CN113244244A - Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis - Google Patents

Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis Download PDF

Info

Publication number
CN113244244A
CN113244244A CN202110666788.XA CN202110666788A CN113244244A CN 113244244 A CN113244244 A CN 113244244A CN 202110666788 A CN202110666788 A CN 202110666788A CN 113244244 A CN113244244 A CN 113244244A
Authority
CN
China
Prior art keywords
liver
fibrosis
hepatic fibrosis
hepatic
demethyleularmin
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
CN202110666788.XA
Other languages
Chinese (zh)
Inventor
王淑珍
陈珂
郭蔚然
李荣欣
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.)
China Pharmaceutical University
Original Assignee
China Pharmaceutical 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 China Pharmaceutical University filed Critical China Pharmaceutical University
Priority to CN202110666788.XA priority Critical patent/CN113244244A/en
Publication of CN113244244A publication Critical patent/CN113244244A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses application of demethylzeylaspartic acid (T-96) in preventing and treating hepatic fibrosis and in preparing a medicament for preventing or treating hepatic fibrosis, belonging to the technical field of biological medicines. In-vivo and in-vitro experiments prove that the demethyleularmin can inhibit the activation of hepatic stellate cells; reduce collagen deposition in liver of liver fibrosis mice, inhibit fibroplasia, improve liver function, and reduce the expression of liver fibrosis related protein, which shows that the demethyleularwood aldehyde has the function of anti-liver fibrosis. In addition, the transcriptome sequencing and experimental results show that the demethyleularmin can inhibit the activation of hepatic stellate cells by regulating the expression of proteins such as AGAP2 and the like so as to improve hepatic fibrosis, and the demethyleularmin has the advantages of small toxic and side effects, wide raw material source, low production cost and the like. Therefore, the demethyleularmaldehyde can be used for preparing medicines and pharmaceutical compositions for preventing/treating hepatic fibrosis and has good development and application values.

