CN111398447A - Kit for detecting 12 kinds of steroid hormones in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology - Google Patents

Kit for detecting 12 kinds of steroid hormones in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology Download PDF

Info

Publication number
CN111398447A
CN111398447A CN202010172191.5A CN202010172191A CN111398447A CN 111398447 A CN111398447 A CN 111398447A CN 202010172191 A CN202010172191 A CN 202010172191A CN 111398447 A CN111398447 A CN 111398447A
Authority
CN
China
Prior art keywords
solution
dehydroepiandrosterone
methanol
estradiol
progesterone
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.)
Granted
Application number
CN202010172191.5A
Other languages
Chinese (zh)
Other versions
CN111398447B (en
Inventor
成晓亮
李美娟
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.)
Fujian Pinsheng Wanfu Medical Testing Co ltd
Fuzhou Pinsheng Medical Technology Co ltd
Nanjing Pinsheng Medical Laboratory Co ltd
Nanjing Pinsheng Medical Technology Co ltd
Original Assignee
Nanjing Pinsheng Medical Laboratory 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 Nanjing Pinsheng Medical Laboratory Co ltd filed Critical Nanjing Pinsheng Medical Laboratory Co ltd
Priority to CN202010172191.5A priority Critical patent/CN111398447B/en
Publication of CN111398447A publication Critical patent/CN111398447A/en
Application granted granted Critical
Publication of CN111398447B publication Critical patent/CN111398447B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • G01N30/50Conditioning of the sorbent material or stationary liquid
    • 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

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)
  • Steroid Compounds (AREA)

Abstract

The invention relates to a kit for detecting 12 kinds of steroid hormones in serum by an ultra-high performance liquid chromatography tandem mass spectrometry technology, wherein a serum sample is mixed with mixed internal standard solutions of all substances to be detected, the sample does not need derivatization treatment, and the solution to be detected can be obtained by only one-step liquid-liquid extraction; the ionization efficiency of certain target compounds can be effectively improved by adding the electrolyte ammonium fluoride into the mobile phase; and a mass spectrum positive-negative switching scanning mode is adopted to simultaneously detect 12 hormones, the detection types are multiple, the sensitivity is high, the specificity is strong, the cost is low, the pretreatment process is simple and quick, solid-phase extraction or derivation is not needed, the separation and detection of the 12 hormones can be completed within 5.0min, and a reliable detection kit is provided for the clinical health assessment of endocrine diseases.

