CN112294800A - Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury - Google Patents

Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury Download PDF

Info

Publication number
CN112294800A
CN112294800A CN201910709404.0A CN201910709404A CN112294800A CN 112294800 A CN112294800 A CN 112294800A CN 201910709404 A CN201910709404 A CN 201910709404A CN 112294800 A CN112294800 A CN 112294800A
Authority
CN
China
Prior art keywords
nrf2
liver
nafld
mice
moa
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
CN201910709404.0A
Other languages
Chinese (zh)
Inventor
余璐
王凌
蒋学华
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.)
Sichuan University
Original Assignee
Sichuan 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 Sichuan University filed Critical Sichuan University
Priority to CN201910709404.0A priority Critical patent/CN112294800A/en
Publication of CN112294800A publication Critical patent/CN112294800A/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
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • 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

Landscapes

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

Abstract

The invention provides application of ellagic acid in preparing a medicine for preventing and/or treating non-alcoholic fatty liver disease and liver injury. The invention proves that the ellagic acid is an Nrf2 agonist, can increase the level of GSH in liver by up-regulating the expression of Nrf2 and downstream antioxidant factors and transporters thereof, simultaneously reduces the level of ROS in liver cells, reduces oxidative stress reaction and inflammatory reaction, maintains the function of the liver cells, and thus plays a role in treating NAFLD. In addition, ellagic acid can increase levels of GSH in the liver by up-regulating expression of Nrf2 and its downstream antioxidant factors and transporters, thereby serving to prevent hepatotoxicity due to increased normal doses of APAP by NAFLD. Ellagic acid has good application prospect in preparing NAFLD therapeutic drugs and APAP hepatotoxicity prevention drugs.

