CN113876797A - Application of astragalin in repairing oxidative damage of muscles - Google Patents

Application of astragalin in repairing oxidative damage of muscles Download PDF

Info

Publication number
CN113876797A
CN113876797A CN202111391366.2A CN202111391366A CN113876797A CN 113876797 A CN113876797 A CN 113876797A CN 202111391366 A CN202111391366 A CN 202111391366A CN 113876797 A CN113876797 A CN 113876797A
Authority
CN
China
Prior art keywords
astragalin
exercise
gsh
application
mouse
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
CN202111391366.2A
Other languages
Chinese (zh)
Inventor
张伟伟
杨清竹
黄鑫
付学鹏
邵淑丽
吴秋雪
祝锴烨
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.)
Qiqihar University
Original Assignee
Qiqihar 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 Qiqihar University filed Critical Qiqihar University
Priority to CN202111391366.2A priority Critical patent/CN113876797A/en
Publication of CN113876797A publication Critical patent/CN113876797A/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
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants

Abstract

The invention provides an application of astragalin in repairing oxidative damage of muscles, in particular to an application of astragalin in reducing liver damage caused by exhaustion exercise, namely reducing the activities of glutamic-pyruvic transaminase, glutamic-oxalacetic transaminase and alkaline phosphatase; in addition, the astragalin is applied to enhancing the function of resisting oxidation by exhaustive movement, namely enhancing the scavenging capacity of hydroxyl free radicals and the scavenging capacity of superoxide anions, and improving GSH-Px activity, GR enzyme activity, SOD enzyme activity and GSH enzyme activity; the application of astragalin in up-regulating the expression of Nrf2 and downstream gene mRNA and protein thereof; the astragalin can reduce the liver injury of an organism caused by exhaustive exercise and can enhance the oxidation resistance of the organism, so that the astragalin regulates the oxidative stress level in the muscle contraction process through the Nrf2, has the oxidation resistance activity and can partially repair the oxidative injury of a mouse after the exhaustive exercise.

