METHOD FOR TARGETED MODIFICATION OF SEQUENCE OF PLANT GENOME

Information

  • Patent Application
  • 20230075587
  • Publication Number
    20230075587
  • Date Filed
    September 25, 2020
    3 years ago
  • Date Published
    March 09, 2023
    a year ago
Abstract
The present invention relates to the field of plant genetic engineering. Specifically, the present invention relates to a method for targeted modification of a plant genome sequence. More specifically, the present invention relates to a method for targeted modification of a specific sequence in the plant genome into a target sequence of interest by a nuclease-reverse transcriptase fusion protein guided by a guide RNA, a genetically-modified plant produced by said method, and progenies of said plant.
Description
TECHNICAL FIELD

The present invention relates to the field of plant genetic engineering. Specifically, the present invention relates to a method for targeted modification of a plant genome sequence. More specifically, the present invention relates to a method for targeted modification of a specific sequence in the plant genome into a target sequence of interest by a nuclease-reverse transcriptase fusion protein guided by a guide RNA, a genetically-modified plant produced by said method, and progenies of said plant.


BACKGROUND ART

Many important agronomic traits depend on sequences in the genome. By directionally changing specific sequences of the genome, new heritable traits may be conferred to organisms, providing possibility for disease treatment and breeding improvement. Currently, cleavage of a specific sequences can be achieved by the genome editing techniques (e.g., a CRISPR/Cas technique), so that a repairing pathway of cells is activated to repair injury site, and in turn the sequence at the target site is changed. Precise change to a genome sequence can be achieved based on a homologous recombination (HR) repairing pathway. However, in most higher organisms, especially plants, the homologous recombination efficiency is very low, so that wide application of the method is greatly limited. Furthermore, efficient conversion from cytosine to thymine (C→T) and efficient conversion from adenine to guanine (A→G) within the target site can be achieved by using a single base editing system. However, the method has limited base transition types, and precise insertions or deletions of fragments also cannot be achieved. Thus, there remains a need in the art for an efficient method capable of achieving precise directed modification of the plant genome sequences.


SUMMARY OF THE INVENTION

The present invention comprises a novel plant DNA precise editing system which is comprised of a Cas nuclease having target strand nicking activity (Cas9-H840A) and fused with a reverse transcriptase, and a pegRNA (prime editing gRNA) with a repair template (RT template) and a primer binding site (PBS) of a free single strand at the 3′ end. According to the system, the PBS binds to the free single strand generated by a Cas nickase such as Cas9-H840A, a single-stranded DNA sequence is transcribed according to the given RT template, and any change of the DNA sequence in the genome located downstream part of −3 position of the PAM sequence can be achieved through cell repair. In addition, by introducing a new nicking sgRNA, nicks are generated in non-target strands of the pegRNA, which promote the cell to repare according to the donor template. Experimental results show that the system effectively induces precise modification at target sites in plants.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows a schematic diagram of mechanism underlying the present invention.



FIG. 2 shows a working schematic diagram of three different kinds of PPE (plant prime editor) systems. The system that does not provide an additional nicking sgRNA is named (PPE2); the system that provides an additional nicking sgRNA that facilitates cleavage of opposite strand of pegRNA is named (PPE3); and when the PAM sequence of the nicking sgRNA that cleaves the opposite strand is located within the spacer sequence of the pegRNA, the system is named (PPE3b).



FIG. 3 shows a schematic diagram of the PPE constructs and pegRNA constructs.



FIG. 4 shows a working mechanism of a BFP-to-GFP reporting system for detecting precise editing in plant protoplasts.



FIG. 5 shows a result diagram of fluorescence intensity measured by a flow cytometer for the PPE systems. “CK” is a protoplast control without plasmid transformation, “PBE” is a BE3 single base editing system, and “PPE3b (ΔM-MLV)” represents a control group lacking an M-MLV reverse transcriptase.



FIG. 6 shows efficiency the PPE systems measured by Flow Cytometry. The “CK” is the protoplast control without plasmid transformation, the “PBE” is the BE3 single base editing system, and the “PPE3b (ΔM-MLV)” represents the control group lacking the M-MLV reverse transcriptase.



FIG. 7 shows editing by the PPE systems at rice endogenous targets.



FIG. 8 shows editing by the PPE systems at wheat endogenous targets.



FIG. 9 shows byproducts produced by the PPE systems and their ratios.



FIG. 10 shows editing by a PPE-CaMV system in plant endogenous targets.



FIG. 11 shows a schematic diagram of the pegRNAs initiated by type II promoters and processed by ribozymes.



FIG. 12 shows editing by a PPE-R system in plant endogenous genes.



FIG. 13 shows temperature treatments for improving editing efficiency of the PPE systems.



FIG. 14 shows influences of different PBS lengths on the PPE systems.



FIG. 15 shows influences of different RT template lengths on the PPE systems.



FIG. 16 shows influences of the different RT template lengths on the precise editing ratio of the PPE systems.



FIG. 17 shows influences of different nicking sgRNA locations on the PPE systems.



FIG. 18 shows different types of mutations achieved by the PPE systems in the plant endogenous genes.



FIG. 19 shows insertions of fragments of different lengths by the PPE systems in the plant endogenous genes.



FIG. 20 shows deletions of fragments of different lengths by the PPE systems in the plant endogenous genes.



FIG. 21 shows a schematic diagram of the PPE constructs for rice agrobacterium infection.



FIG. 22 shows obtaining of rice mutants and sequencing results thereof by using the PPE systems, and the arrow indicates the location of a target mutation.



FIG. 23 shows monoclonal sequencing results of T0-9 mutant plants.



FIG. 24 shows comparison of influences of different Tm-guided PBS lengths on editing efficiency by using published data for three target sites and newly obtained data for ten new target sites in rice protoplasts.



FIG. 25 shows normalization of priming editing frequencies with different PBS melting temperatures. The highest editing frequency obtained at each target is normalized to 1, and the frequencies obtained at other PBS Tms are adjusted accordingly.



FIG. 26 shows a schematic diagram of priming editing by using single-pegRNA and double-pegRNA strategies. (a) Only an NGG-pegRNA is used for editing (a forward DNA strand is edited). (b) Only a CCN-pegRNA is used for editing (a reverse DNA strand is edited). (c) The double-pegRNA strategy is used for editing. Double-pegRNAs create two edits in the two DNA strands simultaneously.



FIG. 27 shows comparison of editing efficiency induced by NGG-pegRNA, CCN-pegRNA and double-pegRNAs at 15 target sites.



FIG. 28 shows product purity of editing at 15 endogenous sites of the rice protoplasts using NGG-pegRNA, CCN-pegRNA and double-pegRNAs.



FIG. 29 shows the percentage of rice genomic bases that theoretically can be targeted by priming editing with single pegRNAs and double-pegRNAs.





DETAILED DESCRIPTION OF THE INVENTION
1. Definition

In the present invention , unless indicated otherwise, the scientific and technological terminologies used herein refer to meanings commonly understood by a person skilled in the art. Also, the terminologies and experimental procedures used herein relating to protein and nucleotide chemistry, molecular biology, cell and tissue cultivation, microbiology, immunology, all belong to terminologies and conventional methods generally used in the art. For example, the standard DNA recombination and molecular cloning technology used herein are well known to a person skilled in the art, and are described in details in the following references: Sambrook, J., Fritsch, E. F. and Maniatis, T., Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, 1989. In the meantime, in order to better understand the present invention, definitions and explanations for the relevant terminologies are provided below.


As used herein, the term “and/or” encompasses all combinations of items connected by the term, and each combination should be regarded as individually listed herein. For example, “A and/or B” covers “A”, “A and B”, and “B”. For example, “A, B, and/or C” covers “A”, “B”, “C”, “A and B”, “A and C”, “B and C”, and “A and B and C”.


When the term “comprise” is used herein to describe the sequence of a protein or nucleic acid, the protein or nucleic acid may consist of the sequence, or may have additional amino acids or nucleotide at one or both ends of the protein or nucleic acid, but still have the activity described in this invention. In addition, those skilled in the art know that the methionine encoded by the start codon at the N-terminus of the polypeptide will be retained under certain practical conditions (for example, when expressed in a specific expression system), but does not substantially affect the function of the polypeptide. Therefore, when describing the amino acid sequence of specific polypeptide in the specification and claims of the present application, although it may not include the methionine encoded by the start codon at the N-terminus, the sequence containing the methionine is also encompassed, correspondingly, its coding nucleotide sequence may also contain a start codon; vice versa.


“Genome” as used herein encompasses not only chromosomal DNA present in the nucleus, but also organelle DNA present in the subcellular components (e.g., mitochondria, plastids) of the cell.


A “genetically modified plant” means a plant which comprises an exogenous polynucleotide or comprises a modified gene or expression regulatory sequence within its genome. For example, the exogenous polynucleotide can be stably integrated into the genome of the plant and inherited in successive generations. The exogenous polynucleotide may be integrated into the genome alone or as a part of a recombinant DNA construct. The modified gene or expression regulatory sequence is a gene or expression regulatory sequence comprising one or more nucleotide substitutions, deletions and additions in the plant genome.


The term “exogenous” with respect to sequence means a sequence that originates from a foreign species, or, if from the same species, is substantially modified from its native form in composition and/or genomic locus by deliberate human intervention.


“Polynucleotide”, “nucleic acid sequence”, “nucleotide sequence”, or “nucleic acid fragment” are used interchangeably to refer to a polymer of RNA or DNA that is single- or double-stranded, optionally containing synthetic, non-natural or altered nucleotide bases. Nucleotides (usually found in their 5′-monophosphate form) are referred to by their single letter designation as follows: “A” for adenylate or deoxyadenylate (for RNA or DNA, respectively), “C” for cytidylate or deoxycytidylate, “G” for guanylate or deoxyguanylate, “U” for uridylate, “T” for deoxythymidylate, “R” for purines (A or G), “Y” for pyrimidines (C or T), “K” for G or T, “H” for A or C or T, “I” for inosine, and “N” for any nucleotide.


“Polypeptide”, “peptide”, “amino acid sequence” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical analogue of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers. The terms “polypeptide”, “peptide”, “amino acid sequence”, and “protein” are also inclusive of modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation.


As used herein, an “expression construct” refers to a vector suitable for expression of a nucleotide sequence of interest in an organism, such as a recombinant vector. “Expression” refers to the production of a functional product. For example, the expression of a nucleotide sequence may refer to transcription of the nucleotide sequence (such as transcribe to produce an mRNA or a functional RNA) and/or translation of RNA into a protein precursor or a mature protein.


“Expression construct” of the invention may be a linear nucleic acid fragment, a circular plasmid, a viral vector, or, in some embodiments, an RNA that can be translated (such as an mRNA).


“Expression construct” of the invention may comprise regulatory sequences and nucleotide sequences of interest that are derived from different sources, or regulatory sequences and nucleotide sequences of interest derived from the same source, but arranged in a manner different than that normally found in nature.


“Regulatory sequence” or “regulatory element” are used interchangeably and refer to nucleotide sequences located upstream (5′ non-coding sequences), within, or downstream (3′ non-coding sequences) of a coding sequence, and which influence the transcription, RNA processing or stability, or translation of the associated coding sequence. Regulatory sequences may include, but are not limited to, promoters, translation leader sequences, introns, and polyadenylation recognition sequences.


