CN109402115B - gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit - Google Patents

gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit Download PDF

Info

Publication number
CN109402115B
CN109402115B CN201811038238.8A CN201811038238A CN109402115B CN 109402115 B CN109402115 B CN 109402115B CN 201811038238 A CN201811038238 A CN 201811038238A CN 109402115 B CN109402115 B CN 109402115B
Authority
CN
China
Prior art keywords
grna
crispr
sequence
complex
rna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201811038238.8A
Other languages
Chinese (zh)
Other versions
CN109402115A (en
Inventor
陈侃
罗镇华
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangzhou Pushi Lihua Technology Co ltd
Original Assignee
Guangzhou Pushi Lihua Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Guangzhou Pushi Lihua Technology Co ltd filed Critical Guangzhou Pushi Lihua Technology Co ltd
Priority to CN201811038238.8A priority Critical patent/CN109402115B/en
Publication of CN109402115A publication Critical patent/CN109402115A/en
Application granted granted Critical
Publication of CN109402115B publication Critical patent/CN109402115B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Abstract

The invention provides gRNA for targeting a Rett syndrome related mutant gene RNA, and also provides a human Rett syndrome related mutant gene detection method and a detection kit based on a clustered short palindromic repeat (CRISPR) -C2C2 system at regular intervals. The invention provides a detection method, integrates the advantages of gRNA targeting recognition of a transcription product RNA (target RNA sequence) of a mutation gene related to the Rett syndrome and the characteristics that when a CRISPR-C2C2 complex detects the target RNA sequence, the complex cuts a report RNA with a detection mark and releases a detectable signal, and the CRISPR-C2C2 system is applied to detection of the mutation gene related to the Rett syndrome, so that the detection method and the detection kit have high sensitivity and high accuracy and are of great commercial application value.

