a multi-purpose toolkit to enable advanced genome ... · 1 large-scale biology article a...

61
1 LARGE-SCALE BIOLOGY ARTICLE A Multi-purpose Toolkit to Enable Advanced Genome Engineering in Plants Tomáš Čermák 1 , Shaun J. Curtin 2,3,4 , Javier Gil-Humanes 1,5 , Radim Čegan 6 , Thomas J. Y. Kono 3 , Eva Konečná 1 , Joseph J. Belanto 1 , Colby G. Starker 1 , Jade W. Mathre 1 , Rebecca L. Greenstein 1 , Daniel F. Voytas 1 1 Department of Genetics, Cell Biology & Development and Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455 2 Department of Plant Pathology, University of Minnesota, St Paul, MN 55108 3 Department of Agronomy & Plant Genetics, University of Minnesota, St. Paul, MN 55108 4 Present address: United States Department of Agriculture, Agricultural Research Service, Cereal Disease Laboratory, St Paul, MN 55108 5 Present address: Calyxt Inc., New Brighton, MN 55112 6 Department of Plant Developmental Genetics, Institute of Biophysics of the CAS, v.v.i., CZ-61265, Brno, Czech Republic Corresponding author, Daniel F. Voytas, [email protected] Short title: Toolkit for plant genome engineering One-sentence summary: This integrated reagent toolkit and streamlined protocols work across diverse plant species to enable sophisticated genome edits. The author responsible for distribution of materials integral to the findings presented in this article in accordance with the policy described in the Instructions for Authors (www.plantcell.org) is: Daniel F. Voytas ([email protected]). ABSTRACT We report a comprehensive toolkit that enables targeted, specific modification of monocot and dicot genomes using a variety of genome engineering approaches. Our reagents, based on Transcription Activator-Like Effector Nucleases TALENs and the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 system, are systematized for fast, modular cloning and accommodate diverse regulatory sequences to drive reagent expression. Vectors are optimized to create either single or multiple gene knockouts and large chromosomal deletions. Moreover, integration of geminivirus-based vectors enables precise gene editing through homologous recombination. Regulation of transcription is also possible. A web-based tool streamlines vector selection and construction. One advantage of our platform is the use of the Csy-type (CRISPR system yersinia) ribonuclease 4 Csy4 and tRNA processing enzymes to simultaneously express multiple guide RNAs (gRNAs). For example, we demonstrate targeted deletions in up to six genes by expressing twelve gRNAs from a single transcript. Csy4 and tRNA expression systems are almost twice as effective in inducing mutations as gRNAs expressed from individual RNA polymerase III promoters. Mutagenesis can be further enhanced 2.5-fold by incorporating the Trex2 exonuclease. Finally, we demonstrate that Cas9 nickases induce gene targeting at frequencies comparable to native Cas9 when they are delivered on geminivirus replicons. The reagents have been successfully validated in tomato (Solanum lycopersicum), tobacco (Nicotiana tabacum), Medicago truncatula, wheat (Triticum aestivum), and barley (Hordeum vulgare). Plant Cell Advance Publication. Published on May 18, 2017, doi:10.1105/tpc.16.00922 ©2017 American Society of Plant Biologists. All Rights Reserved

Upload: others

Post on 27-May-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    LARGE-SCALEBIOLOGYARTICLE

    AMulti-purposeToolkittoEnableAdvancedGenomeEngineeringinPlants

    TomášČermák1,ShaunJ.Curtin2,3,4,JavierGil-Humanes1,5,RadimČegan6,ThomasJ.Y.Kono3,EvaKonečná1,JosephJ.Belanto1,ColbyG.Starker1,JadeW.Mathre1,RebeccaL.Greenstein1,DanielF.Voytas1

    1DepartmentofGenetics,CellBiology&DevelopmentandCenterforGenomeEngineering,UniversityofMinnesota,Minneapolis,MN554552DepartmentofPlantPathology,UniversityofMinnesota,StPaul,MN551083DepartmentofAgronomy&PlantGenetics,UniversityofMinnesota,St.Paul,MN551084Presentaddress:UnitedStatesDepartmentofAgriculture,AgriculturalResearchService,CerealDiseaseLaboratory,StPaul,MN551085Presentaddress:CalyxtInc.,NewBrighton,MN551126DepartmentofPlantDevelopmentalGenetics,InstituteofBiophysicsoftheCAS,v.v.i.,CZ-61265,Brno,CzechRepublicCorrespondingauthor,DanielF.Voytas,[email protected]

    Shorttitle:Toolkitforplantgenomeengineering

    One-sentencesummary:Thisintegratedreagenttoolkitandstreamlinedprotocolsworkacrossdiverseplantspeciestoenablesophisticatedgenomeedits.

    TheauthorresponsiblefordistributionofmaterialsintegraltothefindingspresentedinthisarticleinaccordancewiththepolicydescribedintheInstructionsforAuthors(www.plantcell.org)is:DanielF.Voytas([email protected]).

    ABSTRACT

    We report a comprehensive toolkit that enables targeted, specificmodificationofmonocot anddicotgenomes using a variety of genome engineering approaches. Our reagents, based on TranscriptionActivator-Like Effector Nucleases TALENs and the Clustered Regularly Interspaced Short PalindromicRepeats (CRISPR)/Cas9 system, are systematized for fast, modular cloning and accommodate diverseregulatory sequences to drive reagent expression. Vectors are optimized to create either single ormultiplegeneknockoutsandlargechromosomaldeletions.Moreover,integrationofgeminivirus-basedvectorsenablesprecisegeneeditingthroughhomologousrecombination.Regulationoftranscriptionisalso possible. Aweb-based tool streamlines vector selection and construction.One advantage of ourplatformistheuseoftheCsy-type(CRISPRsystemyersinia)ribonuclease4Csy4 andtRNAprocessingenzymes to simultaneously express multiple guide RNAs (gRNAs). For example, we demonstratetargeteddeletionsinuptosixgenesbyexpressingtwelvegRNAsfromasingletranscript.Csy4andtRNAexpression systems are almost twice as effective in inducing mutations as gRNAs expressed fromindividual RNA polymerase III promoters. Mutagenesis can be further enhanced 2.5-fold byincorporatingtheTrex2exonuclease.Finally,wedemonstratethatCas9nickasesinducegenetargetingat frequencies comparable to native Cas9 when they are delivered on geminivirus replicons. Thereagents have been successfully validated in tomato (Solanum lycopersicum), tobacco (Nicotianatabacum),Medicagotruncatula,wheat(Triticumaestivum),andbarley(Hordeumvulgare).

    Plant Cell Advance Publication. Published on May 18, 2017, doi:10.1105/tpc.16.00922

    ©2017 American Society of Plant Biologists. All Rights Reserved

  • 2

    INTRODUCTION

    Genome engineering is a rapidly emerging discipline that seeks to develop strategies and

    methods for the efficient, targeted modification of DNA in living cells. Most genome

    engineering approaches use sequence-specific nucleases (SSNs), such as TALENs and

    CRISPR/Cas9reagents,tocreateatargetedDNAdouble-strandbreak(DSB)inagenome.DNA

    repair then achieves the desired DNA sequence modification. DSBs are most frequently

    repaired by non-homologous end joining (NHEJ), which can create insertion/deletion (indel)

    mutations at the break site that knockout gene function. TALEN- or CRISPR/Cas9-mediated

    geneknockoutshavebeenmadeindiverseplantspeciesamenabletotransformation(Brooks

    etal.,2014;Fauseretal.,2014;Forneretal.,2015;Gaoetal.,2014;Christianetal.,2013;Liang

    etal.,2014;Shanetal.,2013;Suganoetal.,2014;Xuetal.,2015;Zhangetal.,2014).Traitsof

    commercialvaluehavealsobeencreatedbytargetedknockoutofselectedgenes(Wangetal.,

    2014;Clasenetal.,2015;Haunetal.,2014;Lietal.,2015a,2012).Alternatively,DSBscanbe

    repairedbyhomologydependentrepair (HDR),whichcopies information froma ‘donor’DNA

    template. HDR typically occurs at lower frequencies thanNHEJ in somatic cells, and ismore

    challengingtoachievebecause it requires introducing intotheplantcellboththedonorDNA

    molecule and the SSNexpression cassette. As describedbelow, however, novel strategies to

    deliver these reagents have recently realized some improvements in efficiency (Baltes et al.,

    2014;Čermáketal.,2015;Fauseretal.,2012;Schimletal.,2014).

    Since the development of TALENs and CRISPR/Cas9 reagents, improvements in the

    technologyhaveoccurredatarapidpace.Forexample,indelfrequencieshavebeenincreased

    by coupling SSNs with exonucleases (Certo et al., 2012; Kwon et al., 2012), altering gRNA

  • 3

    architecture (Chenetal.,2013)orusingspecializedpromoters (Wangetal.,2015;Yanetal.,

    2015).IncreasedspecificityhasbeenachievedusingpairedTALENorCRISPR/Cas9nickases(Ran

    etal.,2013),truncatedgRNAs(Fuetal.,2014)ordimericCRISPR/Cas9nucleases(Guilingeret

    al., 2014; Tsai et al., 2014). Other diverse applications have emerged, including targeted

    regulation of gene expression, targeted epigenetic modification or site-specific base editing

    through deamination (Russa and Qi, 2015; Kungulovski and Jeltsch, 2016; Zong et al., 2017;

    Shimatanietal.,2017).

    CRISPR/Cas9 is particularly useful for multiplexed gene editing, because target

    specificityisdeterminedbyshortgRNAsratherthanproteinsthatmustbeengineeredforeach

    newtarget(BaltesandVoytas,2015).Severalsystemsforsimultaneousexpressionofmultiple

    gRNAshavebeendeveloped,andthesemostofteninvolvetheassemblyofmultiple,individual

    gRNA expression cassettes, each transcribed from a separate RNA Polymerase III (Pol III)

    promoter(Xingetal.,2014;Maetal.,2015;Lowderetal.,2015).MultiplegRNAscanalsobe

    expressed from a single transcript. Such polycistronic mRNAs are processed post-

    transcriptionally into individual gRNAs by RNA-cleaving enzymes. These enzymes include the

    CRISPR-associated RNA endoribonuclease Csy4 from Pseudomonas aeruginosa (Tsai et al.,

    2014), the tRNAprocessingenzymesnaturallypresent in thehost cells (Xie et al., 2015) and

    ribozymes(GaoandZhao,2014).

    One significant remaining challenge in plant genome engineering is achieving high

    frequencygeneeditingbyHDR.Asmentionedabove,HDRrequiresintroducingintotheplant

    cellbothadonorDNAmoleculeandaSSNexpressioncassette.DNAsequencemodifications

    incorporated into the donor template are then copied into the broken chromosome. The in

  • 4

    planta gene targeting (GT) approach stably integrates the donor template into the plant

    genome.Thisensuresthateachcellhasatleastonecopyofthedonor,whichisreleasedfrom

    thechromosomebyaSSN,makingitavailableforHDR(Schimletal.,2014;Fauseretal.,2012).

    Anotherapproachinvolvesincreasingdonortemplateavailabilitybyreplicatingittohighcopy

    numberusinggeminivirusreplicons(GVR)(Baltesetal.2014).GVRsmakeitpossibletocreate

    precise modifications without the need to stably integrate editing reagents into the plant

    genome(Cermaketal.2015).

    Notallof themethodsandapproachesdescribedabovehavebeen implementedand

    optimized for use in plants. Although there are several toolsets available for plant genome

    engineering(Maetal.,2015;Xingetal.,2014;Lowderetal.,2015),theyarelimitedinthatthey

    useasingletypeofDSB-inducingreagent(e.g.CRISPR/Cas9),asingletypeofexpressionsystem

    (e.g.PolIIIpromotersforexpressionofgRNAs)ortheywerespecializedforasingleapplication

    (e.g.geneknockouts).Here,weintroduceacomprehensive,modularsystemforplantgenome

    engineering that makes it possible to accomplish gene knockouts, replacements, altered

    transcriptionalregulationormultiplexedmodifications. Inaddition,theplatformcanbeeasily

    upgraded and adjusted to accommodate novel reagents and approaches as they arise. Our

    toolkitusesGoldenGatecloningforfastandeasyassemblyofgenomeengineeringconstructs

    and flexibility in combining different functional modules for different applications. The

    functional modules include TALE-based reagents (TALENs and TALE activators), CRISPR/Cas9

    reagents (nucleases, nickases, catalytically inactive enzymes and activators), various gRNA

    expression approaches (single and multi-gRNA expression systems using Pol III or Pol II

    promoters), donor template plasmids for gene targeting, and a variety of other expression

  • 5

    cassettes (Trex2, Csy4 and GFP). Further, a large set of promoters, terminators, codon

    optimizedgenesandselectionmarkersfacilitateengineeringdicotormonocotgenomesusing

    T-DNA or non-T-DNA vectors for Agrobacterium-mediated, biolistic or protoplast

    transformation.Wealsoprovideauser-friendlywebportalwithprotocols,DNAsequencesof

    all reagentsandweb-basedtoolstoassist inconstructdesign. Finally,wehavevalidatedour

    reagentsinsixdifferentplantspeciesandreportthefirstuseofgenomeengineeringtocreate

    modifiedMedicagotruncatulaplants.

