Sei sulla pagina 1di 17

Review Article

https://doi.org/10.1038/s41477-019-0461-5

The emerging and uncultivated potential of


CRISPR technology in plant science
Yingxiao Zhang1, Aimee A. Malzahn1, Simon Sretenovic1 and Yiping Qi   1,2*

The application of clustered regularly interspaced short palindromic repeats (CRISPR) for genetic manipulation has revolution-
ized life science over the past few years. CRISPR was first discovered as an adaptive immune system in bacteria and archaea,
and then engineered to generate targeted DNA breaks in living cells and organisms. During the cellular DNA repair process,
various DNA changes can be introduced. The diverse and expanding CRISPR toolbox allows programmable genome editing,
epigenome editing and transcriptome regulation in plants. However, challenges in plant genome editing need to be fully appre-
ciated and solutions explored. This Review intends to provide an informative summary of the latest developments and break-
throughs of CRISPR technology, with a focus on achievements and potential utility in plant biology. Ultimately, CRISPR will not
only facilitate basic research, but also accelerate plant breeding and germplasm development. The application of CRISPR to
improve germplasm is particularly important in the context of global climate change as well as in the face of current agricultural,
environmental and ecological challenges.

G
enerating targeted genetic changes in living cells and organ- engineering will likely head. We hope this Review provides a com-
isms has historically been a great challenge in many species. prehensive guide on the current status and some future directions of
The basic strategy to introduce genetic changes is to gen- CRISPR technology in plants.
erate double-strand breaks (DSBs) by a sequence-specific nucle-
ase (SSN) at the targeted genomic site. Repair of DSBs in somatic The diverse and expanding CRISPR toolbox
cells of higher eukaryotes is predominantly done through the non- CRISPR arrays exist in bacteria and archaea DNA sequences,
homologous end joining (NHEJ) pathway, which results in inser- consisting of short repeats separated by unique spacer sequences.
tions/deletions (indels). Homology-directed repair (HDR), as a Small clusters of genes encoding Cas proteins can be found around
minor pathway, helps achieve precise genomic changes. Early SSNs, CRISPR arrays. Cas proteins are used to classify diverse CRISPR
including zinc finger nucleases (ZFNs)1 and transcription activator- systems based on their phylogenetic, structural and functional
like effector nucleases (TALENs)2, rely on protein–DNA interac- characteristics. During CRISPR antiviral defence, if the pre-
tions to determine specificity, which are difficult to engineer and CRISPR RNA (pre-crRNA) processing and interference stages are
multiplex. In 2012, the clustered regularly interspaced short palin- accomplished by one single multifunctional protein, the CRISPR
dromic repeat (CRISPR)–CRISPR-associated (Cas) system was first system is grouped as class 2; otherwise, it is grouped as a class 1
used for programmable RNA-guided genome editing in vitro3. The system17. Each class can be divided into multiple types according
following year, genome editing using CRISPR was successfully dem- to their signature proteins: type I, III and IV belong to class 1, with
onstrated in a mammalian system4,5. CRISPR, as a versatile, simple Cas3, Cas10 and Csf1 as their respective signature proteins17; while
and inexpensive system for genetic manipulation, has since domi- type II (Cas9), type V (Cas12a–e (Cas12d and Cas12e are also
nated the genome editing field. known as CasY and CasX, respectively), Cas12g–i and Cas14a–c)
Since the first demonstration of CRISPR in plant genome edit- and type VI (Cas13a–d) belong to class 2 (refs. 18–23). Each type of
ing in 2013 (refs. 6–8) there has been much progress in basic plant CRISPR system can be further grouped into subtypes based on
science and crop improvement9,10. Numerous molecular tools and operon organization and Cas proteins at the CRISPR loci. Although
platforms have been developed to achieve plant genetic engineering, the classification of CRISPR systems is constantly under develop-
including targeted mutagenesis11–13, base editing14, precise editing by ment, the current established classification method will facilitate
HDR15 and transcriptional regulation16. In this Review, we describe our understanding of CRISPR systems and the discovery of new
the state-of-the-art development of CRISPR technologies that have Cas proteins.
been applied in plants or remain to be fully explored, which are
partly illustrated in Fig. 1. We cover available CRISPR nucleases and CRISPR–SpCas9 genome editing system. Cas9 is a class 2, type
their variants, expression systems for multiplexing, diverse ways II CRISPR system and has been adapted for genome editing in the
to achieve precise genome editing, epigenome editing and tran- vast majority of organisms. Targeted genome cleavage requires Cas9
scriptome regulation. We address specific considerations in plant to assemble with a single-guide RNA (sgRNA; a fusion of crRNA
genome editing, such as temperature sensitivity, transgene-free and trans-activating crRNA (tracrRNA)), then recognize and bind
editing, polyploid genome editing and germline editing through to desired DNA sequences followed by a protospacer adjacent motif
floral dip. Recent breakthroughs that combine CRISPR with other (PAM). Streptococcus pyogenes Cas9 (SpCas9) recognizes a very sim-
cutting-edge technologies in plant breeding are also presented. We ple PAM (NGG), making it the most commonly used CRISPR–Cas9
envision some new frontiers where CRISPR-based plant genome system. SpCas9 has been codon-optimized for different species,

Department of Plant Science and Landscape Architecture, University of Maryland, College Park, MD, USA. 2Institute for Bioscience and Biotechnology
1

Research, University of Maryland, Rockville, MD, USA. *e-mail: yiping@umd.edu

778 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
Plasmid dsDNA RNA RNP

Attachment Viral vector


Nanoparticle
PEG
Particle
Agrobacterium bombardment O H
H O
n

RNA editing

ºC Temperature sensitivity
Organelle genome editing
90
High activity 80
70
RNA base editing 60
50
40
Low activity 30
20

C
as
C
10

as
Mitochondria

12
C

12

b
as
Plastid

a
9
Cyto
plas
m
Introduce Nuc
leus
stop codon
Gene knockout
D
Regulatory elements Promoter 5′ UTR Exon1 Intron Exon2 3′ UTR Terminator

Precise genome editing (base editing)


D D D D D
Regulatory elements Promoter 5′ UTR Exon1 Intron Exon2 3′ UTR Terminator

Precise genome editing (HDR)


Regulatory elements Promoter 5′ UTR Exon1 Intron Exon2 3′ UTR Terminator

LHA RHA
Transcriptional regulation
D Edit splicing sites
Regulatory elements Promoter 5′ UTR Exon1 Intron Exon2 3′ UTR Terminator

D R

Cas Cas Dead FnCas9 Cas13 gRNA Deaminase Regulator ADAR


nickase Cas

Fig. 1 | Applications of CRISPR technology in plant cells. Outside the cell, CRISPR reagents can be delivered as plasmids, dsDNA, RNA and RNPs
through PEG-mediated transformation, particle bombardment or nanoparticles. Plasmids can also be delivered by Agrobacterium and viral vectors. In the
cytoplasm, CRISPR can cleave and edit RNA molecules through RNA-targeting Cas proteins, such as FnCas9 and Cas13, and the ADAR-derived RNA base
editor, respectively. CRISPR may also be introduced into organelles to edit organelle genomes. Commonly used Cas systems (Cas9, Cas12a and Cas12b)
are temperature sensitive, requiring higher temperatures for optimal activities. In the nucleus, CRISPR technology is applied to achieve gene knockout,
precise genome editing through base editing, HDR and transcriptional regulation. To knock out genes, CRISPR–Cas nucleases can be used to target coding
regions or regulatory elements. Base editors can be used to target coding regions (for example, to introduce stop codons) or regulatory elements. When
the DNA repair donor with the left homology arm (LHA) and the right homology arm (RHA) are introduced along CRISPR–Cas, precise genome editing
can be achieved through HDR. To regulate transcription, CRISPR–Cas nucleases and base editors can be used to edit regulatory elements or splicing sites.
Transcriptional regulation can also be achieved by using dead Cas proteins to recruit regulators to the promoter region, including activators, repressors,
DNA methyltransferase, demethylase and so on.

including human (Homo sapiens; hCas9)5, plant (pcoCas9 and (nCas9) is generated that only cleaves the targeting or non-targeting
Cas9p)6,24, Arabidopsis thaliana (AteCas9)25, maize (Zea mays; strand, respectively4,29,30. When both mutations are introduced, the
zCas9)26,27 and soybean (Glycine max; GmCas9)28, to improve the nuclease activity is abolished, resulting in a catalytically inactive or
expression level in vivo. By introducing the point mutations D10A dead Cas9 (dCas9)31. Cas9 nuclease, nCas9 and dCas9 can all be
in the RuvCI domain or H840A in the HNH domain, a Cas9 nickase applied to develop many versatile genome engineering tools.

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 779


Review Article NATURe PlAnTS

Expanding target ranges with Cas9 variants and orthologues. and Nicotiana tabacum)68,73,74, tomato (Solanum lycopersicum)73,
The PAM requirement of SpCas9 limits its targeting scope in a soybean74, cotton (Gossypium hirsutum)75 and citrus76.
genome. The PAM-interacting (PI) domain can be engineered Similar to Cas9, Cas12a also contains a recognition (REC) lobe
through rationale design and directed evolution to obtain mutated and a nuclease (NUC) lobe63. However, the introduction of muta-
versions with altered PAM requirements (Table 1). The SpCas9 tions at the catalytic residues in the RuvC domain of Cas12a abol-
VQR, EQR and VRER variants can robustly recognize NGA, ishes the cleavage of both DNA strands at target sites62,63. Therefore,
NGAG and NGCG PAMs, respectively32. Similarly, the QQR1 there is no Cas12a nickase currently available. Multiple versions
variant was engineered with highly specific binding to the NAAG of catalytically dead Cas12a (dCas12a) have been engineered and
PAM33. Using phage-assisted continuous evolution, xCas9 has been repurposed for different applications, including dAsCas12a, dLb-
developed, recognizing NG, GAA and GAT PAM sites34. In addi- Cas12a and DNase-dead Cas12a (namely ddCas12a)62,63,66,77. To
tion, a rationally engineered SpCas9-NG recognizes relaxed NG broaden the target ranges of Cas12a, mutations have been intro-
PAMs and outperforms xCas9 at sites with NGH PAMs35. Many of duced to the wedge and PI domains to generate RR and RVR vari-
these SpCas9 variants have been demonstrated in plants, such as ants78,79 (Table 1). Similar mutations have been further introduced
the VQR variant36,37 and the VRER variant36 in rice (Oryza sativa), into rice-codon-optimized LbCas12a and FnCas12a for genome
as well as xCas9 and SpCas9-NG in rice and Arabidopsis38–46. Both editing in plants; only LbCas12a variants successfully edit sites with
xCas9 and SpCas9-NG have shown lower editing activity than wild- altered PAMs in plants67,80. Recently, an enhanced AsCas12a variant
type SpCas9 at NGG PAMs while exhibiting higher activity at non- (enAsCas12a) has been engineered to recognize a broader range of
canonical NG PAMs39,40,46. Specifically, SpCas9-NG is superior to PAM sites and also exhibits enhanced activity at low temperatures81.
xCas9 at most target sites with NG PAMs, as well as AT-rich PAM To further expand the utility of Cas12a in genome editing, ortho-
sites such as GAT, GAA and CAA39,40. Although the performance of logues from diverse bacteria species have been surveyed62,82 (Table 1).
some Cas9 variants may be suboptimal in plants, further optimiza- Cas12a orthologues with editing activity at loosened or shorter
tion could help restore editing activity in a plant cellular environ- PAM sites include Moraxella bovoculi 237 Cas12a (MbCas12a),
ment to achieve efficient genome editing. M. bovoculi AAX08_00205 Cas12a (Mb2Cas12a), M. bovoculi
Alternative PAM recognition can also be achieved by harnessing AAX11_00205 Cas12a (Mb3Cas12a), Thiomicrospira sp. XS5 Cas12a
Cas9 orthologues from other bacterial and archaeal species (Table 1). (TsCas12a), Btyrivibrio sp. NC3005 Cas12a (BsCas12a), Helcococcus
Staphylococcus aureus Cas9 (SaCas9) recognizes an NNGRRT kunzii ATCC 51366 Cas12a (HkCas12a), Lachnospira pectinoschiza
PAM and is smaller in size compared to SpCas9, making it pre- strain 2789STDY5834886 Cas12a (LpCas12a), Pseudobutyrivibrio
ferred for virus-based delivery47. SaCas9 nickase48, dead SaCas9 ruminis CF1b Cas12a (PrCas12a) and Pseudobutyrivibrio xylaniv-
(ref. 47) and a KKH variant49 with a loosened PAM requirement orans strain DSM 10317 Cas12a (PxCas12a)62,79,82,83, which could
have been developed to expand the application of this system. potentially be useful for plant genome editing.
SaCas9 has been demonstrated in multiple plant species, including
Nicotiana benthamiana50, rice50, Arabidopsis51 and citrus52. Other Emerging DNA-targeting CRISPR nucleases. Cas12b, formerly
Cas9 orthologues that have been used for genome editing in mam- known as C2c1, is a class 2 type V-B endonuclease. Cas12b recog-
malian systems include Streptococcus thermophilus Cas9 (St1Cas9) nizes target sequences with a distal 5′-T-rich PAM sequence and
and St3Cas9 (ref. 53); Neisseria meningitidis Cas9 (NmCas9)54; generates staggered DSBs with 7-nt 5′-overhangs21,84. Like Cas9,
Francisella novicida Cas9 (FnCas9) and its RHA variant55, Cas12b requires both crRNA and tracrRNA for target recogni-
Treponema denticola Cas9 (TdCas9)56; Campylobacter jejuni Cas9 tion, which can be engineered as a sgRNA21. Like Cas12a, Cas12b
(CjCas9)57 and Streptococcus canis Cas9 (ScCas9)58, of which contains a REC lobe and a NUC lobe without a HNH domain85.
only St1Cas9 has been demonstrated to edit plant genomes Two Cas12b nucleases, Alicyclobacillus acidoterrestris Cas12b
(Arabidopsis)51. Brevibacillus laterosporus Cas9 (BlatCas9) has also (AacCas12b) and Bacillus thermoamylovorans Cas12b (BthCas12b),
been characterized and is used for maize genome editing59. Notably, have initially shown editing activity in vitro with a preference for
Cas9s from class 2 type II-A and II-C have shown RNA-guided sin- higher temperatures (48–50 ℃)21, which is not ideal for mamma-
gle-stranded RNA cleavage activity60. Moreover, by grafting the PI lian and plant genome editing. By exploring diverse Cas12b ortho-
domain of a Streptococcus macacae Cas9 (SmacCas9) to SpCas9, logues, Alicyclobacillus acidiphilus Cas12b (AaCas12b) has been
iSpy-macCas9 has been developed to recognize NAA PAM61. In discovered and shows efficient editing activity at a broader range
general, the availability of Cas9 orthologues increases the accessible of temperatures86. Later, Bacillus hisashii Cas12b (BhCas12b) was
target sites for genome editing. Cas9 orthologues can be used for screened from multiple Cas12b orthologues and further engineered
orthogonal genome engineering, benefiting from their distinctive to increase activity at lower temperatures87. The Cas12b orthologues
sgRNA structures. that possess nuclease activity at lower temperatures may be applied
for plant genome editing.
Cas12a is a distinct CRISPR system. Cas12a (formerly known as CasX (also known as Cas12e), which was discovered from pre-
Cpf1, CRISPR from Prevotella and Francisella 1), a class 2 type V viously uncultivated environmental microbial communities, is a
endonuclease, is a genome editing system that is distinct from Cas9 group of dual-RNA-guided Cas proteins targeting double-stranded
(ref. 62). First, the PAM requirement allows Cas12a to target T-rich DNA (dsDNA) with a 5′-TTCN PAM18,20. CasX requires a 20 nt
regions62. Second, Cas12a only requires a short crRNA (~42 nt), guide RNA (gRNA) and a tracrRNA, and its cleavage generates a
making Cas12a crRNA easy to synthesize, multiplex and engineer62. staggered-end break with an overhang of approximately 10 nt20.
In addition to DNA nuclease activity, Cas12a also possesses RNase Deltaproteobacteria CasX (DpbCasX) and Planctomycetes CasX
activity, which is able to process a CRISPR array for multiplexed (PlmCasX) have been shown to edit the genomes of Escherichia
genome editing62,63. Moreover, Cas12a generates a DSB with stag- coli and human cells20. Mutations can be introduced to the RuvC
gered ends distal from the PAM site, which allows continuous domain to generate deactivated CasX20. Additionally, another
cleavage of DNA and may promote NHEJ-based gene insertion62. CRISPR system, CasY (also known as Cas12d), has also been identi-
Cas12a is also considered more specific than wild-type SpCas9 fied but is less well characterized. CasY may recognize a 5′-TA PAM
in several biological systems64–67. Three Cas12a nucleases from and cleave dsDNA18.
Acidaminococcus spp. BV3L6 (AsCpf1), Lachnospiraceae bacterium Researchers further exploited class 1 CRISPR systems for genome
ND2006 (LbCpf1) and Francisella novicida U112 (FnCpf1) have been editing. A type I-E CRISPR system from Thermobifida fusca has
demonstrated in rice66–72, Arabidopsis73, tobacco (N. benthamiana been shown to achieve 13–60% editing efficiency in mammalian

