmid DNA was purified for verification by restriction enzyme digestion, and sequencing analysis using Plasmid Mini-Prep Kit procured from BioFact Co. Ltd. (Daejeon, South Korea).
Generation of rpoB mutant library and rifampicin resistance (RifR) assay.
The desired sgRNA-expression cassettes were cloned together with iACBE4 or iACBE4-NG depending on the target site-PAM compositions in the rpoB gene. In particular, sites 1 to 4 consisted of sgRNA spacers with NGG and sites 5 to 7 with NGN PAM. Seven independent iACBE4 constructs were transformed into E. coli DH5-alpha strain and individual clones were grown on LB-agar plates containing kanamycin (50 μg/mL) were further screened for RifR. Single colony cultures were streaked or dotted on LB plates containing 50 μg/mL rifampicin to verify the RifR. Also, multisite editing iACBE4 constructs comprising two (site 1 and 2) or three (site 1, 2, and 3) sgRNAs were screened as described earlier. The genotypes of individual RifR colonies that grew on rifampicin-containing plates were determined by Sanger sequencing, and mutations with amino acid changes were mapped across the targeted sites.
Analysis of base substitution activities.
To map the mutagenesis patterns in synthetic targets (T1 and T2) by Sanger sequencing, Plasmid Mini-Prep Kit was used for extracting plasmid DNA purchased from BioFact Co. Ltd. (Daejeon, South Korea). To investigate the mutations induced by different iACBE types at E. coli genomic sites, the colonies were randomly picked, cultured in LB broth with necessary antibiotics, and then incubated for 24 h at 180 rpm at 37°C. The genetic fragments were amplified using target region-specific oligos by PCR method and then subjected to Sanger sequencing. SnapGene software was used for Sanger data analysis (GSL Biotech; available at snapgene.com). The base editing efficiencies were estimated by the proportion of nonedited to mutated clones from the analyzed colonies. The frequency of base change (from C-to-T and A-to-G) was calculated using the EditR, an online base-editing analysis tool (28). The data were statistically analyzed and plotted in GraphPad Prism 9.3.1 (www.graphpad.com, last accessed on April 3, 2022).
Genome-wide evaluation of off-targets of iACBE4 systems.
To analyze the off-target effects induced by the iACBE system, total genomic DNA was extracted from the laboratory DH5α strain and three edited clones (iACBE4 with galK gRNA1, iACBE4-NG with galK NG-gRNA1 containing native or esgRNA scaffold) using QIAamp DNA minikit (Qiagen, Germany). Whole-genome sequencing was performed at SEEDERS Inc. (SEEDERS, Daejeon, South Korea) with the Illumina NGS platform. Raw sequence reads were aligned to the reference E. coli genome (NCBI accession: GCA_022221385.1) or laboratory DH5α strain. The Illumina reads were preprocessed using Trimmomatic (v. 0.39) (54). SEEDERS in-house script (55) was utilized to map SNVs using BWA (0.7.17 r1188) and SAMtools (v 0.1.16) programs (56, 57). The alignment accuracy to the reference genome was 99.84% for all four samples. The SNV for a nucleobase was counted as homozygous (read rate 90%) or heterozygous (40%≤ read rate ≤60%) based on the read rate. Mutation calls found in all the samples and parent DH5α were excluded and not considered off-targets.
Data availability.
The Illumina sequencing data generated for off-target evaluation has been deposited to NCBI: NCBI BioProject PRJNA906298; SRA accessions SRR22439416, SRR22452371, SRR22452578, and SRR22455127.
ACKNOWLEDGMENTS
This work was supported by the National Research Foundation of Korea (grants NRF 2020M3A9I4038352, 2021R1A5A8029490, 2020R1A6A1A03044344, and 2021R1I1A3057067), and by the Program for New Plant Breeding Techniques, Rural Development Administration (NBT, PJ01686702).
R.M.S. and J.Y.-K. conceived the idea. R.M.S. researched and designed the manuscript structure. R.M.S. wrote the initial draft. R.M.S., D.P., and J.Y.-K. contributed to writing and editing. All authors have made direct and intellectual contributions to the editing before final submission and approved the manuscript.
We declare no conflicts of interest that might be perceived as affecting the objectivity of this paper.
Contributor Information
Rahul Mahadev Shelake, Email: rahultnau@gmail.com.
Jae-Yean Kim, Email: kimjy@gnu.ac.kr.
Jeremy M. Rock, The Rockefeller University
Sabine Ehrt, Weill Cornell Medicine.
