CN113376267A - Method and kit capable of rapidly detecting TMAO biomarker - Google Patents

Method and kit capable of rapidly detecting TMAO biomarker Download PDF

Info

Publication number
CN113376267A
CN113376267A CN202110508077.XA CN202110508077A CN113376267A CN 113376267 A CN113376267 A CN 113376267A CN 202110508077 A CN202110508077 A CN 202110508077A CN 113376267 A CN113376267 A CN 113376267A
Authority
CN
China
Prior art keywords
tmao
sample
standard
plasma sample
internal standard
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.)
Pending
Application number
CN202110508077.XA
Other languages
Chinese (zh)
Inventor
赵良才
董俊一
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.)
Lanli Biotechnology Suzhou Co ltd
Original Assignee
Lanli Biotechnology Suzhou Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lanli Biotechnology Suzhou Co ltd filed Critical Lanli Biotechnology Suzhou Co ltd
Priority to CN202110508077.XA priority Critical patent/CN113376267A/en
Publication of CN113376267A publication Critical patent/CN113376267A/en
Pending legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/62Detectors specially adapted therefor
    • G01N30/72Mass spectrometers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/04Preparation or injection of sample to be analysed
    • G01N2030/042Standards
    • G01N2030/045Standards internal

Landscapes

  • Physics & Mathematics (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)

Abstract

The invention belongs to the technical field of biological sample detection, and particularly relates to a method and a kit capable of quickly detecting a TMAO biomarker. The method is based on liquid chromatography-mass spectrometry, and relates to (1) setting liquid chromatography conditions; (2) setting mass spectrum conditions; (3) preparing a standard sample; (4) establishing a standard curve; (5) pretreating blood plasma to be detected; (6) pre-treating plasma sample, detecting and the like. The invention further provides a kit and a using method thereof, which are convenient to operate. According to the method for rapidly detecting the TMAO biomarker, provided by the invention, the sample is reasonably pretreated, and reasonable conditions of liquid chromatography and mass spectrum are optimized, so that a detection result with high accuracy and good stability is obtained; the kit provided by the invention provides the internal standard and the standard sample with accurate concentration, so that the actual operation steps are reduced, and the detection efficiency is improved.