Description

Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury
Technical Field
The invention belongs to the field of medicines, and particularly relates to application of ellagic acid in preparing a medicine for preventing and/or treating non-alcoholic fatty liver disease and liver injury.
Background
Nonalcoholic fatty liver disease (NAFLD) is a metabolic stress liver injury associated with genetic predisposition and insulin resistance. The pathological feature of the clinical disease is hepatic steatosis and the patient has no history of excessive alcohol consumption. The disease spectrum of NAFLD includes: nonalcoholic simple fatty liver, Nonalcoholic steatohepatitis (NASH) and its associated cirrhosis and Hepatocellular carcinoma (HCC). NAFLD can cause not only disability and death associated with liver disease but also has a strong correlation with the high prevalence of metabolic syndrome, type II diabetes, arteriosclerotic cardiovascular disease, and the like. NAFLD is one of the globally important public health problems of the 21 st century. According to the latest data of 2018 edition of Chinese guide on prevention and treatment of nonalcoholic fatty liver diseases, NAFLD has become the first chronic liver disease in China at present and seriously harms the life health of people.
NAFLD is mainly caused by accumulation of free fatty acids in the human body due to high-fat high-sugar diet, obesity, diabetes, sedentary and sedentary life style and other life styles. Oxidative Stress (OS) caused by excessive free radical formation by peroxidation of accumulated fatty acids, mitochondrial dysfunction and generation of more Reactive Oxygen Species (ROS) caused by cardiolipin peroxidation, and formation of proinflammatory cytokines, etc., ultimately leading to necrosis and apoptosis of hepatocytes. NAFLD can provide some relief through dietary and exercise interventions, but currently there is no approved drug therapy.
Nrf2(Nuclear factor 2-related factor 2, NF-E2-related factor 2) is a key regulatory factor in antioxidant stress and anti-inflammatory reaction, and is mainly responsible for regulating the expression of antioxidant proteins such as quinone oxidoreductase (NADPH: quinone oxido reductase-1, NQO1) and Glutathione (GSH) synthesis and metabolism-related enzymes, Heme oxygenase 1 (HO-1), ATP-transporter (ATP binding cassette transporter), phase II detoxifier and transporter, and is helpful for clearing up endogenous/toxic substances and active oxygen free radicals, thereby playing a role in protecting cells. There are studies that show that: nrf2 can prevent and treat NAFLD by inhibiting fat accumulation in liver, regulating cholesterol and bile acid metabolism, and resisting Free Fatty Acid (FFAs) induced liver cell steatosis. In addition, NAFLD can significantly induce levels of CYP2E1 (cytochrome P4502E 1) in liver tissue, resulting in a significant increase in toxic harmful substances produced by its metabolism.
Acetaminophen (APAP) is a nonsteroidal antipyretic analgesic and is one of the main active ingredients of most OTC anti-cold drugs. As a first-line treatment for colds, nearly 80% of these drugs contain APAP. Although APAP has good antipyretic and analgesic effects and high safety, liver injury is frequently caused by excessive APAP administration clinically. According to the clinical medication need of pharmacopoeia of the people's republic of China (2015 edition), the dosage of APAP orally taken by an adult at one time is 0.3-0.6 g, and the maximum dose is not more than 2.0g per day. Clinical studies report that a daily dose of APAP exceeding 4g can cause significant liver damage and serious life risks. When NAFLD patients take normal doses of acetaminophen (APAP), the amount of N-acetyl-p-benzoquinone imine (NAPQI) produced by CYP2E1 metabolism is significantly increased. The metabolite can bind to the reducing GSH in the liver and is eventually excreted to the outside of the body through the kidneys, but a large amount of NAPQI will deplete the GSH in the liver, and an excessive amount of NAPQI will bind to cellular proteins, causing oxidative stress, leading to further liver damage. Glutamate cysteine ligase, glutathione reductase, glutathione synthetase and the like regulated by Nrf2 can increase the level of reduced GSH in cells; MRP2 (multidrug resistance-associated protein 2) and MRP4 (multidrug resistance-associated protein 4), both downstream of Nrf2, are the major efflux transporters responsible for the NAPQI glutathione conjugate; furthermore, the antioxidant proteins NQO1 and HO-1 and the like at the downstream can resist the peroxidation stress caused by excessive NAPHQI. Therefore, the activation of the Nrf2 signal path can neutralize excessive NAPHQI, promote the discharge of toxic substances and relieve liver injury of APAP by three ways of antioxidation stress response.
Traditional Chinese medicines and natural medicines are important ways for discovering innovative medicines, and research on effective ingredients of the traditional Chinese medicines and the natural medicines is concerned by researchers. Although many monomeric Nrf2 agonists of traditional Chinese medicines and natural medicines (such as curcumin and resveratrol) are in clinical trials at present, a lot of difficulties and challenges are still faced in the development process.
Therefore, the research and development of new drugs capable of activating the Nrf2 signal pathway are of great significance for improving the current severe NAFLD and preventing the APAP hepatotoxicity increase caused by the disease.
Disclosure of Invention
The invention aims to provide application of ellagic acid in preparing a medicament for preventing and/or treating nonalcoholic fatty liver diseases and liver injury.
The invention provides application of ellagic acid in preparing a medicament for preventing and/or treating non-alcoholic fatty liver disease and liver injury.
Further, the liver injury is a chemical induced liver injury.
Further, the liver damage is caused by acetaminophen administration.
Further, the liver damage is caused by the administration of normal doses of acetaminophen.
"Normal dose acetaminophen": according to the clinical medication need of pharmacopoeia of the people's republic of China (2015 edition), the dosage of acetaminophen (APAP) orally taken once by an adult is 0.3-0.6 g, and the maximum dose is not more than 2.0g per day.
Further, the non-alcoholic fatty liver disease includes non-alcoholic simple fatty liver, non-alcoholic fatty hepatitis, liver cirrhosis, and hepatocellular carcinoma.
Further, the drug can induce the expression of the Nrf2 and the downstream regulatory gene and the efflux transporter thereof.
Further, the downstream regulatory genes of the Nrf2 are NQO1, HO-1, MRP2 and MRP 4; the efflux transporters are MRP2 and MRP 4.
Further, the induction of the expression of the Nrf2 is realized by regulating the promoter region of the Nrf2 to perform DNA methylation transfer, and/or by inducing HAT1 and inhibiting the expression of HDAC1 and HDAC 6.
Further, the drug is capable of counteracting oxidative stress and inflammatory reaction caused by non-alcoholic fatty liver disease.
Further, the drug is capable of reducing ROS levels in liver cells; the drug is capable of increasing GSH levels in liver cells.
Furthermore, the medicine can promote toxic substance discharge and neutralize N-acetyl-p-benzoquinone imine, reduce oxidative stress injury in liver cells and protect liver cells.
Experimental results show that the ellagic acid is an agonist of Nrf2, can increase the level of GSH in liver by up-regulating the expression of Nrf2 and its downstream antioxidant factors and transporters, and simultaneously can reduce the level of ROS in liver cells, reduce oxidative stress reaction and inflammatory reaction, maintain the function of liver cells, thereby playing a role in treating NAFLD. In addition, ellagic acid can increase levels of GSH in the liver by up-regulating expression of Nrf2 and its downstream antioxidant factors and transporters, thereby serving to prevent hepatotoxicity due to increased normal doses of APAP by NAFLD. Further epigenetic studies have also found that ellagic acid induces expression of Nrf2 by regulating the Nrf2 promoter region for DNA methylation transfer, and/or by inducing HAT1 (histone acetyltransferase 1) and inhibiting expression of HDAC1 and HDAC 6. Therefore, the ellagic acid has good application prospect in preparing NAFLD treatment medicines and APAP hepatotoxicity prevention medicines.
Obviously, many modifications, substitutions, and variations are possible in light of the above teachings of the invention, without departing from the basic technical spirit of the invention, as defined by the following claims.
The present invention will be described in further detail with reference to the following examples. This should not be understood as limiting the scope of the above-described subject matter of the present invention to the following examples. All the technologies realized based on the above contents of the present invention belong to the scope of the present invention.
Drawings
Figure 1.2 month high fat diet effect on ALT and AST in sera of wild type and Nrf2 knockout mice (n ═ 6, × p <0.01, compared to control group; # p <0.05, compared to NAFLD (Nrf2 +/+)).
Figure 2.2 month high fat diet feeding effect on liver appearance characteristics of wild type and Nrf2 knockout mice.
Figure 3.2 month high fat diet effect on liver specific gravity of wild type and Nrf2 knockout mice (n ═ 6,. p <0.01, compared to control group; # p <0.05, compared to NAFLD (Nrf2+/+) group).
FIG. 4.2 month after high fat diet feeding, HE staining results of liver tissues of wild type and Nrf2 knockout mice are shown. Red arrow: punctate necrosis; black arrows: inflammatory cell infiltration; green arrow: performing vacuole denaturation; blue arrow: and (4) fatty degeneration.
FIG. 5.2 month after high fat diet feeding, oil red O staining results of liver tissues of wild type and Nrf2 knockout mice are shown. Yellow arrow: nuclear cavitations; green arrow: and (4) fat dripping.
Figure 6. effect of EA, MoA, CUR on ALT in serum of wild-type and Nrf2 knockout NAFLD mouse model sera, administered by gavage for 1 week, respectively (n ═ 6,. p <0.05,. p <0.01, compared to control group of the same genotype;. p <0.05, compared to NAFLD group of the same genotype).
Figure 7. effect of EA, MoA, CUR on AST in sera of wild-type and Nrf2 knockout mice models of NAFLD given by gavage for 1 week respectively (n ═ 6, # p <0.05, # p <0.01, compared to control group of the same genotype; # p <0.05, compared to NAFLD group of the same genotype).
Figure 8 effect of EA, MoA, CUR on appearance characteristics of wild type and Nrf2 knockout NAFLD mouse model liver given by gavage for 1 week, respectively.
Figure 9. effect of EA, MoA, CUR on liver specific gravity in wild-type and Nrf2 knockout mice model liver for 1 week gavage, respectively (n 6, # p <0.05, # p <0.01, compared to control group of the same genotype; # p <0.05, # p <0.01, compared to NAFLD group of the same genotype).
FIG. 10 is a graph of the results of HE staining of liver tissue in wild-type and Nrf2 knockout mice models of NAFLD after 1 week of intragastric administration of EA, MoA, CUR, respectively. Red arrow: punctate necrosis; black arrows: inflammatory cell infiltration; green arrow: performing vacuole denaturation; blue arrow: and (4) fatty degeneration.
FIG. 11 is a graph of the results of oil red O staining of liver tissue in wild type and Nrf2 knockout NAFLD mouse models after 1 week intragastric administration of EA, MoA, and CUR, respectively. Yellow arrow: nuclear cavitations; green arrow: and (4) fat dripping.
Figure 12 effects of 1 week intragastric administration of EA, MoA, CUR on GSH levels in liver tissues of wild-type and Nrf2 knockout mice model mice, respectively (n ═ 6,. p <0.05,. p <0.01, compared to control group of the same genotype;. p <0.05, compared to NAFLD group of the same genotype).
Figure 13. effect of EA, MoA, CUR on Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4mRNA levels in wild-type (a) and Nrf2 knockout (B) NAFLD mouse model liver tissues given intragastric administration for 1 week, respectively (n ═ 6, × p <0.05, × p <0.01, compared to control group of the same genotype; # p <0.05, compared to NAFLD group of the same genotype).
Figure 14 effects of gavage administration of EA, MoA, CUR for 1 week on the levels of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4 protein in NAFLD mouse model liver tissue of wild type (a, Nrf2(+/+)) and Nrf2 knockouts (B, Nrf2(-/-)) (n ═ 6, # p <0.05, # p <0.01, compared to control group of the same genotype; # p <0.05, compared to NAFLD group of the same genotype), respectively.
FIG. 15 nucleotide sequence of mouse Nrf2 (SEQ ID NO. 41).
FIG. 16.100, 500, 1000. mu.M FFAs treated for 24h, L02, THLE-3 and HepG2 are graphs showing the results of oil red O staining.
FIG. 17.0-100 μ M effect of EA, MoA and CUR on L02, THLE-3 and HepG2 cell viability (n-5).
FIG. 18 effects of EA, MoA and CUR on ROS levels in NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) (n 3, # p <0.05, # p <0.01, vs. control; # p <0.05, # p <0.01 vs. NAFLD; # p <0.05 vs. 50 μ M EA).
FIG. 19.EA, MoA and CUR effect on GSH levels in NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) (n 3, p <0.05, p <0.01, compared to control; # p <0.05, # p <0.01, compared to NAFLD group; $ p <0.05, compared to 50 μ M EA group; @ @ p <0.01, compared to 10/25 μ M MMoA group)).
FIG. 20. the effects of EA, MoA and CUR on the expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1mRNA in the NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) (n ═ 3,. p <0.05,. p <0.01, compared to the control group,. p <0.05,. #. p <0.01, compared to the NAFLD group,. p <0.05,. $ p <0.01, compared to the 50 μ M EA group,. p <0.05,. @ p <0.01, compared to the 10/25 μ M group).
FIG. 21. effects of EA, MoA and CUR on expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1 proteins in NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) (n ═ 3,. p <0.05,. p <0.01, compare control;. p <0.05,. p <0.01, compare NAFLD group,. p <0.05,. $ p <0.01, compare 50. mu.M EA group;. p <0.05,. @ p <0.01, compare 10/25. mu. MMoA group).
FIG. 22.500 μ M FFAs treated for 24h, oil red O staining results for NRF2 knockdown L02, THLE-3 and HepG2 cells (transfected with NC-siRNA or NRF 2-siRNA).
FIG. 23 Effect of EA, MoA and CUR on ROS levels in NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) NRF2 knockdown (n ═ 3,. p <0.05,. p <0.01, compared to control;. p <0.05,. # p <0.01, compared to NAFL + NC-siRNA).
Figure 24 effect of EA, MoA and CUR on GSH levels in the NAFLD cell model of NRF2 knockdown of L02(a), THLE-3(B) and HepG2(C) (n ═ 3,. p <0.05,. p <0.01, compared to control group;. p <0.05, compared to NAFL + NC-siRNA group).
FIG. 25. effects of EA, MoA and CUR on expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1mRNA in NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) (n ═ 3,. p <0.05,. p <0.01, compare control;. p <0.05,. p;. 0.01, compare NAFL + NC-siRNA group).
FIG. 26. effects of EA, MoA and CUR on expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1 proteins in NAFLD cell models of L02(A), THLE-3(B) and HepG2(C) (n ═ 3,. p <0.05,. p <0.01,. compared to control,. p <0.05,. p # p <0.01, compared to NAFLD group).
FIG. 27. Effect of gavage administration of 150mg/kg APAP on serum ALT and AST concentrations in wild type mice at 9AM and 5PM, respectively, for 3 consecutive days (n 6,. p <0.01, vs. control;. p <0.05 vs. AM).
FIG. 28. Effect of gavage of 150mg/kg APAP on liver appearance characteristics in wild type mice at 9AM and 5PM, respectively, for 3 consecutive days.
FIG. 29 is a graph of HE staining results of liver tissue from wild type mice after gavage administration of 150mg/kg APAP at 9AM and 5PM, respectively, for 3 consecutive days. Black arrows: infiltration of inflammatory cells; green arrow: vacuolar degeneration.
FIG. 30 shows the change in serum ALT concentration in mice when intragastrically administered EA, MoA and CUR for 1 week consecutively and 150mg/kg of APAP was intragastrically administered 1h after the administration on the third day from the last, and 3 days after the consecutive administration (n ═ 6,. sup.p <0.05,. sup.p <0.01, compared to the control group;. sup.p <0.05,. sup.p <0.01, compared to the APAP group;. sup.p <0.05, compared to the NAFLD + APAP group;. sup.p <0.01, compared to the APAP (Nrf2+/+) group;. sup.p <0.05, compared to the NAFLD + APAP (Nrf2+/+) group).
FIG. 31 shows changes in AST concentration in mice serum when the mice were gavaged with EA, MoA and CUR for 1 week consecutively and 150mg/kg of APAP was gavaged 1h after the administration on the third day after the last, and 3 days after the consecutive administration (n ═ 6,. beta.p <0.05,. beta.p <0.01, compared to the control group,. beta.p <0.05,. beta.p <0.01, compared to the APAP group,. beta.p <0.05, compared to the NAFLD + APAP group,. beta.p <0.01, compared to the APAP (Nrf2+/+) group,. beta.p <0.05, compared to the NAFLD + APAP (Nrf2+/+) group).
FIG. 32 Effect of intragastric administration of EA, MoA and CUR, respectively, for 1 week consecutively, and 150mg/kg APAP for 1h intragastric administration on the third last day after the administration, on appearance of liver characteristics of mice for 3 days consecutively.
FIG. 33 is a graph of HE staining results of mouse liver tissue following intragastric administration of EA, MoA, and CUR for 1 week, respectively, and 150mg/kg APAP 1h postgavage on the third last day, for 3 days. Red arrow: punctate necrosis; black arrows: inflammatory cell infiltration; green arrow: performing vacuole denaturation; blue arrow: and (4) fatty degeneration.
FIG. 34 is a graph of the results of oil red O staining of mouse liver tissue on the penultimate day after intragastric administration of EA, MoA, and CUR for 1 week, followed by 150mg/kg APAP 1h after intragastric administration and 3 days after continuous administration. Yellow arrow: nuclear cavitations; green arrow: and (4) fat dripping.
FIG. 35. Effect of intragastric administration of EA, MoA and CUR, respectively, for 1 week consecutively, and 150mg/kg of APAP administered intragastric 1h after administration on the third last day, 3 days consecutively on GSH levels in liver tissue of mice (n ═ 6,. p <0.05,. p <0.01, compared to a control group of the same genotype;. p <0.05, compared to a NAFLD + APAP group of the same genotype;. p <0.05, compared to an APAP (Nrf2+/+) group;. p <0.05, compared to a NAFLD + APAP (N2 +/+) group).
FIG. 36. 1 week consecutively gavage EA, MoA and CUR, respectively, and 150mg/kg APAP 1h after the administration on the third last day, 3 days consecutively, the effect on the levels of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4mRNA in liver tissues of wild-type (A) and Nrf2 knockout (B) mice (n ═ 6, $ p <0.05, $ p <0.01, compared to control group of the same genotype, $ p <0.05, compared to NAFLD group of the same genotype, $ p <0.05, $ p <0.01, compared to NAFLD + APAP group of the same genotype).
FIG. 37. Effect of intragastric administration of EA, MoA and CUR, respectively, for 1 week consecutively, and 150mg/kg of APAP administered intragastric administration 1h after administration for 3 days after the last three days on the levels of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4 in liver tissue of wild-type (A) and Nrf2 knockout (B) mice (n ═ 6, $ p <0.05, $ p <0.01, compared to control group of same genotype; # p <0.05, # p <0.01, compared to APAP group of same genotype; $ $ p <0.01, compared to LD + APAP group of same genotype).
Fig. 38 shows the effect of EA on methylation of CpG islands in the promoter region of NRF2(n 3, p <0.05, p <0.01, compared to control).
Figure 39.50,100 μ M EA effect on HAT1 and related HDACs protein expression in L02 cells (n 3, # p <0.05, # p <0.01, vs. control; # p <0.05, # p <0.01 vs. 50 μ M EA).
Figure 40 is a mechanistic diagram of EA treatment for NAFLD and prevention of APAP hepatotoxicity increased by NAFLD.
FIG. 41 is a graph of the mechanism of action of EA and MoA in the treatment of NAFLD.
Detailed Description
The raw materials and equipment used in the invention are known products and are obtained by purchasing commercial products.
Wherein Ellagic Acid (EA), CAS: 476-66-4, purchased from Bailingwei technology.
Ophiopogonin a methyl (moa), CAS: 74805-90-6, available from Doppel scientific development, Inc.
Curcumin (CUR), CAS: 458-37-7, purchased from carbofuran technologies. Curcumin is a known Nrf2 agonist and serves as a positive control drug in the present invention.
Example 1 preliminary study of the therapeutic effects of EA and MoA on NAFLD mouse model and its mechanism
1. Establishment and verification of NAFLD mouse model
1.1 grouping and raising of laboratory animals and sample Collection
C57BL/6 mice (bred by the laboratory) of wild type and Nrf2 knockout homozygotes (Nrf2-/-) were selected for 12 mice each, and the mice were randomly divided into 2 groups (of the same genotype) of 6 mice each, each group being hermaphroditic. After adaptive feeding for 1 week under 12h illumination and 12h darkness, high fat feed is fed for 2 months, and the grouping and feeding scheme is shown in table 2-1.
Table 2-1 grouping and feeding protocol for mice (n ═ 6).
Figure BDA0002153202320000081
After 2 months of breeding, 10% chloral hydrate (0.5g/kg) is injected into the abdominal cavity of the mouse for anesthesia, after the anesthesia is successful, the blood and perfusion of the heart are rapidly carried out, the liver is taken out, part of liver tissue cleaned by normal saline is put into 4% paraformaldehyde fixing solution for soaking, and the rest of liver tissue is stored at-70 ℃ for standby. The whole blood was placed in a clean 1.5mL centrifuge tube, allowed to stand overnight at 4 ℃ and centrifuged at room temperature for 10min (4000rpm) to isolate serum. The body weight and liver weight of each group of mice were recorded.
1.2 Biochemical index analysis of mouse liver function
1.2.1 sample treatment
The concentration of alanine Aminotransferase (ALT) and aspartate Aminotransferase (AST) in the serum obtained under the 1.1 standard was measured by a full-automatic biochemical analyzer.
1.2.2 data processing
The experimental results were performed using Graphpad prism 7 software for data conditioning and one-way analysis of variance (ANOVA), with differences statistically significant when p < 0.05.
1.2.3 results of the experiment
The results of ALT and AST concentrations in the serum of each group of mice are shown in FIG. 1.
As shown in figure 1, compared with a blank control group, the concentration of ALT and AST in the serum of two genotype mice is obviously increased (p is less than 0.01) in the NAFLD model group which is fed with high-fat feed for 2 months, and the indexes of the other NAFLD model groups except the AST of the NAFLD (Nrf2+/+) group mice are higher than the normal level (ALT: 30-110U/L, AST: 60-220U/L). Meanwhile, compared with NAFLD (Nrf2+/+) group, NAFLD (Nrf2-/-) group mice serum AST concentration is higher (p < 0.05). The above results illustrate that: high fat diet feeding for 2 months can cause obvious damage to livers of wild type and Nrf2 knockout mice, and deletion of Nrf2 can aggravate the degree of liver damage of the mice caused by the Nrf 2.
1.3 mouse liver injury and liver tissue section staining analysis
1.3.1 mouse liver injury
The appearance of the liver of the two genotype mice after 2 months of high fat diet feeding is shown in figure 2. From the results in fig. 2, it can be seen that liver tissues of the mice in each group have no visible damage, but compared with the control group, the gall bladder of the wild type mice and NAFLD mice knocked out by Nrf2 fed with high fat feed for 2 months is obviously enlarged. Meanwhile, compared with the NAFLD (Nrf2+/+) group, the degree of gallbladder enlargement of the mice in the NAFLD (Nrf2-/-) group is more obvious. The above results illustrate that: when the mice are fed with high-fat feed for 2 months, gall bladder of wild type and Nrf2 knockout mice can be obviously swollen, and the deletion of Nrf2 can increase the swelling degree of the gall bladder of the mice caused by the deletion.
1.3.2 mouse liver ratio Re-analysis
Liver specific gravity is an index for evaluating the severity of NAFLD, and a larger specific gravity indicates more accumulation of fat in the liver. The calculation formula is as follows:
liver specific gravity (g)/mouse weight (g) × 100%
The results of the liver specific gravity of each group of mice calculated according to the above formula are shown in FIG. 3.
As can be seen from the liver specific gravity statistical result graph, the liver specific gravity of the wild type of the NAFLD group and the Nrf2 knockout mouse is obviously increased compared with the control group with the same genotype, and has statistical difference (p < 0.01). Meanwhile, the liver specific gravity increase of mice in the NAFLD (Nrf2-/-) group was more significant (p <0.05) compared to the NAFLD (Nrf2+/+) group. The above results illustrate that: high fat diet feeding for 2 months can significantly increase liver specific gravity of wild type and Nrf2 knockout mice, and deletion of Nrf2 can aggravate the degree of liver specific gravity increase of the mice caused by the same.
1.3.3 HE staining analysis
Soaking the liver tissues in 4% paraformaldehyde fixing solution for 24-48 h, and then dehydrating, trimming, embedding, slicing, HE staining and sealing. The morphological change of the liver tissue cells was observed under an optical microscope, and the observation result is shown in fig. 4.
As can be seen from fig. 4, in comparison with the control group, vacuolar degeneration and steatosis occurred in liver tissues of both wild type mice fed with high fat diet for 2 months and NAFLD mice knocked out with Nrf2, and a small amount of lipid droplets with different sizes, round shapes and obvious outlines were observed in cytoplasm of the steatosis-occurring hepatocytes. Meanwhile, liver cells of mice in the NAFLD (Nrf2-/-) group also have a small amount of inflammatory cell infiltration, and part of liver cells show different degrees of punctate necrosis. The above results illustrate that: high fat feed for 2 months can cause liver lesions of wild type and Nrf2 knockout mice to different degrees, and the loss of Nrf2 can aggravate liver injury of the mice caused by the liver lesions.
1.3.4 oil Red O staining analysis
1.3.4.1 frozen sections
Adjusting the temperature of the low-temperature constant-cooling box to-20 ℃, taking out the collected liver tissue from the refrigerator at-70 ℃ after the specimen placing platform refrigerates to reach the required temperature, and placing the liver tissue on a propeller of a microtome. And adjusting the angle of the slicing knife, and setting the slicing thickness to be 10-15 mu m, so that slicing can be carried out. And placing the cut sample slice in the center of an anti-falling glass slide, and sealing and storing the sample slice to-20 ℃ for later use.
1.3.4.2 oil Red O staining