“Promoter” refers to a nucleic acid fragment capable of controlling the transcription of another nucleic acid fragment. In some embodiments of the present invention, the promoter is a promoter capable of controlling the transcription of a gene in a cell, whether or not it is derived from the cell. The promoter may be a constitutive promoter or a tissue-specific promoter or a developmentally-regulated promoter or an inducible promoter.


“Constitutive promoter” refers to a promoter that may cause expression of a gene in most circumstances in most cell types. “Tissue-specific promoter” and “tissue-preferred promoter” are used interchangeably, and refer to a promoter that is expressed predominantly but not necessarily exclusively in one tissue or organ, but that may also be expressed in one specific cell or cell type. “Developmentally regulated promoter” refers to a promoter whose activity is determined by developmental events. “Inducible promoter” selectively expresses a DNA sequence operably linked to it in response to an endogenous or exogenous stimulus (environment, hormones, or chemical signals, and so on).


Examples of promoters that can be used in the present invention include but are not limited to polymerase (pol) I, pol II or pol III promoters. Examples of pol I promoters include chicken RNA pol I promoter. Examples of pol II promoters include but are not limited to cytomegalovirus immediate early(CMV) promoter, rous sarcoma virus long terminal repeat(RSV-LTR) promoter and simian virus 40(SV40) immediate early promoter. Examples of pol III promoters include U6 and H1 promoter. Inducible promoter such as metalothionein promoter can be used. Other examples of promoters include T7 bacteriophage promoter, T3 bacteriophage promoter, β-galactosidase promoter and Sp6 bacteriophage promoter etc. When used for plants, promoters that can be used include but are not limited to cauliflower mosaic virus 35S promoter, maize Ubi-1 promoter, wheat U6 promoter, rice U3 promoter, maize U3 promoter and rice actin promoter etc.


As used herein, the term “operably linked” means that a regulatory element (for example but not limited to, a promoter sequence, a transcription termination sequence, and so on) is associated to a nucleic acid sequence (such as a coding sequence or an open reading frame), such that the transcription of the nucleotide sequence is controlled and regulated by the transcriptional regulatory element. Techniques for operably linking a regulatory element region to a nucleic acid molecule are known in the art.


“Introduction” of a nucleic acid molecule (e.g., plasmid, linear nucleic acid fragment, RNA, etc.) or protein into an organism means that the nucleic acid or protein is used to transform a cell of the organism such that the nucleic acid or protein functions in the cell. As used in the present invention, “transformation” includes both stable and transient transformations. “Stable transformation” refers to the introduction of an exogenous nucleotide sequence into the genome, resulting in the stable inheritance of foreign genes. Once stably transformed, the exogenous nucleic acid sequence is stably integrated into the genome of the organism and any of its successive generations. “Transient transformation” refers to the introduction of a nucleic acid molecule or protein into a cell, performing its function without the stable inheritance of an exogenous gene. In transient transformation, the exogenous nucleic acid sequence is not integrated into the genome.


“Trait” refers to the physiological, morphological, biochemical, or physical characteristics of a cell or an organism.


“Agronomic trait” is a measurable parameter including but not limited to, leaf greenness, yield, growth rate, biomass, fresh weight at maturation, dry weight at maturation, fruit yield, seed yield, total plant nitrogen content, fruit nitrogen content, seed nitrogen content, nitrogen content in a vegetative tissue, total plant free amino acid content, fruit free amino acid content, seed free amino acid content, free amino acid content in a vegetative tissue, total plant protein content, fruit protein content, seed protein content, protein content in a vegetative tissue, drought tolerance, nitrogen uptake, root lodging, harvest index, stalk lodging, plant height, ear height, ear length, disease resistance, cold resistance, salt tolerance, and tiller number and so on.


II. Plant Genome Editing System

In one aspect, the present invention relates to a genome editing system for targeted modification of a genomic DNA sequence of an organism, comprising


i) a fusion protein and/or an expression construct comprising a nucleotide sequence encoding the fusion protein, wherein the fusion protein comprises a CRISPR nickase and a reverse transcriptase; and/or


ii) at least one pegRNA and/or an expression construct comprising nucleotide sequence(s) encoding the at least one pegRNA,


wherein each of the at least one pegRNA comprises a guide sequence, a scaffold sequence, a reverse transcription (RT) template sequence and a primer binding site (PBS) sequence in the direction from 5′ to 3′.


In some embodiments, the at least one pegRNA is capable of forming a complex with the fusion protein and targeting the fusion protein to the target sequence in the genome, resulting in a nick in the target sequence.


In some embodiments, the organism is a plant.


As used herein, a “genome editing system” refers to a combination of components required for genome editing of the genome in a cell. The individual components, e.g., the fusion protein, the gRNA, etc., of the system may be present independently of each other, or may be present in any combination as a composition.


As used herein, a “target sequence” refer to a sequence which is approximately 20 nucleotides in length in the genome and is represented by the 5′ or 3′ flanking PAM (protospacer adjacent motif) sequence. In general, PAM is required for recognizing the target sequence by the complex formed by the CRISPR nuclease or a variant thereof and the guide RNA. For example, for the Cas9 nuclease and a variant thereof, the target sequence is, at the 3′ end, closely adjacent to the PAM , for example, 5′-NGG-3′. Based on the presence of PAM, those skilled in the art may readily determine the target sequences available in the genome for targeting. In addition, depending on the locations of the PAMs, the target sequences may be located on any strand of genome DNA molecule. For Cas9 or derivatives thereof such as the Cas9 nickase, the target sequences are preferably 20 nucleotides in length.


In some embodiments, the CRISPR nickase in the fusion protein is capable of forming a nick within the target sequence in the genome DNA. In some embodiments, the CRISPR nickase is a Cas9 nickase.


In some embodiments, the Cas9 nickase is derived from Streptococcus pyogenes (S. pyogenes) SpCas9, and comprises at least an amino acid substitution H840A relative to wild type SpCas9. The exemplary wild type SpCas9 comprises an amino acid sequence set forth in SEQ ID NO: 1. In some embodiments, the Cas9 nickase comprises an amino acid sequence set forth in SEQ ID NO: 2. In some embodiments, the Cas9 nickase in the fusion protein is capable of forming a nick between the −3 position nucleotide (the first nucleotide at the 5′ end of each PAM sequence is considered as the +1 site nucleotide) and the −4 position nucleotide of the PAM of the target sequence.


In some embodiments, the Cas9 nickase is a Cas9 nickase variant capable of recognizing an altered PAM sequence. In some preferred embodiments, the Cas9 nickase is a Cas9 variant that recognizes the PAM sequence 5′-NG-3′. In some embodiments, the Cas9 nickase variant that recognizes the PAM sequence 5′-NG-3′ comprises the following amino acid substitutions H840A, R1335V, L1111R, D1135V, G1218R, E1219F, A1322R and T1337R relative to the wild type Cas9, wherein the amino acid numbering refers to SEQ ID NO: 1.


The nick formed by the Cas9 nickase of the present invention is capable of causing the target sequence to form a free single strand at the 3′ terminal (3′ free single strand) and a free single strand at the 5′ terminal (5′ free single strand).


In some embodiments, the reverse transcriptase in the fusion protein of the present invention may be derived from different sources. In some embodiments, the reverse transcriptase is a bacteria-derived reverse transcriptase. For example, in some embodiments, the reverse transcriptase is an M-MLV reverse transcriptase or a functional variant thereof. An exemplary wild type M-MLV reverse transcriptase sequence is set forth in SEQ ID NO: 3. In some embodiments, the reverse transcriptase is an enhanced M-MLV reverse transcriptase, for example, the amino acid sequence of the enhanced M-MLV reverse transcriptase is set forth in SEQ ID NO: 4. In some embodiments, the reverse transcriptase is CaMV-RT from Cauliflower mosaic viruses (CaMV), and an amino acid sequence of the CaMV-RT is set forth in SEQ ID NO: 5. In some embodiments, the reverse transcriptase is the bacteria-derived reverse transcriptase, for example, retron-RT from Escherichia coli, and an amino acid sequence of the retron-RT is set forth in SEQ ID NO: 6.


In some embodiments, the CRISPR nickase and the reverse transcriptase in the fusion protein are linked by a linker. As used herein, a “linker” may be a non-functional amino acid sequence which is 1-50 (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 20-25 or 25-50) or more amino acids in length and is free of secondary or higher structures. For example, the linker may be a flexible linker, for example, GGGGS, GS, GAP, (GGGGS)x3, GGS, (GGS) x7, etc. For example, the linker may be a linker set forth in SEQ ID NO: 7.


In some embodiments, the CRISPR nickase in the fusion protein is fused to the N-terminus of the reverse transcriptase directly or through a linker. In some embodiments, the CRISPR nickase in the fusion protein is fused to the C-terminus of the reverse transcriptase directly or through a linker.


In some embodiments of the present invention, the fusion protein of the present invention may further comprise a nuclear localization sequence (NLS). In general, one or more NLSs in the fusion protein should be of sufficient strength to drive accumulation of the fusion protein in nuclei of the cells in an amount that can realize its editing function. In general, the strength of nuclear localization activity is determined by the number of NLS in the fusion protein, location of NLS in the fusion protein, the one or more specific NLSs as used in the fusion protein, or a combination of these factors.


The guide sequence (also referred to as seed sequence or spacer sequence) in at least one pegRNA of the present invention is configured to have sufficient sequence identity (preferably 100% identity) to the target sequence so as to achieve sequence-specific targeting by binding to complementary strand of the target sequence through base pairing.


A variety of scaffold sequences of the gRNAs suitable for genome editing based on the CRISPR nuclease (e.g. Cas9) are known in the field and may be used in the pegRNA of the present invention. In some embodiments, the scaffold sequence of the gRNA is set forth in SEQ ID NO: 8.


In some embodiments, the primer binding sequence is configured to be complementary to at least a portion of the target sequence (preferably fully paired with at least a portion of the target sequence), and preferably, the primer binding sequence is complementary to (preferably fully paired with at least a portion of the 3′ free single strand) at least a portion of the 3′ free single strand caused by the nick in the DNA strand where the target sequence is located, in particular complementary to (preferably fully paired with) the nucleotide sequences at the 3′ terminal of the 3′ free single strand. When the 3′ free single strand of the strand binds to the primer binding sequence by base pairing, the 3′ free single strand can serve as a primer, and the reverse transcription (RT) template sequence closely adjacent to the primer binding sequence can serve as a template for reverse transcription by the reverse transcriptase in the fusion protein, and the DNA sequence corresponding to the reverse transcription (RT) template sequence is obtained through extension.


The primer binding sequence depends on the length of the free single strand formed in the target sequence by the CRISPR nickase, however, they should have a minimum length capable of ensuring specific binding. In some embodiments, the primer binding sequence may be 4-20 nucleotides in length, for example, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 and 20 nucleotides in length.


In some embodiments, the primer binding sequence is configured to have a Tm (melting temperature) of no more than about 52° C. In some embodiments, the Tm (melting temperature) of the primer binding sequences is about 18° C-52° C., preferably about 24° C-36° C., more preferably about 28° C-32° C., and most preferably about 30° C.