Description

Method and kit capable of rapidly detecting TMAO biomarker
Technical Field
The invention belongs to the technical field of biological sample detection, and particularly relates to a method and a kit capable of quickly detecting a TMAO biomarker.
Background
Trimethylamine oxide (TMAO) is involved in many important biological functions in the human body, and after eating choline-containing foods such as beans and eggs, common people convert to trimethylamine in the large intestine, and the trimethylamine is further converted to trimethylamine oxide by trimethylamine oxidase in the liver. In recent years, much attention has been paid to the research on TMAO, and researchers have associated TMAO concentration with various chronic cardiovascular and cerebrovascular diseases and used TMAO as one of early warning indicators for chronic cardiovascular and cerebrovascular diseases. In addition, the team is working on researching and constructing the action relationship between TMAO and common diseases such as diabetic complications, bladder cancer, kidney cancer, prostate cancer, breast cancer and the like. Therefore, how to rapidly and accurately detect the concentration of TMAO in a sample is the basis for a series of studies in practice.
Disclosure of Invention
Aiming at the defects in the prior art, the invention provides a method and a kit capable of rapidly detecting a TMAO biomarker.
The method for rapidly detecting the TMAO biomarker comprises the following steps:
(S1) establishing a standard curve: taking a blank plasma sample, adding methanol, performing vortex oscillation, then loading into a centrifuge for centrifugal treatment, and taking supernatant and adding water for dilution; then adding TMAO and an internal standard to prepare a series of standard samples containing the internal standard with the same concentration and different TMAO concentrations; testing a series of standard samples by liquid chromatography-mass spectrometry, calculating to obtain a ratio (Y) of a TMAO peak area corresponding to each standard sample to an internal standard peak area and a ratio (X) of a TMAO concentration corresponding to each standard sample to an internal standard concentration, and making a change curve of Y along with X to obtain a standard curve;
(S2) pretreatment sample detection: detecting the pretreated plasma sample by liquid chromatography-mass spectrometry, calculating to obtain the ratio of the TMAO peak area to the internal standard peak area, substituting into a standard curve to obtain the ratio of the TMAO concentration in the pretreated sample to the internal standard concentration; calculating the concentration of TMAO in the original plasma sample according to the dilution times in the pretreatment process;
the plasma sample pretreatment method comprises the following steps: and adding a methanol solution containing an internal standard into a plasma sample, performing vortex oscillation, then loading into a centrifuge for centrifugal treatment, and adding water into a certain amount of supernatant for dilution to obtain a pretreated plasma sample.
Further, the plasma sample pretreatment method comprises the following steps: adding the plasma sample into methanol solution containing internal standard for protein precipitation, centrifuging to separate the sample, and diluting with water.
Furthermore, the plasma sample pretreatment method comprises the following steps: taking 35 mu L of plasma sample, adding 100 mu L of methanol solution containing the internal standard, performing vortex oscillation for 30-45s, then placing the plasma sample into a centrifuge for centrifugal treatment, wherein the separation factor RCF of the centrifuge is 3600-8000, and taking 50 mu L of supernatant, adding 200 mu L of deionized water for dilution to obtain the pretreated plasma sample.
In the pretreatment process of the plasma sample, the methanol solution of the internal standard is a methanol solution with d9-TMAO concentration of 30ng/mL (d 9-TMAO refers to a deuterated trimethylamine oxide isotope label).
Further, in the step (S1), 350 μ L of blank plasma sample is taken, 1000 μ L of methanol is added, vortex oscillation is carried out for 30-45S, and then the blank plasma sample is placed into a centrifuge for centrifugal treatment, wherein the separation factor RCF of the centrifuge is 3600-8000; adding 2000 mu L of deionized water into 500 mu L of supernatant for dilution, then adding TMAO, gradually diluting with water to obtain 10 solutions containing different concentrations of TMAO, wherein the TMAO concentrations are as follows: 10. 25, 55, 80, 100, 200, 400, 500, 800, 1000 ng/mL; mu.L of each solution was taken and 100. mu.L of a methanol solution containing 30ng/mL of d9-TMAO was added to each solution to obtain a series of standard samples.