Description

Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis
Technical Field
The invention specifically relates to an application of demethyleularmin in preparation of a medicine for preventing or treating hepatic fibrosis, and belongs to the technical field of biological medicines.
Background
Hepatic fibrosis is a chronic liver injury caused by various causes of viral infection, autoimmunity, excessive alcohol intake, bile duct obstruction and the like. When hepatocytes are damaged, continued liver damage results in excessive deposition of extracellular matrix (ECM) with concomitant inflammatory and compensatory repair reactions that lead to liver fibrosis. Early hepatic fibrosis is a reversible process, and if timely and effective intervention treatment is not obtained, cirrhosis can develop, and is accompanied by a series of fatal complications, such as hepatic failure, portal hypertension, ascites, hepatic encephalopathy and the like, and finally liver cancer is caused. However, no ideal anti-hepatic fibrosis drug is found clinically at present, and hepatic fibrosis becomes a clinical problem which seriously threatens human health globally. Therefore, intensive research and drug development on the mechanism associated with early treatment of hepatic fibrosis to improve the quality of life of patients with hepatic fibrosis are imminent.
Hepatic Stellate Cells (HSCs) are non-parenchymal cells of the liver located in the space periantral space, accounting for 5-8% of the total number of hepatocytes. In the course of hepatic fibrosis, quiescent HSCs are activated by various cytokines to transdifferentiate into highly activated Myofibroblasts (MF), gradually lose the ability to store vitamin a, release a range of pro-inflammatory factors such as interleukin 6 (IL-6) and pro-fibrotic factors such as transforming growth factor beta (TGF- β), and secrete excessive ECM proteins and alpha smooth muscle actin (alpha-SMA), resulting in scar tissue formation. Preventing the activation of HSCs can reduce ECM protein deposition, slow down or even reverse the progression of liver fibrosis. Therefore, the activation of HSCs is the key to the development of liver fibrosis, and blocking the activation of this cell is the current major strategy for liver fibrosis treatment.
ADP-ribosylation factor (Arf) is a member of the Ras superfamily of small G proteins and belongs to GTP-binding proteins. AGAP2 belongs to a family member of GTPase activating protein (Arf GAP), and participates in regulating various signal transduction pathways of cells as a GTPase activating protein switch of Arfs. Thus, AGAP2 is a multifunctional protein that has GTPase activity and is involved in the signaling pathways regulating apoptosis, cell migration, receptor transport and recycling, and integrin activation. Increased expression of AGAP2 is closely associated with the development of cancer and fibrotic diseases. Recent studies have found that AGAP2 has a role in regulating the progression of liver fibrosis in the TGF-beta 1 signaling pathway. AGAP2 in HSC cell responds to TGF-beta 1 cytokine, promotes proliferation and migration of hepatic stellate cell, and improves expression of hepatic fibrosis related gene such as alpha-SMA, collagen, etc., and simultaneously reduces expression of anti-fibrosis gene MMP-1, MMP-9. Therefore, AGAP2 is expected to become a potential therapeutic target for resisting hepatic fibrosis, and brings a brand-new therapeutic strategy for hepatic fibrosis patients. However, no specific inhibitor aiming at AGAP2 is developed at present, and a medicament capable of targeting AGAP2 protein is found to have good application prospect.
The active ingredients of natural products have the characteristics of low toxicity, diverse activity, multi-target synergistic effect and the like, and become a main source for new drug discovery. The comprehensive domestic and foreign experimental research finds that: the natural medicine shows better curative effect and lower toxic and side effect in the aspect of treating hepatic fibrosis, so that the development of a new anti-fibrosis medicine from the natural medicine has great significance. The demethyleulaldehyde is an effective monomer compound extracted from radix Tripterygii Wilfordii, and has wide pharmacological activities of resisting inflammation, suppressing immunity, resisting fertility, resisting tumor, etc. For example, noreulaldehyde can inhibit the growth of glioma cells by down-regulating expression of MYBL2, causing the cell cycle to stop at G1/S phase, impeding DNA replication; in addition, the demethyleulaldehyde has kidney protection effect on MRL/lpr lupus nephritis mice, can inhibit NF-kB activity, and reduce the level of downstream proinflammatory factors so as to inhibit macrophage infiltration and relieve kidney injury. For example, the Chinese patent "application of demethyleularmin