CN111154837B - Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range - Google Patents

Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range Download PDF

Info

Publication number
CN111154837B
CN111154837B CN201910822658.3A CN201910822658A CN111154837B CN 111154837 B CN111154837 B CN 111154837B CN 201910822658 A CN201910822658 A CN 201910822658A CN 111154837 B CN111154837 B CN 111154837B
Authority
CN
China
Prior art keywords
rna
allyl
cells
site
sequencing
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
CN201910822658.3A
Other languages
Chinese (zh)
Other versions
CN111154837A (en
Inventor
刘建钊
冯新华
舒潇
曹婕
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 CN201910822658.3A priority Critical patent/CN111154837B/en
Publication of CN111154837A publication Critical patent/CN111154837A/en
Priority to PCT/CN2020/102643 priority patent/WO2021042883A1/en
Application granted granted Critical
Publication of CN111154837B publication Critical patent/CN111154837B/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
    • 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
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • 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
    • C12Q1/6869Methods for sequencing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5308Immunoassay; Biospecific binding assay; Materials therefor for analytes not provided for elsewhere, e.g. nucleic acids, uric acid, worms, mites

Abstract

The invention provides a method for detecting RNA N with single base resolution in the full transcriptome range6-methyl adenine modification method and kit. The method is based on N of ribonucleic acid (RNA) adenine in vivo6Allyl labelling and chemical treatment to induce base mutation during reverse transcription into DNA, and identifying the mutation site by nucleic acid sequencing to obtain6A site, which is the original m in cellular RNA6A site of modification. The method realizes N in the cells for the first time6-a marker specific for allyladenine which can be used not only to replace N in cells6-methyladenine sites and can be localized by means of sequencing of mutations. The method of the invention and the existing application to m6Compared with the gene sequencing technology of A detection, the mutation site can be accurate to the resolution of a single base, so that the currently commonly adopted m-based gene sequencing technology is improved6Antibody A immunoprecipitation and large-scale parallel sequencing method detection m6The precision of the A site is a direct high-throughput single-base identification method.