1) Taking out the pre-made frozen section from a refrigerator at-20 deg.C, placing on a slicing rack, and standing at room temperature for 10min for dyeing.
2) Preparing oil red O dyeing application liquid, comprising the following operation steps: according to the ratio (volume ratio) of oil red O stock solution to diluent to 5:2, a proper amount of application solution is prepared according to the amount of the sample. The prepared application liquid needs to be filtered by slow filter paper to remove impurities and is used within 2 h.
3) Directly putting the cooled slice into a dye vat filled with oil red O dyeing application liquid for dyeing for 10-15 min, and rinsing for 5-20 s by UP water at the temperature of about 37 ℃.
4) And (3) placing the slices into a dye vat filled with a re-dyeing solution for dyeing for 3-5 min, and rinsing with UP water for 30-60 s.
5) When the water on the surface of the slide is not dried completely, the three-water-based sealing agent (heated to liquid state in water bath at 60 ℃) can be dripped on the surface of the slide for sealing.
6) The staining condition and morphological change of the liver tissue section are observed under an optical microscope, fat is stained bright red, cell nucleus is stained dark blue, and other tissues are stained light blue. The observation results are shown in FIG. 5.
As can be seen from fig. 5, compared to the control group, the liver cells of both wild type mice fed with high fat diet for 2 months and NAFLD mice knocked out with Nrf2 showed different degrees of nuclear cavitations and steatosis, and the steatosis-occurring liver cells showed different sizes, were rounded, had a clear outline and were stained with red lipid droplets in the cytoplasm. Meanwhile, nuclear caverns and steatosis in liver cells of mice in the NAFLD (Nrf2-/-) group were more obvious compared with the NAFLD (Nrf2+/+) group. The above results illustrate that: high fat feed for 2 months can cause liver lesions of wild type and Nrf2 knockout mice to occur to different degrees, and deletion of Nrf2 can aggravate the degree of liver injury of the mice caused by the same.
In conclusion, combining the results of biochemical indexes of the liver, liver damage and liver proportion analysis, HE staining and oil red O staining of the mouse, it can be fully proved that the NAFLD model of the wild type mouse and the Nrf2 knockout mouse can be successfully constructed by the method of feeding the high-fat feed for 2 months.
2. Therapeutic effects of EA and MoA on NAFLD and mechanism exploration thereof
2.1 grouping, administration and sample Collection of laboratory animals
Wild type (Nrf2+/+) and Nrf2 knockout homozygote (Nrf2-/-) C57BL/6 mice obtained by breeding in the laboratory are respectively selected for 30 mice, the mice with the same genotype are respectively and randomly divided into 5 groups, and each group comprises 6 mice and male and female halves. After adaptive feeding for 1 week with 12h illumination and 12h darkness, feeding with high fat feed for 2 months. Administration was started after 2 months, and the groups and schedule are shown in tables 2-2, with the administration schedule being 1 week consecutively for intragastric administration of EA, MoA or CUR, respectively, once daily.
Table 2-2 grouping and dosing schedule of mice (n ═ 6).
Figure BDA0002153202320000121
Figure BDA0002153202320000131
Because EA, MoA and CUR are all difficult to dissolve in water, in order to avoid the inaccurate intragastric dose caused by the uneven dispersion of the medicines, the three medicines are solubilized by adopting an emulsification mode, and the formula of the emulsion is shown in tables 2-3.
Table 2-3 emulsion formulations.
Figure BDA0002153202320000132
After 2 months of breeding, 10% chloral hydrate (0.5g/kg) is injected into the abdominal cavity of the mouse for anesthesia, after the anesthesia is successful, the blood and perfusion of the heart are rapidly carried out, the liver is taken out, part of liver tissue cleaned by normal saline is put into 4% paraformaldehyde fixing solution for soaking, and the rest of liver tissue is stored at-70 ℃ for standby. The whole blood was placed in a clean 1.5mL centrifuge tube, allowed to stand overnight at 4 ℃ and centrifuged at room temperature for 10min (4000rpm) to isolate serum. The body weight and liver weight of each group of mice were recorded.
2.2 Biochemical index analysis of mouse liver function
2.2.1 sample treatment
The operation is the same as under item "1.2.1".
2.2.2 data processing
The operation is the same as under item "1.2.2".
2.2.3 results of the experiment
The results of the data for the biochemical markers ALT and AST of liver function are shown in FIGS. 6 and 7, respectively.
As can be seen from the results of fig. 6 and 7, compared with the blank control group with the same genotype, the concentrations of ALT and AST in the serum of the mice in the NAFLD group are both significantly increased, and the difference has statistical significance (p <0.05), indicating that the NAFLD mouse model is successfully constructed. After continuing to gavage mice models EA, MoA and CUR, respectively, the serum ALT and AST concentrations in wild-type NAFLD mice models were reduced to different degrees compared to the simple NAFLD group of the same genotype. The changes were statistically significant (p <0.05) except for ALT in the NAFLD + EA (Nrf2+/+) group of mice. However, for Nrf2 knockout mice, there was no significant change in serum concentrations of ALT and AST following continued gavage of mouse models EA, MoA, and CUR, respectively. The above results illustrate that: intragastric administration of EA, MoA and CUR reduced ALT and AST concentrations in serum of wild-type NAFLD mouse models to varying degrees, and Nrf2 played an important role in this process.
2.3 mouse liver injury and liver tissue section staining
2.3.1 mouse liver injury
After the thoracic cavity of the mouse is opened, the appearance characteristics of the liver of the mouse are visually observed, and the result of the liver injury of the mouse is shown in fig. 8.
As can be seen from the results of fig. 8, the gall bladder of the mice in the NAFLD group was significantly enlarged compared to the control group of the same genotype, indicating that the NAFLD mouse model was successfully constructed. After 1 week of gavage with EA, MoA and CUR, the gall bladder was significantly smaller in mice of the wild type NAFLD group compared to the NAFLD only group of the same genotype, but not much changed in Nrf2 knockout mice. The results show that: the gall bladder swelling condition of a wild type NAFLD mouse model can be obviously relieved by intragastric administration of EA, MoA and CUR for 1 week, and Nrf2 plays an important role in the process.
2.3.2 mouse liver ratio Re-analysis
The operation is the same as under item "1.3.2". The results of the liver gravity analysis of the mice are shown in FIG. 9.
As can be seen from the results in fig. 9, compared with the control group with the same genotype, the liver ratio of the mice in the NAFLD group is significantly increased, and the difference has statistical significance (p <0.01), indicating that the NAFLD mouse model is successfully constructed. After 1 week of gavage administration of EA, MoA and CUR, liver specific gravity of mice in the wild type NAFLD group tended to be significantly lower than that of the NAFLD group alone of the same genotype, and the difference between EA and CUR treated group was statistically significant (p <0.05), but there was not much change in the Nrf2 knockout mouse group. The results show that: gavage administration of EA, MoA and CUR for 1 week significantly reduced liver specific gravity in wild type NAFLD mouse models, and Nrf2 played an important role in this process.
2.3.3 HE staining analysis
The operation is the same as under item "1.3.3". The results of analysis of HE stained lesions in mouse liver are shown in fig. 10.
As can be seen from the results in fig. 10, compared with the control group with the same genotype, the mouse liver cells of NAFLD group all showed different degrees of vacuolar degeneration, steatosis and inflammatory cell infiltration, indicating that NAFLD mouse model was successfully constructed. Meanwhile, hepatic cells of mice in the NAFLD (Nrf2-/-) group also show different degrees of punctate necrosis, which indicates that the deletion of Nrf2 can increase the severity of NAFLD. The phenomena of vacuolar degeneration, steatosis and inflammatory cell infiltration in the hepatocytes of mice in the wild-type NAFLD group were improved compared to the NAFLD-only group of the same genotype 1 week after gavage administration of EA, MoA and CUR, but the phenomena were not much changed in the Nrf2 knockout group. The results show that: intragastric administration of EA, MoA and CUR for 1 week improved lesions such as vacuolar degeneration, steatosis and inflammatory cell infiltration that occurred in liver cells of a wild-type NAFLD mouse model, and Nrf2 played an important role in this process.
2.3.4 oil Red O staining analysis
The operation is the same as under item "1.3.4". The results of mouse liver oil red O staining are shown in fig. 11.
From the results in fig. 11, it can be seen that the liver cells of the NAFLD mice showed different degrees of nuclear lacunae and steatosis compared to the control group with the same genotype, indicating that the NAFLD mouse model was successfully constructed. After 1 week of gavage administration of EA, MoA and CUR, the phenomenon of nuclear voiding and steatosis in hepatocytes of mice in the wild type NAFLD group was improved compared to the simple NAFLD group of the same genotype, but the phenomenon was not much changed in the Nrf2 knockout group. The results show that: intragastric administration of EA, MoA and CUR for 1 week improved the nuclear voiding and steatosis phenomena occurring in liver cells of the wild type NAFLD mouse model, and Nrf2 played an important role in this process.
2.4 Effect of EA and MoA on GSH levels in liver tissues of mice
The glutathione test kit was used to detect the levels of GSH in mouse liver tissue, the principle of which was as follows:
upon reaction of dithiodinitrobenzoic acid with a mercapto compound, a yellow compound is produced which can be determined colorimetrically.
2.4.1 reagent preparation
1) A first reagent: 1 bottle of powder A and 1 bottle of 50mL of liquid B, and storing for 6 months at 4 ℃. When the powder A is used for the first time, 170mL of UP water preheated to 90-100 ℃ is added into the powder A to be fully dissolved.
2) And a second reagent: the powder is added into 1 bottle, 200mL UP water is added when the powder is used, the solution is fully dissolved, and the powder can be stored for 6 months in a sealing way at room temperature.
3) And (3) reagent III: adding 50mL UP water into 1 powder, dissolving completely, sealing at room temperature, and storing in dark for 6 months.
4) And (4) reagent IV: 1 powder is added with 10mL UP water when in use, fully dissolved and can be stored for 5 days in a sealed and refrigerated way in the dark.
5) And a fifth reagent: GSH standard powder 3.07mg, and storing at 4 deg.C for 6 months.
6) Reagent six: the GSH standard substance solvent stock solution is 10mL, and is stored for 6 months at 4 ℃.
7) Preparation of reagent I application liquid: mixing the prepared solution A and solution B. The solution is supersaturated, and can precipitate after standing and cooling at room temperature, and the supernatant is taken for experiment. The solution can be stored at room temperature for 6 months under sealed condition.
8) Preparation of GSH standard solvent application liquid: diluting according to the ratio (volume ratio) of GSH standard solvent stock solution and UP water to 1:9, and preparing for use.
9) Preparation of 1mM GSH standard solution: before each measurement, 3.07mg of GSH standard substance is added into 10mL of GSH standard substance solvent application liquid, and the mixture is fully and uniformly mixed to be prepared for use.
10) Preparation of 20 μ M GSH standard solution: 0.2mL of 1mM GSH standard solution is added into the application solution of the 9.8mLGSH standard solvent and is prepared for use.
2.4.2 sample treatment
1) Preparation of 10% homogenate: accurately weighing the tissue, mechanically homogenizing under ice-water bath condition according to the ratio of the tissue weight (g) to the volume of physiological saline (mL) to 1:9, centrifuging at 2500rpm at 4 ℃ for 10min, and taking the supernatant for determination.
2) Preparation of the supernatant: taking 0.5mL of the supernatant of the 10% homogenate obtained above, adding 2mL of reagent-application liquid, mixing well, centrifuging at 4000rpm for 10min at 4 ℃, taking 1mL of the supernatant, and performing a display reaction.
3) And (3) color development reaction: corresponding reagents were added according to tables 2-4 to prepare blank, standard and assay tubes, respectively. After the preparation, the mixture is fully mixed, kept stand for 5min at room temperature, adjusted to zero by UP water at 420nm and the absorbance value of each tube is measured.
TABLE 2-4 solution composition of color reaction
Figure BDA0002153202320000161
Figure BDA0002153202320000171
2.4.4.3 data processing
Calculating the content of GSH in the tissue according to the formula:
Figure BDA0002153202320000172
the dilution ratio before sample test was equal to the dilution ratio (5-fold) in the supernatant preparation process. And detecting the protein content in a 10% mouse liver homogenate sample by using a BCA kit.
The obtained data are sorted and counted by using Microsoft Excel 2019, the experimental result is subjected to one-factor analysis of variance (ANOVA) by using Graphpad prism 7 software, and the difference has statistical significance when the p is less than 0.05.
2.4.4 results of the experiment
The results of the GSH concentration in the liver tissues of the groups of mice obtained according to the above procedure are shown in fig. 12. .
As can be seen from the results of fig. 12, the levels of GSH in liver tissues of mice of the NAFLD model group were all significantly decreased compared to the control group of the same genotype, and the difference was statistically significant (p < 0.05). After 1 week of gavage administration of EA, MoA and CUR, GSH levels were increased in liver tissues of mice of the wild type NAFLD group to varying degrees compared to the NAFLD-only group of the same genotype and the changes were statistically significant (p <0.05), but not too great for the Nrf2 knockout mouse group. The results show that: gavage administration of EA, MoA and CUR for 1 week significantly increased GSH levels in liver tissues of wild type NAFLD mouse models, and Nrf2 played an important role in this process.
2.5 Effect of EA and MoA on mRNA levels of various genes of interest in liver tissues of mice
2.5.1 design and Synthesis of primers
Mouse-derived Nrf2, Keap1 (Kelch-like epichlorohydrin-related protein 1), Ho-1, Nqo1, Mrp2, Mrp4, Cyp2e1, and Gapdh mRNA NCBI Reference Sequences Reference numbers were obtained from NCBI's Gene (https:// www.ncbi.nlm.nih.gov/Gene /), and qPCR primers were designed via Integrated DNAtechnologies (IDT, https:// sg. idtdna. com/pages) website, and the primer Sequences are shown in tables 2-5.
TABLE 2-5 qPCR primer sequences for mouse Nrf2, Keap1, Ho-1, Nqo1, Gapdh, Mrp4, Cyp2e1, and Mrp2
Figure BDA0002153202320000181
Figure BDA0002153202320000191
2.5.2 tissue Total RNA extraction
Various consumables required by the experiment are soaked in 0.1% DEPC water for 24-48 h, and are dried for use after high-temperature and high-pressure sterilization. The operation steps are as follows:
1) the frozen mouse liver tissue is restored to the room temperature, the excess water on the tissue is sucked dry by using clean filter paper, precooled normal saline is added according to the proportion of the tissue weight (g) to the normal saline volume (mL) of 1:1, a sample is placed on ice, a hand-held electric tissue homogenizer is used for homogenizing, and the homogenization is carried out for 10s at intervals of 10-15 s until a uniform system is obtained.
2) To 500. mu.L TRNzol was added 50. mu.L of the homogenate and vortexed until no significant precipitation occurred. Standing at room temperature for 5 min.
3) Centrifuge at 4 ℃ for 5min (12000 rpm).
4) The supernatant obtained after centrifugation was added to 100. mu.L of chloroform, vortexed and shaken until the solution was sufficiently emulsified, and allowed to stand at room temperature for 3 min.
5) Centrifuge at 4 ℃ for 15min (12000 rpm).
6) After the centrifugation was completed, the transparent liquid at the uppermost layer of the three layers was transferred to a 1.5mL centrifuge tube.
7) Add 500. mu.L of isopropanol to the tube, mix well and let stand at room temperature for 10 min.
8) Centrifuge at 4 ℃ for 10min (12000 rpm).
9) The supernatant was discarded, 700. mu.L of 75% ethanol was added, and the tubes were washed by inversion.
10) Centrifuge at 4 ℃ for 5min (7500 rpm).
11) Absorb ethanol in spite of the fact that the ethanol is dissolved in the water.
12) The sample was dried for 5min and the precipitate was dissolved with 20 μ LRNasefree water.
13) RNA concentration and OD260/280 values of each tissue sample were determined using Nanodrop 2000. The final RNA concentration of each sample was adjusted to about 1. mu.g/mL and stored at-70 ℃ until use.
2.5.3 Synthesis of cDNA
By using
Figure BDA0002153202320000201
II 1st Strand cDNA Synthesis Supermix for qPCR kit was used to synthesize cDNA by reverse transcription of RNA from each sample obtained under section "2.4.5.2". The details of the specific procedures and volumes of reagents added are described in the kit instructions (https:// www.yeasen.com/products/detail/105). The resulting samples were then stored at-20 ℃ until use.
2.5.4 fluorescent polymerase chain reaction (Real-time Quantitative PCR, qPCR)
By using
Figure BDA0002153202320000202
The cDNA sample obtained under section "2.4.5.3" was subjected to the qPCR SYBR Green Master Mix (No Rox) kitAnd (4) performing fluorescence quantitative detection. The details of the specific procedures and volumes of reagents added are described in the kit instructions (https:// www.yeasen.com/products/detail/861). And (3) putting the eight connecting pipes containing the prepared samples into a fluorescent quantitative PCR instrument for amplification. The three-step amplification procedure recommended by the instruction was used. And collecting Ct values for calculation after the amplification is finished.
2.5.5 data processing
Data sorting was performed using Microsoft Excel 2019, calculation 2△△CtTo compare the differences in expression of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4mRNA in liver tissues of mice in each experimental group. Experimental results Single-factor analysis of variance (ANOVA), p, was performed using Graphpad prism 7 software<The difference is statistically significant at 0.05.
2.5.6 results of the experiment
The expression of mRNA of Keap1, Cyp2e1, Nrf2, Ho-1, Nqo1, Mrp2 and Mrp4 in liver tissues of each group of mice is shown in FIG. 13.
As can be seen from the results in fig. 13, mRNA levels of Nrf2 were significantly increased in the liver of the wild-type NAFLD mouse model after 2 months of high fat diet feeding compared to the control group, and the difference was statistically significant (p <0.05), indicating: NAFLD induces mRNA expression of Nrf2 in the liver of wild type mice. After EA, MoA and CUR treatments were given, Nrf2 mRNA levels in the liver of wild-type NAFLD mouse models were significantly increased compared to both control and NAFLD alone of the same genotype, with statistical differences (p <0.01), but there was no significant change in Nrf2 mRNA levels in the liver of Nrf2 knockout mice; similarly, in wild-type NAFLD mouse model livers treated with EA, MoA and CUR, respectively, the downstream gene of Nrf 2: nqo1, Ho-1, Mrp2 and Mrp4 have different degrees of mRNA level increase, the difference has statistical significance (p is less than 0.05), and the variation trend is similar to that of Nrf2, but the variation rule is not obvious in the liver of an Nrf2 knockout mouse. The above results illustrate that: EA. MoA, like CUR, is an agonist of Nrf 2.
Compared with the blank control group, the mRNA levels of Cyp2e1 in wild-type and Nrf2 knockout NAFLD mouse model livers were significantly increased on average, the difference was statistically significant (p <0.05), and there was not much change after EA, MoA and CUR treatment, indicating that NAFLD can indeed induce the expression of Cyp2e1 in mouse livers, and EA, MoA and CUR did not have much influence on the mRNA levels of Cyp2e 1.
After the above treatments, no significant changes in the levels of Keap 1mRNA were observed in the livers of the groups of mice.
2.6 Effect of EA and MoA on levels of proteins of genes of interest in liver tissues of mice
2.6.1 preparation of solution
1) RIPA protein lysate: 1mL RIPA lysine Buffer + 10. mu.L PMSF, placed on ice for use.
2)5 XSDS-PAGE buffer: 0.5g SDS, 0.15g Tris-HCl, 25mg BPB and 2.5mL glycerol were added to 2.5mL UP water, respectively, and then mixed well.
3)1640 complete medium: the medium powder, 2.435g NaHCO3, 100mg streptomycin, 10000U penicillin and 3.571g HEPES were added to 900mL UP water, respectively, and then sufficiently dissolved and mixed. Filtering with 0.22 μm filter, packaging, sealing, and storing. Before use, 10% (volume ratio) fetal bovine serum was added.
4) Co-IP (Co-immunoprecipitation) lysis buffer: 0.5. mu.L of protease inhibitor cocktail, 1. mu.L of PMSF and 500. mu.L of NP40 lysate were mixed on ice until ready for use (this is the amount used to prepare a 10cm dish of cells).
5) 0.25% pancreatin: 0.25% pancreatin: adding 20mg of EDTA-2Na into 100mL of PBS, dissolving by ultrasonic, adding 250mg of pancreatin powder, shaking up gently, filtering by using a 0.22 mu m filter membrane after fully dissolving, and subpackaging for later use.
6)1 × electrophoresis buffer: 3g of Tris-HCl, 1g of SDS and 14.4g of glycine are respectively added into 1L of UP water, then the mixture is fully mixed and stored at room temperature for standby.
7)1 × transfer membrane buffer: 14.4g of glycine, 3g of Tris-HCl and 100mL of methanol are respectively added into 900mL of UP water, fully mixed and stored at room temperature for standby.
8) TBS buffer: adding 8g NaCl and 2.42g Tris-HCl into 1L UP water respectively, fully mixing uniformly, and storing at room temperature for later use.
9) TBST buffer: adding 8g NaCl, 2.42g Tris-HCl and 1mL Tween-20 into 1L UP water respectively, fully mixing uniformly, and storing at room temperature for later use.
2.6.2 extraction of Total tissue protein and sample preparation
The method adopts RIPA Lysis Buffer to perform Lysis on mouse liver tissues, and comprises the following specific steps:
1) the frozen mouse liver tissue is restored to the room temperature, the excess water on the tissue is sucked dry by using clean filter paper, precooled normal saline is added according to the proportion of the tissue weight (g) to the normal saline volume (mL) of 1:1, a sample is placed on ice, a hand-held electric tissue homogenizer is used for homogenizing, and the homogenization is carried out for 10s at intervals of 10-15 s until a uniform system is obtained.
2) To 200. mu.L of RIPA lysate containing PMSF was added 50. mu.L of the liver tissue homogenate obtained above. (the final concentration of PMSF was 1 mM).
3) Each tissue sample was vortexed for 30 seconds and then placed on ice, and vortexed for 30 seconds after standing for 3min, for a total of 30 min.
4) Centrifuge at 4 ℃ for 10min (10000 rpm).
5) Protein quantification was performed on each sample using the BCA kit.
6) The appropriate PBS and 5 × loading buffer were added to the sample and mixed well to give a final protein concentration of 10 μ g/. mu.L.
7) Placing each obtained sample in boiling water for 10min, and storing at-70 deg.C.
2.6.3 Western blotting test (Western Blot)
Western blot assays were performed using the Bio-Rad Mini-Protean System. The operation steps are as follows:
1) preparation of SDS-PAGE gels: 12% separation gel (5 mL/gel) and 4% concentration gel (2 mL/gel) were formulated according to gel formulation instructions.
2) Electrophoresis: and adding 20 mu L of sample into each sample loading hole, performing constant voltage electrophoresis at 90V until different bands appear in a Marker lane, increasing the electrophoresis voltage to 120V, and continuing the electrophoresis until a satisfactory band distance is obtained.
3) Film transfer: cutting a PVDF membrane with a proper size, and soaking in methanol for 1 min; soaking the PVDF membrane, the filter paper, the SDS-PAGE gel and the sponge in a membrane transferring buffer solution in a certain sequence, and then placing the membrane transferring buffer solution in a plastic splint for transferring the membrane; thirdly, the assembled plastic splint is placed in an electrophoresis tank filled with the membrane transferring liquid. Film transferring conditions: constant current 300mA, 60 min.
4) And (3) sealing: putting the PVDF membrane subjected to membrane transfer into a TBST solution containing 2% BSA, and blocking at 37 ℃ for 1-3 h (80 rpm).
5) Primary antibody incubation: the blocked PVDF membrane was placed in an antibody incubation cassette containing a primary anti-diluent (diluted with 2% BSA). Shaking the shaker at 4 ℃ overnight.
6) And (3) secondary antibody incubation: rinsing the incubated PVDF membrane of the primary antibody for 3 times by TBST, 5min each time; ② placing the PVDF membrane into an antibody incubation box filled with a proper amount of secondary antibody diluent (diluted by TBST), and shaking for 90min at the temperature of 4 ℃ by a shaking table.
7) Rinsing: the PVDF membrane incubated with the secondary antibody was rinsed 3 times with TBST and 2 times with TBS, each time for 5 min.
Developing and photographing: and taking out the cleaned PVDF membrane, sucking the redundant liquid on the membrane by using a clean paper towel, dripping ECL developing solution (solution A: solution B is 1:1) on the membrane, developing on a gel imager after the ECL developing solution is uniformly soaked on the PVDF membrane, and collecting a developed image through SageCapture.
2.6.4 data processing
The resulting protein bands were subjected to gray scale analysis using LANE1D software.
The data were collated using Microsoft Excel 2019 to compare the expression differences of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4 proteins in liver tissues of mice in each experimental group. Experimental results single-factor analysis of variance (ANOVA) was performed using Graphpad prism 7 software, with differences of statistical significance when p < 0.05.
2.6.5 results of the experiment
The protein expression of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4 in liver tissues of each group of mice is shown in fig. 14.