Further, the liquid chromatography conditions were as follows:
a chromatographic column: an Agilent ZORBAX SB-C18 liquid chromatography column;
mobile phase: acetonitrile and 0.1% formic acid aqueous solution with the volume ratio of 88: 12;
flow rate: 0.3-0.5 mL.min-1
Column temperature: 35-40 ℃;
sample introduction amount: 5 mu L of the solution;
and (3) an elution mode: isocratic elution.
Further, the mass spectrometry conditions were as follows:
mass spectrometry: a triple quadrupole mass spectrometer;
scanning mode: electrospray positive ion mode;
an acquisition mode: mass spectrometry Multiple Reaction Monitoring (MRM);
ion source voltage: 4000V;
ion source temperature: 400 ℃;
air curtain air: 25 psi;
atomizing gas (gas 1): 50 psi;
auxiliary gas (gas 2): 40 psi.
The invention not only provides the method capable of rapidly detecting the TMAO biomarker, but also provides a kit which can be matched with the method for use, wherein the kit comprises the following reagents:
reagent A: d9-TMAO methanol solution with concentration of 30 ng/mL;
a series of reagents B: the standard sample used in claim 5.
The application of the kit is as follows: the kit is used for detecting the TMAO content in blood by liquid chromatography-mass spectrometry, the reagent A is used as an internal standard substance, and the series reagent B is used as a standard sample to obtain a standard curve under the determined chromatographic condition.
Has the advantages that: the method for rapidly detecting the TMAO biomarker is a high-efficiency detection method which is based on liquid chromatography-mass spectrometry and can carry out batch rapid detection, and a detection result with high accuracy and good stability is obtained by reasonably preprocessing a sample and matching and optimizing reasonable conditions of liquid chromatogram and mass spectrum; in addition, the invention also provides a kit which can be used by matching with the method, and provides an internal standard and a standard sample with accurate concentration so as to reduce actual operation steps and improve detection efficiency.
Detailed Description
The invention is further illustrated by the following specific examples, which are illustrative and intended to illustrate the problem and explain the invention, but not limiting.
Example 1
(1) Setting the conditions of liquid chromatography
A chromatographic column: an Agilent ZORBAX SB-C18 liquid chromatography column;
mobile phase: acetonitrile and 0.1% formic acid aqueous solution with the volume ratio of 88: 12;
flow rate: 0.4 mL.min-1
Column temperature: 35 ℃;
sample introduction amount: 5 mu L of the solution;
and (3) an elution mode: isocratic elution.
(2) Setting Mass Spectrometry conditions
Mass spectrometry: a triple quadrupole mass spectrometer;
scanning mode: electrospray positive ion mode;
an acquisition mode: mass spectrometry Multiple Reaction Monitoring (MRM);
ion source voltage: 4000V;
ion source temperature: 400 ℃;
air curtain air: 25 psi;
atomizing gas (gas 1): 50 psi;
auxiliary gas (gas 2): 40 psi.
(3) Preparing a standard sample
Taking 350 mu L of blank plasma sample, adding 1000 mu L of methanol, performing vortex oscillation for 45s, and then loading into a centrifuge for centrifugal treatment, wherein the separation factor RCF of the centrifuge is 8000; adding 2000 mu L of deionized water into 500 mu L of supernatant for dilution, and then adding TMAO to make the concentration of the TMAO 1000 ng/mL; gradually diluting with water to obtain 10 solutions containing TMAO with different concentrations, wherein the TMAO concentrations are respectively as follows: 10. 25, 55, 80, 100, 200, 400, 500, 800, 1000 ng/mL; a series of standards were prepared by adding 100. mu.L of each internal standard solution to 300. mu.L of each solution.
Internal standard solution: d9-TMAO was used as an internal standard to prepare a 30ng/mL methanol solution.
(4) Establishing a standard curve
And (3) testing a series of standard samples by liquid chromatography-mass spectrometry, calculating to obtain the ratio (Y) of the TMAO peak area corresponding to each standard sample to the internal standard peak area and the ratio (X) of the TMAO concentration corresponding to each standard sample to the internal standard concentration, and drawing a change curve of Y along with X to obtain the standard curve.
Linear regression equation: y = 1.256X-0.01192;
coefficient of correlation r2:0.9995;
Linear range of X: 1-100.
(5) Pretreatment of blood plasma to be tested