REFERENCES
- 1.Shelake RM, Pramanik D, Kim J-Y. 2019. Exploration of plant-microbe interactions for sustainable agriculture in CRISPR era. Microorganisms 7:269. doi: 10.3390/microorganisms7080269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Anzalone AV, Koblan LW, Liu DR. 2020. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol 38:824–844. doi: 10.1038/s41587-020-0561-9. [DOI] [PubMed] [Google Scholar]
- 3.Pramanik D, Shelake RM, Kim MJ, Kim J-Y. 2021. CRISPR-mediated engineering across the central dogma in plant biology for basic research and crop improvement. Mol Plant 14:127–150. doi: 10.1016/j.molp.2020.11.002. [DOI] [PubMed] [Google Scholar]
- 4.Li C, Zhang R, Meng X, Chen S, Zong Y, Lu C, Qiu JL, Chen YH, Li J, Gao C. 2020. Targeted, random mutagenesis of plant genes with dual cytosine and adenine base editors. Nat Biotechnol 38:875–882. doi: 10.1038/s41587-019-0393-7. [DOI] [PubMed] [Google Scholar]
- 5.Nishida K, Arazoe T, Yachie N, Banno S, Kakimoto M, Tabata M, Mochizuki M, Miyabe A, Araki M, Hara KY, Shimatani Z, Kondo A. 2016. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353:aaf8729. doi: 10.1126/science.aaf8729. [DOI] [PubMed] [Google Scholar]
- 6.Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. 2016. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533:420–424. doi: 10.1038/nature17946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, Liu DR. 2017. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551:464–471. doi: 10.1038/nature24644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Grünewald J, Zhou R, Lareau CA, Garcia SP, Iyer S, Miller BR, Langner LM, Hsu JY, Aryee MJ, Joung JK. 2020. A dual-deaminase CRISPR base editor enables concurrent adenine and cytosine editing. Nat Biotechnol 38:861–864. doi: 10.1038/s41587-020-0535-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sakata RC, Ishiguro S, Mori H, Tanaka M, Tatsuno K, Ueda H, Yamamoto S, Seki M, Masuyama N, Nishida K, Nishimasu H, Arakawa K, Kondo A, Nureki O, Tomita M, Aburatani H, Yachie N. 2020. Base editors for simultaneous introduction of C-to-T and A-to-G mutations. Nat Biotechnol 38:865–869. doi: 10.1038/s41587-020-0509-0. [DOI] [PubMed] [Google Scholar]
- 10.Tao W, Liu Q, Huang S, Wang X, Qu S, Guo J, Ou D, Li G, Zhang Y, Xu X, Huang X. 2021. CABE-RY: a PAM-flexible dual-mutation base editor for reliable modeling of multi-nucleotide variants. Mol Ther Nucleic Acids 26:114–121. doi: 10.1016/j.omtn.2021.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Xie J, Huang X, Wang X, Gou S, Liang Y, Chen F, Li N, Ouyang Z, Zhang Q, Ge W, Jin Q, Shi H, Zhuang Z, Zhao X, Lian M, Wang J, Ye Y, Quan L, Wu H, Wang K, Lai L. 2020. ACBE, a new base editor for simultaneous C-to-T and A-to-G substitutions in mammalian systems. BMC Biol 18:1–14. doi: 10.1186/s12915-020-00866-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Xiong X, Li Z, Liang J, Liu K, Li C, Li J-F. 2022. A cytosine base editor toolkit with varying activity windows and target scopes for versatile gene manipulation in plants. Nucleic Acids Res 50:3565–3580. doi: 10.1093/nar/gkac166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Xu R, Kong F, Qin R, Li J, Liu X, Wei P. 2021. Development of an efficient plant dual cytosine and adenine editor. J Integr Plant Biol 63:1600–1605. doi: 10.1111/jipb.13146. [DOI] [PubMed] [Google Scholar]
- 14.Zhang X, Zhu B, Chen L, Xie L, Yu W, Wang Y, Li L, Yin S, Yang L, Hu H, Han H, Li Y, Wang L, Chen G, Ma X, Geng H, Huang W, Pang X, Yang Z, Wu Y, Siwko S, Kurita R, Nakamura Y, Yang L, Liu M, Li D. 2020. Dual base editor catalyzes both cytosine and adenine base conversions in human cells. Nat Biotechnol 38:856–860. doi: 10.1038/s41587-020-0527-y. [DOI] [PubMed] [Google Scholar]