in preparing a medicament for treating pancreatic cancer" (publication No. CN106727603A) discloses that demethyleularmin plays a role in resisting pancreatic cancer by inducing autophagic death and Caspase-3 dependent apoptosis; and has better effect of inhibiting pancreatic cancer cells when being combined with gemcitabine. For example, Chinese patent "potential application of demethyleularmin in resisting mycobacterium tuberculosis infection" (publication No. CN112294825A) discloses that demethyleularmin has the function of inhibiting 3-dehydroquinate dehydratase in mycobacterium tuberculosis and can be used as a potential drug for resisting mycobacterium tuberculosis infection. However, to date, there is no disclosure of noreulaldehyde for treating or preventing liver fibrosis, nor that it effectively inhibits AGAP2 to exert pharmacological activity against liver fibrosis.
Disclosure of Invention
The purpose of the invention is as follows: aiming at the defects in the prior art, the invention provides a new application of the demethyleularmin, namely an application of the demethyleularmin in preparing a medicament for preventing or treating hepatic fibrosis.
The invention also provides application of the demethyleulaldehyde in inhibiting hepatic stellate cell activation and preparing a medicament for inhibiting AGAP2 expression.
The invention constructs a mouse hepatic fibrosis model by using carbon tetrachloride, and perfuses different doses of the demethylzelarwood aldehyde to further analyze the treatment effect of the demethylzelarwood aldehyde on hepatic fibrosis. The results show that the noreulaldehyde can obviously reduce the collagen deposition in the liver, inhibit fibroplasia, improve the liver function of a liver fibrosis mouse and reduce the expression of liver fibrosis related genes, thereby achieving the purpose of treating liver fibrosis.
The hepatic fibrosis diseases comprise: alcoholic liver disease, non-alcoholic fatty liver disease, chronic viral hepatitis, autoimmune liver disease, and hepatic blood stasis.
The invention also discloses a molecular mechanism of treating hepatic fibrosis by the demethyleulaldehyde, namely the demethyleulaldehyde is proved to play a role in resisting hepatic fibrosis by effectively inhibiting the expression of the AGAP2 gene by combining transcriptome sequencing analysis and in vitro and in vivo experiments.
The demethyleularmaldehyde is medicinal salt or crystal form, preparation and/or a pharmaceutical composition consisting of a drug delivery carrier.
The medicinal salt forms of the demethyleularmaldehyde comprise various acid salts formed by inorganic acid and organic acid or various alkaline salts formed by inorganic base and organic base and various crystal forms.
The invention claims and protects a pharmaceutical composition for resisting hepatic fibrosis, which is characterized in that the pharmaceutical composition takes demethyleularmaldehyde as a main active ingredient and contains one or more pharmaceutically acceptable carriers or auxiliary materials. Wherein the drug carrier comprises microcapsules, microspheres, nanoparticles and liposomes. The auxiliary materials comprise conventional diluents, cosolvent, emulsifier, excipient, filler, adhesive, colorant, wetting agent, disintegrant, absorption enhancer, surfactant, lubricant, synergist, preservative and the like in the pharmaceutical field. The carrier or adjuvant for sustained and controlled release comprises hydrogel (such as cellulose derivatives, non-cellulose polysaccharide, natural gum, and synthetic polymer), biodegradable polymer (such as polylactic acid, polyglycolic acid, and polyacrylic acids), and ion exchange resin (microporous ion exchange resin, macroporous ion exchange resin, homogeneous porous ion exchange resin, and macroporous network adsorption resin).
The medicine can be made into injection, oral liquid, tablet, pill, capsule, suspension, suppository, lyophilized powder for injection, controlled release preparation, sustained release preparation, etc., and can be administered by appropriate route according to disease condition and the part to be treated. The administration route can be divided into oral administration, oral inhalation, cutaneous administration, rectal administration, subcutaneous, intravenous or intramuscular injection, etc.
The dose of the pharmaceutical composition of the present invention as an active ingredient to be administered depends on various factors such as the nature and severity of the disease to be prevented or treated, the age, sex, body weight, race, administration time, administration frequency, administration route, individual reaction and the like of the patient or animal, and thus the therapeutic dose of the present invention may be varied widely. Appropriate adjustments may be made to the dosage depending on the condition of the patient being treated, and the physician will determine the appropriate dosage for the individual patient.