    RESULTS

    Directandmodularassemblyofgenomeengineeringconstructs

    Our plant genome engineering toolkit provides two sets of vectors, designated ‘direct’ and

    ‘modular’ (Figure1).Bothvectorsetscandeliver toplantcellseitherTALENsorCRISPR/Cas9

    reagents for creating targeted DNA sequence modifications. The TALEN vectors are fully

    compatiblewithourGoldenGateTALENandTALEffectorKit2.0(Cermaketal.,2011),andthey

    use the highly active Δ152/+63 TALEN architecture (Miller et al., 2011). The CRISPR/Cas9

    vectorsincludeeitherwheatorArabidopsisthalianacodon-optimizedversionsofCas9foruse

    inmonocotsanddicots,respectively.BothversionsofCas9containasingleC-terminalnuclear

    localizationsignal(NLS).

    The direct cloning vectors (numbering 31) enable rapid construction of reagents for

    making targeted gene knock-outs. They consist of vector backbones (T-DNA or non-T-DNA),

    selectable marker expression cassettes and nucleases (i.e. Cas9 or the TALEN backbone).

    However, they lack determinants for DNA targeting (i.e. gRNAs or TALE repeats). Single or

  • 6

    multiplegRNAscanbeaddedinasinglestepusingaGoldenGateprotocol(Engleretal.,2008,

    2009) (Figure 1A). Addition of TALENs uses a three-step Golden Gate protocol (see

    SupplementalMethodsforassemblyprotocols).

    ThemodularvectorsetusesGoldenGatecloningtocreatevectorsforadiverserangeof

    geneeditingapplications(Figure1B,SupplementalMethods–Protocol5).Therearethreesets

    ofmodularcloningvectors.ModulesetAcontainsready-to-useplasmids(currentlynumbering

    61)withCas9orGFPexpression cassettes.ModuleAplasmidsarealsoavailable that canbe

    used when assembling TALENs with our Golden Gate kit (Cermak et al., 2011). Module B

    plasmids(currentlynumbering22)areusedeithertoaddsingleormultiplegRNAsinavariety

    of expression formats or to add a second TALEN monomer. Module C plasmids (currently

    numbering 22) may be used to add donor templates for gene targeting, additional gRNA

    cassettesorother expression cassettes suchasGFP, Trex2orCsy4. Eachexpression cassette

    hasa similararchitecture (SupplementalFigure1): a commonsetof restrictionenzymesites

    are used that allow for easy swapping of regulatory elements (promoters, terminator

    sequences)orcodingsequences.Ifareagentfromagivensetisnotdesired,thereareempty

    modules to serve as placeholders and still allow for Golden Gate assembly. Finally, a

    transformationbackbone canbe selected froma set (currently numbering 33) of T-DNAand

    non-T-DNA vectors. Standard or geminivirus replicon (GVR) vectors are available with or

    without one of the commonly used antibiotic selectable markers. For most applications,

    creatingatransformationvectorusingthemodularvectorsetisatwo-stepcloningprocessthat

    canbecompletedinfivedays;assemblyofTALEN-basedvectorsusuallyrequiresanadditional

    cloningstep.

  • 7

    Tofacilitateuseofourtoolkit,wedevelopedonlineresourcestoaidinvectorselection

    and construct design (http://cfans-pmorrell.oit.umn.edu/CRISPR_Multiplex/). Our website

    provides a complete list of both direct andmodular cloning vectorswith descriptions of key

    features,DNAsequence filesand relevantprotocols fordownloading (seealsoSupplemental

    DataSet1forthelistofvectors). Alsoavailableonlineisa‘vectorselection’toolthatusesa

    dropdownmenutohelpidentifysuitableintermediaryplasmidsneeded(e.g.moduleA,BorC)

    or to select a transformationbackbone.Thevector selection tool thenprovidesa link to the

    DNA sequence file of the selectedplasmid that canbedownloaded inGenBank format. For

    multiplexedgeneediting,our‘primerdesignandmapconstruction’tooltakesasinputaFASTA

    filewiththegRNAtargetsequences.Alististhenpreparedofalltheprimersequencesneeded

    tocreateavectorthatexpressesmultiplegRNAsusingourtRNA,Csy4orribozymeexpression

    formats,andaGenBankfileiscreatedwiththeannotatedsequenceofthespecifiedgRNAarray

    intheselectedplasmidbackbone.

    TargetedmutagenesisofgenesinMedicagotruncatulausingdirectvectors

    To test the functionality of the direct vectors, we designed a TALEN pair to target two

    paralogous genes in Medicago (Medtr4g020620 and Medtr2g013650) (Figure 2A). The

    Arabidopsis ortholog is known to play a role in phosphate regulation, and mutants hyper-

    accumulatephosphate(Parketal.,2014).AT-DNAvectorwiththeTALENswasassembledusing

    ourGoldenGateassemblyprotocol (SupplementalMethods–Protocol1B)and transformed

    intoMedicago.ElevenT0plantswererecoveredandscreenedfortargetedmutationsatboth

    Medtr4g020620andMedtr2g013650usingaPCRdigestionassay.TwoT0plants(WPT52-4and

    WPT52-5)producedPCRampliconsthatwereresistanttorestrictionenzymedigestion(atboth

  • 8

    targets inWPT52-4 and at one target inWPT52-5). PlantWPT52-4was self-fertilized, and T1

    progenywerescreenedusingthesamePCRdigestionassay.Mutationswereconfirmedatboth

    loci (Figure 2B).Mutations inMedtr4g020620were likely somatic, as only twoof the tested

    plants produced digestion-resistant amplicons of varying intensity. However, mutations in

    Medtr2g013650segregatedasexpectedforatraittransmittedthroughthegermline,andone

    outofeighttestedplantswasahomozygousmutant.ThiswasconfirmedbyDNAsequencing,

    whichrevealedfourdifferentMedtr4g020620deletionallelesinasingleplant,andanidentical

    63bpdeletioninMedtr2g013650inthreeofthetestedT1plants(Figure2C).FromtheeightT1

    plants,weidentifiedasingleplantwithmutationsinbothgenes.Thus,TALENsworkefficiently

    inourdirectvectorsandinduceheritablemutations.

    ExpressingmultiplegRNAsbyCsy4,tRNA-processingenzymesandribozymes

    To harness one of the biggest advantages of the CRISPR/Cas9 system – the ability to target

    multiple sites simultaneously – ourmodular cloning systemprovides vectors formultiplexed

    genome editing. Typically in multiplexed editing, independent Pol III promoters are used to

    drive expression of each gRNA. One limitation is that Pol III expression requires a specific

    nucleotide in the first position of the transcript. Further, the size of the final array can be

    cumbersome due to multiple, repeating promoter sequences. Processing polycistronic

    transcriptscontainingmultiplegRNAsprovidesanalternativetotheuseofindependentPolIII

    promoters.ThepolycistronicmessagecanbeproducedfromaPolIIpromoterthatisprocessed

    eitherbytheCsy4protein,tRNAprocessingenzymesorribozymes(Nissimetal.,2014;Aebyet

    al.,2010;Tangetal.,2016).PolIIpromotersprovideflexibilityintermsofspatialandtemporal

    controlofexpressioninvivo,andtheyaremorelikelytoproducelongtranscripts,sincePolIIis

  • 9

    not hampered by the presence of short internal termination sites, as is the case for Pol III

    (Arimbasserietal.,2013).Wesoughttocomparetheefficiencyoftargetedmutagenesisusing

    five different gRNA expression systems, including three different methods for processing

    polycistronic gRNA transcripts (Figure 3A). All vectors were assembled using our modular

    cloningsystemandusedtwogRNAstotargettwositesinthetomatoAUXINRESPONSEFACTOR

    8AARF8Agene,apositiveregulatorofflowerdevelopmentandfruitset(Liuetal.,2014).

    We created three vectors that use individual Pol III promoters to express each gRNA:

    two AtU6 promoters, an AtU6 and At7SL promoter or AtU6 and At7SL promoters combined

    with structurally optimized gRNA scaffolds (Chen et al., 2013). The remaining three vectors

    produceasingle transcriptwithgRNAcassettesseparatedby20bpCsy4hairpins (Tsaietal.,

    2014),77bptRNAGlygenes(Xieetal.,2015)or15bpribozymecleavagesites(inadditiontoa

    synthetic ribozyme at the 3’ end of the transcript) (Haseloff and Gerlach, 1988; Tang et al.,

    2016).ThankstotheadvantagesoverPolIIIpromotersmentionedabove,wechosetouseaPol

    IIpromotertodrivetheexpressionofthepolycistronicgRNAs.SinceCas9wasexpressedfrom

    a 35S promoter, we chose Cestrum Yellow Leaf Curling Virus promoter (CmYLCV) for gRNA

    expressiontopreventuseofduplicatepromoters.TheCmYLCVpromoterdrivescomparableor

    higher levelsofexpression than the35Sormaizeubiquitin (ZmUbi)promoters inbothdicots

    andmonocots(Stavoloneetal.,2003),andtheintensityofGFPexpressedfromthispromoter

    was similar to 35S-driven GFP in tomato protoplasts (Supplemental Figure 2). Final vectors

    were co-transformed into tomato protoplasts along with a yellow fluorescent protein (YFP)

    expressionplasmid. Transformationefficiencyapproximated60%, asdeterminedby counting

    YFPpositivecells.

  • 10

    Toassessmutationfrequencies, thetargetsitewasPCR-amplifiedfromDNAprepared

    from the transformed protoplasts and subjected to Illumina DNA sequencing (Supplemental

    Table1).AlloftheexpectedtypesofmutationsintheARF8Alocuswererevealedinallsamples

    except for thenon-transformedcontrol (Figure3B,3C;SupplementalTable2; Supplemental

    Figures 3-7). Interestingly, the overall frequency ofmutagenesiswas approximately two fold

    higher with the Csy4 and tRNA expression systems compared to constructs with two Pol III

    promoters.AlthoughtheoverallfrequencyofmutagenesisintheCsy4samplewasconsistently

    higher than in the tRNA sample across three replicates, the difference was not statistically

    significant(Figure3C,SupplementalTable3).Theribozymeshowedlowerfrequenciesofmost

    mutation types, particularly at the 3’-most gRNA target site. We did not observe any

    enhancement in overall mutation frequencies with the previously reported improved gRNA

    architecture(Chenetal.,2013).Itisimportanttonotethatthemutationfrequencieswerenot

    adjusted to account for the ~60% transformation efficiency, and therefore they are

    underestimates.

    DeletionbetweenthetwoCRISPR/Cas9cleavagesiteswasthemostcommonmutation

    inallsamples(SupplementalFigure3).Shortinsertions/deletions(indels)ateitheroneorboth

    cleavagesitesweremorethan10-foldlessabundant(SupplementalFigures3-6).Inversionsof

    theinterveningsequenceanddeletionscombinedwithinsertionsweremorethan100-foldless

    frequent(SupplementalFigure3).Mostinsertionsmappedeithertothetomatogenomeorto

    thevector,coveringalmosteveryregionofthevectorsequence(SupplementalFigure7).

    Cas9mostfrequentlycreatesablunt-endedDSBthreenucleotidesupstreamofthePAM

    sequence(Jineketal.,2012).Inourexperiments,preciseligationofCas9-cleavedDNAwithout

  • 11

    theinterveningsequencewouldresultinadeletionof85bp.Gainorlossofnucleotidesdueto

    staggeredCas9cleavage(Jineketal.,2012;Lietal.,2015b)and/orexonucleaseactivitycould

    result in shorteror longerdeletions.Strikingly,weobservedaveryhigh frequencyofprecise

    ligation: on average, 83.5% of all reads with long deletions were deletions of 85 bp

    (SupplementalFigure8).TheseresultssuggestthatifasinglegRNAwasused,mostinstances

    ofNHEJrepairwouldnotresult inamutation.Thereforecreatingdeletionswithtwoormore

    gRNAsshouldbeamoreefficientapproachtoachievetargetedmutagenesis.