780 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
Table 1 | Variants and orthologues of Cas9 and Cas12a
Cas Size (amino PAM Mutations Plants Features
acids)
SpCas9 (refs. 3–5) 1,368–1,424 NGG – Many plant species –
SpCas9 VQR 32
1,372 NGA D1135V/R1335Q/T1337R Rice 36,37
Altered PAM
SpCas9 EQR32 1,372 NGAG D1135E/R1335Q/T1337R – Altered PAM
SpCas9 VRER32 1,372 NGCG D1135V/G1218R/R1335E/T1337R Rice5 Altered PAM
SpCas9 D1135E4 1,372 NAG and NGA D1135E – Altered PAM
SpCas9 QQR1 (ref. 33) 1,372 NAAG G1218R/N1286Q/I1331F/D1332K/ – Altered PAM
R1333Q/R1335Q/T1337R
SpCas9-NG35 1,372 NG R1335V/L1111R/D1135V/G1218R/ Rice39,40,42,43 and Arabidopsis42 Altered PAM
E1219F/A1322R/T1337R
iSpy-macCas9 (ref. 61) 1,359 NAA R221K/N394K – Altered PAM
SpCas9-HF1 (ref. 253) 1,368 NGG N497A/R661A/Q695A/Q926A Rice259,260 and Arabidopsis261 Enhanced specificity
SpCas9 (K855A)254 1,424 NGG K855A – Enhanced specificity
eSpCas9 (1.0)254 1,424 NGG K810A/K1003A/R1060A Rice259 and Arabidopsis261 Enhanced specificity
eSpCas9 (1.1)254 1,424 NGG K848A/K1003A/R1060A Rice259 and Arabidopsis261 Enhanced specificity
HypaCas9 (ref. 255) 1,368 NGG N692A/M694A/Q695A/H698A Rice260 Enhanced specificity
eHF1-Cas9 (ref. 260
) 1,368 NGG N497A/R661A/Q695A/K848A/ Rice 260
Enhanced specificity
Q926A/K1003A/R1060A
eHypa-Cas9 (ref. 260) 1,368 NGG N692A/M694A/Q695A/H698A/ Rice260 Enhanced specificity
K848A/K1003A/R1060A
EvoCas9 (ref. 256) 1,368 NGG M495V/Y515N/K526E/R661Q – Enhanced specificity
Sniper-Cas9 (ref. 257) 1,372 NGG F539S/M763I/K890N – Enhanced specificity
HiFi Cas9 (ref. 258) 1,368 NGG R691A – Enhanced specificity
xCas9 3.7 (ref. 34) 1,368 NG, GAA A262T/R324L/S409I/E480K/ Rice38–40 Enhanced specificity
and GAT E543D/M694I/E1219V and altered PAM
SaCas9 (refs. 47,297) 1,053 NNGRRT – N. benthamiana50,298, –
Arabidopsis51, rice50 and citrus52
SaCas9 KKH49 1,053 NNNRRT E782K/N968K/R1015H – –
St1Cas9 (ref. 53) 1,122 NNAGAAW – Arabidopsis51 –
St3Cas9 (ref. 53) 1,393 NGGNG – – –
NmCas9 (ref. 54) 1,109 NNNNGATT – – –
FnCas9 (ref. 55) 1,629 NGG – – –
FnCas9 RHA55 1,632 YG E1369R/E1449H/R1556A – –
TdCas9 (ref. ) 56
1,423 NAAAAN – – –
CjCas9 (ref. 57) 984 NNNNACAC or – – –
NNNNRYAC
ScCas9 (ref. 58) 1,379 NNG – – –
SmacCas9 (ref. 61) 1,338 NAA – – –
BlatCas9 (ref. 59) 1,092 NNNNCND – Maize59 –
AsCas12a 62
1,307 TTTV – Rice66, N. benthamiana and –
tomato73, soybean and wild
tobacco74
AsCas12a RR78 1,307 TYCV and S542R/K607R – Altered PAM
CCCC
AsCas12a RVR78 1,307 TATV S542R/K548V/N552R – Altered PAM
enAsCas12a81 1,307 VTTV, TTTT, E174R/S542R/K548R – Altered PAM and
TTCN and TATV enhanced activity at
low temperature
LbCas12a62 1,228 TTTV – Rice66–72, Arabidopsis73,211, –
N. benthamiana and tomato73,
soybean and wild tobacco74,
cotton75, citrus76 and maize27
LbCas12a RR78 1,228 TYCV and G532R/K595R Rice67,80 Altered PAM
CCCC
Continued

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 781


Review Article NATURe PlAnTS

Table 1 | Variants and orthologues of Cas9 and Cas12a (continued)


Cas Size (amino PAM Mutations Plants Features
acids)
LbCas12a RVR78 1,228 TATV G532R/K538V/Y542R Rice67,80 Altered PAM
FnCas12a62,79
1,300 TTV, TTTV and – Rice67,68,70,72 and tobacco68 –
KYTV
FnCas12a RR79 1,300 TYCV and N607R/K671R Rice67 –
TCTV
FnCas12a RVR79 1,300 TWTV N607R/K613V/N617R Rice67 –
MbCas12a 62,79
1,373 TTV and TTTV – – –
MbCas12a RR79 1,373 TYCV and N576R/K637R – Altered PAM
TCTV
MbCas12a RVR79 1,373 TWTV N576R/K582V/N586R – Altered PAM

cells88. This system requires a multi-subunit cascade (CRISPR- RNA-targeting CRISPR nucleases. In addition to the aforemen-
associated complex for antiviral defence) and a crRNA to recognize tioned Cas systems that target DNA, Cas13 (formerly known as C2c2)
target sequences with a 5′-PAM; the helicase-nuclease enzyme Cas3 is a class 2 type VI CRISPR system targeting RNA101. Leptotrichia
is then recruited to degrade DNA. Cas3 generates a range of large shahii Cas13a (LshCas13a) recognizes a 22–28 nt target sequence
deletions up to 100 kb, which may be potentially useful for large- followed by a protospacer flanking sequence (PFS) of H (H denotes
scale genome manipulation and deletion screens. A, U or C), while no specific PFS requirement for Leptotrichia
Cas14 is a family of miniature nucleases (400–700 amino acids wadei Cas13a (LwaCas13a) and Prevotella sp. P5-125 Cas13b
long) that can cleave targeted single-stranded DNA (ssDNA) (PspCas13b)102,103 has been discovered. Like Cas12a, the RNase activ-
without PAM requirements19. Cas14 can be used for diagnosing ity of Cas13a allows it to process crRNA arrays, which can be used
diseases19 and may be also applied for ssDNA virus interference to target multiple RNAs simultaneously101. Interestingly, Cas13a
in plants. shows non-specific RNase activity that cleaves collateral RNA fol-
lowing initial binding to its target RNA in vitro and in bacteria101,104.
Repurposing CRISPR as a recruiting platform. The programmable By harnessing this special feature of Cas13a, the specific high-sensi-
DNA-binding property of CRISPR systems provides a powerful plat- tivity enzymatic reporter unlocking (SHERLOCK) method has been
form to recruit functional domains to desired regions of the genome developed to detect specific RNA and DNA sequences104. A catalyti-
through protein fusion or sgRNA–protein interactions (Fig. 2). cally deactivated Cas13 or dCas13 can be generated by introducing
Such functional domains can be fused to the C terminus or N ter- an alanine substitution to any of the four HEPN catalytic residues
minus of Cas proteins and may include FokI nuclease domains, (R597A, H602A, R1278A or H1283A)101. dCas13 has been used as a
transcriptional activators or repressors, epigenetic modifiers, deam- programmable RNA binding protein to achieve in vivo RNA track-
inases, fluorescent protein tags and so on. To increase the dosage ing102 and base editing103. In plants, gene expression knockdown
of the functional domains, the SUperNova tagging system (SunTag) using LwaCas13a has been achieved in rice protoplasts102. RNA virus
uses a repeat of GCN4 epitopes fused to dCas9, which are bound by interference using LshCas13a has been demonstrated in N. ben-
scFv antibodies fused to effectors89. Protein recruitment can also be thamiana105 and Arabidopsis106. Potentially more Cas13 orthologues
achieved using self-complementing split green fluorescent protein can be tested in plants to develop RNA targeting tools for transcrip-
(GFP)90 (Fig. 2a). In contrast to the protein fusion strategy, func- tional regulation, RNA base editing, RNA tracking and functional
tional domains can be recruited through RNA–protein interactions studies, pathogen detection and disease control.
(Fig. 2b). The versatility of Cas9 sgRNA allows for the modification
of the structural modules, including the upper stem, the first and Versatile and multiplexed CRISPR expression systems
the second hairpins and the 3'-end of the sgRNA, without compris- There are generally four strategies for in planta expression of the
ing binding efficiency. Several RNA-binding protein–RNA aptamer two CRISPR components, Cas protein and gRNA, and each strategy
systems have been used for CRISPR protein recruitment, includ- has its advantages and disadvantages (Fig. 3a). The most commonly
ing MCP–MS2 (refs. 29,91–93), PCP–PP7 (refs. 93,94), Com–Com95, λ used system is the mixed dual promoter system in which the Cas
N22 peptide–boxB96, as well as streptavidin and its binding RNA gene is driven by an RNA polymerase II (Pol II) promoter while
aptamer S1 (ref. 97), and so on. For the most widely used MCP– the gRNA is driven by an RNA polymerase III (Pol III) promoter107.
MS2 system, up to 16 MS2 loops have been added to the 3′-end Due to limitations of Pol III promoters, a dual Pol II promoter sys-
and hairpins of the sgRNA98. Moreover, an octet array of MS2 is tem has been developed, which resulted in a high transcription
added to the upper stem of the sgRNA to create the CRISPR–Sirius level of gRNAs66,108,109. Pol II promoters allow for potential spatial
system, resulting in more stable sgRNA secondary structures99. By and temporal control of expression, and usually outperform Pol III
fusing SunTag epitopes to MCP and using MS2 to recruit activators, promoters for generating long transcripts with multiplexed gRNAs.
a three-component repurposed technology for enhanced expres- This system can be further simplified to a compact single transcript
sion (TREE) system has been developed100. These versatile DNA- unit using only one promoter to drive both the Cas protein and
targeting and protein-recruiting CRISPR systems demonstrate that the gRNA110–113. A similar compact system expresses gRNAs in the
multiple approaches can be used simultaneously to maximize the intron region, allowing the gRNAs to be excised during splicing113,114.
number and types of functional domains, expanding genome engi- Finally, a bidirectional promoter can be used to drive Cas and gRNA
neering outcomes in plants for base editing and transcriptional reg- expression in opposite directions, with different 3′-untranslated
ulation, which are discussed later in this Review (in sections titled regions (UTRs) or terminators to regulate both components inde-
“Precise genome editing by base editors” and “Transcription regula- pendently. In the future, these four expression strategies may be fur-
tion and epigenome editing”). ther evaluated and applied for tailored applications in plants.

782 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
a Effector protein recruitment through protein fusions and interactions
C-terminus fusion N-terminus fusion SunTag GFP11

Antibody GFP1–10

Cas Cas Cas Cas


Peptide epitopes GFP11

b Effector protein recruitment through protein-RNA interactions


Stem loop modification

3′-end modification

RNA aptamer RNA interacting protein Effector protein

Examples: Examples: Examples: •Methyltransferase


•PP7 •BoxB •PCP •MCP •Activator •Histone modification protein
•Com •MS2 •Com •λ N22 peptide •Repressor •Biotinylated protein and nucleic acid
•Streptavidin •Deaminase •Florescent protein

Fig. 2 | Repurposing CRISPR as a recruiting platform. a, Effector protein recruitment through protein fusions and interactions. Effector proteins can
be fused to the C- or N-terminus of Cas. In the SunTag system, a repeat of epitopes is fused to the Cas protein, which is bound by antibodies fused to
effectors. In the self-complementing split fluorescent protein system, the 11th β-strand of GFP (GFP11) can be tandemly arrayed to recruit the rest of the
fluorescent protein (GFP1–10) fused to effectors. b, Effector protein recruitment through protein–RNA interactions. Protein-binding RNA aptamers (such
as PP7, MS2, Com and BoxB) can be incorporated into the upper stem, the first and second hairpins, and the 3′-end of the sgRNA. In the CRISPR–Sirius
system, an array of aptamers are added into the upper stem loop of the sgRNA. In the TREE system, the repeat of epitopes used in SunTag is recruited
through sgRNA.

