CN111718931B - Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell - Google Patents

Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell Download PDF

Info

Publication number
CN111718931B
CN111718931B CN202010553835.5A CN202010553835A CN111718931B CN 111718931 B CN111718931 B CN 111718931B CN 202010553835 A CN202010553835 A CN 202010553835A CN 111718931 B CN111718931 B CN 111718931B
Authority
CN
China
Prior art keywords
cell
gene
culture medium
protein
sequence
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
CN202010553835.5A
Other languages
Chinese (zh)
Other versions
CN111718931A (en
Inventor
陈宝惠
徐海月
邹炜
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.)
Zhejiang University ZJU
Original Assignee
Zhejiang University ZJU
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 Zhejiang University ZJU filed Critical Zhejiang University ZJU
Priority to CN202010553835.5A priority Critical patent/CN111718931B/en
Publication of CN111718931A publication Critical patent/CN111718931A/en
Application granted granted Critical
Publication of CN111718931B publication Critical patent/CN111718931B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/65Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression using markers
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/66General methods for inserting a gene into a vector to form a recombinant vector using cleavage and ligation; Use of non-functional linkers or adaptors, e.g. linkers containing the sequence for a restriction endonuclease
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention discloses a triple label and a method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell. The label comprises a CRISPR-Tag DNA sequence consisting of 12 TS1 repeated units, an MS2 DNA sequence consisting of 12 MS2V5 unit sequences and a blue fluorescent protein sequence containing an intron. The DNA sequence is short in length and can be inserted into the N end or the C end of the target protein coding gene in a CRISPR-Cas9 mediated gene recombination mode; the preparation method comprises the steps of firstly preparing a TriTag label, constructing a visual stable cell line, then constructing a visual system of a target endogenous gene, transfecting and sorting, and finally realizing dynamic imaging of living cells. The TriTag can be labeled, realizes the dynamic imaging of chromatin, can visually visualize the chromatin dynamics and gene expression of endogenous specific target genes, is favorable for the integration and analysis of data, and helps to analyze the dynamic regulation mechanism of gene expression.

Description

Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell
Technical Field
The invention relates to a method for establishing a TriTag marker imaging system in the field of biotechnology, in particular to a label and a method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell, and realizes real-time imaging and analysis of gene expression regulation.
Background
Gene expression is the process of transmitting genetic information from gene to protein, transcription and translation are two key steps of gene expression, and the whole process is tightly regulated. However, few studies have visualized the entire gene expression regulation process: that is, in living cells, DNA, mRNA and protein of the gene are visualized simultaneously, and the expression level of the gene is controlled at the chromatin level, so that the spatiotemporal control of the protein expression level is realized.
There has been much work done to visualize RNA in living cells. In these works, the MS2 phage capsid protein binding site (MS2) was localized to either the 5 'noncoding region or the 3' noncoding region of the gene, both locations enabling visualization of mature mRNA and nascent RNA simultaneously. However, work has shown that the insertion of the MS2 sequence into the 5' non-coding region of a gene affects gene expression to some extent. Therefore, in combination with the above two aspects, the prior art cannot intuitively and objectively capture the dynamic change of the gene transcription level in real time.
One published work has used exogenous reporter gene systems to visualize the entire gene expression regulation process in living cells: i.e. visualizing DNA, mRNA and protein of the gene simultaneously. The exogenous reporter gene is connected with 3 elements in series: (1)256 copies of LacO sequence to visualize the DNA of the gene; (2)24 copies of the MS2 sequence to visualize the mRNA; (3) cyan Fluorescent Protein (CFP) to visualize the protein of interest. This work visualizes both mature mRNA and nascent RNA, and therefore does not clearly capture the production of nascent RNA and quantitatively analyze transcriptional dynamics. In addition, the total length of DNA of 3 elements of the report system reaches 20kb, and the report system can only be used as an exogenous report gene, thereby limiting the application of the technology to a certain extent.
Therefore, the prior art lacks a living cell imaging technology which can be widely used and can realize the visualization of DNA, mRNA and protein of genes, and lacks a gene visualization tool which can better study the expression regulation of endogenous genes and provide powerful research work for disease pathology, regenerative medicine and the like.
Disclosure of Invention
In order to solve the bottlenecks and problems of the background art, the present invention aims to provide a label and an imaging method that can be widely used and can simultaneously visualize DNA, mRNA and protein of an endogenous gene. The invention is mainly applied to the visualization work of gene expression regulation.
Therefore, the technical scheme adopted by the invention is as follows:
a TriTag label for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell is composed of three parts of DNA sequences:
for gene visualizationThe CRISPR-Tag DNA sequence of (1), TS1 consisting of 12 TS1 repeating units12xA DNA sequence, such as SEQ ID No.1, as a CRISPR-Tag DNA sequence for assembling a Tritag label; the CRISPR-Tag DNA sequence for gene visualization is a living cell gene visualization label by combining dCas9-GFP14xThe fluorescent fusion protein realizes gene visualization.
The MS2 DNA sequence for RNA visualization consists of 12 MS2V5 unit sequences, such as SEQ ID No.2, as the MS2 DNA sequence for assembling a TriTag label;
the blue fluorescent protein sequence containing the intron, such as SEQ ID No.5, used for protein visualization is formed by inserting a segment of intron sequence, such as SEQ ID No.4, into the blue fluorescent protein sequence, such as SEQ ID No.3, and can visualize the nascent RNA generated by the target endogenous gene.
The TriTag label is a DNA/RNA double-visualized sequence formed by alternately inserting 12 TS1 repetitive unit sequences and 12 MS2V5 unit sequences into a sequence of SEQ ID No.6, and then the double-visualized sequence is inserted into an intron in a blue fluorescent protein sequence containing the intron by an enzyme digestion connection method to form the TriTag label of SEQ ID No. 7.
Secondly, a method for simultaneously visualizing DNA, mRNA and protein of genes in living cells:
(ii) (a) preparing a TriTag tag constructed according to any one of claims 1-2;
and (II) simultaneously visualizing the DNA, mRNA and protein of the target endogenous gene in the living cell by adopting the TriTag label.
(1) Constructing a visual stable cell line:
(1.1) on the day, culturing HEK293T cells by using a PS-free culture medium and paving the cells into a 12-well plate to form a first cell culture medium, wherein the HEK293T cells are added at a concentration which enables the cell density on the next day to be more than 80%;
(1.2) the following day, lentivirus packaging was performed: 750ng of each dCas9-GFP were pre-mixed with 75. mu.l of serum-reduced medium (Opti-medium)14xThe virus expression plasmid and stdMCP-tdTomato virus expression plasmid, 705ng pCMV-dR8.91 plasmid and 87ng PMD2.G plasmid were added, and 4.5. mu.l Fugene transientForming liposome by using a transfection reagent (Promega), dripping the liposome into the supernatant of a first cell culture medium, and performing transient transfection on HEK293T cells in the first cell culture medium to prepare cells containing virus particles;
(1.3) after 12 hours of transient transfection in step (1.2), aspirating the first cell culture medium supernatant and replacing the first cell culture medium with fresh medium;
(1.4) culturing the cells to be infected 48 hours after transient transfection in the step (1.2) and paving the cells to be infected into a 24-well plate to form a second cell culture medium, wherein the concentration of the cells to be infected can enable the next day cell density to reach about 60%;
(1.5) after 24 hours of forming the second cell culture medium, sucking the first cell culture medium supernatant obtained by replacing the fresh culture medium in the step (1.3), then placing the first cell culture medium supernatant into a centrifuge for 8 minutes at the rotating speed of 800g, taking the supernatant after centrifugation, and extracting the cell supernatant containing viruses;
(1.6) preparing a PS-free culture medium containing a gene transfection enhancer (polybrene) with the concentration of 5 mug/mL as a third culture medium to replace a second cell culture medium, so that the cells to be infected are cultured and transferred to the third culture medium, and then taking 30-90 mul of cell supernatant containing viruses to be dripped into the third culture medium supernatant of the cells to be infected to obtain infected cells; the cells to be infected are cells to be infected by virus, and are experimental samples in specific experiments, and human HeLa cells are adopted as the experimental samples in the experiments.
(1.7) after 12 hours of infection in the step (1.6), replacing a fresh culture medium for the third culture medium of the infected cells, continuing culturing until the third culture medium can be passaged to an 8-well plate for microscope imaging observation, screening a cell infected population according to the fluorescence intensity and the infection efficiency, and separating single cell clones of a 96-well plate;
the screening of the single cell clone specifically screens out cell infected groups with proper fluorescence intensity and higher infection efficiency: selecting cells in a single hole of the pore plate with the reserved fluorescence intensity smaller than the first fluorescence intensity threshold, and otherwise, removing the cells; and meanwhile, selecting the cells in a single hole of the pore plate with the reserved infection efficiency larger than the infection efficiency threshold, and otherwise, removing the cells. The infection efficiency threshold is typically 30-40%.
(1.8) when the cells in the single hole of the 96-hole plate grow to a cell group state, observing the fluorescence intensity under a microscope, and screening single-cell clone;
and specifically, screening the cells in the single hole of the pore plate with the retained fluorescence intensity larger than the second fluorescence intensity threshold, and otherwise, removing the cells.
(2) Constructing a target endogenous gene visualization system in a stable cell line, and carrying out transfection and sorting;
(3) simultaneously visualizing the DNA, mRNA and protein of the target endogenous gene and live cell dynamic imaging.
The step (one) is as follows:
s1, designing and constructing a blue fluorescent protein sequence containing an intron:
adopting the 4 th intron sequence of the human HSPA5 gene, adding BstXI restriction enzyme cutting sites in the intron sequence, and directly synthesizing by a nucleic acid synthesis mode;
s2, designing and synthesizing a DNA/RNA double visualization sequence:
alternately interleaving 12 TS1 repeating unit sequences and 12 MS2V5 unit sequences for 12 times by means of nucleic acid synthesis;
s3, building a TriTag label: and (3) inserting the DNA/RNA double-visual sequence into an intron of a blue protein sequence containing the intron by using an enzyme digestion connection method to obtain a TriTag label, and particularly inserting the TriTag label into a BstXI enzyme digestion site of the intron sequence. Under the condition that the TriTag label obtained by the assembly is fused and expressed with a target endogenous gene protein, the chromatin dynamics and the transcription level change of the target endogenous gene can be visualized.
The step (2) is specifically as follows:
(2.1) according to the condition of a target endogenous gene, designing and constructing a HDR repair template plasmid which is specific in gene and contains a TriTag label and an sgRNA expression plasmid for targeting the target endogenous gene to carry out knock-in editing of cells, and inserting the TriTag label into the N end or the C end of the target endogenous gene through knock-in editing;
(2.2) culturing the cell to be edited by using a PS-free culture medium and paving the cell to a 24-well plate to form a fourth cell culture medium, wherein the adding concentration of the cell can enable the cell density of the next day to reach more than 80%;
(2.3) on the day after 24 hours from the start of the culturing in step (2.2), premixing 500ng of sgRNA expression plasmid for targeting the target endogenous gene, 100ng of Cas9 protein expression plasmid and 400ng of HDR repair template plasmid with 40 μ l of serum-reduced medium (Opti-medium), adding 2.4 μ l of Fugene transient transfection reagent (Promega) to form liposomes, slowly adding dropwise to the supernatant of the fourth cell culture medium, and transiently transfecting the cells to be knottin-editing;
(2.4) performing cell flow sorting after the transient transfection for 96 hours, recovering the BFP fluorescence expression positive cells during the cell flow sorting, continuously culturing, and discarding the rest. I.e., cell retention in the cell channel that exhibits a fluorescent blue color.
The cell to be knock-in edited refers to a working cell of a visualization system.
The specific construction method and process of the HDR repair template plasmid are as follows:
(1) selecting the N end or the C end of the protein as an insertion site of a TriTag label through protein structure prediction;
(2) extracting a human HeLa cell genome, amplifying homologous arm sequences at two sides of an N end or a C end of a human target gene by using a PCR (polymerase chain reaction) method, namely a5 'homologous arm sequence and a 3' homologous arm sequence, wherein the length of the homologous arm sequences is about 500bp, and then inserting a TriTag label between the homologous arm sequences at two sides in a homologous recombination mode to form a tandem sequence of the 5 'homologous arm sequence-TriTag label-3' homologous arm sequence as an HDR (high-density lipoprotein lipase) repair template plasmid. The result after preparation is shown in SEQ ID No. 8.
The step (3) is specifically as follows:
(3.1) culturing the cells obtained by sorting in the step (2) by using a PS-free culture medium and paving the cells into an 8-well plate to form a fifth cell culture medium, wherein the adding concentration of the cells can enable the cell density of the next day to reach about 60%;
(3.2) on the day after 24 hours from the start of the culture in step (3.1), 500ng of TS1-sgRNA expression plasmid was premixed with 25. mu.l of serum-reduced medium (Opti-medium), and after liposome formation with the addition of 1.2. mu.l of Fugene transient transfection reagent (Promega), cells were transiently transfected by slowly dropping into the fifth cell culture supernatant;
(3.3) after 24 hours from the completion of step (3.2), the cells obtained in step (3.2) were placed in a microscope, the microscope was adjusted to specific environmental conditions, and the imaging parameters of the microscope were set for live cell imaging visualization.
In a specific implementation, the microscope live cell workstation settings were adjusted to a temperature of 37 ℃ and contained 5% by volume CO2
The microscope comprises a microscope living cell workstation, a microscope light source and an imaging camera, the microscope living cell workstation is adjusted to a specific environmental condition, and living cell imaging visualization is carried out by setting and adjusting the laser intensity of the microscope light source and the exposure time of the imaging camera.
In the step (3.3), the DNA of the target endogenous gene itself is displayed in green, mRNA produced by transcription of the target endogenous gene is displayed in red, and protein produced by translation of the target endogenous gene is displayed in blue.
Therefore, the invention can observe the transcriptional dynamic regulation of the gene in real time in a living cell state through the fusion of the TriTag label and the target endogenous gene.
The DNA sequence of the tag of the invention is about 1.5kb in length, and can be inserted into the N-terminal or C-terminal of a specific protein encoding gene through a CRISPR-Cas9 mediated gene recombination mode. The label is small and relatively easy to handle. At the DNA level, TriTag can be labeled with dCas9-GFP14x, thereby enabling dynamic imaging of chromatin. The target gene is transcribed into new mRNA, and can be recorded in real time through an MCP-FP system; mRNA is translated into fluorescent fusion protein, so that subcellular localization imaging of the target protein is facilitated. Therefore, the invention can visually visualize the chromatin dynamics and the gene expression of the endogenous specific target gene, is beneficial to the integration and analysis of data and helps to analyze the dynamic regulation and control mechanism of the gene expression.
Compared with the prior art, the invention has the beneficial effects that:
1. the TriTag label with shorter length is beneficial to the occurrence of a knock-in editing process mediated by a CRISPR-Cas9 system, so that the visualization of DNA, mRNA and protein of an endogenous gene is realized, an exogenous reporter gene system is developed to the endogenous gene, and the application range of a visualization system is expanded;
2. after the intron of the nascent RNA is removed by an RNA shearing mechanism, mature mRNA is formed by a subsequent processing process. The MS2 DNA sequence is placed in an intron of TriTag, so that the nascent RNA can be visualized intuitively, and the quantification of transcription level change is more accurate;
3. the design and use strategy of the TriTag label does not obviously influence the expression of the protein of the gene product, so that the gene regulation process can be visualized under the condition of not influencing the normal expression of the gene;
4. the DNA, mRNA and protein of the gene are visualized simultaneously, the gene expression regulation and control process can be observed and researched more carefully, and an important technical method is established for analyzing the relationship between chromatin dynamics and gene expression regulation and control.
Drawings
Fig. 1 is a schematic diagram of the design and construction of a TriTag tag. Subfigure 1(a) inserting the 4 th intron sequence of the human HSPA5 gene into the blue fluorescent protein sequence by nucleic acid synthesis to obtain the blue fluorescent protein sequence containing intron for protein visualization; subgraph 1(b) the TS112x sequence for gene DNA visualization and the MS2V512x sequence for RNA visualization were assembled into a DNA/RNA double visualization sequence by nucleic acid synthesis; subgraph 1(c) inserts a DNA/RNA double visualization sequence into an intron region in a blue fluorescent protein sequence containing an intron by enzyme digestion ligation to obtain a TriTag tag.
FIG. 2 is a technical route for simultaneously visualizing target endogenous gene DNA, mRNA and protein in living cells by using TriTag tags. Sub-figure 2(a) obtains virus particles in HEK293T cells by lentivirus packaging, and then obtains a stable cell line capable of expressing dCas9-GFP14x and stdMCP-tdTomato by lentivirus infection and screening of single cell clones; FIG. 2(b) is a schematic diagram of HDR repair template plasmid containing TriTag tag; sub-graph 2(c) constructs a target endogenous gene visualization system in a stable cell line by knock-in editing and cell flow sorting, namely positive cells obtained after TriTag label knock-in editing is carried out on the target endogenous gene; panel 2(d) transient transfection and microscopic imaging of the TS1-sgRNA expression plasmid, while visualizing DNA, mRNA and protein of the endogenous gene of interest and live cell dynamic imaging.
Fig. 3 is a result of simultaneously visualizing DNA, mRNA, and protein of the human histone H2B gene and the nuclear membrane protein LMNA gene using a visualization system. FIG. 3(a) is a schematic diagram of HDR repair template containing TriTag label, specific to H2B gene and LMNA gene; subgraph 3(b) is the visualization result of two genes; FIG. 3(c), is a diagram showing the co-localization analysis of mRNA (red) and gene DNA (green) signals in FIG. 3 (b); panel (d) is a cell flow analysis of protein expression in two gene-visualized cell lines.
Fig. 4 is an analysis of the transcriptional burst pattern of the human histone H2B gene and the nuclear membrane protein LMNA gene using the TriTag system, and an example graph is drawn for both transcriptional patterns. FIG. 4(a) shows the result of mRNA imaging of the human histone H2B gene and the nuclear membrane protein LMNA gene.
FIG. 5 is a TriTag system used for simultaneously visualizing human nuclear membrane protein LMNA gene DNA, mRNA and protein, and then performing correlation analysis on transcription level and movement state of chromatin region where gene locus is located. Subgraph 5(a) is the visualization result of the human nuclear membrane protein LMNA gene; sub-diagram 5(b) is the motion trail of two gene loci obtained after a pair of LMNA allelic gene loci at different transcription levels are tracked for a short time; sub-graph 5(c) is two MSD curves obtained after analyzing the motion trajectories of a plurality of pairs of LMNA allelic loci under different transcription levels; subfigure 5(d) is two parameters obtained after performing motion state analysis on multiple pairs of LMNA allelic sites at different transcription levels: comparative analysis of the macroscopic diffusion coefficient (D) with the confined area (A).
FIG. 6 is a TriTag system used to visualize DNA, mRNA and protein of the human heat shock protein HSPA1A gene and to correlate the transcription level with the open status of the chromatin region in which the locus is located. FIG. 6(a) is a schematic diagram of a specific HDR repair template containing a TriTag tag of the HSPA1A gene; FIG. 6(b) is the result of visualizing the human heat shock protein HSPA1A gene at different heat shock durations of 42 ℃; panel 6(c) is a statistic of the fluorescence intensity of the mRNA (red) signal and the area of the DNA (green) signal at the gene locus generated by the human heat shock protein HSPA1A gene at different heat shock durations of 42 ℃.
Detailed Description
The invention is further illustrated by the following figures and examples.
The examples of the invention are as follows:
example 1:
s1, designing and constructing a blue fluorescent protein sequence containing an intron:
extracting human HeLa cell genome, and obtaining a blue fluorescent protein sequence containing an intron by adopting a nucleic acid synthesis mode, as shown in figure 1 (a);
s2, TS1 screened from nematode genome capable of being efficiently targeted and edited12xMS2V5 with reduced sequence and repeatability12xThe sequence is formed by the way of nucleic acid synthesis, and the TS1 repeating unit and the MS2V5 repeating unit are alternately connected with each other to form a DNA/RNA double visualization sequence, as shown in figure 1 (b);
s3, building a TriTag label: the double-visualized sequence is inserted into the intron region in the blue fluorescent protein sequence containing the intron by using an enzyme digestion ligation method to obtain a TriTag tag, as shown in FIG. 1(c), specifically inserted into a BstxI restriction enzyme site of the intron sequence.
Example 2
(1) Constructing a visual stable cell line as shown in fig. 2 (a):
(1.1) on the day, culturing HEK293T cells by using a PS-free culture medium and paving the cells into a 12-well plate to form a first cell culture medium, wherein the HEK293T cells are added at a concentration which enables the cell density on the next day to be more than 80%;
(1.2) the following day, lentivirus packaging was performed: 750ng of each dCas9-GFP were pre-mixed with 75. mu.l of serum-reduced medium (Opti-medium)14xAfter the virus expression plasmid and stdMCP-tdTomato virus expression plasmid, 705ng of pCMV-dR8.91 plasmid and 87ng of PMD2.G plasmid, 4.5. mu.l of plasmid was addedForming a liposome by a Fugene transient transfection reagent (Promega), dripping the liposome into the supernatant of a first cell culture medium, and performing transient transfection on HEK293T cells in the first cell culture medium to prepare cells containing virus particles;
(1.3) after 12 hours of transient transfection in step (1.2), aspirating the first cell culture medium supernatant and replacing the first cell culture medium with fresh medium;
(1.4) culturing the cells to be infected 48 hours after transient transfection in the step (1.2) and paving the cells to be infected into a 24-well plate to form a second cell culture medium, wherein the concentration of the cells to be infected can enable the next day cell density to reach about 60%;
(1.5) on the day after 24 hours of forming the second cell culture medium, sucking the first cell culture medium supernatant obtained by replacing the fresh culture medium in the step (1.3), then placing the first cell culture medium supernatant into a centrifuge for 8 minutes at the rotating speed of 800g, taking the supernatant after centrifugation, and extracting the cell supernatant containing the virus;
(1.6) preparing a PS-free culture medium containing a gene transfection enhancer (polybrene) with the concentration of 5 mu g/mL as a third culture medium to replace a second cell culture medium, so that a cell to be infected by adopting a human HeLa cell is cultured and transferred to the third culture medium, and then 30 mu l-90 mu l of cell supernatant containing the virus is taken and dripped into the third culture medium supernatant of the cell to be infected to obtain an infected cell;
(1.7) after 12 hours of infection in the step (1.6), replacing a fresh culture medium for the third culture medium of the infected cells, continuing culturing until the third culture medium can be passaged to an 8-well plate for microscope imaging observation, screening a cell infected population according to the fluorescence intensity and the infection efficiency, and separating single cell clones of a 96-well plate;
the screening of the single cell clone specifically screens out cell infected groups with proper fluorescence intensity and higher infection efficiency: selecting cells in a single hole of the pore plate with the reserved fluorescence intensity smaller than the first fluorescence intensity threshold, and otherwise, removing the cells; meanwhile, selecting the cells in a single hole of the pore plate with the preserved infection efficiency of more than 30-40%, and otherwise, removing the cells.
(1.8) when the cells in the single well of the 96-well plate grow to the state of cell groups, observing the fluorescence intensity under a microscope, and screening the single-cell clone.
And specifically, screening the cells in the single hole of the pore plate with the retained fluorescence intensity larger than the second fluorescence intensity threshold, and otherwise, removing the cells.
(2) Constructing a target endogenous gene visualization system in a stable cell line, and carrying out transfection and sorting, as shown in FIG. 2 (c):
(2.1) according to the condition of a target endogenous gene, designing and constructing a HDR repair template plasmid which is specific in gene and contains a TriTag label and an sgRNA expression plasmid for targeting the target endogenous gene to carry out knock-in editing of cells, and inserting the TriTag label into the N end or the C end of the target endogenous gene through knock-in editing;
(2.2) culturing the cell to be edited by using a PS-free culture medium and paving the cell to a 24-well plate to form a fourth cell culture medium, wherein the adding concentration of the cell can enable the cell density of the next day to reach more than 80%;
(2.3) on the day 12 hours after the start of the culture in step (2.2), premixing 500ng of sgRNA expression plasmid for targeting the target endogenous gene, 100ng of Cas9 protein expression plasmid and 400ng of HDR repair template plasmid with 40 μ l of serum-reduced medium (Opti-medium), adding 2.4 μ l of Fugene transient transfection reagent (Promega) to form liposomes, slowly adding dropwise to the supernatant of the fourth cell culture medium, and transiently transfecting the cells to be knottin-editing;
(2.4) performing cell flow sorting after the transient transfection for 96 hours, recovering the BFP fluorescence expression positive cells during the cell flow sorting, continuously culturing, and discarding the rest. I.e., cell retention in the cell channel that exhibits a fluorescent blue color.
The cells to be knotted-in edited in this example were visualized cell lines stably expressing dCas9-GFP14x and stdMCP-tdTomato obtained by screening.
The specific construction method and process of the HDR repair template plasmid are as shown in FIG. 2 (b): and selecting the N end or the C end of the protein as an insertion site of the TriTag label through protein structure prediction.
Extracting human HeLa cell genome, and amplifying homologous arm sequences (namely 5 'homologous arm sequence and 3' homologous arm sequence) at two sides of N end (or C end) of a human target gene by using a PCR method, wherein the length of the homologous arm sequences is about 500 bp. And then inserting the TriTag tag between the homologous arm sequences on two sides by adopting a homologous recombination mode to form a series sequence of a5 'homologous arm sequence-TriTag tag-3' homologous arm sequence.
The result after preparation is shown in SEQ ID No. 8.
(3) Simultaneously visualizing DNA, mRNA and protein of the target endogenous gene and performing live cell dynamic imaging, as shown in fig. 2 (d):
(3.1) culturing the cells obtained by sorting in the step (2) by using a PS-free culture medium and paving the cells into an 8-well plate to form a fifth cell culture medium, wherein the adding concentration of the cells can enable the cell density of the next day to reach about 60%;
(3.2) on the day after 24 hours from the start of the culture in step (3.1), 500ng of TS1-sgRNA expression plasmid was premixed with 25. mu.l of serum-reduced medium (Opti-medium), and after liposome formation with the addition of 1.2. mu.l of Fugene transient transfection reagent (Promega), cells were transiently transfected by slowly dropping into the fifth cell culture supernatant;
(3.3) on the day 24 hours after completion of step (3.2), the cells obtained in step (3.2) were placed in a live microscope cell station set to a temperature of 37 ℃ and containing a volume fraction of 5% CO2
And the microscope also comprises a microscope light source and an imaging camera, and live cell imaging visualization is carried out by setting and adjusting the laser intensity of the microscope light source and the exposure time of the imaging camera.
Finally, the target endogenous gene DNA itself appears green, mRNA produced by the target endogenous gene appears red, and protein produced by the target endogenous gene appears blue.
Therefore, the invention can observe the transcriptional dynamic regulation of the gene in real time in a living cell state through the fusion of the TriTag label and the target endogenous gene.
Example 3
(1) Constructing a target endogenous gene (human histone H2B gene and nuclear membrane protein LMNA gene) visual system in a stable cell line, and carrying out transfection and sorting:
(1.1) according to the protein structure prediction of target endogenous gene products H2B histone and LMNA nuclear membrane protein, inserting a TriTag label into the C end of human histone H2B gene, and inserting the TriTag label into the N end of human nuclear membrane protein LMNA gene. Extracting human HeLa cell genome, respectively amplifying homologous arm sequences (5 'homologous arm sequence and 3' homologous arm sequence) at both sides of C end of H2B gene or N end of LMNA gene, wherein the length of the homologous arm sequences is about 500 bp. And constructing an HDR repair template of the 5 'homologous arm sequence-TriTag tag-3' homologous arm sequence by adopting a homologous recombination mode. As shown in fig. 3 (a).
(1.2) Tritag tag knock-in editing was performed on the human histone H2B gene and the nuclear membrane protein LMNA gene as in the steps (2.2) to (2.4) in example 2, and BFP fluorescent protein expression positive cells were sorted by cell flow.
(2) Simultaneously visualizing the DNA, mRNA and protein of the target endogenous gene and performing dynamic imaging of living cells:
(2.1) culturing and paving the cells (positive cells obtained after Tritag label knock-in editing of human histone H2B gene and nuclear membrane protein LMNA gene) obtained by sorting in the step (1) into an 8-hole plate by using a PS-free culture medium to form a fifth cell culture medium, wherein the adding concentration of the cells can enable the cell density of the next day to reach about 60%;
(2.2) on the day after 24 hours from the start of the culture in step (2.1), 500ng of TS1-sgRNA expression plasmid was premixed with 25. mu.l of serum-reduced medium (Opti-medium), and after liposome formation with the addition of 1.2. mu.l of Fugene transient transfection reagent (Promega), cells were transiently transfected by slowly dropping into the fifth cell culture supernatant;
(2.3) on the day 24 hours after completion of step (2.2), the cells obtained in step (2.2) were placed in a live microscope cell station set to a temperature of 37 ℃ and containing a volume fraction of 5% CO2. For the human histone H2B gene, 561 laser intensity was set to 10%, exposure time was 100ms, 488 laser intensity was set to 20%, exposure time was 100ms, 405 laser intensity was set to 10%, exposure time was 100 ms; for the human nuclear membrane protein LMNA gene, 561 laser intensity is set as 10%, and exposure time isThe laser intensity was set at 20% for 100ms and 488, the exposure time was 100ms, the laser intensity was set at 10% for 405, and the exposure time was 100 ms. The results are shown in FIG. 3 (b). In the figure, the DNA of the target endogenous gene itself is shown in green, the mRNA produced by the target endogenous gene is shown in red, and the protein produced by the target endogenous gene is shown in blue.
(2.4) the co-localization analysis of the DNA and mRNA signals of the endogenous gene of the cell shown in FIG. 3(b) was carried out by ImageJ analysis software, and the results are shown in FIG. 3 (c). The figure shows that the green color displayed by DNA can co-localize with the red color displayed by mRNA: the fluorescence intensity peaks at the same position.
(3) The visualization system establishes comparative analysis of gene protein expression conditions in cells:
and (3) collecting the cells obtained by sorting in the step (1) (positive cells obtained after Tritag label knock-in editing of human histone H2B gene and nuclear membrane protein LMNA gene) and performing flow cytometry analysis, as shown in figure 3 (d). The figure shows that the insertion of TriTag has no influence on gene expression.
Therefore, the invention can observe the transcriptional dynamic regulation of the gene in real time under the condition of not influencing the gene expression by fusing the TriTag label and the target endogenous gene.
Example 4
(1) Visualization of target endogenous gene mRNA and live cell dynamic imaging:
(1.1) culturing the positive cells obtained by sorting in the step (1) in the above example 3, paving the cells on an 8-well plate, and paving the plates the next day to obtain dynamic images of living cells for a long time so as to visualize the mRNA production and change conditions of two genes in the long time: the microscope live cell workstation settings were adjusted to a temperature of 37 ℃ and to contain 5% by volume of CO2(ii) a For human histone H2B gene, 561 laser intensity is set as 10%, exposure time is 100ms, total shooting time is 2 hours, and shooting interval is 1 minute; for the human nuclear membrane protein LMNA gene, 561 laser intensity is set as 10%, exposure time is 100ms, total shooting time is 3 hours, and shooting interval is 2 minutes.
(1.2) As shown in FIG. 4(a), the fluorescence intensity of the mRNA signal at each time point in the long-term imaging process is plotted as a line graph which changes with time, and thus the transcription dynamics of the two genes can be quantified. As shown, both genes, although housekeeping, are transcribed in an intermittent burst mode: i.e., alternating transcription-non-transcription patterns.
The result shows that the TriTag visualization system can realize simultaneous marking of the DNA, mRNA and protein of the endogenous gene without influencing the gene expression, can visually present the transcription dynamics of the endogenous gene, and is more favorable for analyzing the transcription burst mode of the gene and the transcription regulation mechanism of the gene.
Example 5
(1) A visual system of an endogenous gene of interest (human nuclear membrane protein LMNA gene) was constructed in a stable cell line and sorted by transfection as in (1) in example 3.
(2) Simultaneously visualizing target endogenous gene DNA, mRNA and protein and performing live cell dynamic imaging:
(2.1) culturing the cells obtained by sorting in the step (1) (positive cells obtained after Tritag label knock-in editing of human nuclear membrane protein LMNA genes) by using a PS-free culture medium and paving the cells into an 8-hole plate to form a fifth cell culture medium, wherein the addition concentration of the cells can enable the cell density of the next day to reach about 60%;
(2.2) on the day after 24 hours from the start of the culture in step (2.1), 500ng of TS1-sgRNA expression plasmid was premixed with 25. mu.l of serum-reduced medium (Opti-medium), and after liposome formation with the addition of 1.2. mu.l of Fugene transient transfection reagent (Promega), cells were transiently transfected by slowly dropping into the fifth cell culture supernatant;
(2.3) on the day 24 hours after completion of step (2.2), the cells obtained in step (2.2) were placed in a live microscope cell station set to a temperature of 37 ℃ and containing a volume fraction of 5% CO2. For the human nuclear membrane protein LMNA gene, 561 laser intensity was set to 10%, exposure time was 100ms, 488 laser intensity was set to 20%, exposure time was 100ms, 405 laser intensity was set to 10%, and exposure time was 100 ms. The results are shown in FIG. 5 (a). It is shown that in this cell, the two allelic positions of the human nuclear membrane protein LMNA geneThe dots are all visualized and displayed as green; the nascent RNA produced by the LMNA gene is shown in red, the two allelic loci are shown in different transcription states (i.e., not transcribed; ii.e., transcribed), and the nuclear membrane protein produced by the LMNA gene is shown in blue.
(2.4) setting the 488 laser intensity as 40%, setting the exposure time as 100ms, turning off 561, 405 laser light source, and shooting two allelic loci of the cell human nuclear membrane protein LMNA gene in short time and without interval: the total shooting time is 1 minute and 33 seconds, the shooting interval is 0.25 seconds, and 400 frames are shot in total. The green signals displayed by the two allelic loci are tracked and analyzed by an Object Tracking module of CellSens software (Olympus), and the locus motion tracks under different transcription states are obtained. As shown in fig. 5(b), the locus of the gene in the transcribed state has a compact motion trajectory and a small motion range compared to the untranscribed locus.
(2.5) tracking the human nuclear membrane protein LMNA gene allelic locus in a plurality of cells by the imaging and analyzing method to 44 pairs of allelic loci, analyzing the obtained plurality of tracks in MatLab software to obtain two Mean Square Displacement (MSD) curves in the transcription and non-transcription states, and comparing with the untranscribed loci, the locus in the transcription state has slower movement speed and smaller movement range as shown in figure 5 (c).
(2.6) analyzing the resulting plurality of traces in MatLab software by fitting the following functions:
MSD(t)=A(1-e-t/τ)+4Dmacrot+v2t2
wherein MSD (t) represents mean square displacement, A represents the area size of the locus movement track, t represents time difference, tau represents the characteristic time constant of microscopic diffusion, and DmacroRepresents the maximum diffusion coefficient, v represents the speed of movement of the locus, and e represents the base of the natural logarithm.
Obtaining the parameters related to the movement of the gene locus under two states of transcription and non-transcription: macroscopic diffusion coefficient (D)macro) Numerical distribution with restricted area (A), as shown in FIG. 5(d), compared to untranscribedThe gene locus in the transcription state has slow motion speed and small motion range.
The result shows that the movement state of the chromatin region where the gene locus in different transcription states is located can be observed through a TriTag visualization system, so that observation and analysis of the correlation between chromatin dynamics and transcription regulation are enriched, and the regulation mechanism of chromatin space-time dynamics change on gene expression is further understood.
Example 6
(1) Constructing a target endogenous gene (human heat shock protein HSPA1A gene) visualization system in a stable cell line and carrying out transfection and sorting:
(1.1) As shown in FIG. 6(a), the TriTag tag is selectively inserted into the C-terminal of the human heat shock protein HSPA1A gene according to the structural prediction of the target endogenous gene product HSPA1A heat shock protein. Extracting a human HeLa cell genome, respectively amplifying homologous arm sequences (5 'homologous arm sequence and 3' homologous arm sequence) on two sides of the C end of the human heat shock protein gene HSPA1A gene, and constructing an HDR repair template of the 5 'homologous arm sequence-TriTag tag-3' homologous arm sequence by adopting a homologous recombination mode.
(1.2) TriTag tag knock-in editing of the human heat shock protein HSPA1A gene was performed as in step (2) in example 2, and positive cells expressing BFP fluorescent protein were sorted by cell flow.
(2) Simultaneously visualizing target endogenous gene DNA, mRNA and protein and performing live cell dynamic imaging:
(2.1) culturing and paving the cells (positive cells obtained after Tritag label knock-in editing of human heat shock protein HSPA1A gene) obtained by sorting in the step (1) into an 8-hole plate by using a PS-free culture medium to form a fifth cell culture medium, wherein the addition concentration of the cells can enable the cell density of the next day to reach about 60%;
(2.2) on the day after 24 hours from the start of the culture in step (2.1), 500ng of TS1-sgRNA expression plasmid was premixed with 25. mu.l of serum-reduced medium (Opti-medium), and after liposome formation with the addition of 1.2. mu.l of Fugene transient transfection reagent (Promega), cells were transiently transfected by slowly dropping into the fifth cell culture supernatant;
(2.3) completion of step (2.2)The day 24 hours later, the cells obtained in step (2.2) were pretreated in a 42 ℃ water bath for 30 minutes, and then placed in a microscope live cell workstation, which was set to a temperature of 42 ℃ and CO was turned off2The supply of (2). For the human heat shock protein HSPA1A gene, 561 laser intensity was set to 10%, exposure time was 100ms, 488 laser intensity was 20%, exposure time was 100ms, 405 laser intensity was 10%, and exposure time was 100 ms. Carrying out long-time visualization: the total photographing time was 4 hours, and the photographing interval was 20 minutes. The results are shown in FIG. 6 (b). In this cell, the DNA of the human heat shock protein HSPA1A gene is shown in green, the nascent RNA produced by the HSPA1A gene is shown in red, and the HSPA1A heat shock protein produced by the HSPA1A gene is shown in blue. During the heat shock, the red signal is increased from zero to zero, and the green signal is changed from compact to loose.
(2.4) a total of 3 cells were followed by the above imaging and analysis method, and time-dependent line graphs were made for both the fluorescence intensity of the red signal visualized by the nascent RNA and the area of the green signal visualized by the gene DNA at 5 time points of 0 hour, 1 hour, 2 hours, 3 hours and 4 hours of the heat shock process. As shown in fig. 6 (c). The line chart shows that the fluorescence intensity of the red signal representing the new RNA is increased from zero to zero in the heat shock process, and the area of the green signal representing the chromatin DNA is increased from small to large, which indicates the opening of the chromatin structure state.
The result shows that the structure openness degree of the chromatin region where the gene locus is located can be directly observed when the gene locus is in different transcription levels through a TriTag visualization system, and can be associated with the corresponding transcription levels, so that the transcription regulation and control can be more carefully, comprehensively and deeply analyzed.
The nucleotide and amino acid sequences involved in the invention are as follows:
SEQ ID No.1:
name: CRISPR-Tag DNA sequence
The source is as follows: artificial Sequence (Artificial Sequence)
gcaccgatgctctccgaggaggcgtgaccgcaccgatgctctccgaggaggagcaagcaccgatgctctccga ggaggcagctgtgcaccgatgctctccgaggaggcacgggctaattcactcccaacgagcaagcaccgatgctctccga ggaggcgtgaccgcaccgatgctctccgaggaggagcaagcaccgatgctctccgaggaggcagctgtgcaccgatg ctctccgaggaggcacccttgatctgtggatctaccagaacgcaccgatgctctccgaggaggcgtgaccgcaccgatg ctctccgaggaggagcaagcaccgatgctctccgaggaggcagctgtgcaccgatgctctccgaggaggcac
SEQ ID No.2:
Name: MS2 DNA sequence
The source is as follows: artificial Sequence (Artificial Sequence)
ccggtaacctacaaacgggtggaggatcaccccacccgacacttcacaatcaaggggtacaatacacaagggtg gaggaacaccccaccctccagacacattacacagaaatccaatcaaacagaagcaccatcagggcttctgctaccaaattt atctcaaaaaactacaacaaggaatcaccatcagggattccctgtgcaatatacgtcaaacgagggccacgacgggagg acgatcacgcctcccgaatatcggcatgtctggctttcgaattcagtgcgtggagcatcagcccacgcagccaatcagagt cgaatacaagtcgactttcgcgaagagcatcagccttcgcgccattcttacacaaaccacactctcccctacaggaacagc atcagcgttcctgcccagtacccaactcaagaaaatttatgtccccatgcagcatcagcgcatgggccccaagaatacatc cccaacaaaatcacatccgagcaccaacagggctcggagtgttgtttcttgtccaactggacaaaccctccatggaccatc aggccatggactctcaccaacaagacaaaaactactcttctcgaagcagcatcagcgcttcgaaacactcgagcatacatt gtgcctatttcttgggtggac
SEQ ID No.3:
Name: blue fluorescent protein sequence
The source is as follows: artificial Sequence (Artificial Sequence)
Atgagcgagctgattaaggagaacatgcacatgaagctgtacatggagggcaccgtggacaaccatcacttcaa gtgcacatccgagggcgaaggcaagccctacgagggcacccagaccatgagaatcaaggtggtcgagggcggccctc tccccttcgccttcgacatcctggctactagcttcctctacggcagcaagaccttcatcaaccacacccagggcatccccga cttcttcaagcagtccttccctgagggcttcacatgggagagagtcaccacatacgaagacgggggcgtgctgaccgcta cccaggacaccagcctccaggacggctgcctcatctacaacgtcaagatcagaggggtgaacttcacatccaacggccc tgtgatgcagaagaaaacactcggctgggaggccttcaccgagacgctgtaccccgctgacggcggcctggaaggcag aaacgacatggccctgaagctcgtgggcgggagccatctgatcgcaaacatcaagaccacatatagatccaagaaaccc gctaagaacctcaagatgcctggcgtctactatgtggactacagactggaaagaatcaaggaggccaacaacgagacct acgtcgagcagcacgaggtggcagtggccagatactgcgacctccctagcaaactggggcacaagcttaattaa
SEQ ID No.4:
Name: intron sequence
The source is as follows: artificial Sequence (Artificial Sequence)
gtaagtatgaaattcagggatacggccaccttgttggcatatttgccaaatagtggaaatgtgaagtactgacaaaac ttttccctttttcaatctaatag
SEQ ID No.5:
Name: intron-containing blue fluorescent protein sequence
The source is as follows: artificial Sequence (Artificial Sequence)
atgagcgagctgattaaggagaacatgcacatgaagctgtacatggagggcaccgtggacaaccatcacttcaag tgcacatccgagggcgaaggcaagccctacgagggcacccagaccatgagaatcaaggtggtcgagggcggccctct ccccttcgccttcgacatcctggctactagcttcctctacggcagcaagaccttcatcaaccacacccagggcatccccga cttcttcaagcagtccttccctgagggcttcacatgggagagagtcaccacatacgaagacgggggcgtgctgaccgcta cccaggacaccagcctccaggacgtaagtatgaaattcagggatacggccaccttgttggcatatttgccaaatagtggaa atgtgaagtactgacaaaacttttccctttttcaatctaatagggctgcctcatctacaacgtcaagatcagaggggtgaactt cacatccaacggccctgtgatgcagaagaaaacactcggctgggaggccttcaccgagacgctgtaccccgctgacgg cggcctggaaggcagaaacgacatggccctgaagctcgtgggcgggagccatctgatcgcaaacatcaagaccacata tagatccaagaaacccgctaagaacctcaagatgcctggcgtctactatgtggactacagactggaaagaatcaaggagg ccaacaacgagacctacgtcgagcagcacgaggtggcagtggccagatactgcgacctccctagcaaactggggcaca agcttaattaagcggccgcgactctag
SEQ ID No.6:
Name: DNA/RNA double visualization sequence
The source is as follows: artificial Sequence (Artificial Sequence)
ccggtaacctacaaacgggtggaggatcaccccacccgacacgcaccgatgctctccgaggaggcacaagggt ggaggaacaccccaccctccaggcaccgatgctctccgaggaggcaaacagaagcaccatcagggcttctgctacgca ccgatgctctccgaggaggaacaaggaatcaccatcagggattccctgtggcaccgatgctctccgaggaggacgacg ggaggacgatcacgcctcccgaatagcaccgatgctctccgaggaggtcagtgcgtggagcatcagcccacgcagcca gcaccgatgctctccgaggaggctttcgcgaagagcatcagccttcgcgccatgcaccgatgctctccgaggaggccta caggaacagcatcagcgttcctgcccagcaccgatgctctccgaggaggtgtccccatgcagcatcagcgcatgggccc cgcaccgatgctctccgaggaggcacatccgagcaccaacagggctcggagtgtgcaccgatgctctccgaggaggac cctccatggaccatcaggccatggactctgcaccgatgctctccgaggaggcttctcgaagcagcatcagcgcttcgaaa cagcaccgatgctctccgaggaggttct
SEQ ID No.7:
Name: TriTag label
The source is as follows: artificial Sequence (Artificial Sequence)
atgagcgagctgattaaggagaacatgcacatgaagctgtacatggagggcaccgtggacaaccatcacttcaag tgcacatccgagggcgaaggcaagccctacgagggcacccagaccatgagaatcaaggtggtcgagggcggccctct ccccttcgccttcgacatcctggctactagcttcctctacggcagcaagaccttcatcaaccacacccagggcatccccga cttcttcaagcagtccttccctgagggcttcacatgggagagagtcaccacatacgaagacgggggcgtgctgaccgcta cccaggacaccagcctccaggacgtaagtatgaaattcagggatacggccaccttgttggccggtaacctacaaacgggt ggaggatcaccccacccgacacgcaccgatgctctccgaggaggcacaagggtggaggaacaccccaccctccaggc accgatgctctccgaggaggcaaacagaagcaccatcagggcttctgctacgcaccgatgctctccgaggaggaacaa ggaatcaccatcagggattccctgtggcaccgatgctctccgaggaggacgacgggaggacgatcacgcctcccgaat agcaccgatgctctccgaggaggtcagtgcgtggagcatcagcccacgcagccagcaccgatgctctccgaggaggct ttcgcgaagagcatcagccttcgcgccatgcaccgatgctctccgaggaggcctacaggaacagcatcagcgttcctgcc cagcaccgatgctctccgaggaggtgtccccatgcagcatcagcgcatgggccccgcaccgatgctctccgaggaggc acatccgagcaccaacagggctcggagtgtgcaccgatgctctccgaggaggaccctccatggaccatcaggccatgg actctgcaccgatgctctccgaggaggcttctcgaagcagcatcagcgcttcgaaacagcaccgatgctctccgaggag gttctccaccttgttggcatatttgccaaatagtggaaatgtgaagtactgacaaaacttttccctttttcaatctaatagggctg cctcatctacaacgtcaagatcagaggggtgaacttcacatccaacggccctgtgatgcagaagaaaacactcggctggg aggccttcaccgagacgctgtaccccgctgacggcggcctggaaggcagaaacgacatggccctgaagctcgtgggc gggagccatctgatcgcaaacatcaagaccacatatagatccaagaaacccgctaagaacctcaagatgcctggcgtcta ctatgtggactacagactggaaagaatcaaggaggccaacaacgagacctacgtcgagcagcacgaggtggcagtggc cagatactgcgacctccctagcaaactggggcacaagcttaattaa
SEQ ID No.8:
Name: HDR repair template plasmid sequences
The source is as follows: artificial Sequence (Artificial Sequence)
gaggctgtcccaggctggatcacggccgcgactgcgagggacgccaaggagtcctctttcccctggccgcagg cgtgcgccttcgcagcccatctggcaccgcagatgccggtcgccggggtcagcggcgaagcccgaacccatgcggcc gggaggggacgcccacaaacccctgcccgccggccgggaagcgctcctacgggaccccagccccgttggggaggc ggggcggccacgcgggttccaccaatcagcggccaatgttcgaattcgcgtcctggcgcggccaatggcgggtcccgg agcaggcggggagggcgtggagggcggtaaatgaggcgagcacagggcgggacatgggcggggccggcggcgg cccccccgggcaggccaatgggagggccgggtgcgtttgaaaactggggtgggcggcggggcggggtctgcgcctg cgcgagggctacgcgcgctccggccggggcgcgggcgcgctctcaggcgggctccggcggcagcgacgcgagcg cggcgggatcgggtggtaccagtggcatgagcgagctgattaaggagaacatgcacatgaagctgtacatggagggca ccgtggacaaccatcacttcaagtgcacatccgagggcgaaggcaagccctacgagggcacccagaccatgagaatca aggtggtcgagggcggccctctccccttcgccttcgacatcctggctactagcttcctctacggcagcaagaccttcatcaa ccacacccagggcatccccgacttcttcaagcagtccttccctgagggcttcacatgggagagagtcaccacatacgaag acgggggcgtgctgaccgctacccaggacaccagcctccaggacgtaagtatgaaattcagggatacggccaccttgtt ggccggtaacctacaaacgggtggaggatcaccccacccgacacgcaccgatgctctccgaggaggcacaagggtgg aggaacaccccaccctccaggcaccgatgctctccgaggaggcaaacagaagcaccatcagggcttctgctacgcacc gatgctctccgaggaggaacaaggaatcaccatcagggattccctgtggcaccgatgctctccgaggaggacgacggg aggacgatcacgcctcccgaatagcaccgatgctctccgaggaggtcagtgcgtggagcatcagcccacgcagccagc accgatgctctccgaggaggctttcgcgaagagcatcagccttcgcgccatgcaccgatgctctccgaggaggcctaca ggaacagcatcagcgttcctgcccagcaccgatgctctccgaggaggtgtccccatgcagcatcagcgcatgggccccg caccgatgctctccgaggaggcacatccgagcaccaacagggctcggagtgtgcaccgatgctctccgaggaggaccc tccatggaccatcaggccatggactctgcaccgatgctctccgaggaggcttctcgaagcagcatcagcgcttcgaaaca gcaccgatgctctccgaggaggttctccaccttgttggcatatttgccaaatagtggaaatgtgaagtactgacaaaacttttc cctttttcaatctaatagggctgcctcatctacaacgtcaagatcagaggggtgaacttcacatccaacggccctgtgatgca gaagaaaacactcggctgggaggccttcaccgagacgctgtaccccgctgacggcggcctggaaggcagaaacgaca tggccctgaagctcgtgggcgggagccatctgatcgcaaacatcaagaccacatatagatccaagaaacccgctaagaa cctcaagatgcctggcgtctactatgtggactacagactggaaagaatcaaggaggccaacaacgagacctacgtcgag cagcacgaggtggcagtggccagatactgcgacctccctagcaaactggggcacaagcttaatggaaccggtgctggat ccggtgggagcggcggcggtgagtgcggggcgatgtccgctggtttctgccccacaccccttctgcctgccctgcgggg cggacggtgggtcccgcgggaggggaggccctggcggcctgaagagggctggctcgagctctttaacccggggcgg atgtcgcgtcccgcgcagggagccccggcggccgggcgcgcggtttaaatgcccggcgggcgcccgagcccctcgg agccttcccccgcggcgacgttttccggccctttcctggggcaacgatcgggtcccggggcagcgatccggcgatcggg tcccggggcggcgatcgggtctcggggagccaaacaccagcgcctcctggtcggggaaagggctgcggagcgcgtg ggggcgaagcgatcccgcagcccggccgggccctgaccgagcctcagtccagcggggccgtggacctagctctgtgg aaagcagcgttcggccccgcgcgcgttgaggcacaggaaacacgaactgtggtttcagctattatcatagtatctgtcctg gagcc。
Sequence listing
<110> Zhejiang university
<120> Label and method for simultaneously visualizing DNA, mRNA and protein of Gene in Living cell
<160> 8
<170> SIPOSequenceListing 1.0
<210> 1
<211> 380
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 1
gcaccgatgc tctccgagga ggcgtgaccg caccgatgct ctccgaggag gagcaagcac 60
cgatgctctc cgaggaggca gctgtgcacc gatgctctcc gaggaggcac gggctaattc 120
actcccaacg agcaagcacc gatgctctcc gaggaggcgt gaccgcaccg atgctctccg 180
aggaggagca agcaccgatg ctctccgagg aggcagctgt gcaccgatgc tctccgagga 240
ggcacccttg atctgtggat ctaccagaac gcaccgatgc tctccgagga ggcgtgaccg 300
caccgatgct ctccgaggag gagcaagcac cgatgctctc cgaggaggca gctgtgcacc 360
gatgctctcc gaggaggcac 380
<210> 2
<211> 660
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 2
ccggtaacct acaaacgggt ggaggatcac cccacccgac acttcacaat caaggggtac 60
aatacacaag ggtggaggaa caccccaccc tccagacaca ttacacagaa atccaatcaa 120
acagaagcac catcagggct tctgctacca aatttatctc aaaaaactac aacaaggaat 180
caccatcagg gattccctgt gcaatatacg tcaaacgagg gccacgacgg gaggacgatc 240
acgcctcccg aatatcggca tgtctggctt tcgaattcag tgcgtggagc atcagcccac 300
gcagccaatc agagtcgaat acaagtcgac tttcgcgaag agcatcagcc ttcgcgccat 360
tcttacacaa accacactct cccctacagg aacagcatca gcgttcctgc ccagtaccca 420
actcaagaaa atttatgtcc ccatgcagca tcagcgcatg ggccccaaga atacatcccc 480
aacaaaatca catccgagca ccaacagggc tcggagtgtt gtttcttgtc caactggaca 540
aaccctccat ggaccatcag gccatggact ctcaccaaca agacaaaaac tactcttctc 600
gaagcagcat cagcgcttcg aaacactcga gcatacattg tgcctatttc ttgggtggac 660
<210> 3
<211> 702
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 3
atgagcgagc tgattaagga gaacatgcac atgaagctgt acatggaggg caccgtggac 60
aaccatcact tcaagtgcac atccgagggc gaaggcaagc cctacgaggg cacccagacc 120
atgagaatca aggtggtcga gggcggccct ctccccttcg ccttcgacat cctggctact 180
agcttcctct acggcagcaa gaccttcatc aaccacaccc agggcatccc cgacttcttc 240
aagcagtcct tccctgaggg cttcacatgg gagagagtca ccacatacga agacgggggc 300
gtgctgaccg ctacccagga caccagcctc caggacggct gcctcatcta caacgtcaag 360
atcagagggg tgaacttcac atccaacggc cctgtgatgc agaagaaaac actcggctgg 420
gaggccttca ccgagacgct gtaccccgct gacggcggcc tggaaggcag aaacgacatg 480
gccctgaagc tcgtgggcgg gagccatctg atcgcaaaca tcaagaccac atatagatcc 540
aagaaacccg ctaagaacct caagatgcct ggcgtctact atgtggacta cagactggaa 600
agaatcaagg aggccaacaa cgagacctac gtcgagcagc acgaggtggc agtggccaga 660
tactgcgacc tccctagcaa actggggcac aagcttaatt aa 702
<210> 4
<211> 101
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 4
gtaagtatga aattcaggga tacggccacc ttgttggcat atttgccaaa tagtggaaat 60
gtgaagtact gacaaaactt ttcccttttt caatctaata g 101
<210> 5
<211> 819
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 5
atgagcgagc tgattaagga gaacatgcac atgaagctgt acatggaggg caccgtggac 60
aaccatcact tcaagtgcac atccgagggc gaaggcaagc cctacgaggg cacccagacc 120
atgagaatca aggtggtcga gggcggccct ctccccttcg ccttcgacat cctggctact 180
agcttcctct acggcagcaa gaccttcatc aaccacaccc agggcatccc cgacttcttc 240
aagcagtcct tccctgaggg cttcacatgg gagagagtca ccacatacga agacgggggc 300
gtgctgaccg ctacccagga caccagcctc caggacgtaa gtatgaaatt cagggatacg 360
gccaccttgt tggcatattt gccaaatagt ggaaatgtga agtactgaca aaacttttcc 420
ctttttcaat ctaatagggc tgcctcatct acaacgtcaa gatcagaggg gtgaacttca 480
catccaacgg ccctgtgatg cagaagaaaa cactcggctg ggaggccttc accgagacgc 540
tgtaccccgc tgacggcggc ctggaaggca gaaacgacat ggccctgaag ctcgtgggcg 600
ggagccatct gatcgcaaac atcaagacca catatagatc caagaaaccc gctaagaacc 660
tcaagatgcc tggcgtctac tatgtggact acagactgga aagaatcaag gaggccaaca 720
acgagaccta cgtcgagcag cacgaggtgg cagtggccag atactgcgac ctccctagca 780
aactggggca caagcttaat taagcggccg cgactctag 819
<210> 6
<211> 651
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 6
ccggtaacct acaaacgggt ggaggatcac cccacccgac acgcaccgat gctctccgag 60
gaggcacaag ggtggaggaa caccccaccc tccaggcacc gatgctctcc gaggaggcaa 120
acagaagcac catcagggct tctgctacgc accgatgctc tccgaggagg aacaaggaat 180
caccatcagg gattccctgt ggcaccgatg ctctccgagg aggacgacgg gaggacgatc 240
acgcctcccg aatagcaccg atgctctccg aggaggtcag tgcgtggagc atcagcccac 300
gcagccagca ccgatgctct ccgaggaggc tttcgcgaag agcatcagcc ttcgcgccat 360
gcaccgatgc tctccgagga ggcctacagg aacagcatca gcgttcctgc ccagcaccga 420
tgctctccga ggaggtgtcc ccatgcagca tcagcgcatg ggccccgcac cgatgctctc 480
cgaggaggca catccgagca ccaacagggc tcggagtgtg caccgatgct ctccgaggag 540
gaccctccat ggaccatcag gccatggact ctgcaccgat gctctccgag gaggcttctc 600
gaagcagcat cagcgcttcg aaacagcacc gatgctctcc gaggaggttc t 651
<210> 7
<211> 1466
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 7
atgagcgagc tgattaagga gaacatgcac atgaagctgt acatggaggg caccgtggac 60
aaccatcact tcaagtgcac atccgagggc gaaggcaagc cctacgaggg cacccagacc 120
atgagaatca aggtggtcga gggcggccct ctccccttcg ccttcgacat cctggctact 180
agcttcctct acggcagcaa gaccttcatc aaccacaccc agggcatccc cgacttcttc 240
aagcagtcct tccctgaggg cttcacatgg gagagagtca ccacatacga agacgggggc 300
gtgctgaccg ctacccagga caccagcctc caggacgtaa gtatgaaatt cagggatacg 360
gccaccttgt tggccggtaa cctacaaacg ggtggaggat caccccaccc gacacgcacc 420
gatgctctcc gaggaggcac aagggtggag gaacacccca ccctccaggc accgatgctc 480
tccgaggagg caaacagaag caccatcagg gcttctgcta cgcaccgatg ctctccgagg 540
aggaacaagg aatcaccatc agggattccc tgtggcaccg atgctctccg aggaggacga 600
cgggaggacg atcacgcctc ccgaatagca ccgatgctct ccgaggaggt cagtgcgtgg 660
agcatcagcc cacgcagcca gcaccgatgc tctccgagga ggctttcgcg aagagcatca 720
gccttcgcgc catgcaccga tgctctccga ggaggcctac aggaacagca tcagcgttcc 780
tgcccagcac cgatgctctc cgaggaggtg tccccatgca gcatcagcgc atgggccccg 840
caccgatgct ctccgaggag gcacatccga gcaccaacag ggctcggagt gtgcaccgat 900
gctctccgag gaggaccctc catggaccat caggccatgg actctgcacc gatgctctcc 960
gaggaggctt ctcgaagcag catcagcgct tcgaaacagc accgatgctc tccgaggagg 1020
ttctccacct tgttggcata tttgccaaat agtggaaatg tgaagtactg acaaaacttt 1080
tccctttttc aatctaatag ggctgcctca tctacaacgt caagatcaga ggggtgaact 1140
tcacatccaa cggccctgtg atgcagaaga aaacactcgg ctgggaggcc ttcaccgaga 1200
cgctgtaccc cgctgacggc ggcctggaag gcagaaacga catggccctg aagctcgtgg 1260
gcgggagcca tctgatcgca aacatcaaga ccacatatag atccaagaaa cccgctaaga 1320
acctcaagat gcctggcgtc tactatgtgg actacagact ggaaagaatc aaggaggcca 1380
acaacgagac ctacgtcgag cagcacgagg tggcagtggc cagatactgc gacctcccta 1440
gcaaactggg gcacaagctt aattaa 1466
<210> 8
<211> 2580
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 8
gaggctgtcc caggctggat cacggccgcg actgcgaggg acgccaagga gtcctctttc 60
ccctggccgc aggcgtgcgc cttcgcagcc catctggcac cgcagatgcc ggtcgccggg 120
gtcagcggcg aagcccgaac ccatgcggcc gggaggggac gcccacaaac ccctgcccgc 180
cggccgggaa gcgctcctac gggaccccag ccccgttggg gaggcggggc ggccacgcgg 240
gttccaccaa tcagcggcca atgttcgaat tcgcgtcctg gcgcggccaa tggcgggtcc 300
cggagcaggc ggggagggcg tggagggcgg taaatgaggc gagcacaggg cgggacatgg 360
gcggggccgg cggcggcccc cccgggcagg ccaatgggag ggccgggtgc gtttgaaaac 420
tggggtgggc ggcggggcgg ggtctgcgcc tgcgcgaggg ctacgcgcgc tccggccggg 480
gcgcgggcgc gctctcaggc gggctccggc ggcagcgacg cgagcgcggc gggatcgggt 540
ggtaccagtg gcatgagcga gctgattaag gagaacatgc acatgaagct gtacatggag 600
ggcaccgtgg acaaccatca cttcaagtgc acatccgagg gcgaaggcaa gccctacgag 660
ggcacccaga ccatgagaat caaggtggtc gagggcggcc ctctcccctt cgccttcgac 720
atcctggcta ctagcttcct ctacggcagc aagaccttca tcaaccacac ccagggcatc 780
cccgacttct tcaagcagtc cttccctgag ggcttcacat gggagagagt caccacatac 840
gaagacgggg gcgtgctgac cgctacccag gacaccagcc tccaggacgt aagtatgaaa 900
ttcagggata cggccacctt gttggccggt aacctacaaa cgggtggagg atcaccccac 960
ccgacacgca ccgatgctct ccgaggaggc acaagggtgg aggaacaccc caccctccag 1020
gcaccgatgc tctccgagga ggcaaacaga agcaccatca gggcttctgc tacgcaccga 1080
tgctctccga ggaggaacaa ggaatcacca tcagggattc cctgtggcac cgatgctctc 1140
cgaggaggac gacgggagga cgatcacgcc tcccgaatag caccgatgct ctccgaggag 1200
gtcagtgcgt ggagcatcag cccacgcagc cagcaccgat gctctccgag gaggctttcg 1260
cgaagagcat cagccttcgc gccatgcacc gatgctctcc gaggaggcct acaggaacag 1320
catcagcgtt cctgcccagc accgatgctc tccgaggagg tgtccccatg cagcatcagc 1380
gcatgggccc cgcaccgatg ctctccgagg aggcacatcc gagcaccaac agggctcgga 1440
gtgtgcaccg atgctctccg aggaggaccc tccatggacc atcaggccat ggactctgca 1500
ccgatgctct ccgaggaggc ttctcgaagc agcatcagcg cttcgaaaca gcaccgatgc 1560
tctccgagga ggttctccac cttgttggca tatttgccaa atagtggaaa tgtgaagtac 1620
tgacaaaact tttccctttt tcaatctaat agggctgcct catctacaac gtcaagatca 1680
gaggggtgaa cttcacatcc aacggccctg tgatgcagaa gaaaacactc ggctgggagg 1740
ccttcaccga gacgctgtac cccgctgacg gcggcctgga aggcagaaac gacatggccc 1800
tgaagctcgt gggcgggagc catctgatcg caaacatcaa gaccacatat agatccaaga 1860
aacccgctaa gaacctcaag atgcctggcg tctactatgt ggactacaga ctggaaagaa 1920
tcaaggaggc caacaacgag acctacgtcg agcagcacga ggtggcagtg gccagatact 1980
gcgacctccc tagcaaactg gggcacaagc ttaatggaac cggtgctgga tccggtggga 2040
gcggcggcgg tgagtgcggg gcgatgtccg ctggtttctg ccccacaccc cttctgcctg 2100
ccctgcgggg cggacggtgg gtcccgcggg aggggaggcc ctggcggcct gaagagggct 2160
ggctcgagct ctttaacccg gggcggatgt cgcgtcccgc gcagggagcc ccggcggccg 2220
ggcgcgcggt ttaaatgccc ggcgggcgcc cgagcccctc ggagccttcc cccgcggcga 2280
cgttttccgg ccctttcctg gggcaacgat cgggtcccgg ggcagcgatc cggcgatcgg 2340
gtcccggggc ggcgatcggg tctcggggag ccaaacacca gcgcctcctg gtcggggaaa 2400
gggctgcgga gcgcgtgggg gcgaagcgat cccgcagccc ggccgggccc tgaccgagcc 2460
tcagtccagc ggggccgtgg acctagctct gtggaaagca gcgttcggcc ccgcgcgcgt 2520
tgaggcacag gaaacacgaa ctgtggtttc agctattatc atagtatctg tcctggagcc 2580

Claims (6)

1. A TriTag label for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell is characterized in that: consists of three parts of DNA sequence:
CRISPR-Tag DNA sequence of TS1 consisting of 12 TS1 repeating units12x DNA sequence as SEQ ID No. 1;
MS2 DNA sequence consisting of 12 MS2V5 unit sequences as shown in SEQ ID No. 2;
the blue fluorescent protein sequence containing an intron, such as SEQ ID No.5, is formed by inserting a segment of intron sequence, such as SEQ ID No.4, into the blue fluorescent protein sequence, such as SEQ ID No. 3;
the TriTag label is formed by alternately inserting 12 TS1 repetitive unit sequences and 12 MS2V5 unit sequences to form a DNA/RNA double-visual sequence shown as SEQ ID No.6, and then inserting the double-visual sequence into an intron in a blue fluorescent protein sequence containing the intron by an enzyme digestion connection method to form the TriTag label shown as SEQ ID No. 7.
2. A method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell, comprising:
preparing and constructing a TriTag tag of claim 1;
(II) adopting a TriTag label to visualize DNA, mRNA and protein of a target endogenous gene in a living cell simultaneously;
(1) constructing a visual stable cell line:
(1.1) on the day, HEK293T cells were cultured with PS-free medium and plated into 12-well plates to form a first cell culture medium;
(1.2) the following day, lentivirus packaging was performed: 750ng of each dCas9-GFP were premixed with 75. mu.l of serum-reduced medium, Opti-medium14xAfter virus expression plasmid and stdMCP-tdTomato virus expression plasmid, 705ng pCMV-dR8.91 plasmid and 87ng PMD2.G plasmid, adding 4.5 mul Fugene transient transfection reagent Promega to form liposome, dripping into the supernatant of the first cell culture medium, and carrying out transient transfection on HEK293T cells in the first cell culture medium;
(1.3) after 12 hours of transient transfection in step (1.2), aspirating the first cell culture medium supernatant and replacing the first cell culture medium with fresh medium;
(1.4) culturing the cells to be infected 48 hours after transient transfection in the step (1.2) and spreading the cells into a 24-well plate to form a second cell culture medium;
(1.5) after 24 hours of forming the second cell culture medium, sucking the first cell culture medium supernatant obtained by replacing the fresh culture medium in the step (1.3), then placing the first cell culture medium supernatant into a centrifuge for 8 minutes at the rotating speed of 800g, taking the supernatant after centrifugation, and extracting the cell supernatant containing viruses;
(1.6) preparing a PS-free culture medium containing a gene transfection enhancer polybrene with the concentration of 5 mug/mL as a third culture medium to replace a second cell culture medium, transferring the cell culture to be infected to the third culture medium, and then dropwise adding 30-90 mul of cell supernatant containing viruses into the third culture medium supernatant of the cell to be infected;
(1.7) after 12 hours of infection in the step (1.6), replacing a fresh culture medium for the third culture medium, continuously culturing until the third culture medium can be passaged to an 8-well plate for microscope imaging observation, screening out a cell infection population according to fluorescence intensity and infection efficiency, and separating single cell clones of a 96-well plate;
(1.8) when the cells in the single hole of the 96-hole plate grow to a cell group state, observing the fluorescence intensity under a microscope, and screening single-cell clone;
(2) constructing a target endogenous gene visualization system containing a TriTag label in the stable cell line, and carrying out transfection and sorting;
(3) simultaneously visualizing the DNA, mRNA and protein of the target endogenous gene and live cell dynamic imaging.
3. The method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell as claimed in claim 2, wherein: the step (one) is as follows:
s1, designing and constructing a blue fluorescent protein sequence containing an intron:
adopting the 4 th intron sequence of the human HSPA5 gene, adding BstXI restriction enzyme cutting sites in the intron sequence, and directly synthesizing by a nucleic acid synthesis mode;
s2, designing and synthesizing a DNA/RNA double visualization sequence: alternately interleaving 12 TS1 repeating unit sequences and 12 MS2V5 unit sequences for 12 times by means of nucleic acid synthesis;
s3, building a TriTag label: and inserting the DNA/RNA double visualization sequence into an intron of a blue protein sequence containing the intron by using an enzyme digestion connection method to obtain the TriTag label.
4. The method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell as claimed in claim 2, wherein:
the step (2) is specifically as follows:
(2.1) designing and constructing HDR repair template plasmids and sgRNA expression plasmids which are specific in genes and contain TriTag labels according to the condition of target endogenous genes so as to edit knock-in of cells;
(2.2) culturing the cell to be knotted-in edited by using the PS-free culture medium and paving the cell to a 24-well plate to form a fourth cell culture medium;
(2.3) on the day after 24 hours from the start of the culturing in step (2.2), premixing 500ng of sgRNA expression plasmid, 100ng of Cas9 protein expression plasmid and 400ng of HDR repair template plasmid with 40. mu.l of serum-reduced medium Opti-medium, adding 2.4. mu.l of Fugene transient transfection reagent Promega to form liposomes, and adding dropwise into the supernatant of the fourth cell culture medium to transiently transfect the cells to be transfected-in edited;
(2.4) performing cell flow sorting after the transient transfection for 96 hours, recovering the BFP fluorescence expression positive cells during the cell flow sorting, continuously culturing, and discarding the rest.
5. The method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell as claimed in claim 4, wherein: the specific construction method and process of the HDR repair template plasmid are as follows:
(1) selecting the N end or the C end of the protein as an insertion site of a TriTag label through protein structure prediction;
(2) extracting a human HeLa cell genome, amplifying homologous arm sequences on two sides of the N end or the C end of a human target gene by using a PCR (polymerase chain reaction) method, and then inserting a TriTag label between the homologous arm sequences on two sides by using a homologous recombination mode to form a series sequence of a5 'homologous arm sequence-TriTag label-3' homologous arm sequence as an HDR (high-density lipoprotein receptor) repair template plasmid.
6. The method for simultaneously visualizing DNA, mRNA and protein of a gene in a living cell as claimed in claim 2, wherein: the step (3) is specifically as follows:
(3.1) culturing the cells sorted in the step (2) by using a PS-free culture medium and paving the cells into an 8-well plate to form a fifth cell culture medium;
(3.2) on the day after 24 hours from the start of the culture in step (3.1), 500ng of the TS1-sgRNA expression plasmid was premixed with 25. mu.l of the serum-reduced medium Opti-medium, and after liposome formation with the addition of 1.2. mu.l of Fugene transient transfection reagent Promega, the mixture was added dropwise to the supernatant of the fifth cell culture medium to transiently transfect the cells;
(3.3) after 24 hours from the completion of step (3.2), the cells obtained in step (3.2) were placed in a microscope, the microscope was adjusted to specific environmental conditions, and the imaging parameters of the microscope were set for live cell imaging visualization.
CN202010553835.5A 2020-06-17 2020-06-17 Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell Active CN111718931B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010553835.5A CN111718931B (en) 2020-06-17 2020-06-17 Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010553835.5A CN111718931B (en) 2020-06-17 2020-06-17 Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell

Publications (2)

Publication Number Publication Date
CN111718931A CN111718931A (en) 2020-09-29
CN111718931B true CN111718931B (en) 2021-12-07

Family

ID=72567126

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010553835.5A Active CN111718931B (en) 2020-06-17 2020-06-17 Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell

Country Status (1)

Country Link
CN (1) CN111718931B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022188054A1 (en) * 2021-03-10 2022-09-15 Nanjing University Methods and reagents for sample multiplexing for high throughput single-cell rna sequencing
CN116949039A (en) * 2022-04-15 2023-10-27 西湖实验室(生命科学和生物医学浙江省实验室) Imaging marking system based on CRISPR and application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018017738A1 (en) * 2016-07-19 2018-01-25 Altius Institute For Biomedical Sciences Methods for fluorescence imaging microscopy
CN109507416B (en) * 2018-12-27 2022-02-15 杭州迪相实业有限公司 Rapid detection kit for exosome tumor marker PDL1

Also Published As

Publication number Publication date
CN111718931A (en) 2020-09-29

Similar Documents

Publication Publication Date Title
CA3139122C (en) Curing for recursive nucleic acid-guided cell editing
US10835869B1 (en) Instruments, modules, and methods for improved detection of edited sequences in live cells
CN111718931B (en) Label and method for simultaneously visualizing DNA, mRNA and protein of gene in living cell
IL290801B2 (en) Instruments, modules, and methods for improved detection of edited sequences in live cells
CN112955540A (en) Improved detection of nuclease edited sequences in automated modules and instruments
US11236441B2 (en) Nucleic acid-guided editing of exogenous polynucleotides in heterologous cells
US20200071657A1 (en) Instruments, modules, and methods for improved detection of edited sequences in live cells
Robertson et al. Creating custom synthetic genomes in Escherichia coli with REXER and GENESIS
Gilles et al. Clonal analysis by tunable CRISPR-mediated excision
CA3167640A1 (en) Cell populations with rationally designed edits
US20210381007A1 (en) Methods and compositions for crispr editing of cells and correlating the edits to a resulting cellular nucleic acid profile
CN105695509B (en) Method for obtaining high-purity myocardial cells
WO2020037051A1 (en) Instruments, modules, and methods for improved detection of edited sequences in live cells
Sabate et al. Versatile CRISPR-Based Method for Site-Specific Insertion of Repeat Arrays to Visualize Chromatin Loci in Living Cells
Ochiai Real-time observation of localization and expression (ROLEX) system for live imaging of the transcriptional activity and nuclear position of a specific endogenous gene
US12054520B2 (en) Control of and imaging techniques for protein nanoscaffolds
WO2020183984A1 (en) Method for introducing substance into protoplast or alga by electroporation and power supply device for electroporator
WO2024068845A1 (en) New loxpsym sites for large-scale orthogonal cre-mediated recombination
Tasan New tools for live cell imaging of endogenous loci in mammalian cells
Van den Broeck et al. Establishing a reproducible approach for the controllable deposition and maintenance of plants cells with 3D bioprinting
CN113574175A (en) Gene knock-in method, gene knock-in cell production method, gene knock-in cell, canceration risk evaluation method, cancer cell production method, and kit for use in these methods
Smeele et al. Lineage tracing of zebrafish spinal cord neurons using barcode editing

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