As can be seen from the results of fig. 14, in the wild-type NAFLD mouse model liver after 2 months of high fat diet, the level of Nrf2 was significantly increased compared to the control group, and the difference was statistically significant (p <0.05) because: NAFLD induces expression of Nrf2 protein in the liver of wild type mice. The protein level of Nrf2 in the liver of the wild-type NAFLD mouse model was significantly increased after EA, MoA and CUR treatments, respectively, compared to both the control and NAFLD alone groups of the same genotype, with statistical significance for the difference (p <0.01), but there was no significant change in the protein level of Nrf2 in the liver of Nrf2 knockout mice; similarly, in wild-type NAFLD mouse model livers treated with EA, MoA and CUR, respectively, the downstream gene of Nrf 2: the protein levels of Mrp2, Mrp4, Nqo1 and Ho-1 were all increased to different degrees, the differences were statistically significant (p <0.05), and showed a similar trend to Nrf2, but there was no obvious change in the liver of Nrf2 knockout mice. The above results illustrate that: EA. MoA, like CUR, is an agonist of Nrf 2.
In wild-type and Nrf2 knockout NAFLD mouse model livers, Cyp2e1 protein levels were significantly increased, the differences were statistically significant (p <0.05), and there were not much change after EA, MoA, and CUR treatment, compared to the blank control group, indicating that NAFLD can indeed induce Cyp2e1 expression in mouse livers, and EA, MoA, and CUR did not have much effect on Cyp2e1 protein levels.
The experiments show that EA and MoA are really agonists of Nrf2, and the EA and MoA are likely to up-regulate the expression of downstream antioxidant proteins, II-phase detoxification enzymes and transporters thereof by inducing Nrf2, thereby playing a role in resisting oxidative stress reaction, inflammatory reaction and the like caused by NAFLD.
Example 2, in vitro cell model validation of therapeutic effects of EA and MoA on NAFLD via Nrf2 pathway the sources of the drugs in this example were the same as above except for the following:
reactive Oxygen Species (ROS) test box (E004, buika, nanjing); fatty acid-free-BSA (B2064, SIGMA, usa); oleic acid (O1383, SIGMA, usa); palmitic acid (P0500, SIGMA usa).
1. Establishment and verification of NAFLD cell model
1.1 solution preparation
1)1640 complete medium: the same as in example 1.
2) DMEM complete medium: the same as in example 1.
3) 0.25% pancreatin: the same as in example 1.
4) PBS solution: a bag of PBS powder is taken, 1L of UP water is added according to the instruction requirements, and the mixture is fully and uniformly mixed for standby.
5) 20% fatty acid-free-BSA solution: 0.6g of fatty acid-free BSA is weighed and placed in a 50mL centrifuge tube, 3mL of PBS solution preheated to 55 ℃ is added, and the mixture is directly centrifuged at 8000rpm at room temperature (about 25 ℃) for 20min to be completely dissolved, so that a brown yellow clear solution is obtained. Care was taken that shaking or mixing was contraindicated immediately upon addition of the PBS solution, which would cause the BSA to form a lumpy crystal that would not dissolve.
6) 40% fatty acid-free-BSA solution: weighing 1.2g of fatty acid-free BSA, placing in a 50mL centrifuge tube, adding 3mL of PBS solution preheated to 55 ℃, and directly centrifuging at 8000rpm for 20min at room temperature to completely dissolve to obtain a brownish yellow clear solution.
7)0.1M NaOH solution: 0.04g NaOH powder is weighed, 10mL UP water is added, and the mixture is fully and uniformly mixed, thus obtaining 10mL 0.1M NaOH solution.
8)20mM oleic acid solution: 19.04. mu.L of oleic acid was measured at 25 ℃ using a pipette and added to 3mL of 0.1M NaOH solution, followed by sufficient saponification in a 75 ℃ water bath for about 30min until a colorless, clear solution was obtained, i.e., 20mM sodium oleate saponified solution. The incubated sodium oleate saponification solution was added rapidly to 3mL of 20% fatty acid-free BSA solution to give 6mL of 20mM oleic acid + 20% BSA solution. Mixing, and dissolving at 55 deg.C for 30min to obtain brown yellow clear sample solution with stable properties, no solid or flocculent impurities, and no difference from original BSA solution. The solution was filtered (0.22. mu.M) in a clean bench, sealed and stored at 4 ℃ for a long period of time.
9)40mM palmitic acid solution: 0.0307g of palmitic acid was weighed into 3mL of 0.1M NaOH solution, and then placed in a 75 ℃ water bath for sufficient saponification for about 30min until a colorless transparent clear solution was obtained, i.e., 40mM sodium palmitate saponification solution. The incubated sodium palmitate saponification solution was added rapidly to 3mL of 40% fatty acid-free BSA solution to give 6mL of 40mM oleic acid + 40% BSA solution. Mixing, and dissolving at 55 deg.C for 30min to obtain brown yellow clear sample solution with stable properties, no solid or flocculent impurities, and no difference from original BSA solution. The solution was filtered (0.22. mu.M) in a clean bench, sealed and stored at 4 ℃ for a long period of time.
10) Standard investigation solution: 3mL of 20% fatty acid-free BSA solution was mixed well with 3mL of 0.1M NaOH solution, filtered (0.22. mu.M) in a clean bench, sealed and stored at 4 ℃ for a long period of time. Used for checking the influence of BSA, pH value and other substances in the solution on cells.
11) Free fatty acid stocks (Free fatty acids, FFAs): stock solutions of 4mM FFAs were prepared at a ratio of 20mM oleic acid solution to 40mM palmitic acid solution to 4:1, sealed and stored at 4 ℃ for a long period of time.
1.2 cell lines and cell cultures
THLE-3 cells, purchased from Shanghai cell biology institute of Chinese academy of sciences, were cultured in 1640 complete medium.
HepG2 cells were purchased from Shanghai institute of cell biology, Chinese academy of sciences and cultured in DMEM complete medium.
1.3 establishment and validation of NAFLD cell model
1.3.1 cell administration
Respectively mixing L02, THLE-3 and HepG2 cells at a ratio of 1 × 106And (3) inoculating the cells/mL into a six-hole plate, when the cell growth area accounts for about 80% of the total area of the culture dish, respectively carrying out administration treatment by using a blank culture medium (blank control) containing a standard examination solution and a drug-containing culture medium of 100/500/1000 mu M FFAs, placing the six-hole plate after administration into a cell culture box, continuously culturing for 24 hours, and taking out for carrying out subsequent experiments.
1.3.2 validation of NAFLD cell model by oil Red O staining
1) Sample processing
Firstly, oil red O dyeing application liquid is prepared according to the method in the embodiment 1.
② use PBS 6 hole plate cells gently rinse 3 times, completely residual liquid. Add 200. mu.L of oil Red O staining solution to each well and stain for 15 min. Followed by rinsing with UP water at around 37 c for 15 s.
And thirdly, sucking UP the UP water, adding 200 mu L of re-dyeing liquid into each hole, continuously dyeing for 3-5 min, and then rinsing for 30-60 s by using the UP water.
Adding a proper amount of UP water seal, and observing the dyeing condition and morphological change of the cells under an inverted optical microscope as soon as possible.
2) Results of the experiment
The results of L02, THLE-3 and HepG2 cell oil red O staining obtained according to the above procedure are shown in FIG. 16.
As can be seen from the results of oil red O staining in FIG. 16, after 24h of FFAs treatment at 100, 500 and 1000. mu.M, lipid droplets with different sizes, circles, clear outlines and red staining appeared in the cytoplasm of L02, THLE-3 and HepG2 cells, indicating that the NAFLD hepatocyte models of the three cell lines were successfully constructed. Meanwhile, when the treatment concentration of the FFAs is 500 mu M, red lipid drops in cytoplasm are dense and well-defined, so that the FFAs with the concentration of 500 mu M is selected as a treatment condition for subsequently constructing the NAFLD hepatocyte model.
2. Effect of EA and MoA on NAFLD human hepatocyte model
2.1 Effect of EA and MoA on L02, THLE-3 and HepG2 cell viability
MTT experiments were performed on L02, THLE-3 and HepG2 cells, and the effect of different concentrations of EA, MoA and CUR (positive control, Nrf2 agonist) on the viability of L02, THLE-3 and HepG2 cells after 24h treatment was examined. The results are shown in FIG. 17.
The MTT experimental result shows that after 24 hours of treatment, the cell viability of the L02 cells treated by 0-100 mu M EA, 0-25 mu M MoA and 0-25 mu M CUR is more than 90%, which indicates that EA, MoA and CUR have no obvious growth inhibition effect on L02 cells in the concentration ranges, so that 50 mu MEA, 100 mu MEA, 10 mu MMoA and 10 mu M CUR are selected as the treatment conditions of the subsequent L02 cells. Similarly, after 24 hours of treatment, the cell viability of the THLE-3 cells treated by 0-100 mu M EA, 0-50 mu M MoA and 0-25 mu M CUR is more than 90%, which indicates that EA, MoA and CUR have no obvious growth inhibition effect on the THLE-3 cells in the concentration ranges, so 50,100 mu M EA, 25, 50 mu M MoA and 10 mu M CUR are selected as the treatment conditions of the subsequent THLE-3 cells. Similarly, 50, 100. mu.M EA, 10, 25. mu.M MMoA and 10. mu.M CUR were selected as the treatment conditions for the subsequent HepG2 cells.
2.2 Effect of EA and MoA on ROS levels in NAFLD cell models
2.2.1 administration of cells
Respectively mixing L02, THLE-3 and HepG2 cells at 5X 105Inoculating the cells in a 12-well plate at a density of one/mL, respectively performing administration treatment by using a blank culture medium (blank control) containing a standard examination solution and a 500-mu M FFAs (fringe field acting as a drug) medicine-containing culture medium when the cells are attached to the wall and grow to 70-80% fusion, and placing the 12-well plate after administration in a cell culture box for further culture for 24 hours. Taking out the 12-hole plate, adding 0.1% DMSO (blank control), 50 μ M EA, 100 μ M EA, 10 μ M MMoA and 10 μ M CUR into the culture medium to treat L02 cells; ② adding 0.1 percent DMSO (blank control), 50,100 mu M EA, 25, 50 mu M MMoA and 10 mu M CUR into the culture medium respectively to carry out drug administration treatment on the THLE-3 cells; ③ 0.1 percent DMSO (blank control), 50,100 mu M EA, 10, 25 mu M MMoA and 10 mu M CUR were added into the culture medium respectively to carry out drug administration treatment on HepG2 cells. Then placing the mixture in a cell culture box to continue culturing for 24 h.
2.2.2 sample treatment
1) Adding a fluorescent probe: an appropriate amount of 10mM DCFH-DA was added to each well to a final concentration of 500 nM. An equal amount of DMSO was added as a blank. An appropriate amount of 12mM active oxygen donor was added to give a final concentration of 20. mu.M, which served as a positive control. The 12-well plate was placed in a cell incubator and incubated at 37 ℃ for 1 h.
2) Cell collection: the cells were digested with 0.25% trypsin, the digestion was stopped by adding the corresponding medium, a cell suspension was prepared, the cells were washed 2 times with PBS, and centrifuged at 1000rpm for 5min at room temperature. The cells were resuspended with the appropriate amount of PBS.
3) Fluorescence detection: 200. mu.L of the cell suspension obtained above was added to a 96-well plate using VarioskanTMThe LUX multifunctional microplate reader and the operating software perform the fluorescent inspection on the cell sample. Optimum excitation wavelength: 500 ± 15nm, optimal emission wavelength: 530 +/-20 nm.
4) The protein concentration of the cell suspension is detected by using a BCA kit, namely when the L02 cells in a 10cm culture dish grow to be about 80% fused, the cells are treated by adding the monomer with proper concentration for 24h, and then a Co-IP experiment is carried out. The specific operation steps are as follows:
a) the treated cells were removed, washed three times with pre-cooled PBS and the remaining liquid was aspirated.
b) And adding pre-cooled 500 mu L Co-IP lysis buffer solution on ice, taking out the solution every 1-3 min, and vortexing for 1min for 30min in total.
c) After lysis was complete, centrifugation was carried out at 4 ℃ for 15min (14000 rpm).
d) The supernatant from the centrifugation was transferred to a 1.5mL EP tube.
e) The UV absorbance was measured at 562nm using a BCA protein quantification kit according to the instructions (http:// www.wanleibio.cn/index. phpm. content & c. window & cat. 199& id. 798) using a microplate reader. The protein concentration of each sample was calculated from the standard curve calculated from the standard.
f) Adding a proper amount of PBS, diluting the total protein concentration to about 10 mug/muL, reserving a proper volume of cell lysate per tube as an Input control, and storing at-70 ℃ for later use.
g) 500 μ L of cell lysate from each sample was mixed with the appropriate volume of NRF2 primary antibody (according to antibody instructions) and the appropriate volume of IgG primary antibody was added to the negative control tube according to antibody instructions.
h) Each sample was supplemented with 2.5. mu.L protease inhibitor followed by shaking overnight at 4 ℃.
i) Preparation of Protein A/G magnetic beads: gently blow the Protein A/G magnetic bead stock solution with a tip with the front end cut off until the solution is uniformly dispersed, take out an appropriate volume and place in another 1.5mL EP tube.
j) The aspirated Protein A/G magnetic beads were washed twice with PBS, the magnetic beads were adsorbed using a magnetic rack, the supernatant was discarded, and then an appropriate volume of PBS was added as needed to prepare it to a 50% concentration for use.
k) Taking out the incubated cell lysate, adding 50% Protein A/G magnetic beads, mixing uniformly, and shaking at 4 ℃ for 2 h.
l) standing the sample tube on a magnetic frame for 2min, discarding the supernatant when the magnetic bead-antigen-antibody complex is agglomerated, adding an appropriate amount of precooled PBS for resuspension and mixing, and shaking in a shaking table at 4 ℃ for 5 min. This procedure was repeated three times
m) discarding the supernatant, resuspending and mixing the magnetic bead-antigen-antibody complex with an appropriate amount of 2 × Loading buffer, and heating in boiling water bath for 10 min.
n) taking out the Input sample stored at-70 ℃, returning to room temperature, adding a proper amount of 5 × Loading buffer, uniformly mixing, and putting the sample in a boiling water bath at 100 ℃ for boiling for 10 min.
o) centrifugation at 4 ℃ for 1min (12000rpm), supernatant was collected and stored at-20 ℃ until use.
2.2.3 data processing
The level of ROS in the cells is calculated using the fluorescence intensity of the unit protein, and the formula is as follows:
ROS fluorescence intensity in protein Unit ═ sampleIntensity of fluorescence-blankIntensity of fluorescence) SampleProtein concentration
The data were collated using Microsoft Excel 2019, the experimental results were analyzed for one-way variance (ANOVA) using Graphpad prism 7 software, and differences were statistically significant when p < 0.05.
2.2.4 results of the experiment
The ROS levels in the cells treated with different dosing regimens for L02, THLE-3 and HepG2 cells are shown in FIG. 18.
As can be seen from the results in fig. 18, ROS levels were significantly increased in L02, THLE-3 and HepG2 cells after 24h treatment with FFAs, the differences being statistically significant (p <0.01), indicating: oxidative stress damage is significantly increased in NAFLD cell models constructed from FFAs. After EA, MoA and CUR treatments, respectively, ROS levels in all three cell lines declined to different degrees compared to the NAFLD group alone, and some of the differences were statistically significant (p <0.05), but still higher than the ROS levels in the control group. The above results suggest: EA. MoA and CUR can reduce ROS level in NAFLD cell model, reduce oxidative stress injury in cell, and protect liver cell.
2.3 Effect of EA and MoA on GSH levels in NAFLD cell models
2.3.1 cell administration
The operation is carried out with reference to the above under item "2.2.1".
2.3.2 solution preparation
The procedure was followed as in example 1.
2.3.3 sample treatment
1) Taking out 12-well plate, digesting cells with 0.25% pancreatin, adding corresponding culture medium to terminate digestion, making cell suspension, washing cells with PBS for 2 times, centrifuging at 1000rpm for 3min at room temperature. Adding appropriate amount of UP water, and mixing well until it is transparent to obtain cell lysate.
2) And (3) taking 100 mu L of the cell lysate, adding 400 mu L of reagent application liquid, fully and uniformly mixing, centrifuging at 4000rpm and 4 ℃ for 10min, and taking 200 mu L of supernatant for display reaction.
3) And (3) color development reaction: corresponding reagents are added according to the proportion of tables 2-4, and a blank sample, a standard sample and a determination sample are respectively prepared.
4) After the preparation, the mixture is fully mixed, kept stand at room temperature for 5min, and 200 mu L of sample is added into a 96-well plate.
5) Using VarioskanTMThe absorbance of each sample was measured at 420nm using a LUX microplate reader. The instrument was zeroed using UP water.
6) The protein concentration of the cell suspension was measured using the BCA kit.
2.3.4 data processing
Calculating the content of GSH in the tissue according to the formula:
Figure BDA0002153202320000301
wherein the dilution factor before sample test is (5 times) in the supernatant preparation process and (5 times) in the cell lysate preparation process.
The obtained data are sorted and counted by using Microsoft Excel 2019, the experimental result is subjected to one-way analysis of variance (ANOVA) by using Graphpadprism 7 software, and the difference has statistical significance when p is less than 0.05.
2.3.5 results of the experiment
GSH levels in L02, THLE-3 and HepG2 cells after treatment with different dosing regimens are shown in FIG. 19.
As can be seen from the results in fig. 19, GSH levels were significantly reduced in L02, THLE-3 and HepG2 cells after 24h treatment with FFAs, the differences being statistically significant (p <0.01), indicating: there was a significant increase in GSH consumption in NAFLD cell models constructed from FFAs. After EA, MoA and CUR treatments, respectively, GSH levels in all three cell lines appeared to rise to different degrees compared to the NAFLD group alone, with some differences statistically significant (p <0.05), but still lower than the GSH levels in the control group. The above results suggest: EA. MoA and CUR can improve excessive consumption of GSH in NAFLD cell model, increase GSH level, and protect liver cell.
2.4 Effect of EA and MoA on mRNA levels of various genes of interest in NAFLD cell models
2.4.1 design and Synthesis of primers
mRNA NCBI Reference sequence Reference numbers for human NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1, and GAPDF were obtained from NCBI's Gene (https:// www.ncbi.nlm.nih.gov/Gene /), and qPCR primers were designed via the Integrated DNAtechnologies (IDT, https:// sg. idtna. com/pages) website, and the primer Sequences are shown in Table 3-1.
TABLE 3-1 qPCR primer sequences for humanized NRF2, KEAP1, HO-1, NQO1, CYP2E1, MRP2, GAPDF, and MRP 4.
Figure BDA0002153202320000311
Figure BDA0002153202320000321
2.4.2 cell administration
The operation is carried out with reference to the contents under "2.2.1".
2.4.3 Total RNA extraction from cells
Various consumables required by the experiment are soaked in 0.1% DEPC water for 24-48 h, and are dried for use after high-temperature and high-pressure sterilization. The operation steps are as follows:
1) the media in the 6-well plate was discarded, rinsed 3 times with PBS and excess liquid was aspirated.
2) mu.L of TRNzol was added to each well, and the mixture was allowed to stand at room temperature for 5 min.
3) The above liquid was transferred to a 1.5mL centrifuge tube and 50 μ L chloroform was added, vortexed until the solution was sufficiently emulsified, and allowed to stand at room temperature for 3 min.
4) Centrifuge at 4 ℃ for 15min (12000 rpm).
5) After the centrifugation was completed, the transparent liquid at the uppermost layer of the three layers was transferred to a 1.5mL centrifuge tube.
6) Add 200. mu.L of isopropanol to the tube, mix well and let stand at room temperature for 10 min.
7) Centrifuge at 4 ℃ for 10min (12000 rpm).
8) The supernatant was discarded, 700. mu.L of 75% ethanol was added, and the tubes were washed by inversion.
9) Centrifuge at 4 ℃ for 5min (7500 rpm).
10) Absorb ethanol in spite of the fact that the ethanol is dissolved in the water.
11) The sample was dried for 5min and the precipitate was dissolved with 20 μ LRNasefree water.
12) RNA concentration and OD260/280 values of each tissue sample were determined using Nanodrop 2000. The final RNA concentration of each sample was adjusted to about 1. mu.g/mL and stored at-70 ℃ until use.
2.4.4 Synthesis of cDNA
The procedure was followed as in example 1.
2.4.5 fluorescent polymerase chain reaction
The procedure was followed as in example 1.
2.4.6 data processing
The procedure was followed as in example 1.
2.4.7 results of the experiment
mRNA expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1 in L02, THLE-3, and HepG2 cells treated with different dosing regimens is shown in FIG. 20.
As can be seen from the results in FIG. 20, the mRNA level of NRF2 in L02, THLE-3 and HepG2 cells was increased to a different extent than that in the control group after FFAs treatment for 24h, and some of the differences were statistically significant (p < 0.05); NRF2 mRNA levels were significantly increased in all three cell lines after different concentrations of EA, MoA and CUR treatment, respectively, with statistical differences (p <0.05), and EA and MoA concentration-dependent upregulation of NRF2, compared to the control and NAFLD alone. The above results suggest: NAFLD increases mRNA levels of NRF2 in three liver cell lines; EA and MoA are likely agonists of NRF2, and their upregulation is concentration dependent.
Similarly, the genes downstream of NRF2 in three cell lines after FFAs, EA, MoA and CUR treatments, respectively: the mRNA levels of NQO1, HO-1, MRP2 and MRP4 all increased to different degrees and showed similar trend to NRF2, with some differences being statistically significant (p < 0.05).
After FFAs treatment, CYP2E1mRNA level in L02, THLE-3 and HepG2 cells is increased by 2-4 times compared with that in a control group, the difference has statistical significance (p is less than 0.05), and the difference is not changed too much after EA, MoA and CUR treatment, which indicates that NAFLD can really induce CYP2E1 expression in hepatocytes, and EA, MoA and CUR have no great influence on CYP2E1mRNA level.
There was no significant change in KEAP 1mRNA levels in the three cell lines after FFAs, EA, MoA and CUR treatments, respectively.
2.5 Effect of EA and MoA on levels of proteins of Gene of interest in NAFLD cell models
2.5.1 preparation of solution
The procedure was followed as in example 1.
2.5.2 administration of cells
Respectively mixing L02, THLE-3 and HepG2 cells at a ratio of 1 × 106Inoculating the cells in a 6-well plate at a density of one/mL, and performing cell adherence and cell growth until 70-80% fusion is achieved according to the condition of 2.2.1The operation is carried out.
2.5.3 extraction of Total cellular protein and sample preparation
The cell Lysis is carried out by RIPA Lysis Buffer, and the specific steps are as follows:
1) the media in the 6-well plate was discarded, rinsed 3 times with PBS and excess liquid was aspirated.
2) mu.L of RIPA lysate containing PMSF (final concentration of PMSF is 1mM) was added to each well.
3) The 6-hole plate is placed on ice after being oscillated and vortexed for 30s, and the oscillating and vortexing are continued for 30s after standing for 3min, and the operation is carried out for 30min in total.
4) The cell lysate obtained above was transferred to a 1.5mL centrifuge tube and centrifuged at 4 ℃ for 10min (10000 rpm).
5) Protein quantification was performed on each sample using the BCA kit, with reference to the procedure under "1.5.2.3".
6) The appropriate PBS and 5 × loading buffer were added to the sample and mixed well to give a final protein concentration of 10 μ g/. mu.L.
7) Placing each obtained sample in boiling water for 10min, and storing at-70 deg.C.
2.5.4 Western blot assay
The procedure was followed as in example 1.
2.5.5 data processing
The procedure was followed as in example 1.
2.5.6 results of the experiment
Protein expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1 in L02, THLE-3 and HepG2 cells treated with different dosing regimens is shown in FIG. 21.
As can be seen from the results in FIG. 21, the protein level of NRF2 in L02, THLE-3 and HepG2 cells was increased to a different extent than that in the control group after FFAs treatment for 24h, and some of the differences were statistically significant (p < 0.05); the protein level of NRF2 was significantly increased in the three cell lines compared to the control and NAFLD alone after different concentrations of EA, MoA and CUR treatment, respectively, with statistical differences (p <0.05), and EA and MoA were concentration-dependent on the up-regulation of NRF 2. The above results suggest: NAFLD increases the protein levels of NRF2 in three liver cell lines; EA and MoA are likely agonists of NRF2, and their upregulation is concentration dependent.
Similarly, the genes downstream of NRF2 in three cell lines after FFAs, EA, MoA and CUR treatments, respectively: the protein levels of NQO1, HO-1, MRP2 and MRP4 were all increased to different extents and showed similar trend to NRF2, with some differences statistically significant (p < 0.05).
After FFAs treatment, the protein levels of CYP2E1 in L02, THLE-3 and HepG2 cells are increased by 2-3 times compared with the control group, the difference has statistical significance (p is less than 0.05), and no obvious change trend exists after EA, MoA and CUR treatment, which indicates that NAFLD can really induce the expression of CYP2E1 in hepatocytes, and EA, MoA and CUR have no great influence on the protein levels of CYP2E 1.
No significant changes in the protein level of KEAP1 occurred in the three cell lines after FFAs, EA, MoA and CUR treatments, respectively.
3. Establishment and verification of NRF 2-knocked down NAFLD cell model
The application of liposome transfection technology to introduce NRF2 specific small interfering RNA (NRF2-siRNA) into L02, THLE-3 and HepG2 cells, utilizes siRNA to inhibit the expression of intracellular NRF2 with high efficiency and specificity, thereby observing whether the treatment effect of EA, MoA and CUR (positive drugs) on NAFLD is also affected, so as to verify again that EA and MoA are indeed agonists of NRF2, and can play a role in treating NAFLD by activating NRF2 pathway.
The three cell lines L02, THLE-3 and HepG2 were treated as follows: (iii) blank control group (addition only)