Advantageous effects
The invention discovers that the demethyleulasialdehyde has the effect of preventing or treating hepatic fibrosis for the first time, and the method specifically comprises the following steps: the pharmacological activity of the demethyl eulaldehyde is examined in the TGF-beta 1 induced and activated LX-2 cells by adopting an RT-qPCR and Western Blot experiment method. The result shows that the noreulaldehyde can effectively inhibit the activation of LX-2 cells, namely the expression of liver fibrosis related genes COL1A1 and alpha-SMA, and has dose dependence. Then by applying to CCl4Inducing a hepatic fibrosis mouse model to carry out drug treatment, and exploring the hepatic fibrosis resisting activity of the demethyleularwood aldehyde. Pathology detection finds that compared with a model group, the noreulaldehyde can obviously reduce the liver collagen deposition of liver fibrosis mice and improve liver injury. The determination of the contents of blood biochemical indexes of alanine Aminotransferase (ALT), aspartate Aminotransferase (AST), total bilirubin (T-Bil) and Hydroxyproline (Hydroxyproline) proves that the noreulaldehyde has the function of protecting the liver function. RT-qPCR and Western Blot detection show that the demethylzelaronal can obviously reduce the expression level of hepatic fibrosis related genes in the liver of a hepatic fibrosis mouse. And then, carrying out data analysis on liver samples of the tested mice in a normal control group, a hepatic fibrosis model group and a noreulaldehyde drug treatment group by adopting transcriptome sequencing, and further discussing potential molecular mechanisms of the liver samples. According to the transcriptome sequencing analysis result and literature research, the function of the AGAP2 protein capable of promoting hepatic fibrosis in the anti-hepatic fibrosis process of the demethyleularmaldehyde is investigated. The result shows that the expression level of the AGAP2 gene in a hepatic fibrosis mouse model and an LX-2 cell can be obviously reduced by the demethylzelarstyle, so that the AGAP2 can be used as a drug action target of the demethylzelarstyle for resisting hepatic fibrosis. In conclusion, the demethyleularmaldehyde has new pharmacological activity of resisting hepatic fibrosis and is expected to become a potential therapeutic drug for resisting hepatic fibrosis.
Drawings
FIG. 1 is a graph showing the effect of T-96 on the expression of liver fibrosis-associated genes in activated LX-2 cells. Analyzing protein expression levels of hepatic fibrosis related genes Col1A1 and alpha-SMA in LX-2 cells of a normal control group, a model group and a noreulaldehyde treatment group with different concentrations by using an AWestern Blot; b RT-qPCR experiment analyzes the mRNA expression level of hepatic fibrosis related genes Col1a1 and alpha-SMA in LX-2 cells of a normal control group, a model group and a noreulaldehyde treatment group with different concentrations.
FIG. 2 shows hematoxylin-eosin staining (H)&E) Evaluation of T-96 treatment vs CCl4Induced liver fibrosis the effect of liver injury in mice is 100 μm.
FIG. 3 shows T-96 vs CCl4Induced liver fibrosis and the degree of liver fibrosis in mice. A results of Sirius Red Staining (Sirius Red Staining) of liver tissues of mice in a normal control group, a model group, a low-dose noreulaldehyde treatment group and a high-dose noreulaldehyde treatment group were 100 μm in a specific ruler; b is the semi-quantitative analysis of the collagen area of sirius red in the A picture.
FIG. 4 shows T-96 vs CCl4Induced liver fibrosis the effect of fibrosis-associated gene expression in the liver of mice. AWestern Blot analysis of protein expression of fibrosis-associated genes Col1A1, MMP-2 and alpha-SMA in liver tissues of mice in a normal control group, a model group, a low-dose noreulaldehyde treatment group and a high-dose noreulaldehyde treatment group; b RT-qPCR experiments analyzed the mRNA expression levels of fibrosis-associated genes Col1a1, Mmp-2 and alpha-SMA in liver tissues of mice in the normal control group, the model group, the low-dose noreulaldehyde-treated group and the high-dose noreulaldehyde-treated group.
FIG. 5 shows T-96 vs CCl4Induced liver fibrosis effects of liver function in mice. The serum biochemical indexes ALT, AST and T-Bil levels and the liver Hydroxyproline content of mice in an A-D normal control group, a model group, a low-dose noreulaldehyde treatment group and a high-dose noreulaldehyde treatment group.