One of the most significant advantages of the CRISPR system in many species with Cas9, including rice121, wheat (Triticum aes-
is its flexibility for multiplexing, typically achieved by simultane- tivum)122 and maize123. The fourth strategy is the Csy4-based exci-
ous delivery and expression of multiple gRNAs. It is possible to sion system where sgRNAs or crRNAs are flanked by Csy4 excision
deliver many gRNAs through ribonucleoproteins (RNPs) or particle sites and processed by the Csy4 endonuclease111,124. Furthermore, a
bombardment. However, since most plants rely on Agrobacterium- Drosha-mediated multiplexing system using sgRNA–short hairpin
mediated transfer DNA (T-DNA) transformation, the development RNA (shRNA) has been developed for genome editing in mamma-
of efficient multiplexing expression systems is required. The most lian cells125, but has not yet been demonstrated in plant cells. The
straightforward approach is to stack multiple gRNA expression aforementioned efficient multiplexing systems can be used to target
units where each unit contains a promoter and a terminator24,115–117. multiple genes and gene families simultaneously for knockout, base
This approach can result in large constructs with many repetitive editing and regulation as well as large DNA fragment deletion, gene
elements, which increases cloning difficulty while decreasing trans- insertion and replacement.
formation efficiency. Therefore, multiple alternative strategies have
been developed to achieve multiplexed genome editing in plants Precise genome editing by base editors
(Fig. 3b). One strategy is to harness the native CRISPR array expres- Base editing, using cytidine base editors (CBE) and adenine base
sion system62,118. This is the most compact expression system and editors (ABE), refers to programmable and irreversible conversion
has been successfully demonstrated in plants using Cas9 (refs. 112,119) of one target nucleotide into another without DSBs or a donor tem-
and Cas12a70. Notably, Cas9 CRISPR arrays may contain tandem plate126–128. The first generation of CBE (BE1) was engineered by fus-
sgRNAs112,119 or only crRNA, with the tracrRNA expressed sepa- ing cytidine deaminase rat apolipoprotein B mRNA editing enzyme
rately4. Another strategy is to use the ribozyme–gRNA–ribozyme (rAPOBEC1)129 with dCas9 (ref. 126). Cytosine (C) deamination con-
(RGR) system in which the gRNA is flanked by a hammerhead (HH) verts C to uracil (U) which is recognized by cell replication machin-
ribozyme and a hepatitis delta virus (HDV) ribozyme sequence108. ery as thymine (T), resulting in C-to-T transition126. However, the
Tandemly arraying the RGR units allows the expression of a single conversion of C-to-U is usually reversed by base excision repair
transcript followed by the precise processing of each unit, as dem- where the U is transformed to an apurinic/apyrimidinic (AP) site
onstrated in rice120. The third strategy is the polycistronic T-RNA– by DNA glycosylases, such as uracil DNA glycosylase (UDG)130.
gRNA system in which T-RNA is fused to each gRNA in a single To increase editing efficiency and purity, the second generation of
transcript unit, and the endogenous RNaseZ and RNaseP recognize CBE (BE2) incorporates a uracil DNA glycosylase inhibitor (UGI)
and cut out the T-RNAs after transcription, allowing for the forma- into BE1 to block UDG126,131, causing a shift to the mismatch repair
tion of each mature gRNA121. This T-RNA system has been used (MMR) pathway. Editing fidelity and efficiency can be enhanced

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 783


Review Article NATURe PlAnTS

a
CRISPR–Cas expression systems
Mixed dual promoter system
Pol II promoter Cas Terminator Pol II promoter gRNA Terminator
Dual Pol II promoter system
Pol II promoter Cas Terminator Pol II promoter gRNA Terminator
Single bidirectional promoter
Terminator Cas Bidirectional promoter gRNA Terminator

Single transcript unit system


Single promoter
Pol II promoter Cas PolyA gRNA Terminator
Single promoter with an intron
Pol II promoter Exon Intron gRNA Intron Exon Cas Terminator

b
Multiplexed gRNA expression systems
CRISPR array 6/12 nt linker 6/12 nt linker
Cas9 sgRNA
Promoter gRNA Scaffold gRNA Scaffold gRNA Scaffold Terminator
RNase III RNase III

Cas9 crRNA and tracrRNA


Promoter gRNA Scaffold cRNA Scaffold cRNA Scaffold Terminator Promoter tracrRNA
RNase III RNase III

Cas12a crRNA
Promoter Scaffold cRNA Scaffold cRNA Scaffold cRNA Scaffold Terminator

Cas12a Cas12a Cas12a

Polycistronic T-RNA–gRNA
Promoter T-RNA gRNA Scaffold T-RNA gRNA Scaffold T-RNA gRNA Scaffold Terminator
R

R
R

R
N

N
N

N
as

as

as
as

as

as
e

e
e

e
P

P
Z

HH–gRNA–HDV
Promoter HH gRNA Scaffold HDV HH gRNA Scaffold HDV HH gRNA Scaffold HDV Terminator

Cys4 system
Promoter Cys4 site gRNA Scaffold Cys4 site gRNA Scaffold Cys4 site gRNA Scaffold Cys4 site Terminator Promoter Cys4

Cys4 Cys4 Cys4

Drosha-mediated gRNA–shRNA
Promoter gRNA Scaffold shRNA gRNA Scaffold shRNA gRNA Scaffold Terminator
Drosha Drosha Drosha Drosha

Fig. 3 | Diverse CRISPR expression and multiplex systems. a, CRISPR–Cas expression systems. Cas and gRNA can be expressed under mixed dual
promoters, dual RNA Polymerase II (Pol II) promoters, a single bidirectional promoter and as a single transcript unit. b, Multiplexed gRNA expression
systems. Multiplexed gRNA can be expressed using CRISPR array, polycistronic T-RNA–gRNA, HH–gRNA–HDV, Cys4 and drosha-mediated
gRNA–shRNA systems.

with the addition of more free or fused UGIs, further demonstrating base editing132. Efficiencies of base editing can be also improved by
its importance132,133. Later generations of CBE (BE3 and BE4)126,132 use linker modification132, codon optimization and the incorporation of
nCas9 with the D10A mutation to activate MMR and induce cells to additional nuclear localization sequences134. The most commonly
repair the non-edited strand using the edited strand as a template. used cytidine deaminases include APOBEC1 (ref. 126), activation-
Further optimization of BEs includes the fusion of the Gam protein induced cytidine deaminase (AID)135, Petromyzon marinus cytosine
from bacteriophage Mu, which binds to the ends of DSBs and pro- deaminase 1 (PmCDA1)127 and APOBEC3A136,137. Since no natu-
tects them from degradation, thus reducing indel formation during rally known enzymes deaminate adenine (A) in DNA, E. coli TadA

784 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
(ecTadA) evolved to catalyse adenine deamination in ssDNA128. In to the non-target strand, probably due to asymmetrical Cas9 dis-
addition, many variants of CBE and ABE have been developed to sociation158. In general, longer homology arms promote HDR effi-
overcome limitations of current base editors, thus expanding the ciency159. Symmetric and asymmetric donor templates may have
toolbox for base editing. Diverse base editors and their composi- different effects on knock-in efficiencies157,158. Interestingly, the
tions, editing windows, types and efficiencies have been compre- gRNA-to-mutation distance may affect the efficiency and the zygos-
hensively reviewed recently138,139. ity of HDR consequences157. With limited HDR research in plants, it
Base editors have been optimized and utilized in many plant would be useful to compare donor types as well as lengths and posi-
species and, presently, the most widely used base editor is the cyti- tions of homology arms to improve HDR in plant cells.
dine base editor BE3 (refs. 140–144). In addition, many other base Another approach to increase donor availability is to sim-
editing systems have been demonstrated in plants. A cereal plant ply increase the amount of donor DNA delivered to the cell. In
codon-optimized APOBEC3A is able to generate C-to-T conver- plants, when CRISPR reagents are delivered into cells through
sions efficiently in wheat, rice and potato (Solanum tuberosum)145. Agrobacterium-mediated transformation, donor DNA fragments
SpCas9 and SaCas9 variants have also been incorporated into BEs can be flanked by the target sequences so that even if the donors
to expand the targeting scope in plants140. A human AID variant are integrated into the genome, they can be liberated by the nucle-
lacking a nuclear export signal has been successfully used for rice ase160,161. This method is considered much more efficient than using
base editing, both with and without a UGI (rBE9 and rBE5, respec- a circular plasmid162. To increase donor amount, free dsDNA or
tively)146. A PmCDA1-based CBE termed Target-AID has resulted ssDNA donors can be delivered alongside the CRISPR reagents
in efficient editing in rice and tomato147. A separate study has shown through particle bombardment155,163. A geminivirus-based rep-
that PmCDA1 has higher base editing activity in rice than rAPO- licon system can be used to amplify DNA donor in plant cells.
BEC1111. ABEs have also been demonstrated in rice140,148, Arabidopsis Initially delivered through Agrobacterium, the geminivirus system
and rapeseed (Brassica napus) protoplasts149. allows rolling circle replication of the donor to increase HDR effi-
In addition to DNA base editing, RNA base editing offers a prom- ciency154,164–166. With the development of CRISPR delivery methods,
ising tool for post-transcriptional regulation. Currently, RNA base more approaches, such as nanoparticles, will be available to deliver
editors utilize the adenosine deaminase acting on RNA (ADAR) to large quantities of DNA donors167,168.
convert A to inosine (I)150, which are guided by antisense oligoribo- Direct recruitment of donor DNA to the target site represents
nucleotides151 or the Cas13 system102,103. Application of RNA base another effective approach for improving HDR. One strategy is to
editors in plants has not yet been reported. facilitate the transportation of the donor sequence into the nucleus.
Optimization of base editing is still warranted to minimize or The 5′-end of donors can be modified by adding an NLS peptide
eliminate off-target effects152, confer more flexible targeting win- through the interaction between an RNA adaptor and a peptide
dows and increase editing activity. Engineering of novel base editors nucleic acid, which increases donor potency and HDR efficiency169.
would offer base changes in addition to C-to-T and A-to-G transi- Additionally, biotinylated donor DNA can be recruited to tar-
tions. Recently, a hypothetical model for a transversion (CG-to-GC) get sites through streptavidin–biotin interactions, where an S1m
base editor has been proposed by using an AID–UDG–enCas9– aptamer is added to the SpCas9 sgRNA stem loop to recruit strep-
Rev1* (where Rev1* indicates evolved Rev1 with PolI-like 5'-to-3' tavidin97. Similarly, avidin can be directly fused to Cas9 to recruit
exonuclease activity and desired processivity) fusion protein153, biotinylated donor DNA, thus increasing local donor availability170.
which could potentially further revolutionize base editing. More recently, a new covalent tethering method has been developed
through a fusion between HUH endonucleases and Cas9 (ref. 171).
Gene replacement and insertion When the recognition sequence is added to the ssODN, it can cova-
Although base editing allows the conversion of nucleotides within lently bind to the HUH endonucleases and increase HDR efficiency
a small window of plant genomes, larger, precise genomic changes, up to 30-fold171. While no similar studies have been reported in
such as gene replacements and targeted gene insertions, cannot be plant cells, these targeted donor recruitment strategies represent
achieved through base editing. HDR, also known as gene targeting promising avenues for achieving high HDR efficiency in plants.
(GT), is the major approach to introduce foreign genes or alter gene Interestingly, RNA molecules highly expressed from a DNA tem-
contents15. While GT in higher plants was extremely difficult for plate may also serve as donors for HDR in yeast172 and plants173,
decades, the utilization of SSNs has made GT much more acces- albeit with relatively low efficiencies, encouraging further explora-
sible25,154,155. When SSNs are transformed into plants to create DSBs, tion of using RNA donors to improve HDR in plants.
DNA fragments harbouring desired sequences can be introduced as
the donor templates to repair the DSBs. After DSB induction, gene Improving HDR by modulating repair pathways. HDR enhance-
replacements or insertions can be achieved through one of the fol- ment can be achieved by overexpression of key enzymes involved
lowing pathways: canonical NHEJ (cNHEJ), microhomology-medi- in this pathway. For instance, RS-1 treatment and RAD51 mRNA
ated end-joining (MMEJ) or HDR156. Cas9 and Cas12a have been injection have been shown to enhance Cas9- and TALEN-mediated
widely used to generate DSBs and co-introduced with repair tem- knock-in efficiency in rabbit embryos174,175. In plants, it has not been
plates for precise genome editing through HDR. Given HDR-based thoroughly investigated whether manipulating RAD51 expression
gene replacement is essential but challenging for precise genome can increase DSB-induced HDR. With a better understanding of
editing, more efforts have focused on improving HDR over utilizing plant HDR pathways, more enzymes involved in DNA repair, such
cNHEJ and MMEJ for gene replacement and insertion. as RAD54, RAD50, CAF1 and SMC6B (also known as MIM), may
be manipulated to enhance DSB-induced HDR efficiency176,177.
Improving HDR through donor design. Using a donor format HDR can also be promoted by using a nCas9 (ref. 158) since
can influence the rate of HDR and editing outcomes. Donor tem- single-stranded breaks induced by the nickase play a critical role
plates can be delivered as dsDNA, such as PCR products, linear- in promoting the HDR pathway. Moreover, nicks are not the sub-
ized or non-linearized plasmids or ssDNA. In mammalian systems, strates of the competing NHEJ pathway, which is usually triggered
PCR products and ssDNA outperform supercoiled plasmids in by DSBs. An in trans-paired nicking system has been developed
total knock-in efficiency, while ssDNA induces less off-target and to introduce nicks in both targeted genomic DNA and the donor
undesired integration than using dsDNA as the donor157. Notably, plasmid to achieve seamless homology-directed gene inser-
Cas9-induced DSB repair through HDR with single-stranded oligo- tion at a higher efficiency than using Cas9 nuclease178. nCas9-
deoxynucleotide (ssODN) is bias to use donor DNA complementary induced HDR has also been successfully demonstrated in plants25.