Description

Kit for detecting 12 kinds of steroid hormones in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology
Technical Field
The invention belongs to the technical field of blood detection, and particularly relates to a kit for detecting 12 steroid hormones in serum by using an ultra-high performance liquid chromatography tandem mass spectrometry technology.
Background
The steroid hormones are lipid-soluble small molecular hormones, also called steroid hormones, which are generated by cholesterol through a series of enzyme-catalyzed metabolism and play an important role in the aspects of regulating the metabolism, the immunity, the sexual function, the growth and development of organisms and the like.
Progesterone (P4), also known as progesterone, from the source of the adrenal gland, corpus luteum and placenta, progesterone assays can determine ovulatory phase, assess infertility, assess abnormal uterine bleeding, high risk placental health in pregnancy, etc. pathological increases in progesterone are seen in toxemia of pregnancy, preeclampsia, essential hypertension, congenital disorders of 17-hydroxyprogesterone production, etc. pathological decreases in progesterone are seen in threatened abortion, ectopic pregnancy, premature birth, amenorrhea, retarded fetal development, stillbirth, infertility, corpus luteum insufficiency, severe disturbances of adrenal or thyroid gland function, etc. 17 α -hydroxyprogesterone and progesterone are the prerequisite substances for other steroid hormones, whose increased levels contribute to conception.
Aldosterone (a L D) is a mineralocorticoid hormone that acts primarily on the kidneys to effect reabsorption of sodium ions and water, and in general, aldosterone is a hormone that enhances the reabsorption of ions and water by the kidneys and is part of the renin-angiotensin system.
Cortisol (F) is a glucocorticoid and also the most important anti-stress hormone in the body, protecting the body from excessive stress damage. Cortisol raises blood glucose levels, supplies the body with the energy needed to escape injury, and works in concert with insulin to supply the cells with the glucose needed to produce energy. Humans with high cortisol content are very immunocompromised because cortisol can suppress most immune cells, especially leukocytes. In summary, cortisol maintains life through two opposing but interrelated regulatory activities: first, release and activate the body's defense mechanisms; the second is to shut down and regulate the same mechanism to protect against over-reaction causing cell damage or death.
Dehydroepiandrosterone sulfate (DHEA-S) is secreted mainly by the adrenal cortex and also by the ovaries in small amounts, and is a precursor of all sex hormones. Research shows that DHEA-S has the effects of resisting aging, maintaining cardiovascular health, reducing blood fat, lowering blood pressure, improving immune system function, enhancing bone, smoothing and refining skin, enhancing memory, increasing vitality and the like. DHEA-S peaks between 20 and 30 years of age and decreases significantly with age.
Testosterone (T) is mainly secreted by ovary and adrenal gland, and its content is increased mostly in idiopathic precocious puberty (adult level in male children), adrenal hyperplasia (male children), adrenal cortex tumor, female testosterone increase, and hirsutism, acne, hypomenorrhea or infertility, etc. Testosterone can be directly or first converted into more active Dihydrotestosterone (DHT), and can bind to androgen receptor of spermatogenic cells to promote sperm production, stimulate reproductive organ development, and promote male secondary sexual characteristics and maintain normal form. After male adults, excessive dihydrotestosterone can cause pathological phenomena such as whelk, alopecia and the like. In addition, prostatic hyperplasia in the middle-aged and the elderly is also closely related to dihydrotestosterone.
Androstenedione (AD) is an androgen with prohormone properties, primarily used to assess adrenal, ovarian, or testicular function. In men, 90% are secreted by the adrenal cortex. In women, androstenedione is derived from the ovary in 50% and adrenal gland in 50%. Hyperandrogenism in women may cause hyperandrogenism. Slightly lower levels of androstenedione in adult males, with significantly higher levels in age-matched females, may indicate adrenal cortical hyperplasia and gonadal tumors.
Estrone (E1) and estradiol (E2) are two estrogens mainly synthesized by the ovary, and their physiological actions are those of estrogens. The content of estrone in women of childbearing age is lower than that of estradiol, but the content of estrone in obese men and women of near menopause is relatively higher than that of estradiol, and the ratio of E1/E2 is obviously increased after menopause. Estrone is the major estrogen in postmenopausal women and is transformed by adipocytes. Estradiol is the major female hormone and is responsible for regulating female characteristics, the maturation of the accessory sexual organs and the menstrual-ovulation cycle, promoting the production of the ductal system of the mammary gland. Estradiol not only has an important role in reproductive and sexual function, but also affects other organs such as the bones.
The detection of the steroid hormones can help to diagnose some endocrine disorders, such as congenital adrenal cortical hyperplasia, aldosteronism, Cushing's syndrome and other diseases, at present, the immunoassay is the most commonly used kit in domestic clinical laboratories, but has the defects of poor specificity, false positive or false negative results caused by interferents such as heterophilic antibodies and the like, the immunoassay has low flux, only one hormone can be detected at one time, and the L C-MS/MS technology simultaneously detects retention time and target parent ions and fragment ions, has higher specificity and less potential interference, is particularly suitable for quantitative detection of complex biological matrixes, but the current L C-MS/MS technology still has a plurality of problems when detecting the steroid hormones, and the pretreatment mostly adopts a solid phase extraction or derivation method, has complex operation and high cost, and is relatively limited in clinical application.
Disclosure of Invention
The invention aims to provide a kit for detecting 12 steroid hormones in serum by using an ultra-high performance liquid chromatography tandem mass spectrometry technology on the basis of the prior art.
The invention also aims to provide application of the kit in detecting the 12 kinds of steroid hormones in serum by using an ultra performance liquid chromatography tandem mass spectrometry technology.
The technical scheme of the invention is as follows:
the 12 steroid hormones are progesterone (P4), 17 α -hydroxyprogesterone (17 α -OHP4), Corticosterone (CORT), Dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), testosterone (T), Dihydrotestosterone (DHT), Androstenedione (AD), estrone (E1), estradiol (E2), aldosterone (A L D) and cortisol (F);
the 12 steroid hormones corresponding isotope internal standard substances are progesterone-D9 (P4-D9), 17 α -hydroxyprogesterone-D8 (17 α -OHP4-D8), corticosterone-D8 (CORT-D8), dehydroepiandrosterone-D2 (DHEA-D2), dehydroepiandrosterone sulfate-D6 (DHEAS-D6), testosterone-D3 (T-D3), dihydrotestosterone-D3 (DHT-D3), androstenedione-13C 3(AD-13C3), estrone-D4 (E1-D4), estradiol-D4 (E2-D4), aldosterone-D4 (A L D-D4) and cortisol-D4 (F-D4);
the kit comprises the following reagents:
(1) eluent:
eluent A: an aqueous solution containing 50 to 400. mu.M ammonium fluoride; eluent B: methanol;
(2) calibration solution:
a mixed standard S0 solution containing 200ng/m L progesterone, 200ng/m L17 α -hydroxyprogesterone, 400ng/m L0 corticosterone, 400ng/m L1 dehydroepiandrosterone, 200000ng/m L2 dehydroepiandrosterone sulfate, 200ng/m L testosterone, 100ng/m L dihydrotestosterone, 200ng/m L androstenedione, 40ng/m L estrone, 40ng/m L estradiol, 40ng/m L aldosterone and 4000ng/m L cortisol is prepared into calibrator solutions with eight different concentration points by using a blank serum matrix solution, wherein the eight concentration points of the calibrator solutions are as follows:
the concentrations of the progesterone, the 17 α -hydroxyprogesterone, the testosterone and the androstenedione are the same, and the eight concentrations are 0.05ng/ml, 0.1ng/ml, 0.2ng/ml, 0.5ng/ml, 1ng/ml, 2.5ng/ml, 5ng/ml and 10ng/ml in sequence;
the concentrations of corticosterone and dehydroepiandrosterone are the same, and the eight concentrations are as follows: 0.1ng/ml, 0.2ng/ml, 0.4ng/ml, 1ng/ml, 2ng/ml, 5ng/ml, 10ng/ml and 20 ng/ml;
the eight concentrations of dehydroepiandrosterone sulfate are as follows in sequence: 50ng/ml, 100ng/ml, 200ng/ml, 500ng/ml, 1000ng/ml, 2500ng/ml, 5000ng/ml and 10000 ng/ml;
the eight concentrations of dihydrotestosterone are as follows: 0.025ng/ml, 0.05ng/ml, 0.1ng/ml, 0.25ng/ml, 0.5ng/ml, 1.25ng/ml, 2.5ng/ml and 5 ng/ml;
the eight concentrations of estrone, estradiol and aldosterone are in sequence: 0.01ng/ml, 0.02ng/ml, 0.04ng/ml, 0.1ng/ml, 0.2ng/ml, 0.5ng/ml, 1ng/ml and 2 ng/ml;
the eight concentrations of cortisol are as follows: 1ng/ml, 2ng/ml, 4ng/ml, 10ng/ml, 20ng/ml, 50ng/ml, 100ng/ml and 200 ng/ml;
(3) mixing internal standard working solution:
contains 10ng/m L progesterone-d 9, 5ng/m L17 α -hydroxyprogesterone-d 8, 20ng/m L0 corticosterone-d 8, 20ng/m L1 dehydroepiandrosterone-d 2, 10000ng/m L2 dehydroepiandrosterone sulfate-d 6, 10ng/m L testosterone-d 3, 5ng/m L dihydrotestosterone-d 3, 10ng/m L androstenedione-13C 3, 1ng/m L estrone-d 4, 2ng/m L estradiol-d 4, 4ng/m L aldosterone-d 4 and 100ng/m L cortisol-d 4;
(4) extracting liquid: methyl tert-butyl ether;
(5) compounding the solution: 40-60% methanol water solution;
(6) quality control product:
blank serum matrix solution containing 12 steroid hormones at three concentrations, low, medium and high, QC (L), QC (m) and QC (h), respectively, wherein:
QC (L) was performed to dilute the mixed standard S0 solution 2000-fold with blank serum matrix solution (1: 1999);
qc (m) was 400-fold dilution of the mixed standard S0 solution with a blank serum matrix solution (1: 399);
qc (h) was 50-fold (1:49) dilution of the mixed standard S0 solution with a blank serum matrix solution.
In a preferred embodiment, the eluent A is an aqueous solution containing 50-100 μ M ammonium fluoride. In a more preferred embodiment, eluent A is an aqueous solution containing 50. mu.M ammonium fluoride.
In one scheme, the blank serum substrate is 5-20% methanol water solution; preferably 5 to 15 percent of methanol aqueous solution; more preferably 10% aqueous methanol.
In one scheme, the redissolution provided by the invention is a 45-55% methanol aqueous solution; preferably 50% aqueous methanol.
The mixed standard S0 solution is prepared by preparing progesterone, 17 α -hydroxyprogesterone, corticosterone, dehydroepiandrosterone sulfate, testosterone, dihydrotestosterone, androstenedione, estrone, estradiol, aldosterone and cortisol into standard mother liquor with the concentrations of 2mg/m L, 2mg/m L0, 4mg/m L1, 1mg/m L2, 10mg/m L, 2mg/m L, 1mg/m L, 4mg/m L, 2mg/m L, 1mg/m L, 1mg/m L and 4mg/m L in sequence by using pure methanol;
the mother liquor of each standard substance is prepared into mixed standard S0 solution containing 200ng/m L progesterone, 200ng/m L17 α -hydroxyprogesterone, 400ng/m L0 corticosterone, 400ng/m L1 dehydroepiandrosterone, 200000ng/m L2 dehydroepiandrosterone sulfate, 200ng/m L testosterone, 100ng/m L dihydrotestosterone, 200ng/m L androstenedione, 40ng/m L estrone, 40ng/m L estradiol, 40ng/m L aldosterone and 4000ng/m L cortisol by using methanol aqueous solution.
When preparing the mixing standard S0 solution, the methanol aqueous solution is 30-70% methanol aqueous solution, preferably 45-55% methanol aqueous solution, and more preferably 50% methanol aqueous solution.
The mixed internal standard working solution is prepared by weighing various isotope internal standards including progesterone-D9 (P4-D9), 17 α -hydroxyprogesterone-D8 (17 α -OHP4-D8), corticosterone-D8 (CORT-D8), dehydroepiandrosterone-D2 (DHEA-D2), dehydroepiandrosterone-D6 (DHEAS-D6), testosterone-D6 (T-D6), dihydrotestosterone-D6 (DHT-D6), androstenedione-13C 6 (AD-13C 6), estrone-D6 (E6-D6), estradiol-D6 (E6-D6), aldosterone-D6 (A6D 6) and cortisol-D6 (F-D6), pure methanol are added to completely dissolve isotope internal standards such as isotope concentrations of progesterone-D581/6, isotope internal standard working solution/6, isotope internal standard isotope concentration of 361/6, isotope concentration of isotope internal standard isotope 1/6, isotope/6, isotope concentration of isotope internal standard isotope 1/6, isotope/361/6 and isotope internal standard isotope concentration of isotope/361/6, isotope/361/72, isotope/365 mg/6, isotope internal standard isotope content of isotope/6 and isotope of isotope internal standard isotope/6.
The mother liquor of each isotope internal standard is prepared into internal standard solutions of 100ng/m L progesterone-D9 (P4-D9), 50ng/m L17 α -hydroxyprogesterone-D8 (17 α -OHP4-D8), 200ng/m L0 corticosterone-D8 (CORT-D8), 200ng/m L1 dehydroepiandrosterone-D L (DHEA-D L), 100000ng/m L2 dehydroepiandrosterone sulfate-D L (DHEAS-D L), 100ng/m L3 testosterone-D L (T-D L), 50ng/m L4 dihydrotestosterone-D L (DHT-D L), 100ng/m L androstenedione-13C L (AD-13C L), 10ng/m L-D L (E72-D L), 20ng/m L-estradiol-13C L (SI-D L) and 10ng/m L F-D L (SI-D L).
And adding a methanol aqueous solution of 900 mu L into a SI solution of 100 mu L, and uniformly mixing to obtain a mixed internal standard working solution.
When the internal standard working solution is mixed, the methanol aqueous solution is 30-70% methanol aqueous solution, preferably 45-55% methanol aqueous solution, and more preferably 50% methanol aqueous solution.
In a preferred scheme, the kit for detecting 12 kinds of steroid hormones in serum by the ultra performance liquid chromatography tandem mass spectrometry technology comprises the following reagents:
(1) eluent:
eluent A: containing 50 μ M ammonium fluoride aqueous solution; eluent B: methanol;
(2) calibration solution:
preparing progesterone, 17 α -hydroxyprogesterone, corticosterone, dehydroepiandrosterone sulfate, testosterone, dihydrotestosterone, androstenedione, estrone, estradiol, aldosterone and cortisol into standard mother liquor with the concentration of 2mg/m L, 2mg/m L0, 4mg/m L1, 1mg/m L2, 10mg/m L, 2mg/m L, 1mg/m L, 4mg/m L, 2mg/m L, 1mg/m L, 1mg/m L and 4mg/m L in sequence by using pure methanol;
preparing the mother liquor of each standard product into mixed standard S0 solution containing 200ng/m L progesterone, 200ng/m L17 α -hydroxyprogesterone, 400ng/m L0 corticosterone, 400ng/m L1 dehydroepiandrosterone, 200000ng/m L2 sulfuric acid dehydroepiandrosterone, 200ng/m L testosterone, 100ng/m L dihydrotestosterone, 200ng/m L androstenedione, 40ng/m L estrone, 40ng/m L estradiol, 40ng/m L aldosterone and 4000ng/m L cortisol by using 50% methanol aqueous solution;
preparing the mixed standard S0 solution into calibration substance solutions with eight different concentration points by using a 10% methanol solution;
(3) mixing internal standard working solution:
weighing isotope internal standard substances including progesterone-d 9, 17 α -hydroxyprogesterone-d 8, corticosterone-d 8, dehydroepiandrosterone-d 2, dehydroepiandrosterone-d 6 sulfate, testosterone-d 3, dihydrotestosterone-d 3, androstenedione-13C 3, estrone-d 4, estradiol-d 4, aldosterone-d 4 and cortisol-d 4, respectively adding pure methanol to completely dissolve, and preparing into isotope internal standard mother liquor with the concentration of 1mg/m L, 1mg/m L0, 5mg/m L1, 2mg/m L2, 1mg/m L, 0.1mg/m L, 1mg/m L, 1mg/m L, 2.5mg/m L, 1mg/m L, 1mg/m L and 1mg/m L;
the isotope internal standard mother liquor is prepared into an isotope internal standard SI solution containing 100ng/m L progesterone-d 9, 50ng/m L17 α -hydroxyprogesterone-d 8, 200ng/m L0 corticosterone-d 8, 200ng/m L1 dehydroepiandrosterone-d 2, 100000ng/m L2 dehydroepiandrosterone sulfate-d 6, 100ng/m L testosterone-d 3, 50ng/m L dihydrotestosterone-d 3, 100ng/m L androstenedione-13C 3, 10ng/m L estrone-d 4, 20ng/m L estradiol-d 4, 40ng/m L aldosterone-d 4 and 1000ng/m L cortisol-d 4 by 50% methanol aqueous solution;
adding 100 mu L SI solution into 900 mu L50% methanol water solution, and uniformly mixing to obtain mixed internal standard working solution;
(4) extracting liquid: methyl tert-butyl ether;
(5) compounding the solution: 50% aqueous methanol;
(6) quality control product:
10% aqueous methanol solution containing 12 steroid hormones at three concentrations, low, medium and high, QC (L), QC (M) and QC (H), respectively, wherein:
QC (L) contains 0.1ng/m L progesterone (P4), 0.1ng/m L017 α -hydroxyprogesterone (17 α -OHP4), 0.2ng/m L1 Corticosterone (CORT), 0.2ng/m L2 Dehydroepiandrosterone (DHEA), 100ng/m L3 dehydroepiandrosterone sulfate (DHEAS), 0.1ng/m L4 testosterone (T), 0.05ng/m L5 Dihydrotestosterone (DHT), 0.1ng/m L Androstenedione (AD), 0.02ng/m L estrone (E1), 0.02ng/m L estradiol (E2), 0.02ng/m L aldosterone (A L D) and 2ng/m L cortisol (F).
QC (M) comprises 0.5ng/m L progesterone (P4), 0.5ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 1ng/m L0 Corticosterone (CORT), 1ng/m L1 Dehydroepiandrosterone (DHEA), 500ng/m L2 dehydroepiandrosterone sulfate (DHEAS), 0.5ng/m L3 testosterone (T), 0.25ng/m L4 Dihydrotestosterone (DHT), 0.5ng/m L Androstenedione (AD), 0.1ng/m L estrone (E1), 0.1ng/m L estradiol (E2), 0.1ng/m L aldosterone (A L D) and 10ng/m L cortisol (F).
QC (H) comprises 5ng/m L progesterone (P4), 5ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 10ng/m L0 Corticosterone (CORT), 10ng/m L1 Dehydroepiandrosterone (DHEA), 5000ng/m L2 sulfuric acid Dehydroepiandrosterone (DHEAS), 5ng/m L3 testosterone (T), 2.5ng/m L4 Dihydrotestosterone (DHT), 5ng/m L Androstenedione (AD), 1ng/m L estrone (E1), 1ng/m L estradiol (E2), 1/m L aldosterone (A L D) and 100ng/m L cortisol (F).
Specifically, the concentrations of QC (L), QC (m), QC (h) corresponding to 12 steroid hormones in the quality control are shown in table 1;
TABLE 1 corresponding concentration of quality control (unit ng/m L)
Figure BDA0002409572100000071
In a preferred embodiment, the calibrator solution is prepared as follows:
accurately weighing 3-5mg of each standard substance powder to be detected in a 5m L centrifuge tube, preparing the mother liquor concentration of the standard substance in the following table by using pure methanol, except dehydroepiandrosterone sulfate (DHEAS), diluting the mother liquor of each other standard substance into the use concentration of 10 mu g/m L by using 50% methanol aqueous solution, preparing the use concentrations into a mixed standard solution S0 (detailed in table 2), and uniformly mixing for later use.
TABLE 2 preparation of the Mixed reference solution S0
Figure BDA0002409572100000072
A50 μ L mixed standard S0 solution was added to 950 μ L10% aqueous methanol as the first high concentration point S8 and diluted stepwise to S1 (see Table 3 for details), with the concentrations of the various points listed in Table 3.
TABLE 3 formulation and concentration of the standard
Figure BDA0002409572100000081
(Note: concentration units are ng/m L)
The concentration of the aqueous methanol solution referred to in the present invention generally means a volume concentration.
In a preferred embodiment, the mixed internal standard working solution is prepared according to the following method:
accurately weighing 3-5mg of each isotope internal standard substance in a 5m L centrifuge tube, preparing the isotope internal standard mother liquor concentration in the following table by using pure methanol, except dehydroepiandrosterone-d 6(DHEAS-d6) sulfate, diluting the rest isotope internal standard mother liquor into the use concentration of 10 mu g/m L by using 50% methanol aqueous solution, preparing the use concentrations into mixed internal standard SI solution (detailed in table 4), finally taking 100 mu L SI solution, adding 900 mu L50% methanol aqueous solution, and uniformly mixing to obtain the mixed internal standard working solution.
TABLE 4 preparation of SI solution as internal Mixed standard
Figure BDA0002409572100000082
The application of the kit in detecting the 12 kinds of steroid hormones in the serum by using the ultra performance liquid chromatography tandem mass spectrometry technology is also within the protection scope of the invention.
The specific detection method comprises the following steps:
a method for detecting 12 steroid hormones in serum by an ultra-high performance liquid chromatography tandem mass spectrometry technology,
mixing a serum sample with internal standard solutions of all objects to be detected, obtaining the solution to be detected through one-step liquid-liquid extraction, detecting 12 kinds of steroid hormones in the preprocessed serum by adopting an ultra-high performance liquid chromatography tandem mass spectrometry technology, firstly separating a target object to be detected from interfering components in a serum matrix by utilizing the ultra-high performance liquid chromatography, then detecting the mass-to-charge ratio (m/z) of the target object and the corresponding isotope internal standard thereof by utilizing mass spectrometry, quantifying by using an isotope internal standard method, and accurately calculating the content of the 12 kinds of steroid hormones, wherein the specific chromatographic conditions are as follows:
(1) ultra-high performance liquid chromatography conditions:
mobile phase A: an aqueous solution containing 50 to 400. mu.M ammonium fluoride; mobile phase B: methanol;
column model Kinetex XB-C18(3.0 × 50mm,2.6 μm); (Phenomenex, USA)
And (3) performing gradient elution by adopting the mobile phase A and the mobile phase B as a mixed mobile phase, wherein the gradient elution process is as follows: the volume ratio of the mobile phase A to the mobile phase B is gradually changed from 60:40 to 2:98 at a constant speed within 0-3.0 minutes; the volume ratio of the mobile phase A to the mobile phase B is 2:98 within 3.0-3.5 minutes; the volume ratio of the mobile phase A to the mobile phase B is gradually changed from 2:98 to 60:40 at a constant speed within 3.5-5.0 minutes.
(2) Mass spectrum conditions:
in an electrospray ionization (ESI) mode, Multiple Reaction Monitoring (MRM) is adopted to perform positive and negative switching scanning, the spraying voltage is 3.0kV (ESI +)/2.5kV (ESI-), the ion source temperature is 120 ℃, the atomizing gas temperature is 400 ℃, the atomizing gas flow rate is 800L/h, the cone hole gas flow rate is 150L/h, and 12 steroid hormones and corresponding isotope internal standards thereof are monitored.
The invention adds ammonium fluoride into the mobile phase A, which can effectively improve the ionization efficiency of some target compounds, under the coordination of other conditions, compared with the prior art that an L C-MS/MS method is adopted to detect steroid hormones, the invention has higher sensitivity, the pretreatment only needs one-step liquid-liquid extraction, the invention is simple, fast and low in cost, and 12 important steroid hormones can be simultaneously detected within 5 minutes.
In the chromatography, the selection of the chromatographic column is very important, and the requirements on the chromatographic column are high column efficiency, good selectivity, high analysis speed and the like, the invention adopts ammonium fluoride aqueous solution containing 50-400 MuM and methanol as mobile phases, the type of the chromatographic column is KinetexXB-C18(3.0 × 50mm,2.6 Mum), endogenous substances do not interfere the determination of a sample under the coordination of other conditions, the sensitivity is high, the specificity is strong, the cost is low, the pretreatment process is simple, the separation and the detection can be completed within 5.0min, and the precision and the accuracy meet the requirements.
The present invention uses progesterone-D9 (P4-D9), 17 α -hydroxyprogesterone-D8 (17 α -OHP 4-D4), corticosterone-D4 (CORT-D4), dehydroepiandrosterone-D4 (DHEA-D4), dehydroepiandrosterone-D4 sulfate (DHEAS-D4), testosterone-D4 (T-D4), dihydrotestosterone-D4 (DHT-D4), enedione-13C 4 (AD-13C 4), deuterone-D4 (DHEAS-D4), estradiol-D4 (T-D4), estradiol-D4 (OHA-D4), estradiol-72 (estradiol-72), estradiol-72, estradiol-D4, estradiol-72, estradiol-b 4, and serum, and the like, and the internal standard method is used to determine the accuracy of the internal standard method.
In one scheme, the flow rate is 0.2-1.0 m L/min, and preferably 0.5m L/min.
Further, the column temperature is 40-60 ℃, preferably 50 ℃.
Furthermore, the injection volume is 2-10 mu L, preferably 5 mu L.
In a preferred scheme, when the ultra-high performance liquid chromatography tandem mass spectrometry technology is adopted to detect 12 kinds of steroid hormones in serum, the specific chromatographic conditions are as follows:
(1) high performance liquid chromatography conditions:
mobile phase A: water (50 μ M ammonium fluoride);
mobile phase B: methanol;
column model Kinetex XB-C18(3.0 × 50mm,2.6 μm) (Phenomenex, USA);
adopting gradient elution mode, as shown in Table 5, with flow rate of 0.5m L/min, column temperature of 50 deg.C, and injection volume of 5 μ L;
TABLE 5 mobile phase gradient elution parameters
Time of day Flow rate (m L/min) %A %B Curve
0.0 0.5 60 40 -
3.0 0.5 2 98 6
3.5 0.5 2 98 6
5.0 0.5 60 40 1
(2) Mass spectrum conditions:
in an electrospray ionization (ESI) mode, multi-reaction monitoring (MRM) is adopted to perform positive and negative switching scanning, the spraying voltage is 3.0kV (ESI +)/2.5kV (ESI-), the ion source temperature is 120 ℃, the atomizing gas temperature is 400 ℃, the atomizing gas flow rate is 800L/h, the cone hole gas flow rate is 150L/h, 12 steroid hormones and corresponding isotope internal standards thereof are monitored, and the mass spectrum acquisition parameters of each target substance to be detected are shown in Table 6.
TABLE 6 steroid hormone Mass Spectrometry parameters
Figure BDA0002409572100000111
The serum mentioned in the invention is human or animal serum.
In one protocol, pre-treated serum was prepared as follows: adding mixed internal standard working solution into serum, adding the extract liquor for extraction after vortex, taking supernatant after centrifugal treatment, drying the supernatant by nitrogen flow, mixing the supernatant with redissolution, and taking the supernatant after centrifugal treatment again.
Preferably, the extract is methyl tert-butyl ether.
Preferably, the double solution is a 40% to 60% aqueous solution of methanol, for example, a 50% aqueous solution of methanol, without affecting the effect of the present invention.
In a preferred embodiment, the pre-treated serum is prepared by placing 200 μ L serum into a 1.5m L centrifuge tube, adding 20 μ L mixed internal standard working solution, vortexing, extracting with 800 μ L methyl tert-butyl ether, centrifuging at 15 deg.C and 15000r/min for 5min, collecting 700 μ L supernatant, blowing the supernatant with nitrogen, mixing with 80 μ L50% methanol aqueous solution, centrifuging at 15000r/min for 3min, and collecting 60 μ L supernatant.
In a more preferred embodiment, the pre-treated serum is prepared by placing 200 μ L serum into a 1.5m L centrifuge tube, adding 20 μ L mixed internal standard working fluid, vortexing for 5s, adding 800 μ L methyl t-butyl ether, shaking at high speed (max. shaking) for 5min, centrifuging at 15 deg.C at 15000r/min for 5min, transferring 700 μ L supernatant to another centrifuge tube, blowing nitrogen to near dryness (without heating), adding 80 μ L50% methanol (covered with a lid), shaking at high speed for 2min, centrifuging at 15000r/min for 3min, and transferring 60 μ L supernatant to a liner tube for testing, wherein the sample amount is 5 μ L.
In one embodiment, the mixed internal standard working solution is prepared as follows:
weighing various isotope internal standard substances, including progesterone-D9 (P4-D9), 17 α -hydroxyprogesterone-D8 (17 α -OHP4-D8), corticosterone-D8 (CORT-D8), dehydroepiandrosterone-D2 (DHEA-D2), dehydroepiandrosterone-D6 (DHEAS-D6), testosterone-D6 (T-D6), dihydrotestosterone-D6 (DHT-D6), androstenedione-13C 6 (AD-13C 6), estrone-D6 (E6-D6), estradiol-D6 (E6-D6), aldosterone-D6 (A6D 6) and androstenol-D6 (F-D6) (6) by adding pure methanol for complete dissolution, and preparing isotope internal standard solutions with the concentrations of 1mg/m, 361 mg/m, 6 mg/D6, 361 mg/m, 365 mg/m, 6 mg/6, 361 mg/m, 6 mg/m, 365 mg/m and isotope internal standard solutions of cortisone, 6 (F-D6, 361 mg/m, 6, 365 mg/m/6, 361 mg/m/6 and isotope internal standard solutions of;
the mother liquor of the isotope internal standard is prepared into internal standard solution containing 100ng/m L progesterone-D9 (P4-D9), 50ng/m L17 α -hydroxyprogesterone-D8 (17 α -OHP4-D8), 200ng/m L0 corticosterone-D8 (CORT-D8), 200ng/m L1 dehydroepiandrosterone-D L (DHEA-D L), 100000ng/m L2 dehydroepiandrosterone sulfate-D L (DHEAS-D L), 100ng/m L3 testosterone-D L (T-D L), 50ng/m L4 dihydrotestosterone-D L (DHT-D L), 100ng/m L androstenedione-13C L (AD-13C L), 10ng/m L ketone-D L (E L-D L), 100ng/m L-D L, estradiol-72 (SI-L A-L/72), and estradiol-72 (SI-L A-L/1000A);
and adding 100 mu L SI solution into 900 mu L50% methanol water solution, and uniformly mixing to obtain the mixed internal standard working solution.
In one embodiment, the standard solution is prepared as follows:
progesterone (P4), 17 α -hydroxyprogesterone (17 α -OHP4), Corticosterone (CORT), Dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), testosterone (T), Dihydrotestosterone (DHT), Androstenedione (AD), estrone (E1), estradiol (E2), aldosterone (A α 0D) and cortisol (F) are prepared into standard mother solutions with pure methanol, wherein the concentrations of the standard mother solutions are sequentially 2mg/m α 1, 2mg/m α 2, 4mg/m α 3, 1mg/m α 4, 10mg/m α 5, 2mg/m L, 1mg/m L, 4mg/m L, 2mg/m L, 1mg/m L, 1mg/m L and 4mg/m L L.
The mother liquor of each standard product is prepared into mixed standard S0 solution containing 200ng/m L progesterone (P4), 200ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 400ng/m L0 Corticosterone (CORT), 400ng/m L1 Dehydroepiandrosterone (DHEA), 200000ng/m L2 sulfuric acid Dehydroepiandrosterone (DHEAS), 200ng/m L3 testosterone (T), 100ng/m L4 Dihydrotestosterone (DHT), 200ng/m L Androstenedione (AD), 40ng/m L estrone (E1), 40ng/m L estradiol (E2), 40ng/m L aldosterone (A L D) and 4000ng/m L cortisol (F) by 50% methanol aqueous solution.
The mixed standard S0 solution was prepared as a blank serum base (10% methanol in water) into eight calibrator solutions at different concentration points:
the concentrations of progesterone (P4), 17 α -hydroxyprogesterone (17 α -OHP4), testosterone (T) and Androstenedione (AD) are the same, and the eight concentrations are 0.05ng/ml, 0.1ng/ml, 0.2ng/ml, 0.5ng/ml, 1ng/ml, 2.5ng/ml, 5ng/ml and 10ng/ml in sequence;
corticosterone (CORT) and Dehydroepiandrosterone (DHEA) were at the same concentrations, eight concentrations in order: 0.1ng/ml, 0.2ng/ml, 0.4ng/ml, 1ng/ml, 2ng/ml, 5ng/ml, 10ng/ml and 20 ng/ml;
the eight concentrations of dehydroepiandrosterone sulfate (DHEAS) are as follows: 50ng/ml, 100ng/ml, 200ng/ml, 500ng/ml, 1000ng/ml, 2500ng/ml, 5000ng/ml and 10000 ng/ml;
the eight concentrations of Dihydrotestosterone (DHT) are in order: 0.025ng/ml, 0.05ng/ml, 0.1ng/ml, 0.25ng/ml, 0.5ng/ml, 1.25ng/ml, 2.5ng/ml and 5 ng/ml;
eight concentrations of estrone (E1), estradiol (E2) and aldosterone (A L D) are 0.01ng/ml, 0.02ng/ml, 0.04ng/ml, 0.1ng/ml, 0.2ng/ml, 0.5ng/ml, 1ng/ml and 2ng/ml in sequence;
the eight concentrations of cortisol (F) are in order: 1ng/ml, 2ng/ml, 4ng/ml, 10ng/ml, 20ng/ml, 50ng/ml, 100ng/ml and 200 ng/ml.
Taking 200 mu L of each concentration point sample, adding 20 mu L of mixed internal standard working solution into the sample, then whirling for 5s, adding 800 mu L of methyl tert-butyl ether, shaking at high speed (maximum shaking speed) for 5min, centrifuging at the rotating speed of 15000r/min for 5min at 15 ℃, transferring 700 mu L supernatant into another centrifuge tube, blowing nitrogen to be nearly dry (without heating), adding 80 mu L50% of methanol (with a cover covered), shaking at high speed for 2min, centrifuging at the speed of 15000r/min for 3min, transferring 60 mu L supernatant into a lining tube to be subjected to sample detection, and taking the sample with the sample amount of 5 mu L.
In one scheme, the quality control product is prepared according to the following method:
the mixed standard S0 solution was prepared into QC (L), QC (M), QC (H) at three different concentrations with a blank serum substrate (10% methanol in water).
QC (L) contains 0.1ng/m L progesterone (P4), 0.1ng/m L017 α -hydroxyprogesterone (17 α -OHP4), 0.2ng/m L1 Corticosterone (CORT), 0.2ng/m L2 Dehydroepiandrosterone (DHEA), 100ng/m L3 dehydroepiandrosterone sulfate (DHEAS), 0.1ng/m L4 testosterone (T), 0.05ng/m L5 Dihydrotestosterone (DHT), 0.1ng/m L Androstenedione (AD), 0.02ng/m L estrone (E1), 0.02ng/m L estradiol (E2), 0.02ng/m L aldosterone (A L D) and 2ng/m L cortisol (F).
QC (M) comprises 0.5ng/m L progesterone (P4), 0.5ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 1ng/m L0 Corticosterone (CORT), 1ng/m L1 Dehydroepiandrosterone (DHEA), 500ng/m L2 dehydroepiandrosterone sulfate (DHEAS), 0.5ng/m L3 testosterone (T), 0.25ng/m L4 Dihydrotestosterone (DHT), 0.5ng/m L Androstenedione (AD), 0.1ng/m L estrone (E1), 0.1ng/m L estradiol (E2), 0.1ng/m L aldosterone (A L D) and 10ng/m L cortisol (F).
QC (H) comprises 5ng/m L progesterone (P4), 5ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 10ng/m L0 Corticosterone (CORT), 10ng/m L1 Dehydroepiandrosterone (DHEA), 5000ng/m L2 sulfuric acid Dehydroepiandrosterone (DHEAS), 5ng/m L3 testosterone (T), 2.5ng/m L4 Dihydrotestosterone (DHT), 5ng/m L Androstenedione (AD), 1ng/m L estrone (E1), 1ng/m L estradiol (E2), 1/m L aldosterone (A L D) and 100ng/m L cortisol (F).
Respectively taking quality control solution QC (L), QC (M), QC (H) and QC (H) 200 mu L in a centrifuge tube of 1.5m L, adding 20 mu L mixed internal standard working solution into the centrifuge tube, then whirling for 5s, adding 800 mu L methyl tert-butyl ether, shaking at high speed (maximum shaking speed) for 5min, centrifuging at 15 ℃ for 5min at the rotating speed of 15000r/min, transferring 700 mu L supernatant into another centrifuge tube, blowing nitrogen to be nearly dry (without heating), adding 80 mu L50% methanol (covered with a cover), shaking at high speed for 2min, centrifuging at 15000r/min for 3min, transferring 60 mu L supernatant into a lining tube to be subjected to sample detection, and obtaining the sample amount of 5 mu L.
By adopting the technical scheme of the invention, the advantages are as follows:
the kit provided by the invention can be used for detecting 12 kinds of steroid hormones in serum, a serum sample is mixed with the mixed internal standard solution of all substances to be detected, the sample does not need derivatization treatment, and the solution to be detected can be obtained only by one-step liquid-liquid extraction; the ionization efficiency of certain target compounds can be effectively improved by adding the electrolyte ammonium fluoride into the mobile phase; and a mass spectrum positive-negative switching scanning mode is adopted to simultaneously detect 12 hormones, so that the sensitivity is high, the specificity is strong, the cost is low, the pretreatment process is simple, the separation and detection of the 12 hormones can be completed within 5.0min, and a reliable detection method is provided for the clinical health assessment of endocrine diseases.
Drawings
FIG. 1 is a selected ion flow chromatogram of a 12-steroid hormone standard;
FIG. 2 is a selective ion flow chromatogram of 12 steroid hormones in a serum sample.
Detailed Description
The kit of the present invention is further illustrated by the following examples in conjunction with the drawings, but these examples do not limit the present invention in any way.
Example 1:
first, experimental material and instrument
1. Material
The samples were obtained from serum samples collected from the clinic in 2018 and 3 months in the heart disease hospital, wuhan asia.
(1) The instrument comprises a Xevo TQ-S triple quadrupole mass spectrometer (Waters Corporation), an UP L C I-Class ultra-high performance liquid chromatography system (matched with an autosampler, Waters Corporation), a SCI L OGEX D2012 high-speed table centrifuge (USA), an ultrapure water instrument (E L GA L abWater, UK), a multi-tube Vortex mixer (Vortex generator 2, USA), an adjustable pipettor (Eppendorf 0.5-10 mu L, 10-100 mu L, 100-1000 mu L), a glass instrument, a measuring cylinder and the like.
(2) The reagent consumables are MS grade methanol (Fisher, USA), HP L grade C methanol (Honeywell, USA), HP L grade C methyl tert-butyl ether (Fisher, USA), and chromatographic column Kinetex XB-C18(3.0 × 50mm,2.6 μm) (Phenomenex corporation, USA).
(3) The standards P4, 17 α -OHP4, CORT, T, DHT, AD, E1 and F are available from Dr. Ehrensorfer, Germany, A L D, A L D-D4, P4-D9, CORT-D8, 17 α -OHP4-D8, DHEA-D2, F-D4, E1-D4, E2-D4 are available from TRC, DHEA and T-D3 are available from Cerilliant, DHEAS-D6 and AD-13C3 are available from IsoSciences, and E2 and DHT-D3 are available from Sigma.
(4) Quality control the concentrations of QC (L), QC (M), QC (H) corresponding to 12 steroid hormones contained in the quality control are shown in Table 1.
Second, liquid condition
(1) Chromatographic conditions, mobile phase A, water (containing 50 μ M ammonium fluoride-water solution), mobile phase B, methanol, chromatographic column model, Kinetex XB-C18(3.0 × 50mm,2.6 μ M), using gradient elution, see Table 5, flow rate of 0.5M L/min, column temperature of 50 ℃, sample injection volume of 5 μ L.
(2) The mass spectrum conditions are that under an electrospray ionization (ESI) mode, Multiple Reaction Monitoring (MRM) is adopted to perform positive and negative switching scanning, the spraying voltage is 3.0kV (ESI +)/2.5kV (ESI-), the ion source temperature is 120 ℃, the atomizing gas temperature is 400 ℃, the atomizing gas flow rate is 800L/h, the cone hole gas flow rate is 150L/h, 12 steroid hormones and corresponding isotope internal standards thereof are monitored, and the mass spectrum acquisition parameters of each target substance to be detected are shown in Table 6.
Second, the experimental procedure
(1) Preparing a standard substance:
accurately weighing 3-5mg of each standard in a 5m L centrifuge tube (the standard below 3mg does not need to be weighed and is completely dissolved), preparing the mother liquor concentration of the standard with pure methanol, diluting 11 kinds of the mother liquor of the standard with 50% methanol aqueous solution to the use concentration of 10 mu g/m L (DHEAS use concentration is 10000 mu g/m L of the mother liquor of the standard), and preparing the use concentrations into a mixed standard S0 solution (detailed in Table 2).