FIG. 6 shows the effect of T-96 on the expression of AGAP2 gene. Analyzing mRNA expression level of Agap2 in liver tissues of mice in a normal control group, a model group, a low-dose noreulaldehyde treatment group and a high-dose noreulaldehyde treatment group by an ART-qPCR (amplified quantitative polymerase chain reaction) experiment; b, analyzing the mRNA expression level of the Agap2 in LX-2 cells of a normal control group, a model group and a noreulaldehyde treatment group with different concentrations by using a RT-qPCR (reverse transcription-quantitative polymerase chain reaction) experiment; c Western Blot analysis of protein expression levels of AGAP2 in LX-2 cells of a normal control group, a model group and a noreulaldehyde treatment group with different concentrations; d C graph.
Wherein:*P<0.05,**P<0.01,***P<0.001 compared to the normal group;#P<0.05,##P<0.01,###P<0.001 compared to the model group. The experimental data of the invention are processed, counted and analyzed by Excel and GraphPad Prism 7.0 software. All data are expressed as Mean ± standard deviation (Mean ± SD). Differences between two groups of samples were measured using a two-tailed t-test (t-test) with two-sample mean comparisons, and between groups were analyzed using One-way anova (One-way anova).
Detailed Description
The present invention will be described in further detail with reference to specific examples, which are provided only to assist those skilled in the art in fully understanding the present invention and do not limit the scope of the present invention. It will be understood that various modifications, improvements, substitutions and the like in the details or form of the technical solution of the present invention may be made without departing from the spirit and principle of the invention, and these modifications and substitutions should be construed as being included in the scope of the present invention.
Example 1 use of Noreulaldehyde to inhibit LX-2 cell activation
1. Drug treatment of LX-2 cells
LX-2 cells were purchased from the Hunan Yao medical college cell Bank, university of Central and south China. Noreulaldehyde (T-96) was purchased from Med Chem Express as a yellow powder with a purity of 99.90%. LX-2 cells cultured at 37 ℃ under 5% CO2 were induced by TGF-. beta.1 (final concentration 5ng/mL) to stimulate a model of adult cells, and different concentrations of noreulasialdehyde were added as drug treatment groups, and cells cultured in DMEM containing no compound and TGF-. beta.1 were used as normal control groups. Cell samples were collected after 24 h.
2. Real-time fluorescent quantitative polymerase chain reaction detection (RT-qPCR)
Cells were lysed using TRIzol and total RNA was extracted. After reverse transcription, RT-qPCR reaction was performed on a fluorescent quantitative PCR instrument according to the AceQ qPCR SYBR Green Master Mix kit instruction of Nanjing Novozam. The designed primers are shown in Table 1.
TABLE 1 primer sequences of human-derived hepatic fibrosis-associated genes
Figure BDA0003117737850000051
3.Western Blot
After drug treatment, each group of cells was added with an appropriate amount of protein lysate, after fully lysing the cells for 10min, centrifuged at 13000rpm for 15min at 4 ℃, and the supernatant was collected into a new EP tube. Protein concentration was determined using BCA assay kit. Adjusting the protein concentration, adding 6 × loading buffer, and boiling at 100 deg.C for 7min to denature the protein. After preparing the sample, 10% SDS-PAGE gel electrophoresis is used, the target band is transferred to a PVDF membrane, 5% skimmed milk powder is sealed for 2 hours at room temperature, after primary antibody is incubated overnight at 4 ℃, secondary antibody is incubated for 2 hours at room temperature, and ECL chemiluminescence is performed by using a Tanon 2500 gel imaging system.
4. Results of the experiment
As shown in FIG. 1, the expression levels of mRNA and protein of the liver fibrosis-associated genes COL1A1 and alpha-SMA in LX-2 cells were gradually decreased and dose-dependent as compared with the TGF-beta 1-induced model group with the increase in the concentration of compound T-96. Indicating that T-96 inhibits the activation of LX-2 cells.
Example 2 treatment of CCl with noreulaldehyde4Application of induced liver fibrosis in mice
1. Laboratory animals and reagents
The 7-8 week old C57BL/6J mice weighed about 18-22g and were purchased from Jiangsu Jiejiaokang Biotech GmbH. Carbon tetrachloride (CCl)4) Purchased from Sigma company.
2.CCl4Mouse model construction and administration scheme for inducing hepatic fibrosis