Taking 35 mu L of plasma sample, adding 100 mu L of methanol solution containing internal standard, carrying out vortex oscillation for 45s, then loading into a centrifuge for centrifugal treatment, wherein the separation factor RCF of the centrifuge is 8000, and taking 50 mu L of supernatant, adding 200 mu L of deionized water, and diluting to obtain the pretreated plasma sample.
(6) Pretreated plasma sample detection
Detecting the pretreated plasma sample by liquid chromatography-mass spectrometry, calculating to obtain the ratio of the TMAO peak area to the internal standard peak area, substituting into a standard curve to obtain the ratio of the TMAO concentration in the pretreated sample to the internal standard concentration; the concentration of TMAO in the original plasma sample was calculated from the dilution factor during the pretreatment.
Example 2
(1) Setting the conditions of liquid chromatography
A chromatographic column: an Agilent ZORBAX SB-C18 liquid chromatography column;
mobile phase: acetonitrile and 0.1% formic acid aqueous solution with the volume ratio of 88: 12;
flow rate: 0.3 mL.min-1
Column temperature: 40 ℃;
sample introduction amount: 5 mu L of the solution;
and (3) an elution mode: isocratic elution.
(2) Setting Mass Spectrometry conditions
Mass spectrometry: a triple quadrupole mass spectrometer;
scanning mode: electrospray positive ion mode;
an acquisition mode: mass spectrometry Multiple Reaction Monitoring (MRM);
ion source voltage: 4000V;
ion source temperature: 400 ℃;
air curtain air: 25 psi;
atomizing gas (gas 1): 50 psi;
auxiliary gas (gas 2): 40 psi.
(3) Preparing a standard sample
Taking 350 mu L of blank plasma sample, adding 1000 mu L of methanol, performing vortex oscillation for 30s, and then putting the blank plasma sample into a centrifuge for centrifugal treatment, wherein the separation factor RCF of the centrifuge is 3600; adding 2000 mu L of deionized water into 500 mu L of supernatant for dilution, and then adding TMAO to make the concentration of the TMAO 1000 ng/mL; gradually diluting with water to obtain 10 solutions containing TMAO with different concentrations, wherein the TMAO concentrations are respectively as follows: 10. 25, 55, 80, 100, 200, 400, 500, 800, 1000 ng/mL; a series of standards were prepared by adding 100. mu.L of each internal standard solution to 300. mu.L of each solution.
Internal standard solution: d9-TMAO was used as an internal standard to prepare a 30ng/mL methanol solution.
(4) Establishing a standard curve
And (3) testing a series of standard samples by liquid chromatography-mass spectrometry, calculating to obtain the ratio (Y) of the TMAO peak area corresponding to each standard sample to the internal standard peak area and the ratio (X) of the TMAO concentration corresponding to each standard sample to the internal standard concentration, and drawing a change curve of Y along with X to obtain the standard curve.
Linear regression equation: y = 1.249X-0.01204;
coefficient of correlation r2:0.9992;
Linear range of X: 1-100.
(5) Pretreatment of blood plasma to be tested
Taking 35 mu L of plasma sample, adding 100 mu L of methanol solution containing the internal standard, performing vortex oscillation for 30s, then putting the plasma sample into a centrifuge for centrifugation, wherein the separation factor RCF of the centrifuge is 3600, and taking 50 mu L of supernatant, adding 200 mu L of deionized water and diluting to obtain the pretreated plasma sample.
(6) Pretreated plasma sample detection
Detecting the pretreated plasma sample by liquid chromatography-mass spectrometry, calculating to obtain the ratio of the TMAO peak area to the internal standard peak area, substituting into a standard curve to obtain the ratio of the TMAO concentration in the pretreated sample to the internal standard concentration; the concentration of TMAO in the original plasma sample was calculated from the dilution factor during the pretreatment.
As can be seen from the standard curves established in examples 1 and 2, the method provided by the invention can establish a very linear standard curve, and the correlation coefficient r2The TMAO concentration range exceeds 0.999, is optimally 0.9995, and the linear range spans two complete orders of magnitude, so that the detection result with high reliability and accuracy can be obtained in a larger TMAO concentration range. In contrast, example 1 provides the optimum operating parameters.
The foregoing is only a preferred embodiment of the present invention, and it should be noted that modifications can be made by those skilled in the art without departing from the principle of the present invention, and these modifications should also be construed as the protection scope of the present invention.

