CN111351881A - Method for detecting arachidonic acid and metabolite thereof in human plasma - Google Patents

Method for detecting arachidonic acid and metabolite thereof in human plasma Download PDF

Info

Publication number
CN111351881A
CN111351881A CN202010252579.6A CN202010252579A CN111351881A CN 111351881 A CN111351881 A CN 111351881A CN 202010252579 A CN202010252579 A CN 202010252579A CN 111351881 A CN111351881 A CN 111351881A
Authority
CN
China
Prior art keywords
arachidonic acid
acid
methanol
liquid
phase
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
CN202010252579.6A
Other languages
Chinese (zh)
Inventor
骆震
杨超
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.)
Shanghai Applied Protein Technology Co Ltd
Original Assignee
Shanghai Applied Protein Technology Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Applied Protein Technology Co Ltd filed Critical Shanghai Applied Protein Technology Co Ltd
Priority to CN202010252579.6A priority Critical patent/CN111351881A/en
Publication of CN111351881A publication Critical patent/CN111351881A/en
Pending legal-status Critical Current

Links

Images

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
    • G01N30/04Preparation or injection of sample to be analysed
    • G01N30/06Preparation
    • 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
    • G01N30/06Preparation
    • G01N30/14Preparation by elimination of some components
    • 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
    • G01N30/26Conditioning of the fluid carrier; Flow patterns
    • G01N30/28Control of physical parameters of the fluid carrier
    • G01N30/34Control of physical parameters of the fluid carrier of fluid composition, e.g. gradient
    • 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
    • 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
    • G01N30/06Preparation
    • G01N2030/062Preparation extracting sample from raw material

Abstract

The invention aims to provide a method for detecting arachidonic acid in human plasma and metabolites thereof, which comprises the steps of adding isotope arachidonic acid into a biological sample as an internal standard, extracting arachidonic acid in a sample mixture by a liquid-liquid extraction method, separating the arachidonic acid in the sample by using a high-efficiency liquid phase, and performing tandem mass spectrometry by using an internal standard curve method, and has the advantages of good selectivity, high sensitivity, short analysis time, low mass spectrum signal-to-noise ratio and the like.