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 785


Review Article NATURe PlAnTS

Alternatively, NHEJ pathways can be inhibited genetically or However, more research is still needed for plants regarding the dis-
chemically. In mammalian cells when DNA LIGASE 4 (LIG4) covery of new regulators, optimization of targeting sites, develop-
is inhibited by RNA interference, SCR7 or the adenovirus 4 ment of tunable and inducible regulation, and large-scale regulation.
E1B55K and E4orf6 proteins, HDR efficiency can be signifi- Transcription can also be controlled by editing epigenetic marks.
cantly increased179,180. This also holds true when Ku70 and Ku80 Methyltransferases and acetylases can be recruited to alter the epi-
have been knocked down179. Two DNA-dependent protein kinase genetic modifications associated with regulation200–203. In plants,
catalytic subunit inhibitors, NU7441 and KU-0060648, have been DNA methylation193 and demethylation204 have been successfully
demonstrated to reduce NHEJ-mediated repair while increasing achieved using SunTag systems to silence and upregulate gene
HDR-mediated repair181. In plants, HDR enhancement has been expression, respectively. However, genome-wide hypermethylation
observed in Arabidopsis lig4 and ku70 mutants182 as well as in rice has been observed in plants193 and mammalian systems205 when
lig4 mutants183. While it is not desirable to use NHEJ mutants, it is using CRISPR-directed methyltransferases, raising potential con-
feasible to promote HDR with simultaneous knockdown of NHEJ cerns when using this tool for epigenome editing.
genes by RNA interference or CRISPR interference (CRISPRi), Gene expression levels can be regulated at the post-transcrip-
which will be introduced in the next section. tional level by RNA-targeting CRISPR systems such as Cas13 and
RNA-targeting Cas9s. Moreover, SpCas9 can target RNA when
Transcription regulation and epigenome editing presented with a PAM sequence on a separate oligonucleotide
CRISPR–Cas can be used to alter gene expression by editing regula- (PAMmer) alongside a gRNA206. In addition to SpCas9, dCas9 fused
tory elements, including promoters, transcription factors, enhanc- with an RNase can target RNA for degradation207. Furthermore,
ers and so on. For instance, editing regulatory elements can result in RNA sequences can be precisely changed with RNA base editing103.
a wide range of expression levels for a gene of interest, allowing for
the discovery and selection of novel alleles, dissecting the function- Plant genome editing challenges and opportunities
ality of gene regulatory elements and manipulation of quantitative The diverse CRISPR toolbox and advanced technologies provide
trait loci184,185. In addition, editing splicing sites can alter the expres- great platforms for plant genome engineering. However, challenges
sion of different gene isoforms, enabling the discovery of splice vari- still remain in the application of genome editing tools in plants.
ants and splicing mechanisms149,186,187. With the development of new technologies and knowledge, these
CRISPR–Cas can also be engineered for transcriptional regula- challenges could be overcome.
tion. Gene knockdown was first demonstrated with CRISPR–dCas9
by inhibiting the initial binding of the transcription machinery when Temperature sensitivity of Cas9 and Cas12a. Cas9 and Cas12a
targeting the promoter region or blocking RNA polymerase elonga- require higher temperatures to achieve optimal editing efficiency
tion when targeting the coding region31. Repression efficiency can in mammalian cells208, zebrafish209 and plants210,211. In Arabidopsis,
be improved by using CRISPR to recruit repressors, such as SRDX repeated high temperature treatments result in a dramatic increase
in plant cells115,188. These techniques are collectively referred to as of Cas9 efficiency212. Likewise, heat treatments result in effi-
CRISPRi. In contrast, when activators are recruited through CRISPR, cient Cas12a editing in Arabidopsis (29 °C) and maize (28 °C)211.
endogenous genes can be overexpressed in their native environment, Therefore, it is important to consider temperature when applying
which is termed CRISPR activation. Activators that have been used these CRISPR–Cas systems in plants.
for this purpose include the transactivation domain of the nuclear
factor-κB p65 subunit (p65AD), the herpes simplex viral protein Generation of transgene-free edited plants. When CRISPR is used
16 (VP16) and its tandem versions (VP64, VP128 or VP160)189, for crop improvement, it is desirable to obtain final products without
Epstein–Barr virus R transactivator (RTA)190, p300 (ref. 191), transgenes to minimize regulatory burden, foster public acceptance
activation domain from human heat-shock factor 1 (HSF1)91, tran- and mitigate potential ecological consequences. CRISPR transgenes
scription activation domain (TAD) from Xanthamonas transcrip- may be eliminated through breeding and screening the segregat-
tion activator-like effectors (TALEs) as well as plant activators EDLL ing populations, which can be aided by coupling CRISPR-encoding
and ERF2m (modified ERF2)192. Tethering multiple types of effector genes with fluorescent markers or herbicide susceptibility213,214.
proteins to the target site through prementioned protein recruiting However, this approach is not feasible for vegetatively propagated
systems (Fig. 2) can dramatically augment transcriptional regula- plants or plants with long life cycles, such as trees. It could be even
tion. For instance, successful activation has been achieved through more complicated for self-incompatible and polyploid plants.
VP64-p65AD-Rta (VPR)190, synergistic activation mediator (SAM)91, Another approach to achieve transgene-free genome editing is
SunTag89,191,193 and TREE systems100. Moreover, gene regulation is to generate mutated plants without transgene integration. DNA
also affected by the position of the target site194. Another route to or RNA encoding the CRISPR machinery can be introduced into
further modulate expression is to use multiple gRNAs to target one plants and transiently expressed to generate edits215,216. In addition,
gene, maximizing the accessible effector proteins195,196. Interestingly, CRISPR–Cas RNPs can be delivered into plant protoplasts through
gene repression and activation can be achieved at the same time polyethylene glycol (PEG)-mediated transformation, which has
using dead Cas activators targeting different sites197, which allows been demonstrated in Arabidopsis, wild tobacco (Nicotiana attenu-
for the manipulation of more complex pathways. Furthermore, ata), lettuce (Lactuca sativa), rice74,217, apple (Malus pumila), grape
since a shortened gRNA allows Cas binding without cleavage, (Vitis vinifera)218, Petunia × hybrida219 and soybean74. However,
programming gRNA length serves as an elegant strategy to switch regeneration of genome-edited plants from protoplasts poses a
between genome editing and transcriptional regulation with either huge challenge for many species and could potentially introduce
Cas9 (ref. 198) or Cas12a199. unwanted somaclonal variations derived from the lengthy tissue
CRISPRi and CRISPR activation technology also hold great culture process220. Alternatively, RNPs can be delivered into plant
promise in plant transcriptional regulation66,115,188. Tethering activa- tissues that are routinely used for plant regeneration (such as cal-
tors through protein recruiting systems, such as MS2–MCP92 and lus) by particle bombardment221,222. RNPs have been delivered into
SunTag193, have been shown to significantly improve plant transcrip- plant zygotes produced by in  vitro fertilization, which were later
tional activation. A 6TAL–VP128 (TAL refers to TALE TAD motif) used to regenerate mature plants without selection223. RNP delivery
activation system has been demonstrated in plants that outperforms eliminates the introduction of foreign DNA and shortens the expo-
VP64 when delivered by Cas9 fusion or MS2–MCP recruitment, sure time of genomic DNA to CRISPR reagents, hence minimizing
providing a highly efficient platform for plant gene upregulation192. random DNA integration and off-targeting effects in the genome.

786 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
a b c d e f
CRISPR with BBM and WUS Asexual propogation Simultaneous gene editing and Domestication Cas or dCas–spo11 Allele stacking
(co-transformed/pre-transformed) haploid induction by CRISPR promotes meiotic
recombination
2n 2n × 2n
C D
2n × 2n B
A
Gamete Elite line Haploid inducer Elite
mother cell line with CRISPR Cas or dCas– gRNA alleles
Callus Meristem reagent Spo11 lines lines
induction MiMe by CRISPR

Chromosome shuffling
and editing
Wild ancestor

by CRISPR
2n

Domestication
n

by CRISPR
2n

Haploid seeds with edits


Gamete
Subculture 2n
BBM1 Chromosome
MTL doubling
expression
mutation D
in egg cell
C
B
A
Elite
2n 2n alleles
2n 2n

Accelerated tissue culture- Targeted chromosomal Breeding without


dependent and -independent Generation of transgene-free Domesticated breaks promote segregation of
genome editing Maternal clone generation homozygous-edited elite lines crop recombination elite alleles

Fig. 4 | Revolutionizing plant breeding by combining CRISPR with other cutting-edge technologies. a, Transformation efficiency can be elevated
by co-transforming or pre-transforming BBM and WUS2 genes. Genome-edited plants can be generated through tissue culture-dependent regeneration
or tissue culture-independent meristem induction and subculture. b, Asexual propagation of genome-edited plants can be achieved by creating the
genotype MiMe followed by haploid introduction through ectopic expression of BBM1 in the egg cell or knocking out MTL. c, Simultaneous gene editing and
haploid induction can be achieved by crossing elite lines with haploid inducer lines carrying genes encoding the CRISPR system. Edited haploids
without CRISPR genes can be selected and used to make transgene-free double haploid lines. d, De novo domestication of crops can be achieved using
CRISPR in wild species, resulting in improved crops with desirable traits. e, Cas or dCas–spo11 can be used to promote meiotic recombination to break
tight linkage and facilitate breeding. f, Elite alleles located at different chromosomes can be stacked at one locus by CRISPR to avoid segregation
during breeding.

Recently, a transgene killer CRISPR system has been developed to Germline editing by floral dip transformation. Generation of
link suicide transgenes with CRISPR constructs and eliminate all germline-edited plants is important for downstream genetic and
transgenes in the genome of edited T1 rice plants224. trait analysis. Ironically, this remains a challenge for plants where
the convenient Agrobacterium-mediated floral dip method is used
Genome editing of polyploid plants. Polyploid plant species make to deliver CRISPR transgenes, such as Arabidopsis, not only due to
up a significant portion of major crops, including staple food, fruit low editing efficiency in germlines24,238,239, but also the dissociation
and industrial crops. Commonly planted and consumed polyploid of editing results in somatic and germinal cells240. Agrobacterium
crops include triploids (citrus, banana (Musa acuminata, M. balbisi- is thought to deliver CRISPR-carrying T-DNA into the egg cells.
ana and Musa × paradisiaca), seedless watermelon (Citrullus lana- Germline edits can be obtained only if CRISPR makes edits after
tus) and some varieties of apples); tetraploids (pasta wheat (Triticum Agrobacterium infection but before the first embryogenic cell divi-
durum), potato, cotton, canola (B. napus), rapeseed, peanut (Arachis sion. Edits occurring afterwards will most likely result in chimeric
hypogaea), tobacco, switchgrass (Panicum virgatum) and some vari- plants. To overcome this challenge, tissue-specific promoters have
eties of apple); hexaploids (Camelina sativa, bread wheat and oats been used for Cas9 expression to limit or boost genome editing in
(Avena sativa)); and octoploids (sugar cane (Saccharum officinarum) germinal cells, including egg cell-specific promoter EC1.2 (ref. 26),
and strawberry (Fragaria × ananassa)). Gene knockout efficiency is sporogenesis expression promoter SPOROCYTELESS241 and meio-
usually lower in polyploid plant species than diploids, as multiple sis I-specific promoter242, as well as CDC45, DMC1, SOP11, YAO
alleles must be edited simultaneously. Key factors for successful and RPS5A promoters, which are preferentially expressed in actively
polyploid plant genome editing include an efficient expression sys- dividing tissues243–246. HDR was successfully achieved in Arabidopsis
tem and a highly active Cas nuclease. To date, multiallelic genome when a sequential transformation method was used, which takes
editing has been achieved in several polyploid plant species, includ- advantage of preselected transgenic lines with high germline expres-
ing both model systems225,226 and crops221,227–236. Successful gene edit- sion of Cas9 (ref. 247). While the emerging oil crop Camelina faces
ing has been used to introduce valuable agronomic traits such as similar problems to Arabidopsis, it remains to be seen whether any
improved oil quality230 and disease resistance233,237. Many attempts of the aforementioned strategies could improve genome editing in
have even been made to produce transgene-free edited polyploid this species230.
plants through RNP delivery221 and selection-free methods234.
Although both approaches are labour- and time-consuming, they Off-target effects in plants. The off-target effect of Cas proteins
offer the potential to accelerate applied research in crops. However, is usually a major concern of CRISPR technology in applied sci-
in addition to gene knockout, precise genome editing through HDR ences, especially in gene therapies. Cas9-induced DSBs could result
in stable transgenic lines of polyploid plants remains challenging. in large deletions and genome rearrangements248. In plants, inten-
Besides crop improvement, genome editing in polyploid plants sive investigations have been carried out to detect off-target effects
also offers a platform for gene dosage studies in which edited lines by whole genome sequencing in Arabidopsis, rice, cotton and so
with varying copy numbers of functional genes can be obtained on220,249,250. Studies have revealed a high specificity of genome edit-
in one round of transformation and plant regeneration225,232. This ing in plants using wild-type Cas9 and Cas12a, showing that most
allows for quantitative studies of the relationship between genotype mutations found in edited plants are due to somaclonal variations.
and phenotype. However, whole genome sequencing has shown that CBEs, not

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 787


Review Article NATURe PlAnTS

a b

Gene family- or Single gene Whole genome library


Single gene
Whole genome library pathway-specific regulatory element
library CRISPR–Cas
library library Protoplast
transformation CRISPRi
CRISPRa

Plant transformation with CRISPR–Cas

Reporter
cell line

Selection Cell sorting

Expression level
6
4
2
0
W

Li

Li

Li
ne

ne

ne
T

Identify essential Study biological Study gene function Study regulatory Identify genes of interest by sequencing
genes problems and evolve genes elements

Fig. 5 | Genetic screens with CRISPR libraries in whole plants and plant cells. a, Genetic screens with CRISPR libraries in whole plants. gRNA libraries
targeting a whole genome, a gene family or pathway, a single gene coding sequence or regulatory elements can be transformed into plants along with
a chosen Cas nuclease or base editor. Collections of mutants can be generated to identify essential genes or study functionality of genes, regulatory
elements or in planta gene evolution. b, Genetic screens with CRISPR libraries in plant cells. gRNA libraries targeting the whole genome can be
transformed into plant cells such as protoplasts. If traits of interest can be linked to drug-resistant selective markers, protoplasts can be used for plant
regeneration on selective media. If traits of interest can be linked to fluorescent protein expression, cells can be sorted by fluorescence-activated cell
sorting. Selected or sorted cells can be further sequenced to identify genes or regulatory elements contributing to traits of interest.

ABEs, induce substantial genome-wide off-target effects in rice152; Although it is hard to eliminate the possibility of CRISPR-
therefore, their use may necessitate additional screening and purify- induced large chromosomal deletions, insertions, inversions
ing selection. Moreover, recent studies have shown that both CBEs and rearrangement in plant genomes, which are often difficult to
and ABEs induce transcriptome-wide RNA off-target modifications detect with commonly used sequencing technologies, the off-target
in human cells251,252. However, base editors can be engineered to sig- effects should not be an obstacle in plant genome editing based on
nificantly reduce their RNA editing activity251,252. the knowledge generated so far. One can not only avoid off-target
To minimize potential off-target mutations, paired nCas9s can mutations during the process of generating genome-edited plants,
be used30. Protein engineering has been used to reduce Cas bind- but also detect and screen out products with off-target mutations
ing affinity (thus increasing editing specificity), resulting in high through breeding and selection, if necessary.
fidelity versions of SpCas9 such as SpCas9-HF1-4 (ref. 253), SpCas9
(K855A) and eSpCas9 (1.1)254, HypaCas9 (ref. 255), xCas9 (ref. 34), Revolutionizing breeding by combining CRISPR with other
evoCas9 (ref. 256), sniper-Cas9 (ref. 257) and HiFi Cas9 (R691A) in cutting-edge technologies. Generating uniform plants harbour-
the context of RNP delivery258 (Table 1). In rice, eSpCas9 (1.0 and ing CRISPR-induced edits relies heavily on tissue culture-based
1.1) and SpCas9–HF1 have been demonstrated to maintain their embryogenesis or organogenesis, especially for many major crops.
on-target editing activity with enhanced specificity when using the This limits the application of CRISPR in recalcitrant or marginally
T-RNA–sgRNA processing system259. Genome editing with two transformable plant species and varieties. One approach to overcome
Cas9 variants, eHF1-Cas9 and eHypa-Cas9, have also been dem- this difficulty is to express plant growth-stimulating genes to boost
onstrated in rice260. Recently, xCas9 was shown to have a higher plant transformation and regeneration. With the overexpression of
targeting specificity than wild-type Cas9 in rice40. However, owing transcription factors Baby boom (BBM) and Wuschel2 (WUS2), pro-
to intrinsically lower nuclease activities for many high-fidelity found improvement of transformation efficiency was achieved in
SpCas9s in plants259,261, they may not be widely adopted for plant recalcitrant maize lines, sorghum (Sorghum bicolor), sugarcane and
genome editing before further improvement. Other approaches to indica rice262,263. Furthermore, BBM, WUS or similar factors may
minimize off-target effects include designing gRNAs with fewer even allow tissue culture-free genome editing (if they are induced
potential mismatch targets in a given genome. Additionally, limit- or applied ectopically) alongside genome editing reagent delivery
ing the exposure of the genome to CRISPR reagents, for example, to stimulate meristem tissue formation from somatic tissues, repre-
through transient expression and RNP transformation, can reduce senting a novel and effective approach to achieve genome editing in
the potential for off-target activity. many difficult plant species (Fig. 4a).