Intraperitoneal injection of CCl at a dose of 10mL/kg every week4Solution (CCl)4: corn oil is 1: 9 by volume) or corn oil 2 times, two weeks after molding, compound T-96 was administered and drug treatment was initiated. Mice were set as the following 4 groups: controlGroup (2): mice were gavaged daily with 10mL/kg of a cosolvent, i.e., 10% DMSO, 40% PEG300, 5% Tween-80, 45% saline. CCl4 model set: mice were gavaged with 10mL/kg of cosolvent per day. T-96 Low dose treatment group (15 mg/kg): mice were gavaged daily with 15mg/kg T-96 solution. T-96 high dose treatment group (30 mg/kg): mice were gavaged daily with 30mg/kg T-96 solution. After the experiment was completed, sample collection was performed. Blood was collected from the eyeball and placed in a 1.5mL centrifuge tube and allowed to stand on ice. Then, the mouse is killed by removing the neck, after the abdominal cavity is opened, the heart is firstly cut to exsanguinate, the gall bladder is removed, the whole liver tissue is taken out, and the liver shape of the mouse is recorded by photographing. Separating a small piece of liver tissue, fixing in 4% paraformaldehyde, placing the rest tissue in a freezing tube, rapidly freezing with liquid nitrogen, and storing in a refrigerator at-80 deg.C. A small piece of liver tissue is cut, RNA and protein are extracted to detect the expression of related genes. The detection method was the same as described in example 1. The primers used are shown in Table 2.
TABLE 2 primer sequences of mouse hepatic fibrosis-related genes
Figure BDA0003117737850000061
3. Liver pathology detection
Liver tissues were fixed in 4% paraformaldehyde overnight, paraffin-embedded, and sections cut to a thickness of about 5 μm were deparaffinized, then HE-stained and sirius red-stained, and pathological changes were observed under a microscope and image acquisition was performed.
4 liver function testing
The collected blood was placed in a refrigerator at 4 ℃ overnight, divided into two layers of serum and plasma the next day, and centrifuged at 4000rpm at 4 ℃ for 10 min. Taking the upper layer serum, subpackaging into EP tubes, and standing at-80 deg.C for use. The levels of the liver function markers ALT, AST and T-Bil were determined separately according to the instructions of the relevant kits.
5 determination of hepatic hydroxyproline content
In order to evaluate the degree of hepatic fibrosis and detect the content of Hydroxyproline (Hydroxyproline), an index reflecting the content of collagen in liver tissues, the specific steps are described in the specification of a determination kit of Nanjing institute of Biotechnology (alkaline hydrolysis method).
6. Results of the experiment
6.1 Noreulaldehyde improves CCl4Induced liver injury and degree of liver fibrosis in mice
The H & E staining results are shown in FIG. 2, the hepatocyte structures of the mice in the control group are complete and are distributed in a cord-like arrangement by taking the central vein as the center; the hepatic lobule structure is normal, and no pathological change is seen; the size of the hepatic blood sinus clearance in the tissue is uniform and does not expand. And the liver tissue structure of the model group mouse is damaged, the liver lobule structure is disordered, part of liver cells are subjected to fatty degeneration, round vacuoles with regular shapes can be seen in cells, the cell nucleus is deeply contracted, and has ballooning deformation or even cell necrosis, inflammatory cells around the central venous duct are infiltrated, and pseudolobules are formed. The mice in the drug treatment groups with different doses show that the structure of the liver cells and the structure of the hepatic lobules are basically normal and clear, the arrangement of the liver cells is dense, and the infiltration of inflammatory cells is obviously reduced.
The results of sirius red staining are shown in fig. 3A, and compared with the control group of mice, a large amount of collagen fiber hyperplasia was observed in liver tissues of the model group of mice, and collagen fiber deposits were clearly observed around the central venous duct and the manifold duct and connected with adjacent space to surround liver lobules, forming pseudo lobules. In the liver of the drug-treated mice, the deposition of collagen fibers was significantly reduced and the fiber spacing was not significant. More importantly, the hepatic fibrosis of the mice in the high dose treatment group is basically cured. By semi-quantitative analysis of sirius red staining of liver tissue of each group of mice, it was found that the collagen area in the T-96 treated group was significantly reduced compared to the model group and was dose-dependent (see fig. 3B).
6.2 Effect of Noreulaldehyde on expression of fibrosis-related genes in liver of hepatic fibrosis mice
As can be seen from FIG. 4, and CCl4Compared with the induced liver fibrosis model group mice, the mRNA and protein levels of liver fibrosis related genes COL1A1, MMP-2 and alpha-SMA in the liver of the mice treated by the noreulaldehyde medicament are greatly reduced, and the significant difference is achieved. Among them, the inhibition effect of the high-dose treatment group is more remarkable.
6.3 protection of hepatic fibrosis mice from liver function by Noreulaldehyde
After 4 weeks of administration, serum of mice was examined for liver function markers AST, ALT and T-Bil and the content of hydroxyproline in the liver was evaluated. As can be seen from FIG. 5, and CCl4Compared with the induced liver fibrosis model group mice, the serum AST, ALT and T-Bil levels of the mice treated by the drug are obviously reduced. And as the dosage of the drug is increased, the AST, ALT and T-Bil levels are gradually reduced. The collagen formation in the liver tissue is evaluated by measuring the Hydroxyproline content in the liver, and the result is shown in fig. 5D, so that the content of the Hydroxyproline in the liver of the mice in the treatment group is gradually reduced along with the increase of the drug dosage compared with the mice in the liver fibrosis model group, and the method has statistical significance. The results show that the demethyleularmin can reduce liver injury and reduce collagen formation, and has strong liver protection effect.
Example 3 application of Noreulaldehyde to inhibition of AGAP2 Gene expression
According to literature reports, AGAP2 plays an important role in the development process of hepatic fibrosis. It can respond to TGF-beta 1 signal factor to promote the occurrence of hepatic fibrosis. In addition, after comprehensive analysis of transcriptome sequencing results and the like in the early stage, the AGAP2 gene is selected for further research, and the function of the gene in the liver fibrosis resisting process of the demethyleularmaldehyde is examined. RT-qPCR and Western Blot experiments were carried out in the same manner as in example 1, using the primer sequences shown in Table 3.
TABLE 3 primer sequences
Figure BDA0003117737850000081
Firstly, the expression change condition of AGAP2 in the liver of a mice in a noreulaldehyde drug treatment group is verified, and as shown in FIG. 6A, the mRNA expression of the Agap2 in the liver of the mice in the drug treatment group is remarkably reduced compared with the liver fibrosis model group of mice. To further confirm the role of AGAP2 in T-96 drug therapy, RT-qPCR and Western Blot were used to detect changes in expression in LX-2 cells treated with different concentrations of noreulaldehyde, which was found to decrease the level of mRNA and protein expression of AGAP2 induced by TGF-. beta.1 in a dose-dependent manner (see FIGS. 6B-D). Taken together, noreulaldehyde may exert anti-hepatic fibrosis activity by inhibiting expression of AGAP 2.
Sequence listing
<110> university of Chinese pharmacy
Application of <120> demethyleularmaldehyde in preparation of medicine for preventing or treating hepatic fibrosis
<160> 18
<170> SIPOSequenceListing 1.0
<210> 1
<211> 20
<212> DNA
<213> Human β-actin Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 1
ggacttcgag caagagatgg 20
<210> 2
<211> 20
<212> DNA
<213> Human β-actin Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 2
agcactgtgt tggcgtacag 20
<210> 3
<211> 23
<212> DNA
<213> Human α-SMA Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 3
gacaatggct ctgggctctg taa 23
<210> 4
<211> 21
<212> DNA
<213> Human α-SMA Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 4
ctgtgcttcg tcacccacgt a 21
<210> 5
<211> 20
<212> DNA
<213> Human Col1a1 Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 5
gtgctaaagg tgccaatggt 20
<210> 6
<211> 20
<212> DNA
<213> Human Col1a1 Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 6
ctcctcgctt tccttcctct 20
<210> 7
<211> 20
<212> DNA
<213> Mouse β-actin Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 7
agccatgtac gtagccatcc 20
<210> 8
<211> 20
<212> DNA
<213> Mouse β-actin Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 8
ctctcagctg tggtggtgaa 20
<210> 9
<211> 21
<212> DNA
<213> Mouse α-SMA Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 9
cgggagaaaa tgacccagat t 21
<210> 10
<211> 22
<212> DNA
<213> Mouse α-SMA Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 10
agggacagca cagcctgaat ag 22
<210> 11
<211> 22
<212> DNA
<213> Mouse Mmp-2 Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 11
acctgaacac tttctatggc tg 22
<210> 12
<211> 19
<212> DNA
<213> Mouse Mmp-2 Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 12
cttccgcatg gtctcgatg 19
<210> 13
<211> 19
<212> DNA
<213> Mouse Col1a1 Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 13
gctcctctta ggggccact 19
<210> 14
<211> 20
<212> DNA
<213> Mouse Col1a1 Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 14
attggggacc cttaggccat 20
<210> 15
<211> 20
<212> DNA
<213> Mouse Agap2 Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 15
gctgagaagc ggagcttaga 20
<210> 16
<211> 20
<212> DNA
<213> Mouse Agap2 Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 16
ctgggacttt gaccgttgtt 20
<210> 17
<211> 20
<212> DNA
<213> Human Agap2 Forward(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 17
gtgactccga gaaacgaagc 20
<210> 18
<211> 20
<212> DNA
<213> Human Agap2 Reverse(2 Ambystoma laterale x Ambystoma jeffersonianum)
<400> 18
ccgggacttt gactgttgtt 20