Description

gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit
Technical Field
The invention relates to the field of gene detection and gene modification, in particular to a gRNA for specifically targeting a mutant gene RNA related to the Rett syndrome, a detection method and a detection kit for the mutant gene related to the Rett syndrome based on a clustered short palindromic repeat (CRISPR) system at regular intervals; in particular to gRNA for specifically targeting a Rett syndrome resistance gene and/or specific gene RNA, a Rett syndrome resistance gene and/or specific gene detection method based on a clustered short palindromic repeat (CRISPR) system at regular intervals and a detection kit.
Background
CRISPR/Cas is a powerful tool for gene editing, allowing for pinpoint precise editing of genes.
The journal of Science published an important development of research entitled "Nucleic acid detection with CRISPR-Cas13a (i.e., C2)" at month 4 and 13 of 2017. A panel of scientists from the Broad institute, mcGovern institute, etc., has modified the CRISPR system to target RNA, making it a rapid, inexpensive and highly sensitive diagnostic tool. This finding is expected to bring about a revolutionary impact on scientific research and global public health. CRISPR precursor Zhang Feng and James j. Collins from the read institute are co-communicators of this study. Researchers at the Broad institute point out that a new CRISPR technique is utilized: CRISPR-Cas13a/C2C2 can detect diseases including Zika virus infection, dengue virus infection and the like with high sensitivity, and the principle is that CRISPR-Cas13a is combined with isothermal nucleic acid amplification to detect specific RNA and DNA.
Rett Syndrome, is estimated to be about 1,000 girls with Rett Syndrome in china each year. The sick girl looks normal during the period from birth to 6 months, and symptoms begin to appear at the period from 6 to 18 months, including losing the communication ability and purposeful hand functions which are mastered before, and the occurrence of repeated plate cutting actions (such as writing, beating, biting, and the like) of the hand, and other symptoms which are used as auxiliary diagnosis are abnormal breathing rhythm, tooth grinding, scoliosis, epilepsy, and growth retardation. The clinical symptoms of Rett syndrome are variable, in part because of the different types of variation in the MECP gene. Most (80%) adult patients have epileptic symptoms and some women have improved spasticity with age, but some are difficult to treat and most require drug control.
Applicants' studies have found that there is no report on the detection method of Rett syndrome related mutant genes based on the CRISPR-C2 system. Just as the design and preparation of the gRNA of the accurate and specific target gene are key technologies for CRISPR-Cas9 gene knockout, the gRNA of the efficient and specific target gene of the target Rett syndrome related mutant gene is also key for CRISPR-C2C2 to identify the target gene, and the editing, modification and detection of the Rett syndrome related mutant gene based on a CRISPR-C2C2 system are further enabled to be possible.
Therefore, a detection method and a detection kit for a Rett syndrome related mutant gene based on a CRISPR-C2C2 system and a gRNA for specifically targeting the Rett syndrome related mutant gene RNA are to be applied.
Instructions, descriptions, product specifications, and product tables of any manufacturer of any product mentioned herein or incorporated by reference in any document herein are incorporated by reference and may be employed in the practice of the present invention. More specifically, all references are incorporated herein by reference to the same extent as if each individual reference was specifically and individually indicated to be incorporated by reference.
Disclosure of Invention
In order to solve the problems, the invention provides a gRNA for specifically targeting the mutant gene RNA related to the Rett syndrome, a detection method and a detection kit for the mutant gene related to the Rett syndrome based on a CRISPR-C2C2 system.
If not specified, the technical scheme of the invention preferably adopts a CRISPR-C2C2 system.
In a first aspect, the present invention provides a gRNA sequence that specifically recognizes a target nucleotide, where the target nucleotide is an RNA sequence corresponding to a Rett syndrome-associated mutant gene, and the coding DNA sequence corresponding to the gRNA includes one or more of the nucleotide sequences shown in SEQ ID No.1-SEQ ID No.28 in table 1.
In one embodiment of the present invention, the mutation gene related to Rett syndrome is a Rett syndrome resistance gene and/or a Rett syndrome specific gene.
In one embodiment of the present invention, the Rett syndrome associated mutant gene comprises one or more of the genes shown in gene numbers 1 to 14 in table 1.
In an embodiment of the present invention, the "RNA sequence corresponding to the Rett syndrome-related mutant gene" is a transcript corresponding to the Rett syndrome-related mutant gene.
In an embodiment of the present invention, the term "coding DNA sequence corresponding to a gRNA" is transcribed to obtain the gRNA sequence according to the first aspect of the present invention, and specifically, cloning the coding DNA sequence corresponding to a gRNA into a vector containing a T7 promoter, or directly adding a T7 promoter to the front end of the coding DNA corresponding to a gRNA by PCR, annealing, synthesis, or the like, and transcription may be performed to obtain a transcribed product gRNA sequence.
Notably, the applicant designed multiple grnas for each of the Rett syndrome-associated mutant genes shown in table 1 under gene numbers 1-14, each gRNA being capable of independently and specifically recognizing the corresponding Rett syndrome-associated mutant gene of the row.
For example, the Rett syndrome associated mutant gene is MECP2, and the coding DNA sequence corresponding to the gRNA comprises one or more of the nucleotide sequences shown in SEQ ID No.1-SEQ ID No.2 in table 1. The mutation gene related to the Rett syndrome is MECP2, and the coding DNA sequence corresponding to the gRNA comprises one or more of the nucleotide sequences shown in SEQ ID NO3-SEQ ID NO.4 in the table 1. With this in mind, it will be appreciated by those skilled in the art that each of the Rett syndrome-associated mutant genes shown in table 1 gene codes 1-14 correspond to one or more grnas in table 1, respectively. Applicant does not describe one by one.
The abbreviations of bases referred to in the present invention are annotated as follows (it being understood that, if any, other unexplored letters are present, as is conventional in the art):
h is a, c or t, b is g or c or t, d is a or g or t, v is a or g or c, and n is a or g or c or t.
In one embodiment of the invention, the gRNA can specifically recognize the target nucleotide sequence by being fully or substantially complementary, or by being a percentage of complementary.
The gRNA provided herein has sufficient complementarity to the target nucleotide sequence to hybridize to the target nucleotide sequence and direct the specific binding of CRISPR-C2 to the target nucleotide sequence. In some embodiments, the degree of complementarity between a gRNA and its corresponding target nucleotide sequence is about or more than about 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99% or more.
In embodiments of the invention, "complementary" refers to the formation of one or more hydrogen bonds between a nucleic acid and another nucleic acid sequence by means of conventional Watson-Crick base pairing or other non-conventional types. "percent complementary" means the percentage of residues in a nucleic acid molecule that can form hydrogen bonds (e.g., watson-Crick base pairing) with a second nucleic acid sequence (e.g., 5, 6, 7, 8, 9, 10 of 10 are 50%, 60%, 70%, 80%, 90%, and 100% complementary). "fully complementary" means that all consecutive residues of one nucleic acid sequence form hydrogen bonds with the same number of consecutive residues in one second nucleic acid sequence. "substantially complementary" as used herein refers to a degree of complementarity of at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 100% over a region of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more nucleotides.
In one embodiment of the invention, the gRNA is a mature gRNA sequence formed by linking the targeting sequence with the gRNA backbone sequence.
In one embodiment of the present invention, the coding DNA sequence corresponding to the targeting sequence is one or more of the nucleotide sequences shown as SEQ ID NO.1-SEQ ID NO.28 in Table 1.
Common gRNA backbone sequences include, but are not limited to: 5' -GAUUUAGACUACCCCAAAAACGAAGGGGACUAAAAC.
Without the aid of a tracrRNA, the C2 protein (i.e., cas13 a) would complex with the gRNA to form a Cas13a-gRNA complex whose RNase activity would cleave, cleave or label the target nucleotide and a reporter RNA (the reporter RNA is one of the embodiments of the "target nucleotide related sequence" described in the subsequent second, third and fourth aspects of the invention) when the Cas13a-gRNA complex detects a target nucleotide (the invention is denoted as a first target nucleic acid); wherein the first target nucleic acid carries a PFS fragment (PAM-like) that can be specifically recognized by the gRNA in the Cas13a-gRNA complex.
In one embodiment of the present invention, the second target nucleic acid may be a fluorescent-labeled reporter RNA strand, which, after cleavage, fluoresces, such that the first target nucleotide may be detected and determined by detecting the fluorescence.
In one embodiment of the invention, the Cas13a-gRNA complex is specific for the recognition of the first target nucleic acid.
In one embodiment of the invention, the Cas13a-gRNA complex is non-specific for cleavage of the second target nucleic acid.
In embodiments of the invention, the "Cas13a-gRNA complex/complex", "CRISPR-Cas13a complex/complex", "CRISPR-C2 complex/complex" concepts are interchangeable.
In embodiments of the invention, the terms "target nucleotide", "first target nucleic acid", "second target nucleic acid" refer to ribonucleotides or analogues thereof. The following are non-limiting examples of "target nucleic acid", "first target nucleic acid" or "second target nucleic acid": messenger RNA (mRNA), transfer RNA, ribosomal RNA, short interfering RNA (siRNA), short hairpin RNA (shRNA), micro-RNA (miRNA), ssRNA, or any isolated RNA (specifically, including single-stranded RNA or double-stranded RNA with single-stranded RNA). The "target nucleotide", "first target nucleic acid", "second target nucleic acid" may comprise one or more modified nucleotides, such as methylated nucleotides and nucleotide analogs. The nucleotides may be further modified by molecular markers (e.g., fluorescent markers, or other detectable molecular markers).
In embodiments of the invention, the terms "target nucleotide" and "target polynucleotide" are interchangeable.
In a second aspect the present invention provides a CRISPR-C2 system comprising: 1) C2 effector proteins;
2) One or more nucleic acids, wherein the one or more nucleic acids comprise at least one gRNA sequence of the first aspect;
the C2 protein binds to the gRNA to form a CRISPR-C2 complex, and the CRISPR-C2 complex modifies the target nucleotide and/or a sequence associated with the target nucleotide when the CRISPR-C2 complex binds to the target nucleotide of the first aspect.
In a preferred embodiment of the invention, the modification is the introduction of a cleavage, cleavage or label.