- 15.Li M, Huo Y, Guo S. 2022. CRISPR-mediated base editing: from precise point mutation to genome-wide engineering in nonmodel microbes. Biology (Basel) 11:571. doi: 10.3390/biology11040571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Banno S, Nishida K, Arazoe T, Mitsunobu H, Kondo A. 2018. Deaminase-mediated multiplex genome editing in Escherichia coli. Nat Microbiol 3:423–429. doi: 10.1038/s41564-017-0102-6. [DOI] [PubMed] [Google Scholar]
- 17.Rodrigues SD, Karimi M, Impens L, van Lerberge E, Coussens G, Aesaert S, Rombaut D, Holtappels D, Ibrahim HMM, van Montagu M, Wagemans J, Jacobs TB, de Coninck B, Pauwels L. 2021. Efficient CRISPR-mediated base editing in Agrobacterium spp. Proc Natl Acad Sci USA 118:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yu S, Price MA, Wang Y, Liu Y, Guo Y, Ni X, Rosser SJ, Bi C, Wang M. 2020. CRISPR-dCas9 mediated cytosine deaminase base editing in Bacillus subtilis. ACS Synth Biol 9:1781–1789. doi: 10.1021/acssynbio.0c00151. [DOI] [PubMed] [Google Scholar]
- 19.Zhao Y, Tian J, Zheng G, Chen J, Sun C, Yang Z, Zimin AA, Jiang W, Deng Z, Wang Z, Lu Y. 2020. Multiplex genome editing using a dCas9-cytidine deaminase fusion in Streptomyces. Sci China Life Sci 63:1053–1062. doi: 10.1007/s11427-019-1559-y. [DOI] [PubMed] [Google Scholar]
- 20.Kim MS, Kim H-R, Jeong D-E, Choi S-K. 2021. Cytosine base editor-mediated multiplex genome editing to accelerate discovery of novel antibiotics in Bacillus subtilis and Paenibacillus polymyxa. Front Microbiol 12:691839. doi: 10.3389/fmicb.2021.691839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Thuronyi BW, Koblan LW, Levy JM, Yeh W-H, Zheng C, Newby GA, Wilson C, Bhaumik M, Shubina-Oleinik O, Holt JR, Liu DR. 2019. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat Biotechnol 37:1070–1079. doi: 10.1038/s41587-019-0193-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yan D, Ren B, Liu L, Yan F, Li S, Wang G, Sun W, Zhou X, Zhou H. 2021. High-efficiency and multiplex adenine base editing in plants using new TadA variants. Mol Plant 14:722–731. doi: 10.1016/j.molp.2021.02.007. [DOI] [PubMed] [Google Scholar]
- 23.Richter MF, Zhao KT, Eton E, Lapinaite A, Newby GA, Thuronyi BW, Wilson C, Koblan LW, Zeng J, Bauer DE, Doudna JA, Liu DR. 2020. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat Biotechnol 38:883–891. doi: 10.1038/s41587-020-0453-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shelake RM, Pramanik D, Kim JY. 2022. In vivo rapid investigation of CRISPR-based base editing components in Escherichia coli (IRI-CCE): a platform for evaluating base editing tools and their components. Int J Mol Sci 23. doi: 10.3390/ijms23031145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nishimasu H, Shi X, Ishiguro S, Gao L, Hirano S, Okazaki S, Noda T, Abudayyeh OO, Gootenberg JS, Mori H, Oura S, Holmes B, Tanaka M, Seki M, Hirano H, Aburatani H, Ishitani R, Ikawa M, Yachie N, Zhang F, Nureki O. 2018. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 361:1259–1262. doi: 10.1126/science.aas9129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Qin R, Li J, Liu X, Xu R, Yang J, Wei P. 2020. SpCas9-NG self-targets the sgRNA sequence in plant genome editing. Nat Plants 6:197–201. doi: 10.1038/s41477-020-0603-9. [DOI] [PubMed] [Google Scholar]
- 27.Zong Y, Song Q, Li C, Jin S, Zhang D, Wang Y, Qiu J-L, Gao C. 2018. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A. Nat Biotechnol 36:950–953. doi: 10.1038/nbt.4261. [DOI] [PubMed] [Google Scholar]
- 28.Kluesner MG, Nedveck DA, Lahr WS, Garbe JR, Abrahante JE, Webber BR, Moriarity BS. 2018. EditR: a method to quantify base editing from sanger sequencing. Crispr J 1:239–250. doi: 10.1089/crispr.2018.0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Huang TP, Newby GA, Liu DR. 2021. Precision genome editing using cytosine and adenine base editors in mammalian cells. Nat Protoc 16:1089–1128. doi: 10.1038/s41596-020-00450-9. [DOI] [PubMed] [Google Scholar]