788 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
In plant breeding, it is preferable for desirable traits to be sta- editing in mammalian mitochondria has been achieved using
bly inheritable through generations. This is a big challenge for TALENs281 and ZFNs282. In order to implement CRISPR in mito-
hybrid crops benefiting from heterosis as well as self-incompatible chondria, many attempts have been made to import RNA into mito-
crops, since seed production will inevitably cause segregation. With chondria with little success283. One study showed mtDNA editing
CRISPR, asexual propagation of seeds has been achieved by creating using mitochondria localized Cas9 and normal gRNAs, suggesting
the genotype MiMe (mitosis instead of meiosis) followed by hap- that Cas9/gRNA RNPs might be preassembled before transpor-
loid introduction264,265. By knocking out three genes in Arabidopsis tation into mitochondria284. In plastids such as chloroplasts, the
(atosd1 atrec8 atspo11-1 triple mutants)266 and rice (ospair1-4 NHEJ repair pathway is thought to be absent278. While chloroplast
osrec8-3 ososd1-1 triple mutants)267, meiosis can be substituted by genomes can be edited through HDR without the use of any SSNs285,
mitosis to produce diploid gametes that are genetically identical to it is likely that HDR in plastids can be improved by introducing tar-
the parent. Haploid seeds can then be induced through multiple geted DSBs by SSNs, such as CRISPR. Currently, genome editing in
approaches to produce clonal progeny, including using haploid either plant mitochondria or plastids has not been achieved using
inducer lines for crossing, ectopic expression of BBM1 (a BBM-like SSNs, including CRISPR. However, a recent report on efficient chlo-
gene expressed in sperm cells) in the egg cell264 as well as knock- roplast transformation in CRISPR-generated Arabidopsis mutants
ing out the MATRILINEAL (MTL) gene265. CRISPR has facilitated (more sensitive to the selection agent) has provided more opportu-
the development of this new technology and allows for the stable nities in this exciting research area286.
inheritance of elite lines without the risk of segregation (Fig. 4b). Although components of genome editing reagents can be well
To obtain transgene-free homozygous genome-edited germ- designed, the editing outcomes, which highly rely on DNA repair
lines without tissue culture, two similar systems have been devel- choices, cannot be strictly controlled thus far. Edits generated by
oped using haploid induction-edit268 and haploid-inducer mediated cNHEJ and MMEJ pathways are strongly correlated with the target
genome editing (IMGE)269 (Fig. 4c). Haploid inducer lines are used sequence context287,288 and the broken ends generated by SSNs289,290.
to cross with the elite lines. The genome that will not pass to the hap- Through editing data profiling, machine learning and modeling,
loid progeny carries the CRISPR–Cas transgene, which allows for deletion and insertion patterns as well as probabilities can be pre-
the elimination of the transgene in the edited plants. Edited haploids dicted287,291,292. With a thorough understanding of editing results,
can be selected and subsequently used to make double haploid lines. gRNA can be better designed to achieve diverse goals. With the
Without any doubt, plant breeding will further benefit from differences in DNA repair pathways and preference, the prediction
CRISPR technology. Recently, several studies have demonstrated models and algorithms developed in certain types of human cells
the power of CRISPR in de novo plant domestication270–272 (Fig. 4d). may not be readily applicable to a specific plant species. This calls
In the future, CRISPR will speed up the plant breeding process when for future studies to understand DNA repair pathways in plants to
coupled with new ideas. For instance, Spo11, involved in generating better predict and manipulate editing outcomes.
DSBs for meiotic recombination, can be recruited through dead Cas CRISPR is not only a platform for genome editing and genetic
to break tight linkage among genes, which can help overcome link- manipulation, but also serves as a new avenue to localize defined
age drag in breeding273 (Fig. 4e). Moreover, CRISPR can be used to effector molecules (for example, protein, RNA or DNA) to specific
achieve chromosome shuffling to stack multiple elite alleles into one genomic and transcriptomic loci, allowing CRISPR to be repur-
tightly linked locus, thus preventing the segregation of elite alleles posed for many diverse applications (Fig. 2), such as visualizing
during breeding (Fig. 4f). With CRISPR genome editing tools, plant specific regions of DNA and RNA by recruiting fluorescent pro-
breeding will be elevated to a whole new era in a more precise and teins293–295, and purifying and isolating DNA- or RNA-associated
customized manner, which cannot be achieved through conven- proteins and nucleic acids296. In the future, CRISPR technology will
tional breeding and traditional genetic engineering. accelerate plant biology research, ultimately furthering the develop-
ment of new germplasm and even new crops. It will play a critical
Prospects role in addressing agricultural, environmental and ecological issues
The rapid development of CRISPR technology will facilitate numer- while promoting food security in the face of climate change and
ous applications in plant biology. For instance, the use of gRNA librar- population growth.
ies will enable large-scale screening to discover the functions of genes
and regulatory elements (Fig. 5a). With Agrobacterium-based T-DNA Received: 6 April 2019; Accepted: 24 May 2019;
delivery, pooled CRISPR libraries were used to generate a collection Published online: 15 July 2019
of mutants for gene families in tomato274 and nearly all coding genes
in rice275,276, providing valuable genetic resources for basic research References
1. Bibikova, M., Golic, M., Golic, K. G. & Carroll, D. Targeted chromosomal
and breeding. The scale can be shrunk to target a single gene, tilling cleavage and mutagenesis in Drosophila using zinc-finger nucleases. Genetics
the coding sequencing for in planta gene evolution277 or targeting reg- 161, 1169–1175 (2002).
ulatory elements to generate quantitative trait variation184. However, 2. Christian, M. et al. Targeting DNA double-strand breaks with TAL effector
it is time- and labour-consuming to deliver CRISPR libraries into nucleases. Genetics 186, 757–761 (2010).
plants for downstream applications. In the future, we envision the 3. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in
adaptive bacterial immunity. Science 337, 816–821 (2012).
possibility to deliver gRNA libraries into plant cells (such as proto- 4. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems.
plasts) to empower cell-based screens. By linking traits of interest to Science 339, 819–823 (2013).
selectable markers, such as drug resistance and fluorescent protein 5. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science
expression, individual plant cells can be selected or sorted for analysis 339, 823–826 (2013).
6. Li, J.-F. et al. Multiplex and homologous recombination-mediated genome
and regeneration (Fig. 5b). This will present an exciting opportunity
editing in Arabidopsis and Nicotiana benthamiana using guide RNA and
to link genotype to phenotype in a high-throughput manner. Cas9. Nat. Biotechnol. 31, 688–691 (2013).
Plants also store genetic information in two organelles: mito- 7. Nekrasov, V., Staskawicz, B., Weigel, D., Jones, J. D. G. & Kamoun, S.
chondria and plastids. Organelle genome editing could be used to Targeted mutagenesis in the model plant Nicotiana benthamiana using Cas9
manipulate metabolic pathways to gain desired phenotypes. In gen- RNA-guided endonuclease. Nat. Biotechnol. 31, 691–693 (2013).
8. Shan, Q. et al. Targeted genome modification of crop plants using a
eral, mitochondria can repair DSBs through both HDR and NHEJ CRISPR-Cas system. Nat. Biotechnol. 31, 686–688 (2013).
pathways278. However, in mammalian cells, mitochondria cannot 9. Chen, K., Wang, Y., Zhang, R., Zhang, H. & Gao, C. CRISPR/Cas genome
efficiently repair DSBs, leading to the degradation of mitochondrial editing and precision plant breeding in agriculture. Annu. Rev. Plant Biol.
DNA (mtDNA) and a shift of the heteroplasmic ratio279,280. Genome 70, 667–697 (2019).

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 789


Review Article NATURe PlAnTS
10. Zhang, Y., Massel, K., Godwin, I. D. & Gao, C. Applications and potential 42. Endo, M. et al. Genome editing in plants by engineered CRISPR–Cas9
of genome editing in crop improvement. Genome Biol. 19, 210 (2018). recognizing NG PAM. Nat. Plants 5, 14–17 (2018).
11. Ma, X., Zhu, Q., Chen, Y. & Liu, Y.-G. CRISPR/Cas9 platforms for 43. Ren, B. et al. Cas9-NG greatly expands the targeting scope of genome-
genome editing in plants: developments and applications. Mol. Plant 9, editing toolkit by recognizing NG and other atypical PAMs in rice. Mol.
961–974 (2016). Plant https://doi.org/10.1016/j.molp.2019.03.010 (2019).
12. Schindele, P., Wolter, F. & Puchta, H. Transforming plant biology and 44. Negishi, K. et al. An adenine base editor with expanded targeting scope
breeding with CRISPR/Cas9, Cas12 and Cas13. FEBS Lett. 592, using SpCas9-NGv1 in rice. Plant Biotechnol. J. https://doi.org/10.1111/
1954–1967 (2018). pbi.13120 (2019).
13. Yin, K., Gao, C. & Qiu, J.-L. Progress and prospects in plant genome 45. Wang, M. et al. Optimizing baseeditors for improved efficiency and
editing. Nat. Plants 3, 17107 (2017). expanded editing scope in rice. Plant Biotechnol. J. https://doi.org/10.1111/
14. Kim, J.-S. Precision genome engineering through adenine and cytosine base pbi.13124 (2019).
editing. Nat. Plants 4, 148–151 (2018). 46. Ge, Z. et al. Engineered xCas9 and SpCas9-NG variants broaden PAM
15. Huang, T. K. & Puchta, H. CRISPR/Cas-mediated gene targeting in plants: recognition sites to generate mutations in Arabidopsis plants. Plant
finally a turn for the better for homologous recombination. Plant Cell Rep. Biotechnol. J. https://doi.org/10.1111/pbi.13148 (2019).
38, 443–453 (2019). 47. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9.
16. Mahas, A., Neal Stewart, C. & Mahfouz, M. M. Harnessing CRISPR/Cas Nature 520, 186–191 (2015).
systems for programmable transcriptional and post-transcriptional 48. Friedland, A. E. et al. Characterization of Staphylococcus aureus Cas9: a
regulation. Biotechnol. Adv. 36, 295–310 (2018). smaller Cas9 for all-in-one adeno-associated virus delivery and paired
17. Makarova, K. S. et al. An updated evolutionary classification of CRISPR– nickase applications. Genome Biol. 16, 257 (2015).
Cas systems. Nat. Rev. Microbiol. 13, 722–736 (2015). 49. Kleinstiver, B. P. et al. Broadening the targeting range of Staphylococcus
18. Burstein, D. et al. New CRISPR–Cas systems from uncultivated microbes. aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol. 33,
Nature 542, 237–241 (2017). 1293–1298 (2015).
19. Harrington, L. B. et al. Programmed DNA destruction by miniature 50. Kaya, H., Mikami, M., Endo, A., Endo, M. & Toki, S. Highly specific
CRISPR-Cas14 enzymes. Science 362, 839–842 (2018). targeted mutagenesis in plants using Staphylococcus aureus Cas9. Sci. Rep. 6,
20. Liu, J.-J. et al. CasX enzymes comprise a distinct family of RNA-guided 26871 (2016).
genome editors. Nature 566, 218 (2019). 51. Steinert, J., Schiml, S., Fauser, F. & Puchta, H. Highly efficient heritable
21. Shmakov, S. et al. Discovery and functional characterization of diverse plant genome engineering using Cas9 orthologues from Streptococcus
Class 2 CRISPR-Cas systems. Mol. Cell 60, 385–397 (2015). thermophilus and Staphylococcus aureus. Plant J. 84, 1295–1305 (2015).
22. Shmakov, S. et al. Diversity and evolution of class 2 CRISPR–Cas systems. 52. Jia, H., Xu, J., Orbović, V., Zhang, Y. & Wang, N. Editing Citrus genome via
Nat. Rev. Microbiol. 15, 169–182 (2017). SaCas9/sgRNA system. Front. Plant Sci. 8, 2135 (2017).
23. Yan, W. X. et al. Functionally diverse type V CRISPR-Cas systems. Science 53. Müller, M. et al. Streptococcus thermophilus CRISPR-Cas9 systems
363, 88–91 (2019). enable specific editing of the human genome. Mol. Ther. 24,
24. Ma, X. et al. A robust CRISPR/Cas9 system for convenient, high-efficiency 636–644 (2016).
multiplex genome editing in monocot and dicot plants. Mol. Plant 8, 54. Lee, C. M., Cradick, T. J. & Bao, G. The Neisseria meningitidis CRISPR-Cas9
1274–1284 (2015). system enables specific genome editing in mammalian cells. Mol. Ther. 24,
25. Fauser, F., Schiml, S. & Puchta, H. Both CRISPR/Cas-based nucleases and 645–654 (2016).
nickases can be used efficiently for genome engineering in Arabidopsis 55. Hirano, H. et al. Structure and engineering of Francisella novicida Cas9.
thaliana. Plant J. 79, 348–359 (2014). Cell 164, 950–961 (2016).
26. Wang, Z.-P. et al. Egg cell-specific promoter-controlled CRISPR/Cas9 56. Esvelt, K. M. et al. Orthogonal Cas9 proteins for RNA-guided gene
efficiently generates homozygous mutants for multiple target genes in regulation and editing. Nat. Methods 10, 1116–21 (2013).
Arabidopsis in a single generation. Genome Biol. 16, 144 (2015). 57. Kim, E. et al. In vivo genome editing with a small Cas9 orthologue derived
27. Lee, K. et al. Activities and specificities of CRISPR/Cas9 and Cas12a from Campylobacter jejuni. Nat. Commun. 8, 14500 (2017).
nucleases for targeted mutagenesis in maize. Plant Biotechnol. J. 17, 58. Chatterjee, P., Jakimo, N. & Jacobson, J. M. Minimal PAM specificity of a
839–842 (2019). highly similar SpCas9 ortholog. Sci. Adv. 4, eaau0766 (2018).
28. Michno, J.-M. et al. CRISPR/Cas mutagenesis of soybean and Medicago 59. Karvelis, T. et al. Rapid characterization of CRISPR-Cas9 protospacer
truncatula using a new web-tool and a modified Cas9 enzyme. GM Crops adjacent motif sequence elements. Genome Biol. 16, 253 (2015).
Food 6, 243–252 (2015). 60. Rousseau, B. A., Hou, Z., Gramelspacher, M. J. & Zhang, Y. Programmable
29. Mali, P. et al. CAS9 transcriptional activators for target specificity screening RNA cleavage and recognition by a natural CRISPR-Cas9 system from
and paired nickases for cooperative genome engineering. Nat. Biotechnol. Neisseria meningitidis. Mol. Cell 69, 906–914 (2018).
31, 833–838 (2013). 61. Jakimo, N., Chatterjee, P., Nip, L. & Jacobson, J. M. A Cas9 with complete
30. Ran, F. A. et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced PAM recognition for adenine dinucleotides. Preprint at https://www.biorxiv.
genome editing specificity. Cell 154, 1380–1389 (2013). org/node/129129.abstract (2018).
31. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for 62. Zetsche, B. et al. Cpf1 Is a single RNA-guided endonuclease of a Class 2
sequence-specific control of gene expression. Cell 152, 1173–1183 (2013). CRISPR-Cas system. Cell 163, 759–771 (2015).
32. Kleinstiver, B. P. et al. Engineered CRISPR-Cas9 nucleases with altered 63. Yamano, T. et al. Crystal structure of Cpf1 in complex with guide RNA and
PAM specificities. Nature 523, 481–485 (2015). target DNA. Cell 165, 949–962 (2016).
33. Anders, C., Bargsten, K. & Jinek, M. Structural plasticity of PAM 64. Kim, D. et al. Genome-wide analysis reveals specificities of Cpf1
recognition by engineered variants of the RNA-guided endonuclease Cas9. endonucleases in human cells. Nat. Biotechnol. 34, 863–868 (2016).
Mol. Cell 61, 895–902 (2016). 65. Kleinstiver, B. P. et al. Genome-wide specificities of CRISPR-Cas Cpf1
34. Hu, J. H. et al. Evolved Cas9 variants with broad PAM compatibility and nucleases in human cells. Nat. Biotechnol. 34, 869–874 (2016).
high DNA specificity. Nature 556, 57–63 (2018). 66. Tang, X. et al. A CRISPR–Cpf1 system for efficient genome editing and
35. Nishimasu, H. et al. Engineered CRISPR-Cas9 nuclease with expanded transcriptional repression in plants. Nat. Plants 3, 17103 (2017).
targeting space. Science 361, 1259–1262 (2018). 67. Zhong, Z. et al. Plant genome editing using FnCpf1 and LbCpf1 nucleases
36. Hu, X. et al. Expanding the range of CRISPR/Cas9 genome editing in rice. at redefined and altered PAM sites. Mol. Plant 11, 999–1002 (2018).
Mol. Plant 9, 943–945 (2016). 68. Endo, A., Masafumi, M., Kaya, H. & Toki, S. Efficient targeted mutagenesis
37. Hu, X., Meng, X., Liu, Q., Li, J. & Wang, K. Increasing the efficiency of of rice and tobacco genomes using Cpf1 from Francisella novicida. Sci. Rep.
CRISPR-Cas9-VQR precise genome editing in rice. Plant Biotechnol. J. 16, 6, 38169 (2016).
292–297 (2017). 69. Hu, X., Wang, C., Liu, Q., Fu, Y. & Wang, K. Targeted mutagenesis in rice
38. Wang, J. et al. xCas9 expands the scope of genome editing with reduced using CRISPR-Cpf1 system. J. Genet. Genom. 44, 71–73 (2017).
efficiency in rice. Plant Biotechnol. J. 17, 709–711 (2019). 70. Wang, M., Mao, Y., Lu, Y., Tao, X. & Zhu, J. Multiplex gene editing in rice
39. Hua, K., Tao, X., Han, P., Wang, R. & Zhu, J.-K. Genome engineering in using the CRISPR-Cpf1 system. Mol. Plant 10, 1011–1013 (2017).
rice using Cas9 variants that recognize NG PAM sequences. Mol. Plant 71. Yin, X. et al. CRISPR-Cas9 and CRISPR-Cpf1 mediated targeting
https://doi.org/10.1016/j.molp.2019.03.009 (2019). of a stomatal developmental gene EPFL9 in rice. Plant Cell Rep. 36,
40. Zhong, Z. et al. Improving plant genome editing with high-fidelity xCas9 745–757 (2017).
and non-canonical PAM-targeting Cas9-NG. Mol. Plant https://doi. 72. Begemann, M. B. et al. Precise insertion and guided editing of higher plant
org/10.1016/j.molp.2019.03.011 (2019). genomes using Cpf1 CRISPR nucleases. Sci. Rep. 7, 11606 (2017).
41. Li, J. et al. Plant genome editing using xCas9 with expanded PAM 73. Bernabé‐Orts, J. M. et al. Assessment of Cas12a-mediated gene editing
compatibility. J. Genet. Genomics https://doi.org/10.1016/j.jgg.2019.03.004 efficiency in plants. Plant Biotechnol. J. https://doi.org/10.1111/pbi.13113
(2019). (2019).