In an embodiment of the second aspect of the present invention, the target nucleotide comprises the target nucleotide (target RNA) and/or the reporter RNA of the first aspect.
In a third aspect, the invention provides a non-naturally occurring or engineered composition comprising one or more carriers comprising component I and component II:
said component I comprises a first regulatory element, and a coding sequence encoding a C2 protein operably linked to said first regulatory element; the component II comprises a second regulatory element, and a coding sequence operably linked to the second regulatory element that encodes a gRNA, wherein the gRNA comprises a gRNA sequence as described in the first aspect; wherein components I and II are on the same or different carriers;
The C2 protein binds to the gRNA to form a CRISPR-C2 complex, and the CRISPR-C2 complex modifies the target nucleotide and/or a sequence associated with the target nucleotide when the CRISPR-C2 complex binds to the target nucleotide of the first aspect.
In a preferred embodiment of the invention, the modification is the introduction of a cleavage, cleavage or label.
In a first embodiment of the third aspect of the invention, the target nucleotide comprises the target nucleotide (target RNA) and/or the reporter RNA of the first aspect.
In embodiments of the invention, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid molecule linked thereto. Vectors include, but are not limited to, single-stranded, double-stranded, or partially double-stranded nucleic acid molecules; a nucleic acid molecule comprising one or more free ends, free ends (e.g., circular); a nucleic acid molecule comprising DNA, RNA, or both; and other diverse polynucleotides known in the art. Alternatively, one type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA fragments may be inserted, for example, by standard molecular cloning techniques. Alternatively, another type of vector is a viral vector in which a viral-derived DNA or RNA sequence is present in a vector used to package a virus (e.g., retrovirus, replication-defective retrovirus, adenovirus, replication-defective adenovirus, and adeno-associated virus). Viral vectors also comprise polynucleotides carried by a virus for transfection into a host cell. Certain vectors (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors) are capable of autonomous replication in a host cell into which they are introduced. Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operably linked. Such vectors are referred to herein as "expression vectors". The common expression vectors used in recombinant DNA technology are typically in the form of plasmids.
Generally, within a vector, "operably linked" is intended to mean that the nucleotide sequence is linked to one or more regulatory elements in a manner that allows for the expression of the nucleotide sequence (alternatively, the nucleotide sequence may be expressed in an in vitro transcription/translation system; alternatively, the nucleotide sequence may be expressed when the vector is introduced into a host cell).
In embodiments of the invention, the term "expression" refers to the process of transcription from a DNA template into a polynucleotide (e.g., into mRNA or other RNA transcript) and/or the subsequent translation of the transcribed mRNA into a peptide, polypeptide, or protein. Transcripts and encoded polypeptides may be collectively referred to as "gene products". If the polynucleotide is derived from genomic DNA, expression may include splicing of mRNA in eukaryotic cells.
The terms "non-naturally occurring" or "engineered" are used interchangeably herein to refer to a nucleic acid molecule or polypeptide that is at least substantially free from at least one other component with which it is associated in nature or as found in nature.
In preferred embodiments of the invention, the first regulatory element comprises one or more pol III promoters (e.g., 1, 2, 3, 4, 5, or more pol III promoters), one or more pol II promoters (e.g., 1, 2, 3, 4, 5, or more pol II promoters), one or more pol I promoters (e.g., 1, 2, 3, 4, 5, or more pol I promoters), or a combination thereof. Examples of pol III promoters include, but are not limited to, the U6 and H1 promoters. Examples of pol II promoters include, but are not limited to, the reverse transcription Rous Sarcoma Virus (RSV) LTR promoter (optionally with an RSV enhancer), the Cytomegalovirus (CMV) promoter (optionally with a CMV enhancer) [ see, e.g., boshat et al, cell 41:521-530 (1985) ], the SV40 promoter, the dihydrofolate reductase promoter, the beta-actin promoter, the phosphoglycerate kinase (PGK) promoter, and the EF1 alpha promoter.
In some embodiments of the invention, the coding sequence encoding the C2 protein is codon optimized for expression in a particular cell, such as a eukaryotic cell. These eukaryotic cells may be those of or derived from a particular organism, such as a mammal, including but not limited to humans, mice, rats, rabbits, dogs, or non-human primates. Generally, codon optimization refers to a method of modifying a nucleic acid sequence to enhance expression in a host cell of interest by replacing at least one codon of the native sequence with a more or most frequently used codon in the gene of the host cell (e.g., about or more than about 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, or more codons while maintaining the native amino acid sequence). The codon preference (the difference in codon usage between organisms) is often related to the translational efficiency of messenger RNA (mRNA) which is believed to depend on (among other things) the nature of the codon translated and the availability of the particular transfer RNA (tRNA) molecule. The codon advantage of the selected tRNA in the cell generally reflects the codon advantage of the tRNA most frequently used for peptide synthesis. Thus, the gene can be tailored to an optimal gene expression codon usage table based on codon optimization in a given organism can be readily obtained, e.g., by using a "nucleotide optimization" table "as well as a nucleotide" can be obtained in the nucleotide optimization table "by using the nucleotide (35:2000) in the nucleotide sequence of Table of" Naidka "35.2000 (applied to the nucleotide sequence of NadNadIn) and" 2000 ". In 3.2000". In the methods of FIGS.), such as Gene force (Aptagen, inc.; jacobs (Jacobus), pa.) is also available. In some embodiments, one or more codons (e.g., 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, or more, or all codons) in the sequence encoding the CRISPR enzyme correspond to codons most frequently used for a particular amino acid.
In embodiments of the invention, the terms "C2", "C2 protein", "C2 effector protein", "Cas13a protein", "Cas13a effector protein" are interchangeable; the C2 protein is an RNA-targeted RNase that cleaves ssRNA (single-stranded small molecule RNA). The C2 proteins disclosed in the prior literature including, but not limited to: 1. discovery and Functional Characterization of Diverse Class 2 CRISPR-Cas Systems, nature,2016 Oct 13; 2. C2C2 is a single-component programmable RNA-guide RNA-targeting CRISPR effector, science,2016 Aug 5; 3. two distinct RNase activities of CRISPR-C2C2 enable guide-RNA processing and RNA detection, nature,2016 Oct 13; 4. two Distant Catalytic Sites Are Responsible for C2c2 RNase Activities, cell,2017 Jan 12.
In one embodiment of the invention, the C2 protein is a Cas13a protein gene derived from Leptotrichia wadei F0279 or Leptotrichia shahii, i.e. LwCas13a, lshCas13a, respectively.
In one embodiment of the invention, the C2 protein comprises a homolog of the C2 protein or a modified form thereof.
In one embodiment of the invention, the component I also comprises a coding sequence for any other protein or polypeptide domain and optionally a linking sequence between any two domains, which in particular may encode a linking peptide fragment of e.g. a C2 protein with any other protein or polypeptide domain, and a fusion protein is obtained. Examples of protein domains that may be fused to a C2 protein include, but are not limited to, epitope tags, reporter sequences, and protein domains having one or more of the following activities: methylase activity, demethylase activity, transcriptional activation activity, transcriptional repression activity, transcriptional release factor activity, histone modification activity, RNA cleavage activity and nucleic acid binding activity. Non-limiting examples of epitope tags include histidine (His) tags, V5 tags, FLAG tags, influenza virus Hemagglutinin (HA) tags, myc tags, VSV-G tags, and thioredoxin (Trx) tags. Examples of reporter genes include, but are not limited to, glutathione-S-transferase (GST), horseradish peroxidase (HRP), chloramphenicol Acetyl Transferase (CAT), beta-galactosidase, beta-glucuronidase, luciferase, green Fluorescent Protein (GFP), hcRed, dsRed, cyan Fluorescent Protein (CFP), yellow Fluorescent Protein (YFP), autofluorescent proteins including Blue Fluorescent Protein (BFP).
The C2 protein may also be fused to a protein or protein fragment that binds to a DNA molecule or to other cellular molecules, including, but not limited to, maltose Binding Protein (MBP), S-tag, lex a DNA Binding Domain (DBD) fusion, GAL4DNA binding domain fusion, and Herpes Simplex Virus (HSV) BP16 protein fusion. In some embodiments of the invention, the fusion protein is a molecular tag and the use of the tagged C2 protein can be used to identify the position of the target sequence.
The terms "polypeptide", "peptide" and "protein" are used interchangeably herein to refer to a polymer having amino acids of any length. The polymer may be linear or branched, it may contain modified amino acids, and it may be interrupted by non-amino acids. These terms also encompass amino acid polymers that have been modified; such as disulfide bond formation, glycosylation, lipidation (methylation), acetylation, phosphorylation, or any other modification, such as binding to a detection molecule marker component.
In a preferred embodiment of the invention, the second regulatory element is a T7 promoter.
In preferred embodiments of the invention, the first or second regulatory element further includes enhancers, internal Ribosome Entry Sites (IRES), and other expression control elements (e.g., transcription termination signals, such as polyadenylation signals and poly U sequences). Such regulatory sequences are described, for example, in Goeddel: methods of enzymology (GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY) 185, academic Press (Academic Press), san Diego (San Diego), calif., 1990. Regulatory elements include those sequences that direct constitutive expression of a nucleotide sequence in many types of host cells as well as those sequences that direct expression of the nucleotide sequence in only certain host cells (e.g., tissue-specific regulatory sequences). Tissue-specific promoters may primarily direct expression in a desired tissue of interest, such as muscle, neurons, bone, skin, blood, specific organs (e.g., liver, pancreas), or specific cell types (e.g., lymphocytes). Regulatory elements may also direct expression in a time-dependent manner (e.g., in a cell cycle-dependent or developmental stage-dependent manner), which may or may not be tissue or cell type specific.
In a fourth aspect, the invention provides a method of modifying a sequence associated with a target nucleotide according to the first aspect, the method comprising delivering a composition comprising 1) and 2), and bringing the composition comprising 1) and 2) into proximity with the target nucleotide and the sequence associated with the target nucleotide:
1) C2 effector proteins; 2) One or more nucleic acids, wherein the one or more nucleic acids comprise at least one gRNA sequence of the first aspect;
the C2 protein binds to the gRNA to form a CRISPR-C2 complex, and the CRISPR-C2 complex modifies the target nucleotide and/or a sequence associated with the target nucleotide when the CRISPR-C2 complex binds to the target nucleotide of the first aspect.
In a preferred embodiment of the invention, the modification is the introduction of a cleavage, cleavage or label.
In an embodiment of the fourth aspect of the present invention, the target nucleotide comprises the target nucleotide (target RNA) and/or the reporter RNA according to the first aspect.
In one embodiment of the invention, component I further comprises two or more coding sequences encoding grnas operably linked to the first regulatory element, each of which, when expressed, directs the CRISPR-C2 complex to specifically bind to a different target nucleotide sequence (this binding reaction may occur in a host cell, in an in vitro transcription/translation system, or other reaction solution configured by one of skill in the art according to specific experimental requirements).
In embodiments of the invention, the term "bringing a composition comprising 1) and 2) into proximity with the target nucleotide and the reporter RNA" refers to delivering a component into an ex vivo (in vitro) or in vivo (in vivo) environment, such as a reaction solution configured by one of skill in the art according to specific experimental requirements, in vivo environment such as intracellular; the term "proximal" refers to the fact that each component can be contacted with the target nucleotide and the sequence of the reporter RNA in an in vitro (in vitro) or in vivo (in vivo) environment and under conditions that would be expected by one of skill in the art.
In embodiments of the invention, the invention provides methods comprising delivering one or more polynucleotides, one or more vectors, one or more transcripts, and/or one or more transcribed proteins to a host cell. In some aspects, the invention further provides cells produced by such methods and organisms (e.g., animals, plants, or fungi) comprising or produced by such cells.
In an embodiment of the invention, the CRISPR-C2 complex combined with the gRNA is delivered to a cell. Conventional viral and nonviral based gene transfer methods can be used to introduce nucleic acids into mammalian cells or target tissues.
Such methods can be used to administer nucleic acids encoding components of the CRISPR-C2 system to cells in culture or in a host organism. Non-viral vector delivery systems include DNA plasmids, RNA (e.g., transcripts of the vectors described herein), naked nucleic acids, and nucleic acids complexed with delivery vehicles (e.g., liposomes). Viral vector delivery systems include DNA and RNA viruses, which have episomal or integrated genomes after delivery to cells. For reviews of gene delivery systems, see Ande (Anderson), "Science" 256:808-813 (1992); nabel (Nabel) & Felgner (TIBTECH 11:211-217 (1993); three-valley (Mitani) & Caskey (Caskey), TIBTECH 11:162-166 (1993); dilon (Dillon), TIBTECH 11:167-175 (1993); miller (Miller), nature 357:455-460 (1992); fanbu Lant (Van Brunt), "Biotechnology" 6 (10): 1149-1154 (1988); vigne, recovering neurology and neuroscience (Restorative Neurology and Neurosciece) 8:35-36 (1995); kleber (Kremer) & Pelicaudat (Perricaudet), "British medical publication (British Medical Bulletin) 51 (1): 31-44 (1995); dada (Haddada) et al, dulbler (Doerfler) and Bohm (editions) in microbiology and immunology current subject (Current Topics in Microbiologyand Immunology) (1995); and others (Yu) et al, (Gene Therapy) 1:13-26 (1994).
Non-viral delivery methods of nucleic acids include lipofection, nuclear transfection, microinjection, gene gun, viral particles, liposomes, immunoliposomes, polycations or lipids: nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofection is described, for example, in U.S. Pat. nos. 5,049,386, 4,946,787 and 4,897,355 and lipofection reagents are commercially available (e.g., transffectamtm and lipofectin). Useful receptor-recognizing lipofection suitable for polynucleotides include Felgner (Fisher, WO 91/17424; those of WO 91/16024. Delivery may be to cells (e.g., in vitro or ex vivo administration) or to target tissue (e.g., in vivo administration).
The preparation of nucleic acid complexes (including targeted liposomes, such as immunolipid complexes) is well known to those skilled in the art (see, e.g., kelistel (Crystal), "Science" (Science) 270:404-410 (1995), "Blaze (Blaese) et al,", cancer Gene Therapy "(Cancer Gene Therapy) 2:291-297 (1995)," Bell (Behr) et al, ", bioconjugate chem") 5:382-389 (1994), "Remy (Remy) et al,", bioconjugate chem "5:647-654 (1994)," Gao) et al, "Gene Therapy" (Gene Therapy) 2:710-722 (1995), "Emeride (Aad) et al," Cancer research "(Cancer research) 52:4817-483520 (19935, 35, 501, 3235, 35, 024, and/or (U.S. Pat. No. 4, U.S. 4, 35, 501, and/or (U.S. Pat. No. 4, 35, and/or U.S. 4, and" U.S.S.No. 4, and "patent No. 3, and" 8335, and "U.3").
In a fifth aspect the invention provides a eukaryotic host cell comprising component I and/or component II:
said component I comprises a first regulatory element, and a coding sequence encoding a C2 protein operably linked to said first regulatory element; said component II comprises a second regulatory element, and a coding sequence operably linked to said second regulatory element that encodes a gRNA, wherein said gRNA comprises a gRNA sequence of the first aspect;
wherein components I and II are on the same or different carriers;
the C2 protein binds to the gRNA to form a CRISPR-C2 complex, and the CRISPR-C2 complex modifies the target nucleotide and/or a sequence associated with the target nucleotide when the CRISPR-C2 complex binds to the target nucleotide of the first aspect.
In a preferred embodiment of the invention, the modification is the introduction of a cleavage, cleavage or label.
In an embodiment of the fifth aspect of the present invention, the target nucleotide comprises the target nucleotide (target RNA) and/or the reporter RNA according to the first aspect.
In one embodiment of the invention, the eukaryotic host cell comprises component I and component II.
In one embodiment of the invention, component I further comprises two or more coding sequences encoding a gRNA operably linked to the first regulatory element, each of the two or more grnas, when expressed, directs the CRISPR-C2 complex to specifically bind to a different target nucleotide sequence in a eukaryotic host cell.
In a sixth aspect the invention provides a test kit comprising one or more of the gRNA sequence provided in the first aspect, the CRISPR-C2 system provided in the second aspect, the non-naturally occurring or engineered composition provided in the third aspect, the eukaryotic host cell provided in the fifth aspect.
In one embodiment of the invention, the kit further comprises a conventional matched reaction reagent and/or reaction equipment. For example, the kit may provide one or more reaction or storage buffers. The reagents may be provided in a form useful in the particular assay or in a form (e.g., in concentrated or lyophilized form) in which one or more other components are added as needed prior to use. The buffer may be any buffer including, but not limited to, sodium carbonate buffer, sodium bicarbonate buffer, borate buffer, tris buffer, MOPS buffer, HEPES buffer, and combinations thereof. In some embodiments, the buffer is alkaline. In some embodiments, the buffer has a pH of from about 7 to about 10. In some embodiments, the kit comprises one or more oligonucleotides, the one or more nucleic acids comprising at least one gRNA comprising a gRNA sequence as described in the first aspect.
The components of the kits of the invention may be provided individually or in combination and may be provided in any suitable container, such as vials, bottles, tubes or cardboard.
The seventh aspect of the present invention provides a detection method of a Rett syndrome related mutant gene based on a CRISPR-C2 system, comprising:
1) Preparing or providing a sample to be tested, wherein the sample to be tested comprises DNA and/or RNA;
2) Providing a composition comprising a), b), and C), component a) comprising a C2 effector protein; component b) comprises one or more nucleic acids, wherein the one or more nucleic acids comprise at least one gRNA comprising a gRNA sequence of the first aspect; component c) comprises a reporter RNA modified with a molecular detection label;
3) Contacting a composition comprising a) b) C) with the sample to be tested in a reaction system, wherein the C2C2 protein is combined with gRNA to form a CRISPR-C2C2 complex, the CRISPR-C2C2 complex is combined with the target nucleotide, and shearing the reporter RNA modified with the molecular detection mark to generate a detectable molecular detection mark;
4) And detecting the molecular detection mark to obtain a detection result.
In one embodiment of the present invention, the target nucleotide is RNA obtained by transcription of a T7 polymerase from a DNA fragment to which a T7 promoter is linked.
Alternatively, the DNA fragment is obtained by extraction or purification and modified with a T7 promoter. Optionally, the extracted or purified DNA fragment is subjected to PCR amplification, NASBA isothermal amplification or recombinase polymerase RPA amplification treatment and modified with a T7 promoter.
In one embodiment of the invention, the reaction system comprises a Cas13a detection system. In a specific embodiment of the present invention, the Cas13a detection system includes: 45nM purified LwCas13a,22.5nM gRNA,125nM, a reporter RNA strand that fluoresces upon cleavage of LwCas13a (RNAse Alert v2, thermo Scientific), 2. Mu.L mouse-derived RNase inhibitor (New England Biolabs), 100ng total human RNA (purified from HEK293FT medium), different amounts of target nucleic acid, and nuclease detection buffer (40 mM Tris-HCl,60mM NaCl,6mM MgCl 2,pH 7.3).
In one embodiment of the invention, the reaction system comprises an RPA-DNA amplification system, a reaction system in which a T7 polymerase transcribes DNA into RNA, and a Cas13a detection system. In one embodiment of the present invention, the reaction system (50. Mu.L system) comprises: 0.48. Mu.M forward primer, 0.48. Mu.M reverse primer, 1 XRPA make-up buffer, different amounts of DNA,45nM LwCas13a recombinant protein, 22.5nM gRNA,250ng total human RNA,200nM RNA reporter (RNase alert v 2), 4. Mu.L murine RNase inhibitor (New England Biolabs), 2mM ATP, 2mM GTP,2mM UTP,2mM CTP,1. Mu.LT 7 polymerase mixture (New England Biolabs), 5mM MgCl 2 And 14mM MgAc.
In an eighth aspect the invention provides a kit for detection of a Rett syndrome associated mutant gene based on a CRISPR-C2 system comprising one or more of the gRNA sequence provided in the first aspect, the CRISPR-C2 system provided in the second aspect, the non-naturally occurring or engineered composition provided in the third aspect, the eukaryotic host cell provided in the fifth aspect.
In an embodiment of the present invention, the kit provided in the eighth aspect further includes: one or more reagents from the group consisting of PCR amplification, NASBA isothermal amplification or recombinase polymerase RPA, loop-mediated isothermal amplification (LAMP), strand Displacement Amplification (SDA), helicase Dependent Amplification (HDA), and Nicking Enzyme Amplification Reaction (NEAR).
In one embodiment of the present invention, the kit provided in the eighth aspect further comprises T7 polymerase.
In a ninth aspect, the invention provides the use of a gRNA sequence as described in the first aspect, comprising:
(i) Forming a complex with C2, and visualizing the intracellular transport and/or localization of the target RNA specifically bound to the gRNA sequence in combination with a molecular labeling technique, such as a fluorescent labeling technique;
(ii) Forming a complex with C2, capturing specific transcripts that bind specifically to the gRNA sequence (biotin ligase activity is localized to a specific transcript by direct pull down of dC2C2 or using dC2C 2).
Drawings
FIG. 1 shows the detection results of the C2C 2-based Rett syndrome related mutant gene according to the examples of the present invention.
Detailed Description
The following description is of the preferred embodiments of the present invention, and it should be noted that, for those skilled in the art, it is possible to make several improvements and modifications without departing from the principle of the embodiments of the present invention, and these improvements and modifications are also considered as the protection scope of the embodiments of the present invention.
Unless otherwise specified, reagents and consumables used in the examples of the present invention are commercially available.
The practice of the present invention employs, unless otherwise indicated, conventional techniques of immunology, biochemistry, chemistry, molecular biology, microbiology, cell biology, genomics and recombinant DNA, which are within the skill of the art. See Sambrook (Sambrook), friech (Fritsch) and manitis (Maniatis), molecular cloning: laboratory Manual (MOLECULAR CLONING: A LABORATORY MANUAL), edit 2 (1989); the handbook of contemporary molecular biology (CURRENT PROTOCOLS IN MOLECULAR BIOLOGY) (edited by f.m. ausubel (f.m. ausubel) et al, (1987)); series (academic publishing company) of methods in enzymology (METHODS IN ENZYMOLOGY): PCR2 practical methods (PCR 2:A PRACTICAL APPROACH) (M.J. MaxFrisson (M.J. MacPherson), B.D. Black (B.D. Hames) and G.R. Taylor (G.R. Taylor) editions (1995)), harlow and Lane editions (1988) antibodies: laboratory Manual (ANTIBODIES, A LABORATORY MANUAL), animal cell CULTURE (ANIMAL CELL CULTURE) (R.I. French Lei Xieni (R.I. Freshney) eds. (1987)).
In one embodiment of the invention, the present examples provide a gRNA for targeting a mutant gene RNA associated with Rett syndrome. The embodiment of the invention also provides a detection method and a detection kit of the Rett syndrome related mutant gene based on C2C2, and one or more steps of an experimental method disclosed in a paper titled "Nucleic acid detection with CRISPR-Cas13a" published in the Science journal (hereinafter referred to as "document 1") are incorporated into the embodiment by reference. Including but not limited to one or more of the following:
1. cas13a (i.e., C2C 2) gene clone and protein expression
The Cas13a protein genes derived from Leptotrichia wadei F0279 and Leptotrichia shahii are used, and are codon optimized to make the genes more suitable for expression in mammalian cells. The optimized Cas13a protein gene was cloned into the pACYC184 backbone (this backbone includes a spacer sequence that is expressed driven by the J23119 promoter, which is a β -lactamase targeting or non-targeting spacer region).
The Cas13a protein gene subjected to codon optimization is cloned to a prokaryotic expression plasmid vector, and the prokaryotic expression plasmid vector can adopt a pET plasmid with a 6-His histidine tag, so that the protein can be conveniently purified and expressed. The expression strain is Rosetta2 (DE 3).
Plasmids used in the examples of the present invention include: pC004 plasmid map: https:// benchling.com/s/lPJ cCwR (i.e., pACYC184 with beta-lactamase scanning site); pC009 plasmid map: https:// benchling.com/s/seq-ylkMuglYmiG4A3VhShZg (LshCas 13a gene inserted into pACYC184 plasmid with β -lactamase scanning site); pC010 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LshCas 13a gene inserted into pACYC184 plasmid without β -lactamase scanning site); pC011 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LwCas 13a gene inserted into pACYC184 plasmid with β -lactamase scanning site); pC012 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LwCas 13a gene inserted into pACYC184 plasmid without β -lactamase scanning site); pC013 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LwCas 13a gene inserted into the pACYC184 plasmid with Twin-Strep tag); after the recombinant expression vector of the Cas13a protein is converted, protein expression, SDS-PAGE detection and gel column purification are carried out, and the obtained purified Cas13a protein is preserved at the temperature of minus 80 ℃.
2. Preparation of target RNA gRNA preparation of target nucleotide:
The target nucleotide is extracted and amplified by PCR amplification, recombinase Polymerase Amplification (RPA), NASBA isothermal amplification or loop-mediated isothermal amplification (LAMP), strand Displacement Amplification (SDA), helicase Dependent Amplification (HDA) and Nicking Enzyme Amplification Reaction (NEAR). The target RNA was obtained by gel separation and purification (using MinElute gel extraction kit (Qiagen) kit), incubation of the purified dsDNA with T7 polymerase overnight at 30℃using HiScribe T7 Quick High Yield RNA Synthesis kit (New England Biolabs) kit, and then RNA purification using MEGAclear Transcription Clean-up kit (Thermo Fisher).
NASBA isothermal amplification
At 4 ℃, an amplification system was configured as follows:
placing the mixed system at 65 ℃ for 2min; then at 41 ℃ for 2 minutes;
to the above mixed system was further added 5ul of enzyme mixture (Life Sciences, NEC-1-24) to obtain 20. Mu.L of the total reaction system. The reaction was carried out at 65℃for 2 hours.
Recombinase polymerase amplification RPA (Recombinase Polymerase Amplification)
Designing an RPA primer by adopting NCBI Primer blast, wherein the amplified fragment size is 80-180nt, the denaturation temperature of the primer can be 54-67 ℃, the length is 30-35nt, the Opt=32, the GC content in the primer is 40-60%, and the DNA primer is synthesized according to a designed sequence.
Reference is made toBasic and +.>BasicRT (TwistDx) the RPA reaction was performed with the exception that 280mM MgAc, magnesium acetate, was added prior to the addition of the template fragment. The reaction was carried out at 37℃for 2 hours.
3. Preparation of gRNA
Preparation of gRNA referring to HiScribet7 Quick High Yield RNA Synthesis kit (New England Biolabs) kit instructions, mixing the DNA fragment with T7 promoter, T7 primer, T7 polymerase, and incubating overnight at 37 ℃; purified gRNAs were obtained by further purification using a RNAXP clean beads (Beckman Coulter) kit.
4. Detection of Rett syndrome-associated mutant Gene
The detection system of the mutation gene related to the Rett syndrome comprises the following steps: 45nM purified LwCas13a,22.5nM gRNA,125nM, a reporter RNA strand that fluoresces upon cleavage of LwCas13a (RNAse Alert v2, thermo Scientific), 2. Mu.l mouse-derived RNase inhibitor (New England Biolabs), target RNA, and nuclease detection buffer (40 mM Tris-HCl,60mM NaCl,6mM MgCl 2,pH 7.3). The reaction system was placed in a fluorescence analyzer (BioTek) and reacted at 37 ℃ (unless otherwise indicated) for 1-3 hours, and fluorescence kinetics was detected once for 5 minutes.
5. SHERLOCK (Specific High Sensitivity Enzymatic Reporter UnLOCKing) one-step method for detecting mutation gene related to Rett syndrome
Alternatively, the aforementioned DNA amplification, transcription of DNA to RNA by T7 polymerase, and Cas13a detection system may be configured to react in the same system. Optionally, the integrated architecture configuration comprises:
in a 50 μl system:
0.48. Mu.M forward primer, 0.48. Mu.M reverse primer, 1 XRPA make-up buffer, different amounts of DNA,45nM LwCas13a recombinant protein, 22.5nM gRNA,250ng total human RNA,200nM RNA reporter (RNase alert v 2), 4. Mu.l murine RNase inhibitor (New England Biolabs), 2mM ATP,2mM GTP,2mM UTP,2mM CTP,1. Mu. l T7 polymerase mixture (New England Biolabs), 5mM MgCl 2 And 14mM MgAc. The reaction system was placed in a fluorescence analyzer (BioTek) and reacted at 37 ℃ (unless otherwise indicated) for 1-3 hours, and fluorescence kinetics was detected once for 5 minutes.
6. SHERLOCK (Specific High Sensitivity Enzymatic Reporter UnLOCKing) freeze drying and paper deposition
Glass fiber filter paper (Whatman, 1827-021) was autoclaved for 90 min (Consolidated Stills and sterilant, MKII) and blocked in 5% nuclease free BSA (EMD Millipore,126609-10 GM) overnight. After washing the paper once with nuclease-free water (Life technologies, AM 9932), the ribonuclease was removed by incubation (60 ℃) with 4% RNAsecure TM (Life technologies, AM 7006) for 20 minutes, and the paper was rinsed 3 times with nuclease-free water to remove traces of RNAsecure. The treated paper was dried at 80℃for 20 minutes on a hot plate (Cole-Parmer, IKA C-Mag HS 7) before use. The 1.8. Mu. LCas13a reaction mixture (as described previously) was placed on a tray (2 mm) in a black, transparent bottom 384 well plate (Corning, 3544). For the freeze-drying test of SHERLOCK, plates containing reaction mixture trays were flash frozen in liquid nitrogen and freeze-dried overnight as described previously. RPA samples were diluted 1:10 in nuclease free water and 1.8. Mu.L of the mixture was loaded onto a paper tray and incubated at 37℃using a plate reader, bioTek Neo.
7. Analysis of SHERLOCK fluorescence data
To calculate background-removed fluorescence data, the initial fluorescence of the sample is removed, facilitating comparison between different conditions. Background fluorescence (without target nucleotide or without gRNA) is removed from the sample, thereby obtaining data with background fluorescence subtracted.
It will be appreciated by those skilled in the art that conventional alternative methods in the art may be used to replace one or more of the steps of cloning the Cas13a gene, constructing the recombinant expression vector, expressing and purifying the Cas13a protein, amplifying the target nucleotide/target gene fragment, etc., as in the embodiments of the present invention, in order to obtain similar or equivalent effects.
As will be appreciated by those skilled in the art, as disclosed in document 1: the sequence of the gRNA and Protospacer Flanking Site (PFS) is very important for different target nucleotides. PFS is a specific motif present near the target site, necessary for strong ribonuclease activity of Cas13 a. Although this motif is similar to PAM sequences, PAM is an important sequence of the second class of CRISPR-cas systems for DNA targeting, PFS is functionally different from PAM in that PFS is not involved in preventing self-targeting of CRISPR loci in endogenous systems. Importance of PFS to Cas13 a: such as the effect in the formation and cleavage activity of the gRNA target complex, requires further investigation in the future.
The gRNA for targeting the RNA of the REtt syndrome related mutant gene, the detection method of the C2C 2-based REtt syndrome related mutant gene and the detection kit provided by the invention are used for detecting the REtt syndrome related mutant gene including but not limited to the REtt syndrome related mutant gene shown in table 1.
In the examples of the present invention, the case of each base in Table 1 has no special meaning. It will be appreciated by those skilled in the art that the size of each base in Table 1 of the present invention can be changed from upper case to lower case or vice versa, with no change in meaning.
In an embodiment of the present invention, for each Rett syndrome-associated mutant gene, one or more gRNA sequences specifically recognizing the Rett syndrome-associated mutant gene are provided, as shown in table 1:
effect examples
In a specific embodiment of the invention, the embodiment of the invention provides a gRNA for targeting a Rett syndrome related mutant gene RNA, a detection method and a detection kit for a Rett syndrome related mutant gene based on C2C2, which comprise one or more of the following steps:
the embodiment is only one specific implementation manner of the technical scheme of the invention, and does not specifically limit the protection scope of the invention.
1. Cas13a (i.e., C2C 2) gene clone and protein expression
Cas13a (i.e., C2) gene cloning and protein expression, activity detection (assays) reference "Nucleic acid detection with CRISPR-Cas13a, jonathan S Gootenberg, science,2017.4.13" (literature 1) discloses experimental methods and procedures.
Unless otherwise specified, conventional reagents used in the present invention are commercially available.
The related primer description related to the embodiment of the invention:
and a gRNA-related primer sequence:
T7Lwa DRgRNA FP:
TAATACgACTCACTATAggggggATTTAgACTACCCCAAAAACgAAggggACTAAAAC
crRNA/gRNA primers:
reverse complement of the gRNA sequence shown in SEQ ID No.1-28 of 5' -Table 1
GTTTTAGTCCCCTTCGTTTTTGGGGTAGTCT-3’。
Primer sequences related to the target sequence:
T7FP:TAATACgACTCACTATAggg
wt target primer sequence (wt represents the gene sequence of normal human):
5' -TCGAG-corresponds to the gRNA sequences shown in SEQ ID No.1-28 of Table 1 (the gRNA sequences shown in SEQ ID No.1-28 of Table 1 are mutants thereof, respectively, "corresponding" means the wild type corresponding to the mutant, i.e., the gene sequence corresponding to the normal human disclosed in genebank or ncib of Table 1)
ATTTAGCCCTATAGTGAGTCGTATTA-3’。
mut target primer sequence:
5 '-TCGAG-SEQ ID NO.1-28 of Table 1 shows the gRNA sequence-ATTTAGCCCTATAGTGAGTCGTATTA-3'.
The Cas13a protein gene derived from Leptotrichia wadei F0279 is adopted, and is subjected to codon optimization, so that the gene is more suitable for expression in mammalian cells. The optimized Cas13a protein gene was cloned into the pACYC184 backbone (this backbone includes a spacer sequence that is expressed driven by the J23119 promoter, which is a β -lactamase targeting or non-targeting spacer region).
The Cas13a protein gene subjected to codon optimization is cloned to a prokaryotic expression plasmid vector, and the prokaryotic expression plasmid vector can adopt a pET plasmid with a 6-His histidine tag, so that the protein can be conveniently purified and expressed. The expression strain is Rosetta2 (DE 3).
Plasmids used in the examples of the present invention include: pC004 plasmid map: https:// benchling.com/s/lPJ cCwR (i.e., pACYC184 with beta-lactamase scanning site); pC009 plasmid map: https:// benchling.com/s/seq-ylkMuglYmiG4A3VhShZg (LshCas 13a gene inserted into pACYC184 plasmid with β -lactamase scanning site); pC010 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LshCas 13a gene inserted into pACYC184 plasmid without β -lactamase scanning site); pC011 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LwCas 13a gene inserted into pACYC184 plasmid with β -lactamase scanning site); pC012 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LwCas 13a gene inserted into pACYC184 plasmid without β -lactamase scanning site); pC013 plasmid map: https:// benchling.com/s/seq-2WApFr3zni GOACyQY8a (LwCas 13a gene inserted into the pACYC184 plasmid with Twin-Strep tag); after the recombinant expression vector of the Cas13a protein is converted, protein expression, SDS-PAGE detection and gel column purification are carried out, and the obtained purified Cas13a protein is preserved at the temperature of minus 80 ℃.
2. Pretreatment of site template to be detected
10 mu M T primer, 10 mu M target primer, 2x KOD FX buffer,200 mu M dNTP,0.1 mu L KOD FX (KOD FX, KFX-101, TOYOBO), and water to 10 mu L. The PCR procedure was followed: amplification was performed for 6 cycles at 94℃for 10s,60℃for 10s, and 68℃for 20 s. After the reaction was completed, water was added to 50. Mu.L and the mixture was kept at-20℃until it was used.
Target primer sequences were synthesized separately, each comprising the gRNA sequences shown in SEQ ID NOS.1-28 in Table 1.
3. CrRNA preparation
10 mu M T primer, 10 mu M crRNA primer, 2x KOD FX buffer,200 mu M dNTP,0.5 mu L KOD FX (KOD FX, KFX-101, TOYOBO), and water to 50. Mu.L. The PCR procedure was followed: amplification was performed for 6 cycles at 94℃for 10s,60℃for 10s, and 68℃for 20 s. The product was purified with Qiaquick PCR Purification kit (Qiaquick PCR Purification,28104, qiagen) eluting with 15. Mu.L TE. In vitro transcription: mu.g of PCR product, 10. Mu.L of each of 5x transcription buffer,ATP and GTP, UTP, CTP (NTP Set,100mM Solution,R0481,Thermo) 1. Mu.L, 1.5. Mu. L T7 RNA Polymerase (T7 RNA Polymerase (20U/. Mu.L), EP0111, thermo) and water were added to 50. Mu.L. The reaction was carried out at 37℃for 16 hours. After the reaction was completed, 2. Mu.L of Turbo DNase (TURBO DNase (2U/. Mu.L), AM2239, thermo) was added and incubated at 37℃for 2 hours. The reaction products were purified using the RNeasy Mini Kit (Rneasy Mini Kit,76106, qiagen) using the Kit protocol and crRNA was eluted with 20. Mu.L RNase free water and stored at-20℃for further use.
The crRNA primer sequences were synthesized separately, each comprising a sequence that is reverse-complementary to the gRNA sequences shown in SEQ ID NOS.1-28 in Table 1 (specifically, the gRNA sequences shown in SEQ ID NOS.1-28 in Table 1 of the present invention are coding sequences of gRNAs, which require transcription to obtain gRNAs)
4. Detection system (50 mu L)
mu.L of the sample to be tested, 45nM LwCas13a,22.5nM crRNA,25ng human total RNA,125nM substrate reporter (RNaseAlert Lab Test Kit v2, 4479768, thermo), buffer (20mM HEPES,60mM NaCl,6mM MgCl2,pH 6.8), 1. Mu.L of RNase inhibitor (RNasin Ribonuclease Inhibitors, N2515, promega), 1mM ATP,1mM GTP,1mM UTP,1mM CTP (NTP Set,100mM Solution, R0481, thermo), 1.5. Mu. L T7 RNA Polymerase (T7 RNA Polymerase (20U/. Mu.L), EP0111, thermo).
5. Reading board
Recording was performed with a VICTOR X5 plate reader with excitation wavelength 490/emission wavelength 520, reading from the top of the well, reading time 1 second per well, plate temperature set to 37 ℃, reading value once every 5 minutes, and recording was continued for 2 hours.
This example repeats the effect example steps two to five above for each of the gRNA sequences shown in SEQ ID nos. 21-30 in table 2, 3 times for averaging, and the fluorescence intensity after removal of the background is shown in fig. 1. In FIG. 1, the abscissa is the SEQ ID NO. numbers of SEQ ID NO.1-28 of Table 1; the ordinate is the fluorescence intensity with background removed. wt is the gene of normal person corresponding to the target sequence to be detected, mut is the mutant corresponding to the target sequence.
As can be seen from FIG. 1, the degrees of differentiation between the gene and mutant form of the gene to be tested are different from each other between different gRNAs: some gRNAs have very strong specificity, can specifically identify mutant types, such as SEQ ID NO.18 and SEQ ID NO. 26 in figure 1, and have fluorescence difference of more than 2 times; some gRNAs have strong specificity, can well distinguish mutant genes from normal genes, such as SEQ ID NO.6, 12, 16 and 22 in the attached figure 1, and have fluorescence difference of about 1.5-2 times; some gRNAs have poor specificity and recognize mutant and normal genes to a certain extent, for example, SEQ ID NO.2, 8, 24 and 28 in the figure 1 show that the fluorescence difference is about 1.2-1.5 times; the remaining differences are between 1-1.2 times, such as SEQ ID NO.4, 10, 14, 20 in FIG. 1.
In practical application, the fluorescence difference is more than 1.2 times, so that the detection of basic distinguishability can be satisfied, and the specific purpose and requirement to be detected are met.
The method comprises the following steps: to further illustrate the beneficial effects of the present invention, the present invention extracts the reading result of the fifth step, and the statistics are as follows, and the corresponding data in fig. 1 includes:
mean PC >chrX:153296192 >chrX:153296495 >chrX:153296761 >chrX:153296777
Mutant 0 37565.75 12834 36825.25 15492.25
WT 67447 24688.25 11819 22144.5 10021.75
mean >chrX:153296777 >chrX:153296903 >chrX:153296903 >chrX:153296903 >chrX:153296904
Mutant 17430.25 48986.75 46252.75 50515 39465
WT 15943.25 28839.5 42948.25 32532.75 17589.5
mean >chrX:153296904 >chrX:153297761 >chrX:153297889 >chrX:153297889 >chrX:153297971
Mutant 14993.5 29783.5 29211.75 30103.5 14855.25
WT 13140.25 16547.5 22295 14348.5 11454.5
sequence listing
<110> Guangzhou Yongyou investment consultation Limited
<120> method and kit for detecting gRNA targeting REtt mutant gene RNA
<130> 2018
<160> 28
<170> SIPOSequenceListing 1.0
<210> 1
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 1
ctahgggggt gaggaggcgc tgctgctg 28
<210> 2
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 2
ctaghggggt gaggaggcgc tgctgctg 28
<210> 3
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 3
cctabgggtc ggcctcagct tttcgctt 28
<210> 4
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 4
cctaahggtc ggcctcagct tttcgctt 28
<210> 5
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 5
tachtggttt ctgctctcgc cgggaggg 28
<210> 6
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 6
tacgvggttt ctgctctcgc cgggaggg 28
<210> 7
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 7
ctcadcggga ggggctccct ctcccagt 28
<210> 8
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 8
ctcacdggga ggggctccct ctcccagt 28
<210> 9
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 9
ctctdcggga ggggctccct ctcccagt 28
<210> 10
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 10
ctctcdggga ggggctccct ctcccagt 28
<210> 11
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 11
acahtgggga caaacagaaa gacacaag 28
<210> 12
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 12
acagvgggga caaacagaaa gacacaag 28
<210> 13
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 13
acchtgggga caaacagaaa gacacaag 28
<210> 14
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 14
accgvgggga caaacagaaa gacacaag 28
<210> 15
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 15
acghtgggga caaacagaaa gacacaag 28
<210> 16
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 16
acggvgggga caaacagaaa gacacaag 28
<210> 17
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 17
ctavggggac aaacagaaag acacaagg 28
<210> 18
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 18
ctathgggac aaacagaaag acacaagg 28
<210> 19
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 19
ctcvggggac aaacagaaag acacaagg 28
<210> 20
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 20
ctcthgggac aaacagaaag acacaagg 28
<210> 21
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 21
tcadcggtca cggatgatgg agcgccgc 28
<210> 22
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 22
tcacdggtca cggatgatgg agcgccgc 28
<210> 23
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 23
ctcbtggctg cacgggctca tgcttgcc 28
<210> 24
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 24
ctcavggctg cacgggctca tgcttgcc 28
<210> 25
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 25
cttbtggctg cacgggctca tgcttgcc 28
<210> 26
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 26
cttavggctg cacgggctca tgcttgcc 28
<210> 27
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 27
cctahaggcc ctggaggtcc tggtcttc 28
<210> 28
<211> 28
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 28
cctagbggcc ctggaggtcc tggtcttc 28