Claims (3)

1. The application of the demethyleularmin in the preparation of the medicine for preventing or treating hepatic fibrosis or hepatic fibrosis related diseases is disclosed, wherein the structural formula of the demethyleularmin is shown as the formula I:
Figure FDA0003117737840000011
2. the use according to claim 1, wherein the noreulaldehyde is used for inhibiting the occurrence of a liver fibrosis-related disease; and inhibiting activation of hepatic stellate cells.
3. The use according to claim 2, wherein the liver fibrosis related disease comprises: alcoholic liver disease, non-alcoholic fatty liver disease, viral hepatitis, autoimmune liver disease, and hepatic blood stasis.
CN202110666788.XA 2021-06-16 2021-06-16 Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis Pending CN113244244A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110666788.XA CN113244244A (en) 2021-06-16 2021-06-16 Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110666788.XA CN113244244A (en) 2021-06-16 2021-06-16 Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis

Publications (1)

Publication Number Publication Date
CN113244244A true CN113244244A (en) 2021-08-13

Family

ID=77188209

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110666788.XA Pending CN113244244A (en) 2021-06-16 2021-06-16 Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis

Country Status (1)

Country Link
CN (1) CN113244244A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110218143A1 (en) * 2007-11-20 2011-09-08 University Of Florida Research Foundation Compositions and methods for tissue repair
CN103524592A (en) * 2013-09-27 2014-01-22 安徽医科大学 Tripterine derivative, biogenetic salt of derivative, and preparation method and application of biogenetic salt
CN110585217A (en) * 2019-10-30 2019-12-20 中国科学院昆明植物研究所 Application of tripterine in medicine for treating non-alcoholic steatohepatitis
CN111973604A (en) * 2017-03-15 2020-11-24 中国科学院昆明植物研究所 Application of tripterine in preparation of medicine for treating cholestatic liver disease

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110218143A1 (en) * 2007-11-20 2011-09-08 University Of Florida Research Foundation Compositions and methods for tissue repair
CN103524592A (en) * 2013-09-27 2014-01-22 安徽医科大学 Tripterine derivative, biogenetic salt of derivative, and preparation method and application of biogenetic salt
CN111973604A (en) * 2017-03-15 2020-11-24 中国科学院昆明植物研究所 Application of tripterine in preparation of medicine for treating cholestatic liver disease
CN110585217A (en) * 2019-10-30 2019-12-20 中国科学院昆明植物研究所 Application of tripterine in medicine for treating non-alcoholic steatohepatitis

Similar Documents

Publication Publication Date Title
Corcoran et al. Pharmacological inhibition of the nod-like receptor family pyrin domain containing 3 inflammasome with MCC950
CN114191553B (en) Medicine for resisting novel coronavirus SARS-CoV-2 and its application
Chu et al. Paeoniflorin attenuates schistosomiasis japonica-associated liver fibrosis through inhibiting alternative activation of macrophages
EP3349759B1 (en) Dactinomycin compositions and methods for the treatment of acute myeloid leukemia
WO2021052305A1 (en) Use of eight-treasure pill in preparation of medicine for preventing or treating diseases related to il-6 inflammatory cytokine storm
TWI765181B (en) Application of chiglitazar and its related compounds
CN110279697B (en) Application of iron death inducer in medicine for treating or relieving allergic airway inflammation
Sun et al. S100A9 blockade improves the functional recovery after spinal cord injury via mediating neutrophil infiltration
CN113244244A (en) Application of demethyleularmin in preparation of medicine for preventing or treating hepatic fibrosis
US11541030B2 (en) Methods for the treatment of inflammation associated with infection
EP4346831A2 (en) Tgf-beta therapeutics for age disease
CN114129556A (en) Application of wild tectorigenin in treating hepatic fibrosis
WO2021195698A1 (en) Methods for the treatment of inflammation associated with infection
CN113925851A (en) Application of Boropinol-B in preparation of medicine for treating insomnia
TW202200183A (en) Methods for treating inflammatory and fibrotic diseases and disorders
CN108451949B (en) Application of paeoniflorin metabolite I in preparation of colitis treatment drug
CA3176169A1 (en) Compositions and methods for treating cytokine storms
CN110856716B (en) Application of compound and pharmaceutical composition thereof in preventing and treating kidney fibrosis
US8507446B2 (en) Use of PEDF-derived polypeptides for treating liver cirrhosis
WO2020004404A1 (en) IL-1β INHIBITOR
CN117919422A (en) Application of Yoda1 in preparation of medicine for treating liver fibrosis
CN117357511A (en) Application of Ginkgetin in preparation of STING inhibitor
CN116785284A (en) Application of dehydrocorydaline in preparation of medicine for treating idiopathic pulmonary fibrosis
Zou et al. Methylated MFGE8 Promotes Early Brain Injury After Subarachnoid Hemorrhage and Inhibiting Autophagy of Nerve Cell
CN114377133A (en) Targeting renal CD8+Application of TRM formation and activation in glomerular injury

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: 20210813