Figure BDA0002153202320000361
3000) (ii) a ② negative control (NC-siRNA) group: (
Figure BDA0002153202320000362
3000+ NC-siRNA); (iii) Small interfering RNA (NRF2-siRNA) experimental group of NRF2
Figure BDA0002153202320000363
3000+NRF2-siRNA)。
The base sequences of the NRF2-siRNA and NC-siRNA constructed in the early laboratory are shown in Table 3-2.
TABLE 3-2 base sequences of NRF2-siRNA and NC-siRNA
Figure BDA0002153202320000364
3.1 solution preparation
1) MEM medium: to 900mL UP water was added medium powder, 2.431g NaHCO, respectively3And 3.572g of HEPES were thoroughly dissolved and mixed. Filtering with 0.22 μm filter, packaging, sealing, and storing.
2) Preparation of siRNA working solution: the siRNA powder was diluted to a concentration of 10. mu.M with RNasefree water and stored at-20 ℃ in the dark for future use.
3.2 cell lines and cell cultures
L02, THLE-3 and HepG2 cells were cultured. Subculture or counting is carried out when the cells grow to about 80% and are fused, and the diluted cells are spotted for administration experiments.
3.3 cell NRF2-siRNA transfection assay
1) Cell spot plate: 1 day before transfection, L02, THLE-3 and HepG2 cells were treated at 1X 106And (4) inoculating the cells in a 6-well plate at a density of one/mL, and performing subsequent operation when the cells are attached to the wall and grow to 70-80% of fusion.
2) Preparation of siRNA-liposome complexes: using LipofectamineTM3000 transformation Reagent (L3000015, Invitrogen) kit was prepared, and details of the procedures are given in the kit instructions (https:// www.thermofisher.com/order/catalog/product/L3000015SID ═ src-srp-L3000015).
3) And (3) replacing the complete culture medium in the six-well plate, respectively adding the siRNA-liposome compound into corresponding wells, setting a blank control group (Vehicle, only adding a transfection reagent) and placing the blank control group in a cell culture box to continue culturing for 24h, and then carrying out subsequent experiments.
3.4 establishment and validation of NRF2 knockdown NAFLD cell model
3.4.1 cell administration
The NRF2 knocked-down L02, THLE-3 and HepG2 cells obtained under the '3.3' term are taken out from the incubator, a blank culture medium (blank control) containing a standard investigation solution and a 500 mu M FFAs drug-containing culture medium are respectively used for drug administration treatment, and the six-well plate after drug administration is placed in a cell incubator for further culture for 24h and then taken out for subsequent experiments.
3.4.2 validation of NAFLD cell model by oil red O staining
3.4.2.1 sample treatment: the operation was performed as under item "1.3.2".
3.4.2.2 results of the experiment
The results of oil red O staining of L02, THLE-3 and HepG2 cells obtained according to the above procedure are shown in FIG. 22.
As can be seen from the results of oil red O staining in FIG. 22, L02, THLE-3 and HepG2 cells transfected with different siRNAs have different lipid droplets with different sizes, roundness, obvious outlines and red staining in cytoplasm after being treated with 500 μ M FFAs for 24h, which indicates that the three NAFLD hepatocyte models with the knockdown of NRF2 are successfully constructed. Therefore, the above treatment conditions were selected for the subsequent experiments.
4. Effect of EA and MoA on NRF 2-knockdown NAFLD human hepatocyte model
4.1 Effect of EA and MoA on ROS levels in NRF2 knockdown NAFLD cell model
4.1.1 administration of cells
The NAFLD cell models with L02, THLE-3 and HepG2 cell line NRF2 knockdown were constructed by operating under the reference "3.4". After the construction, the administration treatment of the cells was carried out according to the following conditions: adding 0.1% DMSO, 100 mu M EA, 25 mu M MMoA and 10 mu M CUR into a culture medium respectively to carry out administration treatment on L02 cells; adding 0.1% DMSO, 100 μ M EA, 50 μ M MMoA and 10 μ M CUR into the culture medium to treat THLE-3 cells; ③ 0.1 percent DMSO, 100 mu M EA, 25 mu M MeOA and 10 mu M CUR are respectively added into the culture medium to carry out the drug administration treatment on the HepG2 cells.
And placing the medicated cells in a cell culture box for further culture for 24h, and taking out for subsequent experiments.
4.1.2 sample treatment
The operation is carried out with reference to the term "2.2.2".
4.1.3 data processing
The operation is carried out with reference to the item "2.2.3".
4.1.4 results of the experiment
The ROS levels in the cells treated with different dosing regimens for L02, THLE-3, and HepG2 cells are shown in FIG. 23.
As can be seen from the results in fig. 23, after 24h treatment with FFAs, ROS levels in all three cell lines of NAFLD group, NAFLD + NC-siRNA group, and NAFLD + NRF2-siRNA group were significantly increased compared to the blank control group, and the difference was statistically significant (p <0.01), indicating: oxidative stress damage is significantly increased in NAFLD cell models constructed from FFAs. Compared to the NAFLD group alone, ROS levels increased to different degrees in all three cell lines after knockdown of NRF2, and some differences were statistically significant (p <0.05), suggesting: deletion of NRF2 can increase oxidative stress damage in liver tissue caused by NAFLD. Meanwhile, when NRF2 in the cells was knocked down and EA, MoA and CUR treatments were given separately, there was not much change in ROS levels in the three cell lines. The above results suggest: EA, MoA and CUR were unable to reduce ROS levels in NAFLD cell models after NRF2 knockdown, and thus unable to reduce oxidative stress damage in the liver, acting to protect the liver.
4.2 Effect of EA and MoA on GSH levels in NAFLD cell models after NRF2 knockdown
4.2.1 cell administration
The operation is carried out with reference to the contents under "4.1.1".
4.2.2 preparation of solution
The procedure was followed as in example 1.
4.2.3 sample treatment
The operation is carried out with reference to the contents under "2.3.3".
4.2.4 data processing
The operation is carried out with reference to the contents under "2.3.4".
4.2.5 results of the experiment
GSH levels in NRF 2-knocked-down L02, THLE-3 and HepG2 cells after treatment with different dosing regimens are shown in FIG. 24.
As can be seen from the results in fig. 24, after 24h treatment with FFAs, GSH levels were significantly reduced in three cell lines of NAFLD, NAFLD + NC-siRNA and NAFLD + NRF2-siRNA groups compared to the blank control group, with statistical differences (p <0.05), indicating: there was a significant increase in GSH consumption in NAFLD cell models constructed from FFAs. After knockdown of NRF2, GSH levels in all three cell lines were reduced to different degrees compared to the NAFLD group alone, with partial differences of statistical significance (p <0.05), suggesting: the absence of NRF2 may exacerbate the excessive consumption of GSH in liver tissue caused by NAFLD. Meanwhile, when NRF2 was knocked down in the cells and EA, MoA and CUR treatments were given separately, GSH levels did not change much in the three cell lines. The above results suggest: when NRF2 was knocked down, EA, MoA and CUR were unable to alleviate GSH consumption in NAFLD cell models, keeping GSH at a lower level and thus unable to protect the liver.
4.3 Effect of EA and MoA on mRNA levels of various genes of interest in NAFLD cell models after NRF2 knockdown
4.3.1 design and Synthesis of primers
The operation is carried out with reference to the contents under "2.4.1".
4.3.2 cell administration
The operation is carried out with reference to the contents under "4.1.1".
4.3.3 Total RNA extraction from cells
The operation is carried out with reference to the contents under "2.4.3".
4.3.4 Synthesis of cDNA
The procedure was followed as in example 1.
4.3.5 fluorescent polymerase chain reaction
The procedure was followed as in example 1.
4.3.6 data processing
The procedure was followed as in example 1.
4.3.7 results of the experiment
mRNA expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1 in NRF 2-knocked-down L02, THLE-3 and HepG2 cells treated with different dosing regimens is shown in FIG. 25.
As can be seen from the results of FIG. 25, after FFAs treatment for 24h, the mRNA levels of NRF2 in L02, THLE-3 and HepG2 cells of NAFLD group and NAFLD + NC-siRNA group were increased to different degrees compared with the blank control group, and some differences were statistically significant (p < 0.05); after transfection with NRF2-siRNA, mRNA levels of NRF2 were significantly reduced in all three cell lines, with the difference statistically significant (p < 0.05); after transfection with NRF2-siRNA, different concentrations of EA, MoA and CUR treatments were administered, respectively, and the mRNA levels of NRF2 were significantly reduced in all three cell lines compared to the placebo and NAFLD groups, with statistical significance (p <0.05), but no significant change compared to the NAFLD + NRF2-siRNA group. The above results suggest: NAFLD increases mRNA levels of NRF2 in three liver cell lines; transfection with NRF2-siRNA successfully reduced mRNA levels of NRF2 in three cell lines; the transfection of NRF2-siRNA attenuated the induction of NRF2 by EA with MoA and CUR (known as NRF2 agonists).
NRF2 downstream genes in three cell lines transfected with NRF2-siRNA after FFAs, EA, MoA and CUR treatments, respectively: no obvious change rule exists in mRNA levels of NQO1, HO-1, MRP2 and MRP 4.
Similarly, CYP2E1mRNA levels in L02, THLE-3 and HepG2 cells were 2-4 fold increased after FFAs treatment compared to the control group, the differences were statistically significant (p <0.05), and there was no significant trend after EA, MoA and CUR treatment and siRNA transfection, indicating: NAFLD indeed induced CYP2E1 expression in hepatocytes and EA, MoA, CUR and siRNA did not have much effect on CYP2E1mRNA levels.
There was no significant change in KEAP 1mRNA levels in the three cell lines after each of the above treatments.
4.4 Effect of EA and MoA on levels of protein of Gene of interest in NAFLD cell model after NRF2 knockdown
4.4.1 solution preparation
The procedure was followed as in example 1.
4.4.2 cell administration
The operation is carried out with reference to the contents under "4.1.1".
4.4.3 extraction of Total cellular protein and sample preparation
The operation is carried out with reference to the contents under "2.5.3".
4.4.4 Western blot assay
The procedure was followed as in example 1.
4.4.5 data processing
The procedure was followed as in example 1.
4.4.6 results of the experiment
Protein expression of NRF2, KEAP1, HO-1, NQO1, MRP2, MRP4, CYP2E1 in L02, THLE-3 and HepG2 cells treated with different dosing regimens is shown in FIG. 26.
As can be seen from the results in FIG. 26, after FFAs treatment for 24h, the protein levels of NRF2 in L02, THLE-3 and HepG2 cells of NAFLD group and NAFLD + NC-siRNA group were increased to different degrees compared with blank control group, and some differences were statistically significant (p < 0.05); after transfection with NRF2-siRNA, the protein levels of NRF2 were significantly reduced in the three cell lines, with the differences being statistically significant (p < 0.05); after transfection with NRF2-siRNA, and administration of different concentrations of EA, MoA, and CUR treatments, respectively, the protein level of NRF2 was significantly reduced in all three cell lines compared to the placebo and NAFLD groups, with statistical significance for the difference (p <0.05), but there was no significant change compared to the NAFLD + NRF2-siRNA group. The above results suggest: NAFLD increases the protein expression level of NRF2 in three liver cell lines; transfection with NRF2-siRNA successfully reduced the protein levels of NRF2 in three cell lines; the transfection of NRF2-siRNA can obviously reduce the induction of NRF2 by EA, MoA and CUR (known as NRF2 agonist).
NRF2 downstream genes in three cell lines transfected with NRF2-siRNA after FFAs, EA, MoA and CUR treatments, respectively: the protein levels of NQO1, HO-1, MRP2 and MRP4 have no obvious change rule.
Similarly, after FFAs treatment, CYP2E1 protein levels in L02, THLE-3 and HepG2 cells were increased 2-3 fold compared to the control group, the difference was statistically significant (p <0.05), and there was no significant trend after EA, MoA and CUR treatment and siRNA transfection, indicating: NAFLD indeed induced CYP2E1 expression in hepatocytes and EA, MoA, CUR and siRNA did not have much effect on CYP2E1 protein levels.
No significant changes in the protein level of KEAP1 occurred in the three cell lines after each of the above treatments.
In summary, it is shown by the above experiments that EA, MoA and CUR all reduce the intracellular increased ROS levels caused by FFAs, and that NRF2 is a key factor for EA, MoA and CUR to play this role. EA. Both MoA and CUR reduce the increased levels of GSH consumption in cells caused by FFAs, and NRF2 is a key factor for EA, MoA and CUR to play this role. EA and MoA were indeed agonists of Nrf2, both of which were dependent on the presence of Nrf2 for induction of NQO1, HO-1, MRP2 and MRP 4; EA and MoA up-regulate the expression of downstream antioxidant protein, II phase detoxification enzyme and transporter by inducing NRF2, reduce the generation of ROS in cells, and increase the level of reducing GSH, thereby playing a role in resisting oxidative stress reaction and inflammatory reaction caused by NAFLD.
Example 3 prevention of liver injury of NAFLD mouse model APAP by EA and MoA and mechanism research thereof
The sources of the other drugs in this example were the same as above except that: acetaminophen suspension drops (tenolin, shanghai qiangsheng pharmaceutical limited).
1.1 determination of APAP dosing time
1.1.1 Experimental animal grouping, administration and sample Collection
18 wild type C57BL/6 mice bred in the laboratory are selected and randomly divided into 3 groups of 6 mice each with half of male and female. After 1 week of alternating 12h illumination and 12h dark acclimation, treatment groups were gavaged with maximum safe dose of APAP (150mg/kg) at 9AM (APAP-AM) and 5PM (APAP-PM), respectively, for 3 consecutive days, once a day. The grouping and dosing regimen are shown in Table 4-1.
Table 4-1 grouping and dosing regimen of mice (n ═ 6)
Figure BDA0002153202320000431
Injecting 10% chloral hydrate (0.5g/kg) into abdominal cavity of mouse at 9am of the last administration, rapidly performing heart blood sampling and perfusion after anesthesia is successful, taking out liver, soaking part of liver tissue cleaned by normal saline in 4% paraformaldehyde fixing solution, and storing the rest liver tissue at-70 ℃ for later use. The whole blood was placed in a clean 1.5mL centrifuge tube, allowed to stand overnight at 4 ℃ and centrifuged at room temperature for 10min (4000rpm) to separate serum. The weight of each group of mice and the weight of the liver were recorded.
1.1.2 Biochemical index analysis of mouse liver function
1.1.2.1 sample treatment
The procedure is as in example 1.
1.1.2.2 data processing
The procedure is as in example 1.
1.1.2.3 results of the experiment
The results of data processing for the biochemical markers ALT and AST for liver function are shown in FIG. 27.
As can be seen from the results in FIG. 27, the serum concentrations of ALT and AST in all APAP-treated mice were within the normal range (ALT: 30-110U/L, AST: 60-220U/L), indicating that the administration dose of 150mg/kg did not cause significant damage to the liver of the mice, and was a safe administration dose. Compared with a control group, the concentrations of ALT and AST in the serum of mice of a 5PM group which is administrated at 5PM every day are remarkably increased, and the difference has statistical significance (p is less than 0.01); while there was a tendency for the serum concentrations of ALT and AST to increase in the 9AM group of mice administered at 9AM, the changes were not statistically significant. The above results suggest: a continuous intragastric administration of 150mg/kg APAP for 3 days caused a slight damage to the mouse liver. Meanwhile, compared with the 9AM group, the concentrations of ALT and AST in the serum of the mice in the 5PM group were significantly increased, and the difference had statistical significance (p <0.05), suggesting that: the degree of damage to the mouse liver by the administration at 5pm was greater.
1.1.3 mouse liver injury and liver tissue section staining
1.1.3.1 conditions of liver injury in mice
After the thoracic cavity of the mouse was opened, the appearance of the liver of the mouse was visually observed, and the analysis result of the liver injury of the mouse is shown in FIG. 28.
As can be seen from the results in FIG. 28, the livers of the mice were dark red, the surfaces were bright and smooth, and the gall bladder was not swollen. The results suggest that: at different time points, there was no significant effect on liver appearance in wild type mice after continuous 3-day gavage of 150mg/kg APAP.
1.1.4 HE staining analysis
The procedure is as in example 1. Results of HE staining analysis of mouse liver are shown in fig. 29.
As can be seen from the results of fig. 29, liver cells of mice in the APAP-treated group were slightly inflamed and liver cells of mice in the 5PM group were also slightly vacuolized, compared to the control group. The above results suggest: administration of 150mg/kg APAP by gavage for 3 consecutive days caused mild damage to the mouse liver and more severe damage caused by administration in the afternoon.
In conclusion, 150mg/kg of APAP administered by gavage for three consecutive days did not cause significant damage to the liver of the mice, so this dose was a safe dose. In addition, the liver damage of mice caused by the administration of the drug at different time points is different, and the liver damage of mice caused by the administration of the drug at 5pm is more serious. Because mice are nocturnal animals and mostly eat at the evening and night, in order to better simulate the actual situation that most human beings take medicines in the daytime, the subsequent experiments all adopt a dosing scheme of gavage administration of 150mg/kg APAP at 5 pm.
1.2 prevention of liver injury in NAFLD mouse model caused by APAP by EA and MoA
1.2.1 Experimental animal grouping, administration and sample Collection
36 wild type and Nrf2 knockout homozygote (Nrf2-/-) C57BL/6 mice obtained by breeding in the laboratory are selected, and the mice with the same genotype are randomly divided into 6 groups, wherein each group comprises 6 mice and each half of the mice is male and female. After adaptive feeding for 1 week with 12h illumination and 12h darkness, feeding with high fat feed for 2 months. Dosing was started after 2 months and the dosing schedule was: respectively gavage and administering EA, MoA or CUR at 4 pm every day for 1 week, once a day; on the third last day, gavage was continued 1h after dosing (5 pm) with a maximum safe dose of APAP (150mg/kg) for 3 consecutive days, once daily. The grouping and dosing regimen are shown in Table 4-2. EA. The MoA and CUR are still solubilized by emulsification, and the emulsion is formulated as "2.4.1" in tables 2-3 below.
Table 4-2 grouping and dosing regimens for mice (n ═ 6)
Figure BDA0002153202320000451
Figure BDA0002153202320000461
Injecting 10% chloral hydrate (0.5g/kg) into abdominal cavity of mouse at 9am of the last administration, rapidly performing heart blood sampling and perfusion after anesthesia is successful, taking out liver, soaking part of liver tissue cleaned by normal saline in 4% paraformaldehyde fixing solution, and storing the rest liver tissue at-70 ℃ for later use. The whole blood was placed in a clean 1.5mL centrifuge tube, allowed to stand overnight at 4 ℃ and centrifuged at room temperature for 10min (4000rpm) to separate serum. The weight of each group of mice and the weight of the liver were recorded.
1.2.2 Biochemical index analysis of mouse liver function
1.2.2.1 sample treatment
The procedure is as in example 1.
1.2.2.2 data processing
The procedure is as in example 1.
1.2.2.3 results of the experiment
The results of data processing for the biochemical markers ALT and AST for liver function are shown in FIGS. 30 and 31, respectively.
From the results of FIGS. 30 and 31, it can be seen that the serum concentrations of ALT and AST were both increased in APAP-treated mice compared to the control group of the same genotype, with statistical differences (p <0.05), but only the serum concentrations of ALT and AST in wild-type mice remained within the normal range. This phenomenon suggests: APAP 150mg/kg had little effect on wild type mice, but had significant hepatotoxicity in Nrf2 knockout mice.
Compared with the APAP group with the same genotype, the concentration of ALT and AST in the blood serum of the mice of the NAFLD + APAP group is obviously increased and exceeds the normal range, and the difference has statistical significance (p is less than 0.01). This phenomenon suggests: NAFLD can indeed significantly increase the hepatotoxicity of APAP. Meanwhile, the ALT and AST concentrations in the serum of the Nrf2 knockout mouse of the NAFLD + APAP group are obviously higher than those in the serum of a wild mouse, and the statistical significance is achieved (p is less than 0.05), which indicates that the Nrf2 has a certain effect of relieving the hepatotoxicity of APAP.
After respectively gavage administration of EA, MoA and CUR to mice, the serum concentrations of ALT and AST in the wild-type NAFLD + APAP mouse model were all reduced to different degrees compared to the simple NAFLD + APAP group of the same genotype, and the changes were statistically significant (p < 0.05). However, in Nrf2 knockout mice, after intragastric administration of EA, MoA and CUR, respectively, the concentrations of ALT and AST in the serum did not change significantly compared to the NAFLD + APAP group alone of the same genotype. The above results suggest: gavage administration of EA, MoA and CUR significantly reduced APAP liver damage due to NAFLD in wild type mice, and Nrf2 played an important role in this process.
1.2.3 mouse liver injury and liver tissue section staining
1.2.3.1 mouse liver injury
The appearance of the mouse liver was visually observed, and the result of the liver injury of the mouse is shown in FIG. 32.
From the results in fig. 32, it can be seen that the liver of the wild-type mouse treated by APAP alone was dark red and smooth, as compared with the control group, but the liver of the Nrf2 knockout mouse showed a large-area white spot. The above results suggest: APAP 150mg/kg had little effect on wild type mice, but had significant hepatotoxicity in Nrf2 knockout mice. When APAP is given to mice of NAFLD group, obvious large-area small white spots appear on livers of wild type and Nrf2 knockout mice, which indicates that NAFLD can increase liver toxicity of APAP indeed. After EA, MoA and CUR are respectively given, the small white spots on the liver of a wild mouse basically disappear, the appearance characteristics are basically recovered to be normal, and the small white spots with obvious large area can still be seen on the liver of an Nrf2 knockout mouse. The results suggest that: EA. MoA and CUR can significantly reduce the APAP liver injury of wild type mice due to NAFLD, and Nrf2 plays an important role in this process.
1.2.3.2 HE staining analysis results
The procedure is as in example 1. Results of HE staining analysis of mouse liver are shown in fig. 33.
As can be seen from the results in fig. 33, the liver cells of the mice treated with APAP alone all showed different degrees of vacuolar degeneration compared to the control group, and the liver cells of the Nrf2 knockout mice also showed more obvious inflammatory cell infiltration, suggesting that: APAP of 150mg/kg has no great influence on the liver of wild type mice, but has certain hepatotoxicity on Nrf2 knockout mice. After APAP is given to mice of the NAFLD group, the liver cells of wild type and Nrf2 knockout mice have serious phenomena of vacuole degeneration, steatosis, punctate necrosis and inflammatory cell infiltration, which shows that NAFLD can increase the hepatotoxicity of APAP. The punctate necrosis on hepatocytes of wild-type mice was substantially eliminated and the phenomena of vacuolar degeneration, steatosis, and inflammatory cell infiltration were improved after administration of EA, MoA, and CUR, respectively, whereas the Nrf2 knockout mouse group was not much changed. The results suggest that: EA. MoA and CUR can significantly reduce the APAP liver injury of wild type mice due to NAFLD, and Nrf2 plays an important role in this process.
1.2.3.3 oil Red O staining analysis results
The procedure is as in example 1. The results of mouse liver oil red O staining are shown in fig. 34.
As can be seen from the results in fig. 34, liver cells of mice treated with APAP alone all exhibited different degrees of nuclear voiding compared to the control group, and Nrf2 knockout mice were more severe, suggesting that: 150mg/kg APAP had not much effect on the liver of wild type mice, but had some hepatotoxicity to Nrf2 knockout mice. After APAP is given to mice of the NAFLD group, the phenomena of severe nuclear cavern and steatosis appear in liver cells of wild type mice and Nrf2 knockout mice, and the NAFLD can increase the hepatotoxicity of the APAP. The phenomenon of nuclear cavern and steatosis in hepatocytes of wild type mice was improved after administration of EA, MoA and CUR, respectively, whereas the Nrf2 knockout mice group did not change much. The results suggest that: EA. MoA and CUR can significantly reduce APAP liver damage in wild type mice due to NAFLD, and Nrf2 plays an important role in this process.
1.2.4 Effect of EA and MoA on GSH levels in liver tissues of mice
1.2.4.1 reagent preparation
The procedure is as in "example 1.
1.2.4.2 sample treatment
The procedure is as in example 1.
1.2.4.3 data processing
The procedure is as in example 1.
1.2.4.4 results of the experiment
The results of GSH concentrations in liver tissues of the mice in each group are shown in fig. 35.
As can be seen from the results in fig. 35, the GSH concentration in the livers of the APAP-treated mice was significantly reduced compared to the blank control group of the same genotype, the difference was statistically significant (p <0.05), and the GSH concentration of the Nrf2 knockout mice was also significantly lower than that of the wild-type mice (p <0.05), suggesting that: APAP at 150mg/kg significantly reduced GSH levels in the liver of mice and had a greater effect on Nrf2 knockout mice.
The concentration of GSH in the liver of mice in the NAFLD + APAP group was significantly reduced compared to the APAP group of the same genotype, with statistical differences (p <0.01), suggesting: NAFLD can indeed significantly increase the hepatotoxicity of APAP, producing more NAPQI and thus consuming more GSH. Meanwhile, the concentration of GSH in the liver of the Nrf2 knockout mouse of the NAFLD + APAP group is obviously lower than that of GSH in the liver of a wild type mouse, and the concentration has statistical significance (p is less than 0.05), which indicates that the consumption of GSH in the liver by APAP can be relieved by the existence of Nrf 2.
