Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)—CRISPR-associated protein (Cas) and base editors are fundamental tools in plant genome editing. Cas9 from Streptococcus pyogenes (SpCas9), recognizing an NGG protospacer adjacent motif (PAM), is a widely used nuclease for genome editing in living cells. Cas12a nucleases, targeting T-rich PAMs, have also been recently demonstrated in several plant species. Furthermore, multiple Cas9 and Cas12a engineered variants and orthologs, with different PAM recognition sites, editing efficiencies and fidelity, have been explored in plants. These RNA-guided sequence-specific nucleases (SSN) generate double-stranded breaks (DSBs) in DNA, which trigger non-homologous end-joining (NHEJ) repair or homology-directed repair (HDR), resulting in insertion and deletion (indel) mutations or precise gene replacement, respectively. Alternatively, genome editing can be achieved by base editors without introducing DSBs. So far, several base editors have been applied in plants to introduce C-to-T or A-to-G transitions, but they are still undergoing improvement in editing window size, targeting scope, off-target effects in DNA and RNA, product purity and overall activity. Here, we summarize recent progress on the application of Cas nucleases, engineered Cas variants and base editors in plants.
Keywords: CRISPR, SpCas9, Cas12a, Cas12b, PAM, Cytidine/adenine base editors
Introduction
CRISPR (Clustered regularly interspaced short palindromic repeats)—Cas (CRISPR associated protein), derived from a bacterial innate immune system, is a leading sequence-specific nuclease (SSN) used for genome editing. The CRISPR locus is characterized by direct repeats of varying sizes (21–48 bp), separated by non-repetitive spacer sequences of defined sizes (26–72 bp), in bacterial and archaeal genomes (Jansen et al. 2002; Haft et al. 2005; Rath et al. 2015). Spacer sequences can be transcribed and processed into individual CRISPR-RNAs (crRNAs), which represent the basic component of the archaeal and bacterial adaptive immune response against invading genetic elements (e.g. phage DNA and plasmids). The second key component of CRISPR-mediated adaptive immunity is Cas proteins, encoded by Cas genes (Jansen et al. 2002). Cas endonucleases are expressed from a locus adjacent to the CRISPR arrays. Even though Cas proteins exhibit polymorphism within genomes, they are all known to have the ability to interact with nucleic acids. Cas core genes have been well characterized and shown to mainly encode nucleases, helicases or RecB-family exonucleases (Haft et al. 2005). Based on the signature Cas genes and the organization of the CRISPR loci, the CRISPR-Cas system is classified into two classes and six types (Type I to Type VI), each of which has several subtypes (~ 20 subtypes) (McGinn and Marraffini 2019). While CRISPR systems of types I, III and IV (Class 1) involve multiple Cas proteins to degrade DNA/RNA, Type II, V and VI (Class 2) require single multifunctional proteins to achieve nucleic acid degradation (Jiang and Doudna 2015).
Streptococcus pyogenes Cas9 (SpCas9), the first well-characterized endonuclease from Class 2 Type II-A CRISPR system, has been widely used for genome editing in various organisms, including plants. SpCas9 assembles with a single-guide RNA (sgRNA), which is engineered by fusing crRNA and trans-activating crRNA (tracrRNA) (Jinek et al. 2012). Ribonucleoprotein complex then recognizes and binds targeted DNA sequences followed by a 5′-NGG-3′ PAM. SpCas9 cleaves DNA, resulting in blunt-ended double-stranded breaks (DSBs). SpCas9 is a multidomain protein that forms two lobes, a recognition (REC) lobe and a nuclease (NUC) lobe (Nishimasu et al. 2014). The REC lobe is responsible for sgRNA and DNA binding, while the HNH and RuvC nuclease domains of the NUC lobe cleave the complementary (targeting) and non-complementary (non-targeting) strands of DNA, respectively. The PI domain (PAM-interacting domain) of NUC lobe is essential for the recognition of the PAM sequence. But PAM requirement also reduces the number of potential target sites in a genome. To address this limitation, Cas9 orthologs or engineered Cas9 proteins recognizing non-canonical PAMs have been explored.
DNA repair pathways are triggered upon the formation of DSBs induced by Cas endonucleases (Fig. 1). Two major DSB repair pathways are the non-homologous end joining (NHEJ) pathway and the homology directed repair (HDR) pathway. The classical (or canonical) NHEJ (C-NHEJ) pathway is the predominant mechanism, which joins two broken DNA ends (Lieber 2010). Repair by C-NHEJ is error-prone and can result in small deletions or insertions at the junction, often causing a reading frame shift and the disruption of gene function. In the alternative NHEJ pathway, also known as the microhomology-mediated end joining (MMEJ) pathway, 5–25 bp microhomology sequences at DSB sites lead to the annealing of broken DNA ends before the ligation. MMEJ may also allow the insertion of desired double-stranded oligodeoxynucleotides at the DSB site (Fig. 1A) (Waterworth et al. 2011). In contrast to NHEJ, HDR pathway requires a template with homology arms for the repair process (Puchta 2004). Homologous recombination is restricted to the S and G2 phases of the eukaryotic cell cycle and is essential for sister chromatid exchange, while NHEJ remains active throughout the cell cycle. At the beginning of HDR (pre-synapsis stage) end resection of DNA surrounding the DSB is initiated by MRN (Mre11–Rad50–Nbs1) complex including RAD50 protein to produce 3′ ssDNA (single-stranded DNA) overhangs, subsequently covered by proteins for stabilization (Waterworth et al. 2011). RAD51 and its paralogs are essential in the HDR repair. Assembly of the RAD51 nucleoprotein searches for DNA homology and mediates DNA strand invasion, which leads to DNA synthesis from the 3′-end of the invading strand followed by DNA ligation (Holthausen et al. 2010; Dexheimer 2013). Although multiple mechanisms for HDR repair have been proposed, three of them are well-known: DSBR (Double-Strand Break Repair), SDSA (Synthesis-Dependent Strand Annealing) and SSA (Single-strand annealing) (Fig. 1A) (Waterworth et al. 2011). HDR allows precise genome editing, including gene insertions. Genome editing is a consequence of the SSN (e.g. CRISPR-Cas) cleavage and DNA repair. While we acknowledge the importance of DNA repair pathways in genome editing (Fig. 1), few studies have explored DNA repair pathways for enhancing genome editing efficiencies and manipulating editing outcomes in plants (Qi et al. 2013; Endo et al. 2016a) and thus this topic will not be a focus.
Base editing is another CRISPR-mediated genome engineering method that enables direct, irreversible conversion of one base pair to another without the need for introducing DSBs or template DNA (Komor et al. 2016; Nishida et al. 2016; Gaudelli et al. 2017). Base editors, such as Cytosine base editors (CBEs) and Adenine base editors (ABEs), are mainly composed of a deaminase fused to a Cas9 variant [nickase (nCas9) or catalytically deactivated Cas9 (dCas9)]. Base editors allow the generation of precise mutations, including the introduction of single nucleotide polymorphisms (SNPs), alternative splicing sites and stop codons. The application of base editing in plants will undoubtedly facilitate basic research and plant breeding. In this mini-review, we will summarize recent developments of CRISPR-Cas technology in plant genome editing with a focus on Cas nucleases and base editors.
Targeting altered PAMs with Cas9 orthologs and engineered variants
The PAM (5′-NGG-3′) requirement is a limitation in the utility of SpCas9 endonuclease. Interestingly, it was demonstrated in rice that SpCas9 could also recognize the 5′-NAG-3′ PAM (Meng et al. 2018). To enable targeting of alternative PAMs, the PI domain of wild-type SpCas9 has been mutated, resulting in the generation of PAM-altered SpCas9 variants with amino acid substitutions, namely VQR, EQR and VRER (Kleinstiver et al. 2015a). VQR, EQR and VRER recognize 5′-NGAN-3′, 5′-NGAG-3′ and 5′-NGCG-3′ PAMs, respectively. In rice, the SpCas9 variants VQR and VRER have been demonstrated to recognize 5′-NGA-3′ and 5′-NGCG-3 PAM sequences, respectively (Hu et al. 2016, 2018a).