Methods for calculating the Tm of a nucleic acid sequence is well known in the field, for example, an Oligo Analysis Tool online analytical tool may be used for calculation. An exemplary calculation formula is Tm=NG:C*4+NA:T*2, wherein NG:C is the number of G and C bases in the sequence, and NA:T is the number of A and T bases in the sequence. The suitable Tm may be obtained by selecting a suitable PBS length. Alternatively, PBS sequence with the appropriate Tm may be obtained by selecting the appropriate target sequence.


In some embodiments, the RT template sequence may be any sequence. Through the above reverse transcription, sequence information thereof may be integrated into the DNA strand where the target sequence is located (namely, the strand containing the PAM of the target sequence), and then DNA double strands containing the sequence information of the RT template are formed through DNA repairing of the cell. In some embodiments, the RT template sequence comprises desired modification(s). For example, the desired modification comprises substitution, deletion and/or addition of one or more nucleotide. For example, the modification comprises one or more substitutions selected from: C to T substitution, C to G substitution, C to A substitution, G to T substitution, G to C substitution, G to A substitution, A to T substitution, A to G substitution, A to C substitution, T to C substitution, T to G substitution and T to A substitution; and/or comprises deletion of one or more nucleotides, for example, deletion of 1 to about 100 or more nucleotides, e.g. deletion of 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100 nucleotides; and/or comprises insertion of one or more nucleotides, for example, insertion of 1 to about 100 or more nucleotides, for example, insertion of 1 to about 100 or more nucleotides, for example, insertion of 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100 nucleotides.


In some embodiments, the RT template sequence is configured as corresponding to the sequence downstream the nick of the target sequence (e.g., being complementary to at least a portion of the sequence downstream the nick of the target sequence) and contain desired modification(s). For example, the desired modification comprises substitution, deletion and/or addition of one or more nucleotide. For example, the modification comprises one or more substitutions selected from: C to T substitution, C to G substitution, C to A substitution, G to T substitution, G to C substitution, G to A substitution, A to T substitution, A to G substitution, A to C substitution, T to C substitution, T to G substitution and T to A substitution; and/or comprises deletion of one or more nucleotides, for example, deletion of 1 to about 100 or more nucleotides, e.g. deletion of 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100 nucleotides; and/or comprises insertion of one or more nucleotides, for example, insertion of 1 to about 100 or more nucleotides, for example, insertion of 1 to about 100 or more nucleotides, for example, insertion of 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100 nucleotides.


In some embodiments, the RT template sequence may be about 1-300 or more nucleotides in length, for example, 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, about 300 nucleotides or more nucleotides in length. Preferably, the RT template sequence is 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 and 23 nucleotides in length.


In some embodiments, the plant genome editing system further comprises a nicking gRNA (the nicking gRNA is used for generating an additional nick) and/or an expression construct comprising a nucleotide sequence encoding the nicking gRNA, wherein the nicking gRNA comprises a guide sequence and a scaffold sequence. In some preferred embodiments, the nicking gRNA does not contain the reverse transcription (RT) template sequence and the primer binding site (PBS) sequence.


The guide sequence (also referred to as seed sequence or spacer sequence) in the nicking gRNA of the present invention is configured to have sufficient sequence identity (preferably 100% identity) to a nicking target sequence in the genome, so that the fusion protein of the present invention is targeted to the nicking target sequence, resulting in a nick in the nicking target sequence, wherein the nicking target sequence and the target sequence (pegRNA target sequence) targeted by the pegRNA are located on the opposite strand of the genome DNA. In some embodiments, the nick formed by the nicking RNA and the nick formed by the pegRNA are about 1 to about 300 or more nucleotides apart, e.g., 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, about 300 nucleotides or more nucleotides apart. In some embodiments, the nick created by the nicking RNA is located upstream or downstream of the nick formed by the pegRNA (upstream or downstream refers to the DNA strand on which the pegRNA target sequence is located). In some embodiments, the guide sequence in the nicking gRNA has sufficient sequence identity (preferably 100% identity) to the pegRNA target sequence on the opposite strand after editing event occur (modified pegRNA target sequence), so that the nicking gRNA targets only the nicking target sequence that is generated after pegRNA-induced sequence targeting and modification. In some embodiments, the PAM of the nicking target sequence is located within the complementary sequence of the pegRNA target sequence.


In some embodiments, the sequence of the pegRNA and/or the nicking gRNA may be precisely processed by using a self-processing system. In some specific embodiments, the 5′ end of the pegRNA and/or the nicking gRNA are linked to the 3′ end of a first ribozyme, wherein the first ribozyme is designed to cleave the fusion at the 5′ end of the pegRNA and/or the nicking gRNA; and/or the 3′ end of the pegRNA and/or the nicking gRNA is linked to the 5′ end of a second ribozyme, wherein the second ribozyme is designed to cleave the fusion at the 3′ end of the pegRNA and/or the nicking gRNA. The design of the first or second ribozyme is within the capabilities of those skilled in the art. For example, see Gao et al. JIPB, apr, 2014; Vol 56, Issue 4, 343-349. As for a method for precise processing of gRNA, see, for example, WO 2018/149418.


In some embodiments, the genome editing system comprises at least one pair of pegRNAs and/or expression construct comprising the nucleotide sequence encoding the at least one pair of pegRNAs. In some embodiments, the two pegRNAs of the pair of pegRNAs are configured to target different target sequences on the same strand of the genome DNA. In some embodiments, the two pegRNAs of the pair of pegRNAs are configured to target the target sequences on different strands of the genome DNA. In some embodiments, the PAM for the target sequence of one pegRNA of the pair of pegRNAs is located on a sense strand, and the PAM for the other pegRNA is located on an antisense strand. In some embodiments, the induced nicks of the two pegRNAs are located on both sides of a site to be modified respectively. In some embodiments, the pegRNA-induced nick for the sense strand is located upstream (5′ direction) of the site to be modified, and the pegRNA-induced nick for the antisense strand is located downstream (3′ direction) of the site to be modified. The “upstream” or “downstream” refers to the sense strand. In some embodiments, the induced nicks of the two pegRNAs are about 1 to about 300 or more nucleotides apart, for example, 1-15 nucleotides apart.


In some embodiments, the two pegRNAs in the pair of pegRNAs are configured to introduce the same desired modification. For example, one pegRNA is configured to introduce a A to G substitution in the sense strand, and the other pegRNA is configured to introduce a T to C substitution at the corresponding position of the antisense strand. For another example, one pegRNA is configured to introduce two nucleotide deletions in the sense strand, and the other pegRNA is configured to similarly introduce two nucleotide deletions at the corresponding position of the antisense strand. Other types of modification may be made in a similar manner. Designing appropriate RT template sequences can allow the pegRNAs targeting two different strands to achieve the same desired modification.


In order to obtain efficient expression in plants, in some embodiments of the present invention, the nucleotide sequence encoding the fusion protein is subjected to codon optimization for plant species whose genome is to be modified.


Codon optimization refers to a process of modifying a nucleic acid sequence for enhanced expression in the host cells of interest by replacing at least one codon (e.g. about or more than about 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, or more codons) of the native sequence with codons that are more frequently or most frequently used in the genes of that host cell while maintaining the native amino acid sequence. Various species exhibit particular bias for certain codons of a particular amino acid. Codon bias (differences in codon usage between organisms) often correlates with the efficiency of translation of messenger RNA (mRNA), which is in turn believed to be dependent on, among other things, the properties of the codons being translated and the availability of particular transfer RNA (tRNA) molecules. The predominance of selected tRNAs in a cell is generally a reflection of the codons used most frequently in peptide synthesis. Accordingly, genes can be tailored for optimal gene expression in a given organism based on codon optimization. Codon usage tables are readily available, for example, at the“Codon Usage Database” available at www.kazusa.orjp/codon/ and these tables can be adapted in a number of ways. See Nakamura, Y, et al.“Codon usage tabulated from the international DNA sequence databases: status for the year 2000” Nucl. Acids Res. 28:292 (2000).


In some embodiments, the fusion protein of the present invention is encoded by nucleotide sequence of any one of SEQ ID NOs: 9-11, or comprises amino acid sequence of any one of SEQ ID NOs: 12-14.


Plants that can be genome-modified by the genome editing system of the invention include monocots and dicots. For example, the plant may be a crop plant such as wheat, rice, corn, soybean, sunflower, sorghum, canola, alfalfa, cotton, barley, millet, sugar cane, tomato, tobacco, tapioca or potato.


In another aspect, the present invention provides a method of determining the PBS sequence in the pegRNA of the genome editing system of the present invention, comprising the steps of:


a) determining at least one candidate target sequence based on the PAM recognized by the CRISPR nickase as used and the site to be modified;


b) obtaining a series of PBS sequences based on the nick location which can be generated by the CRISPR nickase at the at least one candidate target sequence;


c) calculating the Tm of the PBS sequences; and


d) selecting the PBS sequences with Tm of not more than 52° C., for example, Tm of about 18° C. to 52° C., preferably about 24° C. to 36° C., more preferably about 28° C. to 32° C., and most preferably about 30° C.


III. Method of Producing a Genetically Modified Plant

In another aspect, the present invention provides a method of producing a genetically modified plant, comprising the step of introducing the genome editing system of the present invention into at least one plant, resulting in modification in genome of the at least one plant. The modification comprises substitution, deletion and/or addition of one or more nucleotide. For example, the modification comprises one or more substitutions selected from: C to T substitution, C to G substitution, C to A substitution, G to T substitution, G to C substitution, G to A substitution, A to T substitution, A to G substitution, A to C substitution, T to C substitution, T to G substitution and T to A substitution; and/or comprises deletion of one or more nucleotides, for example, deletion of 1 to about 100 or more nucleotides, e.g. deletion of 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100 nucleotides; and/or comprises insertion of one or more nucleotides, for example, insertion of 1 to about 100 or more nucleotides, for example, insertion of 1 to about 100 or more nucleotides, for example, insertion of 1, 2, 3, 4, 5, about 10, about 20, about 30, about 40, about 50, about 75, about 100 nucleotides.


In some embodiments, the method further comprises the step of screening the plant with the desired modification from the at least one plant.


In the method of the present invention, the genome editing system may be introduced into the plants in a variety of methods well known to those skilled in the art. The methods used for introducing the genome editing system of the present invention into a plant include, but are not limited to a gene gun method, PEG-mediated protoplast transformation, Agrobacterium-mediated transformation, plant virus-mediated transformation, pollen tube pathway and ovary injection method. Preferably, the genome editing system is introduced into the plant by transient transformation.


In the method of the present invention, modification of the genome may be achieved by only introducing or producing the fusion protein and the gRNAs in the plant cells, and the modification can be stably inherited without stably transforming exogenous polynucleotides encoding the components of the genome editing system into the plant. This avoids the potential off-target effects due to the stably existing (continuously produced) genome editing system and also avoids the integration of the exogenous nucleotide sequences in the plant genome, thereby providing greater biosafety.


In some preferred embodiments, the introduction is carried out in the absence of selection pressure to avoid integration of the exogenous nucleotide sequence into the plant genome.