790 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
74. Kim, H. et al. CRISPR/Cpf1-mediated DNA-free plant genome editing. Nat. 107. Lowder, L., Malzahn, A. & Qi, Y. Rapid evolution of manifold CRISPR
Commun. 8, 14406 (2017). systems for plant genome editing. Front. Plant Sci. 7, 1683 (2016).
75. Li, B. et al. Robust CRISPR/Cpf1(Cas12a) mediated genome editing in 108. Gao, Y. & Zhao, Y. Self-processing of ribozyme-flanked RNAs into guide
allotetraploid cotton (G. hirsutum). Plant Biotechnol. J. https://doi. RNAs in vitro and in vivo for CRISPR-mediated genome editing. J. Integr.
org/10.1111/pbi.13147 (2019). Plant Biol. 56, 343–349 (2014).
76. Jia, H., Orbović, V. & Wang, N. CRISPR-LbCas12a-mediated modification 109. Gao, Y. et al. Auxin binding protein 1 (ABP1) is not required for either
of citrus. Plant Biotechnol. J. https://doi.org/10.1111/pbi.13109 (2019). auxin signaling or Arabidopsis development. Proc. Natl Acad. Sci. USA 112,
77. Zhang, X. et al. Multiplex gene regulation by CRISPR-ddCpf1. Cell Discov. 2275–2280 (2015).
3, 17018 (2017). 110. Tang, X. et al. A single transcript CRISPR-Cas9 system for efficient genome
78. Gao, L. et al. Engineered Cpf1 variants with altered PAM specificities. Nat. editing in plants. Mol. Plant 9, 1088–1091 (2016).
Biotechnol. 35, 789 (2017). 111. Tang, X. et al. Single transcript unit CRISPR 2.0 systems for robust Cas9
79. Tóth, E. et al. Mb- and FnCpf1 nucleases are active in mammalian cells: and Cas12a mediated plant genome editing. Plant Biotechnol. J. https://doi.
activities and PAM preferences of four wild-type Cpf1 nucleases and org/10.1111/pbi.13068 (2018).
of their altered PAM specificity variants. Nucleic Acids Res. 46, 112. Wang, M. et al. Multiplex gene editing in rice with simplified CRISPR-Cpf1
10272–10285 (2018). and CRISPR-Cas9 systems: simplified single transcriptional unit CRISPR
80. Li, S. et al. Expanding the scope of CRISPR/Cpf1-mediated genome editing systems. J. Integr. Plant Biol. 60, 626–631 (2018).
in rice. Mol. Plant 11, 995–998 (2018). 113. Yoshioka, S., Fujii, W., Ogawa, T., Sugiura, K. & Naito, K. Development of a
81. Kleinstiver, B. P. et al. Engineered CRISPR–Cas12a variants with increased mono-promoter-driven CRISPR/Cas9 system in mammalian cells. Sci. Rep.
activities and improved targeting ranges for gene, epigenetic and base 5, 18341 (2015).
editing. Nat. Biotechnol. 37, 276 (2019). 114. Ding, D., Chen, K., Chen, Y., Li, H. & Xie, K. Engineering introns to
82. Zetsche, B. et al. A survey of genome editing activity for 16 Cpf1 orthologs. express RNA guides for Cas9- and Cpf1-mediated multiplex genome
Preprint at https://www.biorxiv.org/content/10.1101/134015v1.article-info editing. Mol. Plant 11, 542–552 (2018).
(2017). 115. Lowder, L. G. et al. A CRISPR/Cas9 toolbox for multiplexed plant genome
83. Teng, F. et al. Enhanced mammalian genome editing by new Cas12a editing and transcriptional regulation. Plant Physiol. 169, 971–985 (2015).
orthologs with optimized crRNA scaffolds. Genome Biol. 20, 15 (2019). 116. Vazquez-Vilar, M. et al. A modular toolbox for gRNA–Cas9 genome
84. Liu, L. et al. C2c1-sgRNA complex structure reveals RNA-guided DNA engineering in plants based on the GoldenBraid standard. Plant Methods
cleavage mechanism. Mol. Cell 65, 310–322 (2017). 12, 10 (2016).
85. Yang, H., Gao, P., Rajashankar, K. R. & Patel, D. J. PAM-dependent target 117. Xing, H.-L. et al. A CRISPR/Cas9 toolkit for multiplex genome editing in
DNA recognition and cleavage by C2c1 CRISPR-Cas endonuclease. Cell plants. BMC Plant Biol. 14, 327 (2014).
167, 1814–1828 (2016). 118. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems.
86. Teng, F. et al. Repurposing CRISPR-Cas12b for mammalian genome Science 339, 819–823 (2013).
engineering. Cell Discov. 4, 63 (2018). 119. Mikami, M., Toki, S. & Endo, M. In Planta processing of the SpCas9–gRNA
87. Strecker, J. et al. Engineering of CRISPR-Cas12b for human genome editing. complex. Plant Cell Physiol. 58, 1857–1867 (2017).
Nat. Commun. 10, 212 (2019). 120. Li, S. et al. Synthesis-dependent repair of Cpf1-induced double strand
88. Dolan, A. E. et al. Introducing a spectrum of long-range genomic deletions DNA breaks enables targeted gene replacement in rice. J. Exp. Bot. 69,
in human embryonic stem cells using Type I CRISPR-Cas. Mol. Cell 74, 4715–4721 (2018).
1–15 (2019). 121. Xie, K., Minkenberg, B. & Yang, Y. Boosting CRISPR/Cas9 multiplex
89. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A editing capability with the endogenous tRNA-processing system. Proc. Natl
protein-tagging system for signal amplification in gene expression and Acad. Sci. USA 112, 3570–3575 (2015).
fluorescence imaging. Cell 159, 635–646 (2014). 122. Wang, W., Akhunova, A., Chao, S. & Akhunov, E. Optimizing multiplex
90. Leonetti, M. D., Sekine, S., Kamiyama, D., Weissman, J. S. & Huang, B. A CRISPR/Cas9-based genome editing for wheat. Preprint at https://www.
scalable strategy for high-throughput GFP tagging of endogenous human biorxiv.org/content/10.1101/051342v1 (2016).
proteins. Proc. Natl Acad. Sci. USA 113, E3501–E3508 (2016). 123. Qi, W. et al. High-efficiency CRISPR/Cas9 multiplex gene editing using the
91. Konermann, S. et al. Genome-scale transcriptional activation by an glycine tRNA-processing system-based strategy in maize. BMC Biotechnol.
engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015). 16, 58 (2016).
92. Lowder, L. G. et al. Robust transcriptional activation in plants using 124. Tsai, S. Q. et al. Dimeric CRISPR RNA-guided FokI nucleases for highly
multiplexed CRISPR-Act2.0 and mTALE-Act systems. Mol. Plant 11, specific genome editing. Nat. Biotechnol. 32, 569–576 (2014).
245–256 (2018). 125. Yan, Q. et al. Multiplex CRISPR/Cas9-based genome engineering enhanced
93. Shao, S. et al. Long-term dual-color tracking of genomic loci by modified by Drosha-mediated sgRNA-shRNA structure. Sci. Rep. 6, 38970 (2016).
sgRNAs of the CRISPR/Cas9 system. Nucleic Acids Res. 44, e86 (2016). 126. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R.
94. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Programmable editing of a target base in genomic DNA without
Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. double-stranded DNA cleavage. Nature 533, 420–424 (2016).
Nat. Methods 12, 664–670 (2015). 127. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and
95. Zalatan, J. G. et al. Engineering complex synthetic transcriptional programs vertebrate adaptive immune systems. Science 353, aaf8729 (2016).
with CRISPR RNA scaffolds. Cell 160, 339–350 (2015). 128. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in
96. Ma, H. et al. Multiplexed labeling of genomic loci with dCas9 and engineered genomic DNA without DNA cleavage. Nature 551, 464 (2017).
sgRNAs using CRISPRainbow. Nat. Biotechnol. 34, 528–530 (2016). 129. Conticello, S. G. The AID/APOBEC family of nucleic acid mutators.
97. Carlson-Stevermer, J. et al. Assembly of CRISPR ribonucleoproteins with Genome Biol. 9, 229 (2008).
biotinylated oligonucleotides via an RNA aptamer for precise gene editing. 130. Kunz, C., Saito, Y. & Schär, P. DNA Repair in mammalian cells:
Nat. Commun. 8, 1711 (2017). Mismatched repair: variations on a theme. Cell. Mol. Life Sci. CMLS 66,
98. Qin, P. et al. Live cell imaging of low- and non-repetitive chromosome loci 1021–1038 (2009).
using CRISPR-Cas9. Nat. Commun. 8, 14725 (2017). 131. Mol, C. D. et al. Crystal structure of human uracil-DNA glycosylase in
99. Ma, H. et al. CRISPR-Sirius: RNA scaffolds for signal amplification in complex with a protein inhibitor: Protein mimicry of DNA. Cell 82,
genome imaging. Nat. Methods 15, 928 (2018). 701–708 (1995).
100. Kunii, A. et al. Three-component repurposed technology for enhanced 132. Komor, A. C. et al. Improved base excision repair inhibition and
expression: highly accumulable transcriptional activators via branched tag bacteriophage Mu Gam protein yields C:G-to-T: Abase editors with higher
arrays. CRISPR J. 1, 337–347 (2018). efficiency and product purity. Sci. Adv. 3, eaao4774 (2017).
101. Abudayyeh, O. O. et al. C2c2 is a single-component programmable 133. Wang, L. et al. Enhanced base editing by co-expression of free uracil DNA
RNA-guided RNA-targeting CRISPR effector. Science 353, aaf5573 (2016). glycosylase inhibitor. Cell Res. 27, 1289–1292 (2017).
102. Abudayyeh, O. O. et al. RNA targeting with CRISPR–Cas13. Nature 550, 134. Zafra, M. P. et al. Optimized base editors enable efficient editing in cells,
280–284 (2017). organoids and mice. Nat. Biotechnol. 36, 888–893 (2018).
103. Cox, D. B. T. et al. RNA editing with CRISPR-Cas13. Science 358, 135. Kuscu, C. & Adli, M. CRISPR-Cas9-AID base editor is a powerful
1019–1027 (2017). gain-of-function screening tool. Nat. Methods 13, 983–984 (2016).
104. Gootenberg, J. S. et al. Nucleic acid detection with CRISPR-Cas13a/C2c2. 136. Gehrke, J. M. et al. An APOBEC3A-Cas9 base editor with minimized
Science 356, 438–442 (2017). bystander and off-target activities. Nat. Biotechnol. 36, 977–982 (2018).
105. Aman, R. et al. RNA virus interference via CRISPR/Cas13a system in 137. Wang, X. et al. Efficient base editing in methylated regions with a human
plants. Genome Biol. 19, 1 (2018). APOBEC3A-Cas9 fusion. Nat. Biotechnol. 36, 946–949 (2018).
106. Aman, R. et al. Engineering RNA virus interference via the CRISPR/Cas13 138. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome
machinery in Arabidopsis. Viruses 10, 732 (2018). and transcriptome of living cells. Nat. Rev. Genet. 19, 770 (2018).