Description

Method for detecting arachidonic acid and metabolite thereof in human plasma
Technical Field
The invention relates to the technical field of biological detection, in particular to a method for accurately and efficiently extracting and detecting pentadecanoic acid and metabolites thereof in human plasma.
Background
Arachidonic acid is an important substance for the development of human brain and optic nerve, and has important effects on improving intelligence, improving memory and enhancing visual acuity. Arachidonic acid also has a series of physiological activities of reducing cholesterol, increasing blood vessel elasticity, inhibiting platelet aggregation, reducing blood viscosity, regulating blood cell function and the like. Arachidonic acid has 3 metabolic pathways: (1) cyclooxygenase COX; (2) lipoxygenase LOX; (3) cytochrome P450 enzymes (CYP) are direct precursors for synthesizing icosic derivatives such as prostaglandins (prostaglandins), thromboxanes (thromboxanes) and leukotrienes (leukotrines), and these bioactive substances are involved in the regulation of inflammatory reaction and immune system, regulating vascular tension and platelet aggregation, regulating the growth of tumor cells, influencing the synthesis and release of hormones in endocrine system, and playing an important role in the development of central nervous system and the development of retinal nerve, so that the cytochrome P450 enzymes are related to various diseases such as atherosclerosis, hypertension, allergy, arthritis, and the like.
However, the existing pretreatment preparation method for the substances has the problems of high cost, long extraction time, low sample yield, difficulty in detection and the like. Therefore, how to develop a method which is low in cost, quick and capable of improving the yield of a target product, improving the detection limit of a mass spectrum and reducing detection noise is a key technical problem to be solved in the prior art.
Disclosure of Invention
The invention aims to provide a method for detecting arachidonic acid in human plasma and metabolites thereof, which comprises the steps of adding isotope arachidonic acid into a biological sample as an internal standard, extracting arachidonic acid in a sample mixture by a liquid-liquid extraction method, separating the arachidonic acid in the sample by using a high-efficiency liquid phase, and performing tandem mass spectrometry by using an internal standard curve method, and has the advantages of good selectivity, high sensitivity, short analysis time, low mass spectrum signal-to-noise ratio and the like.
In order to achieve the purpose, the technical scheme adopted by the invention is as follows:
a method for detecting arachidonic acid and metabolites thereof in human plasma comprises the following steps:
step S1, dissolving an isotope internal standard product arachidonic acid by using methanol, preparing a mixed internal standard working solution, dissolving the standard product arachidonic acid by using a methanol aqueous solution, and preparing a standard product mixed solution;
step S2, using ethanol to dilute the standard substance mixed liquor obtained in the step S1 into a series of concentration standard substance mixed liquor with a plurality of concentration gradients in a gradient concentration manner, wherein the concentration gradients are more than three; respectively adding the mixed internal standard working solution in the step S1 and methanol containing 1% of FA into the mixed solution of the standard substance with each gradient concentration, carrying out ice bath ultrasonic treatment, centrifuging at low temperature, and then taking the supernatant to carry out liquid-liquid extraction to obtain an eluted filtrate after extraction for later use;
step S3, taking a sample to be detected, adding mixed internal standard working solution and methanol containing 1% of FA into the sample to be detected by the same operation method of step S2, carrying out ice bath ultrasonic treatment, centrifuging at low temperature, taking supernatant, and carrying out liquid-liquid extraction to obtain extracted filtrate for later use;
step S4, carrying out high performance liquid chromatography-tandem mass spectrometry on the extracted samples obtained in the steps S2 and S3, wherein the high performance liquid chromatography conditions comprise sample introduction at 4 ℃, column temperature of 35 ℃, flow rate of 400 mu L/min and sample introduction amount of 2 mu L; the liquid phase gradient is as follows, 0-1min, the B phase is maintained at 30%; 1-7min, the phase B changes linearly from 30% to 80%; 7-9min, phase B changes linearly from 80% to 90%; 9-11min, keeping phase B at 90%; 11-11.2min, phase B changes linearly from 90% to 30%; 11.2-14min, keeping phase B at 30%;
the mass spectrum detection is in a multi-reaction monitoring mode, and the ion source condition is that the ion source temperature (sourcetemperature) is 500 ℃; an atomization Gas pressure (ion Source Gas1) of 50; assist Gas pressure (Ion Source Gas 2): 50; air Curtain gas (curtaingas): 30, of a nitrogen-containing gas; spray Voltage (ionsafe Voltage flowing): 4500V.
According to the above protocol, the isotopic internal standard Arachidonic acid in step S comprises Thromboxane B-D (Thromboxane B-D), Leukotriene B-D (Leukotriene B-D), Leukotriene D-D (Leukotriene D-D), Prostaglandin F-4 (Prostagladin F-4), 8-isoprostane F0-D (8-iso-prostagladin F1-D), 6-keto-prostagladin F-D (6-keto-prostagladin F-D), 12 hydroxyeicosatetraenoic acid-D (12S-HETE-D), Arachidonic acid-D (Arachidonic acid-D), Prostaglandin D-D (Prostagladin D-D), (9S,10E,12Z) -9-hydroxyoctadecane-10, 12-dienoic acid-D (9S-DE-D), (9Z,11E) -13-hydroxy-9, 11-octadecane-D, 9S-12-hydroxyeicosatetraenoic acid-D (9S-D), Arachidonic acid-D (2E-12E, 12E-hydroxy octadecane-10, 12-10-octadecane-10, 12-dienoic acid-D (9S-D, 10E, 12-13-oxadocosatetraenoic acid) and 15-15% of each of a mixture of a working mixture solution, a mixture of Thromboxane-15-oxadocosatetraenoic acid (15-7-15-oxadiene-7-15 and 15-oxadiene-2-7-15-2-oxadiene-2-oxatrine mixture of a working mixture of a Thromboxane-oxadiene-oxatriene-2-7-2-oxatriene-7-15-one of a working mixture of a Thromboxane, a working mixture of a Thromboxane-oxatriene-2-oxatriene-2-one of a working mixture of a thrombodode-2-oxatriene-2-oxatriene-oxatrie.
According to the above scheme, the methanol containing 1% of FA in the steps S2 and S3 further contains an antioxidant; the ice bath ultrasonic treatment is 500W ultrasonic treatment in an ice bath for 30 min; the low-temperature centrifugation is performed for 10min at 4 ℃ at 14000 g.
According to the scheme, the antioxidant is 2, 6-di-tert-butyl-p-cresol, and the preparation method comprises the steps of weighing 2, 6-di-tert-butyl-p-cresol, adding methanol according to the proportion of 2.32mg/mL for dissolving, adding formic acid with the volume of 1/100 methanol, and uniformly mixing.
According to the above scheme, the liquid-liquid extraction in steps S2 and S3 is specifically implemented by using Oasis HLB solid phase extraction 96-well Plate (96-well Plate), adding 3mL of methanol activated well Plate in 3 times, each time adding 1 mL; adding 3mL of pure water balance pore plate for 3 times, wherein 1mL of pure water balance pore plate is added each time; adding the supernatant prepared in the step S2 or S3 into a pore plate; adding 2mL of cleaning solution A (washing solution A) in 2 times, and adding 1mL each time; adding 2mL of cleaning solution B (washing solution A) in 2 times, and adding 1mL of cleaning solution B each time; and finally, adding 1mL of methanol to elute the sample to obtain eluted filtrate.
According to the scheme, the cleaning solution A (washing solution A) is prepared by mixing the following components in a volume ratio of 19: 1-water: the antioxidant (2, 6-ditertbutyl-p-cresol solution) is mixed according to the proportion; the cleaning solution B (washing solution B) is prepared from the following components in a volume ratio of 10000: 1-cleaning liquid a: mixing with formic acid.
The invention has the beneficial effects that:
1) the sample is pretreated by liquid-liquid extraction, and the pretreatment of the sample is simple and quick.
2) The method has the advantages that the pretreatment step of the sample can be completed only by simple liquid-liquid extraction on the body fluid sample, 15 arachidonic acid can be effectively separated by using a Waters ACQUITY UPLC I-Class system, the quantification is carried out by 13 kinds of site internal standards, the important arachidonic acid and metabolites thereof in human plasma can be accurately detected, the method has higher precision and accuracy, can be used for quantitative analysis of various clinical samples, is simple in experimental operation and short in experimental period, provides a reliable detection method for the clinical health assessment of the arachidonic acid level, and provides an effective research means for scientific research.
Drawings
FIG. 1 is a diagram of a standard XIC of arachidonic acid and its metabolites in examples.
Fig. 2 is a graph of the DHA standard curve results in the examples.
The two drawings are display graphs of the result of the instrument analysis, only the result of the current experiment is displayed, the change can occur according to the difference of each actual experiment, and the characters in the drawings do not influence the repeated implementation of the patent scheme.
Detailed Description
The technical solution of the present invention is described below with reference to the accompanying drawings and examples.
A method for detecting arachidonic acid and metabolites thereof in human plasma comprises the following steps:
step S1, preparing a mixed internal standard working solution, namely dissolving an isotope internal standard product arachidonic acid comprising thromboxane B2-D4, leukotriene B4-D4, leukotriene D4-D5, prostaglandin F2 α -4, 8-isoprostaglandin F2 α -D4, 6-ketone-prostaglandin F1 α -D4, 12-hydroxyeicosatetraenoic acid-D8, arachidonic acid-D8, prostaglandin D2-D4, (9S,10E,12Z) -9-hydroxyoctadeca-10, 12-dienoic acid-D4, (9Z,11E) -13-hydroxy-9, 11-octadecadienoic acid-D4, 15-hydroxyeicosatetraenoic acid-D8 and docosahexaenoic acid-D5 by using methanol, and diluting the mixed internal standard working solution with water to obtain mixed solution of which the final concentration of each component in the mixed solution is 200 ng/ml;
preparing a standard substance mixed solution, namely dissolving various standard substances of arachidonic acid including thromboxane B2, leukotriene B4, leukotriene D4, prostaglandin E2, 8-isoprostane F2 α, prostaglandin F2 α, 6-keto-prostaglandin F1 α, prostaglandin D2, (9S,10E,12Z) -9-hydroxyoctadeca-10, 12-dienoic acid, (9Z,11E) -13-hydroxy-9, 11-octadecadienoic acid, 14, 15-epoxyeicosatetraenoic acid, 12-hydroxyeicosatetraenoic acid, 15-hydroxyeicosatetraenoic acid, arachidonic acid and docosahexaenoic acid by 50 percent methanol aqueous solution, and mixing the components according to the component proportion of the following table 1 to prepare the standard substance mixed solution of which the final concentration of each component in the mixed solution is 1000 ng/ml;
table 1 arachidonic acid 15 standard mixed solutions were prepared with ingredient concentrations.
Figure BDA0002436024820000041
Step S2, the standard substance mixture obtained in step S1 is diluted with ethanol in a gradient concentration to a series of concentration standard substance mixtures of 10 concentration gradients, as shown in table 2 below.
TABLE 2 gradient dilution Table for arachidonic acid standard solution
Figure BDA0002436024820000051
Respectively adding 20 mu L of mixed internal standard working solution obtained in the step S1 and 500 mu L of methanol containing 1% of FA into each gradient concentration standard product mixed solution, carrying out 500W ultrasonic treatment in an ice bath for 30min, centrifuging for 10min at 14000g and 4 ℃, taking the supernatant, and carrying out liquid-liquid extraction, wherein the specific liquid-liquid extraction step is to use an Oasis HLB solid phase extraction 96-well plate, add 3mL of methanol activated well plates for 3 times, and add 1mL of methanol each time; adding 3mL of pure water balance pore plate for 3 times, wherein 1mL of pure water balance pore plate is added each time; adding the supernatant prepared in the step S2 or S3 into a pore plate; adding 2mL of cleaning solution A for 2 times, and adding 1mL of cleaning solution A for each time; adding 2mL of cleaning solution B for 2 times, and adding 1mL of cleaning solution B for each time; finally, adding 1mL of methanol to elute the sample to obtain eluted filtrate, and drying the eluted filtrate by using nitrogen for later use;
step S3, unfreezing the plasma sample at 4 ℃, taking 200 microliter of sample, adding 20 microliter of mixed internal standard working solution in the step S1 and 500 microliter of methanol containing 1% of FA, carrying out ultrasonic treatment for 30min at 500W in an ice bath, centrifuging for 10min at 4 ℃ by 14000g, taking supernatant, and carrying out liquid-liquid extraction, wherein the specific liquid-liquid extraction step is to use an Oasis HLB solid phase extraction 96 pore plate, add 3mL methanol activation pore plates for 3 times, and add 1mL each time; adding 3mL of pure water balance pore plate for 3 times, wherein 1mL of pure water balance pore plate is added each time; adding the supernatant prepared in the step S2 or S3 into a pore plate; adding 2mL of cleaning solution A for 2 times, and adding 1mL of cleaning solution A for each time; adding 2mL of cleaning solution B for 2 times, and adding 1mL of cleaning solution B for each time; finally, adding 1mL of methanol to elute the sample to obtain eluted filtrate, and drying the eluted filtrate by using nitrogen for later use;
step S4, samples obtained in step S2 and step S3 were mixed with 100ul acetonitrile: water: re-dissolving formic acid solution (80: 19: 1, v/v/v), performing high performance liquid chromatography-tandem mass spectrometry, loading sample amount of 4ul, performing chromatographic separation by adopting a Waters acquisition UPLC I-Class system, introducing sample at 4 ℃, column temperature of 35 ℃, flow rate of 400 muL/min, and sample amount of 2 muL; the liquid phase gradient is as follows, 0-1min, the B phase is maintained at 30%; 1-7min, the phase B changes linearly from 30% to 80%; 7-9min, phase B changes linearly from 80% to 90%; 9-11min, keeping phase B at 90%; 11-11.2min, phase B changes linearly from 90% to 30%; 11.2-14min, keeping phase B at 30%;
performing mass spectrometry by using 5500QTRAP mass Spectrometer (SCIEX) in negative ion mode, and using multiple reaction monitoring mode under the condition that the temperature of the ion source is 500 ℃; the atomization air pressure is 50; auxiliary air pressure: 50; air curtain air: 30, of a nitrogen-containing gas; spraying voltage: 4500V.
Each ion pair and its corresponding declustering voltage, collision voltage and collision cell exit voltage parameters are as in table 3 below.
Table 3 mass spectrometry ion pair declustering voltage, collision voltage and collision cell exit voltage parameter tables.
Figure BDA0002436024820000061
Figure BDA0002436024820000071
As a preferable example, the methanol containing 1% FA in steps S2 and S3 further contains an antioxidant solution of 2, 6-di-tert-butyl-p-cresol, and the preparation method is as follows, weighing 11.6mg of 2, 6-di-tert-butyl-p-cresol, adding 5mL of methanol to dissolve, adding 50. mu.L of formic acid, and mixing well.
As a preferred embodiment, the cleaning solution A is prepared by measuring 38mL of pure water, adding 2mL of 2, 6-di-tert-butyl-p-cresol and mixing; the cleaning solution B is prepared by measuring 20mL of the cleaning solution A and adding 200 mu L of formic acid for mixing.
The detection result of this embodiment is further analyzed to verify the feasibility of the method provided by the present invention:
and establishing a standard curve by adopting an isotope internal standard quantitative method and taking the concentration of the arachidonic acid standard as an x axis and the peak area ratio of the arachidonic acid standard to the internal standard as a y axis. The concentration of arachidonic acid in plasma was calculated from this curve and the results were as follows:
Figure BDA0002436024820000072
15 the arachidonic acid has good linear relation in respective concentration linear range, meets the quantitative requirement, and can rapidly, conveniently, stably and accurately quantitatively detect the arachidonic acid in the blood plasma.
The above embodiments are only used for illustrating but not limiting the technical solutions of the present invention, and although the above embodiments describe the present invention in detail, those skilled in the art should understand that: modifications and equivalents may be made thereto without departing from the spirit and scope of the invention and any modifications and equivalents may fall within the scope of the claims.

Claims (6)

1. A method for detecting arachidonic acid and metabolites thereof in human plasma is characterized by comprising the following steps:
step S1, dissolving an isotope internal standard product arachidonic acid by using methanol, preparing a mixed internal standard working solution, dissolving the standard product arachidonic acid by using a methanol aqueous solution, and preparing a standard product mixed solution;
step S2, using ethanol to dilute the standard substance mixed liquor obtained in the step S1 into a series of concentration standard substance mixed liquor with a plurality of concentration gradients in a gradient concentration manner, wherein the concentration gradients are more than three; respectively adding the mixed internal standard working solution in the step S1 and methanol containing 1% of FA into the mixed solution of the standard substance with each gradient concentration, carrying out ice bath ultrasonic treatment, centrifuging at low temperature, and then taking the supernatant to carry out liquid-liquid extraction to obtain an eluted filtrate after extraction for later use;
step S3, taking a sample to be detected, adding mixed internal standard working solution and methanol containing 1% of FA into the sample to be detected by the same operation method of step S2, carrying out ice bath ultrasonic treatment, centrifuging at low temperature, taking supernatant, and carrying out liquid-liquid extraction to obtain extracted filtrate for later use;
step S4, carrying out high performance liquid chromatography-tandem mass spectrometry on the extracted samples obtained in the steps S2 and S3, wherein the high performance liquid chromatography conditions comprise sample introduction at 4 ℃, column temperature of 35 ℃, flow rate of 400 mu L/min and sample introduction amount of 2 mu L; the liquid phase gradient is as follows, 0-1min, the B phase is maintained at 30%; 1-7min, the phase B changes linearly from 30% to 80%; 7-9min, phase B changes linearly from 80% to 90%; 9-11min, keeping phase B at 90%; 11-11.2min, phase B changes linearly from 90% to 30%; 11.2-14min, keeping phase B at 30%;
the mass spectrum detection is in a multi-reaction monitoring mode, and the ion source condition is that the temperature of the ion source is 500 ℃; the atomization air pressure is 50; auxiliary air pressure: 50; air curtain air: 30, of a nitrogen-containing gas; spraying voltage: 4500V.
2. A method as claimed in claim 1, wherein the step S1 comprises mixing arachidonic acid as an isotope, including thromboxane B2-D4, leukotriene B4-D4, leukotriene D4-D5, prostaglandin F2 α -4, 8-isoprostane F2 α -D4, 6-keto-prostaglandin F1 α -D4, 12 hydroxyeicosatetraenoic acid-D8, arachidonic acid-D8, prostaglandin D2-D4, (9S,10E,12Z) -9-hydroxyoctadeca-10, 12-dienoic acid-D4, (9Z,11E) -13-hydroxy-9, 11-octadecadienoic acid-D4, 15 hydroxyeicosatetraenoic acid-D4 and docosahexenoic acid-D4, dissolving the respective components with methanol, diluting the respective components with water to a final concentration of the respective components in 200ng/ml, arachidonic acid-D4 and docosahexaenoic acid-D4, dissolving the respective concentrations of arachidonic acid, and its metabolites in water as a final concentration, including arachidonic acid B2-D, arachidonic acid, and docosahexaenoic acid-D4, and docosahexaenoic acid solutions as a final concentration, and docosahexaenoic acid solutions as solutions, as solutions of step S465, and as solutions of step S466-5, 6-5-6-5, 6-13-5-13, 13-5, 13-6-5.
3. The method of claim 1, wherein the methanol containing 1% FA in steps S2 and S3 further contains an antioxidant; the ice bath ultrasonic treatment is 500W ultrasonic treatment in an ice bath for 30 min; the low-temperature centrifugation is performed for 10min at 4 ℃ at 14000 g.
4. The method for detecting arachidonic acid and its metabolites in human plasma as claimed in claim 3, wherein the antioxidant is 2, 6-di-tert-butyl-p-cresol, which is prepared by weighing 2, 6-di-tert-butyl-p-cresol, dissolving in methanol at a ratio of 2.32mg/mL, adding 1/100 volumes of formic acid, and mixing well.
5. The method of claim 4, wherein the liquid-liquid extraction in steps S2 and S3 is performed by 3 times adding 1mL of 3mL methanol-activated well plates using Oasis HLB solid-phase extraction 96-well plates; adding 3mL of pure water balance pore plate for 3 times, wherein 1mL of pure water balance pore plate is added each time; adding the supernatant prepared in the step S2 or S3 into a pore plate; adding 2mL of cleaning solution A for 2 times, and adding 1mL of cleaning solution A for each time; adding 2mL of cleaning solution B for 2 times, and adding 1mL of cleaning solution B for each time; and finally, adding 1mL of methanol to elute the sample to obtain eluted filtrate.
6. The method for detecting arachidonic acid and its metabolites in human plasma according to claim 5, wherein the washing solution A is prepared by mixing the following raw materials in a volume ratio of 19: 1-water: the antioxidant is mixed in proportion; the cleaning solution B is prepared from the following components in percentage by volume of 10000: 1-cleaning liquid a: mixing with formic acid.
CN202010252579.6A 2020-04-01 2020-04-01 Method for detecting arachidonic acid and metabolite thereof in human plasma Pending CN111351881A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010252579.6A CN111351881A (en) 2020-04-01 2020-04-01 Method for detecting arachidonic acid and metabolite thereof in human plasma