    Wefurthertestedtheeffectivenessofthepolycistronicexpressionsystemsusingeight

    gRNAstargetingfourgenesfordeletionintomatoprotoplasts.Wealsomonitoredtheeffectof

    thepositionofthegRNAinthearray.gRNAs#49-56(SupplementalTable4)wereassembled

    directly intonon-T-DNAprotoplastexpressionvectors in twodifferentorders (Figure4A). In

    onearray,thegRNAswereorderedfrom49to56,andtheorderwasreversed inthesecond

    array. In addition, a vector expressing each gRNA from a separate Pol III promoter was

    constructed. Each pair of gRNAs was designed to create a ~3 kb deletion to prevent

    amplificationofthenon-deletedlociinsubsequentPCRanalysis(Figure4B).Thesevenvectors

    were transformed into tomato protoplasts, and the frequencies of deletions induced by the

    first,lastandoneoftheinternalgRNApairsineacharrayweremeasuredbyquantitativePCR

    (Figure4C). Consistentwith thepreviousexperiment, theCsy4expression systemperformed

    bestregardlessofthepositionofthegRNAinthearray.ThetRNAsystemwasthenextmost

    effective,followedbytheuseofindividualPolIIIpromoters;theribozymesystemwastheleast

    effective.gRNAsneartheendoftheCsy4andtRNAarrayswere lessefficient inmutagenesis

    relative to gRNAs in earlier positions for themajority of tested loci, suggesting that position

  • 12

    doeshaveaneffectongRNAactivity.Nevertheless, regardlessofpositioneffects,whenCsy4

    was used, the frequencyof deletionswas as highor higher than theuseof individual Pol III

    promoters,confirmingtheCsy4isthemostefficientsystemformultiplexedgeneediting.

    Multiplexedmutagenesisintomato,wheat,andbarleyprotoplasts

    WefurtherexploredtheeffectivenessoftheCsy4systemformultiplexingbytargetingsixgenes

    in tomato (Solyc06g074350, Solyc02g085500, Solyc02g090730, Solyc11g071810,

    Solyc06g074240andSolyc02g077390),threegenesinwheat(ubiquitin,5-Enolpyruvylshikimate-

    3-phosphatesynthase-TaEPSPS,andMildewlocusO-TaMLO),andonegeneinbarley(Mildew

    locusO-HvMLO).EachgenewastargetedfordeletionusingtwogRNAs.Toprovideexpression

    in cereals, we created vectors with the maize ubiquitin (ZmUbi), switchgrass (Panicum

    virgatum) ubiquitin 1 (PvUbi1) and switchgrass ubiquitin 2 (PvUbi2) promoters (Mann et al.,

    2011).ThesequencesofPvUbi1andPvUbi2weremodifiedtoremoveAarI,SapI,andBsaIsites,

    and themodifiedpromoterswere confirmed todrivehigh levelsofGFPexpression inwheat

    scutella(SupplementalFigure9).Next,wedesignedCsy4arraysoftwelvegRNAstargetingthe

    sixgenesintomato,sixgRNAstargetingthethreegenesinwheat,andtwogRNAstargetingthe

    singlegeneinbarley.Intomato,the35SpromoterwasusedtoexpressCas9,andtheCmYLCV

    promoter drove expression of the gRNAs. In wheat and barley, ZmUbi was used to drive

    expression of Cas9, and the gRNA array was under control of PvUbi1. To test whether the

    PvUbi1 promoter could be substituted with the significantly shorter viral CmYLCV for gRNA

    expressioninmonocots,wealsodesignedavectorinwhichthetwogRNAstargetingthebarley

    MLOgenewerecontrolledbytheCmYLCVpromoter.ThefinalvectorsexpressingCas9andthe

    gRNA array were transformed into tomato, wheat and barley protoplasts, and PCR was

  • 13

    conducted two days later to detect targeted deletions. Deletions of the expected size were

    foundineachtargetedgeneandverifiedbyDNAsequencing(Figures5-7).BothCmYLCVand

    PvUbi1-driven gRNA arrays induced targeted deletion of the barleyMLO gene with similar

    efficiencies (Figure 7B). Thus, we confirmed that our system is efficient in simultaneously

    creating multiplexed, targeted gene deletions in both dicots and monocots using up to 12

    gRNAs.

    Heritable,multiplexedmutagenesisinMedicagotruncatula

    To demonstrate the efficiency of our Csy4multiplexing vectors inwhole plants,we used six

    gRNAstotargetfourMedicagogenesthatencodenodule-specificcysteine-rich(NCR)peptides

    (SupplementalFigure10).Morethan500genesmakeuptheNCRfamily,andtheirsmallsize,

    sequence identity and overall number pose challenges to traditional knock-out/knock-down

    platformssuchas transposableelements (Tnt1),RNA interferenceandchemicalmutagenesis.

    Three gRNA pairs were designed to each delete one of three loci: NCR53, NCR54

    (Medtr4g026818) andNCR55 (Medtr3g014705). TheNCR53 locus contains two homologous

    NCR genes differing by several SNPs (Supplemental Data Set 2) and separated by

    approximately 4 kb. The vector carrying the gRNA array, Cas9 expression cassette and bar

    resistance gene was transformed into Medicago, and 46 phosphinothricin resistant T0

    transformantswererecoveredandscreenedbyPCRanddirectsequencing.Forty-threeplants

    hadmutations in inNCR53,eight inNCR54and12 inNCR55 (Table1,SupplementalTable5,

    SupplementalDataSet2).Themutations includeddeletionsof theNCR53andNCR55genes,

    resultingfromsimultaneouscleavageat twosites,aswellasshort indels inall three lociata

    giventargetsite.Threeplantshadmutationsinallthreegenes,13weredoublemutantsand27

  • 14

    hadmutationsinasinglegene.Despiteitsrepetitiveness,mutationsattheNCR53 locuswere

    exclusively long deletions or inversions between the two cleavage sites in seven out of 46

    plants.Nomutationswere recoveredat thegRNA3 site, likelydue to twoSNPs foundat the

    targetsite.gRNA1,whichtargetsasiteatNCR53,hastwomismatchesintheseedregiontoa

    siteatNCR54andaPAM-disruptingmutation.Similarly,gRNA2hastwomismatchestoasiteat

    NCR54, one in eachof the 5’ and3’ endsof the seed region. Nooff-targetmutationswere

    observedfromgRNA1andgRNA2at theNCR54 locus in the46plants, further indicatingthat

    twomismatchesinthetargetsitearesufficienttopreventoff-targetactivity.

    To confirm transmission of the mutations to the next generation, plant #16 (with

    mutations inNCR53 andNCR54), plant#17 (withmutations inNCR53 andNCR55), plant#27

    (with mutations in all three targets) and plant #40 (with mutations in NCR53) were self-

    fertilized.SevenT1seedlingsfromeachT0parentwerescreenedbyPCRandDNAsequencing

    formutations in the respective genes and for presence of thebar transgene (Supplemental

    Figure 11A, B). Mutations identical to those observed in the T0 parents were found in the

    NCR53 locusintheprogenyofallfourtestedplants(SupplementalFigure11A).Intwoofthe

    fourplants(plant#17andplant#40),all testedprogenyonlyshowedthemutatedversionof

    theNCR53 locus. Both thedoublemutant (plant #16) and the triplemutant (plant #27) also

    transmittedtheexpectedmutationsinNCR54totheirprogeny,whereasonlythetriplemutant

    (plant #27) and not the double mutant (plant #17) transmitted the mutations in NCR55.

    Althoughwedidnotdetectthewild-typealleleofNCR53inT0plant#16orthewild-typeallele

    ofNCR55intheT0plant#27,wild-typealleleswererecoveredintheT1progeny,suggestingthe

    T0 plants might have been chimeric. Nevertheless, the T-DNA insertion segregated away in

  • 15

    severalT1progenyofT0plants#16,#27(includingatriplemutant27-6)and#40(Supplemental

    Figure 11B), confirming that the mutations in these plants were transmitted through the

    germline rather than created de novo in the T1. Thus, we have been able to show that

    mutationsineachofthegenesareheritable.Inaddition,weshowedthatmutationsinallthree

    genescanbetransmittedsimultaneouslyfromatriplymutantparent(plant#27)toitsprogeny.

    These results show the feasibility of using the Csy4 system to effectively recover heritable

    mutationsinmultiplegenes.

    Targeteddeletionof58kbinMedicagousingthetRNAandCsy4multiplexingsystems

    To further evaluate the Csy4 and tRNA systems for creating chromosomal deletions in

    Medicago,wedesignedanothersetofsixgRNAs(MPC1toMPC6)thattargeta58kbregionon

    chromosome2(Figure8A).Theregioncontains fivecandidategenes identifiedbyagenome-

    wideassociation study tobeassociatedwith symbioticnitrogen fixation (Curtinetal., 2017).

    ThegRNAswereassembled(intheorderfrom1to6)intothedirectcloningT-DNAvectorsthat

    useeitherCsy4ortRNAprocessingenzymes(seeSupplementalMethods–Protocols3Aand

    3B),andaftertransformation,DNAfromrandomlyselectedcalliwastestedbyPCRforpotential

    chromosomaldeletions. Primersweredesigned200 to300bpupstreamanddownstreamof

    varioustargetsitesthroughoutthelocus(Figure8A,SupplementalTable6).Resultsfromthis

    preliminaryassay revealedall fourpossibledeletions (detectableusing this setofprimers) in

    the Csy4-transformed calli, whereas only the 27 kb deletion between gRNA sitesMPC1 and

    MPC4wasdetected in thecalli transformedwiththetRNAmultiplexingreagents (Figure8B).

    Amplicons generated from putative 58 kb deletions were sequence confirmed (Figure 8C).

    While still in tissueculture,wepre-screened46T0plantlets fromeachof theCsy4and tRNA

  • 16

    transformations for the 27 kb and 58 kb deletions. The putatively positive plantlets were

    transferredtosoil,grownintomatureplantsandscreenedagainbyPCR.ThreeCsy4T0plants

    wereidentifiedwiththe58kbdeletionandtwoplantswiththe27kbdeletion.ForthetRNAT0

    plants,oneplanthadthe27kbdeletion(Figure8B).Thewild-typesequencewithinthe58kb

    deletedregioncouldstillbeamplifiedwithadifferentsetofprimers,suggestingthatallplants

    wereheterozygousorchimeric.Oneplant(WPT228-5)appearedtobeaT0homozygousmutant

    for the 27 kb deletion, since we failed to recover a PCR amplicon from the wild-type allele

    (Figure8B).

    To test for heritable transmission of the deletions, T0 plants WPT228-28 (Csy4,

    heterozygote or chimeric for the 58 kb deletion) and WPT227-28 (tRNA, heterozygote or

    chimeric for the 27 kb deletion) were self-fertilized, and T1 plants were screened for the

    segregating chromosomal deletions. Eight out of 12 T1 seedlings produced bands of the

    expectedsizeforthe58kbdeletioninWPT228-28progeny,andsevenoutofsevenT1seedlings

    showed bands expected for the 27 kb deletion inWPT227-28 progeny, confirming germline

    transmissionintheselines(Figure8B).Inaddition,thewild-typeallelewasnotdetectedintwo

    out of the eight WPT228-28 T1 seedlings, suggesting the two seedlings are homozygous

    mutants. The bar transgene was not detected in three heterozygous WPT228-28 seedlings,

    indicatingthe lossofT-DNAinsertionandconfirmingthatthemutationswerenotcreatedde

    novo intheT1(Figure8B).Todeterminewhetherwecouldrecoverhomozygousmutants ina

    transgene-freebackground,T1 plantWPT228-28-1–heterozygous for the58kbdeletionand

    lackingtheT-DNAinsertion–wasself-fertilized,andsevenT2seedlingswerescreenedforthe

  • 17

    presence of the deletion. PCR and sequencing identified two homozygous and three

    heterozygousnon-transgenicplants(Figure8B).

    TheloweroverallmutationfrequenciescomparedwiththoseobservedintheNCRgeneediting

    experimentare likelyduetothesignificantly largersizeofdeletionsbeingsought.An inverse

    relationshipbetweenCas9-mediateddeletionfrequencyandsizehasbeenobservedpreviously

    inbothmammalianandplantcells(Canveretal.,2014;Xieetal.,2015;Ordonetal.,2016).

    EnhancingtargetedmutagenesiswithTrex2

    To increase the frequency of NHEJ-induced mutations, sequence-specific nucleases can be

    coupledwithexonucleasestopromoteendresectionandpreventprecise ligation.Previously,

    co-expressionofthehuman3’repairexonuclease2(Trex2)withameganucleaseresultedina

    25-fold enhancement in gene disruption frequencies in human cells (Certo et al., 2012). In

    plants, overexpression of exonuclease 1 (Exo1) significantly enhanced the frequency of DSB

    resectioninricecalli(Kwonetal.,2012),andTrex2increasedthemutagenesisfrequencywhen

    co-deliveredtoplantcellswithameganuclease(Luoetal.,2015).Basedonthisdataandour

    observation that the majority of Cas9-induced breaks are rejoined precisely, we tested the

    effectonmutagenesisofexpressingTrex2withCas9andasinglegRNA.WeusedgRNAsthat

    target two different sites in the tomato ANT1 gene and one site on the barleyMLO gene.