In some embodiments, the introduction comprises transforming the genome editing system of the present invention into an isolated plant cell or tissue and then regenerating the transformed plant cell or tissue into an intact plant. Preferably, the regeneration is carried out in the absence of selection pressure, i.e., no selection agent for the selectable gene on the expression vector is used during tissue culture. Avoiding the use of a selection agent can increase the regeneration efficiency of the plant, obtaining a modified plant free of exogenous nucleotide sequences.


In other embodiments, the genome editing system of the present invention can be transformed into specific parts of an intact plant, such as leaves, shoot tips, pollen tubes, young ears or hypocotyls. This is particularly suitable for the transformation of plants that are difficult to regenerate in tissue culture.


In some embodiments of the invention, the in vitro expressed protein and/or the in vitro transcribed RNA molecule (e.g., the expression construct is in vitro transcribed RNA molecule) are directly transformed into the plant. The protein and/or RNA molecule is capable of performing genome editing in plant cells and is subsequently degraded by the cell, avoiding integration of the exogenous nucleotide sequence in the plant genome.


Thus, in some embodiments, using the method of the present invention for plant genetic modification and breeding can obtain plants with the genome free of exogenous polynucleotide integration, namely transgene-free modified plants.


In some embodiments, the method further comprises the step of culturing the plant cell, tissue or whole plant into which the genome editing system has been introduced at an elevated temperature, for example, the elevated temperature is 37° C.


In some embodiments of the present invention, the modified genome region is associated with plant traits, such as the agronomic traits, so that modified results in a plant having altered (preferably improved) traits, such as agronomic traits, relative to a wild type plant.


In some embodiments, the method further comprises the step of screening a plant having the desired modification and/or desired traits, such as agronomic traits.


In some embodiments of the present invention, the method further comprises the step of obtaining progenies of the genetically modified plant. Preferably, the genetically modified plant or the progenies thereof have the desired modification and/or the desired traits, such as agronomic traits.


In another aspect, the invention also provides a genetically modified plant or progenies or a part thereof, wherein the plant is obtained by the method according to the present invention as described above. In some embodiments, the genetically modified plant or the progenies or a part thereof is transgene-free. Preferably, the genetically modified plant or progenies thereof have the desired genetic modification and/or the desired traits, such as agronomic traits.


In another aspect, the present invention provides a method of plant breeding comprising crossing a first genetically modified plant obtained by the above method of the present invention with a second plant not containing the modification, thereby the genetic modification is introduced into the second plant. Preferably, the first genetically modified plant has the desired traits, such as the agronomic traits.


EXAMPLES
Materials and Methods
1. Vector Construction

The nCas9 (H840A)-M-MLV construct, the nCas9 (H840A)-CaMV construct and the nCas9 (H840A)-retron construct were constructed by Suzhou Jinweizhi Co. Ltd. The M-MLV used in the examples has 5 amino acid mutations compared to the wild type M-MLV reverse transcriptase. The M-MLV, the RT-CaMV and the RT-retron are all subjected to codon optimization for monocots.


The pegRNA fragments (including the RT and PBS sequences) were constructed into the vectors driven by OsU3 promoter by using a Gibson method to obtain OsU3-pegRNA constructs suitable for use in rice. The pegRNA fragments (including the RT and PBS sequences) were constructed into vectors driven by TaU6 promoter by using the Gibson method to obtain TaU6-pegRNA constructs suitable for use in wheat. The pegRNA fragments (including the RT and PBS sequences) with ribozymes at both the 5′ end and the 3′ end were constructed into vectors driven by maize Ubiquitin-1 (Ubi-1) promoter by using the Gibson method to obtain Ubi-pegRNA-R constructs.


The nicking gRNAs were constructed into vectors driven by TaU3 promoter through T4 ligase to obtain TaU3-nick vectors.









TABLE 1







List of the pegRNA targeting sites and the RT and PBS sequences














Target site 
RT template and 

PBS
RT
Homologous


pegRNA
sequence
PBS sequence
Editing type
length
length
arm length





BFP-peg0l
CTCGTGACCACCTT
TGCACGCCGTACGTGA
+1-2 CC to GT
14
12
10



CACCCACGG
AGGTGGTCAC









OsAAT-peg0l
CAAGGATCCCAGCC
AGATTGACCTGGACGG
+2-3 GA to CC
12
13
10



CCGTGAAGG
GGCTGGGAT









OsALS-Tl-peg01
GGGTATGGTGGTGC
TAAAACCTATCCTCCAA
+1 G to T
12
16
15



AATGGGAGG
TTGCACCACCA









OsALS-T2-peg01
GTGCTGCCTATGATC
TGAATGCGCCCCCAATT
+3 G to T
10
17
14



CCAAGTGG
GGGATCATAG









OsALS-T2-peg02
GTGCTGCCTATGATC
TGAATGCGCCCCCACCT
+2 A to G
10
17
15



CCAAGTGG
GGGATCATAG









OsALS-T2-pegAT
GTGCTGCCTATGATC
TGAATGCGCCCCCACAT
+2 A to T
10
17
15



CCAAGTGG
GGGATCATAG









OsALS-T2-pegAC
GTGCTGCCTATGATC
TGAATGCGCCCCCACG
+2 A to C
10
17
15



CCAAGTGG
TGGGATCATAG









OsALS-T2-pegTA
GTGCTGCCTATGATC
TGAATGCGCCCCCTCTT
+4 T to A
10
17
13



CCAAGTGG
GGGATCATAG









OsALS-T2-pegTC
GTGCTGCCTATGATC
TGAATGCGCCCCCGCTT
+4 T to C
10
17
13



CCAAGTGG
GGGATCATAG









OsALS-T2-pegTG
GTGCTGCCTATGATC
TGAATGCGCCCCCCCTT
+4 T to G
10
17
13



CCAAGTGG
GGGATCATAG









OsALS-T2-pegGA
GTGCTGCCTATGATC
TGAATGCGCCCCCATTT
+3 G to A
10
17
14



CCAAGTGG
GGGATCATAG









OsALS-T2-pegAGCA
GTGCTGCCTATGATC
TGAATGCGCCCCCATGT
+2-3 AG to CA
10
17
14



CCAAGTGG
GGGATCATAG









OsALS-T2-pegPBS6
GTGCTGCCTATGATC
TGAATGCGCCCCCACCT
+2 A to G
 6
17
15



CCAAGTGG
GGGATC









OsALS-T2-pegPBS8
GTGCTGCCTATGATC
TGAATGCGCCCCCACCT
+2 A to G
 8
17
15



CCAAGTGG
GGGATCAT









OsALS-T2-pegPBS12
GTGCTGCCTATGATC
TGAATGCGCCCCCACCT
+2 A to G 
12
17
15



CCAAGTGG
GGGATCATAGGC









OsALS-T2-pegPBS14
GTGCTGCCTATGATC
TGAATGCGCCCCCACCT
+2 A to G
14
17
15



CCAAGTGG
GGGATCATAGGCAG









OsALS-T2-pegPBS16
GTGCTGCCTATGATC
TGAATGCGCCCCCACCT
+2 A to G
16
17
15



CCAAGTGG
GGGATCATAGGCAGCA









OsALS-T2-pegRT10
GTGCTGCCTATGATC
GCCCCCACCTGGGATCA
+2A to G
10
10
 8



CCAAGTGG
TAG









OsALS-T2-pegRT11
GTGCTGCCTATGATC
CGCCCCCACCTGGGAT
+2 A to G
10
11
 9



CCAAGTGG
CATAG









OsALS-T2-pegRT13
GTGCTGCCTATGATC
TGCGCCCCCACCTGGG
+2 A to G
10
13
11



CCAAGTGG
ATCATAG









OsALS-T2-pegRT15
GTGCTGCCTATGATC
AATGCGCCCCCACCTG
+2 A to G
10
15
13



CCAAGTGG
GGATCATAG









OsALS-T2-pegRT18
GTGCTGCCTATGATC
TTGAATGCGCCCCCACC
+2 A to G 
10
18
16



CCAAGTGG
TGGGATCATAG









OsALS-T2-pegRT19
GTGCTGCCTATGATC
CTTGAATGCGCCCCCAC
+2 A to G
10
19
17



CCAAGTGG
CTGGGATCATAG









OsALS-T2-pegRT21
GTGCTGCCTATGATC
TCCTTGAATGCGCCCCC
+2 A to G
10
21
19



CCAAGTGG
ACCTGGGATCATAG









OsALS-T2-pegRT23
GTGCTGCCTATGATC
TGTCCTTGAATGCGCCC
+2 A to G
10
23
21



CCAAGTGG
CCACCTGGGATCATAG









OsCDC48-T1-peg0l
GCTAGCTTTGACAT
ATTAATGGCATTATGTC
+1-6 CTCCGG
12
 9
 9



AATCTCCGG
AAAG
del








OsCDC48-T1-peg02
GCTAGCTTTGACAT
ATTAATGGCCTTAAGAT
+3-5 AG to CA
12
15
10



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-peg03
GCTAGCTTTGACAT
ATTAATGGCCCGGATCC
+2 GGA ins
12
18
11



AATCTCCGG
GATTATGTCAAAG









OsCDC48-T1-pegAT
GCTAGCTTTGACAT
TTCTTCTTTCTGATTAA
+10 Ato T
12
27
17



AATCTCCGG
AGGCCCGGAGATTATGT








CAAAG









OsCDC48-T1-pegAC
GCTAGCTTTGACAT
TTCTTCTTTCTGATTAA
+10 Ato C
12
27
17



AATCTCCGG
GGGCCCGGAGATTATGT








CAAAG









OsCDC48-T1-pegAG
GCTAGCTTTGACAT
TTCTTCTTTCTGATTAA
+10 Ato G
12
27
17



AATCTCCGG
CGGCCCGGAGATTATGT








CAAAG









OsCDC48-T1-pegCA
GCTAGCTTTGACAT
ATTAATGGCCCGGATAT
+1 C to A
12
15
14



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegCT
GCTAGCTTTGACAT
ATTAATGGCCCGGAAAT
+1 C to T
12
15
14



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegCG
GCTAGCTTTGACAT
ATTAATGGCCCGGACAT
+1 C to G
12
15
14



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegGA
GCTAGCTTTGACAT
ATTAATGGCCTGGAGAT
+5 G to A
12
15
10



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegGC
GCTAGCTTTGACAT
ATTAATGGCCGGGAGAT
+5 G to C
12
15
10



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegGT
GCTAGCTTTGACAT
ATTAATGGCCAGGAGAT
+5 G to T
12
15
10



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegTA
GCTAGCTTTGACAT
ATTAATGGCCCGGTGAT
+2 T to A
12
15
13



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegTC
GCTAGCTTTGACAT
ATTAATGGCCCGGGGAT
+2 T to C
12
15
13



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegTG
GCTAGCTTTGACAT
ATTAATGGCCCGGCGAT
+2 T to G
12
15
13