Recently, xCas9 was developed by directed evolution to expand the range of PAM recognition and to reduce off-target binding in mammalian cells (Hu et al. 2018b). xCas9 recognizes 5′-NG-3′, 5′-GAA-3′ and 5′-GAT-3′ PAM sequences. To evaluate xCas9 in plants, Wang et al. (2019) generated two variants (xCas9 3.6 and xCas9 3.7) of xCas9 using the rice codon optimized SpCas9 and tested them in rice, targeting 63 genomic sites. xCas9 recognized 5′-GAA-3′ and 5′-NG-3′ PAMs in the rice genome with a relatively low targeting efficiency compared to the canonical 5′-NGG-3′ PAM. According to recent studies in rice, xCas9 had weak or no targeting activity at 5′-NGH-3′ (H = A, T and C) PAM sites (Ren et al. 2019). Similarly, xCas9 was found to have lower editing efficiency at 5′-NGH-3′ sites, while possessing similar activity to SpCas9 at the canonical 5′-NGG-3′ PAM (Zhong et al. 2019). Hua et al. (2019a) also showed induced mutations at 5′-NG-3′ and 5′-GAT-3′ PAM sites in rice by xCas9, albeit with low activity.
Nishimasu et al. (2018) reported another engineered variant of SpCas9 (SpCas9-NG) recognizing 5′-NG-3′ PAMs, which showed higher cleavage kinetics than xCas9 by in vitro assays and higher editing activity in human cells. SpCas9-NGv1 has been demonstrated to target 5′-NG-3′ PAMs in Arabidopsis and rice with enhanced specificity (Endo et al. 2019). SpCas9-NG was shown to successfully edit rice genomic sites with 5′-NGN-3′ PAMs, for which wild-type SpCas9 had little activity (Ren et al. 2019; Hua et al. 2019a; Zhong et al. 2019). However, both SpCas9-NG and its variant SpCas9-NGv1 had lower activity targeting 5′-NGG-3′ PAM sites than wild-type SpCas9 (Zhong et al. 2019). Furthermore, SpCas9-NG showed better editing rates at AT-rich PAM sites (5′-GAT-3′, 5′-GAA-3′, 5′-CAA-3′) than xCas9 (Zhong et al. 2019). More recently, genome editing with xCas9 and Cas9-NG, albeit with low efficiency, was also reported in Arabidopsis (Ge et al. 2019).
Cas9 endonucleases obtained from different bacterial species (e.g. Staphylococcus, Francisella, Brevibacillus) likewise recognize different PAMs. Ran et al. (2015) characterized the Cas9 ortholog from Staphylococcus aureus (SaCas9) and defined its 5′-NNGRRT-3′ PAM site. A SaCas9 variant, referred to as KKH SaCas9, was engineered to recognize the 5′-NNNRRT-3′ PAM (Kleinstiver et al. 2015b). SaCas9 showed a comparable editing efficiency to SpCas9 in Arabidopsis, rice and tobacco (Steinert et al. 2015; Kaya et al. 2016). Both SaCas9 and its KKH variant have been shown to induce high mutagenesis frequencies in rice (Qin et al. 2019). In another study, a 1092 aa Cas9 ortholog was identified in Brevibacillus laterosporus (BlatCas9), with the PAM preference of 5′-NNNNCNDD-3′ (D = A, G or T) (Karvelis et al. 2015). BlatCas9 was able to achieve genome editing in maize cells (Karvelis et al. 2015). St1Cas9 from Streptococcus thermophilus was identified with a PAM requirement of 5′-NNAGAAW-3′ (W = A or T) and was shown to efficiently induce mutations in Arabidopsis at frequencies comparable to SpCas9 (Esvelt et al. 2013; Steinert et al. 2015).
In addition, multiple Cas9 orthologs have been used for genome editing in mammalian systems, but not in plants. The Cas9 from Neisseria meningitidis (NmCas9), recognizing a 5′-NNNNGATT-3′ PAM, was shown to mediate efficient genome editing in human cells (Hou et al. 2013). Hirano et al. (2016) characterized the high-resolution structure of the largest Cas9 protein (FnCas9; ~ 1629 aa) from Francisella novicida with the PAM of 5′-NGG-3′. A triple mutant of FnCas9 referred to as RHA was obtained in the same study, which has a PAM requirement of 5′-YG-3′. In addition, Cas9 from Streptococcus canis (ScCas9) can target a 5′-NNG-3′ PAM in both bacterial and human cells (Chatterjee et al. 2018). More recently, the PAM recognition domain of Streptococcus macacae Cas9 (Smac Cas9) and the nuclease domain of Streptococcus pyogenes (Spy Cas9) were fused to generate SpyMacCas9, which had high editing activity at 5′-NAA-3′ PAM sites (Jakimo et al. 2018). These Cas9 orthologs and engineered variants are likely to further expand targeting scope in plants (Fig. 2).
SpCas9 variants with improved targeting specificity
Cas9 may have off-target effects when used in genome editing and such off-target activities could be reduced by protein engineering. A number of single amino acid mutants of SpCas9 were generated by Slaymaker et al. (2016) and two of them, SpCas9(K855A) and eSpCas9(1.1), have been shown to reduce genome-wide off-target activity. The authors observed that neutralization of positively charged groups in SpCas9 led to a dramatic reduction in unwanted indel formation. It was proposed that wild-type Cas9 possesses more Gibbs free energy than is needed for optimal recognition of the target site and thus will be prone to off-target activity. Similarly, a triple mutant referred to as SpCas9-HF1 (R661A/Q695A/Q926A) has been engineered, which retained its on-target activities (90–140% editing efficiencies of wild type Cas9), while dramatically reducing off-target effects (Kleinstiver et al. 2016). It was found that both SpCas9-HF1 and eSpCas9(1.1) were very sensitive to a mismatch at the 5′ end of the protospacer, and only perfectly matched protospacers (e.g. through precise ribozyme processing) resulted in high on-target activities (Kim et al. 2017a).
Both eSpCas9 and SpCas9-HF1 have been tested in plants. In rice, it was found that optimal on-target activity of these high-fidelity Cas9 variants required a perfectly matched 20-nucleotide protospacer, which was achieved using a tRNA-sgRNA scaffold (Zhang et al. 2017). In Arabidopsis, it was also shown that a tRNA-sgRNA(m) (e.g. modified sgRNA) scaffold aided eSpCas9(1.1) mediated editing, while SpCas9-HF1 barely worked (Zhang et al. 2018). Recently, it was demonstrated in rice that xCas9 possesses higher targeting specificity than the wild-type SpCas9. However, the generally low editing activities of these high-fidelity SpCas9 variants in plants may prevent their wide use by the plant research community. It will be interesting to see whether other high-fidelity Cas9 proteins, such as HiFi Cas9 (Vakulskas et al. 2018) and Sniper-Cas9 (Lee et al. 2018), could enable highly efficient genome editing in plants.
Cas12a orthologs and variants
In 2015, a Class 2 Type V-A enzyme, referred to as Cas12a (formerly Cpf1), was discovered and characterized in detail (Zetsche et al. 2015). There are several differences between Cas12a and SpCas9. First, Cas12a-mediated genome editing only requires a crRNA (~ 43 nt), which is significantly shorter than the sgRNA of SpCas9 (~ 100 nt). Second, Cas12a requires a T-rich (5′-TTTV-3′ or 5′-TTV-3′) PAM, which is suitable for targeting AT-rich genomic regions. Third, Cas12a produces staggered ends of 4–5 bp, which may facilitate NHEJ-mediated gene replacement. Fourth, a single RuvC-like domain of Cas12a is responsible for DNA cleavage (Zetsche et al. 2015), unlike SpCas9 that has two functional catalytic domains for DNA strand cleavage. This feature makes it a challenge to generate a Cas12a nickase. Finally, Cas12a cleaves DNA in distant locations from the seed sequence, which may allow continuous cleavage of the target DNA. This feature makes Cas12a more suitable for donor insertions, as proven in Zebrafish embryos, in which a higher HDR frequency has been obtained by LbCas12a than by SpCas9 (Moreno-Mateos et al. 2017).