    IndividualgRNAswereclonedunderthecontroloftheAtU6orTaU6promoters,combinedwith

    a dicot or monocot optimized Trex2-P2A-Cas9 fusion, and tested in tomato and barley

    protoplasts. We saw a modest 1.5-2.5-fold increase in indel frequencies in the presence of

    Trex2comparedtothesampleswithoutTrex2inbarley(asmeasuredbytheT7EIassay)(Figure

  • 18

    9A).Similarresultswereobtainedintomato(asmeasuredbybothT7EIandDNAsequencing)

    (Figure9A,B).Sequencingrevealeda trendtowards longerdeletions in theTrex2samples in

    both tomato and barley (Figure 9B, C).Whereas no deletions >10 bp were detected in the

    samples lacking Trex2, they represented 77% (tomato) and 100% (barley) of all mutations

    recoveredinsamplesexpressingTrex2.Insertionsofupto42bpwerefoundinsampleswithout

    Trex2 (Supplemental Figure 12) but not in the samples expressing Trex2, suggesting that

    deletionswereinducedpreferentiallybythepresenceoftheexonuclease.

    GenetargetingwithCas9nickases

    Severalstrategieshavebeendevelopedtodecreasethefrequencyofoff-targetmutagenesisby

    CRISPR/Cas9. Among these are the use of paired Cas9 nickases, which create a DSB by

    introducingnicksonoppositeDNAstrands.Pairednickaseshaveon-targetcleavageefficiency

    comparabletowildtypeCas9inhumancellsandplants(i.e.Arabidopsisandrice),andreduce

    off-targetactivityupto1500-fold(Ranetal.,2013;Schimletal.,2014;Mikamietal.,2016).We

    createdCas9nickasevariantsbymutatingthecatalyticresiduesintheconservedRuvC(D10A)

    andHNH (H840A)nucleasedomains.ModuleA vectorswere then created thatexpressCas9

    variantswithindividualorcombinedmutations(thelatterresultinginacatalyticallydeadCas9

    enzyme).

    Wewere interested inexploringwhetherpairedCas9nickasescanalsopromotegene

    targeting(GT)inplantsusingapreviouslydescribedtobaccotransientGTassay.Inthisassay,a

    defectivegus:nptII transgene is restoredby cleavage andhomologous recombination (Figure

    10A) (Wright et al., 2005; Baltes et al., 2014). We designed a pair of gRNAs in PAM-out

    orientationthatoverlappedby5bp(-5bpoffset)andshouldcreatea29bpoverhang(Figure

  • 19

    10A).TostimulateGT,thereagentsweredeliveredtotheplanttissueontheBeanyellowdwarf

    virus (BeYDV) replicons (Baltesetal.,2014;Čermáketal.,2015)available inour toolkitasT-

    DNAtransformationbackbones.Thereadoutwasthenumberofblue(GUSpositive)spotsand

    intensityofstainingintobaccoleaves(SupplementalFigure13A).

    WefirstcomparedtheGTfrequencies inducedbyasinglenickorDSB.Thenumberof

    GTeventsinducedbysinglenickswasaround70%oftheGTeventsinducedbyaDSB(Figure

    10B).NosignificantdifferencesinGTfrequencieswereobservedbetweentheD10AandH840A

    nickases.Then,wecomparedtheGTefficienciesusingpairednickasesthatinducedoublenicks

    resulting in 29 bp 5’ overhangs (AtD10A) and 3’ overhangs (AtH840A). We observed 2-fold

    increaseinGTwith5’overhangscomparedto3’overhangs.ThenumberofGTeventsinduced

    bydoublenickscreating5’overhangswasevenhigherthanthenumberofGTeventsinduced

    bythewild-typeCas9nuclease.Thisis inagreementwithobservationsinhumancells(Ranet

    al.,2013).

    ToconfirmthenatureoftheGTeventsonthemolecularlevel,weextractedDNAfrom

    infiltrated tobacco leaves and PCR amplified both junctions of the inserted sequence

    (Supplemental Figure 13B). All samples except the one that was infiltrated with the vector

    expressingnon-cuttinggRNAs,yieldedproductsofexpectedsizeforbothjunctions.Sixtoten

    bacterial clones for each junction from each sample were analyzed by sequencing, which

    revealed precise integration of the donor template in all clones (Figure 10C). These data

    suggestthattheoverallfrequencyandprecisionofGTinducedbynickasesiscomparabletothe

    nuclease-basedapproach,whichwehavepreviouslyshowncanbeusedtoefficientlyrecover

    plantswithheritablemodificationscreatedbyGT(Baltesetal.,2014;Čermáketal.,2015).

  • 20

    To see whether the nickase approach is applicable for GT in a monocot species, we

    targetedanin-frameinsertionofaT2A-GFPcassetteintothethirdexonoftheubiquitingenein

    wheat scutella, usingWheat dwarf virus (WDV) replicons as described in Gil-Humanes et al.

    (2017). We observed GFP expression consistent with insertion into the ubiquitin locus

    (Supplemental Figure 14A-C), suggesting the feasibility of this approach across different

    species.

    DISCUSSION

    Genome engineering promises to advance both basic and applied plant research, and we

    developed a comprehensive reagent toolkit to make the latest genome engineering

    technologiesavailable to theplant sciencecommunity.BothTALENsandCRISPR/Cas9canbe

    easilycombinedwithavarietyofregulatorysequences,transformationvectorsandotherDNA

    modifyingenzymes,making itpossibletorapidlyassemblereagentsnecessarytomakesingle

    andmulti-geneknockoutsaswellasgenereplacements.

    Usingour flexible,modularsystemweevaluated fourdifferentmulti-gRNAexpression

    strategiesintomatoprotoplaststoachievemultiplexedediting.ApolycistronicgRNAtranscript,

    when processed by the bacterial Csy4 ribonuclease, resulted in almost two times higher

    mutation frequencies compared with the commonly used strategy in which each gRNA is

    expressedfroman individualPol IIIpromoter.Further, thepositionofthegRNAs inthearray

    hasonlyamodesteffectonactivity,andthelastgRNAsinanarrayofeightperformcomparably

    togRNAsexpressedindependentlyfromPollIIIpromoters.Althoughthefrequenciesobserved

    with Csy4 were comparable to the previously described tRNA processing system (Xie et al.,

  • 21

    2015),wefavortheCsy4systembecauseitconsistentlygaveushigherfrequenciesofdeletions

    intomatoprotoplasts,andthemulti-gRNAtranscriptsareshorterandlessrepetitive(i.e.Csy4

    recognitionsitesare20bpvs.77bpfortRNAs).AlthoughtheCsy4systemrequiresexpression

    oftheCsy4ribonucleaseintheplantcell,andnoextrafactorsarerequiredfortRNAprocessing,

    theCsy4geneisrelativelyshort(561bp=187aa),andwetypicallyexpressitasP2Afusionto

    Cas9withoutanyobservablephenotypicconsequencestotransgenicplants.

    Although we have previously shown that gRNA transcripts processed by ribozymes

    efficiently induce mutagenesis in rice, tobacco, and Arabidopsis (Tang et al., 2016), of the

    polycistronicRNAprocessingmechanismstestedinthepresentstudy,ribozymesresultedinthe

    lowestmutation frequencies. One difference between the two studies is that we expressed

    Cas9 and thegRNAarray fromseparatepromoters,whereasTangetal. (2016)useda single

    transcription unit to express both the Cas9 and the gRNAs. Furthermore, the frequency of

    deletionscreatedbytwogRNAswasnotdirectlymeasuredinthepreviousstudy.Itispossible

    that coordinate expression ofCas9 and gRNAs from a single transcription unit, which is not

    possible in the current version of our cloning systemdue to itsmodular structure,might be

    essentialtoachievehighergeneeditingfrequenciesusingribozymeprocessing.Whetherthisis

    true remains to be elucidated. Nevertheless, we demonstrate Csy4 and tRNA expression

    systemsefficientlyprocessmulti-gRNAtranscriptsandenablehighefficiencymultiplexedgene

    editing.

    AnalysisofmutationsresultingfromcleavageofasinglegenewithtwogRNAsrevealed

    that approximately 85% of DSBs were precisely ligated. This is consistent with the high

    frequencyofpreciserepairobservedinmouseandhumancells(Lietal.,2015b;Geisingeretal.,

  • 22

    2016).SinceDSBsinducedbyasinglegRNAarealsolikelyaccuratelyrepaired,wefavortheuse

    of two gRNAs to induce loss-of-function mutations. Precise repair can be used to your

    advantagetomakethemutationofchoice,andtherearenoconcernsaboutcreatingfunctional

    geneisoformsthroughnon-frameshiftindelsoralternativesplicing.Furthermore,thetargeted

    deletionscanbedetectedbyPCR,makingthescreeningformutantssignificantlyeasier.Thisis

    particularly valuable, if not essential, when simultaneously mutagenizing several genes, as

    screening for loss of restriction enzyme sites, T7 assays or direct DNA sequencing is time

    consuming and costly. Targeted deletions might also be valuable for editing non-coding

    sequences, including promoters and regulatory sequences,where short indelmutationsmay

    not be effective. Moreover, engineering complex traits will likely require an efficient

    multiplexededitingplatform.Hereweshowthatcombinationsofdifferentgenedeletionscan

    be induced in a single experiment using a single vector, highlighting the potential of this

    approachtospeedupcandidategeneevaluation.UsingourCsy4-basedsystem,wesuccessfully

    deleteduptosixgenes intomato,wheat,andbarleyprotoplastsand inMedicagotruncatula

    plants,demonstratingtheversatilityofthissystemacrossavarietyofplantspecies.Finally,the

    high level of precision inNHEJ repair also opens up an opportunity for targeted and precise

    geneinsertion/replacement,independentofHDR(Geisingeretal.,2016).

    As an alternative to knocking out geneswith twoormore gRNAs,we also show that

    Trex2, in combination with Cas9, increases the length and frequency (2.5 fold) of NHEJ-

    mediateddeletions inbothtomatoandbarley.AsinglestudythatcombinedTrex2withCas9

    forgeneeditinginhumancellsreportedasimilar2.5-foldincreaseinmutagenesis(Charietal.,

    2015). When Trex2 was used in combination with TALENs, a 3-4-fold enhancement was

  • 23

    observed(Certoetal.,2012;Luoetal.,2015),whereasa25-foldincreasewasachievedwhena

    meganucleasewasusedastheDSB-inducingagent(Certoetal.,2012).Thedifferencesinfold-

    enhancementbetweenthethreetypesofnucleasesmaybecausedbydifferentlevelsofTrex2

    activity on different types of DNA ends induced by Cas9 (blunt), TALENs (5’ overhang) or

    meganucleases(3’overhang,thepreferredsubstrateofTrex2).Interestingly,Certoetal.(2012)

    detectedabias towards smalldeletionswhen they combinedTrex2withmeganucleasesand

    TALENs,butthestudyofCharietal.(2015)aswellasourdataindicatethatthedeletionlength

    increaseswhenTrex2isusedwiththeCRISPR/Cas9system.Nevertheless,deletionsseemtobe

    induced preferentially in the presence of Trex2, regardless of the type of nuclease used to

    createtheDSBsincethelowerfrequencyof insertionscomparedwiththenoTrex2control is

    consistentinallstudies.