AATCTCCGG
TATGTCAAAG









OsCDC48-T1-pegPBS8
GCTAGCTTTGACAT
ATTAATGGCATTATGTC
+1-6 CTCCGG
 8
 9
 9



AATCTCCGG

del








OsCDC48-T1-pegPBS10
GCTAGCTTTGACAT
ATTAATGGCATTATGTC
+1-6 CTCCGG
10
 9
 9



AATCTCCGG
AA
del








OsCDC48-T1-pegPBS14
GCTAGCTTTGACAT
ATTAATGGCATTATGTC
+1-6 CTCCGG
14
 9
 9



AATCTCCGG
AAAGCT
del








OsCDC48-T1-pegPBS16
GCTAGCTTTGACAT
ATTAATGGCATTATGTC
+1-6 CTCCGG
16
 9
 9



AATCTCCGG
AAAGCTAG
del








OsCDC48-T1-pegRT7
GCTAGCTTTGACAT
TAATGGCATTATGTCAA
+1-6 CTCCGG
 8
 7
 7



AATCTCCGG
AG
del








OsCDC48-T1-pegRT11
GCTAGCTTTGACAT
TGATTAATGGCATTATG
+1-6 CTCCGG
 8
11
11



AATCTCCGG
TCAAAG
del








OsCDC48-T1-pegRT13
GCTAGCTTTGACAT
TCTGATTAATGGCATTAT
+1-6 CTCCGG
 8
13
13



AATCTCCGG
GTCAAAG
del








OsCDC48-T1-pegRT15
GCTAGCTTTGACAT
TTTCTGATTAATGGCATT
+1-6 CTCCGG
 8
15
15



AATCTCCGG
ATGTCAAAG
del








OsCDC48-T1-pegins3
GCTAGCTTTGACAT
ATTAATGGCCCGGAGA
+1 GGT ins
12
18
16



AATCTCCGG
CCATTATGTCAAAG









OsCDC48-T1-pegins6
GCTAGCTTTGACAT
ATTAATGGCCCGGAGA
+1 GTTGGT
12
21
16



AATCTCCGG
CCAACATTATGTCAAAG
ins








OsCDC48-T1-pegins15
GCTAGCTTTGACAT
ATTAATGGCCCGGAGA
+1 15 nt ins
12
30
16



AATCTCCGG
CCAACCTGTCCGACATT








ATGTCAAAG









OsCDC48-T1-pegins30
GCTAGCTTTGACAT
ATTAATGGCCCGGAGA
+1 30 nt ins
12
45
16



AATCTCCGG
CCAACCTGTCCGACATC








ATCGAGAAGGAGATTAT








GTCAAAG









OsCDC48-T1-pegins60
GCTAGCTTTGACAT
ATTAATGGCCCGGAGA
+1 60 nt ins
12
75
16



AATCTCCGG
CCAACCTGTCCGACATC








ATCGAGAAGGAGACGG








GCAAGCAACTCGTGAT








CCAGGAGAGCATTATGT








CAAAG









OsCDC48-T1-pegde112
GCTAGCTTTGACAT
TTTCTGATTATTATGTCA
+1 12-nt del
12
9
9



AATCTCCGG
AAG









OsCDC48-T1-pegde120
GCTAGCTTTGACAT
GCTTTCTTCTATTATGTC
+1 20-nt del
12
9
9



AATCTCCGG
AAAG









OsCDC48-T1-pegde140
GCTAGCTTTGACAT
TGTTGCTAAATTATGTC
+1 40-nt del
12
9
9



AATCTCCGG
AAAG









OsCDC48-T1-pegde170
GCTAGCTTTGACAT
TGGCTCTGGATTATGTC
+1 70-nt del
12
9
9



AATCTCCGG
AAAG









OsCDC48-T2-peg01
GACCAGCCAGCGTC
GGCGCCGGCTTTGCCA
+2 AAA ins
12
13
9



TGGCGCCGG
GACGCTGGC









OsCDC48-T2-peg02
GACCAGCCAGCGTC
TCAGCGGCGGCGCTGG
+5 G to A
12
18
13



TGGCGCCGG
CGCCAGACGCTGGC









OsCDC48-T2-pegGT
GACCAGCCAGCGTC
TCAGCGGCGGCGCAGG
+5 G to T
12
18
13



TGGCGCCGG
CGCCAGACGCTGGC









OsCDC48-T2-pegGC
GACCAGCCAGCGTC
TCAGCGGCGGCGCGGG
+5 G to C
12
18
13



TGGCGCCGG
CGCCAGACGCTGGC









OsCDC48-T2-pegCA
GACCAGCCAGCGTC
TCAGCGGCGGCGCCTG
+4 C to A
12
18
14



TGGCGCCGG
CGCCAGACGCTGGC









OsCDC48-T2-pegCT
GACCAGCCAGCGTC
TCAGCGGCGGCGCCAG
+4 C to T
12
18
14



TGGCGCCGG
CGCCAGACGCTGGC









OsCDC48-T2-pegDel
GACCAGCCAGCGTC
TCAGCGGCGGCGGCGC
+4-6 CGG del
12
15
10



TGGCGCCGG
CAGACGCTGGC









OsCDC48-T2-pegPBS8
GACCAGCCAGCGTC
GGCGCCGGCTTTGCCA
+2 AAA ins
 8
13
 9



TGGCGCCGG
GACGC









OsCDC48-T2-pegPBS10
GACCAGCCAGCGTC
GGCGCCGGCTTTGCCA
+2 AAA ins
10
13
 9



TGGCGCCGG
GACGCTG









OsCDC48-T2-pegPBS14
GACCAGCCAGCGTC
GGCGCCGGCTTTGCCA
+2 AAA ins
14
13
 9



TGGCGCCGG
GACGCTGGCTG









OsCDC48-T2-pegPBS16
GACCAGCCAGCGTC
GGCGCCGGCTTTGCCA
+2 AAA ins
16
13
 9



TGGCGCCGG
GACGCTGGCTGGT









OsCDC48-T2-pegRT10
GACCAGCCAGCGTC
GCCGGCTTTGCCAGAC
+2 AAA ins
12
10
 6



TGGCGCCGG
GCTGGC









OsCDC48-T2-pegRT11
GACCAGCCAGCGTC
CGCCGGCTTTGCCAGA
+2 AAA ins
12
11
 7



TGGCGCCGG
CGCTGGC









OsCDC48-T2-pegRT15
GACCAGCCAGCGTC
GCGGCGCCGGCTTTGC
+2 AAA ins
12
15
11



TGGCGCCGG
CAGACGCTGGC









OsCDC48-T2-pegRT16
GACCAGCCAGCGTC
GGCGGCGCCGGCTTTG
+2 AAA ins
12
16
12



TGGCGCCGG
CCAGACGCTGGC









OsCDC48-T2-pegRT17
GACCAGCCAGCGTC
CGGCGGCGCCGGCTTT
+2 AAA ins
12
17
13



TGGCGCCGG
GCCAGACGCTGGC









OsCDC48-T2-pegRT19
GACCAGCCAGCGTC
AGCGGCGGCGCCGGCT
+2 AAA ins
12
19
15



TGGCGCCGG
TTGCCAGACGCTGGC









OsCDC48-T2-pegRT21
GACCAGCCAGCGTC
TCAGCGGCGGCGCCGG
+2 AAA ins
12
21
17



TGGCGCCGG
CTTTGCCAGACGCTGG








C









OsCDC48-T3-peg01
TAGCACCCATGACA
AGCTCGTTCCCGGGTCA
+2-4 ATG del
12
15
11



ATGACATGG
TTGTCATGGG









OsDEPl-peg0l
GTGGGTTTGTTTCTA
ATTTCCTCCTTAAGCTA
+2 C to T
13
13
11



GCTCAAGG
GAAACAAAC









OsDEPl-peg02
GTGGGTTTGTTTCTA
ATTTCCTCCTCGAGCTA
+3 A to G
13
13
10



GCTCAAGG
GAAACAAAC









OsEPSPS-T1-peg01
TACTAAATATACAAT
ATAAACCCATGGGATTG
+2 T to A
13
11
 9



CCCTTGGG
TATATTT









OsEPSPS-T1-peg02
TACTAAATATACAAT
TGGAAACAAATAAACA
+5 G to T
13
20
16



CCCTTGGG
CAAGGGATTGTATATTT









OsEPSPS-T2-peg01
GAAAATATGTATGG
ATAAAACATACCCCATT
+2 C to A
13
17
15



AATTCATGG
AATTCCATACATAT









OsGAPDH-peg01
GAGTATGTCGTGGA
AGTGAAGACACCGTTG
+3 C to A
12
16
13



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegCT
GAGTATGTCGTGGA
AGTGAAGACACCGATG
+3 C to T
12
16
13



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegCG
GAGTATGTCGTGGA
AGTGAAGACACCGCTG
+3 C to G
12
16
13



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegAT
GAGTATGTCGTGGA
AGTGAAGACACCGGAG
+2 A to T
12
16
14



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegAC
GAGTATGTCGTGGA
AGTGAAGACACCGGGG
+2 A to C
12
16
14



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegAG
GAGTATGTCGTGGA
AGTGAAGACACCGGCG
+2 A to G
12
16
14



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegGA
GAGTATGTCGTGGA
AGTGAAGACACTGGTG
+5 G to A
12
16
11



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegGT
GAGTATGTCGTGGA
AGTGAAGACACAGGTG
+5 G to T
12
16
11



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegGC
GAGTATGTCGTGGA
AGTGAAGACACAGGTG
+5 G to C
12
16
11



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegTA
GAGTATGTCGTGGA
AGTGAAGACTCCGGTG
+7 T to A
12
16
 9



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegTC
GAGTATGTCGTGGA
AGTGAAGACGCCGGTG
+7 T to C
12
16
 9



GTCCACCGG
GACTCCACGACA









OsGAPDH-pegTG
GAGTATGTCGTGGA
AGTGAAGACCCCGGTG
+7 T to G
12
16
 9



GTCCACCGG
GACTCCACGACA









OsLDMAR-peg01
GTGCATCCTGGGAA
AAACTTGCTACCACTAG
+2 T to A
12
15
14



AGCTTGTGG
CTTTCCCAGGA









TaGW2-peg01
CACAAGAAAATCCA
ATGCCCCATCGTGGTGG
+1 G to C
11
11
10



CCAGGATGG
ATTTT









TaUbi10-T1-peg01
GCGTATGTATGAAA
AGAAAAGGCTCCAGTA
+4 A to T
13
16
12



CTGTACAGG
CAGTTTCATACAT









TaUbi10-T2-peg01
GCTGGTGCAACTGG
AGCCCTCCTGGCCCAC
+1 C to G
12
12
11



TGGCCCAGG
CAGTTGCA









TaLOX2-peg01
GTGCCGCGCGACGA
TGAGGTGGCCGAACAG
+1 C to G
12
14
13



GCTCTTCGG
CTCGTCGCGC









TaMLO-peg01
GGAGATTGGGTCCT
TGCTCGCCGTCCCGCA
+1 T to G
12
12
11



GCGTGACGG
GGACCCAA









TaDME- peg01
TTTGATGGTTTCAG
AGCACAGGCCAAATGT
+1 C to A
13
14
13



GACCTTTGG
CCTGAAACCAT









TaGASR7-peg01
GCCGGGCACCTACG
AGGGGCAGGCGCGCTT
+6 G to C
12
18
12



GCAACAAGG
GTTGCCGTAGGTGC





The PAM sequence is shown in bold.






2. Protoplast Isolation and Transformation

The protoplasts used in the present invention were derived from the rice variety Zhonghua 11 and the wheat variety Kenong 199.


