CN113140266A - Screening method of xanthine oxidase inhibitor for reducing uric acid - Google Patents

Screening method of xanthine oxidase inhibitor for reducing uric acid Download PDF

Info

Publication number
CN113140266A
CN113140266A CN202110548996.XA CN202110548996A CN113140266A CN 113140266 A CN113140266 A CN 113140266A CN 202110548996 A CN202110548996 A CN 202110548996A CN 113140266 A CN113140266 A CN 113140266A
Authority
CN
China
Prior art keywords
xanthine oxidase
model
uric acid
oxidase inhibitor
reducing uric
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
CN202110548996.XA
Other languages
Chinese (zh)
Inventor
张英华
周倩
王小慧
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.)
Northeast Agricultural University
Original Assignee
Northeast Agricultural University
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 Northeast Agricultural University filed Critical Northeast Agricultural University
Priority to CN202110548996.XA priority Critical patent/CN113140266A/en
Publication of CN113140266A publication Critical patent/CN113140266A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/60In silico combinatorial chemistry
    • G16C20/64Screening of libraries
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06NCOMPUTING ARRANGEMENTS BASED ON SPECIFIC COMPUTATIONAL MODELS
    • G06N20/00Machine learning
    • G06N20/10Machine learning using kernel methods, e.g. support vector machines [SVM]
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06NCOMPUTING ARRANGEMENTS BASED ON SPECIFIC COMPUTATIONAL MODELS
    • G06N20/00Machine learning
    • G06N20/20Ensemble learning
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/70Machine learning, data mining or chemometrics
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/90Programming languages; Computing architectures; Database systems; Data warehousing
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients

Landscapes

  • Engineering & Computer Science (AREA)
  • Theoretical Computer Science (AREA)
  • Computing Systems (AREA)
  • Software Systems (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medical Informatics (AREA)
  • Health & Medical Sciences (AREA)
  • Evolutionary Computation (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Artificial Intelligence (AREA)
  • Computer Vision & Pattern Recognition (AREA)
  • Data Mining & Analysis (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Databases & Information Systems (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Physics & Mathematics (AREA)
  • Mathematical Physics (AREA)
  • Epidemiology (AREA)
  • Primary Health Care (AREA)
  • Public Health (AREA)
  • Library & Information Science (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention belongs to the technical field of functional component separation, and particularly relates to a xanthine oxidase inhibitor for reducing uric acid.