- 30.Tong Y, Whitford CM, Robertsen HL, Blin K, Jørgensen TS, Klitgaard AK, Gren T, Jiang X, Weber T, Lee SY. 2019. Highly efficient DSB-free base editing for Streptomycetes with CRISPR-BEST. Proc Natl Acad Sci USA 116:20366–20375. doi: 10.1073/pnas.1913493116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sun J, Lu L-B, Liang T-X, Yang L-R, Wu J-P. 2020. CRISPR-assisted multiplex base editing system in Pseudomonas putida KT2440. Front Bioeng Biotechnol 8:905. doi: 10.3389/fbioe.2020.00905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wu Y, Ren Q, Zhong Z, Liu G, Han Y, Bao Y, Liu L, Xiang S, Liu S, Tang X, Zhou J, Zheng X, Sretenovic S, Zhang T, Qi Y, Zhang Y. 2022. Genome-wide analyses of PAM-relaxed Cas9 genome editors reveal substantial off-target effects by ABE8e in rice. Plant Biotechnol J doi: 10.1111/pbi.13838.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tan J, Zeng D, Zhao Y, Wang Y, Liu T, Li S, Xue Y, Luo Y, Xie X, Chen L, Liu Y-G, Zhu Q. 2022. PhieABEs: a PAM-less/free high-efficiency adenine base editor toolbox with wide target scope in plants. Plant Biotechnol J 20:934–943. doi: 10.1111/pbi.13774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Xiao M, Zhu X, Xu H, Tang J, Liu R, Bi C, Fan F, Zhang X. 2017. A novel point mutation in RpoB improves osmotolerance and succinic acid production in Escherichia coli. BMC Biotechnol 17:10. doi: 10.1186/s12896-017-0337-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Garibyan L, Huang T, Kim M, Wolff E, Nguyen A, Nguyen T, Diep A, Hu K, Iverson A, Yang H, Miller JH. 2003. Use of the rpoB gene to determine the specificity of base substitution mutations on the Escherichia coli chromosome. DNA Repair (Amst) 2:593–608. doi: 10.1016/S1568-7864(03)00024-7. [DOI] [PubMed] [Google Scholar]
- 36.Jin DJ, Gross CA. 1988. Mapping and sequencing of mutations in the Escherichia coli rpoB gene that lead to rifampicin resistance. J Mol Biol 202:45–58. doi: 10.1016/0022-2836(88)90517-7. [DOI] [PubMed] [Google Scholar]
- 37.Choudhury A, Fenster JA, Fankhauser RG, Kaar JL, Tenaillon O, Gill RT. 2020. CRISPR/Cas9 recombineering-mediated deep mutational scanning of essential genes in Escherichia coli. Mol Syst Biol 16:e9265. doi: 10.15252/msb.20199265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Jatsenko T, Tover A, Tegova R, Kivisaar M. 2010. Molecular characterization of Rif(r) mutations in Pseudomonas aeruginosa and Pseudomonas putida. Mutat Res 683:106–114. doi: 10.1016/j.mrfmmm.2009.10.015. [DOI] [PubMed] [Google Scholar]
- 39.Zeng D, Liu T, Tan J, Zhang Y, Zheng Z, Wang B, Zhou D, Xie X, Guo M, Liu Y-G, Zhu Q. 2020. PhieCBEs: plant high-efficiency cytidine base editors with expanded target range. Mol Plant 13:1666–1669. doi: 10.1016/j.molp.2020.11.001. [DOI] [PubMed] [Google Scholar]
- 40.Ren Q, Sretenovic S, Liu G, Zhong Z, Wang J, Huang L, Tang X, Guo Y, Liu L, Wu Y, Zhou J, Zhao Y, Yang H, He Y, Liu S, Yin D, Mayorga R, Zheng X, Zhang T, Qi Y, Zhang Y. 2021. Improved plant cytosine base editors with high editing activity, purity, and specificity. Plant Biotechnol J 19:2052–2068. doi: 10.1111/pbi.13635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Walton RT, Christie KA, Whittaker MN, Kleinstiver BP. 2020. Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science 368:290–296. doi: 10.1126/science.aba8853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ren Q, Sretenovic S, Liu S, Tang X, Huang L, He Y, Liu L, Guo Y, Zhong Z, Liu G, Cheng Y, Zheng X, Pan C, Yin D, Zhang Y, Li W, Qi L, Li C, Qi Y, Zhang Y. 2021. PAM-less plant genome editing using a CRISPR-SpRY toolbox. Nat Plants 7:25–33. doi: 10.1038/s41477-020-00827-4. [DOI] [PubMed] [Google Scholar]