2.1 Cultivation of Rice Seedlings


Seeds were first rinsed with 75% ethanol for 1 min, then treated with 4% sodium hypochlorite for 30 min, and washed with sterile water for more than 5 times. The treated seeds were cultured on a M6 culture medium for 3-4 weeks at 26° C., and protected from light.


2.2 Isolation of Rice Protoplasts


(1) Rice stalks were cut off and the middle parts thereof were cut into 0.5-1 mm filaments with a blade. The filaments were treated with a 0.6 M mannitol solution for 10 min in the dark, filtered with a filter, put into 50 mL an enzymolysis solution (filtered with a 0.45 μm membrane), vacuumized (pressure intensity of about 15 Kpa) for 30 min, taken out and placed on a shaker (10 rpm) for enzymolysis at room temperature for 5 h.


(2) The enzymolysis product was diluted by adding 30-50 mL W5 and filtered with a 75 μm nylon filter membrane into a round-bottomed centrifuge tube (50 mL).


(3) Centrifugation was conducted at 23° C. for 3 min at 250 g (rcf) prior to discarding the supernatant.


(4) The cells were slightly suspended with 20 mL W5, and step (3) was repeated.


(5) An appropriate amount of MMG was added to suspend the cells for transformation.


2.3 Transformation of Rice Protoplasts


(1) 10 μg vector to be transformed was added to each 2 mL centrifuge tube and mixed well. 200 μL protoplasts were taken with a tipped pipette and mixed well by flicking. 220 μL PEG4000 solution was added and mixed well by flicking, followed by inducing transformation for 20-30 min at room temperature in the absence of light.


(2) 880 μL W5 was added and mixed well by slight inverting, and centrifugation was conducted at 250 g (rcf) for 3 min prior to discarding the supernatant.


(3) 1 mL WI solution was added and mixed well by gently inverting before being gently transferred to a flow tube and cultured in the dark at 26° C. for 48 h. Protoplasts to be treated at 37° C. should be transferred to the flow tube for culturing in the dark at 26° C. for 12 h, then transferred to a dark place at 37° C. for 8 h, and finally transferred to a place at 26° C. for culturing for 48 h.


2.4 Cultivation of Wheat Seedlings


Potted wheat seeds were planted in a culture room for culturing (about 10 days) at 25±2° C. with a light intensity of 1000 Lx and illumination time of 14-16 h/d for about 1-2 weeks.


2.5 Isolation of Wheat Protoplasts


(1) The young leaves of wheat were obtained and the middle parts thereof were cut into 0.5-1 mm filaments with a blade. The filaments were treated with a 0.6 M mannitol solution for 10 min in the dark, filtered with a filter screen, put into 50 mL enzymolysis solution (filtered with a 0.45 μm filter membrane), vacuumized (pressure intensity of about 15 Kpa) for 30 min, taken out and placed on a shaker (10 rpm) for enzymolysis at room temperature for 5 h.


(2) The enzymolysis product was diluted by adding 30-50 mL W5 and filtered with 75 μm nylon filter membrane into a round-bottomed centrifuge tube (50 mL).


(3) Centrifugation was conducted at 23° C. for 3 min at 100 g (rcf) prior to discarding the supernatant. (4) The cells were slightly suspended with 10 mL W5 and placed on ice for 30 min. The protoplasts were gradually settled, and the supernatant was discarded. (5) The resulting product was suspended with an appropriate amount of MMG and placed on ice until transformation.


2.6 Transformation of Wheat Protoplasts


(1) 10 μg plasmids to be transformed were added into each 2 mL centrifuge tube, and mixed well.


(2) 200 μL protoplasts were sucked with a tipped pipette and mixed well by flicking. 250 μL PEG4000 solution was added immediately and mixed well by flicking, followed by inducing transformation for 20-30 min at room temperature in the absence of light.


(3) 800 μL W5 (room temperature) was added and mixed well by slight inverting, and centrifugation was conducted at 100 g (rcf) for 3 min prior to discarding the supernatant.


(4) 1 mL W5 was added, mixed well by slight inverting and gently transferred to a 6-well plate wrapped with foil in which 1 mL W5 has been added. The cells were incubated in the dark at 26° C. for 48 h.


3. Observation of Cell Fluorescence by Flow Cytometer

The FACSAria III (BD Biosciences) instrument for the flow cytometry analysis of protoplasts has the following specific operation steps:


(1) After the instrument was turned on, the BD FACSDiva Software was started for the operation such as instrument calibration.


(2) “New Protocol” was clicked to create a suitable experimental protocol.


(3) “Density plot” was selected to draw a FSC/SSC scatter diagram and a GFP/PE-Texas Red scatter diagram.


(4) The FSC/SSC voltage was adjusted to make the cell population appear in the center of the scatter diagram, the FL1 voltage was adjusted to make the wild-type control protoplast population appear in the center of the scatter diagram, and the GFP-positive protoplast population will appear in the location where the GFP fluorescence channel signal is higher (there is no such population in the wild-type control sample). If necessary, compensated regulation was made, so that the GFP-negative and GFP-positive protoplast populations are more distinct.


(5) A gate was set to circle the GFP-positive population. A protoplast-negative control needs to be prepared to determine the boundary of the gate.


(6) The cell population to be sorted was clicked by the right button, the “left sort” was selected, and the analysis condition and the analysis mode were set according to the experimental needs and the percentage of target cells.


(7) The prepared protoplast samples cultured in the flow tube were sampled sequentially, and relevant data was recorded for analysis.


(8) Both the software and the instrument were shut down.


4. Protoplast DNA Extraction and Amplicon Sequencing Analysis 3.1 Protoplast DNA Extraction

The protoplasts were collected in a 2 mL centrifuge tube, and protoplast DNA (−30 μL) with a concentration (30-60 ng/μL) measured by a NanoDrop ultra-micro spectrophotometer was extracted by a CTAB method and stored at −20° C.


3.2 Amplicon Sequencing Analysis


(1) PCR amplification was conducted on a protoplast DNA template using genomic primers. A 20 μL amplification system comprised 4 μL 5× Fastpfu buffer, 1.6 μL dNTPs (2.5 mM), 0.4 μL Forward primer (10 μM), 0.4 μL Reverse primer (10 μM), 0.4 μL FastPfu polymerase (2.5 U/μL), and 2 μL DNA template (−60 ng). The amplification condition was as followings: pre-denaturation at 95° C. for 5 min; denaturation at 95° C. for 30 s, annealing at 50-64° C. for 30 s, extension at 72° C. for 30 s, for 35 cycles; fully extending at 72° C. for 5 min, and storing at 12° C.


(2) The above amplification product was diluted by 10 times, 1 μL amplification product was taken as a second PCR amplification template, and the amplification primers were sequencing primers containing Barcode. A 50 μL amplification system comprised 10 μL 5× Fastpfu buffer, 4 μL dNTPs (2.5 mM), 1 μL Forward primer (10 μM), 1 μL Reverse primer (10 μM), 1 μL FastPfu polymerase (2.5U/μL), and 1 μL DNA template. The amplification condition was the same as that mentioned above, and the number of amplification cycles was 35 cycles.


(3) The PCR product was separated by 2% agarose gel electrophoresis, and gel extraction was conducted on a target fragment using the AxyPrep DNA Gel Extraction kit, and the resulting product was quantitatively analyzed using a NanoDrop ultra-micro spectrophotometer. The 100 ng recovered product was taken, mixed and sent to the Sangon Bioengineering Co., Ltd. for amplicon sequencing library construction and amplicon sequencing analysis.


(4) After sequencing was completed, the original data were split according to the sequencing primers, and WT was used as a control to compare and analyze the editing type and editing efficiency of the products at different genetic target loci in three repeated trials.


Example 1 BFP-to-GFP Reporter System in Rice Protoplasts Precisely Modified by Cas9 (H840A) Nickase-Reverse Transcriptase Fusion System

In order to test whether Cas9 (H840A) nickase-reverse transcriptase fusion (PPEs, plant prime editors) can be used to precisely modify a target sequence (FIGS. 1-2) or not, an nCas9(H840A)-M-MLV construct (PPE-M-MLV), an nCas9(H840A)-CaMV construct (PPE-CaMV), an nCas9(H840A)-retron (PPE-retron) construct, an Os U3/Ta U6-promoter-driven pegRNA construct with target guide RNA and RT and PBS sequences, and a TaU3-promoter-driven nicking gRNA construct that nicks a non-target strand (FIG. 3) were constructed. The performance of PPEs in the protoplasts was observed by the BFP-to-GFP reporter system (FIG. 4). When the “CC” in the BFP gene sequence was changed into “GT”, the 66th amino acid can be changed from histidine (H, Histidine) to tyrosine (Y, Tyrosine), so that the gene encods GFP fluorescent protein and the cells emited green fluorescence. The efficiency of PPEs can then be analyzed by a flow cytometry. The results show that PPE lacking a reverse transcriptase (i.e., PPE3b (AM-MLU)) failed to make the cells emit green fluorescence, while green fluorescence can be clearly observed in the cells after co-transforming the PPE-M-MLV, the pegRNA, the nicking gRNA and the BFP-to-GFP reporter system (i.e., PPE3b) into the protoplasts, and the average efficiency is 4.4% (FIGS. 5-6). Replacing the M-MLV reverse transcriptase in this system with a reverse transcriptase (i.e., PPE3b-CaMV) derived from CaMV viruses or a reverse transcriptase (i.e., PPE3b-retron) derived from a bacterial retron system can also make the cells emit green fluorescence, and the efficiency was 3.7% and 2.4%, respectively (FIGS. 5-6). Therefore, the above results demonstrate the ability of PPEs in plants to allow desired specified modifications of the target sequence of the reporter system, which preliminarily demonstrates that PPEs can work in plants. PPE may comprise reverse transcriptase of other forms and sources.


Example 2 Genome Sequences in Rice and Wheat Protoplasts Precisely Modified by Cas9(H840A) Nickase-Reverse Transcriptase Fusion System

In order to test whether the PPE system can work on endogenous loci in rice or not, 21 pegRNAs were designed and constructed for 10 endogenous loci in rice (OsCDC48-T1, OsCDC48-T2, OsCDC48-T3, OsALS-T1, OsALS-T2, OsDEP1, OsEPSPS-T1, OsEPSPS-T2, OsLDAMR and OsGAPDH) and 7 endogenous loci in wheat (TaUbi10-T1, TaUbi10-T2, TaGW2, TaGASR7, TaLOX2, TaMLO and TaDME) to test whether PPE2 and PPE3 (or PPE3b) can work on these endogenous loci or not. The results indicate that both the PPE2 and PPE3 (or PPE3b) systems can achieve specific single-base changes including C-to-T, G-to-T, A-to-G, G-to-A, T-to-A and C-to-A on the endogenous loci in rice, as well as addition or deletion of specific bases, and the system has the highest efficiency of 8.2% in rice (FIG. 7). The PPE2 and PPE3 (or PPE3b) systems can also achieve single-base changes including A to T, C to G, G to C, T to G and C to A on the endogenous loci in wheat, and the highest efficiency is 1.4% (FIG. 8). The overall efficiency of PPE2 and PPE3 (or PPE3b) did not differ significantly on the loci tested. In addition, a proportion of by-products were also observed in the PPE system, mainly including insertion or substitution of the pegRNA skeleton (FIG. 9). The above results indicate that the PPE systems including PPE2, PPE3 and PPE3b can achieve targeted modification including precise single-base mutation, precise insertion and precise deletion on the endogenous loci in plants.