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010252579.6A CN111351881A (en) 2020-04-01 2020-04-01 Method for detecting arachidonic acid and metabolite thereof in human plasma

Publications (1)

Publication Number Publication Date
CN111351881A true CN111351881A (en) 2020-06-30

Family

ID=71196318

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010252579.6A Pending CN111351881A (en) 2020-04-01 2020-04-01 Method for detecting arachidonic acid and metabolite thereof in human plasma

Country Status (1)

Country Link
CN (1) CN111351881A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113049802A (en) * 2021-03-23 2021-06-29 广州医科大学附属第一医院(广州呼吸中心) Serological marker for evaluating allergen-specific immunotherapy effect
CN115876935A (en) * 2022-12-29 2023-03-31 大连博源医学科技有限公司 Method for detecting 11-dehydrothromboxane B in blood 2 Liquid chromatography-tandem mass spectrometry method and kit
CN115980223A (en) * 2022-12-29 2023-04-18 大连博源医学科技有限公司 Method for detecting 11-dehydrothromboxane B in blood 2 Liquid chromatography-tandem mass spectrometry method and kit

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104316609A (en) * 2014-10-13 2015-01-28 上海市第一人民医院宝山分院 Application of arachidonic acid metabolite in preparing preeclampsia detection kit
CN109212087A (en) * 2018-10-22 2019-01-15 嘉兴迈维代谢生物科技有限公司 A kind of quasi-arachidonic acid substance detection method
CN111337609A (en) * 2020-03-16 2020-06-26 上海中科新生命生物科技有限公司 Human serum determination method for simultaneously determining multiple neurotransmitters based on LC-MC

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104316609A (en) * 2014-10-13 2015-01-28 上海市第一人民医院宝山分院 Application of arachidonic acid metabolite in preparing preeclampsia detection kit
CN109212087A (en) * 2018-10-22 2019-01-15 嘉兴迈维代谢生物科技有限公司 A kind of quasi-arachidonic acid substance detection method
CN111337609A (en) * 2020-03-16 2020-06-26 上海中科新生命生物科技有限公司 Human serum determination method for simultaneously determining multiple neurotransmitters based on LC-MC

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
AYUMI ANDO等: "A Simple and Highly Sensitive Quantitation of Eicosanoids in Biologica Samples Using Nano-flow Liquid Chromatography/Mass Spectrometry", 《ANALYTICAL SCIENCES》 *
GABRIEL DASILVA等: "Lipidomic analysis of polyunsaturated fatty acids and their oxygenated metabolites in plasma by solid-phase extraction followed by LC-MS", 《ANAL BIOANAL CHEM》 *
MICHIEL G.J. BALVERS等: "Time-dependent effect of in vivo inflammation on eicosanoid and endocannabinoid levels in plasma, liver, ileum and adipose tissue in C57BL/6 mice fed a fish-oil diet", 《INTERNATIONAL IMMUNOPHARMACOLOGY》 *
XU ZHANG等: "Systematic Metabolomic Analysis of Eicosanoids after Omega ‑ 3 Polyunsaturated Fatty Acid Supplementation by a Highly Specific Liquid Chromatography − Tandem Mass Spectrometry-Based Method", 《J. PROTEOME RES.》 *
张晓冬等: "依布硒及其类似物对大鼠血小板和腹腔白细胞脂氧合酶产物生成的影响", 《中国药理学与毒理学杂志》 *
谷艳等: "高效液相色谱-电喷雾串联四级杆质谱法检测小鼠组织中花生四烯酸及其代谢产物", 《分析化学》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113049802A (en) * 2021-03-23 2021-06-29 广州医科大学附属第一医院(广州呼吸中心) Serological marker for evaluating allergen-specific immunotherapy effect
CN115876935A (en) * 2022-12-29 2023-03-31 大连博源医学科技有限公司 Method for detecting 11-dehydrothromboxane B in blood 2 Liquid chromatography-tandem mass spectrometry method and kit
CN115980223A (en) * 2022-12-29 2023-04-18 大连博源医学科技有限公司 Method for detecting 11-dehydrothromboxane B in blood 2 Liquid chromatography-tandem mass spectrometry method and kit
CN115876935B (en) * 2022-12-29 2023-11-14 大连博源医学科技有限公司 A method for detecting 11-dehydrothromboxane B in blood 2 Liquid chromatography-tandem mass spectrometry method and kit