Description

Screening method of xanthine oxidase inhibitor for reducing uric acid
Technical Field
The invention belongs to the technical field of functional component separation, and particularly relates to a screening method of a xanthine oxidase inhibitor for reducing uric acid.
Background
Gout is an inflammatory disease caused by increased purine metabolism or abnormal uric acid excretion, and causes kidney diseases, cardiovascular diseases, metabolic syndrome, and the like. In recent decades, the incidence of gout has been increasing and is in a trend of youthfulness, both increased uric acid synthesis and decreased uric acid excretion can cause the increase of uric acid level in blood, thereby inducing gout, the uric acid in human bodies is derived from food intake at least in part, about 80% of uric acid is a product of purine metabolism catalyzed by xanthine oxidase in human bodies, the xanthine oxidase is a key restriction enzyme for producing uric acid in human bodies, and is also a drug action target point for treating gout, the activity of the xanthine oxidase is inhibited, the conversion of xanthine to uric acid is reduced, and the method becomes a main way for clinically treating gout.
Xanthine oxidase inhibitors have become the main drugs for treating gout, which are mainly classified into purine and non-purine analogues, however, some xanthine oxidase inhibitors such as allopurinol and febuxostat are accompanied by adverse reactions in gout treatment, including fever, rash, renal function deterioration and Stevens-Johnson syndrome, and some natural compounds from plant sources can effectively inhibit xanthine oxidase and can be used for gout treatment, so that the search for the xanthine oxidase inhibitors with high activity and small toxic and side effects from natural plants is particularly urgent and important.
The invention screens potential xanthine oxidase inhibitor candidates by combining a quantitative structure-activity relationship model and molecular docking through establishing a xanthine oxidase inhibitor molecular database, and obtains the xanthine oxidase inhibitor for reducing uric acid through in vitro experiment verification.
Disclosure of Invention
The invention aims to establish a xanthine oxidase inhibitor molecular database, combine a quantitative structure-activity relationship model with molecular docking to screen potential xanthine oxidase inhibitor candidates, and obtain the xanthine oxidase inhibitor for reducing uric acid through in vitro experimental verification. The product and the preparation method of the invention are as follows:
a screening method of a xanthine oxidase inhibitor for reducing uric acid is characterized by comprising the following steps of: (1) establishing a database of xanthine oxidase inhibitors of plant origin, the database comprising 150 compounds that inhibit xanthine oxidase; (2) calculating smile of the molecule in the step (1), generating a molecular descriptor for capturing molecular structure and property, and comparing; (3) the four machine learning methods comprise random forests, support vector machines, k nearest neighbor and linear discriminant analysis, a quantitative structure-activity relationship model capable of screening xanthine oxidase inhibitors is established according to the descriptors generated in the step (2), and an optimal model is selected by comparing the accuracy of the model; (4) taking allopurinol as a positive control, obtaining a crystal structure of xanthine oxidase, taking the crystal structure as a protein target, performing molecular pairing to verify a model prediction result, and further determining the xanthine oxidase inhibitory activity of the compound by using an ultraviolet spectrophotometry; (5) the potential xanthine oxidase inhibitor is discovered through model screening and experimental verification.
The MOE descriptor has the best modeling effect.
The optimal model is a random forest model with the accuracy of 0.9605.
The experimental conditions are that 50 mu L of sample solution, 50 mu L of xanthine oxidase solution (0.02U/mL) and phosphate buffer solution (0.075mol/L and pH 7.5) are sequentially added on a 96-well plate, incubation is carried out for 5min in a 25 ℃ microplate reader, then 50 mu L of xanthine stock solution (0.001mol/L) is added, reaction is carried out for 15min at 25 ℃, zero adjustment is carried out by a blank group at 295nm, and the absorbance A of the sample is measured0(ii) a Then, the negative control group is used for zero setting, and the absorbance A of the positive control group is measured1The XO suppression ratio (IR) is calculated by the following formula: IR (%) - (a)1-A0)/A1]×100%。
The characteristics of the contents of the final product of the invention are as follows: the candidate with xanthine oxidase inhibitory activity is vanillic acid, IC50It was 0.593. mu.g/mL.
Drawings
FIG. 1 is a process flow diagram of the present invention;
FIG. 2 is an importance diagram of MOE descriptors;
FIG. 3 is a graph of accuracy of the RF model;
FIG. 4 is a molecular docking diagram of vanillic acid and xanthine oxidase.
Detailed Description
Specific embodiments are further described below in conjunction with the appended drawings.
A screening method of a xanthine oxidase inhibitor for reducing uric acid is characterized by comprising the following steps of: (1) establishing a database of xanthine oxidase inhibitors of plant origin, the database comprising 150 compounds that inhibit xanthine oxidase; (2) calculating smile of the molecule in the step (1), generating a molecular descriptor for capturing molecular structure and property, and comparing; (3) the four machine learning methods comprise random forests, support vector machines, k nearest neighbor and linear discriminant analysis, a quantitative structure-activity relationship model capable of screening xanthine oxidase inhibitors is established according to the descriptors generated in the step (2), and an optimal model is selected by comparing the accuracy of the model; (4) taking allopurinol as a positive control, obtaining a crystal structure of xanthine oxidase, taking the crystal structure as a protein target, performing molecular pairing to verify a model prediction result, and further determining the xanthine oxidase inhibitory activity of the compound by using an ultraviolet spectrophotometry; (5) the potential xanthine oxidase inhibitor is discovered through model screening and experimental verification.
The MOE descriptor has the best modeling effect.
The optimal model is a random forest model with the accuracy of 0.9605.
The experimental conditions are that 50 mu L of sample solution, 50 mu L of xanthine oxidase solution (0.02U/mL) and phosphate buffer solution (0.075mol/L and pH 7.5) are sequentially added on a 96-well plate, incubation is carried out for 5min in a 25 ℃ microplate reader, then 50 mu L of xanthine stock solution (0.001mol/L) is added, reaction is carried out for 15min at 25 ℃, zero adjustment is carried out by a blank group at 295nm, and the absorbance A of the sample is measured0(ii) a Then, the negative control group is used for zero setting, and the absorbance A of the positive control group is measured1The XO suppression ratio (IR) is calculated by the following formula: IR (%) - (a)1-A0)/A1]×100%。
The candidate with xanthine oxidase inhibitory activity is vanillic acid, IC50It was 0.593. mu.g/mL.
Example 1
(1) A database of plant-derived xanthine oxidase inhibitors is established.
(2) Calculating the MOE descriptor of the compound collected in step (1).
(3) And (3) establishing comparison accuracy of the RF model, the SVM model, the kNN model and the LDA model according to the descriptor generated in the step (2).
(4) Taking allopurinol as a positive control, obtaining the crystal structure of xanthine oxidase, taking the crystal structure as a protein target, performing molecular pairing to verify the result of model prediction, and further determining the xanthine oxidase inhibitory activity of the compound by using an ultraviolet spectrophotometry.
(5) A candidate having xanthine oxidase inhibitory activity was found to be vanillic acid, IC50It was 0.593. mu.g/mL.
Example 2
(1) A database of plant-derived xanthine oxidase inhibitors is established.
(2) Calculating the Mordred descriptor of the compound collected in step (1).
(3) And (3) establishing comparison accuracy of the RF model, the SVM model, the kNN model and the LDA model according to the descriptor generated in the step (2).
(4) Taking allopurinol as a positive control, obtaining the crystal structure of xanthine oxidase, taking the crystal structure as a protein target, performing molecular pairing to verify the result of model prediction, and further determining the xanthine oxidase inhibitory activity of the compound by using an ultraviolet spectrophotometry.
(5) A candidate having xanthine oxidase inhibitory activity was found to be vanillic acid, IC50It was 0.593. mu.g/mL.
Example 3
(1) A database of plant-derived xanthine oxidase inhibitors is established.
(2) Calculating the ChemoPy descriptor of the compound collected in step (1).
(3) And (3) establishing comparison accuracy of the RF model, the SVM model, the kNN model and the LDA model according to the descriptor generated in the step (2).
(4) Taking allopurinol as a positive control, obtaining the crystal structure of xanthine oxidase, taking the crystal structure as a protein target, performing molecular pairing to verify the result of model prediction, and further determining the xanthine oxidase inhibitory activity of the compound by using an ultraviolet spectrophotometry.
(5) A candidate having xanthine oxidase inhibitory activity was found to be vanillic acid, IC50It was 0.593. mu.g/mL.