Example 3 Improvement of Cas9 (H840A) Nickase-Reverse Transcriptase Fusion System

In order to further improve the PPE editing system, the performance of the PPE-CaMV system on the endogenous loci in rice was tested. The results showed that PPE-CaMV can also achieve precise targeted modification of endogenous sequences in rice, with the highest efficiency of 5.8% (FIG. 10), and indicated that PPE systems derived from other species or other forms of reverse transcriptases also can modify endogenous genes in plants in a targeted manner.


In addition, the performance of a ribozyme-processed pegRNA in the PPE system was also tested. An Ubi-1-driven ribozyme-processed pegRNA construct with target guide RNA and RT and PBS sequences was constructed to replace an OsU3-driven pegRNA construct in the original system, and this system was named as PPE-R (R represents Ribozyme) (FIG. 11). The results of endogenous targets showed that the strategy of using ribozyme processing can also achieve precise change of the endogenous sequences. PPE-R was improved compared with PPE on some loci, and the highest efficiency can be 9.7% (FIG. 12). This result suggests that both the ribozyme-processed pegRNA and using type II promoters to drive the transcription of pegRNA are suitable for use in the PPE system.


In order to improve the editing efficiency of the PPE system, the protoplasts were cultured at 37° C. to test whether it can improve the editing efficiency or not. 2 endogenous loci (OsCDC48-T2 and OsALS-T2) in rice were selected for testing. The transformed protoplasts were cultured at 26° C. overnight, incubated at 37° C. for 8 h and cultured at 26° C. again, and then compared with the treatment group in the aspect of efficiency. The results showed that treatment at 37° C. can significantly improve the editing efficiency of the PPE systems (including PPE2, PPE3 and PPE3b), the average efficiency was increased by 1.6 times (from 3.9% to 6.3%), and the highest editing efficiency was increased by 2.9 times (FIG. 13).


Example 4 Influences of Different Lengths of PBS and RT Template and Different Nicking gRNA Positions on the PPE System

The influences of different lengths of PBS and RT templates and different nicking gRNA positions on the PPE system were tested. The results using OsCDC48-T1 as a test locus showed that different lengths of PBS (6-16nt) and RT templates (7-23nt) tested can lead to a targeted sequence modification on the specific loci (FIGS. 14-15), and the efficiency is 3.4% to 15.3% on the OsCDC48-T1 locus, 0.9% to 8.1% on the OsCDC48-T2 locus, and 1.1% to 10.5% on the OsALS-T2 locus. The results showed that different lengths of RT templates and PBS have a significant effect on the editing efficiency, and it was found that RT templates of different lengths have a relatively significant effect on the proportion and type of by-products produced in the PPE system (FIG. 16). In addition, different nicking gRNAs also affected the efficiency of the PPE system, and the efficiency is 3.2% to 19.2% on the OsCDC48-T1 locus and 2.9% to 8.6% on the OsCDC48-T2 locus (FIG. 17).


Example 5 Various Types of Precise Modifications of Endogenous Loci Achieved by PPE System

In order to test whether the PPE systems can achieve various types of precise modifications or not, 12 types of single-base changes (N to N, wherein N represents four base types A, T, C or G), multiple bases change, and insertion and deletion of bases of different lengths were designed using 4 loci (OsCDC48-T1, OsCDC48-T2, OsALS-T2 and OsGAPDH) in rice as examples. The results showed that the PPE system can achieve all types of single-base changes with the highest efficiency of 8.0%. The system can also achieve simultaneous change of multiple bases, with the highest efficiency of 1.5% (FIG. 18). The efficiency of base insertion and deletion is decreased with the increase of the length needed to be modified, and the highest targeted insertion efficiency can be 3.0%, and the longest insertion length can be 15 nt (FIG. 19). The highest targeted deletion efficiency can be 19.2%, and the longest deletion length can be 40 nt (FIG. 20). Therefore, the system can efficiently achieve addition and deletion of small segments. Therefore, the PPE system can enable various types of targeted modification of endogenous loci.


Example 6 Targeted Edited Plants Obtained by PPE System

In order to obtain targeted edited plants, OsCDC48-T1 and OsALS-T2 loci were selected for testing, a binary vector PPE3 was constructed (FIG. 21), and the rice was infected by agrobacterium-mediated transformation. Finally, 12 rice seedlings with the expected precise deletion on the OsCDC48-T1 locus with an editing efficiency of 21.8% (12/55), 2 rice seedlings with the expected single-base mutation of G to T on the OsALS-T2 locus with an editing efficiency of 14.3% (2/14), and 1 rice seedling with the expected precise simultaneous mutation of 3 pairs of bases on the OsCDC48-T1 locus with an editing efficiency of 2.6% (1/38) (FIG. 22) (Table 2) were detected. It was also found that a very small amount of unintended by-products were produced in plants (FIG. 23). The above results indicated that the PPE system can obtain mutant plants with targeted modifications therein.









TABLE 2







Targeted edited rice obtained by PPE system





















Editing







Expected

efficiency






Nicking
number
Number of
(%) of




PBS
RT
gRNA
of edited
transgenic
the PPE


Target locus
Target type
length
length
position
plants
plants
system

















OsCDC48-T1
+1-6
12
 9
+37
12
55
21.8 



CTCCGG del








OsALS-T2
+3 G to T
10
17
+33
 2
14
14.3 


OsCDC48-T1
+3-5 AG to CA
12
15
+76
 1
38
2.6









Example 7 Influence of Tm of PBS on the Efficiency of PPE System

The influence of the PBS length which controls the Tm on prime editing in plants was first assessed based on published data (FIG. 24). The results show that the editing efficiency is very high when PBS Tm was close to 30° C. (30° C. for OsCDC48-T1, 28° C. for OsCDC48-T2 and 30° C. for OsALS-T1).


Then, the editing efficiency of PPE using PBS (corresponding to the PBS length of 6 nt to 17 nt) with a Tm of 18° C. to 52° C. was evaluated on the four target loci (OsACC-T1, OsCDC48-T3, OsEPSPS-T1 and OsPDS-T1) in rice protoplasts. The results are shown in FIG. 24. When PBS Tm was close to 30° C. (24-30° C. in OsACC-T1, 26-34° C. in OsEPSPS-T1, 28-36° C. in OsCDC48-T3, and 30° C. in OsPDS-T1), pegRNA had higher activity. The editing efficiency at these PBS Tm was 1.5 to 4.3 times higher than that at other PBS Tm. Additional six target loci were also tested (see FIG. 24), including OsALS-T2, OsDEP1-T1, OsEPSPS-T2, OsAAT-T1, OsGAPDH-T1 and OsLDMAR-T1, and it was found that five (except for OsEPSPS-T2) of the six target loci have similar conditions.


Then, the overall editing efficiency of PPE on the 13 target loci at different PBS Tm was normalized and compared. The results show that the editing efficiency follows a normal distribution (P>0.1) (FIG. 25), typically reaches a maximum value when PBS Tm was 30° C., followed by 32° C. and 28° C., and the efficiency was decreased with increase or decrease of PBS Tm.


From this, it was concluded that the melting temperature of the PBS sequence is closely related to the editing efficiency of the PPE (FIG. 25) and is likely a major factor influencing the pegRNA design for plants. It is recommended to use PBS Tm of 30° C. to guide the design of pegRNA in PPE.


Example 8 Dual pegRNA Strategy to Significantly Improve the Efficiency of PPE System

In order to obtain optimal editing, the inventors developed a dual-pegRNA strategy which uses different pegRNAs against forward and reverse DNA strands (referred to as NGG-pegRNA and CCN-pegRNA, respectively) and simultaneously encoding a same edit (FIG. 26). 15 target loci were selected from ten rice genes, and a pair of pegRNAs were designed for each target (Table 3). Then, the editing activities of NGG-pegRNA, CCN-pegRNA and dual-pegRNA were compared at the same location.









TABLE 3







Target sequence, RT template and PBS sequence of double-pegRNA























Homo-










logous







Edited
PBS
RT
arm 





RT template and
Desired
DNA
length
length
length


Genes
pegRNA
Target sequence
PBS sequence
editing
strands
(nt)
(nt)
(nt)