Claims (9)

1. A method capable of rapidly detecting a TMAO biomarker, comprising the steps of:
(S1) establishing a standard curve: taking a blank plasma sample, adding methanol, performing vortex oscillation, then loading into a centrifuge for centrifugal treatment, and taking supernatant and adding water for dilution; then adding TMAO and an internal standard to prepare a series of standard samples containing the internal standard with the same concentration and different TMAO concentrations; testing a series of standard samples by liquid chromatography-mass spectrometry, calculating to obtain a ratio (Y) of a TMAO peak area corresponding to each standard sample to an internal standard peak area and a ratio (X) of a TMAO concentration corresponding to each standard sample to an internal standard concentration, and making a change curve of Y along with X to obtain a standard curve;
(S2) pretreatment sample detection: detecting the pretreated plasma sample by liquid chromatography-mass spectrometry, calculating to obtain the ratio of the TMAO peak area to the internal standard peak area, substituting into a standard curve to obtain the ratio of the TMAO concentration in the pretreated sample to the internal standard concentration; calculating the concentration of TMAO in the original plasma sample according to the dilution times in the pretreatment process;
the plasma sample pretreatment method comprises the following steps: and adding a methanol solution containing an internal standard into a plasma sample, performing vortex oscillation, then loading into a centrifuge for centrifugal treatment, and adding water into a certain amount of supernatant for dilution to obtain a pretreated plasma sample.
2. The method of claim 1, wherein the plasma sample pretreatment method comprises: adding the plasma sample into methanol solution containing internal standard for protein precipitation, centrifuging to separate the sample, and diluting with water.
3. The method of claim 1, wherein the plasma sample pretreatment method comprises: taking 35 mu L of plasma sample, adding 100 mu L of methanol solution containing the internal standard, performing vortex oscillation for 30-45s, then placing the plasma sample into a centrifuge for centrifugal treatment, wherein the separation factor RCF of the centrifuge is 3600-8000, and taking 50 mu L of supernatant, adding 200 mu L of deionized water for dilution to obtain the pretreated plasma sample.
4. The method of claim 3, wherein the internal standard methanol solution is a methanol solution with d9-TMAO concentration of 30ng/mL during the pretreatment of the plasma sample.
5. The method as claimed in claim 1, wherein in the step (S1), 350 μ L of blank plasma sample is taken, 1000 μ L of methanol is added, vortex oscillation is carried out for 30-45S, and then the blank plasma sample is put into a centrifuge for centrifugal processing, wherein the separation factor RCF of the centrifuge is 3600-8000; adding 2000 mu L of deionized water into 500 mu L of supernatant for dilution, then adding TMAO, gradually diluting with water to obtain 10 solutions containing different concentrations of TMAO, wherein the TMAO concentrations are as follows: 10. 25, 55, 80, 100, 200, 400, 500, 800, 1000 ng/mL; mu.L of each solution was taken and 100. mu.L of a methanol solution containing 30ng/mL of d9-TMAO was added to each solution to obtain a series of standard samples.
6. The method of claim 1, wherein the liquid chromatography conditions are as follows:
a chromatographic column: an Agilent ZORBAX SB-C18 liquid chromatography column;
mobile phase: acetonitrile and 0.1% formic acid aqueous solution with the volume ratio of 88: 12;
flow rate: 0.3-0.5 mL.min-1
Column temperature: 35-40 ℃;
sample introduction amount: 5 mu L of the solution;
and (3) an elution mode: isocratic elution.
7. The method of claim 1, wherein the mass spectrometry conditions are as follows:
mass spectrometry: a triple quadrupole mass spectrometer;
scanning mode: electrospray positive ion mode;
an acquisition mode: mass spectrometry Multiple Reaction Monitoring (MRM);
ion source voltage: 4000V;
ion source temperature: 400 ℃;
air curtain air: 25 psi;
atomizing gas (gas 1): 50 psi;
auxiliary gas (gas 2): 40 psi.
8. A kit comprising the following reagents:
reagent A: d9-TMAO methanol solution with concentration of 30 ng/mL;
a series of reagents B: the standard sample used in claim 5.
9. Use of the kit according to claim 8, wherein the kit is used in LC-MS for detecting TMAO content in blood, reagent A is used as an internal standard substance, and reagent B is used as a standard sample to obtain a standard curve under defined chromatographic conditions.
CN202110508077.XA 2021-05-11 2021-05-11 Method and kit capable of rapidly detecting TMAO biomarker Pending CN113376267A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110508077.XA CN113376267A (en) 2021-05-11 2021-05-11 Method and kit capable of rapidly detecting TMAO biomarker

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110508077.XA CN113376267A (en) 2021-05-11 2021-05-11 Method and kit capable of rapidly detecting TMAO biomarker

Publications (1)

Publication Number Publication Date
CN113376267A true CN113376267A (en) 2021-09-10

Family

ID=77572396

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110508077.XA Pending CN113376267A (en) 2021-05-11 2021-05-11 Method and kit capable of rapidly detecting TMAO biomarker

Country Status (1)

Country Link
CN (1) CN113376267A (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106556656A (en) * 2016-10-26 2017-04-05 南京医科大学 A kind of blood plasma metabolism small molecule mark related to cholelithiasis and its application
CN107340341A (en) * 2017-06-27 2017-11-10 长沙都正生物科技有限责任公司 A kind of TMAO immue quantitative detection reagent box and method
CN109142574A (en) * 2018-08-29 2019-01-04 广东药科大学 Improve the method for the material base of insulin resistance based on SVR research gegen qinlian decoction
CN110531008A (en) * 2019-06-25 2019-12-03 深圳市绿航星际太空科技研究院 A kind of method of trimethylamine oxide in detection urine
CN110687227A (en) * 2019-10-25 2020-01-14 上海阿趣生物科技有限公司 Detection method of TMAO and related metabolites thereof
CN111351885A (en) * 2018-12-20 2020-06-30 长沙都正生物科技有限责任公司 Biological negative sample for TMAO detection and preparation method and application thereof
US20210038550A1 (en) * 2018-02-01 2021-02-11 The Cleveland Clinic Foundation Disease detection and treatment based on trimethyl-lysine levels