Description

Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range
Technical Field
The invention belongs to the field of gene sequencing, and particularly relates to a method for detecting RNA N in a full transcriptome range with single base resolution6-methyl adenine modification method and kit.
Background
RNA is composed of not only cytosine (C), thymine (U), guanine (G) and adenine (A) in combination. N is a radical of6-methyladenine (m)6A) Is an extremely important modified base on RNA, and plays a plurality of biological functions in biological processes, such as regulating the expression of genes and the like. Among them, the identification and sequencing method of the modification is a precondition for the research of the biological significance.
Because the physical and chemical properties of methyl modified adenine are close to those of common adenine, the detection can not be directly carried out by utilizing the existing one-generation or two-generation sequencing technology. At present, based on m6The antibody immunoprecipitation sequencing technology (MeRIP-seq) can make us know which transcripts and genomes contain m6A is modified, but the resolution is only limited to the range of 100 to 200 bases, which A is methylated cannot be distinguished, and whether the A is a single A or a cluster A is methylated cannot be distinguished. In order to increase the resolution, a method of immunoprecipitation (CLIP) after photocrosslinking of antibody/RNA, i.e. m on antibody and RNA, was also used6Optically active uracil or thio-congener adjacent to A is crosslinked, reverse transcription process causes the crosslinked uracil site to mutate or terminate near the crosslinking position, and further analysis of mutation or termination information indirectly identifies A adjacent to uracil as m6And (3) A site. Although the resolution of this method is improved, the location identification is indirect and m cannot be distinguished6And (4) clustering A.
From the above analysis, it was found that m is present on conventional RNA6The reason that the high-throughput analysis means of the A site does not obtain major breakthrough is mainly that the current antibody immunoprecipitation enrichment technology is always based on m enriched by antibodies6A sequence fragment, or antibody and m6Cross-linking of photoactive uracils or thio-homologues on A sequence fragments to indirectly identify m6Site of A, not passing through m directly6The A site is mutated in a manner to identify its site.
We wish to replace m with an identifiable modification group by grafting the modification group to an amino acid, introducing the amino acid with the modification group into cellular metabolism6A is subjected to methylation modification, and then the site where the modification group is located is identified by means of mutation sequencing, so that m is identified6Purpose of A site. But under the conditions of the current culture,amino acids with modifying groups are at a disadvantage in competition with normal amino acids for cellular metabolism and cannot be introduced into cells.
Therefore, a culture condition suitable for introducing a modified amino acid into cells is under urgent development.
Disclosure of Invention
In view of the disadvantages of the prior art, the present invention aims to provide a full transcriptome-wide single base resolution method for detecting RNA N6The method and the kit for modifying the-methyladenine are different from the traditional method for indirectly identifying the m6A site method for direct identification of m of cellular whole transcriptome RNA by means of mutation sequencing with single base resolution6A modification site.
The invention adopts the following technical scheme:
RNA N detection with single base resolution in full transcriptome range6-a method for methyladenine modification comprising the steps of:
(1) allyl-L-seleno/thiohomocysteine is involved in cellular metabolism, labeling nucleic acid adenine: treating cells with methionine analogue allyl-L-seleno/thio-homocysteine, the cells can introduce allyl group into specific adenine N of RNA in cells through natural metabolism under the action of a series of enzymes6On bit, form N6-allyladenine (a)6A) The site should originally be a natural methylation modification, i.e. N6-methyladenine (m)6A);
(2) Containing a6Enrichment of a-modified RNA: extracting total RNA of cells, further extracting mRNA of the cells, then breaking the RNA of the whole transcriptome of the cells into fragmented RNA of 100-300 nt, and combining N with an antibody6-allyladenosine enrichment of a by immunoprecipitation6A modified RNA and elution from the antibody with an eluent a6A, modifying RNA, and purifying the eluted RNA;
(3)RNA a6n on A6Iodine addition and cyclization treatment of allyladenine: RNA a6N on A6Iodine addition reaction of allyl adenine and subsequent induction under alkaline conditionRNA a6N on A6N of allyl adenine1,N6Forming a cyclization structure at the site, and shielding base complementary pairing;
(4) reverse transcription mutation and sequencing recognition of circularized RNA: adding HIV reverse transcriptase, N on RNA to the circularized structure obtained in step (3)1,N6In the process of reverse transcription of cyclic adenine into DNA under the action of in vitro HIV reverse transcriptase, errors are generated by introducing para-complementary base, and mutation sites are identified by means of nucleic acid sequencing, so that a is obtained6A site, which is the original m in cellular RNA6A site of modification.
In the above technical solution, further, in the step (1), the preparation method of allyl-L-seleno/thiohomocysteine comprises: under the protection of nitrogen, selenium powder Se and sodium borohydride NaBH are firstly mixed in a molar ratio of 1:14Taking ethanol as a solvent as a raw material, heating and refluxing at 80 ℃ for 6-24 hours to prepare Na2Se2Heating and refluxing the compound and hydrobromide (compound 1) of (S) - (+) -2-amino-4-bromobutyric acid with a molar weight of one half to one third of selenium powder at 80 ℃ for 6-24 hours, stopping the reaction with acid, filtering to remove insoluble substances, washing off by-products with diethyl ether, adjusting the pH value to be neutral to obtain seleno-homocystine (compound 2), adding sodium borohydride with a molar weight of 1-2 times that of the selenium powder to reduce diselenide bonds, reacting the mixture with allyl bromide with a molar weight of 0.5-2 times that of the selenium powder at room temperature for 6-24 under an alkaline condition (sodium bicarbonate/sodium carbonate with a molar weight of 0.5-1.5 times that of the selenium powder/sulfur powder), and analyzing and purifying by High Performance Liquid Chromatography (HPLC) to obtain allyl-L-seleno-homocysteine (compound 3); sulfur S is used as raw material to prepare thiohomocystine (compound 4) and allyl-L-thiohomocysteine (compound 5).
Figure GDA0002998669840000031
The cells may be, without limitation, normal mammalian cells, mammalian cancer cells, mammalian stem cells, host cells for bacteria, viruses, and cells derived from various types of tissues and organs.
Further, the specific method of cell treatment in step (1) is to replace the normal culture medium of the cell culture system with a methionine-deficient culture medium, add 10% Fetal Bovine Serum (FBS), on the basis of 1% 100x penicillin-streptomycin double antibody, add cysteine (0.1-2 mM) for half an hour, then add allyl-L-seleno/thiohomocysteine (0.1-2 mM), and continue to culture for 12-24 hours, then N on RNA can be obtained6-methyl adenine modification to N6-allyladenine modification. The methionine-deficient medium is available directly from commercial sources.
Further, in the step (2), the sample cell whole transcriptome RNA fragmentation treatment is performed by Zn2+Heating at 70 deg.C for 4-10 min.
Further, in the step (2), for binding N6-allyladenosine antibody is N6An antibody of isopentenyl adenosine, wherein 10 μ g of the antibody is used for enriching 1-20 μ g of fragmented RNA, and the antibody is combined with the sample cell whole transcriptome fragmented RNA through the ProteinA of the magnetic beads.
Further, in the step (2), the eluent is N6-allyl adenosine triphosphate (a)6ATP) in a concentration of 5 to 10mM N6-allyl adenosine triphosphate (a)6ATP) from N6-competitive elution of said cellular whole transcriptome fragmented RNA on antibodies to isopentenyl adenosine, and purification of the resulting RNA by ethanol or isopropanol precipitation.
Further, in step (3), iodine (0.1-0.5M iodine dissolved in 0.2-1M potassium iodide) and N6Iodine addition reaction of allyl group of allyladenine, removal of excess iodine (0.1 to 0.5M sodium thiosulfate), and N addition under alkaline conditions (0.1 to 0.5M sodium carbonate, pH 9 to 10)1,N6The site is self-closing, thus shielding the normal hydrogen bonding pairing of adenine.
Further, in step (4), the method for sequencing the mutation comprises the steps of: i) reverse transcription of the above allyl-L-seleno/thiohomocysteine cellular metabolism using HIV reverse transcriptaseAfter labeling, the antibody enriches the RNA cyclized by iodine addition; ii) adopting RNA library preparation technology and high-throughput sequencing means to carry out identification on the mutation sites of the whole transcriptome so as to obtain m with single base resolution of the whole transcriptome6And (3) distributing the A sites, or identifying and verifying the mutation sites on a specific transcript by adopting PCR and TA-cloning technologies.
Furthermore, the invention provides a full transcriptome range single base resolution detection RNA N6-methods of methyladenine modification, which may also have the following characteristics: amino acids for the cellular metabolism and in vitro enzyme assistance include, but are not limited to, allyl-L-selenohomocysteine (allyl-L-selenohomocysteine), allyl-L-thiohomocysteine (allyl-L-homocysteine), and derivatives and analogs thereof; n is a radical of6Antibodies to allyladenosine include, but are not limited to, N6Isopentenyl adenosine (anti-cytokine. RTM. N)6-isopentenyladenosines) and derivatives, analogues thereof; reverse transcriptase of reverse transcriptase mutations include but are not limited to HIV reverse transcriptase (recombination HIV reverse transcriptase transposase enzyme).
In the step (1), the cells are cultured by adding methionine derivative and cysteine to the methionine-deficient medium, wherein the methionine derivative includes, but is not limited to, allyl-L-selenohomocysteine (allyl-L-selenohomocysteine), allyl-L-thiohomocysteine (allyl-L-homocysteine) and its derivatives and analogs; cysteine in the method includes, but is not limited to, downstream metabolites of methionine in cellular metabolism. The chemical structural formulas of allyl-L-seleno-homocysteine (allyl-L-selenohomocysteine) and allyl-L-thiohomocysteine (allyl-L-selenohomocysteine) are shown in the specification.
Figure GDA0002998669840000041
In the step (1), the method for extracting RNA includes a common purification method or a commercial purification kit. Methods include, but are not limited to: using TRIzolTMReagent, chloroformPhenol extraction, protease K digestion, silica gel membrane centrifugation column method, magnetic bead method, ethanol, isopropanol precipitation and the like. Purification kits include, but are not limited to: GeneEluteTM mRNA Miniprep Kit、RNeasyTM Mini Kit(Qiagen),RNA Clean&Concentrator(Zymo)。
In the step (2), the fragmentation of RNA in the immunoprecipitation method includes, but is not limited to, RNA fragmentation by a metal ion method, and RNA fragmentation by ultrasonication; antibody enrichment in immunoprecipitation methods includes, but is not limited to, the method of protein A/protein G beads; elution in immunoprecipitation methods includes, but is not limited to, N6-allyladenosine monophosphate, N6-allyl adenosine triphosphate (N)6-allyl adenosine-5' -triphosphatate) and TRIzol Reagent. N is a radical of6-allyl adenosine triphosphate (N)6-allyl adenosine-5' -triphosphatate) and the synthesis steps thereof are as follows: under the protection of nitrogen, ethanol is used as a solvent, 6-chloropurine nucleoside (compound 6), allyl amine and triethylamine are heated and refluxed at the temperature of 80 ℃ for 3-6 hours in a molar ratio of 1:3:3, the obtained product is subjected to vacuum rotary evaporation, ether precipitation is carried out, insoluble substances are removed through filtration, and recrystallization is carried out through methanol to obtain N6Allyladenosine (compound 7). Then, N is added under the protection of nitrogen6Allyladenosine (0.2mmol), phosphorus oxychloride (POCl)30.26mmol) was added 0.5mL of anhydrous trimethyl phosphate (MeO)3PO, 0 deg.C for 1.5 hr, adding tributyl ammonium pyrophosphate (TBAPP, 1mmol) dissolved in 2mL anhydrous N, N-Dimethylformamide (DMF) into the reaction system, reacting at 0 deg.C for 20 min, further reacting at room temperature for 5 min, terminating the reaction with 2mL triethylammonium bicarbonate (TEAB, 1mol/L), and purifying by High Performance Liquid Chromatography (HPLC) analysis to obtain N6-allyl adenosine triphosphate (a)6ATP, compound 8). The amounts of the reactants may be adjusted proportionally.
Figure GDA0002998669840000051
In the step (3), the iodine addition method includes, but is not limited to, potassium iodide solution of iodine (0.125M I)20.25MKI), the removal of excess iodine includes but is not limited to sodium thiosulfate treatment (0.2M Na)2S2O3) Methods for inducing cyclization under alkaline conditions include, but are not limited to, sodium carbonate, sodium bicarbonate solution (0.1M Na)2CO3pH 9.5) and the reaction formula is as follows. Under alkaline conditions, N1,N6And (4) a self-closed loop is positioned, and base complementary pairing is shielded.
Figure GDA0002998669840000052
In the step (4), the reverse transcription method includes, but is not limited to, a method of treating with HIV reverse transcriptase (Recombinant HIV reverse transcriptase enzyme).
In step (4) above, the sequencing method after reverse transcription includes, but is not limited to, high-throughput sequencing for constructing a library, and low-throughput sequencing based on TA-cloning. Wherein, the PCR enzyme in TA-cloning method includes but is not limited to KOD-FX DNA polymerase, and the ligation of plasmid vector in TA-cloning method includes but is not limited to T vector; the library construction method includes, but is not limited to, the library construction of the illuma stranded, the library construction of NEB small RNA, eCIP, the improved library construction method and the like.
In the steps (1) to (4), a conventional purification method or a commercial purification kit can be used for the purification step of nucleic acid after each reaction. Methods include, but are not limited to: one or more of the techniques of silica gel membrane centrifugal column method, magnetic bead method, ethanol and isopropanol precipitation, etc. Purification kits include, but are not limited to: the AmpureXP beads are used,
Figure GDA0002998669840000061
PCR purification Kit(Qiagen),RNA Clean&Concentrator(Zymo),DNA Clean&Concentrator(Zymo)。
the invention also provides a method for detecting RNA N by using single base resolution in the full transcriptome range6-methyladenine modifiedThe kit comprises allyl-L-seleno/thiohomocysteine, cysteine and N6Allyl adenosine triphosphate, RNA fragmentation solution containing divalent zinc ions, N6An isopentenyl adenosine antibody, a potassium iodide solution of iodine, a sodium thiosulfate solution, a sodium carbonate solution, an HIV reverse transcriptase, an HIV reverse transcription reaction solution, Tris-HCl, an RNase inhibitor, a sequencing linker and a sequencing primer.
In the present invention:
(1) iodine induced double bond addition after addition to the double bond, iodine has some dissociative properties, and the carbon atom in the immediate vicinity thereof can electrophilically attack the atom with higher electron cloud density, and for adenine, the nitrogen atom on the purine ring has the characteristic of higher electron cloud density, especially the nitrogen atom at the 1-position. Therefore we have designed N6Allyl adenine nucleoside as a molecule, and under the iodine addition and alkaline conditions, allyl iodine at the N6 position can induce carbon atoms to attack N1 at the adjacent position electrophilically after iodine atoms leave to form N1 and N6 ring-closing reactions, and hydrogen bonds used for base complementary pairing at the two positions originally are shielded, so that mutation is caused in the reverse transcription process, and N recognition is achieved by means of sequencing6-the purpose of the allyladenine site;
(2) to apply the above method of mutation sequencing to m6Recognition of A site, we designed a method for cell metabolism, which utilizes the methyl metabolic pathway modified by RNA methylation in cells to replace methyl with allyl, thereby m can be replaced6Replacement of A by N6Allyl adenine, recognition of N by mutational sequencing6-site of allyladenine to achieve single base resolution detection of m6Purpose of A site. The intracellular RNA methylation modified metabolic pathway is methionine, S-adenosylmethionine, N6-methyladenine, we removed the methionine present in the cell itself and then replaced it with allyl-L-seleno/thiohomocysteine (seleno amino acids proved to be more susceptible to group transfer than thio) in culture conditions, successfully modifying the allyl group onto the RNA of the cellular whole transcriptome. Due to the identification of N by quantitative mass spectrometry6The modification efficiency of-allyladenosine is low, and we found that N can be specifically enriched6Antibodies to allyladenosine, enrichment of N by antibody immunoprecipitation6Allyl adenosine modified full transcriptome RNA, and identifying a majority of single bases m in the full transcriptome range by using two ways of high throughput and low throughput by mutation sequencing according to the iodine addition and cyclization method6A site, greatly promoting m6And (3) researching the biological function of the A modification.
The invention has the beneficial effects that:
the invention relates to a method for detecting RNA N with single base resolution in the full transcriptome range6The method and kit for modifying methyl adenine is based on the chemical marking and induced mutation of nucleic acid adenine in vivo and the application in m6Compared with the gene sequencing technology of A detection, the mutation site can be accurate to the resolution of a single base, so that the currently commonly adopted m-based gene sequencing technology is improved6Antibody A immunoprecipitation and large-scale parallel sequencing method detection m6The precision of the A site is a direct high-throughput single-base identification method.
The invention realizes N of ribonucleic acid (RNA) adenine in cells for the first time6Allyl labelling for the subsequent passage m6The way in which the A site is mutated to identify its site provides the possibility.
Due to N6The modification efficiency of-allyladenosine is low, so that N is used in the present invention6Antibodies to isopentenyl adenosine to specifically enrich for N6Antibodies to allyladenosine. Because currently there is no N temporarily6Antibodies specific for allyladenosine, analyzed and screened for some commercial antibodies, we found that N6Antibodies to isopentenyl adenosine to N6-allyladenosine, the antibody having a specificity for N6Allyl adenosine has better binding capacity, and the commercial antibody is more easily obtained, thereby greatly improving the practicability of the method.
The invention adopts N6-allyl radicalAdenosine triphosphate (a)6ATP) as eluent due to a6A on ATP and RNA6The main structure of the A modification is the same, and the water solubility of the A modification is increased by the triphosphate, so that the A modification has stronger binding capacity with the antibody, and the A modification is similar to the a modification6The A modified RNA is in competition in binding to the antibody, so a can be bound to the antibody through competition6Elution of A-modified RNA. Therefore, compared with the common RNA extraction method for eluting RNA, the method enables a6The RNA modified by A is almost completely separated from the antibody, and the yield of the eluted RNA is greatly improved.
In the present invention, HIV Reverse Transcriptase is used for Reverse transcription of RNA subjected to iodine addition and cyclization induction, because it is found in our research that HIV Reverse Transcriptase has a stronger recognition ability for hydrogen bond-shielded cyclization structures of adenine for base complementary pairing, we have selected a number of commercial Reverse transcriptases, including HIV Reverse Transcriptase (Worthington Biochemical Corporation), M-MLV Reverse Transcriptase (PROMEGA, M170A), AMV Reverse Transcriptase (PROMEGA, M510F), RevertAID Reverse Transcriptase (Thermofisher, EP0441), SuperScript II Reverse Transcriptase (Invitrogen,100004925), SuperScript III Reverse Transcriptase (Invitrogen,55549), etc., and finally discovered that HIV Reverse Transcriptase is a in a6The corresponding site is mutated during the reverse transcription of the A iodine addition cyclization RNA.
The invention can be applied to various analysis methods based on gene sequencing on the basis of mutation sequencing, such as m on various types of nucleic acids6Detection of A modification sites, and based on N6Dynamic sequencing of cellular RNA of allyladenosine, etc.
Drawings
FIG. 1 is a single base resolution detection of RNA N in the full transcriptome range6-a schematic representation of a method for methyladenine modification;
FIG. 2 shows N in mRNA obtained by introducing allyl-L-seleno/thiohomocysteine into HeLa cells for metabolism6Content of allyl adenosine, N in mRNA obtained after introduction of methionine into HeLa cells for metabolism6Content of-methyladenine nucleosides and N in mRNA obtained after HeLa cells were cultured normally6-the content of methyladenosine;
FIG. 3 shows RNA bands obtained by fragmenting mRNA of a sample cell with divalent zinc ions;
FIG. 4 is N6Antibodies to isopentenyl adenosine to normal A, m6A modified RNA, a6A dot blot hybridization of modified RNA showing binding ability of corresponding RNA to antibody, methylene blue showing RNA loading;
FIG. 5 shows N before and after mRNA was enriched in HeLa (human cancer cells) and H2.35 (mouse common cells) cells by antibody immunoprecipitation6-content of allyladenosine;
FIG. 6 shows a in HeLa cells6A modified RNA, the result of gene sequencing before and after iodine addition induced cyclization;
FIG. 7 shows m on transcriptome from high throughput sequencing of HeLa cells and H2.35 cells6Conserved sequence of A site;
FIG. 8 shows m on transcriptome from high throughput sequencing of HeLa cells6Site A, exemplified by three mRNA gene sequences;
FIG. 9 shows m on transcriptome from high throughput sequencing of H2.35 cells6Site A, exemplified by four mRNA gene sequences;
FIG. 10 is m on transcriptome from low throughput sequencing of HeLa cells6Site A, exemplified by three mRNA gene sequences;
FIG. 11 is a high resolution mass spectrum of allyl-L-selenohomocysteine;
FIG. 12 is a photograph of allyl-L-seleno-homocysteine1H nuclear magnetic resonance spectroscopy;
FIG. 13 is a photograph of allyl-L-seleno-homocysteine13C nuclear magnetic resonance spectrum;
FIG. 14 is a high resolution mass spectrum of allyl-L-thiohomocysteine;
FIG. 15 is N6-high resolution mass spectrometry of allyl triphosphate.
Detailed Description
The present invention will be described in detail with reference to the drawings and specific examples, but they should not be construed as limiting the scope of the present invention.
FIG. 1 shows a single base resolution method for detecting RNA N in the full transcriptome range according to the present invention6Schematic representation of the process for the modification of methyladenine. The method comprises the following steps:
(1) allyl-L-seleno/thiohomocysteine is involved in cellular metabolism, labeling nucleic acid adenine: the cell can take in the methionine analogue allyl-L-seleno-homocysteine, and the allyl group can be introduced into the specific adenine N of RNA in the cell under the action of a series of enzymes through the natural metabolism of the cell6On bit, form N6-allyladenine (a)6A) The site should originally be a natural methylation modification, i.e. N6Methyl adenine (m)6A);
(2) Containing a6Enrichment of a-modified RNA: extracting total RNA of cells, further extracting mRNA of the cells, then breaking the RNA of the sample cell complete transcriptome into fragmented RNA of 100-300 nt, and utilizing N6-antibodies to allyladenosine enrichment of a by immunoprecipitation6A modified RNA with N6-allyl adenosine triphosphate (a)6ATP) elution from the antibody a6A, modifying RNA, and purifying the eluted RNA;
(3) iodine-added RNA a6N on A6Allyl induced generation of N1,N6Site cyclization: RNA a6N on A6Iodine addition reaction of allyl adenine and subsequent induction of RNA a under alkaline conditions6N on A6N of allyl adenine1,N6Forming a cyclization structure at the site, and shielding base complementary pairing;
(4) reverse transcription mutation and sequencing recognition of circularized RNA: n on RNA1,N6In the process of reverse transcription of cyclic adenine into DNA under the action of in vitro HIV reverse transcriptase, errors are generated by introducing para-complementary base, mutation sites can be identified by means of nucleic acid sequencing, and then a is obtained6A site, which is the original m in cellular RNA6A site of modification.
In the present invention, the specimenAfter the cells are normally cultured to about eighty percent of fusion degree, 1mM cysteine is added into a cell culture medium without methionine for pretreatment for 30 minutes to remove the residual methionine in the cells, 1mM allyl-L-seleno/thiohomocysteine is added into the culture medium, and after the cells are cultured for 12-24 hours, total RNA of sample cells is firstly extracted, and then the RNA of a sample cell complete transcriptome is further extracted. The present invention is not particularly limited in the kind of sample cells that can be used, and the cells may be, but are not limited to, general mammalian cells, mammalian cancer cells, mammalian stem cells, host cells of viruses, bacteria, and cells derived from various types of tissues and organs. In the present invention, the method for collecting, lysing and extracting total RNA and total transcriptome RNA from cells and tissues can be performed by RNA extraction methods conventional in the art without any special requirement, for example, TRIzol Reagent is used for extracting total RNA in the specific implementation process of the present invention, GenEluteTMmRNA was extracted with the mRNA Miniprep Kit.
After the sample cell full transcriptome RNA is obtained, the sample cell full transcriptome RNA is fragmented into 100-300 nt RNA. The fragmentation treatment described in the present invention is preferably achieved using divalent zinc ions which can cleave phosphodiester bonds on RNA without destroying the linked bases. A preferred fragmentation buffer in the present invention is (1. mu.L of 1M ZnCl)21 μ L of Tris-HCl pH 7.0,8 μ L of RNase-free water), 400-700 ng/μ L of the sample cell whole transcriptome RNA and fragmentation buffer were heated at 70 ℃ for 4-10 minutes at a volume ratio of 9: 1. After obtaining the fragmented RNA, the invention preferably further comprises a step of purifying the fragmented RNA, wherein the purification method adopted in the invention is an RNA isopropanol precipitation method which is conventional in the field, namely an RNA sample, a 3M sodium acetate solution, isopropanol and glycogen have a volume ratio of 100:10:110:1, after precipitation overnight, the RNA sample is centrifuged at 15000rpm and 4 ℃ for 45 minutes, and after washing with 80% ethanol, the RNA is dissolved to obtain the fragmented RNA solution of the sample cell complete transcriptome.
After the fragmented RNA of the sample cell complete transcriptome is obtained, the RNA is enriched by an antibody immunoprecipitation method. Antibodies employed in the inventionThe Immunoprecipitation method (IP) is a conventional protein A bead method in the art, and the antibody used is preferably commercial N6Antibodies to isopentenyl adenosine. Every 1-20 mu g of the fragmented RNA, preferably 3-10 mu g, is incubated and combined with 1-20 mu g of antibody, preferably 5-10 mu g, for 2-6 hours, preferably 3-4 hours, and preferably at 4 ℃. Binding the fragmented RNA to the antibody, binding the antibody to ProteinA magnetic beads, and magnetically separating ProteinA magnetic beads and N6Antibodies to isopentenyladenosine, N6After the product of the combination of allyl adenosine-modified RNA and N6-elution with allyl triphosphate of RNA. N is a radical of6The concentration of the eluent of allyl adenosine triphosphate is 5 to 10mM, preferably 6.67 mM. After enriching RNA, the invention preferably further comprises a step of purifying RNA to obtain N6Allyl adenosine modified RNA was purified by the above-described isopropanol precipitation method.
As shown in FIG. 4, three RNAs were obtained by in vitro transcription, each being normal RNA (A-RNA), with substitution of A for m6RNA (m) of A6A-RNA), and replacement of A with a6RNA of A (a)6A-RNA), three RNAs and N6Immunodot hybridization dot-blot of the antibodies to isopentenyl adenosine revealed that only a6A-RNA (compare A-RNA to m)6A-RNA) has a relatively specific binding ability to the antibody, and can be derived from the normal A sequence of cellular mRNA and m6A in the modified sequence6A modified mRNA was identified to provide enrichment function, as a control, Methylene blue methyl blue shows the loading of the corresponding RNA, demonstrating a with the same amount of RNA6The specific binding ability of A-RNA to antibodies.
The invention obtains the sample cell complete transcriptome N6Allyl adenosine-modified RNA was then subjected to iodine addition and cyclization under alkaline conditions. In the present invention, 26. mu.L of the above RNA and 4. mu.L of a 0.125M iodine solution (dissolved in 0.25M potassium iodide) are treated at 4 to 50 ℃ for 15 to 60 minutes, preferably at 37 ℃ for 30 minutes. Then, 2-4 mu L of 0.2M sodium thiosulfate is added until the solution is colorless, and then the solution is addedAdding 6 μ L of 0.1M sodium carbonate (pH 9-10, preferably 9.5), and treating at 4-50 deg.C for 15-60 min, preferably at 37 deg.C for 30 min. In the present invention, after the iodine addition and cyclization treatment of RNA, it is preferable to further include a step of purifying RNA, and the obtained RNA product is purified by the above-mentioned isopropanol precipitation method.
After immunoprecipitated cyclized and unclycled products are obtained, fragmented RNA is subjected to HIV reverse transcription and then connected with a sequencing joint, and a sequencing primer is used for PCR amplification to construct a high-throughput sequencing library. In the invention, the fragmented RNA is subjected to HIV reverse transcription, then is connected with a sequencing joint, and is subjected to PCR amplification by using a sequencing primer to construct a high-throughput library, preferably an illumina Troseq stranded mRNA LT kit, wherein the specific library construction method is carried out according to an instruction.
After the construction of the sequencing library is completed, the sequencing library is sequenced to obtain sequencing data. In the present invention, the sequencing is preferably next generation sequencing, more preferably Illumina double-ended sequencing, and the read length of the sequencing is preferably 150 bp.
The invention analyzes the mutation rate of the samples with cyclization and non-cyclization at the adenylic acid sites after obtaining the sequencing data, if the mutation rate of the samples with cyclization is more than three times of the samples with non-cyclization, and the mutation rate is a6The site is considered as m in the antibody enrichment region A6And (3) A site. The analysis includes quality control of the sequencing data, removal of linker sequences from the data, removal of low quality bases, alignment of the low quality and linker removed data to genomic sequences and enrichment statistics, alignment of the low quality and linker removed data to transcriptome sequences and mutation statistics. In a specific embodiment of the present invention, the analysis comprises the steps of: performing quality control on the original sequencing data through fastqc default parameters, removing a linker sequence and a sequence with less than 25 bases (fastp-F10-F10-x- -detect _ adapter _ for _ pe-l 25-iRaw-R1. fq-I Raw-R2. fq-O Clean-R1. fq-O Clean-R2.fq) through fastp software, performing quality control on the obtained sequence through the fastqc default parameters again, aligning to a transcriptome sequence through the hisat2 default parameters, and converting the aligned SAM file into a BAM text SAM file through samtoolsRemoving repeated sequences by using samtools rmdup, finally counting mutation information in a BAM file by using samtools mplieup, comparing the mutation rates of cyclized and uncyclized samples at the same site, and considering that the mutation rate is m when the fold change is more than three6And (3) A site.
After the immunoprecipitated cyclized and unclycled products are obtained, PCR and TA-cloning technology can be used for low-throughput sequencing of a certain section of transcriptome to identify mutation sites. In the present invention, the DNA polymerase for PCR is preferably KOD-FX DNA polymerase, and the plasmid vector for ligation of the PCR product is preferably a T-vector. As shown in fig. 6, a certain segment passes through a6The result of low-flux sequencing of A-labeled RNA by PCR and TA-cloning technology, wherein X ═ a6A indicates that the site is determined to have a6A is modified, X ═ cyc-A represents a6The A modified site is processed by iodine addition and induced cyclization, and the result shows that HIV reverse transcriptase generates mutation at X ═ cyc-A site and X ═ a6The absence of mutations at the A site demonstrates the necessity for iodine addition and induction of cyclization as well as the necessity for HIV reverse transcriptase. Comparing the mutation rate of cyclized and uncyclized samples at the same locus, finding that the fold change is more than three, and confirming that m can be accurately detected by adopting the method of the invention6And (3) A site.
The invention also provides a method for detecting RNA N by using single base resolution in the full transcriptome range6-methyladenine modified kit comprising allyl-L-seleno/thiohomocysteine, cysteine, N6Allyl adenosine triphosphate, RNA fragmentation solution containing divalent zinc ions, N6Isopentenyl adenosine antibody, 0.125M iodine in potassium iodide, 0.2M sodium thiosulfate, 0.1M sodium carbonate (pH 9.5), HIV reverse transcriptase, HIV reverse transcription reaction solution, Tris-HCl, RNase inhibitor, sequencing linker, sequencing primer.
The synthesis steps of the allyl-L-seleno/thiohomocysteine are as follows: under the protection of nitrogen, selenium powder Se (4mmol) and sodium borohydride NaBH are firstly added4(4mmol) as a raw material and ethanol as a solvent, and heating and refluxing the raw material and the ethanol at the temperature of 80 ℃ for 6 to 24 hours to obtain Na2Se2And then reacting it with (S) - (+) -2-amino-4-bromobutyric acidHeating and refluxing the hydrobromide (2mmol) at 80 ℃ for 6-24 hours, stopping the reaction with acid, filtering to remove insoluble substances, washing off by-products with diethyl ether, and adjusting the pH value to be neutral to obtain seleno homocystine (1 mmol); adding sodium borohydride (6mmol) to reduce diselenide bond, reacting with allyl bromide (3.8mmol) under alkaline condition (sodium bicarbonate, 3mmol) for 6-24, and purifying by high performance liquid chromatography to obtain allyl-L-seleno-homocysteine; in the same way, the allyl-L-thiohomocysteine is prepared by taking the sulfur S as a raw material.
FIG. 11 shows a high resolution mass spectrum of allyl-L-selenohysteine prepared according to the present invention (M/z is 224.0183, [ M + H ]]+Theoretical calculation is 224.0184); FIG. 12 is a drawing thereof1An H nuclear magnetic resonance spectrum, wherein the H nuclear magnetic resonance spectrum,1H NMR(500MHz,D2o) δ 5.84(ddt, J ═ 17.6,9.9,7.7Hz,1H), 5.10-4.90 (m,2H),3.72(dd, J ═ 6.7,5.7Hz,1H), 3.29-3.06 (m,2H), 2.62-2.41 (m,2H), 2.24-1.93 (m, 2H); FIG. 13 is the same13A C nuclear magnetic resonance spectrum of a nuclear magnetic resonance,13C NMR(126MHz,D2o) δ 174.02(s),134.73(s),116.75(s),54.78(s),31.19(s),25.33(s), 17.17(s); as can be seen from FIGS. 11-13, the present invention successfully prepares allyl-L-selenohomocysteine. For the same reason, refer to FIG. 14 (C)7H13NO2S, M/z is 176.0729, [ M + H ]]+Theoretical calculation is 176.0740), it is known that allyl-L-thiohomocysteine is successfully prepared according to this method.
N6The synthesis of allyl adenosine triphosphate comprises the following steps: under the protection of nitrogen, ethanol is used as a solvent, 6-chloropurine nucleoside, allylamine and triethylamine are heated and refluxed at the temperature of 80 ℃ for 3-6 hours in a molar ratio of 1:3:3, the obtained product is subjected to vacuum rotary evaporation, ether precipitation is carried out, insoluble substances are removed through filtration, and recrystallization is carried out through methanol to obtain N6-allyladenosine. Then, N is added under the protection of nitrogen6Allyladenosine (0.2mmol), phosphorus oxychloride (POCl)30.26mmol) was added 0.5mL of anhydrous trimethyl phosphate (MeO)3PO, 0 ℃ for 1.5 hours, then adding tributylammonium pyrophosphate (TBAPP, 1mmol) dissolved in 2mL of anhydrous N, N-Dimethylformamide (DMF) to the above reaction system, reacting at 0 ℃ for 20 minutes, and further reacting at room temperature for 5 minutesFinally, the reaction was quenched with 2mL triethylammonium bicarbonate (TEAB, 1mol/L) and purified by High Performance Liquid Chromatography (HPLC) to yield N6-allyl adenosine triphosphate (a)6ATP)。
FIG. 15 shows that N prepared by the present invention6-allyl adenosine triphosphate (a)6ATP) associated characterization data (high resolution Mass Spectrometry, C13H20N5O13P3M/z is 546.0192, [ M-H ]]-Theoretical calculation 546.0198), it was found that a was successfully prepared6ATP。
Example 1m on transcriptome mRNA in HeLa cells6Detection of A modification sites
1.HeLa cell culture, allyl labeling and mRNA extraction
(1) Culturing HeLa cells under normal culture conditions until the fullness is eighty percent, sucking out the culture medium, and washing away the residual culture medium by Phosphate Buffered Saline (PBS);
(2) adding 10% Fetal Bovine Serum (FBS), 1% 100 Xpenicillin-streptomycin double antibody and 1mM cysteine into methionine-free culture medium, culturing the above HeLa cells in the culture medium for 30 min, and removing methionine remaining in the cells;
(3) removing methionine, adding 1mM allyl-L-seleno/thiohomocysteine into the culture medium for culturing the HeLa cells, and continuously culturing for 16 hours;
(4) after the cells were cultured, the medium was aspirated, the residual medium was washed with PBS, and TRIzol was added to the petri dishTMReagent to cover the bottom of the culture dish, eluting all the adherent HeLa cells into the solution, transferring the HeLa cells into a centrifuge tube, and cracking the HeLa cells for 5 minutes at room temperature;
(5) each 1mL of TRIzolTMReagent, adding 0.2mL of chloroform into the centrifuge tube, violently shaking the centrifuge tube for 15 seconds, extracting at room temperature for 3 minutes, centrifuging at 4 ℃ for 15 minutes under the condition of 12000g of RCF, layering the solution, wherein the supernatant is RNA, the middle white precipitate is DNA, and the lower red liquid is protein;
(6) transferring the upper aqueous phase to a new centrifuge tube, adding isopropanol (isoprostanol) with the same volume, incubating on ice for 10 minutes, and centrifuging at 4 ℃ at RCF 15000g for 15 minutes to obtain total RNA white precipitate;
(7) removing the supernatant to leave white particles of total RNA, washing the precipitate with 1mL 75% ethanol (ethanol)/1mL TRIzol, centrifuging at 4 ℃ at RCF 15000g for 15 minutes, removing the supernatant again, air drying for 3 minutes, dissolving the total RNA with 250 μ L of RNase-free water, heating at 70 ℃ for 10 minutes to dissolve the total RNA sufficiently, and measuring the RNA concentration with Bio Drop to be 1000 ng/. mu.L;
(8) mRNA was further extracted with GenElute mRNA miniprep Kipp. Taking 250 mu L of total RNA (1000 ng/. mu.L) into a 1.5mL centrifuge tube, adding 250 mu L of 2x Binding Solution, and slightly shaking the centrifuge tube to mix the Solution;
(9) adding 20 mu L of evenly dispersed oligo (dT) beads, shaking to fully and evenly mix the system, heating at 70 ℃ for 3 minutes to denature RNA, then incubating at room temperature for 10 minutes to fully combine oligo (dT) with RNA, centrifuging the mixed system at RCF 15000g for 2 minutes to obtain oligo (dT) beads combined with mRNA, carefully removing supernatant, and leaving about 50 mu L of solution to prevent the beads from losing;
(10) adding 500 mu L of Wash Solution into a centrifuge tube to fully suspend oligo (dT), transferring the oligo (dT) into a GenElute centrifugal filter column/collection tube, centrifuging at the RCF 15000g for 2 minutes, and discarding the liquid in the collection tube;
(11) adding 500 mu L of Wash Solution into the filter column again, centrifuging for 2 minutes at the speed of 15000g of RCF, and discarding the liquid in the collecting pipe;
(12) finally, the filtration column was transferred to a new collection tube, 50. mu.L of Solution preheated at 70 ℃ was added to the very center of the filtration membrane of the centrifugal filtration column, brought into full contact with the mRNA complex in the column well, incubated at 70 ℃ for 5 minutes, and centrifuged at RCF 15000g for 1 minute to obtain an mRNA Solution in the collection tube, and the mRNA concentration was measured to be 200 ng/. mu.L by Bio Drop. In addition, can also absorb 50 u L70 degrees of Elution Solution repeated above Elution process, so that the filter column hole in the mRNA fully dissolved, will get the mRNA placed at-80 degrees C storage.
2. Containing adenine N6Site allyl modified RNA enrichment
(1) Mixing 200 μ L of the above RNA sample (200 ng/. mu.L), 20 μ L of 3M sodium acetate solution, 220 μ L of isopropanol, and 2 μ L of glycogen, pumping, mixing, precipitating overnight, centrifuging at 15000rpm for 45 minutes at 4 ℃, washing the precipitate with 440 μ L of 80% ethanol, centrifuging at 15000rpm for 15 minutes at 4 ℃, removing the supernatant again, air drying for 3 minutes, and dissolving RNA with 70 μ L of RNase-free water to a concentration of 550 ng/. mu.L;
(2) mu.L of the above RNA sample (550 ng/. mu.L) was mixed with 5. mu.L of Zn2+Mixing fragmentation buffer (10x Fragment buffer), beating, mixing, heating at 70 deg.C for 7 min, adding 10 μ L0.5M EDTA solution to terminate fragmentation, and adding Zn2+The fragmentation buffer (10x Fragment buffer) composition is shown in table 1;
TABLE 1 Zn2+Fragmentation buffer (10X Fragment buffer)
Figure GDA0002998669840000131
(3) Mixing the 60 μ L solution with 6 μ L3M sodium acetate solution, 66 μ L isopropanol, 1 μ L glycogen, blowing, mixing, precipitating overnight, centrifuging at 15000rpm for 45 min at 4 deg.C, washing the precipitate with 132 μ L80% ethanol, centrifuging at 15000rpm for 15 min at 4 deg.C, removing the supernatant again, air drying for 3 min, dissolving RNA with 200 μ L RNase-free water to a concentration of 100ng/μ L, and obtaining RNA fragments (fragments) with a fragment length of 100-300 nt as shown in FIG. 3;
(4) 50. mu.L of the above-described fragmented RNA (100 ng/. mu.L) was mixed with 80. mu.L of 5 XP buffer (5 XP buffer), 2. mu.L of RNase inhibitor (RNase inhibitor), 1. mu. L N6Mixing the antibody of isopentenyl adenosine and 267 μ L of RNase-free water, beating, mixing, rotating and shaking at 4 deg.C for 4 hr to make N6Allyl adenosine modified RNA with N6Antibodies to isopentenyl adenosine bind well, 5 × IP buffer composition is shown in table 2;
TABLE 25 XIP buffer (5 XIP buffer)
Figure GDA0002998669840000141
(5) Meanwhile, 40. mu.L of ProteinA magnetic beads (ProteinAbeads) were separated on a magnetic stand, washed three times with 200. mu.L of 1 XP buffer (1 XP), the remaining clear solution was aspirated, mixed with 80. mu.L of 5 XP buffer (5 XP), 10. mu.L of 20 mg/. mu.L Bovine Serum Albumin (BSA), 310. mu.L of RNase-free water, and then whipped to mix well, and vortexed at 4 ℃ for 2 hours to prevent nonspecific adsorption of the magnetic beads when they bind to the antibody, and the composition of 1 XP buffer (1 XP buffer) is shown in Table 3;
TABLE 31 XIP buffer (1 XIP buffer)
Figure GDA0002998669840000142
(6) After RNA and antibody are fully combined, separating the mixed solution of the ProteinA magnetic beads and BSA on a magnetic frame, washing the magnetic beads with 200 mu L of 1x IP buffer solution (1x IP buffer) for three times, sucking the residual clear solution, mixing the ProteinA magnetic beads with the mixed solution of the RNA and the antibody, blowing and uniformly mixing the mixture, rotating and uniformly mixing the mixture at 4 ℃ and incubating the mixture for 2 hours to ensure that the antibody combined with the RNA is combined with the ProteinA on the magnetic beads;
(7) the mixture was placed on a magnetic rack to separate the magnetic beads, washed three times with 200. mu.L of 1 XP buffer (1 XP buffer), the remaining supernatant was aspirated, and 100. mu. L N was added6Mixing allyl adenosine triphosphate eluate (Elution buffer) with ProteinA magnetic beads, pumping, shaking at 4 deg.C for 3 hr, and adding N6-allyladenosine modified RNA eluted from antibodies bound to ProteinA magnetic beads into solution, N6The composition of the eluate of allyl triphosphate is shown in table 4;
TABLE 4N6Eluate of allyl adenosine triphosphate (Elution buffer)
Figure GDA0002998669840000151
(8) mu.L of the eluate after immunoprecipitation of 4 50. mu.L of fragmented RNA (100 ng/. mu.L) antibodies was mixed to give 400. mu.L of a solution, and 40. mu.L of a 3M sodium acetate solution, 440. mu.LL isopropanol and 2 mu L glycogen are mixed, then the mixture is beaten and mixed evenly, after the sedimentation is carried out overnight, the mixture is centrifuged for 45 minutes at the rotating speed of 15000rpm and 4 ℃, the sediment is washed by 880 mu L80% ethanol, the mixture is centrifuged for 15 minutes at the rotating speed of 15000rpm and 4 ℃, the supernatant is removed again, the air drying is carried out for 3 minutes, and 20 mu L RNase-free water is used for dissolving RNA to obtain the concentration of 15 ng/mu L. Carrying out enzymolysis on the obtained RNA to obtain single nucleoside, and carrying out liquid mass spectrometry quantification to obtain a before and after IP6The content of A in RNA is shown in FIG. 5, HeLa Input is a in mRNA of HeLa cells before IP6A content, HeLa IP as a after IP6Content of A, H2.35 Input is a in mRNA of IP pre-mouse H2.35 cell6A content, H2.35 IP is a after IP6A content, the results show that IP is indeed a6The a modified mRNA was enriched.
3. N of RNA6Iodine addition reaction and cyclization treatment of-allyladenine
(1) mu.L of the above antibody-enriched RNA (15 ng/. mu.L) was transferred to a PCR tube, diluted to 26. mu.L with RNase-free water, and 4. mu.L of a 0.125M iodine solution (dissolved in 0.25M potassium iodide) was added to turn the solution brown and treated at 37 ℃ for 30 minutes;
(2) the brown solution was transferred to a new PCR tube, 4 μ L of 0.2M sodium thiosulfate was added until the solution was colorless, 6 μ L of 0.1M sodium carbonate (pH 9.5) was added, and treated at 37 ℃ for 30 minutes;
(3) mu.L of the above solution was mixed with 40. mu.L of isopropanol and 1. mu.L of glycogen, and then the mixture was shaken and mixed, and after overnight precipitation, the mixture was centrifuged at 4 ℃ at 15000rpm for 45 minutes, and the precipitate was washed with 880. mu.L of 80% ethanol, and centrifuged at 4 ℃ at 15000rpm for 15 minutes, and the supernatant was removed again, air-dried for 3 minutes, and RNA was dissolved with 15. mu.L of RNase-free water to give a concentration of 10 ng/. mu.L.
4. Constructing a library by using an illumina mRNA library construction kit for RNA fragments obtained without immunoprecipitation and immunoprecipitation (including cyclization treatment and non-cyclization treatment);
(1) configuring a reverse transcription system according to a reaction system shown in the table 5;
TABLE 5 mRNA library construction reverse transcription reaction System
Figure GDA0002998669840000161
(2) The program was run in a PCR instrument after mixing as shown in Table 6;
TABLE 6 mRNA library construction reverse transcription PCR run program
Figure GDA0002998669840000162
(3) Adding 5 μ L of resuspension buffer and 20 μ L of Second Strand Marking Master Mix into the system, mixing uniformly, and incubating at 16 ℃ for 1 h;
(4) purifying the double-stranded DNA fragment obtained above: adding 100 mu L of AMPure XP beads which are restored to the room temperature in advance into a reaction system, blowing and beating the mixture uniformly for 6 to 10 times, then incubating the mixture for 15 minutes at the room temperature, separating the mixture by using a magnetic frame, discarding the supernatant, washing the beads twice by using 80 percent ethanol, standing the mixture for 30 seconds each time for 5 to 10 minutes to volatilize the ethanol, eluting a double-stranded DNA fragment by using 17.5 mu L of resuspension buffer, incubating the mixture for 2 minutes at the room temperature, then separating the mixture by using the magnetic frame, and taking 15 mu L of the supernatant for the next reaction;
(5) the adenylic acid tail was added to the double-stranded DNA obtained above according to the following reaction system configuration of Table 7;
TABLE 7 mRNA library construction with adenylate tail addition reaction System
Figure GDA0002998669840000163
(6) The system is blown, beaten and mixed uniformly, and then the running program in a PCR instrument is shown in Table 8;
table 8 mRNA library construction with addition of adenylate tail PCR run program
Figure GDA0002998669840000164
(7) Add the adaptor according to the following table 9 reaction system configuration;
TABLE 9 mRNA library construction configuration addition linker reaction System
Figure GDA0002998669840000171
(8) Incubating the reaction system at 30 ℃ for 10 minutes;
(9) adding 5 mu L of connection termination mixture into the reaction, and uniformly mixing by blowing;
(10) purification of the adaptor-ligated DNA fragment: adding 42.5 mu L of AMPure XP beads which are restored to room temperature in advance into a reaction system, blowing and uniformly mixing for 6-10 times, then incubating for 15 minutes at room temperature, separating by using a magnetic frame and discarding the supernatant, washing the beads twice by using 80% ethanol, standing for 30 seconds each time, volatilizing the ethanol for 5-10 minutes, eluting double-stranded DNA fragments by using 52.5 mu L of resuspension buffer, separating by using the magnetic frame after incubating for 2 minutes at room temperature, taking 50 mu L of supernatant, adding 50 mu L of AMPure XP beads, repeating the purification step, eluting by using 22.5 mu L of resuspension buffer, and taking 20 mu L of supernatant for next PCR library amplification;
(11) configuring a library amplification system according to the following reaction system shown in Table 10;
TABLE 10 mRNA library construction amplification reaction System
Figure GDA0002998669840000172
(12) The system is blown, beaten and mixed evenly, and then the running program in a PCR instrument is shown in a table 11;
(13) purification of the amplified library: adding 55 mu L of AMPure XP beads which are restored to room temperature in advance into a reaction system, blowing and beating the mixture uniformly for 6-10 times, then incubating the mixture for 15 minutes at room temperature, separating the mixture by using a magnetic frame, discarding the supernatant, washing the beads twice by using 80% ethanol, 30 seconds each time, standing the mixture for 5-10 minutes to volatilize the ethanol, eluting a double-stranded DNA fragment by using 22.5 mu L of resuspension buffer, incubating the mixture for 2 minutes at room temperature, then separating the mixture by using the magnetic frame, and taking 20 mu L of the supernatant, wherein 1 mu L of the supernatant is used for determining the concentration by using Qubit;
TABLE 11 mRNA library construction amplification PCR run program
Figure GDA0002998669840000173
Figure GDA0002998669840000181
(14) Establishing a library of the obtained mRNA, performing double-end 150 sequencing by using an illumina X-10 platform, comparing the obtained data with a transcriptome of a cell source species to be tested after low-quality filtration and joint filtration, and obtaining m on the transcriptome of the HeLa cell and the mouse H2.35 cell by analyzing sequence characteristics before and after a mutation site6Conserved sequence of A site, which is related to the common m6The antibody A immunoprecipitates and the conserved sequences obtained by sequencing technology are approximately the same, which indicates that the site identified by the method is m6A site (as in FIG. 7).
(15) The high throughput sequencing results are shown in FIG. 8, and the sequencing results of the mutations show that LATS1 (NM-001350339) has m at the two sites 2970 and 29916Modification A, ZNF445 (NM-181489) has m at 3451 and 3462 sites6A modification, OTUD1 (NM-001145373) has m at position 14796And (C) modifying.
5. Performing low-flux sequencing on the RNA fragment subjected to iodine addition and cyclization treatment by using a TA cloning technology, verifying mutation of a specific site on the mRNA of the HeLa cell, and further identifying m6A site
(1) Mixing 1 μ L of the above RNA (10 ng/. mu.L) subjected to iodine addition and cyclization treatment with 1 μ L of 10 μ M reverse primer (R-primer) and 12 μ L of RNase-free water, pipetting, mixing, heating at 65 deg.C for denaturation for 5 min, wherein the sequence of the reverse primer (R-primer) is shown in Table 12;
TABLE 12 Forward primer (F-primer) and reverse primer (R-primer) sequences
Figure GDA0002998669840000182
(2) After cooling on ice, 4. mu.L of 5 XTRT Reaction Buffer (5 XT Reaction Buffer, Thermofisiher, EP0441), 1. mu.L of 10mM dNTP, 1. mu.L of 10U/. mu.L HIV Reverse Transcriptase (Reverse Transcriptase Recombinant HIV, Worthington Biochemical Corporation) was added, reacted at 37 ℃ for 1 hour, followed by heating at 70 ℃ for 10 minutes to inactivate the Reverse Transcriptase;
(3) mu.L of the reverse transcription reaction solution was transferred to a new PCR tube, PCR amplification was performed with KOD-FX (TOYOBO, KFX-101) enzyme, 10. mu.L of 2mM dNTP, 1.5. mu.L of 10. mu.M forward primer (F-primer) and reverse primer (R-primer), 25. mu.L of 2 XKOD-FX reaction buffer, and 1. mu.L of KOD-FX enzyme were added, mixed and blown to mix, the sequences of the forward primer (F-primer) and reverse primer (R-primer) are shown in Table 5, and the PCR reaction program is shown in Table 13;
TABLE 13 post reverse transcription PCR amplification reaction procedure
Figure GDA0002998669840000191
(4) After the PCR reaction, the product DNA was purified with 1.8-fold volume of AMPure XP beads (BECKMAN COULTER, A63881), and the DNA was dissolved with 15. mu.L of LRNase-free water to a concentration of 50 ng/. mu.L;
(5) t Vector ligation of 1. mu.L of the above DNA (50 ng/. mu.L) product was performed using pUCM-T Vector (Sangon Biotech, B522213) and reacted at 16 ℃ for 6 hours, and the pUCM-T Vector ligation system is shown in Table 14;
TABLE 14 pUCm-T Vector ligation System
Figure GDA0002998669840000192
(6) Thawing 100 μ L DH5 α competent cells on ice for 5 min, suspending the cells uniformly, adding 5 μ L of the above connecting solution, beating gently, mixing well, and standing on ice for 25 min;
(7) performing heat shock in 42 ℃ water bath for 45 seconds, then placing on ice for 2 minutes, adding 700 mu L of SOC culture medium, and performing shake culture at 37 ℃ and 220rpm for 1 hour;
(8) centrifuging at 5000rpm for 1 min, sucking out 600 μ L of supernatant with a pipette tip, and suspending the cells with the rest of the culture medium;
(9) preparing 50mL LB plate containing 50. mu.L IPTG (100mM), 100. mu. L X-gal (20mg/mL), 50. mu.L 1000 Xampicillin, 15mL each, and uniformly spreading the bacterial suspension on the LB plate;
(10) firstly, culturing the plate at 37 ℃ for 1 hour, then culturing the plate in an inverted mode for 16 hours, screening white single colony extraction plasmids in a blue-white spot, and carrying out Sanger sequencing to obtain a low-pass sequencing result;
(11) the low-throughput sequencing results are identical to the high-throughput sequencing results. As shown in FIG. 10, the results of the mutation showed that LATS1 (NM-001350339) has m at two sites 2970 and 29916Modification A, ZNF445 (NM-181489) has m at 3451 and 3462 sites6A modification, OTUD1 (NM-001145373) has m at position 14796And (C) modifying.
Example 2m on transcriptome mRNA in mouse H2.35 cells6Detection of A modification sites
1. Culture of mouse H2.35 cells, allyl labeling and mRNA extraction
(1) Mouse H2.35 cells were cultured under normal culture conditions (with the addition of 200nM DEX, dexamethasone) to sixty percent fullness, the medium was aspirated, and the residual medium was washed away with Phosphate Buffered Saline (PBS);
(2) adding 10% Fetal Bovine Serum (FBS), 1% 100 Xpenicillin-streptomycin double antibody, 200nM DEX (dexamethasone), and 1mM cysteine to a methionine-free medium, culturing the above mouse H2.35 cells in the medium for 30 minutes, and removing methionine remaining in the cells;
(3) after methionine was removed, 1mM allyl-L-seleno/thiohomocysteine was added to the medium for culturing the above mouse H2.35 cells, and the culture was continued for 16 hours;
(4) - (12) culturing HeLa cells, allyl labeling and mRNA extraction as in example 1.
2. The same as example 1, which contains adenine N6And enriching the RNA modified by allyl.
3. N of RNA as in example 16Iodine addition reaction and cyclization treatment of allyl adenine.
4. In the same example 1, RNA fragments obtained by non-immunoprecipitation and immunoprecipitation (including cyclization treatment and non-cyclization treatment) were used to construct a library using the illumina mRNA library construction kit.
5. The high throughput sequencing results are shown in FIG. 9, and based on the mutation sequencing results, Xist (NR _001463) has m at both positions 11956 and 119646Modification A, Usp42 (NM-029749) with m at position 29736Modification A, Ice1 (NM-144837) has m at position 37776A modification, Eppk1 (NM-144848) has m at two positions 2899 and 29246And (C) modifying.
6. Or performing low-throughput sequencing on the RNA fragment subjected to iodine addition and cyclization treatment by using TA cloning technology in the same way as in example 1, verifying mutation of specific site on HeLa cell mRNA, and further identifying m6And (3) A site.
Example 3 culturing HeLa cells and mouse H2.35 cells under Normal cell culture conditions
Culture of HeLa cells
Adding 10% Fetal Bovine Serum (FBS) and 1% 100x penicillin-streptomycin double antibody into normal cell culture medium, culturing HeLa cell in the culture medium for 16-24 hr to extract mRNA of HeLa cell6The A content is shown as Ctrl in FIG. 2.
Simultaneously, HeLa cells were cultured under the above-mentioned allyl-labeling method, 1mM allyl-L-seleno, allyl-L-thiohomocysteine, or methionine (methyl-L-thiohomocysteine) was added thereto, and the cells were cultured for 16 hours to obtain m of mRNA6The content of A is shown in FIG. 2 as Se-allyl-L-selenohomocysteine, S-allyl-L-homocysteine, and L-Methionine. In FIG. 2, Se-allyl-L-selenohomocysteine/S-allyl-L-homocysteine is obtained by introducing allyl-L-seleno/thiohomocysteine into HeLa cell for metabolism6Content of allyl adenosine, L-Methionine Methionine introduced into HeLa cell for metabolism to obtain N in mRNA6-methyladenine content, Ctrl is N in mRNA obtained after HeLa cells are normally cultured6The content of-methyladenine nucleoside shows that the labeling efficiency of the allyl-L-seleno-homocysteine is higher than that of the thio-homocysteine, and the labeling level is far lower than m in normal cultured cells6A level, and after half an hour of the treatment to remove intracellular methionine, methionine is added again, m of the cell6A level is nearly normal, which indicates thatThe treatment for removing methionine in a short time did not have an excessive influence on the cell state.
2. Culture of mouse H2.35 cells
The mRNA of H2.35 cells was extracted by culturing H2.35 cells in a normal cell culture medium containing 10% Fetal Bovine Serum (FBS), 1% 100 Xpenicillin-streptomycin diabody, 200nM DEX (dexamethasone) for 16-24 hours.
HeLa cells were cultured under the same conditions as described above for the allyl labeling method, and 1mM of allyl-L-seleno, allyl-L-thiohomocysteine, or methionine (methyl-L-thiohomocysteine) was added thereto and cultured for 16 hours to obtain m of each group of mRNA6A. The results were similar to those of HeLa cells, and the labeling efficiency of allyl-L-seleno-homocysteine was higher than that of thio-cysteine, and the labeling level was much lower than that of m in normal cultured cells6A level, and after half an hour of the treatment to remove intracellular methionine, methionine is added again, m of the cell6The level of A was nearly normal, indicating that the treatment to remove methionine in a short time did not have an excessive effect on the cell status.

Claims (8)

1. RNA N detection with single base resolution in full transcriptome range6-a method for methyladenine modification, characterized in that it comprises the following steps:
(1) allyl-L-seleno/thiohomocysteine labeling nucleic acid adenine: treatment of cells with the methionine analogue allyl-L-seleno/thiohomocysteine, which introduces an allyl group into specific adenine N of intracellular RNA by natural metabolism6At the position of methyladenine to form N6-allyladenine a6A;
(2) Containing a6Enrichment of a-modified RNA: extracting total RNA of cells, further extracting mRNA of the cells, then breaking the total transcriptome RNA of the cells into fragmented RNA of 100-300 nt, and combining N with an antibody6-allyladenosine enrichment of a by immunoprecipitation6A modified RNA and elution from the antibody with an eluent a6A is the modified RNA of the RNA gene,purifying the eluted RNA;
(3)RNA a6n on A6Iodine addition and cyclization treatment of allyladenine: RNA a6N on A6Iodine addition reaction of allyl adenine and subsequent induction of RNA a under alkaline conditions6N on A6N of allyl adenine1,N6Forming a cyclization structure at the site, and shielding base complementary pairing;
(4) reverse transcription mutation and sequencing recognition of circularized RNA: adding HIV reverse transcriptase, N on RNA to the circularized structure obtained in step (3)1,N6In the process of reverse transcription of cyclic adenine into DNA under the action of HIV reverse transcriptase, errors are generated by introducing para-complementary base, and mutation sites are identified by means of nucleic acid sequencing, so that a is obtained6A site, which is the original m in cellular RNA6A site of modification;
the methods are not diagnostic or therapeutic methods for disease.
2. The full transcriptome-wide single base resolution detection of RNA N of claim 16The method for modifying methyl adenine is characterized in that in the step (1), the preparation method of allyl-L-seleno/thiohomocysteine comprises the following steps: under the protection of nitrogen, taking selenium powder/sulfur powder and sodium borohydride with a molar ratio of 1:1 as raw materials, taking ethanol as a solvent, and heating and refluxing at 80 ℃ for reaction for 6-24 hours; adding hydrogen bromate of (S) - (+) -2-amino-4-bromobutyric acid with the molar weight of one half to one third of selenium powder/sulfur powder, continuously heating and refluxing for 6-24 hours at 80 ℃, adding acid to stop the reaction, filtering to remove insoluble substances, washing off by-products by using ether, and adjusting the pH value to be neutral; and then adding sodium borohydride with the molar weight 1-2 times that of the selenium powder/sulfur powder to reduce diselenide/disulfide bonds, adding sodium bicarbonate/sodium carbonate with the molar weight 0.5-2 times that of the selenium powder/sulfur powder and allyl bromide with the molar weight 0.5-1.5 times that of the selenium powder/sulfur powder to react at room temperature for 6-24 hours, and purifying by high performance liquid chromatography to obtain the allyl-L-seleno/thiohomocysteine.
3. According toThe full transcriptome-wide single base resolution RNA N detection method of claim 16The method for modifying the methyl adenine is characterized in that the specific method for treating the cells in the step (1) is to culture the cells by adopting a methionine-deficient culture medium, add 0.1-2 mM of cysteine for half an hour, then add 0.1-2 mM of allyl-L-seleno/thiohomocysteine and culture for 12-24 hours.
4. The full transcriptome-wide single base resolution detection of RNA N of claim 16-methyladenine modification method, characterized in that in step (2), said cellular whole transcriptome RNA fragmentation is by Zn2+The ions are heated for 5-10 minutes at 70 ℃.
5. The full transcriptome-wide single base resolution detection of RNA N of claim 16-methyladenine modification method, characterized in that, in step (2), it is used to bind N6-allyladenosine antibody is N6Antibodies to isopentenyl adenosine, 10 μ g of antibodies were used to enrich for 1-20 μ g of fragmented RNA.
6. The full transcriptome-wide single base resolution detection of RNA N of claim 16-methyladenine modification method, wherein in step (2), the eluent is N6Adenosine triphosphate, at a concentration of 5 to 10 mM.
7. The full transcriptome-wide single base resolution detection of RNA N of claim 16-a method for modifying methyladenine, characterized in that said step (3) is specifically: dissolving 0.1-0.5M iodine simple substance in 0.2-1M potassium iodide to obtain potassium iodide solution of iodine, and mixing the potassium iodide solution of iodine with RNA a6N on A6-allylic reaction of allyladenine followed by removal of excess iodine with 0.1-0.5M sodium thiosulfate; adding 0.1-0.5M sodium carbonate, adjusting the pH value to 9-10, and inducing RNA a6N on A6-allyl adenineN of pterin1,N6The site forms a cyclic structure, thus shielding the normal hydrogen bonding pairing of adenine.
8. The full transcriptome-wide single base resolution detection of RNA N of claim 16-a method for methyladenine modification, characterized in that said step (4) is in particular: i) reverse transcribing the iodine addition-cyclized RNA using HIV reverse transcriptase; ii) adopting RNA library preparation technology and high-throughput sequencing means to carry out identification on the mutation sites of the whole transcriptome so as to obtain m with single base resolution of the whole transcriptome6And (3) distributing the A sites, or identifying and verifying the mutation sites on a specific transcript by adopting PCR and TA-cloning technologies.
CN201910822658.3A 2019-09-02 2019-09-02 Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range Active CN111154837B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201910822658.3A CN111154837B (en) 2019-09-02 2019-09-02 Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range
PCT/CN2020/102643 WO2021042883A1 (en) 2019-09-02 2020-07-17 Method and kit for detecting rna n6-methyladenine modification at single-base resolution in range of whole transcriptome

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910822658.3A CN111154837B (en) 2019-09-02 2019-09-02 Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range

Publications (2)

Publication Number Publication Date
CN111154837A CN111154837A (en) 2020-05-15
CN111154837B true CN111154837B (en) 2021-05-18

Family

ID=70555634

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910822658.3A Active CN111154837B (en) 2019-09-02 2019-09-02 Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range

Country Status (2)

Country Link
CN (1) CN111154837B (en)
WO (1) WO2021042883A1 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112176043B (en) * 2019-07-04 2022-07-12 北京大学 Sequencing, enrichment and detection method of modified nucleoside based on chemical marker
CN111154837B (en) * 2019-09-02 2021-05-18 浙江大学 Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range
CN111560423B (en) * 2020-06-05 2023-11-24 中山大学孙逸仙纪念医院 Method for detecting RNA m6A with high flux and high sensitivity and single base resolution and application thereof
CN112301118B (en) * 2020-10-30 2022-05-13 浙江大学 Method and kit for simultaneously obtaining RNA abundance and active RNA polymerase sites in full transcriptome range
CN114507721B (en) * 2020-11-16 2024-04-09 寻鲸生科(北京)智能技术有限公司 Method for detecting full transcriptome RNA structure and application thereof
CN113061648B (en) * 2021-03-24 2022-04-19 中山大学 Method for constructing micro sample m6A modification detection library by aid of Tn5 transposase and application of method
EP4353832A1 (en) * 2021-06-11 2024-04-17 Peking University Method and kit for detecting n6-methyladenosine in nucleic acid molecules
CN114480670B (en) * 2022-01-26 2022-12-06 西北农林科技大学 DNA methylases TET1 and m related to bovine skeletal muscle differentiation 6 Identification and application of A methylase METTL3 interaction
CN114672545B (en) * 2022-04-24 2024-02-23 华中科技大学 Method for detecting N6-isopentenyl adenine modification in RNA with single base resolution
CN117165664B (en) * 2023-10-24 2024-03-15 河北大学 Method for detecting methylation of sixth nitrogen atom of adenylate in DRACH motif at single base level

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106047997A (en) * 2016-05-27 2016-10-26 浙江大学 MRNA methylation high-throughput test method

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4886679B2 (en) * 2004-06-14 2012-02-29 マックス−プランク−ゲゼルシャフト ツール フォーデルング デル ヴィッセンシャフテン エー.ヴェー. Sequence-specific detection of methylation in biomolecules
CN101240341B (en) * 2008-03-14 2010-06-09 东南大学 DNA sequencing method by using thiooligonucleotide probe
CN102533944B (en) * 2010-12-10 2014-04-09 深圳华大基因科技服务有限公司 Hemi methylation connector for enrichment and sequencing of methylation deoxyribonucleic acid (DNA) and application thereof
GB201314695D0 (en) * 2013-08-16 2013-10-02 Oxford Nanopore Tech Ltd Method
CN110029149A (en) * 2019-04-17 2019-07-19 中山大学 A method of identification base modification
CN111154837B (en) * 2019-09-02 2021-05-18 浙江大学 Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106047997A (en) * 2016-05-27 2016-10-26 浙江大学 MRNA methylation high-throughput test method

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
A metabolic labeling method detects m6A transcriptome-wide at single base resolution;Xiao Shu等;《Nature Chemical Biology》;20200427;第16卷;第887-895页 *
Facile Chemoenzymatic Strategies for the Synthesis and Utilization of S-Adenosyl-l-Methionine Analogues;Shanteri Singh等;《Angew. Chem. Int. Ed.》;20140311;第53卷;第3965-3969页 *
N6-Allyladenosine: A New Small Molecule for RNA Labeling Identified by Mutation Assay;Xiao Shu等;《J. Am. Chem. Soc.》;20171108;第139卷;第17213-17216页 *

Also Published As

Publication number Publication date
CN111154837A (en) 2020-05-15
WO2021042883A1 (en) 2021-03-11

Similar Documents

Publication Publication Date Title
CN111154837B (en) Method for detecting RNA N6-methyladenine modification in single base resolution in full transcriptome range
US20210230578A1 (en) Removal of dna fragments in mrna production process
EP4279609A1 (en) Method and kit for labeling nucleic acid molecules
Weinberg et al. Reconstitution of simian virus 40 DNA replication with purified proteins.
Chang et al. Induction of DNA polymerase in mouse L cells
Moeys et al. A sex-inducing pheromone triggers cell cycle arrest and mate attraction in the diatom Seminavis robusta
WO2003002758A1 (en) Nucleic acid ligands to complex targets
US20230022745A1 (en) Methods for Labeling a Population of RNA Molecules
CN106957906B (en) Primer combination and kit applied to high-throughput sequencing detection of T cell leukemia minimal residual disease
Wong-Staal et al. Murine intracisternal type A particles: a biochemical characterization
CN111705135A (en) Method for detecting MGMT promoter region methylation
US20230332219A1 (en) Chemical tagging-based method for modified nucleoside sequencing, enrichment, and measurement
CN110066863B (en) Identification method of BPDE addition gene
CN114149976B (en) Eukaryotic cell S-adenosylmethionine concentration sensing fluorescence reporting system
KR20010103630A (en) Method and test kit for analyzing dna repair
Hilkens et al. Identification of a mouse gene required for binding of Rauscher MuLV envelope gp70
CN113943727A (en) Composition, kit and purification method thereof
CN114807328A (en) Method for marking and identifying single-base resolution DNA
WO2021041519A1 (en) Methods of performing guide-seq on primary human t cells
CN112301118B (en) Method and kit for simultaneously obtaining RNA abundance and active RNA polymerase sites in full transcriptome range
US10370669B2 (en) Nucleic acid compounds for binding growth differentiation factor 8
CN117126921B (en) Library construction method for detecting T cell and B cell immune repertoire, primer and kit thereof
US11535852B2 (en) Nucleic acid compounds for binding growth differentiation factor 11
Washington et al. A comparison of nucleoside (β-S) triphosphates and nucleoside (γ-S) triphosphates as suitable substrates for measuring transcription initiation in preparations of cell nuclei
EP3412777A1 (en) Method for measuring target dna

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