- 43.Ren J, Meng X, Hu F, Liu Q, Cao Y, Li H, Yan C, Li J, Wang K, Yu H, Wang C. 2021. Expanding the scope of genome editing with SpG and SpRY variants in rice. Sci China Life Sci 64:1784–1787. doi: 10.1007/s11427-020-1883-5. [DOI] [PubMed] [Google Scholar]
- 44.Xu Z, Kuang Y, Ren B, Yan D, Yan F, Spetz C, Sun W, Wang G, Zhou X, Zhou H. 2021. SpRY greatly expands the genome editing scope in rice with highly flexible PAM recognition. Genome Biol 22:6. doi: 10.1186/s13059-020-02231-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, Liu DR. 2019. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576:149–157. doi: 10.1038/s41586-019-1711-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tong Y, Jørgensen TS, Whitford CM, Weber T, Lee SY. 2021. A versatile genetic engineering toolkit for E. coli based on CRISPR-prime editing. Nat Commun 12:5206. doi: 10.1038/s41467-021-25541-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Koblan LW, Arbab M, Shen MW, Hussmann JA, Anzalone AV, Doman JL, Newby GA, Yang D, Mok B, Replogle JM, Xu A, Sisley TA, Weissman JS, Adamson B, Liu DR. 2021. Efficient C•G-to-G•C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nat Biotechnol 39:1414–1425. doi: 10.1038/s41587-021-00938-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chen L, Park JE, Paa P, Rajakumar PD, Prekop H-T, Chew YT, Manivannan SN, Chew WL. 2021. Programmable C:G to G:C genome editing with CRISPR-Cas9-directed base excision repair proteins. Nat Commun 12:1384. doi: 10.1038/s41467-021-21559-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhao D, Li J, Li S, Xin X, Hu M, Price MA, Rosser SJ, Bi C, Zhang X. 2021. Glycosylase base editors enable C-to-A and C-to-G base changes. Nat Biotechnol 39:35–40. doi: 10.1038/s41587-020-0592-2. [DOI] [PubMed] [Google Scholar]
- 50.Kurt IC, Zhou R, Iyer S, Garcia SP, Miller BR, Langner LM, Grünewald J, Joung JK. 2021. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat Biotechnol 39:41–46. doi: 10.1038/s41587-020-0609-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liang Y, Xie J, Zhang Q, Wang X, Gou S, Lin L, Chen T, Ge W, Zhuang Z, Lian M, Chen F, Li N, Ouyang Z, Lai C, Liu X, Li L, Ye Y, Wu H, Wang K, Lai L. 2022. AGBE: a dual deaminase-mediated base editor by fusing CGBE with ABE for creating a saturated mutant population with multiple editing patterns. Nucleic Acids Res 50:5384–5399. doi: 10.1093/nar/gkac353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Engler C, Youles M, Gruetzner R, Ehnert T-M, Werner S, Jones JDG, Patron NJ, Marillonnet S. 2014. A golden gate modular cloning toolbox for plants. ACS Synth Biol 3:839–843. doi: 10.1021/sb4001504. [DOI] [PubMed] [Google Scholar]
- 53.Weber E, Engler C, Gruetzner R, Werner S, Marillonnet S. 2011. A modular cloning system for standardized assembly of multigene constructs. PLoS One 6:e16765. doi: 10.1371/journal.pone.0016765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bolger AM, Lohse M, Usadel B. 2014. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30:2114–2120. doi: 10.1093/bioinformatics/btu170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kim JE, Oh SK, Lee JH, Lee BM, Jo SH. 2014. Genome-wide SNP calling using next generation sequencing data in tomato. Mol Cells 37:36–42. doi: 10.14348/molcells.2014.2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R, 1000 Genome Project Data Processing Subgroup . 2009. The sequence alignment/map format and SAMtools. Bioinformatics 25:2078–2079. doi: 10.1093/bioinformatics/btp352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Li H. 2013. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv.
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The Illumina sequencing data generated for off-target evaluation has been deposited to NCBI: NCBI BioProject PRJNA906298; SRA accessions SRR22439416, SRR22452371, SRR22452578, and SRR22455127.