    Cas9nickasesarefrequentlyusedbecausetheyreducetheriskofoff-targetmutations

    (Ranetal.,2013;Choetal.,2014;Shenetal.,2014;Mikamietal.,2016).Singlenickasesdonot

    inducedetectablelevelsofon-targetNHEJ-mediatedmutagenesis(Congetal.,2013;Ranetal.,

    2013; Fauser et al., 2014), whereas paired nickases, which create two adjacent nicks in the

    targetsequence,inducemutationsatefficienciescomparabletonativeCas9(Ranetal.,2013;

    Schimletal.,2014;Mikamietal.,2016).Pairednickasesalsoefficientlyinducegenetargetingin

    humancells(Malietal.,2013;Ranetal.,2013),andherewedemonstratethatpairednickases

    constructed using our modular system induce gene targeting in tobacco and wheat with

    efficienciescomparabletothenuclease.Consistentwiththedatafromhumancells(Malietal.,

    2013;Ranetal.,2013),pairednickasesthatcreate5’overhangs inducedGTmorefrequently

    thanthosecreating3’overhangs.Inaseparatestudy(Fauseretal.,2014),singlenickaseswere

  • 24

    foundcapableofpromotingGTinArabidopsisatfrequenciesexceedingthenuclease;however

    weobservedthatGTfrequencieswithsinglenickaseswereabovebackgroundinbothtobacco

    andwheat,buttheywerelowerthantheGTfrequencyachievedwiththenucleaseandpaired

    nickases.ThehighfrequencyofGTobservedbyFauseretal. (2014)withasinglenickmay in

    partbeduetheproximityofthedonortemplate,whichwasintegratedintothechromosome

    adjacent to thenick site. Indeed, significantly lowerGT frequencieswereobserved inhuman

    cells when single nickases were used with extrachromosomal donors (Ran et al., 2013),

    although other factors such as donor architecture and cell type could contribute to the

    observeddifferencesbetweenstudies.Nevertheless,weshowthatpairednickases,whichhave

    beenshowntohaveminimaloff-targeteffects inplants(Mikamietal.2016),canbeusedfor

    efficientGTinbothmonocotsanddicots.

    Anumberofdifferenttoolkitsforplantgenomeengineeringhavebeenpublishedwithin

    thepasttwoyears(Xingetal.,2014;Xieetal.,2015;Maetal.,2015;Lowderetal.,2015;Zhang

    etal.,2015;Liangetal.,2016;Vazquez-Vilaretal.,2016;Kimetal.2016;Ordonetal.,2016);

    however,allhavelimitations:mostdonotofferflexibilityinthegeneeditingplatform(TALENs

    or CRISPR/Cas9); some reagents are optimized for a non target organism (e.g. humans);

    typicallythereislittlechoiceinpromotersforexpressingtargetingreagents;mostsystemsare

    limited to a single vector type (e.g. T-DNA); applications are limited to NHEJ-mediated gene

    knockouts;mostreagentsarevalidatedinonlyoneortwomodelspecies.Althoughtwostudies

    havemadetheirapproachescompatiblewithawidervarietyofvectorsandreagents,suchas

    theD10ACas9nickaseandCas9-basedtranscriptionalactivatorsandrepressors(Lowderetal.,

    2015;Vazquez-Vilaretal.,2016),theyremainlimitedinotheraspects.Forexample,theLowder

  • 25

    et al. 2015 toolkit limits gRNA expression to Pol III promoters, requires an 8-day assembly

    process,andlackscross-compatibleandadaptableexpressionvectors;theVazquiez-Vilaretal.

    2016 toolkit requires a time-consuming binary cloning approach and lacks substantial

    evaluation of the multiplexed gene editing vectors. In comparison, our plant genome

    engineeringplatformprovides theuserwithanoption to choose thedesired reagent froma

    comprehensivesetofexpressioncassettesincludingTALENs,TALE-VP64,TALE-VPR,Cas9,D10A

    Cas9 nickase, H840A Cas9 nickase, dCas9, dCas9-VP64 and dCas9-VPR, GFP, Trex2 and Csy4

    withversionscodonoptimizedforuseindicotsormonocots.ForCas9-basedgenomeediting,

    the toolkit enables assembly of vectorswith all gRNA expression systems described to date,

    including two dicot and four monocot Pol III promoters. Furthermore, multiplexed systems

    basedonCsy4,tRNAandribozymesareavailablethatcanbecombinedwithPolIIpromoters.

    All reagents can be assembled into T-DNAor non-T-DNA vectorswith orwithout one of the

    threemost common plant selectablemarkers driven by dicot ormonocot promoters. Faster

    direct or flexiblemodular cloning protocols are available.While previous studies focused on

    evaluating reagents in the model species Arabidopsis, rice and tobacco, here we provide

    evidence that inaddition to tobacco,our system is functionalandeffective in two important

    dicotandtwomonocotcropspecies.

    Tothebestofourknowledge,ourtoolkitisthefirstdesignedtofacilitategenetargeting

    through homologous recombination. We include GVR-based vectors, which were first

    demonstrated to work in proof of concept experiments in tobacco (Baltes et al., 2014) and

    morerecentlytoeffectivelyintroduceheritabletargetedinsertionsintothetomatogenomeat

    frequencies3-9-foldhigherthanstandardT-DNA(Čermáketal.,2015).WehavealsousedWDV

  • 26

    repliconstotargetinsertionofaselectablemarkerintothewheatubiquitinlocus(Gil-Humanes

    etal.,2017),andhereweshowGVRsworkincombinationwithpairednickases.Moreover,our

    modularsystemisalsocompatiblewiththeinplantagenetargetingapproachdescribedearlier

    (Fauser et al., 2012). An integral element in a gene targeting strategy is the DNA donor

    template,whichcarriescustommodificationsbeingintroducedinthetargetgenome.Allofthe

    module C plasmids in our toolkit include a unique restriction site for insertion of the donor

    template,andweprovideprotocolsforassemblyofthedonortemplateplasmidsforboththe

    repliconand inplantagenetargetingmethods (seeSupplementalMethods–Protocol4).All

    together, our toolkit enables the most efficient plant gene targeting methods currently

    available.

    In addition to the commonly used 35S and maize ubiquitin promoters, the current

    versionofourcloningsystemincludessevenotherdicotandthreemonocotpromoters.These

    includeconstitutiveviral,bacterial(CmYLCV,M24,FMV,nos)orplant(AtUbi10,OsAct1,PvUbi1,

    PvUbi2)promoters,whichprovidearangeofexpressionstrengthsinavarietyofplantspecies

    (Stavoloneetal.,2003;Sahooetal.,2014;Maitietal.,1997;Norrisetal.,1993;McElroyetal.,

    1990; Mann et al., 2012). We have also included the Arabidopsis thaliana Ec1.2 and YAO

    promoters, which are egg cell and embryo sac/pollen specific, respectively. Although

    Arabidopsiswasnotourtargetspeciesinthisstudy,thesetwopromotershavebeenshownto

    enhancemutagenesis inArabidopsiswhenused todriveCas9expression (Wanget al., 2015;

    Yan et al., 2015). In our toolkit, these promoters can drive either Cas9 expression or the

    polycistronic gRNA arrays. Due to the universal structure of the modular vectors, new

    combinations of promoters and coding sequences canbe easily added. The versatility of the

  • 27

    toolkit is further increased by its optimization for Golden Gate assembly, one of the most

    popular DNA assembly methods. We have made the T-DNA backbones derived from the

    commonly used pCAMBIA vectors free of most relevant type IIs restriction sites, AarI, BsaI,

    Esp3I and SapI. AarI, SapI and BsaI sites were also removed from the CmYLCV, PvUbi1 and

    PvUbi2promoters,andtheactivityofthemodifiedpromoterswasverifiedinplantstoensure

    these modifications do not affect expression levels. Since the Golden Gate modules can be

    easily modified to contain other genes, the system can be adopted for applications beyond

    genomeengineering,suchassyntheticbiology.

    Finally, theability toalter transcription levelsofendogenousgenes isoftendesirable.

    TranscriptionalactivationdomainshavebeenfusedtobothTALEsandCas9toenabletargeted

    gene activation (Lowder et al., 2015; Vazquez-Vilar et al., 2016). Although these custom

    transcriptionfactorshaveproveneffective (Liuetal.,2013;Piateketal.,2015;Lowderetal.,

    2015; Vazquez-Vilar et al., 2016), they havemostly been testedon reporter genes drivenby

    syntheticorminimalviralpromoters.Therefore,theincreaseinexpressionlevelsmightnotbe

    sufficient to inducephenotypicchangeswhenendogenousgenesare targeted (Lowderetal.,

    2015).Toovercomethemodestlevelsofgeneactivation,Chavezetal.(2015)recentlyshowed

    thatfusionsofdCas9orTALEproteinstoastrongtranscriptionalactivatorVPRmediateupto

    320-fold higher activation of endogenous targets compared with fusions to VP64 alone.

    Althoughfurtheroptimizationofartificialplanttranscriptionalregulatorsisneeded,atpresent,

    weprovidemoduleswithfusionsofbothVP64andVPRactivatordomainstoTALEsanddCas9

    fortranscriptionalactivation,aswellasmoduleswithdCas9only,whichhasbeenshowntobe

  • 28

    equallyeffectiveintranscriptionalrepressionasdCas9fusionstorepressordomains(Piateket

    al.,2015;Vazquez-Vilaretal.,2016).

    Inconclusion,wewouldliketonotethatduetoourtoolkit’smodulardesign,itcanbe

    easily updatedwith new functions, andwe intend to incorporate new tools as they become

    available.Giventheeaseandspeedofreagentconstructionandtheavailabilityofuser-friendly

    protocols andonline tools,wehopeourplatformwill accelerateprogress inplant functional

    genomicsandcropimprovement.

    Methods

    Directandmodularvectorlibraryconstruction

    Direct TALEN cloning vectors pDIRECT_37-39J were first constructed by Gibson assembly

    (Gibsonetal.,2009)ofPCRproductscontainingpartsofpCLEAN-G126backbone(Tholeetal.,

    2007,GenBankacc.EU186082), selectionmarker cassettes, theOsADH 5’UTRamplified from

    riceOryza sativa cv.Nipponbare genome (Sugio et al., 2008), 35S promoter,δ152/+63 TALE

    scaffoldsincluding3xFLAGepitopeandSV40NLS,Esp3IflankedccdBcassette,BsaIflankedLacZ

    cassette, ELD andKKRheterodimeric FokI variants (Doyonet al., 2011) andArabidopsis heat

    shock protein 18.2 (HSP) terminator amplified from the A. thaliana genome (Nagaya et al.,

    2010). In theprocess,Esp3I recognitionsiteswere removed fromthenptII geneand theELD

    FokICDS,BsaIfromtheccdBgeneandtheBglIIsitefromtheHSPterminator.Uniquerestriction

    siteswereinsertedtoflankthepromoterandterminatorinthedualTALENexpressioncassette.

    TALE sequences frompTALplasmids (Cermaketal., 2011)wereusedand19bpof sequence

    immediatelyupstreamofthefirstEsp3IsiteinTALEN1anddownstreamofthesecondBsaIsite

  • 29

    inTALEN2werere-codedtoincludeuniqueprimerbindingsitesforcolonyPCRandsequencing

    of both TALE repeat arrays. pCAMBIA-based vectors pDIRECT_21-23J were constructed

    accordingly,excepttheywerefirstmadewithanAscIandSacIflankedtheccdBcassette,later

    replaced with the AscI/SacI fragment from the pCLEAN-based vectors containing the dual

    TALEN expression cassette; and unique restriction sites were inserted to flank the selection

    marker gene. For expression in monocots, the ZmUbi1 promoter was PCR amplified from

    pAHC25(ChristensenandQuail,1996)andclonedbetweenAscIandSbfIsitesofpDIRECT_21J

    andpDIRECT_23Jtoreplacethe35SpromoterandcreatepDIRECT_25KandpDIRECT_26K.To

    createthefirstmoduleplasmids,theBsaILacZcassetteinpTC102(aderivativeofpDIRECT_37J

    with translation initiation sequence optimized for dicots) was replaced with a second Esp3I

    ccdBcassttetocreatepTC163.ThefirstandsecondTALENcassetteswerePCRamplifiedfrom

    pTC163(splittingtheP2AsequencebetweenthetwocassettesandremovingtheAarIsitefrom

    it)andaBaeIsitecontainingfragmentwasamplifiedfrompTC130(Čermáketal.,2015),using

    threepairsofprimerscontainingAarIsiteswithdistinct4bpoverhangs.Thefirstthreemodule

    plasmidspMOD_A1001,pMOD_B2000andpMOD_C0000were constructedby insertingeach

    ofthesethreePCRfragmentsintoBsaXIandPmlI-linearizedpTAL3(Cermaketal.,2011)using

    Gibsonassembly.TogeneratethefirstCRISPR/Cas9modulespMOD_A0101andpMOD_B2515,

    PCRfragmentscontaining35Spromoter,AtCas9genecodon-optimizedforA.thaliana(Fauser

    et al., 2014) and the HSP terminator; or the AtU6 gRNA cassette were inserted into AarI

    linearized pMOD_A1001 and pMOD_B2000 by Gibson assembly. Similarly, an At7SL gRNA

    cassette was inserted into NaeI and XhoI cut pMOD_C0000 to create pMOD_C2516. All

    remaining module plasmids were derived from these initial plasmids through replacing the