To date, three orthologs of Cas12a, FnCas12a (from Francisella tularensis subsp. novicida U112), AsCas12a (from Acidaminococcus sp. BV3L6) and LbCas12a (from Lachnospiraceae bacterium ND2006), have been used in genome editing in plants, including rice, Arabidopsis, soybean, tobacco, maize, tomato, citrus and cotton (Endo et al. 2016b; Kim et al. 2017b; Li et al. 2018a, 2019a; Hu et al. 2017; Xu et al. 2017; Tang et al. 2017; Wang et al. 2017; Yin et al. 2017; Malzahn et al. 2019; Lee et al. 2019; Bernabé-Orts et al. 2019; Jia et al. 2019). Generally, LbCas12a and FnCas12a were shown to be more reliable in plant genome editing than AsCas12a. Elevated temperatures (e.g. 32 °C) improved AsCas12a’s enzymatic activity and resulted in up to 92.8% gene editing frequency in T0 rice lines (Malzahn et al. 2019). The editing frequency is also influenced by the design of the crRNA expression cassette and has reached 100% with the use of the double ribozyme system in T0 rice plants (Tang et al. 2017; Zhong et al. 2018). Furthermore, a tRNA-based processing system and a Cas12a CRISPR array-based processing system were both successfully used for multiplexed plant genome editing (Wang et al. 2017; Ding et al. 2018).
While AsCas12a, FnCas12a and LbCas12a all prefer a 5′-TTTV-3′ PAM, a recent study showed that FnCas12a can target most of 5′-VTTV-3′ PAM sites in rice (Zhong et al. 2018). Relaxed PAM recognition can also be achieved with engineered Cas12a variants (Fig. 2). Altered PAM recognition was previously demonstrated by engineered variants of AsCas12a, namely RR and RVR (Gao et al. 2017), which recognize non-canonical 5′-TYCV-3′ and 5′-CCCC-3′, as well as 5′-TATV-3′ PAMs in human cells, respectively. Recently, Li et al. (2018a) showed that the LbCas12a-RR variant was able to edit 5′-TYCV-3′ PAMs in rice. Zhong et al. (2018) reported that LbCas12a-RR and -RVR variants, but not FnCas12a variants, were able to recognize these altered PAMs in rice.
Cas12b endonucleases
Cas12b (formerly C2c1) is a Class II Type V-B endonuclease that targets double-stranded DNA (Strecker et al. 2019). Cas12b specifically cuts DNA with its single RuvC domain and produces staggered ends with ~ 7-nt overhangs (Wu et al. 2017; Liu et al. 2017). Cas12b recognizes a distal 5′-T rich (5′-TTN-3′) PAM and its catalytic activity depends on both the presence of tracrRNA and crRNA (dual-RNA-guided), similar to SpCas9 (Shmakov et al. 2015). Structural analysis of a Cas12b from Bacillus thermoamylovorans (BthCas12b) showed that recognition of sgRNA and PAM-containing duplex by Cas12b was different from Cas9 and Cas12a (Wu et al. 2017). Liu et al. (2017) reported the crystal structure of AacCas12b isolated from Alicyclobacillus acidoterrestris, a Gram-positive soil bacterium that grows in high temperatures (35–55 °C).
Teng et al. (2018) identified a Cas12b ortholog from Alicylobacillus acidiphilus (AaCas12b) that kept its nuclease activity between the temperatures of 31 °C and 59 °C, enabling genome editing in mammalian cell cultures. Recently, a Cas12b ortholog from Bacillus hisashii (BhCas12b) was characterized in detail and its engineered mutant, BhCas12b v4 was tested for cleavage activity in human cells under lower temperatures than the wild-type form (Strecker et al. 2019). Artificial crRNAs have also been successfully engineered to work with multiple Cas12b orthologs for genome editing in human cells (Teng et al. 2019). Cas12b represents an ideal enzyme by its small size and potential low off-target activity and could be adapted for plant genome editing in the near future.
Base editors
Base editing is a genome editing approach utilizing specific “base editors” to convert a base into another in a targeted manner (Komor et al. 2016; Nishida et al. 2016; Gaudelli et al. 2017). Precise C-to-T or A-to-G changes could be achieved on a desired DNA sequence by base editors, without the formation of DSBs and -induced NHEJ or MMEJ events that eliminates further re-arrangements and without the need for HDR that is restricted to S- and G2-phases of cells. Thus, base-editing represents a powerful and promising tool for point-mutagenesis that can be exploited in crop development, as seen in the manipulation of genes associated with agronomical traits (Hua et al. 2018; Yan et al. 2018; Kang et al. 2018; Endo et al. 2019; Zhang et al. 2019).
At present, there are three major groups of base editors: cytidine base editors (CBEs), adenine base editors (ABEs) and RNA base editors (adenosine deaminases act on RNA: ADARs) (reviewed in Molla and Yang 2019). CBEs catalyze the conversion of C-G to T-A through an initial deamination of cytidine to uracil, which results in a U-G mismatch. U is recognized as T during repair or replication, thus forming a T-A pair. The first generation of CBEs (BE1) comprised of cytidine deaminase rAPOBEC1 fused to dCas9 carrying the D10A and H840A mutations (Komor et al. 2016). BE1 can achieve efficient and targeted deamination in vitro, but not in vivo. This is due to the cellular base excision repair (BER) pathway that usually converts the edited U-G pairs back to C-G pairs. In BER, DNA glycosylases remove the mismatched base by cleaving the N-glycosidic bond between the target base and deoxyribose, creating apurinic or apyrimidinic (AP) sites (also known as abasic sites) on DNA (Schormann et al. 2014). Abasic sites are resolved by an AP endonuclease, followed by gap filling and ligation (Fig. 1B). Uracil DNA Glycosylase (UDG) removes U from DNA in cells and leads to the BER pathway (Komor et al. 2016). To improve base editing efficiency in vivo, a UGI (Uracil-DNA glycosylase inhibitor) domain from Bacillus subtilis bacteriophage PBS1 was fused to the C terminus of a cytidine deaminase to prevent base editor-induced BER, thus the mismatch can be repaired through mismatch repair (MMR) pathway, an alternative route and important mechanism for post-replication repair of bases that are mis-incorporated to DNA. This base editor architecture is referred to as BE2. In BE3 and BE4, dSpCas9 is replaced with nSpCas9 nickase, harboring the D10A mutation abolishing the catalytic activity of the RuvC-like domain, thus inducing cellular repair on non-edited strand using edited DNA as a template. BE3 increases the base editing frequency substantially; however, it generally also results in insertions and deletions (indels), due to the ultimate production of double-stranded breaks. When a nick and an AP site are generated at the same target site, DSBs may occur due to AP endonuclease cleavage or spontaneous breakage, which tend to be repaired by NHEJ. Increasing the number of UGIs that are available at the editing sites can increase the purity of edits (Komor et al. 2017). Fourth-generation base editors (BE4, SaBE4, BE4-Gam, SaBE4-Gam) can boost the C:G to T:A base editing efficiency (approximately 50% in human cells) with reduced indel formation (Komor et al. 2017). Recently, C-to-T base editing purity was improved in yeast by reducing the width of editing window by removing non-essential deaminase sequences and optimizing linker sequences (Tan et al. 2019). Improvement of DNA base editors for PAM compatibility, editing specificity and their current applications can be found in two recent reviews (Rees and Liu 2018; Molla and Yang 2019).
Cytidine deaminases have been used to edit genomes of rice, wheat, maize, tomato, potato and watermelon (Zong et al. 2017; Li et al. 2017; Lu and Zhu 2017; Ren et al. 2017, 2018, 2019; Shimatani et al. 2017; Zong et al. 2018, 2019; Tian et al. 2018; Endo et al. 2019; Hua et al. 2019b; Veillet et al. 2019; Zhang et al. 2019). Optimizations for increasing targeting flexibility, improving editing purity and expanding PAM recognition are provided in Table 1. Target-AID, a base editor utilizing the cytidine deaminase domain of Petromyzon marinus (PmCDA1) has shown a C-to-T editing efficiency of 53% with off-target activity of 0.38% in tomato, suggesting that it is one of the most optimal deaminases for plants (Shimatani et al. 2017). rAPOBEC1 and PmCDA1, hAID and human APOBEC3A (A3A) have been used in C-to-T base editing in plants (Ren et al. 2018; Zong et al. 2018). PmCDA1 has also been fused to Sp nCas9-NGv1 and successfully used to edit the rice genome (Endo et al. 2019). In addition, a combination of cytidine base editing and DNA-free genome editing has been demonstrated in plants (Zong et al. 2018; Veillet et al. 2019).