Claims (5)

1. A screening method of a xanthine oxidase inhibitor for reducing uric acid is characterized by comprising the following steps of: (1) establishing a database of xanthine oxidase inhibitors of plant origin, the database comprising 150 compounds that inhibit xanthine oxidase; (2) calculating smile of the molecule in the step (1), generating a molecular descriptor for capturing molecular structure and property, and comparing; (3) the four machine learning methods comprise random forests, support vector machines, k nearest neighbor and linear discriminant analysis, a quantitative structure-activity relationship model capable of screening xanthine oxidase inhibitors is established according to the descriptors generated in the step (2), and an optimal model is selected by comparing the accuracy of the model; (4) taking allopurinol as a positive control, obtaining a crystal structure of xanthine oxidase, taking the crystal structure as a protein target, performing molecular pairing to verify a model prediction result, and further determining the xanthine oxidase inhibitory activity of the compound by using an ultraviolet spectrophotometry; (5) the potential xanthine oxidase inhibitor is discovered through model screening and experimental verification.
2. The xanthine oxidase inhibitor for reducing uric acid according to claim 1, wherein the MOE descriptor is modeled best.
3. The xanthine oxidase inhibitor for reducing uric acid according to claim 1, wherein the optimal model is a random forest model with an accuracy of 0.9605.
4. The xanthine oxidase inhibitor for reducing uric acid according to claim 1, wherein the experimental conditions are that 50 μ L of sample solution, 50 μ L of xanthine oxidase solution (0.02U/mL), and phosphate buffer (0.075mol/L, pH 7.5) are sequentially added to a 96-well plate, incubated in a 25 ℃ microplate reader for 5min, and thenAdding 50 μ L xanthine stock solution (0.001mol/L) to react at 25 deg.C for 15min, zeroing with blank set at 295nm, and measuring absorbance A of sample0(ii) a Then, the negative control group is used for zero setting, and the absorbance A of the positive control group is measured1The XO suppression ratio (IR) is calculated by the following formula: IR (%) - (a)1-A0)/A1]×100%。
5. The xanthine oxidase inhibitor for reducing uric acid according to claim 1, wherein the candidate substance having xanthine oxidase inhibitory activity is vanillic acid, IC50It was 0.593. mu.g/mL.
CN202110548996.XA 2021-05-20 2021-05-20 Screening method of xanthine oxidase inhibitor for reducing uric acid Pending CN113140266A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110548996.XA CN113140266A (en) 2021-05-20 2021-05-20 Screening method of xanthine oxidase inhibitor for reducing uric acid

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110548996.XA CN113140266A (en) 2021-05-20 2021-05-20 Screening method of xanthine oxidase inhibitor for reducing uric acid

Publications (1)

Publication Number Publication Date
CN113140266A true CN113140266A (en) 2021-07-20

Family

ID=76817338

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110548996.XA Pending CN113140266A (en) 2021-05-20 2021-05-20 Screening method of xanthine oxidase inhibitor for reducing uric acid

Country Status (1)

Country Link
CN (1) CN113140266A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115232855A (en) * 2022-07-08 2022-10-25 华南农业大学 Method for screening drugs influencing xanthine oxidase activity by targeting intestinal flora

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102222178A (en) * 2011-03-31 2011-10-19 清华大学深圳研究生院 Method for screening and/or designing medicines aiming at multiple targets
CN104598772A (en) * 2014-12-25 2015-05-06 南昌大学 Construction method for gout drug effect enzyme target model
CN109524064A (en) * 2018-11-12 2019-03-26 云南省烟草农业科学研究院 A kind of virtual screening method of polyphenol oxidase enzyme inhibitor
CN111627493A (en) * 2020-05-29 2020-09-04 北京晶派科技有限公司 Selective prediction method and computing device for kinase inhibitor

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102222178A (en) * 2011-03-31 2011-10-19 清华大学深圳研究生院 Method for screening and/or designing medicines aiming at multiple targets
CN104598772A (en) * 2014-12-25 2015-05-06 南昌大学 Construction method for gout drug effect enzyme target model
CN109524064A (en) * 2018-11-12 2019-03-26 云南省烟草农业科学研究院 A kind of virtual screening method of polyphenol oxidase enzyme inhibitor
CN111627493A (en) * 2020-05-29 2020-09-04 北京晶派科技有限公司 Selective prediction method and computing device for kinase inhibitor

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115232855A (en) * 2022-07-08 2022-10-25 华南农业大学 Method for screening drugs influencing xanthine oxidase activity by targeting intestinal flora

Similar Documents

Publication Publication Date Title
Maffucci et al. An efficient implementation of the Nwat-MMGBSA method to rescore docking results in medium-throughput virtual screenings
Hamza et al. Novel human mPGES-1 inhibitors identified through structure-based virtual screening
Gershell et al. A brief history of novel drug discovery technologies
Caballero Considerations for docking of selective angiotensin-converting enzyme inhibitors
CN113140266A (en) Screening method of xanthine oxidase inhibitor for reducing uric acid
Kos et al. Synthesis and hybrid SAR property modeling of novel cholinesterase inhibitors
Palmioli et al. First experimental identification of Ras-inhibitor binding interface using a water-soluble Ras ligand
Akbar et al. Characterization of metabolic network of oxalic acid biosynthesis through RNA seq data analysis of developing spikes of finger millet (Eleusine coracana): deciphering the role of key genes involved in oxalate formation in relation to grain calcium accumulation
Pang et al. Comparative phosphoproteomic analysis reveals the response of starch metabolism to high-temperature stress in rice endosperm
CN114081882B (en) A-FABP protein inhibitor and application thereof
Zeng et al. Study on differential protein expression in natural selenium-enriched and non-selenium-enriched rice based on iTRAQ quantitative proteomics
Zhao et al. Novel peptides with xanthine oxidase inhibitory activity identified from macadamia nuts: Integrated in silico and in vitro analysis
Zhou et al. Lactate activates AMPK remodeling of the cellular metabolic profile and promotes the proliferation and differentiation of C2C12 myoblasts
EP4295325A1 (en) Multi-scale spatial transcriptomics analysis
Wang et al. Immunomodulatory protein from nectria haematococca induces apoptosis in lung cancer cells via the P53 pathway
Liao et al. A Novel LSTM-Based Machine Learning Model for Predicting the Activity of Food Protein-Derived Antihypertensive Peptides
Vázquez-Mendoza et al. In silico drug repositioning to target the SARS-CoV-2 main protease as covalent inhibitors employing a combined structure-based virtual screening strategy of pharmacophore models and covalent docking
Romero-Lopez et al. Lung metabolomics profiling of congenital diaphragmatic hernia in fetal rats
Wang et al. Screening and evaluation of flavonoids as xanthine oxidase inhibitors for reducing uric acid through combined cell biology and molecular simulation
Chang et al. The development and validation of a computational model to predict rat liver microsomal clearance
Tanaka et al. Low-Barrier Hydrogen Bond in Fujikurin A–D: A Computational Study
Caballero et al. Docking and quantitative structure–activity relationship studies for sulfonyl hydrazides as inhibitors of cytosolic human branched-chain amino acid aminotransferase
Wang et al. Transcriptome and miRNAs profiles reveal regulatory network and key regulators of secondary xylem formation in “84K” poplar
Wu et al. Slc38a9 deficiency induces apoptosis and metabolic dysregulation and leads to premature death in zebrafish
Abdizadeh et al. Molecular modeling studies of anti-Alzheimer agents by QSAR, molecular docking and molecular dynamics simulations techniques

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: 20210720