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 791


Review Article NATURe PlAnTS
139. Molla, K. A. & Yang, Y. CRISPR/Cas-mediated base editing: technical 169. Ghanta, K. S. et al. 5′ Modifications improve potency and efficacy of DNA
considerations and practical applications. Trends Biotechnol. https://doi. donors for precision genome editing. Preprint at https://www.biorxiv.org/
org/10.1016/j.tibtech.2019.03.008 (2019). content/10.1101/354480v1.full (2018).
140. Hua, K., Tao, X. & Zhu, J.-K. Expanding the base editing scope in rice by 170. Ma, M. et al. Efficient generation of mice carrying homozygous double-
using Cas9 variants. Plant Biotechnol. J. 17, 499–504 (2019). floxp alleles using the Cas9-Avidin/Biotin-donor DNA system. Cell Res. 27,
141. Li, J., Sun, Y., Du, J., Zhao, Y. & Xia, L. Generation of targeted point 578–581 (2017).
mutations in rice by a modified CRISPR/Cas9 system. Mol. Plant 10, 171. Aird, E. J., Lovendahl, K. N., Martin, A. S., Harris, R. S. & Gordon, W. R.
526–529 (2017). Increasing Cas9-mediated homology-directed repair efficiency through
142. Lu, Y. & Zhu, J.-K. Precise editing of a target base in the rice genome using covalent tethering of DNA repair template. Commun. Biol. 1, 54 (2018).
a modified CRISPR/Cas9 system. Mol. Plant 10, 523–525 (2017). 172. Keskin, H. et al. Transcript-RNA-templated DNA recombination and repair.
143. Ren, B. et al. A CRISPR/Cas9 toolkit for efficient targeted base editing to Nature 515, 436–439 (2014).
induce genetic variations in rice. Sci. China Life Sci. 60, 516–519 (2017). 173. Li, S. et al. Precise gene replacement in rice by RNA transcript-templated
144. Zong, Y. et al. Precise base editing in rice, wheat and maize with a homologous recombination. Nat. Biotechnol. 37, 445–450 (2019).
Cas9-cytidine deaminase fusion. Nat. Biotechnol. 35, 438–440 (2017). 174. Jayathilaka, K. et al. A chemical compound that stimulates the human
145. Zong, Y. et al. Efficient C-to-T base editing in plants using a fusion of homologous recombination protein RAD51. Proc. Natl Acad. Sci. USA 105,
nCas9 and human APOBEC3A. Nat. Biotechnol. 36, 950–953 (2018). 15848–15853 (2008).
146. Ren, B. et al. Improved base editor for efficiently inducing genetic variations 175. Song, J. et al. RS-1 enhances CRISPR/Cas9- and TALEN-mediated knock-in
in rice with CRISPR/Cas9-guided hyperactive hAID mutant. Mol. Plant 11, efficiency. Nat. Commun. 7, 10548 (2016).
623–626 (2018). 176. Schuermann, D., Molinier, J., Fritsch, O. & Hohn, B. The dual nature of
147. Shimatani, Z. et al. Targeted base editing in rice and tomato using homologous recombination in plants. Trends Genet. 21, 172–181 (2005).
a CRISPR-Cas9 cytidine deaminase fusion. Nat. Biotechnol. 35, 177. Shaked, H., Melamed-Bessudo, C. & Levy, A. A. High-frequency gene
441–443 (2017). targeting in Arabidopsis plants expressing the yeast RAD54 gene. Proc. Natl
148. Yan, F. et al. Highly efficient A·T to G·C base editing by Cas9n-guided Acad. Sci. USA 102, 12265–12269 (2005).
tRNA adenosine deaminase in rice. Mol. Plant 11, 631–634 (2018). 178. Chen, X. et al. In trans paired nicking triggers seamless genome editing
149. Kang, B.-C. et al. Precision genome engineering through adenine base without double-stranded DNA cutting. Nat. Commun. 8, 657 (2017).
editing in plants. Nat. Plants 4, 427–431 (2018). 179. Chu, V. T. et al. Increasing the efficiency of homology-directed repair for
150. Matthews, M. M. et al. Structures of human ADAR2 bound to dsRNA CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat.
reveal base-flipping mechanism and basis for site selectivity. Nat. Struct. Biotechnol. 33, 543–548 (2015).
Mol. Biol. 23, 426–433 (2016). 180. Maruyama, T. et al. Increasing the efficiency of precise genome editing with
151. Montiel-Gonzalez, M. F., Vallecillo-Viejo, I., Yudowski, G. A. & Rosenthal, CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat. Biotechnol.
J. J. C. Correction of mutations within the cystic fibrosis transmembrane 33, 538–542 (2015).
conductance regulator by site-directed RNA editing. Proc. Natl Acad. Sci. 181. Robert, F., Barbeau, M., Éthier, S., Dostie, J. & Pelletier, J. Pharmacological
USA 110, 18285–18290 (2013). inhibition of DNA-PK stimulates Cas9-mediated genome editing. Genome
152. Jin, S. et al. Cytosine, but not adenine, base editors induce genome-wide Med. 7, 93 (2015).
off-target mutations in rice. Science 364, 292–295 (2019). 182. Qi, Y. et al. Increasing frequencies of site-specific mutagenesis and gene
153. Gajula, K. S. Designing an elusive C•G→G•C CRISPR base editor. Trends targeting in Arabidopsis by manipulating DNA repair pathways. Genome
Biochem. Sci. 44, 91–94 (2019). Res. 23, 547–554 (2013).
154. Čermák, T., Baltes, N. J., Čegan, R., Zhang, Y. & Voytas, D. F. High- 183. Endo, M., Mikami, M. & Toki, S. Biallelic gene targeting in rice. Plant
frequency, precise modification of the tomato genome. Genome Biol. 16, Physiol. 170, 667–677 (2016).
232 (2015). 184. Rodríguez-Leal, D., Lemmon, Z. H., Man, J., Bartlett, M. E. & Lippman, Z.
155. Sun, Y. et al. Engineering herbicide-resistant rice plants through CRISPR/ B. Engineering quantitative trait variation for crop improvement by genome
Cas9-mediated homologous recombination of Acetolactate Synthase. Mol. editing. Cell 171, 470–480 (2017).
Plant 9, 628–631 (2016). 185. Zhang, H. et al. Genome editing of upstream open reading frames enables
156. Malzahn, A., Lowder, L. & Qi, Y. Plant genome editing with TALEN and translational control in plants. Nat. Biotechnol. 36, 894–898 (2018).
CRISPR. Cell Biosci. 7, 21 (2017). 186. Li, Z., Xiong, X., Wang, F., Liang, J. & Li, J.-F. Gene disruption through
157. Li, H. et al. Design and specificity of long ssDNA donors for CRISPR-based base editing-induced messenger RNA missplicing in plants. New Phytol.
knock-in. Preprint at https://www.biorxiv.org/content/10.1101/178905v1 222, 1139–1148 (2019).
(2017). 187. Xue, C., Zhang, H., Lin, Q., Fan, R. & Gao, C. Manipulating mRNA splicing
158. Richardson, C. D., Ray, G. J., DeWitt, M. A., Curie, G. L. & Corn, J. E. by base editing in plants. Sci. China Life Sci. 61, 1293–1300 (2018).
Enhancing homology-directed genome editing by catalytically active and 188. Piatek, A. et al. RNA-guided transcriptional regulation in planta via
inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, synthetic dCas9-based transcription factors. Plant Biotechnol. J. 13, 578–589
339–344 (2016). (2015).
159. Baker, O. et al. The contribution of homology arms to nuclease-assisted 189. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of
genome engineering. Nucleic Acids Res. 45, 8105–8115 (2017). transcription in eukaryotes. Cell 154, 442–451 (2013).
160. Fauser, F. et al. In planta gene targeting. Proc. Natl Acad. Sci. USA 109, 190. Chavez, A. et al. Highly efficient Cas9-mediated transcriptional
7535–7540 (2012). programming. Nat. Methods 12, 326–328 (2015).
161. Schiml, S., Fauser, F. & Puchta, H. The CRISPR/Cas system can be used as 191. Zhang, X. et al. Gene activation in human cells using CRISPR/Cpf1-p300
nuclease for in planta gene targeting and as paired nickases for directed and CRISPR/Cpf1-SunTag systems. Protein Cell 9, 380–383 (2018).
mutagenesis in Arabidopsis resulting in heritable progeny. Plant J. 80, 192. Li, Z. et al. A potent Cas9-derived gene activator for plant and mammalian
1139–1150 (2014). cells. Nat. Plants 3, 930–936 (2017).
162. Zhang, J.-P. et al. Efficient precise knockin with a double cut HDR donor 193. Papikian, A., Liu, W., Gallego-Bartolomé, J. & Jacobsen, S. E. Site-specific
after CRISPR/Cas9-mediated double-stranded DNA cleavage. Genome Biol. manipulation of Arabidopsis loci using CRISPR-Cas9 SunTag systems. Nat.
18, 35 (2017). Commun. 10, 729 (2019).
163. Li, J. et al. Efficient allelic replacement in rice by gene editing: A case study 194. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene
of the NRT1.1B gene. J. Integr. Plant Biol. 60, 536–540 (2018). repression and activation. Cell 159, 647–661 (2014).
164. Baltes, N. J., Gil-Humanes, J., Cermak, T., Atkins, P. A. & Voytas, D. F. 195. Cheng, A. W. et al. Multiplexed activation of endogenous genes by
DNA replicons for plant genome engineering. Plant Cell 26, CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 23,
151–163 (2014). 1163–1171 (2013).
165. Dahan‐Meir, T. et al. Efficient in planta gene targeting in tomato using 196. Farzadfard, F., Perli, S. D. & Lu, T. K. Tunable and multifunctional
geminiviral replicons and the CRISPR/Cas9 system. Plant J. 95, eukaryotic transcription factors based on CRISPR/Cas. ACS Synth. Biol. 2,
5–16 (2018). 604–613 (2013).
166. Wang, M. et al. Gene targeting by homology-directed repair in rice using a 197. Deaner, M., Mejia, J. & Alper, H. S. Enabling graded and large-scale
geminivirus-based CRISPR/Cas9 system. Mol. Plant 10, 1007–1010 (2017). multiplex of desired genes using a dual-mode dCas9 activator in
167. Cunningham, F. J., Goh, N. S., Demirer, G. S., Matos, J. L. & Landry, M. P. Saccharomyces cerevisiae. ACS Synth. Biol. 6, 1931–1943 (2017).
Nanoparticle-mediated delivery towards advancing plant genetic 198. Kiani, S. et al. Cas9 gRNA engineering for genome editing, activation and
engineering. Trends Biotechnol. 36, 882–897 (2018). repression. Nat. Methods 12, 1051–1054 (2015).
168. Demirer, G. S. et al. High aspect ratio nanomaterials enable delivery of 199. Breinig, M. et al. Multiplexed orthogonal genome editing
functional genetic material without DNA integration in mature plants. Nat. and transcriptional activation by Cas12a. Nat. Methods 16,
Nanotechnol. 14, 456–464 (2019). 51–54 (2019).