Table 1.
DNA base editor | References (first description) | Composition | Feature | Plants | References |
---|---|---|---|---|---|
C to T conversion | |||||
BE2 | Komor et al. (2016) | rAPOBEC1-Sp dCas9 (D10A, H840A)-UGI | Inhibition of Uracil N-glycosylase; circumventing cellular DNA repair processes (BER) | Rice, wheat, maize | Zong et al. (2017) |
BE3 | Komor et al. (2016) | rAPOBEC1-Sp nCas9 (D10A)-UGI | Nicking non-edited strand; favoring U to A over U to G outcome | Rice, wheat, maize | Hua et al. (2018); Li et al. (2017); Lu and Zhu (2017); Ren et al. (2017); Zong et al. (2017); Zhang et al. (2019) |
BE3 | Komor et al. (2016) | Sp nCas9 NGv1(D10A)-rAPOBEC-UGI | NGv1 nCas9 recognizing NG PAM; increase targeting flexibility | Rice | Endo et al. (2019) |
NGv1-BE3 | Nishida et al. (2016) | Sp nCas9 NGv1(D10A)-AID-UGI | NGv1 nCas9 recognizing NG PAM; increase targeting flexibility | Rice | Endo et al. (2019) |
BE4 | Komor et al. (2017) | rAPOBEC1-Sp nCas9 (D10A)-UGI-UGI | Addition of second UGI domain, improving editing purity | Rice | Ren et al. (2017) |
rBE5 | Ren et al. (2018) | hAID*Δ-Sp nCas9 (D10A) | hAID deaminase; increase in editing efficiency | Rice | Ren et al. (2018) |
rBE9 | Ren et al. (2018) | hAID*Δ-Sp nCas9 (D10A)-UGI | Addition of the UGI domain, improving editing purity compared to rBE5 | Rice | Ren et al. (2018) |
VQR-BE3 | Kim et al. (2017d) | rAPOBEC1-Sp nCas9 VQR (D10A)-UGI | VQR nCas9 recognizing NGA PAM; increasing targeting flexibility | Rice | Hua et al. (2019b) |
Sa (KKH)-BE3 | Kim et al. (2017d) | rAPOBEC1-Sa nCas9 KKH (D10A)-UGI | Sa nCas9 recognizing NNNRRT PAM, increasing targeting flexibility | Rice | Hua et al. (2019b) |
Target-AID | Nishida et al. (2016) | Sp nCas9 (D10A)-CDA1-UGI | CDA1 provides slightly shifted editing activity window compared to APOBEC1 | Rice, tomato | Shimatani et al. (2017) |
Target-AID | Nishimasu et al. (2018) | Sp nCas9 NGv1(D10A)-CDA1-UGI | CDA1 provides slightly shifted editing activity window compared to APOBEC1, Sp nCas9-NGv1 recognizing NG PAM; increasing targeting flexibility | Rice | Endo et al. (2019) |
BE3 | Komor et al. (2016) | rAPOBEC1-Sp n xCas9 (D10A)-UGI | xCas9 was used for recognizing NGN, GAA, GAT, CAA PAMs; increasing targeting flexibility | Rice | Zhong et al. (2019); Hua et al. (2019a) |
Target AID | Nishida et al. (2016) | Sp nCas9 NG (D10A)-PmCDA1-UGI | Target AID was upgraded to reduce Indels by fusion of UGI to PmCDA1 | Rice | Zhong et al. (2019) |
rBE9 | Ren et al. (2018) | hAID*Δ-Sp nCas9 NG (D10A)-UGI | Expanding the scope of gene editing by recognition of NG PAM | Rice | Ren et al. (2019) |
A3A-PBE | Zong et al. (2018) | APOBEC3A-Sp nCas9 (D10A)-UGI | Replacing rAPOBEC1 with APOBEC3A for increased editing efficiency, specificity, and expanded deamination window | Rice, wheat, potato | Zong et al. (2018) |
A3A-Gam | Zong et al. (2018) | Gam-APOBEC3A-Sp nCas9 (D10A)-2 × UGI | Addition of Gam to A3A-PBE to increase editing efficiency and product purity | Rice, wheat | Zong et al. (2018) |
A to G conversion | |||||
ABE6.3 ABE7.8 ABE7.9 |
Gaudelli et al. (2017) | TadA-TadA mutant-Sp nCas9 (D10A) |
Offer higher editing efficiency at position closer to PAM compared to ABE7.10 |
Arabidopsis thaliana, Brassica napus | Kang et al. (2018) |
ABE7.10 | Gaudelli et al. (2017) | TadA-TadA mutant-Sp nCas9 (D10A) | Most efficient and sequence context-independent ABE, activity window further from PAM compared to ABE6.3, 7.8 and 7.9 | Arabidopsis thaliana, Brassica napus, rice, wheat | Kang et al. (2018); Hua et al. (2019b); Li et al. (2018b) |
ABE7.10 | Gaudelli et al. (2017) | TadA-TadA mutant-Sp n xCas9 (D10A) | xCas9 was used for recognizing NGN, GAA, GAT, CAA PAMs; increasing targeting flexibility | Rice | Hua et al. (2019a) |
ABE7.10 | Gaudelli et al. (2017) | TadA-TadA mutant-Sp nCas9 NGv1 (D10A) | NGv1 nCas9 recognizing NG PAM; increase targeting flexibility | Rice | Negishi et al. (2019) |
ABE7.10* | Yan et al. (2018) | TadA-TadA mutant-Sp nCas9 (D10A) | Broader activity window compared to ABE7.10 | Rice | Yan et al. (2018) |
VQR-ABE | Hua et al. (2018) | TadA-TadA mutant-Sp nCas9 (D10A) VQR | VQR nCas9 recognizing NGA PAM; increasing targeting flexibility | Rice | Hua et al. (2019b) |
VRER-ABE | Hua et al. (2018) | TadA-TadA mutant-Sp nCas9 (D10A) VRER | VRER nCas9 recognizing NGCG PAM; increasing targeting flexibility | Rice | Hua et al. (2019b) |
Sa (KKH)-ABE | Hua et al. (2018) | TadA-TadA mutant-Sa nCas9 (D10A) KKH | Sa nCas9 recognizing NNNRRT PAM, increasing targeting flexibility | Rice | Hua et al. (2019b) |
rBE15 | Yan et al. (2018) | TadA-TadA*7.10-Sp dCas9 (D10A, H840A) | Employs catalytically inactive dCas9 | Rice | Yan et al. (2018) |
rBE18 | Yan et al. (2018) | TadA-TadA*7.8-Sp dCas9 (D10A, H840A) | Employs catalytically inactive dCas9 | Rice | Yan et al. (2018) |
rBE14 | Yan et al. (2018) | TadA-TadA*7.8-Sp nCas9 (D10A) NG | Expanding the scope of gene editing by recognition of NG PAM | Rice | Ren et al. (2019) |
PABE7 | Li et al. (2018b) | TadA-TadA*-Sp nCas9 (D10A)-3xNLS | Optimizes location and number of NLS to increase base editing efficiency | Rice, wheat | Li et al. (2018b) |
Deamination of adenine by adenine base editors produces an inosine (I) base, which is read as guanine by DNA replication or repair machinery. As there are no known natural enzymes that deaminate adenine in dsDNA, Gaudelli et al. (2017) developed deaminase variants based on Escherichia coli tRNA adenine deaminase (TadA), which catalyze the deamination of adenine on ssDNA. The group used directed evolution and protein engineering to produce an improved version of a mutated TadA-dCas9 heterodimeric protein that has high base editing efficiency in mammalian cells. Among adenine base editors, ABE7.10 showed the highest base editing efficiency, of up to 4.1% in Arabidopsis and 8.8% in Brassica protoplasts, and with low undesirable mutation rates (< 0.1%) (Kang et al. 2018). The ABE7.10 architecture was further optimized for base editing of crop plants with respect to the number and locations of nuclear localization signal sequences (Li et al. 2018b).