After respectively gavage administration of EA, MoA and CUR to mice, the concentration of GSH in the liver of wild type mice is increased to a different degree compared with the simple NAFLD + APAP group with the same genotype, but the effect on Nrf2 knockout mice is not too great. The above results suggest: gavage administration of EA, MoA and CUR significantly alleviated the excessive consumption of aphp on GSH by NAFLD in wild-type mice, and Nrf2 played an important role in this process.
1.2.5 Effect of EA and MoA on mRNA levels of various genes of interest in liver tissues of mice
1.2.5.1 design and Synthesis of primers
The procedure is as in example 1.
1.2.5.2 tissue Total RNA extraction
The procedure is as in example 1.
1.2.5.3 Synthesis of cDNA
The procedure is as in example 1.
1.2.5.4 fluorescent polymerase chain reaction
The procedure is as in example 1.
1.2.5.5 data processing
The procedure is as in example 1.
1.2.5.6 results of the experiment
mRNA expression of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4 in liver tissue after treatment of mice with different dosing regimens is shown in FIG. 36.
As can be seen from the results in fig. 36, the mRNA level of Nrf2 in the liver of wild-type mice treated with APAP alone was increased compared to the control group of the same genotype, with statistical significance for the difference (p < 0.05); after wild-type NAFLD mouse model APAP treatment was given, Nrf2 mRNA levels in the liver were increased compared to both control and APAP-only treatment groups, with statistical significance for the difference (p < 0.05); after continuing with EA, MoA and CUR treatments, respectively, Nrf2 mRNA levels in the liver of the wild-type NAFLD mouse model were significantly increased compared to the control, APAP alone and NAFLD + APAP of the same genotype, with statistical significance for the differences (p <0.01), but the above treatments had no significant effect on Nrf2 mRNA levels in the liver of Nrf2 knockout mice.
Similarly, in the liver of wild-type mice treated with EA, MoA and CUR, respectively, the downstream gene of Nrf 2: nqo1, Ho-1, Mrp2 and Mrp4 have different degrees of mRNA level increase, the difference has statistical significance (p is less than 0.05), and the variation trend is similar to that of Nrf2, but the variation rule is not obvious in the liver of an Nrf2 knockout mouse.
The above results suggest: APAP can induce mRNA expression of Nrf2 in the liver of a wild-type mouse; NAFLD can potentiate the induction of aprp on Nrf 2; EA. MoA, like CUR, is an agonist of Nrf 2.
In the livers of wild type and Nrf2 knockout mice treated with APAP alone, Cyp2e1 tended to increase, but Cyp2e1 increased significantly after NAFLD treatment, with a statistically significant difference (p <0.05), and there was no significant change after EA, MoA and CUR treatment, compared to the blank control group. Thus illustrating that: NAFLD can obviously induce the expression of Cyp2e1 in mouse liver, and EA, MoA and CUR have no great influence on the mRNA level of Cyp2e 1. After the above treatments, no significant changes in the levels of Keap 1mRNA were observed in the livers of the groups of mice.
1.2.6 Effect of EA and MoA on levels of proteins of genes of interest in liver tissues of mice
1.2.6.1 preparation of solution
The procedure is as in "example 1.
1.2.6.2 Total protein extraction from tissue and sample preparation
The procedure is as in example 1.
1.2.6.3 Western blot assay
The procedure is as in example 1.
1.2.6.4 data processing
The procedure is as in example 1.
1.2.6.5 results of the experiment
Protein expression of Nrf2, Keap1, Cyp2e1, Ho-1, Nqo1, Mrp2 and Mrp4 in liver tissue after treatment of mice with different dosing regimens is shown in figure 37.
As can be seen from the results in fig. 37, the protein level of Nrf2 in the liver of wild type mice treated with APAP alone was increased compared to the control group of the same genotype, with statistical significance for the difference (p < 0.05); after the wild-type NAFLD mouse model APAP treatment is given, the protein level of Nrf2 in the liver is increased compared with that of a control group and a single APAP treatment group, and the difference is statistically significant (p is less than 0.05), but the treatment has no obvious influence on the protein level of Nrf2 in the liver of an Nrf2 knockout mouse; after continuing with EA, MoA and CUR treatments, respectively, the protein level of Nrf2 in the liver of the wild-type NAFLD mouse model was significantly increased compared to the control group, APAP alone and NAFLD + APAP of the same genotype, with statistical significance (p <0.05), but there was no significant change in the protein level of Nrf2 in the liver of Nrf2 knockout mice.
Similarly, in the liver of wild-type mice treated with EA, MoA and CUR, respectively, the downstream gene of Nrf 2: the protein levels of Mrp2, Mrp4, Nqo1 and Ho-1 were all increased to different degrees, the differences were statistically significant (p <0.05), and showed a similar trend to Nrf2, but there was no obvious change in the liver of Nrf2 knockout mice.
The above results suggest: APAP can induce the protein expression of Nrf2 in the liver of a wild mouse; NAFLD can potentiate the induction of aprp on Nrf 2; EA. MoA, like CUR, is an agonist of Nrf 2.
In the livers of wild type and Nrf2 knockout mice treated with APAP alone, Cyp2e1 levels tended to increase, but Cyp2e1 levels increased significantly after NAFLD treatment, the difference was statistically significant (p <0.05), and there was no significant change after EA, MoA and CUR treatment, compared to the blank control group. Thus, NAFLD can obviously induce the expression of Cyp2e1 in mouse liver, and EA, MoA and CUR have no great influence on the protein level of Cyp2e 1.
There was no significant change in the protein level of Keap1 in the livers of groups of mice after the various treatments described above.
In conclusion, it can be seen from the above experiments that 30mg/kg EA, 10mg/kg MoA and 100mg/kg CUR all can prevent APAP hepatotoxicity of wild type mice caused by NAFLD, and Nrf2 is a key factor for preventing APAP hepatotoxicity of EA and MoA. EA and MoA are indeed agonists of Nrf 2; the induction of Nqo1, Ho-1, Mrp2 and Mrp4 by the two is dependent on the presence of Nrf 2; EA and MoA can increase the level of GSH in liver by inducing the expression of Nrf2 and its downstream antioxidant gene and efflux transporter, and prevent APAP hepatotoxicity caused by the induction of Cyp2e1 by NAFLD through three ways of resisting oxidative stress, promoting the efflux of toxic substances and neutralizing excessive NAPHQI.
Experimental example 4 study on the regulating action of EA on Nrf2 based on epigenetic mechanism
The sources of the other drugs in this example were the same as above except that: Quick-DNATMMiniprep pit (D3024, ZYMO RESEARCH CORP., USA) EZ DNA Methylation-GoldTMKit (D5005, ZYMO RESEARCH CORP. USA) CpG Methyransferase (M.SssI) (M0226S, NEW ENGLAND BIOLABS).
1.1 Effect of EA on the degree of methylation of CpG island regions of the NRF2 promoter in L02 cells
1.1.1 solution preparation
1)1640 complete medium: the same as in example 1.
2) 0.25% pancreatin: the same as in example 1.
3)2 × TBE buffer: 0.1g of NaOH, 10.8g of Tris-HCl, 5.5g of boric acid and 0.74g of EDTA-2Na are respectively weighed, and a proper amount of UP water is added to completely dissolve the components and then the volume is determined to be 500mL, thus obtaining 2 xTBEbuffer.
4)0.5 × TBE buffer: obtained by diluting 2 XTBE buffer with UP water 1:3 (volume ratio), and the solution was used for agarose gel preparation and PCR electrophoresis.
1.1.2 cell lines and cell cultures
L02 cells were cultured. Subculture or counting is carried out when the cells grow to about 80% and are fused, and the diluted cells are spotted for administration experiments.
1.1.3 cell administration
The L02 cells were packed at 1X 106The cells are inoculated in a 6-well plate at a density of one/mL, and when the cells are attached to the wall and grow to 70-80% fusion, the L02 cells are respectively dosed with 0.1% DMSO (blank control, positive control), 50/100 μ M EA, and 1 μ M5-Aza (5-Aza-2' -deoxycytidine, negative control). And placing the 6-hole plate added with the drugs in a cell culture box for further culture for 24h, and taking out for subsequent experiments.
1.1.4 cell DNA extraction and sample preparation
1.1.4.1 extraction of cellular DNA
Using Quick-DNATMThe miniprep kit extracts DNA of L02 cells, and comprises the following specific operation steps:
1) the 6-well plate was taken out and placed in a clean bench, the medium was discarded, and the plate was rinsed 3 times with PBS to remove the residual liquid.
2) Add 500. mu.L Genomic lysine Buffer to each well, vortex for 6s, and let stand at room temperature for 5-10 min.
3) The above mixture was transferred to Zymo-SpinTMIn IIC filter column (on clean collection tube), 10000g centrifugation for 1 min.
4) Zymo-SpinTMThe IIC column was placed in a new collection tube, 200. mu.L of DNAPRE-Wash Buffer was added, and centrifugation was carried out for 1min at 10000 g.
5) Zymo-SpinTMThe IIC cartridge was placed in a fresh collection tube and 500. mu. L g-DNA WashBuffer was added and centrifuged at 10000g for 1 min.
6) Zymo-SpinTMIIC filter column was placed in a new centrifuge tube, 50. mu.L of UP water was added, incubated at room temperature for 2-5min, and centrifuged at 12000g for 30 s. The supernatant (DNA solution) was collected and stored at-20 ℃ until use.
1.1.4.2 preparation of MSP Positive control samples
DNA from the untreated L02 cells was removed and all cytosine residues in CpG islands were recognized and methylated by CpG methylase m.sssi as a positive control to optimize MSP reactions.
The reaction system was prepared according to the following formulation:
Figure BDA0002153202320000531
the obtained methylated solution was thoroughly mixed and centrifuged. Water bath at 37 ℃ for 1 h.
1.1.4.3 sulfite conversion of DNA
Using EZ DNA Methylation-GoldTMKit carries out sulfurous acid transformation on the extracted L02 cell DNA, and the specific operation steps are as follows:
1) mu.L of the DNA was sampled to a value of OD260/280 and the concentration of the DNA measured by Nanodrop 2000, and 500ng of the DNA (20. mu.L) was sampled and placed in a PCR tube for subsequent experiments.
2) Preparing CT Conversion Reagent: adding 900 μ L UP water and 300 μ L LM-Dilution Buffer into CT Conversion Reagent tube, shaking at room temperature in dark for 10min to mix thoroughly (the Reagent is extremely sensitive to light, preferably prepared for use, and stored in dark for 1 day at room temperature, 1 week at 4 deg.C, 1 month at-20 deg.C, and mixed at 37 deg.C).
3) 130 μ L of prepared CT Conversion Reagent was put into the PCR tube containing the DNA, mixed well and centrifuged.
4) And (3) putting the PCR tube into a PCR instrument for sulfurous acid conversion reaction. The PCR amplification procedure was as follows: firstly, the temperature is 98 ℃ and the time is 10 min; ② 64 ℃ for 2.5 h; ③ 4 ℃, standing for 20 h.
5) Taking 600 mu LM-Binding Buffer in Zymo-SpinTMIn an IC cartridge (placed on a clean collection tube).
6) Adding the sample obtained in 4) to Zymo-SpinTMIn the IC filter column, the lid was closed, and the mixture was inverted upside down. 10000g for 30 s.
7) Zymo-SpinTMThe IC cartridge was placed in a fresh collection tube and centrifuged for 30s at 10000g with 100. mu.L-Wash Buffer added.
8) Zymo-SpinTMPlacing the IC filter column into a new collection tube, adding 200 μ M LM-depletion Buffer, standing at 25 deg.C for 15-20min, and centrifuging at 10000g for 30 s.
9) Zymo-SpinTMPlacing the IC filter column into a new collection tube, adding 200 μ M-Wash Buffer, 10000g, centrifuging for 30 s.
10) And repeating the steps.
11) Zymo-SpinTMThe IC cartridge was placed in a fresh centrifuge tube and 10. mu. L M-Elution Buffer was added and centrifuged at 10000g for 30 s. The DNA solution of the lower clear was collected and stored at-20 ℃.
12) Taking 1-4 mu L of the DNA solution for subsequent MSP reaction.
1.1.5 methylation-specific PCR (MSP)
1.1.5.1 Experimental methods
The primer adopts an MSP primer of a human NRF2 promoter designed by the laboratory Wangting classmate, namely, the complete sequence of a human NRF2(Homo sapiens chromosome 2, GRCh38) gene is searched from GeneBank, the transcription initiation site of the complete sequence is taken as an initiation position, and the upstream 2kb is taken as a promoter region. The sequence is subjected to CpG Island prediction by a CpG Island Finder (http:// dbcat. cgm. ntu. edu. tw), and a very dense CpG Island exists in a region of 900-2000 bp. Finally MethPrimer (The Li Lab, http:// www.urogene.org/MethPrimer /)[130]MSP primer design is carried out on the sequence, a proper primer sequence is selected, synthesis of Huada gene is entrusted, and the sequence is purified by PAGE. The primer sequences are shown in Table 5-1.
TABLE 5-1 Methylation Specific PCR (MSP) primer sequences.
Figure BDA0002153202320000541
Figure BDA0002153202320000551
The primers were adjusted to the appropriate concentration (10mM) and the DNA samples obtained above were taken for Polymerase Chain Reaction (PCR) and agarose gel electrophoresis, the procedure was as follows:
polymerase chain reaction:
1) after the plate under the '1.4.2.3' grows monoclonal colonies, a plurality of monoclonal colonies are randomly picked by using an aseptic gun head with a proper size and are quickly dissolved in 10-20 mu L of sterilized water.
2) 0.5. mu.L of the above-mentioned bacterial solution was taken and placed in a PCR tube, and the remaining bacterial solution was added to 1mL of LB liquid medium containing ampicillin and stored at 37 ℃ for later use.
3) By using
Figure BDA0002153202320000552
Max Super-Fidelity DNA Polymerase kit was used to perform colony PCR amplification on each sample. The specific procedures and volumes of reagents added are detailed in the kit instructions (http:// www.vazyme.com/product/115. html).
4) And (3) fully and uniformly mixing the obtained PCR solution, centrifuging, and amplifying each sample by using a PCR instrument, wherein the amplification temperature and time program is as follows: firstly, pre-denaturation is carried out for 3min at 95 ℃; ② denaturation at 95 ℃ for 15 s; ③ annealing at 58 ℃ for 15 s; extension for 1.5min at 72 ℃; fifthly, repeating the steps from the second step to the fourth step for 35 cycles; sixthly, completely extending for 5min at 72 ℃; keeping at 4 deg.C.
Agarose gel electrophoresis:
1) to 40mL of 0.5 XTBE buffer, 0.6g of agarose powder was added and the mixture was heated in a microwave oven for 90 seconds. And (3) when the temperature is reduced to 50-60 ℃, adding 4 mu L of Golden View (EB substitute), fully mixing uniformly, adding into a glue preparation groove, slightly inserting a comb with a proper width to avoid bubbles, and using after the mixture is cooled and solidified.
2) The agarose gel was placed on a scale rack in an electrophoresis tank.
3) After mixing 6 × Loading Buffer with the amplified sample at a ratio of 1:5 (volume ratio), an appropriate volume was added to each well of the agarose gel.
4) And (3) opening a PCR electrophoresis apparatus (with the voltage of 100mV) for 25-30 min.
After the electrophoresis is finished, the agarose gel is placed in a gel imager for photographing.
1.1.5.2 data processing
The resulting gel bands were subjected to gray scale analysis using LANE1D software.
The differences between the degree of methylation and the degree of non-methylation on the NRF2 promoter in each experimental group were compared using Microsoft Excel 2019 for data collation. Experimental results single-factor analysis of variance (ANOVA) was performed using Graphpad prism 7 software, with differences of statistical significance when p < 0.05.
1.1.5.3 results of the experiment
Methylation of the NRF2 promoter region of cells 24h after 50, 100. mu.M EA, 1. mu.M 5-Aza (negative control-demethylation) and M.SssI (positive control-methylation) were examined, respectively, and the results are shown in FIG. 38.
As can be seen from the PCR results, after 50 and 100 μ M EA and 1 μ M5-Aza treatment for 24h, the demethylation degree of the NRF2 promoter region in L02 cells is remarkably increased (p <0.05), which indicates that 50 and 100 μ MEA can reduce the DNA methylation degree of the NRF2 promoter region, thereby inducing the expression of NRF 2.
1.2 Effect of EA on HAT1 and HDACs protein expression in L02 cells
1.2.1 solution preparation
Refer to example 1.
1.2.2 cell lines and cell cultures
L02 cells were cultured. Subculture or counting is carried out when the cells grow to about 80% and are fused, and the diluted cells are spotted for administration experiments.
1.2.3 administration of cells
The L02 cells were packed at 1X 106The cells are inoculated in a 6-well plate at a density of one cell per mL, and when the cells are attached to the wall and grow to 70-80% fusion, the L02 cells are respectively dosed with 0.1% DMSO (blank control) and 50/100 μ M EA. And placing the 6-hole plate added with the drugs in a cell culture box for further culture for 24h, and taking out for subsequent experiments.
1.2.4 extraction of cellular proteins and sample preparation
Refer to example 2.
1.2.5 Western blot assay
Refer to example 1.
1.2.6 data processing
Refer to example 1.
1.2.7 results of the experiment
Protein expression of HAT1 and related HDACs (histone deacetylase) in L02 cells after 24h of 50 and 100. mu.M EA action, respectively, was examined, and the results are shown in FIG. 39.
As can be seen from the results in fig. 39, the protein levels of NRF2 and HAT1 in L02 cells significantly increased (p <0.05) after 50,100 μ M EA treatment for 24h, and there was a concentration-dependent trend. At the same time, protein levels of HDAC1 and HDAC6 were significantly reduced, the difference was statistically significant (p <0.05), and a concentration-dependent trend was exhibited. However, 50 μ M EA had no significant effect on the protein levels of HDAC2, HDAC3, HDAC4, HDAC5, HDAC 8. The above results suggest: EA may induce expression of NRF2 by modulating protein levels of HAT1, HDAC1, and HDAC6 in L02 cells.
The methylation-specific PCR (MSP) experiment results show that EA can regulate the DNA methylation transfer of the NRF2 promoter region, thereby inducing the expression of NRF 2. The results of the Western Blot experiments on the expression of histone acetyltransferase 1(HAT1) and Histone Deacetylase (HDACs) -related proteins show that EA may up-regulate the expression level of NRF2 by inducing HAT1 and inhibiting the expression of HDAC1 and HDAC 6. The experiment discusses the epigenetic mechanism of EA activated Nrf2 pathway, studies the activation of Nrf2 by EA in terms of DNA methylation and histone acetylation, and combines the previous study results to provide the following mechanism of EA treatment NAFLD and prevention of APAP hepatotoxicity increased by NAFLD, as shown in FIG. 40.
In conclusion, the invention proves that the ellagic acid is an agonist of Nrf2, can increase the GSH level in the liver by up-regulating the expression of Nrf2 and downstream antioxidant factors and transporters thereof, simultaneously reduces the ROS level in liver cells, reduces oxidative stress reaction and inflammatory reaction, maintains the function of the liver cells, and plays a role in treating NAFLD. In addition, ellagic acid can increase GSH levels in the liver by up-regulating the expression of Nrf2 and its downstream antioxidant factors and transporters, thereby serving to prevent hepatotoxicity due to increased NAFLD at normal doses of APAP (mechanistic map shown in fig. 41). Therefore, the ellagic acid has good application prospect in preparing NAFLD treatment medicines and APAP hepatotoxicity prevention medicines.
SEQUENCE LISTING
<110> Sichuan university
Application of ellagic acid in preparing medicine for preventing and/or treating non-alcoholic fatty liver disease and liver injury
<130> GYKH1353-2019P017258CC
<160> 41
<170> PatentIn version 3.5
<210> 1
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 1
acagaggaac acaaagacca g 21
<210> 2
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 2
gtgtctggga tgagctagtg 20
<210> 3
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 3
gaacggattt ggccgtattg 20
<210> 4
<211> 23
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 4
ctggaacatg tagaccatgt agt 23
<210> 5
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 5
gaatgctatg acccggacag 20
<210> 6
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 6
tcaggtattc caagtgcttc ag 22
<210> 7
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 7
aggacatgga gcaagtttgg 20
<210> 8
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 8
ttctgtcagt gtggcttctg 20
<210> 9
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 9
cagatccagg accaagagat tt 22
<210> 10
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 10
gacaggtcta tggctgctaa a 21
<210> 11
<211> 24
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 11
tatccttccg agtcatctct agca 24
<210> 12
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 12
tctgcagctt ccagcttctt g 21
<210> 13
<211> 19
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 13
tcactggaca tcaactgcc 19
<210> 14
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 14
tggtctctgt tcctgcaaag 20
<210> 15
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 15
acctctgctc gcgcgtgttc t 21
<210> 16
<211> 24
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 16
ccagtaccgt tgaagctcct ctcc 24
<210> 17
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 17
cctgccattc tgaaaggctg gt 22
<210> 18
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 18
gtggtgatgg aaagcactgc ct 22
<210> 19
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 19
ccaggcagag aatgctgagt tc 22
<210> 20
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 20
aagactgggc tctccttgtt gc 22
<210> 21
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 21
caacttcgct gagcagattg gc 22
<210> 22
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 22
tgatgagggt caccagttgg ca 22
<210> 23
<211> 20
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 23
gcgacggaaa gagtatgagc 20
<210> 24
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 24
agcatctgat ttgggaatgt g 21
<210> 25
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 25
gaaaggatcc tacagtgctc tc 22
<210> 26
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 26
gcccatagtc atcgtcttct t 21
<210> 27
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 27
tgttcttctg gtggctcaat c 21
<210> 28
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 28
tcttctggca gcactgaata c 21
<210> 29
<211> 23
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 29
tcggagtcaa cggatttggt cgt 23
<210> 30
<211> 24
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 30
tgccatgggt ggaatcatat tgga 24
<210> 31
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 31
gagcaccatc aatctctgga cc 22
<210> 32
<211> 22
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 32
cacggtgata ccgtccattg tg 22
<210> 33
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 33
cgcucaguua caacuagaut t 21
<210> 34
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 34
aucuaguugu aacugagcgt t 21
<210> 35
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 35
uucuccgaac gugucacgut t 21
<210> 36
<211> 21
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 36
acgugacacg uucggagaat t 21
<210> 37
<211> 25
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 37
ttttgtaatt ttaaattagg gaggc 25
<210> 38
<211> 25
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 38
gaaaaaccta aaaaaaatct ccgtt 25
<210> 39
<211> 26
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 39
tttgtaattt taaattaggg aggtgt 26
<210> 40
<211> 25
<212> DNA
<213> Artificial sequence (artificial sequence)
<400> 40
aaaaaaccta aaaaaaatct ccatt 25
<210> 41
<211> 2544
<212> DNA
<213> mouse (Mus musculus)
<400> 41
atcagtgata acatgtagtg aactaacccg tgtggaccat cttaaccatg gcttctcctt 60
ccttttctgt tttaaacata ggacatggat ttgattgaca tcctttggag gcaagacata 120
gatcttggag taagtcgaga agtgtttgac tttagtcagc gacagaagga ctatgagctg 180
gaaaaacaga aaaaactcga aaaggaaaga caagagcaac tccagaagga acaggagaag 240
gccttttttg ctcagtttca actggatgaa gaaacaggag aattcctccc aattcagccg 300
gcccagcaca tccagacaga caccagtgga tccgccagct actcccaggt acactcgtcg 360
tggtgggagc taaggaaaac tctagtgaga aagcagactc tctggagttg agttcttggt 420
ctgccattta ctgtgtcttt tgtgaggagg agaagttctc aaacttcgct tcttgataat 480
caggacacag atcacaggga ggactttgtg agttacagaa acatcctctg tggtgatgaa 540
acagcagcag aaatactgtt gggagtaaaa gaagtaggca ttgctcattg tggtaggcag 600
ggccctgatt gtatgggtaa ctgacttaac tgtgttaagt atgattccca ttttatattc 660
ccatgtctaa acaactaaag cattcgccca gaacactcag aaagaaatga agggaggtta 720
ttgcctagca cagagccctg ggttcagtcc cccagcgctg ttcgttaaag gggagggact 780
aataatttga acacaatctg gtttgaatct atgcaaatcg atttctgaaa tgaaaccata 840
ggttatttta tgaacaagtc ttattgctcg ttgtgaccct gtgcttagaa catttcataa 900
atgatgctct ctgtgccttt ccccttcctc caggttgccc acattcccaa acaagatgcc 960
ttgtactttg aagactgtat gcagcttttg gcagagacat tcccatttgt agatgaccat 1020
gaggtataaa aatgtttgtt taacagcaaa actcccttat ctgatattag ttcctttcat 1080
gtgtctccaa ttaagagaag aaaagaaaat tttagaagga aaaaattgat caaagaaatt 1140
gtcaagtaaa ctgtatgaga gctatacaat gcttaaaaat aagacctgta tgggctggtg 1200
agatggctca gttggtaaga gtacccgact gctcttccga aggtccagag ttcaaatccc 1260
agcaaccaca aggtggctca caacatccat aacaagatct gactccctct tctggagtgt 1320
ctgaagacag ctacagtgta cttacatata ataaataaat aaatctttaa aaaaaaaaaa 1380
ataaggcctg taaactacaa gtccatttta ctgtatagct ggaaacagga atcagaataa 1440
ttttccctgg aaactggata taggtatata aaatattttg actagtaaag aacaactatt 1500
aatcagcatt tggattaaaa aaatcttaat ctgttgtttg aagcattctg ctagatatta 1560
tgggtacaga ttaagtccta atgaatgttt ttatccattt tgaagtctgc ctttaaatac 1620
atggagtgaa ataacctagg agtgtattaa tatggagtca ctgggaggag gaaatgtttc 1680
attttataaa agcagcctga gagctgtagg ccctgctgct gtctgttctt catgccttgg 1740
ttctcactca catgaatcaa tgtcacgtca atcttggctt tcttcacttg catttcagtc 1800
gcttgccctg gatatcccca gccacgctga aagttcagtc ttcactgccc ctcatcaggc 1860
ccagtccctc aatagctctc tggaggcagc catgactgat ttaagcagca tagagcagga 1920
catggagcaa gtttggcagg agctattttc cattcccgaa ttacaggtaa gagagctcta 1980
ggagtgtgct gttttctgcg ggccctttta aattagtcat cctagttatt tattatttac 2040
atgctacctc ctcaaaggaa gaaattgatg gtgcatttaa attactcatg agagcttccc 2100
agactcactt aacacacata gtttttaggt aatcagactg aatatttctg gataaattca 2160
ttcaaagact gaaagctaat ttagagttct gacaaagata aaatacttat ctattgaaaa 2220
atgggagttg aaggaattat tgaaaagaac accttggatt tgggggtagg gaattgatct 2280
aaaatgcact tagcctctgc tcatacaatg tgaccttctt tcctagtgtc ttaataccga 2340
aaacaagcag ctggctgata ctaccgctgt tcccagccca gaagccacac tgacagaaat 2400
ggacagcaat taccattttt actcatcgat ctcctcgctg gaaaaagaag tgggcaactg 2460
tggtccacat ttccttcatg gttttgagga ttctttcagc agcatcctct ccactgatga 2520
tgccagccag ctgacctcct taga 2544