OsCDC48
NGG-pegRNA
GACCAGCCAGCGT
TCAGCGGCGGCGCCTG
+4 C to A
Forward
12
18
14




CTGGCGCCGG
CGCCAGACGCTGGC










OsCDC48
CCN-pegRNA

CCGGCGCCAGACG

CAGCGTCTGGCGCAGG
+5 G to T
Reverse
12
18
13




CTGGCTGGTC
CGCCGCCGCTGACC










OsCDC48
NGG-pegRNA
GACCAGCCAGCGT
TCAGCGGCGGCGCTGG
+5 G to A
Forward
12
18
13




CTGGCGCCGG
CGCCAGACGCTGGC










OsCDC48
CCN-pegRNA

CCGGCGCCAGACG

CAGCGTCTGGCGCCAG
+4 C to T
Reverse
12
18
13




CTGGCTGGTC
CGCCGCCGCTGACC










OsALS
NGG-pegRNA
GTGCTGCCTATGAT
TGAATGCGCCCCCAATT
+3 G to T
Forward
10
17
14




CCCAAGTGG

GGGATCATAG











OsALS
CCN-pegRNA

CCAAGTGGGGGCG

TGCCTATGATCCCAATT
+2 C to A
Reverse
10
17
15




CATTCAAGGA

GGGGGCGCAT











OsALS
NGG-pegRNA
GTGCTGCCTATGAT
TGAATGCGCCCCCACC
+2 A to G
Forward
10
17
15




CCCAAGTGG
TGGGATCATAG










OsALS
CCN-pegRNA

CCAAGTGGGGGCG

TGCCTATGATCCCAGGT
+3 T to C
Reverse
10
17
14




CATTCAAGGA

GGGGGCGCAT











OsEPSPS
NGG-pegRNA
TACTAAATATACAA
ATAAACCCATGGGATTG
+2 T to A
Forward
13
11
 9




TCCCTTGGG

TATATTT











OsEPSPS
CCN-pegRNA

CCCTTGGGTTTATT

ACAATCCCATGGGTTTA
+3 A to T
Reverse
13
11
 8




TGTTTCCAA

TTTGTTT











OsEPSPS
NGG-pegRNA
ACTGTGACAGTTC
TGCAAACTGGTAGTAC
+10 G to I
Forward
10
21
11




AAGGTTGTGG
CACAACCTTGAACTG










OsEPSPS
CCN-pegRNA

CCAGTTTGCAGGT

AAGGTTGTGGTACTAC
+8 C to A 
Reverse
11
21
13




ATAACTGTAG
CAGTTTGCAGGTATAA










OsIPA1
NGG-pegRNA
CTCTTCTGTCAACC
AGTATCCCATGCCTGGG
+1 C to G
Forward
10
12
11




CAGCCATGG

TTGAC











OsIPA1
CCN-pegRNA

CCCAGCCATGGGA

TGTCAACCCAGGCATG
+1 G to C
Reverse
10
12
11




TACTACTACC

GGATAC











OsIPA1
NGG-pegRNA
TCTTCTGTCAACCC
GTAGTATCGCATGGCTG
+5 G to C
Forward
15
13
 8




AGCCATTGG

GGTTGACAGAA











OsIPA1
CCN-pegRNA

CCATGGGATACTA

AACCCAGCCATGCGAT
+2 C to G
Reverse
15
13
11




CTACCCACAG

ACTACTACCCAC











OsROC5
NGG-pegRNA
TCTCACCTCTAGTA
TGTAACCTCCCTATGGT
+1-2 AT 
Forward
14
13
13




CCAATTAGG

ACTAGAGGTG

del









OsROC5
CCN-pegRNA

CCAATTAGGGAGG

CCTCTAGTACCATAGGG
+2-3 TA 
Reverse
14
13
13




TTACATTTCT

AGGTTACATT

del









OsDEP1
NGG-pegRNA
GAGACAAGCTTGG
CGCCACGCCCAAGAGG
+2 T del
Forward
12
13
10




CCCTCTTTGG

GCCAAGCTT











OsDEP1
CCN-pegRNA

CCTCTTTGGGCGT

AGCTTGGCCCTCTTGG
+2 A del
Reverse
12
13
10




GGCGCCATGG

GCGTGGCGC











OsDEP1
NGG-pegRNA
AGCTTGGCCCTCTT
ACAGCCATGGCTCCAC
+7 C to A
Forward
10
18
11




TGGGCGTGG
GCCCAAAGAGGG










OsDEP1
CCN-pegRNA

CCATGGCTGTACT

TTGGGCGTGGAGCCAT
+8 G to T
Reverse
10
18
10




ACCTTTGTCG
GGCTGTACTACC










OsNRT1.1B
NGG-pegRNA
ACTAGATATCTAAA
ATTTGACCTACCTTTAA
+2 A ins
Forward
13
17
14




CCATTAAGG

TGGTTTAGATATC











OsNRT1.1B
CCN-pegRNA

CCATTAAGGTAGG

GATATCTAAACCATTAA
+2 T ins
Reverse
13
17
14




TCAAATCAAG

AGGTAGGTCAAAT











OsNRT1.1B
NGG-pegRNA
CCTGTAGGTGTGTA
TAGTGCTGGTTCAAGTA
+6 G to A
Forward
11
16
10




TATACTTGG

TATACACACC











OsNRT1.1B
CCN-pegRNA

CCAGCACTACTAG

TATACTTGAACCAGCAC
+8 C to T
Reverse
11
16
 8




ATATCTAAAC

TACTAGATAT











OsODEV
NGG-pegRNA
CACACACACACTA
AATAGTCCCCTAGGTAC
+3-4 CT to 
Forward
12
13
 9




GTACCTCTGG

TAGTGTGT

AG









OsODEV
CCN-pegRNA

CCTCTGGGACTATT

ACTAGTACCTAGGGGA
+2-3 GA to 
Reverse
12
13
 9




GTGTGTGTC

CTATTGTGT

TC









OsGAPDH
NGG-pegRNA
GAGTATGTCGTGG
AGTGAAGACACCGCTG
+3 C to G
Forward
12
16
13




AGTCCACCGG

GACTCCACGACA











OsGAPDH
CCN-pegRNA

CCACCGGTGTCTT

GTCGTGGAGTCCAGCG
+3 G to C
Reverse
12
16
13




CACTGACAAG

GTGTCTTCACTG






PAM for each target locus is shown in bold, and PBS is underlined.






PAM for each target locus is shown in bold, and PBS is underlined.


As expected, the dual-pegRNA strategy has the highest activity on most of the target loci (13 of 15). Point mutations of C-to-A, G-to-A, G-to-T, A-to-G, T-to-A, C-to-G and CT-AG, 1 bp (T) or 2 bp (AT) deletion and 1 bp (A) insertion were generated, and the highest editing efficiency was 24.5% (FIG. 27).


The editing efficiency of double-pegRNA on all the test loci was about 4.2-times higher than that of single NGG-pegRNA (up to 27.9 times for OsNRT1.1B (insert A)), and 1.8-times higher than the average editing efficiency of single CCN-pegRNA (up to 7.2 times for OsALS (A-to-G)). Also, the proportion of by-products using double-pegRNA was not higher than that of single-pegRNA (FIG. 28).


Computational analysis based on the rice reference genome (Os-Nipponbare reference IRGSP-1.0) show that the dual-pegRNA can theoretically target 20.0% of the genome bases when a prime editing window was from +1 to +15. However, when combined with a SpCas9-NG variant having NG PAM, the dual-pegRNA strategy can target 87.9% of the rice bases (FIG. 29).


Sequence Description



  • >SEQ ID NO: 1 Wild-type SpCas9 amino acid sequence

  • >SEQ ID NO: 2 nCas9(H840A) amino acid sequence

  • >SEQ ID NO: 3 Wild-type M-MLV amino acid sequence

  • >SEQ ID NO: 4 M-MLV amino acid sequence of the present invention

  • >SEQ ID NO: 5 The amino acid sequence of CaMV reverse transcriptase

  • >SEQ ID NO: 6 The amino acid sequence of retron reverse transcriptase

  • >SEQ ID NO: 7 32aa linker amino acid sequence

  • >SEQ ID NO: 8 gRNA scaffold sequence

  • >SEQ ID NO:9 The gene sequence of the fusion protein nCas9(H840A)-M-MLV of the present invention

  • >SEQ ID NO: 10 The gene sequence of the fusion protein nCas9(H840A)-CaMV of the present invention

  • >SEQ ID NO: 11 The gene sequence of the fusion protein nCas9(H840A)-retron of the present invention

  • >SEQ ID NO: 12 The amino acid sequence of the fusion protein nCas9(H840A)-M-MLV of the present invention

  • >SEQ ID NO: 13 The amino acid sequence of the fusion protein nCas9(H840A)-CaMV of the present invention

  • >SEQ ID NO: 14 The amino acid sequence of the fusion protein nCas9(H840A)-retron of the present invention


Claims
  • 1. A plant genome editing system for targeted modification of a plant genome, comprising: i) a fusion protein and/or an expression construct comprising a nucleotide sequence encoding the fusion protein, wherein the fusion protein comprises a CRISPR nickase and a reverse transcriptase; and/orii) at least one pegRNA and/or an expression construct comprising a nucleotide sequence encoding the at least one pegRNA,wherein the at least one pegRNA comprises, in the direction from 5′ to 3′, a guide sequence, a scaffold sequence, a reverse transcription (RT) template sequence and a primer binding site (PBS) sequence,wherein the at least one pegRNA can form a complex with the fusion protein and target the fusion protein to a target sequence in the genome, so that a nick is formed in the target sequence.
  • 2. The system of claim 1, wherein the CRISPR nickase is a Cas9 nickase, e.g., comprising an amino acid sequence shown in SEQ ID NO: 2.
  • 3. The system of claim 1 or 2, wherein the reverse transcriptase is a M-MLV reverse transcriptase, preferably an enhanced M-MLV reverse transcriptase with an amino acid sequence shown in SEQ ID NO: 4, or the reverse transcriptase is a CaMV reverse transcriptase shown in SEQ ID NO: 5 or a retron reverse transcriptase shown in SEQ ID NO: 6.
  • 4. The system of any one of claims 1-3, wherein the guide sequence of the pegRNA is configured to have sufficient sequence identity with the target sequence and thus can bind to a complementary strand of the target sequence by base pairing so as to achieve sequence-specific targeting.
  • 5. The system of any one of claims 1-4, wherein the scaffold sequence of the pegRNA comprises a sequence shown in SEQ ID NO: 8.
  • 6. The system of any one of claims 1-5, wherein the primer binding sequence is configured to be complementary to at least a portion of the target sequence, and preferably the primer binding sequence is complementary to at least a portion of a 3′ free single strand caused by the nick, in particular to a nucleotide sequence at the 3′ end of the 3′ free single strand.
  • 7. The system of any one of claims 1-6, wherein the primer binding sequence has a Tm (melting temperature) of about 18-52° C., preferably about 24-36° C., more preferably about 28-32° C., and most preferably about 30° C.
  • 8. The system of any one of claims 1-7, wherein the RT template sequence is configured to correspond to a sequence downstream of the nick and comprises a desired modification, and the modification comprises substitution, deletion and/or addition of one or more nucleotides.
  • 9. The system of any one of claims 1-8, further comprising a nicking gRNA and/or an expression construct comprising a nucleotide sequence encoding the nicking gRNA, wherein the nicking gRNA comprises a guide sequence and a scaffold sequence, the guide sequence is configured to have sufficient sequence identity with a target sequence of the genome so that the fusion protein can target the target sequence and a nick can be formed in the target sequence, the target sequence of the nicking gRNA and the target sequence of the pegRNA are located on opposite strands of genomic DNA, and about 1 to about 300 nucleotides are present between the nick induced by the nicking gRNA and the nick induced by the pegRNA.
  • 10. The system of any one of claims 1-8, comprising at least a pair of pegRNAs and/or an expression construct comprising a nucleotide sequence encoding the at least a pair of pegRNAs.
  • 11. The system of claim 10, wherein two pegRNAs in the pegRNA pair are configured to target different target sequences on the same strand of the genomic DNA, or the two pegRNAs in the pegRNA pair are configured to target the target sequences on different strands of the genomic DNA.
  • 12. The system of claim 10 or 11, wherein PAM of the target sequence of one pegRNA in the pegRNA pair is located on a sense strand and PAM of the other pegRNA is located on an antisense strand.
  • 13. The system of any one of claims 10-12, wherein nicks induced by the two pegRNAs are located on two sides of the site to be modified, respectively.
  • 14. The system of claim 13, wherein the nick induced by the pegRNA for the sense strand is located upstream (in the 5′ direction) of the site to be modified, and the nick induced by the pegRNA for the antisense strand is located downstream (in the 3′ direction) of the site to be modified.
  • 15. The system of claim 14, wherein about 1 to about 300 or more nucleotides, e.g., 1-15 nucleotides, are presented between the nicks induced by the two pegRNAs.
  • 16. The system of any one of claims 10-15, wherein the two pegRNAs in the pegRNA pair are configured to introduce a same desired modification.
  • 17. A method of producing a genetically modified plant, comprising introducing the genome editing system of any one of claims 1-16 into at least one plant, thereby resulting in a modification in the genome of the at least one plant, for example, the modification comprises substitution, deletion and/or addition of one or more nucleotides.
  • 18. The method of claim 17, wherein the introduction comprises transforming the genome editing system of any one of claims 1-16 into an isolated plant cell or tissue, and then regenerating the transformed plant cell or tissue into an intact plant; or the introduction comprises transforming the genome editing system of any one of claims 1-16 into a specific part of an intact plant, such as a leaf, a shoot apex, a pollen tube, a young ear or a hypocotyl.
  • 19. The method of claim 18, further comprising culturing the plant cell, tissue or intact plant into which the genome editing system has been introduced at an elevated temperature, for example, the elevated temperature is 37° C.
Priority Claims (2)
Number Date Country Kind
201911062005.6 Nov 2019 CN national
202010036374.4 Jan 2020 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/CN2020/117736 9/25/2020 WO