Recently, a series of adenine and cytosine base editors were developed using both SpCas9 and SaCas9 variants to expand the targeting scope in rice (Hua et al. 2019b; Qin et al. 2019). Similarly, adenine and cytosine base editors based on Cas9-NG were also reported to edit relaxed PAM sites in plants (Negishi et al. 2019; Ren et al. 2019; Hua et al. 2019a).
Either loss-of-function or gain-of-function phenotypes can be achieved with base editing. It is possible to introduce a nonsense mutation in the gene of interest by C-to-T base editors. Loss-of-function phenotypes can also be achieved by introduced missense mutations or mutations that alter RNA splicing sites as demonstrated in Arabidopsis (Kang et al. 2018; Li et al. 2019b). Gain-of-function phenotypes were reported such as dominant semi-dwarf phenotype by editing SLR1 in rice (Lu and Zhu 2017) and herbicide resistance by editing ALS in rice and wheat (Shimatani et al. 2017; Zong et al. 2018; Zhang et al. 2019) or editing ACC in rice (Li et al. 2018b). In principle, base editors could also be applied to target upstream open reading frames (uORFs) for improved protein translation, as well as to target promoter regions to introduce quantitative traits, as recently demonstrated with the use of Cas9 nuclease (Rodríguez-Leal et al. 2017; Zhang et al. 2018).
Off-target effects of CRISPR-Cas systems
Off-target effects of CRISPR-Cas systems are potential concerns in many of their applications. Whole-genome sequencing (WGS) conducted in Arabidopsis, tomato and cotton showed that off-target activity induced by SpCas9 has rarely occurred (Feng et al. 2014; Nekrasov et al. 2017; Li et al. 2018c; Tang et al. 2018). High targeting specificity was also recently reported for Cas12a in rice and cotton (Tang et al. 2018; Li et al. 2019a). SpCas9 has some tolerance to 1-nt mismatches in protospacers (Wu et al. 2014; Tang et al. 2018). Such tolerance was drastically diminished with high-fidelity SpCas9 variants. For example, xCas9 was more sensitive to 1-nucleotide mismatch than the wild-type SpCas9 in both human cells and rice (Hu et al. 2018b; Zhong et al. 2019). The simultaneous introduction of 2-nucleotide mismatches in protospacers virtually abolished the editing activities of LbCas12a and FnCas12a (Tang et al. 2017; Zhong et al. 2018). The specificity of wild-type Cas9 and Cas12a can be further improved with engineered variants, making off-target edits by Cas proteins less of a concern in plants. However, lack of high-quality reference genomes for many plant species and cultivars is a major limitation for off-target assessments. Unbiased methods such as WGS and Digenome-seq provide comprehensive analysis (e.g. from small indels to structural variations) of the genome but are costly (Zischewski et al. 2017).
Base editors were recently found to exhibit a tendency to cause genome-wide off-target effects. Similar to nucleases, putative off-target sites for base editing can be identified by tailored methods such as Digenome-seq (Kim et al. 2017c). However, a comprehensive assessment of off-targeting requires WGS. For example, BE3 led to elevated genome-wide C to T base changes in mouse and rice (Zuo et al. 2019; Jin et al. 2019). Such an off-target effect appears to be independent of Cas9, but due to the ectopic expression of the cytidine deaminase. ABE does not seem to cause genome-wide off-target mutations (Jin et al. 2019). Interestingly, both BE3 and ABE were also shown to cause transcriptome-wide off-target effects by editing RNA transcripts (Grünewald et al. 2019; Rees et al. 2019). Fortunately, these off-target effects at DNA and RNA levels can be greatly reduced with engineered deaminase variants (Grünewald et al. 2019; Rees et al. 2019; Zhou et al. 2019).
Conclusion
The scope of genome editing is expanded widely using novel Cas9 and Cas12a engineered variants and orthologs, which recognize a broader range of PAM sequences. To date, only some Cas9 and Cas12a variants and orthologs have been adapted for plant genome editing indicating great potential of further expanding genome editing scope in plants in the near future. Base editing is a powerful technique to generate point mutations in vivo. Given the high number of SNPs associated with agronomic traits in plants, gene editing by base editors could play a significant role in generating favorable allelic combinations. However, improved base editors with reduced off-target effects are needed to allow precise genome editing in plants. The continued evolution of these genome editing tools will further enhance fundamental and translational plant research.
Acknowledgements
We apologize to the authors whose studies were not cited due to limited space. Our plant genome editing research is supported by the National Science Foundation Plant Genome Research Program (IOS-1758745) and USDA-NIFA Biotechnology Risk Assessment Research Program (2018-33522-28789), Foundation for Food and Agriculture Research (593603), and Syngenta Biotechnology. We thank Aimee Malzahn and Dr. Brian Iaffaldano for proofreading this manuscript.
Author contributions
FG, YZ, SS and YQ wrote the manuscript. All authors read and approved the final manuscript.
Compliance with ethical standards
Conflict of interest
The authors have no conflict of interests to declare.
References
- Bernabé-Orts JM, Casas-Rodrigo I, Minguet EG, Landolfi V, Garcia-Carpintero V, Gianoglio S, Vázquez-Vilar M, Granell A, Orzaez D. Assessment of Cas12a-mediated gene editing efficiency in plants. Plant Biotechnol J. 2019 doi: 10.1111/pbi.13113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chatterjee P, Jakimo N, Jacobson JM. Minimal PAM specificity of a highly similar SpCas9 ortholog. Sci Adv. 2018;4(10):eaau0766. doi: 10.1126/sciadv.aau0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dexheimer TS, et al. DNA repair pathways and mechanisms. In: Mathews LA, et al., editors. DNA repair of cancer stem cells. Dordrecht: Springer; 2013. pp. 19–32. [Google Scholar]
- Ding D, Chen K, Chen Y, Li H, Xie K. Engineering introns to express RNA guides for Cas9- and Cpf1-mediated multiplex genome editing. Mol Plant. 2018;11(4):542–552. doi: 10.1016/j.molp.2018.02.005. [DOI] [PubMed] [Google Scholar]
- Endo M, Mikami M, Toki S. Biallelic gene targeting in rice. Plant Physiol. 2016;170:667–677. doi: 10.1104/pp.15.01663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Endo A, Masafumi M, Kaya H, Toki S. Efficient targeted mutagenesis of rice and tobacco genomes using Cpf1 from Francisella novicida. Sci Rep. 2016;6:38169. doi: 10.1038/srep38169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Endo M, Mikami M, Endo A, Kaya H, Itoh T, Nishimasu H, Nureki O, Toki S. Genome editing in plants by engineered CRISPR-Cas9 recognizing NG PAM. Nat Plants. 2019;5(1):14–17. doi: 10.1038/s41477-018-0321-8. [DOI] [PubMed] [Google Scholar]