Claims (13)

1. The gRNA sequence specifically recognizing target nucleotide is used for detecting target nucleotide by SHERLOCK, and is characterized in that the target nucleotide is an RNA sequence corresponding to a mutation gene related to Rett syndrome, and the coding DNA sequence corresponding to gRNA comprises any one of nucleotide sequences shown as SEQ ID NO.2, SEQ ID NO.6, SEQ ID NO.8, SEQ ID NO.12, SEQ ID NO.16, SEQ ID NO.18, SEQ ID NO.22, SEQ ID NO.24, SEQ ID NO.26 and SEQ ID NO. 28.
2. A CRISPR-C2 system, comprising:
1) C2 protein;
2) One or more nucleic acids, wherein the one or more nucleic acids comprise at least one gRNA sequence of claim 1;
the C2 protein binds to gRNA to form a CRISPR-C2 complex, and when the CRISPR-C2 complex binds to the target nucleotide of claim 1, the CRISPR-C2 complex pair:
modifying the target nucleotide; and/or
The reporter RNA is modified.
3. A non-naturally occurring or engineered composition, comprising one or more carriers comprising component I and component II:
Said component I comprises a first regulatory element, and a coding sequence encoding a C2 protein operably linked to said first regulatory element; the component II comprises a second regulatory element, and a coding sequence operably linked to the second regulatory element that encodes a gRNA, wherein the gRNA comprises the gRNA sequence of claim 1;
wherein components I and II are on the same or different carriers;
the C2 protein binds to gRNA to form a CRISPR-C2 complex, and when the CRISPR-C2 complex binds to the target nucleotide of claim 1, the CRISPR-C2 complex pair:
modifying the target nucleotide; and/or
The reporter RNA is modified.
4. Use of the gRNA sequence of claim 1 in the preparation of a kit, wherein the method of use of the kit comprises the steps of: comprising delivering a composition comprising 1) and 2), bringing the composition comprising 1) and 2) into proximity with the target nucleotide:
1) C2 protein;
2) One or more nucleic acids, wherein the one or more nucleic acids comprise at least one gRNA sequence of claim 1;
the C2 protein binds to gRNA to form a CRISPR-C2 complex, and when the CRISPR-C2 complex binds to the target nucleotide of claim 1, the CRISPR-C2 complex pair:
Modifying the target nucleotide; and/or
The reporter RNA is modified.
5. A eukaryotic host cell comprising component I and component II:
said component I comprises a first regulatory element, and a coding sequence encoding a C2 protein operably linked to said first regulatory element; the component II comprises a second regulatory element, and a coding sequence operably linked to the second regulatory element that encodes a gRNA, wherein the gRNA comprises the gRNA sequence of claim 1;
wherein components I and II are on the same or different carriers;
the C2 protein binds to gRNA to form a CRISPR-C2 complex, and when the CRISPR-C2 complex binds to the target nucleotide of claim 1, the CRISPR-C2 complex pair:
modifying the target nucleotide; and/or
The reporter RNA is modified.
6. The CRISPR-C2 system of claim 2, wherein said modification comprises cleavage.
7. The non-naturally occurring or engineered composition of claim 3, wherein said modification comprises cleavage.
8. The use of claim 4, wherein the modification comprises cleavage.
9. The eukaryotic host cell of claim 5, wherein the modification comprises cleavage.
10. A test kit comprising one or more of the gRNA sequences provided in claim 1, the CRISPR-C2 system provided in claim 2, the non-naturally occurring or engineered composition provided in claim 3, the eukaryotic host cell provided in claim 5.
11. The kit of claim 10 for detecting Rett syndrome-related mutant genes based on CRISPR-C2 system, wherein the method of using the kit comprises the steps of:
1) Preparing or providing a sample to be tested, wherein the sample to be tested comprises DNA and/or RNA;
2) Providing a composition comprising a), b), and C), component a) comprising a C2 protein; component b) comprises one or more nucleic acids, wherein the one or more nucleic acids comprise at least one gRNA comprising the gRNA sequence of claim 1; component c) comprises a reporter RNA modified with a molecular detection label;
3) Contacting a composition comprising a) b) C) with the sample to be tested in a reaction system, wherein the C2C2 protein is combined with gRNA to form a CRISPR-C2C2 complex, the CRISPR-C2C2 complex is combined with the target nucleotide, and shearing the reporter RNA modified with the molecular detection mark to generate a detectable molecular detection mark;
4) And detecting the molecular detection mark to obtain a detection result.
12. The kit of claim 10, wherein the kit further comprises T7 polymerase for detecting Rett syndrome-related mutant genes based on the CRISPR-C2 system.
13. A composition comprising the gRNA sequence of claim 1, wherein the method of using the composition comprises the steps of:
(i) The gRNA forms a complex with C2C2, and the target RNA specifically combined with the gRNA sequence is transported and/or positioned in cells by combining a molecular marking technology; or (b)
(ii) The gRNA forms a complex with C2, capturing specific transcripts that specifically bind to the gRNA sequence.
CN201811038238.8A 2018-09-06 2018-09-06 gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit Active CN109402115B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811038238.8A CN109402115B (en) 2018-09-06 2018-09-06 gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811038238.8A CN109402115B (en) 2018-09-06 2018-09-06 gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit

Publications (2)

Publication Number Publication Date
CN109402115A CN109402115A (en) 2019-03-01
CN109402115B true CN109402115B (en) 2024-02-02

Family

ID=65464597

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811038238.8A Active CN109402115B (en) 2018-09-06 2018-09-06 gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit

Country Status (1)

Country Link
CN (1) CN109402115B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110396540B (en) * 2019-04-30 2023-06-09 广州普世利华科技有限公司 Method for detecting hemophilia gene mutation site
CN110396543A (en) * 2019-04-30 2019-11-01 广州普世利华科技有限公司 A kind of tumour associated gene mutation site screening method

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107557455A (en) * 2017-09-15 2018-01-09 国家纳米科学中心 A kind of detection method of the nucleic acid specific fragment based on CRISPR Cas13a
CN108290933A (en) * 2015-06-18 2018-07-17 布罗德研究所有限公司 Reduce the CRISPR enzyme mutants of undershooting-effect

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG10201804976YA (en) * 2013-12-12 2018-07-30 Broad Inst Inc Delivery, Use and Therapeutic Applications of the Crispr-Cas Systems and Compositions for Genome Editing
US9790490B2 (en) * 2015-06-18 2017-10-17 The Broad Institute Inc. CRISPR enzymes and systems

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108290933A (en) * 2015-06-18 2018-07-17 布罗德研究所有限公司 Reduce the CRISPR enzyme mutants of undershooting-effect
CN107557455A (en) * 2017-09-15 2018-01-09 国家纳米科学中心 A kind of detection method of the nucleic acid specific fragment based on CRISPR Cas13a

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CREB Signaling Is Involved in Rett Syndrome Pathogenesis;Qian Bu等;《The Journal of Neuroscience》;20170329;第3671页"摘要"、第3673页右栏第5段、第3676页右栏第1段、第3675页图2 *
Nucleic acid detection with CRISPR-Cas13a/C2c2;Jonathan S. Gootenberg et al.;《Science》;20170428;第2页第1-3段、第4页第2-5段、图1、图3、图4 *
Qian Bu等.CREB Signaling Is Involved in Rett Syndrome Pathogenesis.《The Journal of Neuroscience》.2017,第3671–3685页. *
基于CRISPR系统基因编辑与基因调控技术的发展;杨明辉等;《农业生物技术学报》;20180225(第02期);第183-193页 *

Also Published As

Publication number Publication date
CN109402115A (en) 2019-03-01

Similar Documents

Publication Publication Date Title
JP7239725B2 (en) CRISPR-Cas effector polypeptides and methods of use thereof
US20210340566A1 (en) Compositions and methods for differential cas9 gene labeling and/or editing
AU2015101418A4 (en) Compositions and methods of nucleic acid-targeting nucleic acids
EP2821413B2 (en) Nucleotide-specific recognition sequences for designer tal effectors
EP2984175B1 (en) Dna-guided dna interference by a prokaryotic argonaute
JP6626830B2 (en) Multiple transposase adapters for DNA manipulation
CN111328343A (en) RNA targeting methods and compositions
CN109295054B (en) gRNA for targeting pathogen gene RNA, detection method and kit for pathogen gene based on C2C2
KR20190059966A (en) S. The Piogenes CAS9 mutant gene and the polypeptide encoded thereby
Lampe et al. Targeted DNA integration in human cells without double-strand breaks using CRISPR-associated transposases
CN112823011A (en) Gene targets for T cell-based immunotherapy
JP2022547524A (en) Novel CRISPR DNA targeting enzymes and systems
CN111133100A (en) Multiplexed receptor-ligand interaction screening
US10801017B2 (en) Nucleotide-specific recognition sequences for designer TAL effectors
CN109402115B (en) gRNA of target Rett mutant gene RNA, detection method of Rett mutant gene and detection kit
CN109295055B (en) c2C 2-based gRNA of tumor related mutant gene, detection method and detection kit
JP2023547394A (en) Nucleic acid detection method by oligohybridization and PCR-based amplification
CN114391043B (en) Methylation detection and analysis of mammalian DNA
CN109957568B (en) gRNA for targeting HBB RNA, C2C 2-based HBB mutation detection method and detection kit
JP2022546594A (en) Novel CRISPR DNA targeting enzymes and systems
CN109897852A (en) The gRNA of tumour related mutation gene based on C2c2, detection method, detection kit
CN109913452A (en) For targeting the gRNA and the HBB mutation detection methods based on C2c2, detection kit of HBB RNA
JP7402453B2 (en) Methods of isolating or identifying cells and cell populations
Lampe et al. Targeted DNA integration in human cells without double-strand breaks using CRISPR RNA-guided transposases
CN114901303A (en) Modified endonucleases and related methods

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant