CN113155577B - Preparation method for identifying specific protein acetylation modified sample - Google Patents

Preparation method for identifying specific protein acetylation modified sample Download PDF

Info

Publication number
CN113155577B
CN113155577B CN202110499285.8A CN202110499285A CN113155577B CN 113155577 B CN113155577 B CN 113155577B CN 202110499285 A CN202110499285 A CN 202110499285A CN 113155577 B CN113155577 B CN 113155577B
Authority
CN
China
Prior art keywords
plasmid
protein
transfected
cells
sample
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
CN202110499285.8A
Other languages
Chinese (zh)
Other versions
CN113155577A (en
Inventor
王居平
彭慧
农安娜
黄月艳
高洁
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.)
Youjiang Medical University for Nationalities
Original Assignee
Youjiang Medical University for Nationalities
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 Youjiang Medical University for Nationalities filed Critical Youjiang Medical University for Nationalities
Priority to CN202110499285.8A priority Critical patent/CN113155577B/en
Publication of CN113155577A publication Critical patent/CN113155577A/en
Application granted granted Critical
Publication of CN113155577B publication Critical patent/CN113155577B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/30Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N27/00Investigating or analysing materials by the use of electric, electrochemical, or magnetic means
    • G01N27/62Investigating or analysing materials by the use of electric, electrochemical, or magnetic means by investigating the ionisation of gases, e.g. aerosols; by investigating electric discharges, e.g. emission of cathode
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins

Abstract

The invention belongs to the technical field of biology, and particularly discloses a preparation method for identifying a specific protein acetylation modified sample. The preparation method comprises the following steps: 1) Selecting protein A with acetylation modification and acetylase B; 2) Preparing protein samples of co-transfected plasmids A and B and protein samples of separately transfected plasmid A; 3) Selecting a protein sample of co-transfected plasmid A and plasmid B and separately transfecting the protein sample of plasmid A to complete immunoblotting analysis; 4) And (3) after the protein samples of the other co-transfected plasmids A and B are subjected to SDS-PAGE gel, incubating overnight in coomassie brilliant blue dye solution, decoloring, cutting off a target strip, and placing the target strip into a centrifuge tube to obtain the sample for identifying the specific protein acetylation modification. The protein sample prepared by the method can obviously enhance the success rate of identifying the specific protein acetylation modification site by mass spectrometry.

Description

Preparation method for identifying specific protein acetylation modified sample
[ field of technology ]
The invention relates to the field of biotechnology, in particular to a preparation method for identifying a specific protein acetylation modified sample.
[ background Art ]
Post-translational modification (Post-translational modification, PTM) of proteins refers to chemical modification of proteins after translation, and takes part in almost all normal vital activity processes of cells and plays a very important regulatory role. PTM is a means of protein function regulation and is critical to the structure and function of the protein. Studies have shown that 50% -90% of proteins in humans are post-translationally modified. More than 400 post-translational modifications have been identified, and common modifications include methylation, phosphorylation, ubiquitination, acetylation, glycosylation, SUMO, nitrosylation, oxidation, and the like. Therefore, PTM has become an extremely important field of international protein research. Among these, protein acetylation is a ubiquitous, reversible and highly regulated post-translational modification of proteins, mainly at the epsilon-NH 2 position of protein lysine residues. The acetylation modification is regulated by both acetyl transferase and deacetylase and is involved in almost all biological processes such as transcription, stress, metabolism and protein synthesis and degradation.
Detection of protein acetylation modification is different from phosphorylation, which can be studied by in situ phosphorylation of labels, sensitive phosphorylation-specific antibodies, etc. stable and effective means, and is relatively easy to detect. While the means and methods for detection of protein acetylation modification are very limited, these technical difficulties limit the research and development of protein acetylation.
The main methods for preparing and detecting protein lysine acetylation at present comprise the following steps: (1) Enriching the acetylated peptide fragment by using a lysine acetylation specific antibody, and detecting by using a liquid chromatography-mass spectrometry (HPLC/MS) method; (2) The identification of protein acetylation is completed by adopting a stable isotope labeled amino acid (stable isotope labeling with aminoacids in cell culture, SILAC) technology under the cell culture condition and a high-resolution and high-sensitivity electric field Orbitrap cyclotron resonance combination (LTQ Orbitrap) mass spectrometer. However, all of the above methods are based on proteomics level study of lysine acetylation modification, and cannot be used to detect the acetylation modification of a specific protein.
[ invention ]
In view of the foregoing, it is desirable to provide a method for identifying specific protein acetylation modified samples, wherein the protein samples prepared by the method of the present invention can significantly enhance the success rate of mass spectrometry for identifying specific protein acetylation modified sites.
In order to achieve the above purpose, the technical scheme adopted by the invention is as follows:
a method of preparing a sample for identifying specific protein acetylation modifications, comprising the steps of:
1) Selecting protein A with acetylation modification through an endogenous experiment, and determining an acetylase B of the protein A through an exogenous experiment;
2) Protein samples of co-transfected plasmid a and plasmid B were prepared, and protein samples of plasmid a were transfected alone:
(1) Respectively inoculating 293T cells in at least 20 cell culture dishes, and when the cell density reaches 78-82%, independently transfecting the cells of one cell culture dish with the plasmid A, and simultaneously, co-transfecting the cells of the other cell culture dishes with the plasmid A and the plasmid B by using the transfection reagent;
(2) Placing the transfected cell culture dish into an incubator, culturing for 6 hours at constant temperature and constant carbon dioxide concentration, removing the culture solution containing the transfection reagent-DNA complex, and replacing the new culture solution for continuous culturing for 48 hours;
(3) Collecting the cultured cells in a first centrifuge tube, centrifuging the cell sediment, and adding RIPA lysate into the first centrifuge tube;
(4) Placing the first centrifuge tube on a vertical rotary shaking table for cracking for 1 hour, centrifuging, transferring cell supernatant obtained by centrifuging into a second centrifuge tube, adding an antibody into the second centrifuge tube, and incubating on a vertical rotary shaking table overnight;
(5) Adding agarose beads into the second centrifuge tube the next day, and then continuously incubating on a vertical rotary table for overnight;
(6) Centrifuging the antigen-antibody-agarose bead complex in the second centrifuge tube on the third day, washing 3 times with cold RIPA lysate, and sucking the liquid after the last washing;
(7) Adding a double-concentration loading buffer solution into the obtained compound, boiling at 100 ℃ for 5-8min, and performing instantaneous centrifugation to obtain a protein sample of the single transfected plasmid A and protein samples of at least 19 co-transfected plasmids A and B;
3) Randomly extracting 1 protein sample from the protein samples of the cotransfected plasmid A and the plasmid B, and performing immunoblotting analysis together with the protein sample of the cotransfected plasmid A alone;
4) After running the remaining protein samples of co-transfected plasmid a and plasmid B on SDS-PAGE gel, incubating overnight in coomassie blue dye solution, the next day the gel was decolorized on a shaker until the background of the gel was white and the protein bands in the lanes, particularly the target bands, were blue and the decolorization was terminated; and cutting off a target strip according to the molecular weight of the A, putting the target strip into a third centrifuge tube, and marking to obtain the sample for identifying the specific protein acetylation modification.
Further, in step (1) of step 2), the 293T cells were seeded at an amount of 5.2X10 6 The/each cell culture dish.
Further, in step (1) of step 2), the transfection reagent is an EZ Trans transfection reagent.
Further, in step (2) of step 2), the constant temperature is 37 ℃, and the concentration of carbon dioxide is 5%.
Further, in step (1) of step 2), after the cell density reaches 80%, plasmid A alone transfects cells of one cell culture dish with the transfection reagent, while plasmid A and plasmid B are co-transfected with the transfection reagent into cells of the remaining cell culture dish.
Further, in step 1), the exogenous experiment is: plasmid a and plasmid B were co-transfected in 293T cells, cells were harvested after 48h, first subjected to co-immunoprecipitation and then subjected to immunoblot analysis.
The invention has the following beneficial effects:
firstly, determining that the selected protein has acetylation modification through an endogenous experiment, further determining that the selected protein has acetylation modification through an exogenous experiment, determining that the accuracy reaches 100% in a double manner, and determining that the protein has acetylation transferase through the exogenous experiment; secondly, the protein samples which are subjected to and not subjected to acetylation modification are effectively enriched through an IP technology; thirdly, before mass spectrum identification is carried out on samples, the invention ensures that all samples to be detected are subjected to acetylation modification through immunoblotting analysis, and invalid samples are avoided; fourth, protein samples identified by mass spectrometry are tailored such that they have sufficient abundance to more readily identify sites of acetylation modification. Through the organic combination of the steps, the protein sample prepared by the preparation method can obviously enhance the success rate of identifying the specific protein acetylation modification site by mass spectrometry.
[ description of the drawings ]
FIG. 1 is a graph showing the results of the present invention ODN inducing an acetylation modification of endogenous TLR9 (i.e., graph showing the results of endogenous experiments).
Fig. 2 is a graph of the detection results of exogenous acetylation modification of TLR9 of the invention (i.e., exogenous experimental results).
FIG. 3 is a graph showing the results of an acetylation modification assay performed on randomly sampled protein samples using immunoblot analysis techniques.
FIG. 4 is a Coomassie brilliant blue staining chart of protein samples after running gel co-transfected with HA-A and HA-B.
Fig. 5-11 are secondary mass spectra of TLR9 acetylation sites.
The invention will be further described in the following detailed description in conjunction with the above-described figures.
[ detailed description ] of the invention
In a preferred embodiment of the invention, the identification of the site of the acetylation modification of TLR9 is exemplified.
A method of preparing a sample for identifying specific protein acetylation modifications, comprising the steps of:
1) Determining that the TLR9 has acetylation modification through an endogenous experiment, and determining the acetylase CBP of the TLR9 through an exogenous experiment;
wherein, endogenous experiments specifically are: mice macrophages raw264.7 were stimulated with the same concentration of TLR9 receptor activator ODN (0.5 μm) at different time points (0 min, 15min, 30 min), and the above cells were stimulated with the addition of a deacetylase inhibitor (Trichostatin a, TSA) based on ODN stimulation for 30min, and then the cells were collected for Immunoprecipitation (IP) and immunoblot (WB) analyses, the experimental results are shown in fig. 1: as can be seen from fig. 1, along with the extension of the ODN stimulation time, the TLR9 acetylation level showed a gradual increase trend, and TSA can significantly enhance the ODN-induced TLR9 acetylation level, demonstrating that TLR9 has an acetylation modification;
the exogenous experiment is specifically as follows: in 293T cells, HA-TLR9 and HA-CBP plasmids were co-transfected, cells were harvested after 48h, immunoblot analysis was performed after co-immunoprecipitation was completed, and experimental results are shown in FIG. 2: from fig. 2, it can be seen that the acetylase CBP mediates the acetylating modification of TLR9, again demonstrating that TLR9 (i.e. protein a) has an acetylating modification and that CBP (i.e. acetylase B) is an acetylase of TLR 9;
2) Protein samples co-transfected with HA-TLR9 and HA-CBP were prepared, protein samples transfected with HA-TLR9 alone:
(1) Inoculating 5.2X10 to 20 6cm cell culture dishes 6 The next day when the cell density reached 80%, cells of one of the cell culture dishes were transfected with plasmid 8ug of HA-TLR9 alone with 24ul of EZ Trans transfection reagent, while cells of the remaining 19 cell culture dishes were co-transfected with 4ug of plasmid HA-TLR9 and 4ug of plasmid HA-CBP with 24ul of EZ Trans transfection reagent;
(2) Placing the transfected cell culture dish into an incubator, and adding CO with concentration of 5% at 37 DEG C 2 After culturing for 6 hours, removing the culture solution containing the EZ Trans-DNA complex, and replacing the new culture solution for culturing for 48 hours;
(3) Collecting the cultured cells in a 1.5mL first centrifuge tube, centrifuging the cell sediment at a rotation speed of 5000 revolutions at 4 ℃ for 10min, and adding 500ul of RIPA lysate into each first centrifuge tube;
(4) Placing the first centrifugal tube on a vertical rotary shaking table, cracking for 1h at 4 ℃, centrifuging for 20min at 20000 revolutions at 4 ℃, transferring the cell supernatant obtained by centrifugation into a 1.5mL second centrifugal tube, adding 4 mu L of HA antibody into the second centrifugal tube, and incubating on the vertical rotary shaking table overnight;
(5) After the next day 40ul of Agarose beads (Protein G PLUS-Agarose) was added to the second centrifuge tube, incubated overnight on a vertically rotating shaker at 4 ℃;
(6) Centrifuging the antigen-antibody-agarose bead complex in the second centrifuge tube on the third day, washing 3 times with cold RIPA lysate, and sucking the liquid after the last washing;
(7) Adding 20ul of twice-concentration loading buffer solution into the obtained compound, boiling for 5min at 100 ℃, and performing instantaneous centrifugation to obtain a protein sample of separately transfected HA-A and 19 protein samples of co-transfected HA-TLR9 and HA-CBP, wherein the obtained protein samples can be directly used for WB analysis or frozen in a refrigerator at-80 ℃ for later use;
3) 1 protein sample was randomly extracted from the above 19 protein samples co-transfected with HA-TLR9 and HA-CBP, and immunoblot analysis was performed together with the protein sample transfected with HA-TLR9 alone, and the detection analysis results are shown in FIG. 3: as can be seen from fig. 3, protein samples co-transfected with HA-TLR9 and HA-CBP have undergone acetylation modification;
4) After the remaining 18 protein samples co-transfected with HA-TLR9 and HA-CBP were placed on five lanes on an SDS-PAGE gel to start running, the gel was decolorized on a shaker the next day after incubation overnight in coomassie blue dye solution until the background of the gel was white and the protein bands in the lanes, in particular the band of interest, were blue and the decolorization terminated, the results are shown in fig. 4; and cutting off a target strip according to the molecular weight of the A, putting the target strip into a third centrifuge tube, and marking to obtain the sample for identifying the specific protein acetylation modification.
The prepared sample for identifying the specific protein acetylation modification is sent to qualified biological company for TLR9 acetylation modification site mass spectrometry (LCMS) identification, and the specific identification process comprises the following steps: the sample enzymolysis, mass spectrometry data acquisition and data analysis finally identify 7 TLR9 acetylation modification sites, as shown in figures 5-11.
The foregoing description is directed to the preferred embodiments of the present invention, but the embodiments are not intended to limit the scope of the invention, and all equivalent changes or modifications made under the technical spirit of the present invention should be construed to fall within the scope of the present invention.

Claims (6)

1. A method for preparing a sample for identifying specific protein acetylation modifications, comprising the steps of:
1) Selecting protein A with acetylation modification through an endogenous experiment, and determining an acetylase B of the protein A through an exogenous experiment;
2) Protein samples of co-transfected plasmid a and plasmid B were prepared, and protein samples of plasmid a were transfected alone:
(1) Respectively inoculating 293T cells in at least 20 cell culture dishes, and when the cell density reaches 78-82%, independently transfecting the cells of one cell culture dish with the plasmid A, and simultaneously, co-transfecting the cells of the other cell culture dishes with the plasmid A and the plasmid B by using the transfection reagent;
(2) Placing the transfected cell culture dish into an incubator, culturing for 6 hours at constant temperature and constant carbon dioxide concentration, removing the culture solution containing the transfection reagent-DNA complex, and replacing the new culture solution for continuous culturing for 48 hours;
(3) Collecting the cultured cells in a first centrifuge tube, centrifuging the cell sediment, and adding RIPA lysate into the first centrifuge tube;
(4) Placing the first centrifuge tube on a vertical rotary shaking table for cracking for 1 hour, centrifuging, transferring cell supernatant obtained by centrifuging into a second centrifuge tube, adding an antibody into the second centrifuge tube, and incubating on a vertical rotary shaking table overnight;
(5) Adding agarose beads into the second centrifuge tube the next day, and then continuously incubating on a vertical rotary table for overnight;
(6) Centrifuging the antigen-antibody-agarose bead complex in the second centrifuge tube on the third day, washing 3 times with cold RIPA lysate, and sucking the liquid after the last washing;
(7) Adding a double-concentration loading buffer solution into the obtained compound, boiling at 100 ℃ for 5-8min, and performing instantaneous centrifugation to obtain a protein sample of the single transfected plasmid A and protein samples of at least 19 co-transfected plasmids A and B;
3) Randomly extracting 1 protein sample from the protein samples of the cotransfected plasmid A and the plasmid B, and performing immunoblotting analysis together with the protein sample of the cotransfected plasmid A alone;
4) After running the remaining protein samples of co-transfected plasmid a and plasmid B on SDS-PAGE gel, incubating overnight in coomassie blue dye solution, decolorizing the gel on a shaker the next day until the background of the gel is white and the protein band in the lane is blue and stopping decolorizing; and cutting off a target strip according to the molecular weight of the A, putting the target strip into a third centrifuge tube, and marking to obtain the sample for identifying the specific protein acetylation modification.
2. The method of claim 1, wherein the method comprises the step of: in step (1) of step 2), the 293T cells were seeded at a rate of 5.2X10 6 The/each cell culture dish.
3. The method of claim 1, wherein the method comprises the step of: in step (1) of 2), the transfection reagent is an EZ Trans transfection reagent.
4. The method of claim 1, wherein the method comprises the step of: in step (2) of step 2), the constant temperature is 37 ℃, and the concentration of carbon dioxide is 5%.
5. The method of claim 1, wherein the method comprises the step of: in step (1) of step 2), after the cell density reaches 80%, plasmid A alone transfects cells of one cell culture dish with the transfection reagent, while plasmid A and plasmid B are co-transfected with the transfection reagent into cells of the remaining cell culture dish.
6. The method of claim 1, wherein the method comprises the step of: in step 1), the exogenous experiment is: plasmid a and plasmid B were co-transfected in 293T cells, cells were harvested after 48h, first subjected to co-immunoprecipitation and then subjected to immunoblot analysis.
CN202110499285.8A 2021-05-08 2021-05-08 Preparation method for identifying specific protein acetylation modified sample Active CN113155577B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110499285.8A CN113155577B (en) 2021-05-08 2021-05-08 Preparation method for identifying specific protein acetylation modified sample

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110499285.8A CN113155577B (en) 2021-05-08 2021-05-08 Preparation method for identifying specific protein acetylation modified sample

Publications (2)

Publication Number Publication Date
CN113155577A CN113155577A (en) 2021-07-23
CN113155577B true CN113155577B (en) 2023-07-25

Family

ID=76873792

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110499285.8A Active CN113155577B (en) 2021-05-08 2021-05-08 Preparation method for identifying specific protein acetylation modified sample

Country Status (1)

Country Link
CN (1) CN113155577B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113249382B (en) * 2021-04-12 2023-05-12 右江民族医学院 SiRNA for down regulating TRIM56 gene expression and application thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003102196A1 (en) * 2002-05-30 2003-12-11 Centre National De La Recherche Scientifique (Cnrs) Vectors for expression of biotinylated proteins in mammalian cells, and their use for identification of protein-nucleic acid interactions in vivo
CN101787075A (en) * 2009-09-29 2010-07-28 天津医科大学附属肿瘤医院 Amino acid sequence with characteristic of bonding histone deacetylase 1 and expression vector thereof
CN103900893A (en) * 2013-05-20 2014-07-02 上海华盈生物医药科技有限公司 Kit for gathering protein modified by acetylization, as well as method and application of kit
CN107024588A (en) * 2016-02-01 2017-08-08 上海生物芯片有限公司 Detect the protein chip and kit of protein Acetylation Level
CN108144062A (en) * 2017-12-15 2018-06-12 上海交通大学医学院 Regulate and control SENP1 phosphorylation modifications compound and SIRT3 SUMOization modified compound and its application
CN109738506A (en) * 2019-01-10 2019-05-10 青岛大学附属医院 Autophagy GAP-associated protein GAP acetylation sites research method in a kind of mature fat cell

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080241862A1 (en) * 2007-01-29 2008-10-02 Yingming Zhao Propionyl and butyryl lysine modifications in proteins

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003102196A1 (en) * 2002-05-30 2003-12-11 Centre National De La Recherche Scientifique (Cnrs) Vectors for expression of biotinylated proteins in mammalian cells, and their use for identification of protein-nucleic acid interactions in vivo
CN101787075A (en) * 2009-09-29 2010-07-28 天津医科大学附属肿瘤医院 Amino acid sequence with characteristic of bonding histone deacetylase 1 and expression vector thereof
CN103900893A (en) * 2013-05-20 2014-07-02 上海华盈生物医药科技有限公司 Kit for gathering protein modified by acetylization, as well as method and application of kit
CN107024588A (en) * 2016-02-01 2017-08-08 上海生物芯片有限公司 Detect the protein chip and kit of protein Acetylation Level
CN108144062A (en) * 2017-12-15 2018-06-12 上海交通大学医学院 Regulate and control SENP1 phosphorylation modifications compound and SIRT3 SUMOization modified compound and its application
CN109738506A (en) * 2019-01-10 2019-05-10 青岛大学附属医院 Autophagy GAP-associated protein GAP acetylation sites research method in a kind of mature fat cell

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
5-aza诱导干细胞经乙酰化特异RNAi后心肌细胞发育相关基因检测;朱静等;《第三军医大学学报》(第6期);第535-538页 *
ALS小鼠脊髓组织GFAP蛋白赖氨酸的乙酰化修饰;李俊强等;《山东大学学报(医学版)》;第51卷(第10期);第15-18页 *
川木瓜多糖的化学修饰及其活性研究;李容等;《右江民族医学院学报》;第40卷(第5期);第418-422页 *

Also Published As

Publication number Publication date
CN113155577A (en) 2021-07-23

Similar Documents

Publication Publication Date Title
JP4714584B2 (en) Quantitation method using isotope-labeled internal standard substance, analysis system for executing the quantification method, and program for the analysis
Tannu et al. Comparative proteomes of the proliferating C2C12 myoblasts and fully differentiated myotubes reveal the complexity of the skeletal muscle differentiation program
Baharvand et al. Concise review: trends in stem cell proteomics
He et al. Theoretical and experimental prospects for protein identification based solely on accurate mass measurement
Rubakhin et al. Profiling signaling peptides in single mammalian cells using mass spectrometry
CN111896651B (en) Agkistrodon halys venom thrombin-like enzyme characteristic polypeptide and application thereof
Garden et al. Formation of N‐pyroglutamyl peptides from N‐Glu and N‐Gln precursors in Aplysia neurons
CN113155577B (en) Preparation method for identifying specific protein acetylation modified sample
Kai et al. Direct mass spectrometric screening of antibiotics from bacterial surfaces using liquid extraction surface analysis
Doerge et al. Detection and confirmation of β-agonists in bovine retina using LC/APCI-MS
Tang et al. Indole-3-acetylaspartate and indole-3-acetylglutamate, the IAA-amide conjugates in the diploid strawberry achene, are hydrolyzed in growing seedlings
Dalluge Mass spectrometry for direct determination of proteins in cells: applications in biotechnology and microbiology
CN101581727B (en) Method for efficiently detecting interaction of in vivo proteins
Kjellström et al. In situ liquid− liquid extraction as a sample preparation method for matrix-assisted laser desorption/ionization ms analysis of polypeptide mixtures
An et al. A mass spectrometry‐based method to screen for α‐amidated peptides
JP2005512061A (en) Sphingolipid internal standard
Savaryn et al. Targeted analysis of recombinant NF kappa B (RelA/p65) by denaturing and native top down mass spectrometry
Sebastião et al. Proteomic and Glyco (proteo) mic tools in the profiling of cardiac progenitors and pluripotent stem cell derived cardiomyocytes: Accelerating translation into therapy
John et al. Complete sequencing and oxidative modification of manganese superoxide dismutase in medulloblastoma cells
Albright et al. Identifying gel‐separated proteins using in‐gel digestion, mass spectrometry, and database searching: Consider the chemistry
Cuomo et al. SILAC-based quantitative strategies for accurate histone posttranslational modification profiling across multiple biological samples
Xu et al. Investigating the cell cycle-associated dynamics of histone modifications using quantitative mass spectrometry
WO2024021228A1 (en) Sirt6 h133y protein, method for enriching myristoylation-modified peptide fragments using same, and use thereof
Colas et al. Mass spectrometry in plant proteomic analysis
CN113588856B (en) Method for high-throughput large-scale screening of histone modified binding protein

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