The mixed standard S0 solution was prepared into eight calibrator solutions with different concentration points using a blank serum base (10% methanol in water) according to the following procedure:
a50 μ L mixed standard S0 solution was added to 950 μ L10% aqueous methanol as the first high concentration point S8 and diluted stepwise to S1 (see Table 3 for details), with the concentrations of the various points listed in Table 3.
(2) Preparation of mixed internal standard working solution
Accurately weighing 3-5mg of each isotope internal standard substance into a 5m L centrifuge tube (standard substances with the specification below 3mg are not required to be weighed and are completely dissolved), preparing the concentration of isotope internal standard mother liquor by using pure methanol, then diluting 11 isotope internal standard mother liquors into the use concentration of 10 mu g/m L (DHEAS-d6 use concentration is 1000 mu g/m L of isotope internal standard mother liquor concentration) by using 50% methanol aqueous solution, preparing mixed internal standard SI solutions (detailed in table 4) by using the use concentrations, finally adding 900 mu L50% methanol into 100 mu L SI solutions, and uniformly mixing to obtain the mixed internal standard working solution.
(3) Preparing a quality control product:
the mixed standard S0 solution was mixed with 10% aqueous methanol to prepare QC (L), QC (M), and QC (H) at three different concentrations.
QC (L) contains 0.1ng/m L progesterone (P4), 0.1ng/m L017 α -hydroxyprogesterone (17 α -OHP4), 0.2ng/m L1 Corticosterone (CORT), 0.2ng/m L2 Dehydroepiandrosterone (DHEA), 100ng/m L3 dehydroepiandrosterone sulfate (DHEAS), 0.1ng/m L4 testosterone (T), 0.05ng/m L5 Dihydrotestosterone (DHT), 0.1ng/m L Androstenedione (AD), 0.02ng/m L estrone (E1), 0.02ng/m L estradiol (E2), 0.02ng/m L aldosterone (A L D) and 2ng/m L cortisol (F).
QC (M) comprises 0.5ng/m L progesterone (P4), 0.5ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 1ng/m L0 Corticosterone (CORT), 1ng/m L1 Dehydroepiandrosterone (DHEA), 500ng/m L2 dehydroepiandrosterone sulfate (DHEAS), 0.5ng/m L3 testosterone (T), 0.25ng/m L4 Dihydrotestosterone (DHT), 0.5ng/m L Androstenedione (AD), 0.1ng/m L estrone (E1), 0.1ng/m L estradiol (E2), 0.1ng/m L aldosterone (A L D) and 10ng/m L cortisol (F).
QC (H) comprises 5ng/m L progesterone (P4), 5ng/m L17 α -hydroxyprogesterone (17 α -OHP4), 10ng/m L0 Corticosterone (CORT), 10ng/m L1 Dehydroepiandrosterone (DHEA), 5000ng/m L2 sulfuric acid Dehydroepiandrosterone (DHEAS), 5ng/m L3 testosterone (T), 2.5ng/m L4 Dihydrotestosterone (DHT), 5ng/m L Androstenedione (AD), 1ng/m L estrone (E1), 1ng/m L estradiol (E2), 1/m L aldosterone (A L D) and 100ng/m L cortisol (F).
(4) Sample processing
1) Pretreatment of standard substance, taking 200 mu L of each concentration point sample, adding 20 mu L of mixed internal standard working solution, then whirling for 5s, adding 800 mu L of methyl tert-butyl ether, shaking at high speed (maximum shaking speed) for 5min, centrifuging at the rotating speed of 15000r/min15 ℃ for 5min, transferring 700 mu L supernatant into another centrifuge tube, blowing nitrogen to be nearly dry (without heating), adding 80 mu L50% of methanol (with a cover covered), shaking at high speed for 2min, centrifuging at 15000r/min for 3min, transferring 60 mu L supernatant into a lining tube, and detecting the sample loading amount by 5 mu L.
2) Pretreatment of a serum sample, namely putting 200 mu L serum into a 1.5m L centrifuge tube, adding 20 mu L mixed internal standard working solution into the centrifuge tube, then whirling for 5s, adding 800 mu L methyl tert-butyl ether, shaking at a high speed (maximum shaking speed) for 5min, centrifuging at a rotating speed of 15000r/min at 15 ℃ for 5min, transferring 700 mu L supernatant into another centrifuge tube, blowing nitrogen to be nearly dry (without heating), adding 80 mu L50% methanol (with a cover), shaking at a high speed for 2min, centrifuging at 15000r/min for 3min, transferring 60 mu L supernatant into an inner lining tube, and detecting the sample introduction amount is 5 mu L.
3) Quality control product pretreatment, wherein quality control product solutions QC (L), QC (M), QC (H) and QC (H) are respectively put into centrifuge tubes of 1.5m L with 200 mu L, and then are consistent with the pretreatment of serum samples, and the details are not repeated here.
The upper and lower peripheries of the kit are coated with membranes, the kit is shockproof and heat-insulated, the eluents A and B are placed at the upper left, 12 ampoules are respectively placed at the lower left, which are respectively a calibrator, a mixed internal standard working solution and a quality control product, and the 10m L complex solution and 120m L extract are respectively placed at the right.
The components of the assay kit are shown in Table 7.
TABLE 7 vitamin assay kit Components (100 parts)
Figure BDA0002409572100000171
Figure BDA0002409572100000181
Fourth, method verification
In the detection method of the invention, the peak shapes of the standard substance of 12 kinds of steroid hormones and the serum sample are symmetrical without interfering with other peaks, which indicates that good detection can be obtained under the condition, and FIG. 1 is a selective ion flow chromatogram of the standard substance of 12 kinds of steroid hormones; FIG. 2 is a selective ion flow chromatogram of 12 steroid hormones in a serum sample.
1. Standard curve:
by adopting an isotope internal standard quantitative method, and by using Target L ynx software, the concentration ratio of a standard substance to an internal standard substance is taken as an X axis, the peak area ratio of the standard substance to the internal standard substance is taken as a Y axis, a calibration curve is established, and the concentration of a steroid hormone to be tested substance in serum is calculated.12 linear fitting equations of steroid hormones in respective concentration ranges have good linearity, the correlation coefficient is more than 0.997, and the details are shown in Table 8.
TABLE 812 Retention time and Linear Range of steroid hormones
Figure BDA0002409572100000191
2. Minimum limit of quantitation
Because steroid hormones are low in human body content and have similar structures, and the requirements on the sensitivity and specificity of the method are high, the inventor optimizes and researches the detection method, the current minimum quantitative limit (LL OQ) basically meets the sensitivity requirement of simultaneous detection of 12 male and female sex hormones, and the concentration of LL OQ is specifically shown in Table 9.
TABLE 9 quantitative lower limit data sheet
Figure BDA0002409572100000192
Figure BDA0002409572100000201
3. Accuracy survey: and evaluating the accuracy of the method by adopting a standard recovery rate test. One sample of human serum was randomly selected, 1 of which was not added with the standard, and the other 2 were added with the standard at the low and high concentrations, respectively, and the same procedure was repeated and measured 5 times, and the recovery results were calculated, as shown in table 10. The result shows that the result of the standard recovery rate of the 12 kinds of steroid hormones in the serum is between 85% and 115%, and the results all meet the requirement.
TABLE 10 results of recovery of 12-steroid hormones in serum normalized to ng/m L unit
Figure BDA0002409572100000202
Figure BDA0002409572100000211
4. And (3) precision test: the normal human serum samples were taken and treated for 6 batches within one day and 3 days, the concentration of 12 steroid hormones was quantitatively determined by an isotope internal standard method, the internal and inter-batch precision was counted for three consecutive days, and the calculation results are shown in Table 11.
TABLE 11 results of precision test within and between batches (unit ng/m L)
Figure BDA0002409572100000212
Figure BDA0002409572100000221
Figure BDA0002409572100000231
Fifth, discuss
The method adopts a UP L C-MS/MS method to measure progesterone (P4), 17 α -hydroxyprogesterone (17 α -OHP4), Corticosterone (CORT), Dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), testosterone (T), Dihydrotestosterone (DHT), Androstenedione (AD), estrone (E1), estradiol (E2), aldosterone (A L D) and cortisol (F) in human serum, so 12 steroid hormones are measured.
The invention considers the batch-to-batch precision and the standard recovery rate of 12 steroid hormones in the serum, the result is between 85% and 115%, the requirement is met, meanwhile, the requirement on the sensitivity is very high because the content of the hormones in the human body is very low, the lowest limit of quantitation of the method can reach 10pg/m L, and the requirement of clinical detection is basically met.
The method has the advantages of high sensitivity, strong specificity, low cost and simple pretreatment process, can finish the separation and detection of 12 hormones within 5.0min, and provides a reliable detection method for the clinical health assessment of endocrine diseases.
The above examples are only intended to illustrate the technical solution of the present invention, but not to limit it; although the present invention has been described in detail with reference to the foregoing embodiments, it will be understood by those of ordinary skill in the art that: modifications of the technical solutions described in the foregoing embodiments are still possible, or some technical features may be equivalently replaced; and the modifications or the substitutions do not make the essence of the corresponding technical solutions depart from the scope of the technical solutions of the embodiments of the present invention.