  • 30

    genecassettesandpromotersusingtheuniquerestrictionsitesAscI,SbfI,XhoIandSacI,orin

    case ofmulti-gRNAplasmids, throughGoldenGate assembly of PCR products containing the

    specific elements. TaCas9 was codon optimized for wheat and Gibson assembled from 9

    gBlocks (Integrated DNA Technologies, Coralville, USA). Cas9 nickases and catalytically dead

    variantsweremadebyGibsonassemblyofPCRproductscontainingthespecificmutationsinto

    the Cas9 nuclease modules. Cas9 and TALE transcriptional activators were constructed by

    Gibsonassemblyof fragments intoTALENanddCas9containingmodules.TheVPRactivators

    werebuiltfirst,usingthetripartiteVPRtranscriptionactivationdomaindescribedinChavezet

    al.(2015).TheVP64activatorswerederivedfromtheVPRmodulesbydeletingthep65andRTA

    domains by restriction cloning. The Csy4 genes, codon-optimized for dicots (tomato + A.

    thaliana) and monocots (wheat and rice), were synthesized as gBlocks along with the P2A

    ribosomal skippingsequenceandGibsonassembled intoSbfIandSalIdigestedpMOD_A0101

    and SbfI andBstBI digestedpMOD_A1510, respectively.Trex2was cloned similarly, except it

    wasPCRamplifiedfrompExodusCMV.Trex2plasmid(Certoetal.,2012)containingthemouse

    versionofthegenewhichwepreviouslyobservedhadrobustactivityinplantcells.TheCmYLCV

    (withouttheAarIsite)andFMVpromotersweresynthesizedasgBlocks.ThePvUbi1andPvUbi2

    promoterswereamplifiedfrompANIC10A(Mannetal.,2012)eachasasetofoverlappingPCR

    fragments to enable removal of AarI, BsaI and SapI sites throughGibson assembly. TheNos

    promoter, rbcsE9 and 35S terminators were amplified from pFZ19 (Zhang et al., 2010) and

    pCAMBIAvectors.TheremainingpromoterfragmentswereobtainedbyPCRamplificationfrom

    therespectivegenomicDNAs.ThetRNAGlysequencesusedtocreatethemultigRNAconstructs

    were amplified from tomato cv. MicroTom genomic DNA, while the Csy4 and ribozyme

  • 31

    sequencesweresynthesizedaspartsofprimersforGoldenGateassembly.Finally,theempty

    modulespMOD_A0000andpMOD_B0000werecreatedby ligatingannealedoligonucleotides

    carryinguniquerestrictionsitesintotheAscIandSacIsitesinpMOD_A0101andAscIandAgeI

    sitesinpMOD_B2103.TheT-DNAtransformationbackboneswerederivedfrompCAMBIA1300,

    2300and3300vectorsbyGibsonassemblyoftheCmR/ccdBselectioncassetteamplifiedfrom

    pMDC32 (Curtis and Grossniklaus, 2003) using primers carrying AarI sites specifying 4 bp

    overhangsdesignedtoacceptinsertsfromthethreemodules,intoHindIIIandBamHIdigested

    pCAMBIA backbones. In addition, pTRANS_210d-250d aremodified versions of the resulting

    vectors with AarI, BsaI, Esp3I and SapI sites removed from the vector backbone by Gibson

    assembly of backbone fragments overlapping at the mutated sites. pTRANS_250d and

    pTRANS_260dcontainPvUbi1 andPvUbi2promoters thatweredirectlyassembled into these

    vectors as described above. All non-T-DNA transformation backbones are derived from

    pTRANS_100.ThisvectorwascreateddenovobyGibsonassemblyof twoPCRproducts,one

    containingthespectinomycinresistancegeneandbacterialoriginofreplicationamplifiedfrom

    pTC14 (Cermak et al., 2011) and the other containing the AarI flanked CmR/ccdB selection

    cassette from the T-DNA transformation vector pTRANS_210. The selectionmarker cassettes

    were then inserted into this vector by restriction cloning from the T-DNA transformation

    backbones. The GVR transformation vectors were built by Gibson assembly from modified

    pCAMBIAbackbone(withoutBsaIandEsp3Isites)andgeminiviruselementsdescribedinBaltes

    etal.(2014),Čermáketal.(2015)andGil-Humanesetal.(2017)AllCRISPR/Cas9directcloning

    andCas9onlyvectorswereassembledbyGoldenGateassemblyfrommodulesA,BandC.To

  • 32

    createtheCas9onlyvectors,thetransformationbackbonesweremodifiedtoaccepttheinsert

    fromasinglemodule.

    GeneeditinginMedicagotruncatula

    FortargetedmutagenesisoftheMedtr4g020620andMedtr2g013650Agenes,apairofTALENs

    wasassembledintopDIRECT_39JusingProtocol1B(SupplementalMethods)resultinginpSC1

    and transformed into an Agrobacterium tumefaciens strain EHA105 for whole-plant

    transformation of theMedicago truncatula accession HM340 using an established protocol

    (Cossonet al., 2006). The resultingplantswere screened for putativemutationsusing a PCR

    digestionassaywithprimerslistedinSupplementalTable6andtheHaeIIIrestrictionenzyme.

    Thedigestion-resistantproductswereclonedandseveralclonesweresequencedtoidentifythe

    mutations. For combinatorial deletion of NCR genes, the six gRNAs were assembled into

    pDIRECT_23C along with the CmYLCV promoter using Protocol 3A (SupplementalMethods)

    resulting in pSC2 and transformed into Agrobacterium tumefaciens EHA105. To detect the

    targetedgenedeletionsandshort indelsateachgRNAsite,thethreelociwerePCRamplified

    fromT0andT1plantsusingprimerslistedinSupplementalTable6.ThePCRproductswerefirst

    directly sequenced. PCR products from samples containing both the long deletion and non-

    deletionalleleswere gel purifiedbefore sequencing and sequenced separately. Samples that

    showedoverlappingsequencetracesstartingateithergRNAsiteindicativeof indelmutations

    wereclonedandseveralclonesweresequencedtoidentifythetypeofmutations.Fortargeted

    deletionofthe58kbregiononchromosome2,thesixgRNAswereassembledasCsy4ortRNA

    arrays driven by the CmYLCV promoter into pDIRECT_23C using Protocol 3A and 3B

    (SupplementalMethods)yieldingpSC3andpSC4,respectively,andtransformedintoMedicago

  • 33

    truncatulaaccessionHM340asabove.Targeteddeletions intheresultingT0,T1andT2plants

    weredetectedusingprimerslistedinSupplementalTable6,clonedandsequenced.

    Determiningtheefficiencyofmulti-gRNAexpressionsystemsintomatoprotoplastsbydeep

    sequencing

    To build vector pTC303, expressing the two gRNAs from AtU6 and At7SL promoters; and

    pTC363expressingeachgRNAfromanindividualAtU6promoter,thegRNAspacerswerefirst

    clonedintopMOD_B2515(moduleBwithAtU6promoter,usedforbothpTC303andpTC363),

    pMOD_C2515(moduleCwithAtU6promoter,usedforpTC363)andpMOD_C2516(moduleC

    with At7SL promoter, used for pTC303) using Protocol 2A (Supplemental Methods). The

    resulting plasmids were assembled together with pMOD_A0101 into the pTRANS_100

    protoplast vector using Protocol 5 (SupplementalMethods). pAH750, expressing the gRNAs

    with structurally optimized scaffolds from AtU6 and At7SL promoters was constructed

    accordingly,exceptthegRNAspacerswereclonedintopMOD_B2515bandpMOD_C2516b,the

    respectivemoduleBandCplasmidsthatcarrytheoptimizedgRNArepeats.TheCsy4,tRNAand

    ribozymeexpressioncassetteswerecreateddirectlyinprotoplastvectorspDIRECT_10C(Csy4)

    andpDIRECT_10E(tRNAandribozyme)usingProtocol3A,3Band3C(SupplementalMethods),

    resulting in pTC364 (Csy4), pTC365 (tRNA) and pTC366 (ribozyme). Next, protoplasts were

    isolated from tomato cv. MicroTom as described in Zhang et al. (2012), with minor

    modifications.Leavesfrom1-2sterilegrown(26°Cwith16hlight)~4weeksoldplantletswere

    slicedintothinstripswithasharprazor,transferredtotheenzymesolution(1.0%cellulaseR10,

  • 34

    0.25%macerozymeR10, 0.45Mmannitol, 20mMMES, 1xMS salts and0.1%bovine serum

    albumin)andincubated15hat25°Cand40rpminthedark.Thedigestedproductwasfiltered

    througha40-µmcellstrainer(Falcon)andpipetteddirectlyontopof0.55Msucrosesolution.

    TherestoftheprotocolfollowedZhangetal.(2012),exceptW5solution(2mMMES,154mM

    NaCl,125mMCaCl2and5mMKCl)wasusedaswashingbuffer.TheCRISPR/Cas9constructs

    were co-transformed into ~200,000 cells along with the YFP expression plasmid pZHY162

    (Zhangetal.,2012),theprotoplastswereresuspendedinW5solutionandkeptat25°Cinthe

    dark.All samples2days later, the transformationefficiencywasdeterminedby counting the

    YFP-positivecellsusing imageanalysis,genomicDNAwas isolatedandusedasatemplatefor

    thedeepsequencinglibrarypreparation,asdescribedin(Čermáketal.,2015).Briefly,40ngof

    protoplastgenomicDNAwasusedastemplatetoamplifya354-bpregionoftheARF8A locus

    encompassing the gRNA9 and gRNA10 target sites with primers TC324F and TC324R

    (Supplemental Table 6), which have overhangs complementary to Nextera XT indices. PCR

    productswerepurifiedwithAgencourtAMPureXPBeads(BeckmanCoulter,Brea,USA)and5

    μlofthepurifiedPCRproductwasusedastemplateforthesecondPCRtoattachdualindices

    and Illumina sequencing adapters. PCR products were purified with Agencourt AMPure XP

    Beads, quantified and pooled. The pooled libraries were enriched for fragment sizes in the

    range250 -700bpusingBluePippin (SageScience,Beverly,USA)and sequencedon Illumina

    MiSeq platform (ACGT, Inc., Wheeling, USA). An average of 140,000 reads per sample was

    generated (Supplemental Table 1, http://www.ebi.ac.uk/ena/data/view/PRJEB13819).

    Sequencedataanalysiswasdoneasdescribedpreviously(Čermáketal.,2015),exceptdifferent

    sequence variants were manually sorted into specific groups. All data are presented as the

  • 35

    mean of three biological replicates (three separate pools of protoplasts) plus/minus the

    standarderror.Totestforthesignificanceofthedifferencebetweenthesamples,Tukeytest

    formultiplecomparisonofmeansinR(version3.2.4RC)wasused(SupplementalTable4).

    Testingmulti-gRNAexpressionsystemsintomatoprotoplastsbyqPCR

    The Csy4, tRNA and ribozyme arrays of eight gRNAs (Supplemental Table 4) were created

    directlyinprotoplastvectorspDIRECT_10C(Csy4)andpDIRECT_10E(tRNAandribozyme)using

    Protocol3A, 3B and3C (SupplementalMethods), resulting inpTC634 (Csy4, gRNAorder49-

    56),pTC635(Csy4,gRNAorder56-49),pTC636(tRNA,gRNAorder49-56),pTC637(tRNA,gRNA

    order56-49),pTC638(ribozyme,gRNAorder49-56)andpTC639(ribozyme,gRNAorder56-49).

    To construct the vector with eight gRNAs each expressed from an individual AtU6 (Pol III)

    promoter, four AtU6:gRNA cassettes were first assembled into each pMOD_B2203 and

    pMOD_C2200(resultinginpTC643andpTC644)usingProtocol3,exceptthefirst,lastandeach

    reverse primer were re-designed to bind the AtU6 sequence. The AtU6 cassettes were

    amplifiedfromBanI-digestedpTC363(firstcassette),aHindIIIandSgrAIfragmentfrompTC363

    (intermediarycassettes)andaBsaHIfragmentfrompTC363(lastcassette).The35Sterminator

    wasremovedfrompTC644bySnaBI/Eco53kI-mediatedreleaseoftheterminatorfragmentand

    religationofthebackbone,resultinginpTC645.pTC643andpTC645wereassembledalongwith

    pMOD_A0101 into thenon-T-DNAvectorpTRANS_100usingProtocol5, resulting in the final

    vectorpTC646.

    The CRISPR/Cas9 constructs were transformed into tomato protoplasts as described

    above.PurifiedprotoplastDNAwasextracted48hourslaterandusedwithLunaUniversalqPCR

  • 36

    Master Mix (NEB) following the manufacturer’s protocol. All samples were run in technical

    triplicateoftwobiologicalreplicates(twoseparatepoolsofprotoplasts)ina384-wellformaton

    a LightCycler 480 Instrument (Roche). All qPCR primers are listed in Supplemental Table 6.

    Primers for the genomic locus SGN-U346908 control were previously described (Expósito-

    Rodríguezetal.,2008).Alldataarepresentedasthemeanplus/minusthestandarderrorwith

    eachsamplerelativetothelevelsoftotalDNAasmeasuredbythegenomicslocuscontrol.

    Geneeditingintomato,wheatandbarleyprotoplasts

    TobuildthetomatoconstructpTC362withtwelvegRNAstargetingsixgenes,twoarraysofsix

    gRNAs were cloned into pMOD_B2203 (with CmYLCV promoter) and pMOD_C2200 using

    Protocol 3S1 and the two arrays were assembled together with the Csy4-Cas9 module

    pMOD_A0501 into the non-T-DNA transformation backbone pTRANS_100 using Protocol 5

    (SupplementalMethods).ThewheatvectorpJG612wasbuiltaccordinglyusingtheversionof

    Cas9optimizedformonocotsinpMOD_A1510,asinglearrayofsixgRNAsdrivenbythePvUbi1

    promoter in pMOD_B2112 and an empty module pMOD_C0000. Similarly, pMOD_A1510,

    pMOD_B 2112 (PvUbi1) or pMOD_B2103 (CmYLCV), and the empty pMOD_C0000 modules

    along with the transformation backbone pTRANS_100 were used to construct the barley

    vectors expressing two gRNAs from the PvUbi1 (pTC441) or CmYLCV (pTC437) promoters,

    targeting for deletion theMLO gene. Tomato protoplasts were isolated and transformed as

    described above. Wheat protoplasts were isolated from ~10 day old plantlets of the bread

    wheatcv.Bobwhite208.Seedsweresurfacesterilizedbyrinsingin70%ethanolfor10minand

    soakingfor30minina1%sodiumhypochloritesolution,andasepticallygrowninMSmedium

  • 37

    at24°Cundera16-hlightcycle.Approximately20plantletswereharvestedineachexperiment,

    cutinto~1-mmstripswitharazorblade,anddigestedwithanenzymesolution(1.5%cellulase

    R10, 0.75%macerozyme R10, 0.6Mmannitol, 10mMMES, 10mMCaCl2, and 0.1% bovine

    serumalbumin,pH5.7). The remaining stepswere identical as in theprotocol forprotoplast

    isolation from tomato described above. Barley protoplasts were isolated from cv. Golden

    Promiseusingthesameprotocol,exceptleavesfrom9-to14-day-oldplantlets(22°Cwith16h

    light cycle) were used. The CRISPR/Cas9 constructs were co-transformed into ~200,000 cells

    alongwiththeGFPexpressionplasmidpMOD_A3010theprotoplastswereresuspendedinW5

    solutionandkeptat25°Cinthedark.GenomicDNAwasisolated2daysaftertransformation,

    targeted deletions were detected using primers listed in Supplemental Table 6, cloned and

    sequenced.

    GeneeditingwithTrex2

    Individual gRNAs were cloned into pMOD_B2515 (AtU6 promoter, for tomato) and

    pMOD_B2518 (TaU6promoter, for barley) usingProtocol 2A and assembledwith 35S:Trex2-

    P2A-AtCas9 in pMOD_A0901 (for tomato) or ZmUbi:Trex2-P2A-TaCas9 in pMOD_A1910 (for

    barley) and the empty module pMOD_C0000 into the protoplast transformation backbone

    pTRANS_100 to createpTC391 (gRNA23, tomatoANT1),pTC393 (gRNA24, tomatoANT1) and

    pTC436 (gRNA38, barleyMLO) using PROTOCOL 5. The control plasmids pTC392 (gRNA23,

    tomato ANT1), pTC394 (gRNA24, tomato ANT1) and pTC440 (gRNA38, barleyMLO) without

    Trex2wereconstructedaccordingly,except35S:AtCas9 inpMOD_A0101andZmUbi:TaCas9 in

    pMOD_A1110 were used instead of the Trex2 containing modules. The constructs were

  • 38

    transformed into tomato and barley protoplasts as described above and genomic DNA was

    extracted2dayslater.TheANT1andMLOlociwereamplifiedbyPCR(seeSupplementalTable

    6forprimers)andthegeneeditingratesweredeterminedusingT7endonucleaseIaccordingto

    the manufacturer’s protocol (NEB, Ipswich, USA). Gel images were analyzed using ImageJ

    (Schneider et al., 2012). In addition, theANT1 PCR productswere cloned and sequenced to

    determine both frequency and type of mutations. For barley, the PCR products were first

    digestedwith NcoI to enrich formutations and only the digestion-resistant product was gel

    purified,clonedandsequenced.

    GenetargetingwithCas9nickasesintobaccoandwheat

    IndividualgRNAspacerswereclonedinmoduleB(pMOD_B2515fortobaccoorpMOD_B2518

    forwheat)orC(pMOD_C2516fortobaccoorpMOD_C2518forwheat),downstreamofanAtU6

    andAt7SLor twoTaU6promoters.TheGTdonorswere inserted into the resultingmoduleC

    plasmids usingProtocol 4 (SupplementalMethods). A promoter-less T2A-gfp-nos terminator

    cassette with 747 and 773 bp long flanking homology arms, and a 2530 bp long functional

    gus:nptII gene fusion were used as donor templates for wheat and tobacco, respectively.

    Finally,theBandCmoduleswereassembledwithselectedmoduleACas9variants(nuclease,

    D10Anickase,H840nickaseordeadCas9expressedfromeither35SorZmUbipromoterintoa

    BeYDV (pTRANS_201) or WDV (pTRANS_203) replicon vectors and used for A. tumefaciens

    infiltrationoftobaccoleaves(pJG363,pJG365,pJG367,pJG369,pJG370,pJG380andpJG382)or

    biolistic bombardment ofwheat scutella (pJG455, pJG456, pJG480, pJG481, pJG484, pJG486,

    pJG488 and pJG493). Note that plasmids pJG455-pJG493 above used the octopine synthase

  • 39

    (OCS)terminatorsequenceinsteadofthemoreefficientHSPterminatorusedinthepublished

    modules. The TaCas9 coding sequence and the rest of the plasmidswere identical with the

    newermodulesincludedinthecurrentvectorset.Fourdifferenttransgeniclines(Wrightetal.,

    2005)wereused todeterminegene targeting frequencies in tobaccoaspreviouslydescribed

    (Baltesetal.,2014),withminormodifications.Leavesfrom1to6weeksoldplantswereused

    forAgrobacteriumleafinfiltration.Foreachleaf,onehalfwasinfiltratedwithacontrolplasmid

    pLSLZ.D.R(Baltesetal.,2014)thatservedasareferencetocontrolfortransformationefficiency

    andtheotherhalfwithoneoftheCRISPR/Cas9constructs.Fourtosix leaveswere infiltrated

    witheachconstructineachexperiment.Fivedaysafterinfiltration,theleaftissuewasstained

    in the X-Gluc solution. Whole leaves were scanned and the stained area was quantified by

    imageanalysisusingImageJ(Schneideretal.,2012).TheGTfrequencieswerenormalizedtothe

    pLSLZ.D.RcontrolanddisplayedrelativetotheCas9nucleaseconstruct.Formolecularanalysis,

    genomicDNAwasextracted fromtwo infiltrated leavespersample (three leafpuncheswere

    takenfromeachleafandpooled)andusedasatemplateforPCRtoamplifytheleftandright

    recombinationjunctionsusingprimerslistedinSupplementalTable6.PCRproductsfromboth

    leaf sampleswere cloned and sequenced. To determineGT frequencies inwheat, immature

    wheatscutella(0.5-1.5mm)wereisolatedfromprimarytillersharvested16daysafteranthesis

    and used for biolistic transformation. Scutella isolation and culture conditions were as

    describedinGil-Humanesetal.(2011).BiolisticbombardmentwascarriedoutusingaPDS-1000

    gene gun. Equimolar amounts (1 pmol DNA mg-1 of gold) of plasmid were used for each

    experimentwith60μgofgoldparticles(0.6μmdiameter)pershot.GFPimagesoftransformed

  • 40

    tissue were taken 7 days after transformation and GFP positive cells were counted. The GT

    frequencieswerenormalizedtotheCas9nucleaseconstruct.

    Accessionnumbers

    ThedeepsequencingdataisavailableundertheEuropeanNucleotideArchive(ENA)accession

    PRJEB13819(http://www.ebi.ac.uk/ena/data/view/PRJEB13819).

    AllvectorsarepubliclyavailablefromAddgene(plasmid#90997–91225).AnnotatedvectorsequencesareavailablefromAddgeneandfromhttp://cfans-pmorrell.oit.umn.edu/CRISPR_Multiplex/

    SUPPLEMENTALDATA

    SupplementalFigure1.Universalarchitectureofexpressioncassetteswithuniquerestriction

    sitesshown.

    Supplemental Figure 2. GFP expression mediated by different promoter/terminator

    combinationsintomatoprotoplasts.

    SupplementalFigure3.FrequenciesofmutationsinducedusingdifferentsystemsforgRNA

    expressionintomatoprotoplasts.

    SupplementalFigure4.ShortindelsequencevariantsdetectedatgRNAsite9.

    SupplementalFigure5.ShortindelsequencevariantsdetectedatgRNAsite10.

    SupplementalFigure6.ShortindelsequencevariantsdetectedatbothgRNAsites9and10.

    SupplementalFigure7.InsertionsfromtheARF8AlocusmappedtothevectorpTC364.

    SupplementalFigure8.LongdeletionsequencevariantsdetectedbetweengRNAsites9and

    10.

    SupplementalFigure9.GFPexpressionmediatedbydifferentpromotersinwheatscutella.

  • 41

    SupplementalFigure10.MultiplexedtargetedmutagenesisinMedicagotruncatulausingCas9

    andCsy4.

    SupplementalFigure11.AnalysisofT1progenyfromfourMedicagotruncatulaT0plantswith

    mutationsinNCR53,NCR54andNCR55.

    SupplementalFigure12.SequencesofinsertionsdetectedattheANT1site#2intomato

    protoplastsampleswithoutTrex2expression.

    SupplementalFigure13.Detectionofgenetargetingintobaccoleaves.

    SupplementalFigure14.GenetargetingwithCas9nickasesandgeminivirusreplicons(GVRs)in

    wheat.

    SupplementalTable1.Sequencingandreadfilteringdata.

    SupplementalTable2.FrequenciesofindividualmutationtypesamongallARF8Asequencing

    reads.

    SupplementalTable3.MultipleTukeytestresults.

    SupplementalTable4.SequencesofthegRNAsitesusedintheqPCRexperiment.

    SupplementalTable5.Genotypesof46MedicagotruncatulahygromycinresistantT0plants

    transformedwithaCas9Csy4constructexpressingsixgRNAstargetingthreegenes.

    SupplementalTable6.Listofprimersusedinthisstudy.

    SupplementalMethods.Protocolsforvectorassembly.

    SupplementalDataSet1.Listofvectorsincludedinthetoolset.

    SupplementalDataSet2.SequencesofNCR53,NCR54andNCR55locitargetedfordeletionin

    46MedicagotruncatulaT0plants.

  • 42

    ACKNOWLEDGEMENTS

    WethankmembersoftheVoytaslabforhelpfuldiscussionsandinsights.Weparticularlythank

    LynnHufortechnicalassistance,LázaroPeres(UniversidadedeSãoPaulo,Brazil)andAgustin

    Zsögön (Universidade Federal de Viçosa, Brazil) for input on target genes in tomato, Joseph

    Guhlin (College of Biological Sciences, University of Minnesota) for input onMedicago NCR

    target genes, Aaron Hummel for providing the plasmids with structurally optimized gRNA

    scaffolds, Evan Ellison for the GmUbi promoter reagents, and Kit Leffler for help with the

    figures. FundingwasprovidedbygrantsfromtheNationalScienceFoundation (IOS-1339209

    and IOS-1444511) and the Department of Energy (DE-SC0008769).

    AUTHORS’CONTRIBUTIONS

    TCdesignedthevectorsystemandworkplan.TC,JG,CGSandRLGconstructedthevectors.TC

    performedtheexperimentsintomatoprotoplasts.SJCperformedtheexperimentsinMedicago

    truncatula.JGperformedtheexperimentsinwheatprotoplastsandgenetargetingexperiments

    intobaccoandwheat.EKperformedtheexperimentsinbarleyprotoplastsandcarriedoutthe

    molecularanalysisof thegenetargetingevents in tobacco.RCanalyzedthedeepsequencing

    data.TJYKdesignedandbuiltthesetofonlinetools.JJBperformedtheqPCRanalysistoassess

    deletionfrequencies.JWMperformedtheanalysisofGUSstainingintobaccoleaves.TC,JG,SC

    andDVwrotethemanuscript.

  • 57

    NumberofT0plants(outof46)

    Targetgene

    DeletionbetweengRNAsites

    InversionbetweengRNAsites

    IndelatgRNAsite#1only

    IndelatgRNAsite#2only

    IndelsatbothgRNAsites

    Totalmutant

    NCR53(multicopy) 17 4 11 1 23 43NCR54 0 0 0 8 0 8NCR55 7 0 5 0 1 12

    Table1.MultiplextargetedmutagenesisinMedicagotruncatulausingCas9andCsy4.

  • Golden Gate assembly using Type IIs enzymes

    CCDB

    ORClone gRNA(SINGLE STEP)pF U

    S_A

    pF U

    S_B

    pL

    R

    Assemble a TALEN(THREE STEPS)

    Transforma

    tion Backbone (TB)

    A BGolden Gate assembly using Aarl

    CCDB

    Transforma

    tion Backbone (TB)

    A

    B

    C

    CCTG

    CTAG

    GATC

    GGCCGGAC

    CCGG TCAC

    AGTG

    A B C

    Aarl AarlAarl Aarl Aarl Aarl Aarl Aarl

    DIRECT CLONING VECTORS MODULAR ASSEMBLY VECTORS

    Figure 1. Two sets of vectors for direct cloning or modular assembly of genome engineering reagents. A) Direct cloning vectors were designed to speed-up the cloning process. Specificity determining elements (gRNAs or TAL repeats) are cloned directly into the transformation backbone (e.g. T-DNA). B) The modular assembly vectors enable combination of different functional elements. Specificity determining elements (gRNAs, TAL repeatsand donor templates for gene targeting) are first cloned into intermediate module vectors. Custom-selected modules are then assembled together intothe transformation backbone (e.g. T-DNA).

  • A

    C

    TGAATGTCATAAACACTCATGAATTTTGGCCTCATCAGTTTGTGATGGAAAA

    TGAATGTCATAAACACTCATGAAT-----------------GTGATGGAAAA: C1TGAATGTCATAAACACTCATGAATTT--GCCTCATCAGTTTGTGATGGAAAA: C2TGAATGTCATAAACACTCATGAAT----GCCTCATCAGTTTGTGATGGAAAA: C3TGAATGTCATAAACACTCATGAATTTTG-CCTCATCAGTTTGTGATGGAAAA: C4

    WPT52-4-4 - Medtr4g020620

    ATTTAGTTCCTGTGAATGTCATAAACACTCATGAATTTTGGCCTCATCAGTTTGTGATGGAAAAAG

    A---------------------------------------------------------------AG

    WPT52-4-4 - Medtr2g013650

    4g020620

    2g013650

    bar

    ACTIN

    WPT

    52-4

    -1W

    PT52

    -4-2

    WPT

    52-4

    -3W

    PT52

    -4-4

    WPT

    52-4

    -5W

    PT52

    -4-6

    WPT

    52-4

    -7W

    PT52

    -4-8

    HM34

    0 (D

    )HM

    340

    (UD)

    B

    TGTCATAAACACTCATGAATTTTGGCCTCATCAGTTTGTGATGGA

    Medtr4g020620

    4g020620 F

    4g020620 R

    2g013650 R

    2g013650 F

    Medtr2g013650

    Figure 2. TALEN-mediated mutagenesis in Medicago truncatula using direct cloning vectors. A) Maps of two Medicago genes targeted for mutagenesis using a single TALEN pair. PCR primers used for screening in panel B are shown as green arrowheads. The sequence of the TALEN target site is shown with TALEN binding sites underlined. B) HaeIII PCR digestion screening of eight T1 progeny of plant WPT52-4. The amplified locus is indicated on the right. bar and ACTIN are controls that amplify the transgene and a native gene, respectively. HM340 (D), WT product digested with HaeIII; HM340 (UD), undigested WT product. C) DNA sequences of undigested amplicons from plant WPT52-4-4. The reference sequence of the unmodified locus is shown on the top. TALEN binding sites are underlined. HaeIII restriction site used for the screening is in bold red text.

  • 50

    40

    30

    20

    10

    0

    AtU6+

    At7SL

    Impro

    ved g

    RNA

    AtU6+

    AtU6

    Csy4

    tRNA

    Riboz

    yme

    Non-t

    ransfo

    rmed

    ALL MUTATIONSC

    B

    A

    CCCTCAAAGATCAGGCTGGCAGC...62 bp...CCTAATTAACCAGAAAACTTAC

    Unmodified

    Long deletion

    Short indels at gRNA9 site

    Short indels at gRNA10 site

    Short indels at both sites

    Inversion

    Long deletion plus insertion

    gRNA9 gRNA1085bp

    5’ 3’

    Figure 3. Comparison of different systems for expressing multiple gRNAs. A) Structure of six constructs for expressing gRNA9 and gRNA10, which both target the tomato ARF8A gene. CmYLCV, cestrum yellow leaf curling virus promoter; Csy4, 20 bp Csy4 hairpin; tRNA, 77 bp pre-tRNAGly gene; rb, 15 bp ribozyme cleavage site; ribozyme, 58 bp synthetic ribozyme; AtU6, Arabidopsis U6 promoter; At7SL Arabidopsis 7SL promoter. B) Possible editing outcomes due to expression of both gRNAs. Sequences targeted by gRNA9 and gRNA10 are shown on the top Cleavage sites are indicated by arrows. C) Overall mutation frequencies as determined by deep sequencing. Error bars represent SE of three replicates (pools of protoplasts). Statistical significance was determined by Tukey’s test. *P < 0.05 and ***P < 0.001.

    *Improved gRNA scaffold

    Csy4 Csy4 Csy4CmYLCV gRNA9 gRNA10 35S polyA

    CmYLCV gRNA9 gRNA10 35S polyAtRNA tRNAtRNA

    rb ribozymeCmYLCV gRNA9 rb gRNA10 35S polyA

    AtU6 gRNA9 AtU6 gRNA10 TTTTTTTTTTTT

    gRNA9 At7SL gRNA10 TTTTTTTTTTTTAtU6

    gRNA9 At7SL gRNA10 TTTTTTTTTTTTAtU6

    Ove

    rall

    Mut

    atio

    n Fr

    eque

    ncy

  • C

    Figure 4. Evaluation of eight gRNAs in various polycistronic expression systems and the effect of position in the array on gRNA activity. A) Structure of seven constructs for expressing gRNAs 49 -56 targeting four tomato genes for deletion. B) Detection of deletions between gRNA sites using qPCR. Each gene is targeted for deletion with two gRNAs designed to create a 3 kb deletion to prevent amplification of the non-deleted, WT template. Deletion frequency wsa quantified by qPCR using primers shown as red and blue arrowheads. C) Deletion frequencies in tomato protoplasts as measured by qPCR. Positions of respective gRNAs in the array are specified for each construct. Error bars represent SE of two replicates.

    ACm

    YLCV

    gRNA

    49

    gRNA

    50

    35S p

    olyA

    gRNA

    51

    gRNA

    52

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    gRNA

    53

    gRNA

    54

    gRNA

    55

    gRNA

    56

    CmYL

    CV

    gRNA

    56

    gRNA

    55

    35S p

    olyA

    gRNA

    54

    gRNA

    53

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    Csy4

    gRNA

    52

    gRNA

    51

    gRNA

    50

    gRNA

    49

    CmYL

    CV

    35S p

    olyA

    tRNA

    Gly

    gRNA

    49

    gRNA

    50

    gRNA

    51

    gRNA

    52

    gRNA

    53

    gRNA

    54

    gRNA

    55

    gRNA

    56

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    gRNA

    56

    gRNA

    55

    gRNA

    54

    gRNA

    53

    gRNA

    52

    gRNA

    51

    gRNA

    50

    gRNA

    49

    CmYL

    CV

    35S p

    olyA

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    tRNA

    Gly

    rb rb rb rb rb rb rb rbCmYL

    CV

    gRNA

    49

    gRNA

    50

    35S p

    olyA

    gRNA

    51

    gRNA

    52

    riboz

    yme

    gRNA

    53

    gRNA

    54

    gRNA

    55

    gRNA

    56

    CmYL

    CV

    gRNA

    56

    gRNA

    55

    35S p

    olyA

    gRNA

    54

    gRNA

    53

    rb rb rb rb rb rb rb rbgRNA

    52

    gRNA

    51

    gRNA

    50

    gRNA

    49

    riboz

    yme

    gRNA

    49

    gRNA

    50

    gRNA

    51

    gRNA

    52

    gRNA

    53

    gRNA

    54

    gRNA

    55

    gRNA

    56

    AtU6

    TTTT

    TT

    TTTT

    TT

    TTTT

    TT

    TTTT

    TT

    TTTT

    TT

    TTTT

    TT

    TTTT

    TT

    TTTT

    TT

    AtU6

    AtU6

    AtU6

    AtU6

    AtU6

    AtU6

    AtU6

    BWT locus

    3 kb - no amplification

    Deleted locus

    100-200 bp - amplifies in qPCR

    Del

    etio

    n Fr

    eque

    ncy

    Solyc02g077390 - gRNAs 49/50

    Csy4

    - 1/2

    Csy4

    - 7/8

    tRNA

    - 1/2

    tRNA

    - 7/8

    Ribo -

    1/2

    Ribo -

    7/8 U6

    0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2Solyc06g074240 - gRNAs 55/56

    Del

    etio

    n Fr

    eque

    ncy

    Csy4

    - 7/8

    Csy4

    - 1/2

    tRNA

    - 7/8

    tRNA

    - 1/2

    Ribo -

    7/8

    Ribo -

    1/2 U6

    0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2Solyc02g090730 - gRNAs 53/54

    Del

    etio

    n Fr

    eque

    ncy

    Csy4

    - 5/6

    Csy4

    - 3/4

    tRNA

    - 5/6

    tRNA

    - 3/4

    Ribo -

    5/6

    Ribo -

    3/4 U6

    0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

  • C

    A1930 bp (1306 bp)Solyc06g074350

    gRNA11 gRNA12

    1510 bp (409 bp)Solyc02g085500

    gRNA13 gRNA14

    670 bp (388 bp)Solyc02g090730

    gRNA15 gRNA16

    1497 bp (684 bp)Solyc06g074240

    gRNA17 gRNA18

    1933 bp (1536 bp)Solyc02g077390

    gRNA19 gRNA20

    1551 bp (313 bp)Solyc11g071810

    gRNA21 gRNA22

    Solyc06g074350AGATCATGATAGATCCAGATGTTCCTGGTCCTAGTGATCCATATCT // AGAGGATTTGACCATCTTTACAGGATTGTCACAGACATTCCAGGCACTAGATCATGATAGATCCAG---------------------------- // -------------------------------------------GCACTAGATCATGATAGATCCAG---------------------------- // -------------------------------------------GCACTAGATCATGATAGATCCAG---------------------------- // -------------------------------------------GCACT

    gRNA11 gRNA12

    Solyc02g085500TAGTTCCTTCGTGTCTGAAGAAGAAGAATGTGAAGAGGAGATCAATTT // AGTGAAGAAATCTCAGGACCCGTACTAAGATTTCAAGAGATCGATGTAGTTCCTTCG------------------------------------- // -------------------------GAAGATTTCAAGAGATCGATGTAGTTCCTTCG------------------------------------- // -----------------------------ATTTCAAGAGATCGATG

    gRNA13 gRNA14

    Solyc02g090730ATGTTCCTCCTCACTATGTATCTGCCCCCGGCACCACCACGGCGCGGT // TTAGCATGTGGGAGTAGAGGTGCATTATATTGTTTGCTGGGATTGAATGTTCCTCCT------------------------------------- // -----------------------------------GCTGGGATTGAATGTTCCTCCT------------------------------------- // -----------------------------------GCTGGGATTGAATGTTCCTCCTC------------------------------------ // ----------------------------------TGCTGGGATTGA

    gRNA15 gRNA16

    Solyc11g071810AAATCCCTTCGATTCGTCGTAAGTACTCTCTCTATCTCTCTTAATAAA // GAGAAAAGACAACGTGTTCCTTCTGCGTACAACCGATTCATCAAGTAAATCCCTTCGA------------------------------------ // ------------------------GCGTACAACCGATTCATCAAGTAAATCCCTTCGA------------------------------------ // ----------------------------ACAACCGATTCATCAAGTAAATCCCTTCG------------------------------------- // -------------------------CGTACAACCGATTCATCAAGT

    gRNA21 gRNA22

    Solyc06g074240GATCATTATCGGAGCTGGCCCTGCTGGGCTCAGGCTAGCTGAACAAGT // CTATTGGTGGGAATTCAGGGATAGTTCATCCATCAACAGGGTA