792 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants


NATURe PlAnTS Review Article
200. Braun, S. M. G. et al. Rapid and reversible epigenome editing by 230. Jiang, W. Z. et al. Significant enhancement of fatty acid composition in
endogenous chromatin regulators. Nat. Commun. 8, 560 (2017). seeds of the allohexaploid, Camelina sativa, using CRISPR/Cas9 gene
201. Huang, Y.-H. et al. DNA epigenome editing using CRISPR-Cas SunTag- editing. Plant Biotechnol. J. 15, 648–657 (2017).
directed DNMT3A. Genome Biol. 18, 176 (2017). 231. Liu, Y. et al. Targeted mutagenesis in tetraploid switchgrass (Panicum
202. Liao, H.-K. et al. In vivo target gene activation via CRISPR/Cas9-mediated virgatum L.) using CRISPR/Cas9. Plant Biotechnol. J. 16, 381–393 (2018).
trans-epigenetic modulation. Cell 171, 1495–1507 (2017). 232. Morineau, C. et al. Selective gene dosage by CRISPR-Cas9 genome editing
203. Morita, S. et al. Targeted DNA demethylation in vivo using dCas9–peptide in hexaploid Camelina sativa. Plant Biotechnol. J. 15, 729–739 (2017).
repeat and scFv–TET1 catalytic domain fusions. Nat. Biotechnol. 34, 233. Wang, Y. et al. Simultaneous editing of three homoeoalleles in hexaploid
1060–1065 (2016). bread wheat confers heritable resistance to powdery mildew. Nat.
204. Gallego-Bartolomé, J. et al. Targeted DNA demethylation of the Arabidopsis Biotechnol. 32, 947–951 (2014).
genome using the human TET1 catalytic domain. Proc. Natl Acad. Sci. USA 234. Zhang, Y. et al. Efficient and transgene-free genome editing in wheat
115, E2125–E2134 (2018). through transient expression of CRISPR/Cas9 DNA or RNA. Nat. Commun.
205. Galonska, C. et al. Genome-wide tracking of dCas9-methyltransferase 7, 12617 (2016).
footprints. Nat. Commun. 9, 597 (2018). 235. Wang, W. et al. Transgenerational CRISPR-Cas9 activity facilitates multiplex
206. O’Connell, M. R. et al. Programmable RNA recognition and cleavage by gene editing in allopolyploid wheat. CRISPR J. 1, 65–74 (2018).
CRISPR/Cas9. Nature 516, 263–266 (2014). 236. Zhang, Z. et al. Development of an Agrobacterium-delivered CRISPR/Cas9
207. Batra, R. et al. Elimination of toxic microsatellite repeat expansion RNA by system for wheat genome editing. Plant Biotechnol. J. https://doi.
RNA-targeting Cas9. Cell 170, 899–912 (2017). org/10.1111/pbi.13088 (2019).
208. Xiang, G., Zhang, X., An, C., Cheng, C. & Wang, H. Temperature effect 237. Jia, H. et al. Genome editing of the disease susceptibility gene CsLOB1
on CRISPR-Cas9 mediated genome editing. J. Genet. Genom. 44, in citrus confers resistance to citrus canker. Plant Biotechnol. J. 15,
199–205 (2017). 817–823 (2017).
209. Moreno-Mateos, M. A. et al. CRISPR-Cpf1 mediates efficient homology- 238. Fauser, F., Schiml, S. & Puchta, H. Both CRISPR/Cas-based nucleases and
directed repair and temperature-controlled genome editing. Nat. Commun. nickases can be used efficiently for genome engineering in Arabidopsis
8, 2024 (2017). thaliana. Plant J. 79, 348–359 (2014).
210. LeBlanc Chantal. et al. Increased efficiency of targeted mutagenesis by 239. Feng, Z. et al. Efficient genome editing in plants using a CRISPR/Cas
CRISPR/Cas9 in plants using heat stress. Plant J. 93, 377–386 (2018). system. Cell Res. 23, 1229–1232 (2013).
211. Malzahn, A. A. et al. Application of CRISPR-Cas12a temperature sensitivity 240. Shan, Q. et al. ZFN, TALEN and CRISPR-Cas9 mediated homology
for improved genome editing in rice, maize, and Arabidopsis. BMC Biol. 17, directed gene insertion in Arabidopsis: a disconnect between somatic and
9 (2019). germinal cells. J. Genet. Genom. 45, 681–684 (2018).
212. LeBlanc, C. et al. Increased efficiency of targeted mutagenesis by CRISPR/ 241. Mao, Y. et al. Development of germ-line-specific CRISPR-Cas9 systems to
Cas9 in plants using heat stress. Plant J. 93, 377–386 (2018). improve the production of heritable gene modifications in Arabidopsis.
213. Gao, X., Chen, J., Dai, X., Zhang, D. & Zhao, Y. An effective strategy for Plant Biotechnol. J. 14, 519–532 (2016).
reliably isolating heritable and Cas9-free Arabidopsis mutants generated 242. Eid, A., Ali, Z. & Mahfouz, M. M. High efficiency of targeted mutagenesis
by CRISPR/Cas9-mediated genome editing. Plant Physiol. 171, in Arabidopsis via meiotic promoter-driven expression of Cas9
1794–1800 (2016). endonuclease. Plant Cell Rep. 35, 1555–1558 (2016).
214. Lu, H.-P. et al. CRISPR-S: an active interference element for a rapid and 243. Feng, Z. et al. A highly efficient cell division-specific CRISPR/Cas9 system
inexpensive selection of genome-edited, transgene-free rice plants. Plant generates homozygous mutants for multiple genes in Arabidopsis. Int. J.
Biotechnol. J. 15, 1371–1373 (2017). Mol. Sci. 19, 3925 (2018).
215. Iaffaldano, B., Zhang, Y. & Cornish, K. CRISPR/Cas9 genome editing of 244. Li, H.-J., Liu, N.-Y., Shi, D.-Q., Liu, J. & Yang, W.-C. YAO is a nucleolar
rubber producing dandelion Taraxacum kok-saghyz using Agrobacterium WD40-repeat protein critical for embryogenesis and gametogenesis in
rhizogenes without selection. Ind. Crops Prod. 89, 356–362 (2016). Arabidopsis. BMC Plant Biol. 10, 169 (2010).
216. Zhang, Y. et al. Efficient and transgene-free genome editing in wheat 245. Yan, L. et al. High-efficiency genome editing in Arabidopsis using YAO
through transient expression of CRISPR/Cas9 DNA or RNA. Nat. Commun. promoter-driven CRISPR/Cas9 system. Mol. Plant 8, 1820–1823 (2015).
7, 12617 (2016). 246. Tsutsui, H. & Higashiyama, T. pKAMA-ITACHI vectors for highly efficient
217. Woo, J. W. et al. DNA-free genome editing in plants with preassembled CRISPR/Cas9-mediated gene knockout in Arabidopsis thaliana. Plant Cell
CRISPR-Cas9 ribonucleoproteins. Nat. Biotechnol. 33, 1162–1164 (2015). Physiol. 58, 46–56 (2017).
218. Malnoy, M. et al. DNA-free genetically edited grapevine and apple 247. Miki, D., Zhang, W., Zeng, W., Feng, Z. & Zhu, J.-K. CRISPR/Cas9-
protoplast using CRISPR/Cas9 ribonucleoproteins. Front. Plant Sci. 7, 1904 mediated gene targeting in Arabidopsis using sequential transformation.
(2016). Nat. Commun. 9, 1967 (2018).
219. Subburaj, S. et al. Site-directed mutagenesis in Petunia × hybrida protoplast 248. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks
system using direct delivery of purified recombinant Cas9 induced by CRISPR–Cas9 leads to large deletions and complex
ribonucleoproteins. Plant Cell Rep. 35, 1535–1544 (2016). rearrangements. Nat. Biotechnol. 36, 765–771 (2018).
220. Li, J. et al. Whole genome sequencing reveals rare off-target mutations and 249. Feng, Z. et al. Multigeneration analysis reveals the inheritance, specificity,
considerable inherent genetic or/and somaclonal variations in CRISPR/ and patterns of CRISPR/Cas-induced gene modifications in Arabidopsis.
Cas9-edited cotton plants. Plant Biotechnol. J. 17, 858–868 (2019). Proc. Natl Acad. Sci. USA 111, 4632–4637 (2014).
221. Liang, Z. et al. Efficient DNA-free genome editing of bread wheat using 250. Tang, X. et al. A large-scale whole-genome sequencing analysis reveals
CRISPR/Cas9 ribonucleoprotein complexes. Nat. Commun. 8, 14261 (2017). highly specific genome editing by both Cas9 and Cpf1 (Cas12a) nucleases
222. Svitashev, S., Schwartz, C., Lenderts, B., Young, J. K. & Cigan, A. M. in rice. Genome Biol. 19, 84 (2018).
Genome editing in maize directed by CRISPR–Cas9 ribonucleoprotein 251. Rees, H. A., Wilson, C., Doman, J. L. & Liu, D. R. Analysis and
complexes. Nat. Commun. 7, 13274 (2016). minimization of cellular RNA editing by DNA adenine base editors. Sci.
223. Toda, E. et al. An efficient DNA- and selectable-marker-free genome-editing Adv. 5, eaax5717 (2019).
system using zygotes in rice. Nat. Plants 5, 363–368 (2019). 252. Grünewald, J. et al. Transcriptome-wide off-target RNA editing induced
224. He, Y. et al. Programmed self-elimination of the CRISPR/Cas9 construct byCRISPR-guided DNA base editors. Nature 569, 433 (2019).
greatly accelerates the isolation of edited and transgene-free rice plants. 253. Kleinstiver, B. P. et al. High-fidelity CRISPR-Cas9 variants with
Mol. Plant 11, 1210–1213 (2018). undetectable genome-wide off-targets. Nature 529, 490–495 (2016).
225. Ryder, P., McHale, M., Fort, A. & Spillane, C. Generation of stable nulliplex 254. Slaymaker, I. M. et al. Rationally engineered Cas9 nucleases with improved
autopolyploid lines of Arabidopsis thaliana using CRISPR/Cas9 genome specificity. Science 351, 84–88 (2016).
editing. Plant Cell Rep. 36, 1005–1008 (2017). 255. Chen, J. S. et al. Enhanced proofreading governs CRISPR–Cas9 targeting
226. Shan, S. et al. Application of CRISPR/Cas9 to Tragopogon (Asteraceae), an accuracy. Nature 550, 407–410 (2017).
evolutionary model for the study of polyploidy. Mol. Ecol. Resour. 18, 256. Casini, A. et al. A highly specific SpCas9 variant is identified by in vivo
1427–1443 (2018). screening in yeast. Nat. Biotechnol. 36, 265–271 (2018).
227. Andersson, M. et al. Efficient targeted multiallelic mutagenesis in tetraploid 257. Lee, J. K. et al. Directed evolution of CRISPR-Cas9 to increase its specificity.
potato (Solanum tuberosum) by transient CRISPR-Cas9 expression in Nat. Commun. 9, 3048 (2018).
protoplasts. Plant Cell Rep. 36, 117–128 (2017). 258. Vakulskas, C. A. et al. A high-fidelity Cas9 mutant delivered as a
228. Braatz, J. et al. CRISPR-Cas9 targeted mutagenesis leads to simultaneous ribonucleoprotein complex enables efficient gene editing in human
modification of different homoeologous gene copies in polyploid oilseed hematopoietic stem and progenitor cells. Nat. Med. 24, 1216 (2018).
Rape (Brassica napus). Plant Physiol. 174, 935–942 (2017). 259. Zhang, D. et al. Perfectly matched 20-nucleotide guide RNA sequences
229. Gao, W. et al. Genome editing in cotton with the CRISPR/Cas9 system. enable robust genome editing using high-fidelity SpCas9 nucleases. Genome
Front. Plant Sci. 8, 1364 (2017). Biol. 18, 191 (2017).

Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants 793


Review Article NATURe PlAnTS
260. Liang, Z., Chen, K., Yan, Y., Zhang, Y. & Gao, C. Genotyping genome- 284. Jo, A. et al. Efficient mitochondrial genome editing by CRISPR/Cas9.
edited mutations in plants using CRISPR ribonucleoprotein complexes. BioMed. Res. Int. 2015, 305716 (2015).
Plant Biotechnol. J. 16, 2053–2062 (2018). 285. Piatek, A. A., Lenaghan, S. C. & Neal Stewart, C. Advanced editing of the
261. Zhang, Q. et al. Potential high-frequency off-target mutagenesis induced by nuclear and plastid genomes in plants. Plant Sci. 273, 42–49 (2018).
CRISPR/Cas9 in Arabidopsis and its prevention. Plant Mol. Biol. 96, 286. Ruf, S. et al. High-efficiency generation of fertile transplastomic Arabidopsis
445–456 (2018). plants. Nat. Plants 5, 282 (2019).
262. Lowe, K. et al. Morphogenic regulators Baby boom and Wuschel improve 287. Bae, S., Kweon, J., Kim, H. S. & Kim, J.-S. Microhomology-based choice of
monocot transformation. Plant Cell 28, 1998–2015 (2016). Cas9 nuclease target sites. Nat. Methods 11, 705–706 (2014).
263. Mookkan, M., Nelson-Vasilchik, K., Hague, J., Zhang, Z. J. & Kausch, A. P. 288. van Overbeek, M. et al. DNA repair profiling reveals nonrandom outcomes
Selectable marker independent transformation of recalcitrant maize inbred at Cas9-mediated breaks. Mol. Cell 63, 633–646 (2016).
B73 and sorghum P898012 mediated by morphogenic regulators BABY 289. Bothmer, A. et al. Characterization of the interplay between DNA repair
BOOM and WUSCHEL2. Plant Cell Rep. 36, 1477–1491 (2017). and CRISPR/Cas9-induced DNA lesions at an endogenous locus. Nat.
264. Khanday, I., Skinner, D., Yang, B., Mercier, R. & Sundaresan, V. A Commun. 8, 13905 (2017).
male-expressed rice embryogenic trigger redirected for asexual propagation 290. Chang, H. H. Y. et al. Different DNA end configurations dictate which
through seeds. Nature 565, 91 (2019). NHEJ components are most important for joining efficiency. J. Biol. Chem.
265. Wang, C. et al. Clonal seeds from hybrid rice by simultaneous genome 291, 24377–24389 (2016).
engineering of meiosis and fertilization genes. Nat. Biotechnol. 37, 291. Allen, F. et al. Predicting the mutations generated by repair of Cas9-induced
283–286 (2019). double-strand breaks. Nat. Biotechnol. 37, 64–72 (2018).
266. d’Erfurth, I. et al. Turning meiosis into mitosis. PLoS Biol. 7, 292. Chakrabarti, A. M. et al. Target-specific precision of CRISPR-mediated
e1000124 (2009). genome editing. Mol. Cell 73, 699–713 (2019).
267. Mieulet, D. et al. Turning rice meiosis into mitosis. Cell Res. 26, 293. Dreissig, S. et al. Live-cell CRISPR imaging in plants reveals dynamic
1242–1254 (2016). telomere movements. Plant J. Cell Mol. Biol. 91, 565–573 (2017).
268. Kelliher, T. et al. One-step genome editing of elite crop germplasm during 294. Ma, H. et al. Multiplexed labeling of genomic loci with dCas9 and engineered
haploid induction. Nat. Biotechnol. 37, 287–292 (2019). sgRNAs using CRISPRainbow. Nat. Biotechnol. 34, 528–530 (2016).
269. Wang, B. et al. Development of a haploid-inducer mediated genome 295. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A
editing (IMGE) system for accelerating maize breeding. Mol. Plant 12, protein-tagging system for signal amplification in gene expression and
597–602 (2019). fluorescence imaging. Cell 159, 635–646 (2014).
270. Lemmon, Z. H. et al. Rapid improvement of domestication traits in an 296. Fujita, T. & Fujii, H. Efficient isolation of specific genomic regions and
orphan crop by genome editing. Nat. Plants 4, 766 (2018). identification of associated proteins by engineered DNA-binding molecule-
271. Li, T. et al. Domestication of wild tomato is accelerated by genome editing. mediated chromatin immunoprecipitation (enChIP) using CRISPR.
Nat. Biotechnol. 36, 1160–1163 (2018). Biochem. Biophys. Res. Commun. 439, 132–136 (2013).
272. Zsögön, A. et al. De novo domestication of wild tomato using genome 297. Nishimasu, H. et al. Crystal Structure of Staphylococcus aureus Cas9. Cell
editing. Nat. Biotechnol. 36, 1211–1216 (2018). 162, 1113–1126 (2015).
273. Sarno, R. et al. Programming sites of meiotic crossovers using Spo11 fusion 298. Kaya, H., Ishibashi, K. & Toki, S. A split Staphylococcus aureus Cas9 as a
proteins. Nucleic Acids Res. 45, e164 (2017). compact genome-editing tool in plants. Plant Cell Physiol. 58, 643–649 (2017).
274. Jacobs, T. B., Zhang, N., Patel, D. & Martin, G. B. Generation of a collection
of mutant tomato lines using pooled CRISPR libraries. Plant Physiol. 174,
2023–2037 (2017). Acknowledgements
Due to limited space, we could not cite all the related literature. We apologize to the
275. Lu, Y. et al. Genome-wide targeted mutagenesis in rice using the CRISPR/
authors whose work was not cited in this Review. Our plant genome engineering research
Cas9 system. Mol. Plant 10, 1242–1245 (2017).
is supported by the National Science Foundation Plant Genome Research Program (grant
276. Meng, X. et al. Construction of a genome-wide mutant library in rice using
no. IOS-1758745), Biotechnology Risk Assessment Grant Program (grant no. 2018-
CRISPR/Cas9. Mol. Plant 10, 1238–1241 (2017).
33522-28789) from the US Department of Agriculture, the Foundation for Food and
277. Butt, H. et al. CRISPR directed evolution of the spliceosome for resistance
Agriculture Research (grant no. 593603) and Syngenta Biotechnology.
to splicing inhibitors. Genome Biol. 20, 73 (2019).
278. Boesch, P. et al. DNA repair in organelles: Pathways, organization,
regulation, relevance in disease and aging. Biochim. Biophys. Acta BBA - Author contributions
Mol. Cell Res. 1813, 186–200 (2011). Y.Z., A.A.M., S.S. and Y.Q. wrote the manuscript. Y.Z. prepared the figures. All authors
279. Srivastava, S. & Moraes, C. T. Manipulating mitochondrial DNA read and approved the final manuscript.
heteroplasmy by a mitochondrially targeted restriction endonuclease. Hum.
Mol. Genet. 10, 3093–3099 (2001).
280. Tanaka, M. et al. Gene therapy for mitochondrial disease by delivering Competing interests
restriction endonuclease SmaI into mitochondria. J. Biomed. Sci. 9, The authors declare no competing interests.
534–541 (2002).
281. Bacman, S. R., Williams, S. L., Pinto, M., Peralta, S. & Moraes, C. T. Specific
elimination of mutant mitochondrial genomes in patient-derived cells by Additional information
mitoTALENs. Nat. Med. 19, 1111–1113 (2013). Reprints and permissions information is available at www.nature.com/reprints.
282. Gammage, P. A., Rorbach, J., Vincent, A. I., Rebar, E. J. & Minczuk, M. Correspondence should be addressed to Y.Q.
Mitochondrially targeted ZFNs for selective degradation of pathogenic Peer review information: Nature Plants thanks S. Toki, L. Xia and the other, anonymous,
mitochondrial genomes bearing large‐scale deletions or point mutations. reviewer(s) for their contribution to the peer review of this work.
EMBO Mol. Med. 6, 458–466 (2014).
283. Gammage, P. A., Moraes, C. T. & Minczuk, M. Mitochondrial genome Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
engineering: the revolution may not be CRISPR-Ized. Trends Genet. 34, published maps and institutional affiliations.
101–110 (2018). © Springer Nature Limited 2019

794 Nature Plants | VOL 5 | AUGUST 2019 | 778–794 | www.nature.com/natureplants

Potrebbero piacerti anche