- Esvelt KM, Mali P, Braff JL, Moosburner M, Yaung SJ, Church GM. Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat Methods. 2013;10(11):1116–1121. doi: 10.1038/nmeth.2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng Z, Mao Y, Xu N, Zhang B, Wei P, Yang DL, et al. Multigeneration analysis reveals the inheritance, specificity, and patterns of CRISPR/Cas induced gene modifications in Arabidopsis. Proc Natl Acad Sci USA. 2014;111:4632–4637. doi: 10.1073/pnas.1400822111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao L, Cox DBT, Yan WX, Manteiga JC, Schneider MW, Yamano T, Nishimasu H, Nureki O, Crosetto N, Zhang F. Engineered Cpf1 variants with altered PAM specificities. Nat Biotechnol. 2017;35(8):789–792. doi: 10.1038/nbt.3900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaudelli NM, Komor AC, Rees HA, Packer MS, Badran AH, Bryson DI, Liu DR. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 2017;551:464–471. doi: 10.1038/nature24644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge Z, Zheng L, Zhao Y, Jaing T, et al. Engineered xCas9 and SpCas9-NG variants broaden PAM recognition sites to generate mutations in Arabidopsis plants. Plant Biotechnol J. 2019 doi: 10.1111/pbi.13148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grünewald J, Zhou R, Garcia SP, Iyer S, Lareau CA, Aryee MJ, Joung JK. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature. 2019;569(7756):433–437. doi: 10.1038/s41586-019-1161-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haft DH, Selengut J, Mongodin EF, Nelson KE. A guild of 45 CRISPR-associated (Cas) protein families and multiple CRISPR/Cas subtypes exist in prokaryotic genomes. PLoS Comput Biol. 2005;1(6):e60. doi: 10.1371/journal.pcbi.0010060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirano H, Gootenberg JS, Horii T, Abudayyeh OO, Kimura M, Hsu PD, Nakane T, Ishitani R, Hatada I, Zhang F, et al. Structure and engineering of Francisella novicida Cas9. Cell. 2016;164(5):950–961. doi: 10.1016/j.cell.2016.01.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holthausen JT, Wyman C, Kanaar R. Regulation of DNA strand exchange in homologous recombination. DNA Repair. 2010;9(12):64–72. doi: 10.1016/j.dnarep.2010.09.014. [DOI] [PubMed] [Google Scholar]
- Hou Z, Zhang Y, Propson NE, Howden SE, Chu LF, Sontheimer EJ, Thomson JA. Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc Natl Acad Sci USA. 2013;110(39):15644–15649. doi: 10.1073/pnas.1313587110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu X, Wang C, Fu Y, Liu Q, Jiao X, Wan K. Expanding the range of CRISPR/Cas9 genome editing in rice. Mol Plant. 2016;9:943–945. doi: 10.1016/j.molp.2016.03.003. [DOI] [PubMed] [Google Scholar]
- Hu X, Wang C, Liu Q, Fu Y, Wang K. Targeted mutagenesis in rice using CRISPR-Cpf1 system. J Genet Genom. 2017 doi: 10.1016/j.jgg.2016.12.001. [DOI] [PubMed] [Google Scholar]
- Hu X, Meng X, Liu Q, Li J, Wang K. Increasing the efficiency of CRISPR-Cas9-VQR precise genome editing in rice. Plant Biotechnol J. 2018;16:292–297. doi: 10.1111/pbi.12771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu JH, Miller SM, Geurts MH, Tang W, Chen L, Sun N, Zeina CM, Gao X, Rees HA, Lin Z, Liu DR. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature. 2018;556:57–63. doi: 10.1038/nature26155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hua K, Tao X, Yuan F, Wang D, Zhu JK. Precise A·T to G·C base editing in the rice genome. Mol Plant. 2018;11(4):627–630. doi: 10.1016/j.molp.2018.02.007. [DOI] [PubMed] [Google Scholar]
- Hua K, Tao X, Han P, Wang R, Zhu JK. Genome engineering in rice using Cas9 variants that recognize NG PAM sequences. Mol Plant. 2019 doi: 10.1016/j.molp.2019.03.009. [DOI] [PubMed] [Google Scholar]
- Hua K, Tao X, Zhu J-K. Expanding the base editing scope in rice by using Cas9 variants. Plant Biotechnol J. 2019;17(2):499–504. doi: 10.1111/pbi.12993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jakimo A, Chatterjee P, Nip L, Jacobson JM. Cas9 with complete PAM recognition for adenine dinucleotides. BioRxiv. 2018 doi: 10.1101/429654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jansen R, Embden JD, Gaastra W, Schouls LM. Identification of genes that are associated with DNA repeats in prokaryotes. Mol Microbiol. 2002;43:1565–1575. doi: 10.1046/j.1365-2958.2002.02839.x. [DOI] [PubMed] [Google Scholar]
- Jia H, Orbović V, Wang N. CRISPR-LbCas12a-mediated modification of citrus. Plant Biotechnol J. 2019 doi: 10.1111/pbi.13109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang F, Doudna JA. The structural biology of CRISPR-Cas systems. Curr Opin Struct Biol. 2015;30:100–111. doi: 10.1016/j.sbi.2015.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin S, Zong Y, Gao Q, et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science. 2019;364(6437):292–295. doi: 10.1126/science.aaw7166. [DOI] [PubMed] [Google Scholar]
- Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816–821. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang BC, Yun JY, Kim ST, Shin Y, Ryu J, Choi M, Woo JW, Kim JS. Precision genome engineering through adenine base editing in plants. Nat Plants. 2018;4:427–431. doi: 10.1038/s41477-018-0178-x. [DOI] [PubMed] [Google Scholar]
- Karvelis T, Gasiunas G, Young J, Bigelyte G, Silanskas A, Cigan M, Siksnys V. Rapid characterization of CRISPR-Cas9 protospacer adjacent motif sequence elements. Genome Biol. 2015;16:253. doi: 10.1186/s13059-015-0818-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaya H, Mikami M, Endo A, Endo M, Toki S. Highly specific targeted mutagenesis in plants using Staphylococcus aureus Cas9. Sci Rep. 2016;6:26871. doi: 10.1038/srep26871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim S, Bae T, Hwang J, Kim JS. Rescue of high-specificity Cas9 variants using sgRNAs with matched 5′ nucleotides. Genome Biol. 2017;18(1):218. doi: 10.1186/s13059-017-1355-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim H, Kim ST, Ryu J, Kang BC, Kim JS, Kim SG. CRISPR/Cpf1-mediated DNA-free plant genome editing. Nat Commun. 2017;8:14406. doi: 10.1038/ncomms14406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim D, Lim K, Kim ST, Yoon SH, Kim K, Ryu SM, Kim JS. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat Biotechnol. 2017;35(5):475–480. doi: 10.1038/nbt.3852. [DOI] [PubMed] [Google Scholar]
- Kim YB, Komor AC, Levy JM, Packer MS, Zhao KT, Liu DR. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat Biotechnol. 2017;35(4):371–376. doi: 10.1038/nbt.3803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinstiver BP, Prew MS, Tsai SQ, Topkar VV, Nguyen NT, Zheng Z, et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. 2015;523:481–485. doi: 10.1038/nature14592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinstiver BP, Prew MS, Tsai SQ, et al. Broadening Staphylococcus aureus Cas9 targeting range by modifying PAM recognition. Nat Biotechnol. 2015;33(12):1293–1298. doi: 10.1038/nbt.3404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z, Joung JK. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature. 2016;529:490–495. doi: 10.1038/nature16526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. 2016;533:420–424. doi: 10.1038/nature17946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komor AC, Zhao KT, Packer MS, Gaudelli NM, Waterbury AL, Koblan LW, Kim YB, Badran AH, Liu DR. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T: A base editors with higher efficiency and product purity. Sci Adv. 2017;3(8):eaao4774. doi: 10.1126/sciadv.aao4774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JK, Jeong E, Lee J, Jung M, Shin E, Kim YH, Lee K, Jung I, Kim D, Kim S, Kim JS. Directed evolution of CRISPR-Cas9 to increase its specificity. Nat Commun. 2018;9(1):3048. doi: 10.1038/s41467-018-05477-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee K, Zhang Y, Kleinstiver BP, Guo JA, Aryee MJ, Miller J, Malzahn A, Zarecor S, Lawrence-Dill CJ, Joung JK, Qi Y, Wang K. Activities and specificities of CRISPR/Cas9 and Cas12a nucleases for targeted mutagenesis in maize. Plant Biotechnol J. 2019;17:362–372. doi: 10.1111/pbi.12982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, Sun Y, Du J, Zhao Y, Xia L. Generation of targeted point mutations in rice by a modified CRISPR/Cas9 system. Mol Plant. 2017;10:526–529. doi: 10.1016/j.molp.2016.12.001. [DOI] [PubMed] [Google Scholar]