Claims (11)

1. Application of ellagic acid in preparing medicine for preventing and/or treating non-alcoholic fatty liver disease and liver injury is provided.
2. Use according to claim 1, characterized in that: the liver injury is a chemical induced liver injury.
3. Use according to claim 2, characterized in that: the liver damage is caused by the administration of acetaminophen.
4. Use according to claim 3, characterized in that: the liver damage is caused by the administration of normal doses of acetaminophen.
5. Use according to claim 1, characterized in that: the non-alcoholic fatty liver disease includes non-alcoholic simple fatty liver, non-alcoholic steatohepatitis, liver cirrhosis and hepatocellular carcinoma.
6. Use according to any one of claims 1 to 5, characterized in that: the drug can induce the expression of Nrf2, and the downstream regulatory gene and the efflux transporter thereof.
7. Use according to claim 6, characterized in that: the downstream regulatory genes of the Nrf2 are NQO1, HO-1, MRP2 and MRP 4; the efflux transporters are MRP2 and MRP 4.
8. Use according to claim 6, characterized in that: the induction of the expression of the Nrf2 is realized by regulating the promoter region of the Nrf2 to perform DNA methylation transfer and/or by inducing HAT1 and inhibiting the expression of HDAC1 and HDAC 6.
9. Use according to any one of claims 1 to 5, characterized in that: the medicine can resist oxidative stress reaction and inflammation reaction caused by non-alcoholic fatty liver disease.
10. Use according to any one of claims 1 to 5, characterized in that: the drug is capable of reducing the level of ROS in liver cells; the drug is capable of increasing GSH levels in liver cells.
11. Use according to any one of claims 1 to 5, characterized in that: the medicine can promote toxic substance discharge and neutralize N-acetyl-p-benzoquinone imine, reduce oxidative stress injury in liver cells and protect liver cells.
CN201910709404.0A 2019-08-02 2019-08-02 Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury Pending CN112294800A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910709404.0A CN112294800A (en) 2019-08-02 2019-08-02 Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910709404.0A CN112294800A (en) 2019-08-02 2019-08-02 Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury

Publications (1)

Publication Number Publication Date
CN112294800A true CN112294800A (en) 2021-02-02

Family

ID=74486531

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910709404.0A Pending CN112294800A (en) 2019-08-02 2019-08-02 Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury

Country Status (1)

Country Link
CN (1) CN112294800A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115887542A (en) * 2022-11-22 2023-04-04 中南民族大学 Application of rose ethanol extract in preparation of anti-hepatic fibrosis medicine or health food

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1857259A (en) * 2005-04-30 2006-11-08 韩国生命工学研究院 Medicinal composition for preventing and/or treating liver disease
CN101289453A (en) * 2008-05-16 2008-10-22 西安交通大学 Ellagic acid compounds, preparation thereof and applications in anticancer drugs
CN108743584A (en) * 2018-08-24 2018-11-06 东北林业大学 The new application of ellagic acid

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1857259A (en) * 2005-04-30 2006-11-08 韩国生命工学研究院 Medicinal composition for preventing and/or treating liver disease
CN101289453A (en) * 2008-05-16 2008-10-22 西安交通大学 Ellagic acid compounds, preparation thereof and applications in anticancer drugs
CN108743584A (en) * 2018-08-24 2018-11-06 东北林业大学 The new application of ellagic acid

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
C. GIRISH ET AL.: "Hepatoprotective activity 《of picroliv, curcumin and ellagic acid compared to silymarin on paracetamol induced liver toxicity in mice", 《FUNDAMENTAL & CLINICAL PHARMACOLOGY》 *
SUNIL K. PANCHAL ET AL.: "Ellagic acid attenuates high-carbohydrate, high-fat diet-induced metabolic syndrome in rats", 《EUR J NUTR》 *
YUAN DING ET AL.: "Protective effects of ellagic acid against tetrachloride‑induced cirrhosis in mice through the inhibition of reactive oxygen species formation and angiogenesis", 《EXPERIMENTAL AND THERAPEUTIC MEDICINE》 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115887542A (en) * 2022-11-22 2023-04-04 中南民族大学 Application of rose ethanol extract in preparation of anti-hepatic fibrosis medicine or health food

Similar Documents

Publication Publication Date Title
Chelakkot et al. Modulating glycolysis to improve cancer therapy
Li et al. Andrographolide protects chondrocytes from oxidative stress injury by activation of the Keap1–Nrf2–Are signaling pathway
Wang et al. Curcumin protects against thioacetamide-induced hepatic fibrosis by attenuating the inflammatory response and inducing apoptosis of damaged hepatocytes
Sahai et al. Upregulation of osteopontin expression is involved in the development of nonalcoholic steatohepatitis in a dietary murine model
Zhang et al. Apigenin in the regulation of cholesterol metabolism and protection of blood vessels
Gao et al. Isoacteoside, a dihydroxyphenylethyl glycoside, exhibits anti‐inflammatory effects through blocking toll‐like receptor 4 dimerization
Zhang et al. Pharmacokinetic interaction between JBP485 and cephalexin in rats
Brunet et al. Nitric oxide and the Th2 response combine to prevent severe hepatic damage during Schistosoma mansoni infection
Shi et al. Augmenter of liver regeneration protects against carbon tetrachloride-induced liver injury by promoting autophagy in mice
Wang et al. Therapeutic effect of chitosan on CCl4‑induced hepatic fibrosis in rats
CN108606982A (en) VCP inhibitor and oncolytic virus application in preparation of anti-tumor drugs
Halegoua-DeMarzio et al. Liver injury associated with turmeric—A growing problem: Ten cases from the Drug-Induced Liver Injury Network [DILIN]
Yu et al. Resveratrol activates PI3K/AKT to reduce myocardial cell apoptosis and mitochondrial oxidative damage caused by myocardial ischemia/reperfusion injury
Zhang et al. Soybean glycinin disrupted intestinal structural integrity related to aggravation of apoptosis and downregulated transcription of tight junction proteins in the intestine of juvenile grass carp (Ctenopharyngodon idella)
CN112274523A (en) Medicine and method for treating fatty liver
Zhou et al. Dual TBK1/IKKε inhibitor amlexanox mitigates palmitic acid-induced hepatotoxicity and lipoapoptosis in vitro
Liu et al. 3-Acetyldeoxynivalenol induces lysosomal membrane permeabilization-mediated apoptosis and inhibits autophagic flux in macrophages
Chen et al. Interleukin-22 drives a metabolic adaptive reprogramming to maintain mitochondrial fitness and treat liver injury
Zhu et al. Classical swine fever virus infection induces endoplasmic reticulum stress-mediated autophagy to sustain viral replication in vivo and in vitro
Liu et al. Modulation of HMGB1 release in APAP-induced liver injury: A possible strategy of chikusetsusaponin V targeting NETs formation
Jiang et al. Zinc defends against Parthanatos and promotes functional recovery after spinal cord injury through SIRT3‐mediated anti‐oxidative stress and mitophagy
Wu et al. Pancreatic cancer cell apoptosis is induced by a proteoglycan extracted from Ganoderma lucidum
Shan et al. Caffeine in liver diseases: Pharmacology and toxicology
CN112294800A (en) Application of ellagic acid in preparing medicine for preventing and/or treating nonalcoholic fatty liver disease and liver injury
Yang et al. Carbon monoxide‑releasing molecules protect against blue light exposure and inflammation in retinal pigment epithelial cells

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: 20210202