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106556656A (en) * 2016-10-26 2017-04-05 南京医科大学 A kind of blood plasma metabolism small molecule mark related to cholelithiasis and its application
CN107340341A (en) * 2017-06-27 2017-11-10 长沙都正生物科技有限责任公司 A kind of TMAO immue quantitative detection reagent box and method
US20210038550A1 (en) * 2018-02-01 2021-02-11 The Cleveland Clinic Foundation Disease detection and treatment based on trimethyl-lysine levels
CN109142574A (en) * 2018-08-29 2019-01-04 广东药科大学 Improve the method for the material base of insulin resistance based on SVR research gegen qinlian decoction
CN111351885A (en) * 2018-12-20 2020-06-30 长沙都正生物科技有限责任公司 Biological negative sample for TMAO detection and preparation method and application thereof
CN110531008A (en) * 2019-06-25 2019-12-03 深圳市绿航星际太空科技研究院 A kind of method of trimethylamine oxide in detection urine
CN110687227A (en) * 2019-10-25 2020-01-14 上海阿趣生物科技有限公司 Detection method of TMAO and related metabolites thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
刘燕等: "血清中氧化三甲胺的高效液相色谱串联质谱法测定研究", 《华南预防医学》 *
李天翔等: "肠道菌群及代谢与载脂蛋白E在动脉粥样硬化中的相互作用", 《中国微生态学杂志》 *
陈云明等: "脐疗法治疗腹泻大鼠的血液和粪便代谢组学研究", 《中华中医药杂志》 *

Similar Documents

Publication Publication Date Title
EP4006540A1 (en) Method for simultaneously testing phylloquinone and menaquinone-4 in trace blood
CN114720704B (en) Kit and method for measuring free testosterone in serum
CN110702831B (en) Kit for detecting serum testosterone hormone by ultra-high performance liquid chromatography-tandem mass spectrometry
CN113391001B (en) Detection method of glucosinolate compounds
CN111239267B (en) Method for detecting short-chain fatty acids in serum and lymph tissue based on GC-MS
CN114994214B (en) Method for qualitatively detecting neutral oligosaccharide in breast milk
CN113049719A (en) Method and kit for detecting free testosterone
CN110988193A (en) Method for detecting advanced glycosylation end products in aquatic products
CN109613144B (en) Detection method of catecholamine hormone
CN112014509A (en) Method for synchronously determining angiotensin I and aldosterone in sample
CN108593790B (en) Method for simultaneously detecting 24,25(OH)2D and 25OHD of serum
CN113341027A (en) Method and kit for detecting testosterone in saliva by high performance liquid chromatography tandem mass spectrometry
CN106370741B (en) A kind of measurement screening technique of the laryngocarcinoma serum difference metabolin based on hydrophilic Interaction Chromatography flight time mass spectrum
CN113376267A (en) Method and kit capable of rapidly detecting TMAO biomarker
CN116183801A (en) Liquid chromatography-mass spectrometry method, kit and system for detecting insulin and C peptide
CN110702829A (en) Method for measuring aldosterone content in blood plasma or blood serum
CN115932118A (en) Method for improving catecholamine mass spectrum quantitative detection sensitivity by using vitamin C
CN115508483A (en) LC-MS/MS method for rapidly detecting methylmalonic acid in serum sample
CN111579703A (en) LC-MS/MS-based method for detecting and analyzing degradation products of bilirubin in biological sample
CN114280192A (en) Method for detecting vitamin D and metabolite thereof
CN113376280A (en) Method for simultaneously detecting 94 amino acids in urine sample
CN112485340A (en) Method for detecting 1, 5-sorbitan in plasma by ultra-high performance liquid chromatography tandem mass spectrometry
CN106324173A (en) Method for determining and screening differential metabolites of laryngeal cancer serum based on inverted phase chromatography time-of-flying mass spectrum
CN114563504B (en) Method and kit for determining content of free aldosterone in blood plasma
CN112964814A (en) Method for detecting total homocysteine in biological body fluid

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
RJ01 Rejection of invention patent application after publication

Application publication date: 20210910

RJ01 Rejection of invention patent application after publication