- Li S, Zhang X, Wang W, Guo X, Wu Z, Du W, Zhao Y, et al. Expanding the scope of CRISPR/Cpf1-mediated genome editing in rice. Mol Plant. 2018;11:995–998. doi: 10.1016/j.molp.2018.03.009. [DOI] [PubMed] [Google Scholar]
- Li C, Zong Y, Wang Y, Jin S, Zhang D, Song Q, Zhang R, Gao C. Expanded base editing in rice and wheat using a Cas9-adenosine deaminase fusion. Genome Biol. 2018;19:59. doi: 10.1186/s13059-018-1443-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J, et al. Whole genome sequencing reveals rare off-target mutations and considerable inherent genetic or/and somaclonal variations in CRISPR/Cas9-edited cotton plants. Plant Biotechnol J. 2018 doi: 10.1111/pbi.13020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B, et al. Robust CRISPR/Cpf1 (Cas12a) mediated genome editing in allotetraploid cotton (G. hirsutum) Plant Biotechnol J. 2019 doi: 10.1111/pbi.13147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Z, Xiong X, Wang F, Liang J, Li J-F. Gene disruption through base editing-induced messenger RNA missplicing in plants. New Phytol. 2019;222(2):1139–1148. doi: 10.1111/nph.15647. [DOI] [PubMed] [Google Scholar]
- Lieber MR. The mechanism of double-strand DNA break repair by the nonhomologous DNA end joining pathway. Annu Rev Biochem. 2010;79:181–211. doi: 10.1146/annurev.biochem.052308.093131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu L, Chen P, Wang M, Li X, Wang J, Yin M, Wang Y. C2c1-sgRNA complex structure reveals RNA-guided DNA cleavage mechanism. Mol Cell. 2017;65(2):310–322. doi: 10.1016/j.molcel.2016.11.040. [DOI] [PubMed] [Google Scholar]
- Lu Y, Zhu JK. Precise editing of a target base in the rice genome using a modified CRISPR/Cas9 system. Mol Plant. 2017;10:523–525. doi: 10.1016/j.molp.2016.11.013. [DOI] [PubMed] [Google Scholar]
- Malzahn AA, Tang X, Lee K, Ren Q, Sretenovic S, Zhang Y, Chen H, Kang M, Bao Y, Zheng X, Deng K, Zhang T, Salcedo V, Wang K, Zhang Y, Qi Y. Application of CRISPR-Cas12a temperature sensitivity for improved genome editing in rice, maize, and Arabidopsis. BMC Biol. 2019;17(1):9. doi: 10.1186/s12915-019-0629-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGinn J, Marraffini LA. Molecular mechanisms of CRISPR–Cas spacer acquisition. Nat Rev Microbiol. 2019;17:7–12. doi: 10.1038/s41579-018-0071-7. [DOI] [PubMed] [Google Scholar]
- Meng X, Hu X, Liu Q, Song X, Gao C, Li J, Wang K. Robust genome editing of CRISPR-Cas9 at NAG PAMs in rice. Sci China Life Sci. 2018;61:122–125. doi: 10.1007/s11427-017-9247-9. [DOI] [PubMed] [Google Scholar]
- Molla KA, Yang Y. CRISPR/Cas-mediated base editing: technical considerations and practical applications. Trends Biotechnol. 2019 doi: 10.1016/j.tibtech.2019.03.008. [DOI] [PubMed] [Google Scholar]
- Moreno-Mateos MA, Fernandez JP, Rouet R, Lane MA, Vejnar CE, Mis E, Khokha MK, Doudna JA, Giraldez AJ. CRISPR-Cpf1 mediates efficient homology-directed repair and temperature-controlled genome editing. Nat Commun. 2017;8:2024. doi: 10.1038/s41467-017-01836-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Negishi K, Kaya H, Abe K, Hara N, Saika H, Toki S. An adenine base editor with expanded targeting scope using SpCas9-NG v1 in rice. Plant Biotechnol J. 2019 doi: 10.1111/pbi.13120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nekrasov V, Wang C, Win J, Lanz C, Weigel D, Kamoun S. Rapid generation of a transgene-free powdery mildew resistant tomato by genome deletion. Sci Rep. 2017;7:482. doi: 10.1038/s41598-017-00578-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nishida K, Arazoe T, Yachie N, Banno S, Kakimoto M, Tabata M, et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science. 2016 doi: 10.1126/science.aaf8729. [DOI] [PubMed] [Google Scholar]
- Nishimasu H, et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. 2014;156:935–949. doi: 10.1016/j.cell.2014.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science. 2018;361(6408):1259–1262. doi: 10.1126/science.aas9129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Puchta H. The repair of double-strand breaks in plants: mechanisms and consequences for genome evolution. J Exp Bot. 2004;56(409):1–14. doi: 10.1093/jxb/eri025. [DOI] [PubMed] [Google Scholar]
- Qi Y, et al. Increasing frequencies of site-specific mutagenesis and gene targeting in Arabidopsis by manipulating DNA repair pathways. Genome Res. 2013;23(3):547–554. doi: 10.1101/gr.145557.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin R, Li J, Li H, Zhang Y, Liu X, Miao Y, Zhang X, Wei P. Developing a highly efficient and wildly adaptive CRISPR-SaCas9 toolset for plant genome editing. Plant Biotechnol J. 2019;17(4):706–708. doi: 10.1111/pbi.13047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ran FA, et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015;520:186–191. doi: 10.1038/nature14299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rath D, Amlinger L, Rath A, Lundgren M. The CRISPR-Cas immune system: biology, mechanisms and applications. Biochimie. 2015;117:119–128. doi: 10.1016/j.biochi.2015.03.025. [DOI] [PubMed] [Google Scholar]
- Rees HA, Liu DR. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet. 2018;19(12):770–788. doi: 10.1038/s41576-018-0059-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rees HA, Wilson C, Doman JL, Liu DR. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci Adv. 2019 doi: 10.1126/sciadv.aax5717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren B, Yan F, Kuang Y, Li N, Zhang D, Lin H, Zhou H. A CRISPR/Cas9 toolkit for efficient targeted base editing to induce genetic variations in rice. Sci China Life Sci. 2017;60(5):516–519. doi: 10.1007/s11427-016-0406-x. [DOI] [PubMed] [Google Scholar]
- Ren B, Yan F, Kuang Y, Li N, Zhang D, Zhou X, Lin H, Zhou H. Improved base editor for efficiently inducing genetic variations in rice with CRISPR/Cas9-guided hyperactive hAID mutant. Mol Plant. 2018;11(4):623–626. doi: 10.1016/j.molp.2018.01.005. [DOI] [PubMed] [Google Scholar]
- Ren B, Liu L, Li S, Kuang Y, Wang J, Zhang D, Zhou X, Lin H, Zhou H. Cas9-NG greatly expands the targeting scope of genome-editing toolkit by recognizing NG and other atypical PAMs in rice. Mol Plant. 2019 doi: 10.1016/j.molp.2019.03.010. [DOI] [PubMed] [Google Scholar]
- Rodríguez-Leal D, Lemmon ZH, Man J, Bartlett ME, Lippman ZB. Engineering quantitative trait variation for crop improvement by genome editing. Cell. 2017;171(2):470–480. doi: 10.1016/j.cell.2017.08.030. [DOI] [PubMed] [Google Scholar]
- Schormann N, Ricciardi R, Chattopadhyay D. Uracil-DNA glycosylases—structural and functional perspectives on an essential family of DNA repair enzymes. Protein Sci. 2014;23(12):1667–1685. doi: 10.1002/pro.2554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimatani Z, Kashojiya S, Takayama M, Terada R, Arazoe T, Ishii H, Teramura H, Yamamoto T, Komatsu H, Miura K, Ezura H, Nishida K, Ariizumi T, Kondo A. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat Biotechnol. 2017;35(5):441–443. doi: 10.1038/nbt.3833. [DOI] [PubMed] [Google Scholar]