Claims (10)

1. A kit for detecting 12 kinds of steroid hormones in serum by an ultra-high performance liquid chromatography tandem mass spectrometry technology,
the 12 steroid hormones are progesterone, 17 α -hydroxyprogesterone, corticosterone, dehydroepiandrosterone sulfate, testosterone, dihydrotestosterone, androstenedione, estrone, estradiol, aldosterone and cortisol respectively;
the kit comprises the following reagents:
(1) eluent:
eluent A: an aqueous solution containing 50 to 400. mu.M ammonium fluoride; eluent B: methanol;
(2) calibration solution:
a mixed standard S0 solution containing 200ng/m L progesterone, 200ng/m L17 α -hydroxyprogesterone, 400ng/m L0 corticosterone, 400ng/m L1 dehydroepiandrosterone, 200000ng/m L2 dehydroepiandrosterone sulfate, 200ng/m L testosterone, 100ng/m L dihydrotestosterone, 200ng/m L androstenedione, 40ng/m L estrone, 40ng/m L estradiol, 40ng/m L aldosterone and 4000ng/m L cortisol is prepared into calibrator solutions with eight different concentration points by using a blank serum matrix solution, wherein the eight concentration points of the calibrator solutions are as follows:
the concentrations of the progesterone, the 17 α -hydroxyprogesterone, the testosterone and the androstenedione are the same, and the eight concentrations are 0.05ng/ml, 0.1ng/ml, 0.2ng/ml, 0.5ng/ml, 1ng/ml, 2.5ng/ml, 5ng/ml and 10ng/ml in sequence;
the concentrations of corticosterone and dehydroepiandrosterone are the same, and the eight concentrations are as follows: 0.1ng/ml, 0.2ng/ml, 0.4ng/ml, 1ng/ml, 2ng/ml, 5ng/ml, 10ng/ml and 20 ng/ml;
the eight concentrations of dehydroepiandrosterone sulfate are as follows in sequence: 50ng/ml, 100ng/ml, 200ng/ml, 500ng/ml, 1000ng/ml, 2500ng/ml, 5000ng/ml and 10000 ng/ml;
the eight concentrations of dihydrotestosterone are as follows: 0.025ng/ml, 0.05ng/ml, 0.1ng/ml, 0.25ng/ml, 0.5ng/ml, 1.25ng/ml, 2.5ng/ml and 5 ng/ml;
the eight concentrations of estrone, estradiol and aldosterone are in sequence: 0.01ng/ml, 0.02ng/ml, 0.04ng/ml, 0.1ng/ml, 0.2ng/ml, 0.5ng/ml, 1ng/ml and 2 ng/ml;
the eight concentrations of cortisol are as follows: 1ng/ml, 2ng/ml, 4ng/ml, 10ng/ml, 20ng/ml, 50ng/ml, 100ng/ml and 200 ng/ml;
(3) mixing internal standard working solution:
contains 10ng/m L progesterone-d 9, 5ng/m L17 α -hydroxyprogesterone-d 8, 20ng/m L0 corticosterone-d 8, 20ng/m L1 dehydroepiandrosterone-d 2, 10000ng/m L2 dehydroepiandrosterone sulfate-d 6, 10ng/m L testosterone-d 3, 5ng/m L dihydrotestosterone-d 3, 10ng/m L androstenedione-13C 3, 1ng/m L estrone-d 4, 2ng/m L estradiol-d 4, 4ng/m L aldosterone-d 4 and 100ng/m L cortisol-d 4;
(4) extracting liquid: methyl tert-butyl ether;
(5) compounding the solution: 40-60% methanol water solution;
(6) quality control product:
blank serum matrix solution containing 12 steroid hormones at three concentrations, low, medium and high, QC (L), QC (m) and QC (h), respectively, wherein:
QC (L) was the mixed standard S0 solution diluted 2000-fold with blank serum matrix solution;
qc (m) 400-fold dilution of the mixed standard S0 solution with blank serum matrix solution;
qc (h) was 50-fold dilution of the mixed standard S0 solution with a blank serum matrix solution.
2. The kit according to claim 1,
the eluent A is an aqueous solution containing 50-100 mu M of ammonium fluoride; preferably 50. mu.M ammonium fluoride in water.
3. The kit according to claim 1,
the blank serum matrix is 5-20% methanol water solution; preferably 5 to 15 percent of methanol aqueous solution; more preferably 10% aqueous methanol.
4. The kit according to claim 1,
the complex solution is a 45-55% methanol water solution; preferably 50% aqueous methanol.
5. The kit according to claim 1, 2, 3 or 4,
the mixing standard S0 solution was prepared as follows:
preparing progesterone, 17 α -hydroxyprogesterone, corticosterone, dehydroepiandrosterone sulfate, testosterone, dihydrotestosterone, androstenedione, estrone, estradiol, aldosterone and cortisol into standard mother liquor with the concentration of 2mg/m L, 2mg/m L0, 4mg/m L1, 1mg/m L2, 10mg/m L, 2mg/m L, 1mg/m L, 4mg/m L, 2mg/m L, 1mg/m L, 1mg/m L and 4mg/m L in sequence by using pure methanol;
the mother liquor of each standard substance is prepared into mixed standard S0 solution containing 200ng/m L progesterone, 200ng/m L17 α -hydroxyprogesterone, 400ng/m L0 corticosterone, 400ng/m L1 dehydroepiandrosterone, 200000ng/m L2 dehydroepiandrosterone sulfate, 200ng/m L testosterone, 100ng/m L dihydrotestosterone, 200ng/m L androstenedione, 40ng/m L estrone, 40ng/m L estradiol, 40ng/m L aldosterone and 4000ng/m L cortisol by using methanol aqueous solution.
6. The kit according to claim 5,
the mixed internal standard working solution is prepared according to the following method:
weighing isotope internal standard substances including progesterone-d 9, 17 α -hydroxyprogesterone-d 8, corticosterone-d 8, dehydroepiandrosterone-d 2, dehydroepiandrosterone-d 6 sulfate, testosterone-d 3, dihydrotestosterone-d 3, androstenedione-13C 3, estrone-d 4, estradiol-d 4, aldosterone-d 4 and cortisol-d 4, respectively adding pure methanol to completely dissolve, and preparing into isotope internal standard mother liquor with the concentration of 1mg/m L, 1mg/m L0, 5mg/m L1, 2mg/m L2, 1mg/m L, 0.1mg/m L, 1mg/m L, 1mg/m L, 2.5mg/m L, 1mg/m L, 1mg/m L and 1mg/m L;
preparing isotope internal standard mother liquor containing 100ng/m L progesterone-d 9, 50ng/m L17 α -hydroxyprogesterone-d 8, 200ng/m L0 corticosterone-d 8, 200ng/m L1 dehydroepiandrosterone-d 2, 100000ng/m L2 dehydroepiandrosterone sulfate-d 6, 100ng/m L testosterone-d 3, 50ng/m L dihydrotestosterone-d 3, 100ng/m L androstenedione-13C 3, 10ng/m L estrone-d 4, 20ng/m L estradiol-d 4, 40ng/m L aldosterone-d 4 and 1000ng/m L cortisol-d 4 by using methanol aqueous solution;
and adding a methanol aqueous solution of 900 mu L into a SI solution of 100 mu L, and uniformly mixing to obtain a mixed internal standard working solution.
7. The kit according to claim 6,
when preparing a mixed standard S0 solution and a mixed internal standard working solution, the methanol aqueous solution is a 30-70% methanol aqueous solution, and preferably a 45-55% methanol aqueous solution.
8. The kit according to claim 7,
when preparing the mixed standard S0 solution and the mixed internal standard working solution, the methanol aqueous solution is 50% methanol aqueous solution.
9. The kit according to claim 8,
(1) eluent:
eluent A: containing 50 μ M ammonium fluoride aqueous solution; eluent B: methanol;
(2) calibration solution:
preparing progesterone, 17 α -hydroxyprogesterone, corticosterone, dehydroepiandrosterone sulfate, testosterone, dihydrotestosterone, androstenedione, estrone, estradiol, aldosterone and cortisol into standard mother liquor with the concentration of 2mg/m L, 2mg/m L0, 4mg/m L1, 1mg/m L2, 10mg/m L, 2mg/m L, 1mg/m L, 4mg/m L, 2mg/m L, 1mg/m L, 1mg/m L and 4mg/m L in sequence by using pure methanol;
preparing the mother liquor of each standard product into mixed standard S0 solution containing 200ng/m L progesterone, 200ng/m L17 α -hydroxyprogesterone, 400ng/m L0 corticosterone, 400ng/m L1 dehydroepiandrosterone, 200000ng/m L2 sulfuric acid dehydroepiandrosterone, 200ng/m L testosterone, 100ng/m L dihydrotestosterone, 200ng/m L androstenedione, 40ng/m L estrone, 40ng/m L estradiol, 40ng/m L aldosterone and 4000ng/m L cortisol by using 50% methanol aqueous solution;
preparing the mixed standard S0 solution into calibration substance solutions with eight different concentration points by using a 10% methanol solution;
(3) mixing internal standard working solution:
weighing isotope internal standard substances including progesterone-d 9, 17 α -hydroxyprogesterone-d 8, corticosterone-d 8, dehydroepiandrosterone-d 2, dehydroepiandrosterone-d 6 sulfate, testosterone-d 3, dihydrotestosterone-d 3, androstenedione-13C 3, estrone-d 4, estradiol-d 4, aldosterone-d 4 and cortisol-d 4, respectively adding pure methanol to completely dissolve, and preparing into isotope internal standard mother liquor with the concentration of 1mg/m L, 1mg/m L0, 5mg/m L1, 2mg/m L2, 1mg/m L, 0.1mg/m L, 1mg/m L, 1mg/m L, 2.5mg/m L, 1mg/m L, 1mg/m L and 1mg/m L;
the isotope internal standard mother liquor is prepared into an isotope internal standard SI solution containing 100ng/m L progesterone-d 9, 50ng/m L17 α -hydroxyprogesterone-d 8, 200ng/m L0 corticosterone-d 8, 200ng/m L1 dehydroepiandrosterone-d 2, 100000ng/m L2 dehydroepiandrosterone sulfate-d 6, 100ng/m L testosterone-d 3, 50ng/m L dihydrotestosterone-d 3, 100ng/m L androstenedione-13C 3, 10ng/m L estrone-d 4, 20ng/m L estradiol-d 4, 40ng/m L aldosterone-d 4 and 1000ng/m L cortisol-d 4 by 50% methanol aqueous solution;
adding 100 mu L SI solution into 900 mu L50% methanol water solution, and uniformly mixing to obtain mixed internal standard working solution;
(4) extracting liquid: methyl tert-butyl ether;
(5) compounding the solution: 50% aqueous methanol;
(6) quality control product:
10% aqueous methanol solution containing 12 steroid hormones at three concentrations, low, medium and high, QC (L), QC (M) and QC (H), respectively, wherein:
QC (L) contains 0.1ng/m L progesterone, 0.1ng/m L017 α -hydroxyprogesterone, 0.2ng/m L1 corticosterone, 0.2ng/m L2 dehydroepiandrosterone, 100ng/m L3 dehydroepiandrosterone sulfate, 0.1ng/m L testosterone, 0.05ng/m L dihydrotestosterone, 0.1ng/m L androstenedione, 0.02ng/m L estrone, 0.02ng/m L estradiol, 0.02ng/m L aldosterone and 2ng/m L cortisol;
QC (M) comprises 0.5ng/m L progesterone, 0.5ng/m L17 α -hydroxyprogesterone, 1ng/m L0 corticosterone, 1ng/m L1 dehydroepiandrosterone, 500ng/m L2 dehydroepiandrosterone sulfate, 0.5ng/m L testosterone, 0.25ng/m L dihydrotestosterone, 0.5ng/m L androstenedione, 0.1ng/m L estrone, 0.1ng/m L estradiol, 0.1ng/m L aldosterone and 10ng/m L cortisol;
QC (H) comprises 5ng/m L progesterone, 5ng/m L17 α -hydroxyprogesterone, 10ng/m L0 corticosterone, 10ng/m L1 dehydroepiandrosterone, 5000ng/m L2 dehydroepiandrosterone sulfate, 5ng/m L testosterone, 2.5ng/m L dihydrotestosterone, 5ng/m L androstenedione, 1ng/m L estrone, 1ng/m L estradiol, 1ng/m L aldosterone and 100ng/m L cortisol.
10. Use of the kit of any one of claims 1 to 9 for detecting 12 steroid hormones in serum by ultra performance liquid chromatography tandem mass spectrometry.
CN202010172191.5A 2020-03-12 2020-03-12 Kit for detecting 12 steroid hormone in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology Active CN111398447B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010172191.5A CN111398447B (en) 2020-03-12 2020-03-12 Kit for detecting 12 steroid hormone in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010172191.5A CN111398447B (en) 2020-03-12 2020-03-12 Kit for detecting 12 steroid hormone in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology

Publications (2)

Publication Number Publication Date
CN111398447A true CN111398447A (en) 2020-07-10
CN111398447B CN111398447B (en) 2023-05-05

Family

ID=71434197

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010172191.5A Active CN111398447B (en) 2020-03-12 2020-03-12 Kit for detecting 12 steroid hormone in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology

Country Status (1)

Country Link
CN (1) CN111398447B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112345660A (en) * 2020-09-29 2021-02-09 杭州凯莱谱精准医疗检测技术有限公司 Kit and method for determining activity of human plasma sample renin
CN113009006A (en) * 2021-01-20 2021-06-22 泊迈生物医学检测(苏州)有限公司 Method and kit for detecting dehydroepiandrosterone content in saliva
CN114088859A (en) * 2022-01-19 2022-02-25 北京金域医学检验实验室有限公司 Method for separating multi-component isomer and detecting 29 steroid hormones
CN114609297A (en) * 2022-03-30 2022-06-10 武汉迈特维尔生物科技有限公司 Method and kit for simultaneously detecting 5 kinds of steroid hormones and application thereof
CN114674961A (en) * 2022-04-13 2022-06-28 广东省中医院(广州中医药大学第二附属医院、广州中医药大学第二临床医学院、广东省中医药科学院) Kit for synchronously detecting 17 steroid hormones in serum without derivatization and application thereof
CN114689771A (en) * 2022-03-16 2022-07-01 合肥歆智医疗器械有限公司 Method and kit for simultaneously determining contents of three free androgens in serum
CN114705787A (en) * 2022-04-28 2022-07-05 天津国科医工科技发展有限公司 Method for detecting 12 steroid hormones in dry blood spots based on derivatization
CN115343398A (en) * 2022-09-20 2022-11-15 吉林大学第一医院 Kit and method for simultaneously measuring multiple steroid hormones in serum
CN116519850A (en) * 2023-06-29 2023-08-01 四川大学华西医院 Method for rapidly detecting 16 hormone concentrations in serum sample

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009124570A1 (en) * 2008-04-09 2009-10-15 Ruebben Alexander Method for producing a bioactive surface on an endoprosthesis or on the balloon of a balloon catheter
CN107064400A (en) * 2017-04-20 2017-08-18 博厚健康科技股份有限公司 The method for detecting five steroids hormones in serum simultaneously
CN110187043A (en) * 2019-04-25 2019-08-30 中南民族大学 Method that is a kind of while detecting 13 kinds of steroid hormones in serum

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009124570A1 (en) * 2008-04-09 2009-10-15 Ruebben Alexander Method for producing a bioactive surface on an endoprosthesis or on the balloon of a balloon catheter
CN107064400A (en) * 2017-04-20 2017-08-18 博厚健康科技股份有限公司 The method for detecting five steroids hormones in serum simultaneously
CN110187043A (en) * 2019-04-25 2019-08-30 中南民族大学 Method that is a kind of while detecting 13 kinds of steroid hormones in serum

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LIMIAN ZHAO: "Quantitative Determination of a Panel of Endogenous Steroids in Human Serum by LC/MS/MS", 《APPLICATION NOTE 5994-0949EN》 *
米晓霞等: "猪血浆中33种类固醇激素的高效液相色谱-串联质谱测定", 《分析测试学报》 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112345660A (en) * 2020-09-29 2021-02-09 杭州凯莱谱精准医疗检测技术有限公司 Kit and method for determining activity of human plasma sample renin
CN113009006A (en) * 2021-01-20 2021-06-22 泊迈生物医学检测(苏州)有限公司 Method and kit for detecting dehydroepiandrosterone content in saliva
CN114088859A (en) * 2022-01-19 2022-02-25 北京金域医学检验实验室有限公司 Method for separating multi-component isomer and detecting 29 steroid hormones
CN114689771A (en) * 2022-03-16 2022-07-01 合肥歆智医疗器械有限公司 Method and kit for simultaneously determining contents of three free androgens in serum
CN114609297A (en) * 2022-03-30 2022-06-10 武汉迈特维尔生物科技有限公司 Method and kit for simultaneously detecting 5 kinds of steroid hormones and application thereof
CN114674961A (en) * 2022-04-13 2022-06-28 广东省中医院(广州中医药大学第二附属医院、广州中医药大学第二临床医学院、广东省中医药科学院) Kit for synchronously detecting 17 steroid hormones in serum without derivatization and application thereof
CN114705787A (en) * 2022-04-28 2022-07-05 天津国科医工科技发展有限公司 Method for detecting 12 steroid hormones in dry blood spots based on derivatization
CN115343398A (en) * 2022-09-20 2022-11-15 吉林大学第一医院 Kit and method for simultaneously measuring multiple steroid hormones in serum
CN116519850A (en) * 2023-06-29 2023-08-01 四川大学华西医院 Method for rapidly detecting 16 hormone concentrations in serum sample

Also Published As

Publication number Publication date
CN111398447B (en) 2023-05-05

Similar Documents

Publication Publication Date Title
CN111398446B (en) Method for detecting 12 steroid hormones in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology
CN111398447A (en) Kit for detecting 12 kinds of steroid hormones in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology
CN111366671B (en) Chemical derivatization-ultra-high performance liquid chromatography-tandem mass spectrometry for simultaneously detecting 18 steroid hormones in serum
Keevil LC–MS/MS analysis of steroids in the clinical laboratory
Gosetti et al. Ultra high performance liquid chromatography tandem mass spectrometry determination and profiling of prohibited steroids in human biological matrices. A review
Christeff et al. Elevated oestrogen and reduced testosterone levels in the serum of male septic shock patients
CN104807920B (en) A kind of test kit is using the application in 10 steroids hormones in high performance liquid chromatography tandem mass spectrum technology for detection serum
Matysik et al. Determination of steroid hormones in human plasma by GC–triple quadrupole MS
CN110702831B (en) Kit for detecting serum testosterone hormone by ultra-high performance liquid chromatography-tandem mass spectrometry
du Toit et al. A high-throughput UPC2-MS/MS method for the separation and quantification of C19 and C21 steroids and their C11-oxy steroid metabolites in the classical, alternative, backdoor and 11OHA4 steroid pathways
CN111487328A (en) Method for detecting multiple trace hormones in human body by mass spectrometry
Laszlo et al. A high resolution LC–MS targeted method for the concomitant analysis of 11 contraceptive progestins and 4 steroids
CN110824091A (en) Steroid hormone detection method
Tsakalof et al. Development of GC–MS/MS method with programmable temperature vaporization large volume injection for monitoring of 17β-estradiol and 2-methoxyestradiol in plasma
Qin et al. Parallel derivatization strategy coupled with liquid chromatography-mass spectrometry for broad coverage of steroid hormones
Higashi et al. Methods for determination of fingernail steroids by LC/MS/MS and differences in their contents between right and left hands
CN110208393A (en) A kind of method of 5 kinds of androgens in detection serum
US11536733B2 (en) Methods and systems for the detection of 11-oxo androgens by LC-MS/MS
CN112986433A (en) Method for detecting steroid content in human serum sample
Bai et al. A Method for simultaneous determination of 14 carbonyl-steroid hormones in human serum by ultra high performance liquid chromatography–tandem mass spectrometry
CN113009036A (en) Kit for detecting sex hormone, sex hormone sample pretreatment method and method for simultaneously detecting multiple sex hormones
JP3774888B2 (en) Highly sensitive detection method for steroid compounds by LC-MS
KR101068993B1 (en) Evaluation of metabolic differences between urine samples by quantitative steroid signature
Alvarez-Sanchez et al. Ultrasound-enhanced enzymatic hydrolysis of conjugated female steroids as pretreatment for their analysis by LC–MS/MS in urine
CN115856171A (en) Method for simultaneously detecting multiple fat-soluble vitamins and multiple steroid hormones

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
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20230413

Address after: 210000 east of floor 5, building 3, intelligent manufacturing Industrial Park (Zhicheng Park), No. 6, Zhida Road, Jiangbei new area, Nanjing, Jiangsu Province

Applicant after: Nanjing Pinsheng Medical Technology Co.,Ltd.

Applicant after: Nanjing Pinsheng medical laboratory Co.,Ltd.

Applicant after: Fuzhou Pinsheng Medical Technology Co.,Ltd.

Applicant after: Fujian Pinsheng Wanfu Medical Testing Co.,Ltd.

Address before: 210000 floor 10, building 5, Yangzi science and technology innovation headquarters base, No. 7, Pudong North Road, Jiangbei new area, Nanjing, Jiangsu Province

Applicant before: Nanjing Pinsheng medical laboratory Co.,Ltd.

GR01 Patent grant
GR01 Patent grant