Similar Documents

Publication Publication Date Title
CN111351881A (en) Method for detecting arachidonic acid and metabolite thereof in human plasma
You et al. Air-assisted liquid–liquid microextraction used for the rapid determination of organophosphorus pesticides in juice samples
CN105158394A (en) Method for detecting multiple lipid-soluble vitamins in blood sample simultaneously
Jiang et al. Development of an analytical method for separation of phenolic acids by ultra-performance convergence chromatography (UPC2) using a column packed with a sub-2-μm particle
CN106855545A (en) Liposoluble vitamin simultaneously in detection feed and the method for water soluble vitamin
CN110631875B (en) Method for selectively adsorbing and eluting phospholipid and glycosphingolipid step by step
CN105784894B (en) Pesticide residue detection method for traditional Chinese medicine
Goh et al. Automation of ionic liquid enhanced membrane bag-assisted-liquid-phase microextraction with liquid chromatography-tandem mass spectrometry for determination of glucocorticoids in water
CN112684089A (en) Method for separating and determining carnitine enantiomer in health food based on ultra-high performance synthetic phase chromatography technology
Cao et al. Analysis of volatile compounds in Curcuma wenyujin YH Chen et C. Ling by headspace solvent microextraction–gas chromatography–mass spectrometry
CN114660214A (en) Liquid chromatography detection method of semaglutide and application thereof
CN108362795A (en) Content of homocysteine rapid detection method in dried blood spot
Shahdousti et al. Determination of valproic acid in human serum and pharmaceutical preparations by headspace liquid-phase microextraction gas chromatography-flame ionization detection without prior derivatization
Chen et al. Solid phase extraction based microfluidic chip coupled with mass spectrometry for rapid determination of aflatoxins in peanut oil
CN107688072B (en) Detection method of Xingnaojing injection
CN114184716B (en) High performance liquid chromatography analysis method for determining related substance components in halominosone cream
Zhong et al. Multi-channel purge and trap system coupled with ion chromatography for the determination of alkylamines in cosmetics
CN114034799A (en) Method for detecting fluxapyroxad residual quantity in grapes
CN107976506B (en) Detection method of obeticholic acid related substances
Zhang et al. Combination of pressurized liquid extraction with countercurrent chromatography for systematical isolation of ginsenosides from Panax ginseng
CN110412154B (en) Quantitative determination method for multi-component liquid quality of Qilong capsule and application thereof
CN105651870B (en) The remaining method of pesticide in a kind of gas chromatography-mass spectrum detection liquid beverage
CN111537642A (en) Method for determining alpha-vitamin E in beverage by utilizing solid phase extraction-liquid chromatography
CN110231425A (en) The extracting method and detection method of Co-Q10 in blood
CN113466391B (en) Method for simultaneously determining lactobionic acid and sodium gluconate contents in fermentation liquor

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
RJ01 Rejection of invention patent application after publication

Application publication date: 20200630