- Shmakov S, et al. Discovery and functional characterization of diverse Class 2 CRISPR-Cas systems. Mol Cell. 2015;60:385–397. doi: 10.1016/j.molcel.2015.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Slaymaker IM, Gao L, Zetsche B, Scott DA, Yan WX, Zhang F. Rationally engineered Cas9 nucleases with improved specificity. Science. 2016;351(6268):84–88. doi: 10.1126/science.aad5227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steinert J, Schiml S, Fauser F, Puchta H. Highly efficient heritable plant genome engineering using Cas9 orthologues from Streptococcus thermophilus and Staphylococcus aureus. Plant J. 2015;84(1295–1305):724. doi: 10.1111/tpj.13078. [DOI] [PubMed] [Google Scholar]
- Strecker J, Jones S, Koopal B, Schmid-Burgk J, Zetsche B, Gao L, Makarova KS, Koonin EV, Zhang F. Engineering of CRISPR-Cas12b for human genome editing. Nat Commun. 2019;10(1):212. doi: 10.1038/s41467-018-08224-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan J, Zhang F, Karcher D, Bock R. Engineering of high-precision base editors for site-specific single nucleotide replacement. Nat Commun. 2019;10(1):439. doi: 10.1038/s41467-018-08034-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang X, Lowder LG, Zhang T, Malzahn AA, Zheng X, Voytas DF, Zhong Z, et al. A CRISPR-Cpf1 system for efficient genome editing and transcriptional repression in plants. Nat Plants. 2017;3:17018. doi: 10.1038/nplants.2017.18. [DOI] [PubMed] [Google Scholar]
- Tang X, Liu G, Zhou J, Ren Q, You Q, Tian L, Xin X, et al. A large-scale whole-genome sequencing analysis reveals highly specific genome editing by both Cas9 and Cpf1 nucleases in rice. Genome Biol. 2018 doi: 10.1101/292086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teng F, et al. Repurposing CRISPR-Cas12b for mammalian genome engineering. Cell Dis. 2018;4:63. doi: 10.1038/s41421-018-0069-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teng F, et al. Artificial sgRNAs engineered for genome editing with new Cas12b orthologs. Cell Discov. 2019;5:23. doi: 10.1038/s41421-019-0091-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian S, et al. Engineering herbicide-resistant watermelon variety through CRISPR/Cas9-mediated base-editing. Plant Cell Rep. 2018;37:1353–1356. doi: 10.1007/s00299-018-2299-0. [DOI] [PubMed] [Google Scholar]
- Vakulskas CA, et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat Med. 2018;24:1216. doi: 10.1038/s41591-018-0137-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veillet F, et al. Transgene-free genome editing in tomato and potato plants using Agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. Int J Mol Sci. 2019 doi: 10.3390/ijms20020402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang M, Mao Y, Lu Y, Tao X, Zhu JK. Multiplex gene editing in rice using the CRISPR-Cpf1 system. Mol Plant. 2017;10:1011–1013. doi: 10.1016/j.molp.2017.03.001. [DOI] [PubMed] [Google Scholar]
- Wang J, Meng X, Hu X, Sun T, Li J, Wang K, Yu H. xCas9 expands the scope of genome editing with reduced efficiency in rice. Plant Biotechnol J. 2019;17:1–3. doi: 10.1111/pbi.13053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waterworth MW, Drury GE, Bray CM, West CE. Repairing breaks in the plant genome: the importance of keeping it together. New Phytol. 2011;192:805–822. doi: 10.1111/j.1469-8137.2011.03926.x. [DOI] [PubMed] [Google Scholar]
- Wu X, Kriz AJ, Sharp PA. Target specificity of the CRISPR-Cas9 system. Quant Biol. 2014;2(2):59–70. doi: 10.1007/s40484-014-0030-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu D, Guan X, Zhu Y, Ren K, Huang Z. Structural basis of stringent PAM recognition by CRISPR-C2c1 in complex with sgRNA. Cell Res. 2017;27(5):705–708. doi: 10.1038/cr.2017.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu R, Qin R, Li H, Li D, Li L, Wei P, Yang J. Generation of targeted mutant rice using a CRISPR-Cpf1 system. Plant Biotechnol J. 2017;16:713–717. doi: 10.1111/pbi.12669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan F, Kuang Y, Ren B, Wang J, Zhang D, Lin H, Yang B, Zhou X, Zhou H. Highly efficient A.T to G.C base editing by Cas9n-guided tRNA adenosine deaminase in rice. Mol Plant. 2018;11:631–634. doi: 10.1016/j.molp.2018.02.008. [DOI] [PubMed] [Google Scholar]
- Yin X, Biswal AK, Dionora J, Perdigon KM, Balahadia CP, Mazumdar S, Chater C, et al. CRISPR-Cas9 and CRISPR-Cpf1 mediated targeting as a stomatal developmental gene EPL9 in rice. Plant Cell Rep. 2017;36:745–757. doi: 10.1007/s00299-017-2118-z. [DOI] [PubMed] [Google Scholar]
- Zetsche B, Gootenberg JS, Abudayyeh OO, Slaymaker IM, Makarova KS, et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. 2015;163(3):759–771. doi: 10.1016/j.cell.2015.09.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D, Zhang H, Li T, Chen K, Qiu JL, Gao C. Perfectly matched 20-nucleotide guide RNA sequences enable robust genome editing using high-fidelity SpCas9 nucleases. Genome Biol. 2017;18:191. doi: 10.1186/s13059-017-1325-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Q, Xing HL, Wang ZP, Zhang HY, Yang F, Wang XC, Chen QJ. Potential high-frequency off-target mutagenesis induced by CRISPR/Cas9 in Arabidopsis and its prevention. Plant Mol Biol. 2018;96:445–456. doi: 10.1007/s11103-018-0709-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang R, Liu J, Chai Z, Chen S, Bai Y, Zong Y, Chen K, Li J, Jiang L, Gao C. Generation of herbicide tolerance traits and a new selectable marker in wheat using base editing. Nat Plants. 2019;5(5):480. doi: 10.1038/s41477-019-0405-0. [DOI] [PubMed] [Google Scholar]
- Zhong Z, Zhang Y, You Q, Tang X, Ren Q, Liu S, Yang L, Wang Y, Liu X, Liu B, Zhang T, Zheng X, Le Y, Zhang Y, Qi Y. Plant genome editing using FnCpf1 and LbCpf1 nucleases at redefined and altered PAM sites. Mol Plant. 2018;11(7):999–1002. doi: 10.1016/j.molp.2018.03.008. [DOI] [PubMed] [Google Scholar]
- Zhong Z, Sretenovic S, Ren Q, Yang L, Bao Y, Qi C, Yuan M, He Y, Liu S, Liu X, Wang J, Huang L, Wang Y, Baby D, Wang D, Zhang T, Qi Y, Zhang Y. Improving plant genome editing with high-fidelity xCas9 and non-canonical PAM-targeting Cas9-NG. Mol Plant. 2019 doi: 10.1016/j.molp.2019.03.011. [DOI] [PubMed] [Google Scholar]
- Zhou C, Sun Y, Yan R, Liu Y, Zuo E, et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature. 2019;571(7764):275–278. doi: 10.1038/s41586-019-1314-0. [DOI] [PubMed] [Google Scholar]
- Zischewski J, Fischer R, Bortesi L. Detection of on-target and off-target mutations generated by CRISPR/Cas9 and other sequence-specific nucleases. Biotechnol Adv. 2017;35(1):95–104. doi: 10.1016/j.biotechadv.2016.12.003. [DOI] [PubMed] [Google Scholar]
- Zong Y, Wang Y, Li C, Zhang R, Chen K, Ran Y, Qiu JL, Wang D, Gao C. Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat Biotechnol. 2017;35(5):438–440. doi: 10.1038/nbt.3811. [DOI] [PubMed] [Google Scholar]
- Zong Y, Song Q, Li C, Jin S, Zhang D, Wang Y, Qiu JL, Gao C. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A. Nat Biotechnol. 2018 doi: 10.1038/nbt.4261. [DOI] [PubMed] [Google Scholar]
- Zuo E, Sun Y, Wei W, Yuan T, Ying W, Sun H, Yuan L, Steinmetz LM, Li Y, Yang H. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science. 2019;364(6437):289–292. doi: 10.1126/science.aav9973. [DOI] [PMC free article] [PubMed] [Google Scholar]