Description

Application of astragalin in repairing oxidative damage of muscles
Technical Field
The invention belongs to the technical field of muscle repair, and particularly relates to application of astragalin in repairing oxidative damage of muscles, namely astragalin can repair mouse muscle damage caused by acute exhaustion exercise within the range of 50-200 mg/kg.
Background
Astragalin (AG), also called astragalin, is an important flavonoid substance, is an effective component of plant Chinese herbal medicines such as thesium chinense, eucommia ulmoides, lotus leaves and the like, and astragalin can be extracted from the fruits of thesium chinense and forsythia suspensa by an ethanol extraction method, a chemical method and the like[1][2]. Research results show that astragalin has various pharmacological effects of resisting inflammation, resisting oxidation, resisting allergic dermatitis, regulating organism immunity, protecting nerves and heart and the like[4]
Regular, moderate physical exercise has many health benefits, such as reducing the incidence of cardiovascular disease, reducing the risk of cancer and diabetes[23]. However, intense exercise can cause muscle tissue damage[24]The imbalance between the oxidation and oxidation resistance in the body is caused by the fact that Reactive Oxygen Species (ROS) generated by cells exceed the scavenging capacity of the body's own antioxidant system. The presence of ROS can cause the cells to undergo Oxidative Stress (OS), including reactions with all of DNA, lipids, and proteins[24]. Tissue damage or inflammation caused by exhaustive exercise may be referred to as short-term oxidative stress, but insufficient antioxidant responses may not reverse the toxic effects of overproduced free radicals[25]. During physical exercise, the energy requirements of the muscular system increase the oxygen consumption by 10-20 times the oxygen consumption required by the remaining system[26]This results in a large increase in the accumulation of ROS in the muscle fibers. In addition, studies have shown that body temperature is elevated due to the presence of lactic acidAnd a decrease in blood pH, the presence of this series of related substances accelerates the production of ROS, thereby increasing OS[27]. In addition, it has been suggested that an increase in myoglobin derived from damaged muscle fibers interacting with methemoglobin and peroxide during training is also involved in the production of ROS[26]. When clearance is incomplete, activation of the enzymatic endogenous antioxidant system is induced, which modulates enzymes such as GPx, GR and CAT as well as non-Enzymatic (EAS), including GSH and alpha lipoic acid, among others. Acute training as well as chronic training showed different responses to OS. Studies have shown that a single dose of antioxidant supplement can improve endurance in a trained athlete[27]. Thus, the consumption of antioxidants is important for athletes as well as untrained individuals to prevent oxidative stress caused by exercise, tissue damage and extreme decline in athletic performance.
Regular and appropriate performance of moderate physical exercise can provide many benefits to the health of a person, such as reducing the risk of cardiovascular disease, cancer and diabetes. However, vigorous exercise can cause muscle and tissue damage, where cellular ROS production exceeds the capacity of the endogenous antioxidant defense system. Tissue damage or inflammation caused by exhaustive exercise may be referred to as short-term oxidative stress, but insufficient antioxidant responses may not reverse the toxic effects of overproduced free radicals. Astragalin, also called thesin II, has multiple effective functions, can be used as a medicament and is a research hotspot at present, but the repair function of muscle cell oxidative damage is not researched yet.
Disclosure of Invention
Aiming at the defects in the prior art, the invention aims to provide the application of astragalin in the repair of muscle oxidative damage. Astragalin is a naturally-occurring flavonoid compound with multiple biological activities, and specifically, a mouse with muscle injury caused by acute exercise is taken as a research object, the repair effect of astragalin on the oxidative injury of the mouse with muscle injury is discussed, and the repair and regulation mechanism of astragalin on the oxidative injury of muscle is further researched.
In order to achieve the above purpose, the solution of the invention is as follows:
the invention aims to provide application of astragalin in repairing oxidative damage of muscles.
Furthermore, 50-200mg/kg astragalin has no damage to liver, and can enhance the anti-oxidation function of mouse muscle tissue after exhaustion exercise.
The invention also provides an application of astragalin in reducing liver injury caused by exhaustive exercise.
Further, astragalin decreases the activity of alanine Aminotransferase (ALT), aspartate Aminotransferase (AST), and alkaline phosphatase (AKP).
The invention also provides application of astragalin in enhancing the function of exhaustive exercise against oxidation.
Further, astragalin enhances the scavenging ability of hydroxyl radicals and the scavenging ability of superoxide anions.
Further, astragalin improves GSH-Px activity, GR enzyme activity, SOD enzyme activity, GSH enzyme activity.
The fourth purpose of the invention is to provide application of astragalin in up-regulating mRNA expression of Nrf2 and downstream genes thereof.
The fifth purpose of the invention is to provide application of astragalin in up-regulation of Nrf2 and expression of downstream gene proteins thereof.
Furthermore, the downstream gene is selected from more than one of GCLC, GCLM, HO-1, NQO-1, SOD1 and SOD 2.
Due to the adoption of the scheme, the invention has the beneficial effects that:
the astragalin can reduce the liver injury of the organism caused by exhaustion exercise and can enhance the oxidation resistance of the organism. The expression level of Nrf 2mRNA and protein is increased after the astragalin is perfused, so that the astragalin regulates the oxidative stress level in the muscle contraction process through Nrf2, namely, the astragalin is adopted to promote Nrf2 to regulate the expression of GCLM, GCLC, NQO-1, HO-1, SOD1 and SOD2 genes to increase the oxidation resistance of the body. Therefore, the astragalin has antioxidant activity and can partially repair the oxidative damage of mice after exhaustive exercise.
Drawings
FIG. 1 is a schematic diagram showing the effect of astragalin of the present invention on liver injury in mice.
FIG. 2 is a graph showing the effect of different exercise times of the present invention on the amount of superoxide anion produced in mice.
FIG. 3 is a diagram showing morphological changes before and after exercise in a mouse of the present invention (FIG. A: before Ex; FIG. B: after Ex).
FIG. 4 is a graph of astragalin of the present invention reducing liver damage following sports injury in mice.
FIG. 5 is a graph showing the effect of astragalin of the present invention on the intramuscular OH scavenging ability of mice after sports injury.
FIG. 6 shows the intramuscular O of mice after the astragalin pair sports injury of the invention2-Influence graph of cleaning ability.
FIG. 7 is a graph showing the effect of astragalin of the present invention on the activity of GSH-PX enzyme in mouse muscle after sports injury.
FIG. 8 is a graph showing the effect of astragalin of the present invention on GR enzyme activity in mouse muscle after sports injury.
FIG. 9 is a graph of the effect of astragalin of the present invention on the intramuscular GSH content of mice following exercise injury.
FIG. 10 is a graph showing the effect of astragalin of the present invention on the activity of SOD enzyme in mouse muscle after sports injury.
FIG. 11 is a graph showing that the expression of the mouse muscle Nrf2 and downstream gene mRNA in different treatment groups is detected by the method.
FIG. 12 is a graph showing that the expression of the mouse muscle Nrf2 and downstream gene proteins in different treatment groups are detected by the invention.
FIG. 13 is a schematic view of the approach of astragalin to repair oxidative damage in the body of the invention.
Detailed Description
The invention provides an application of astragalin in repairing oxidative damage of muscles. The invention prepares a mouse muscle injury model through exhaustive exercise, and detects superoxide anions (O) in a mouse body caused by different exercise time by an enzyme chemistry method2-) The content of the extract is detected by detecting whether liver injury occurs after gastric lavage of astragalin, and OH and O in muscle tissue are detected by an enzyme chemical method2-The content of GSH, the enzyme activities of SOD, GSH-Px and GR, and the repairing effect of astragalin on the oxidative damage of muscle tissues.
Astragalin, also known as kaempferol-3-O-glucoside, has a relative molecular mass of Mr.448.38 and a molecular formula of C21H20O11The molecular structural formula is shown as follows. The product is yellow needle crystal at normal temperature, has melting point of 218-220 deg.C and boiling point of 823.2 deg.C, is insoluble in water, and is easily soluble in organic solvents, such as dimethyl sulfoxide (DMSO), methanol and ethanol.
Figure BDA0003368957900000031
1. Pharmacological action of astragalin
(1) Antioxidant effect of astragalin
Overproduction of free radicals may affect the balance of antioxidant and antioxidant systems in the human body, leading to various pathological conditions such as arterial hypertension, rheumatism, inflammation, diabetes, cancer, neurodegenerative diseases and genetic mutations. However antioxidants can act as radical scavengers and chelators in biological systems[6]. At present, natural plants with antioxidant properties are considered to have great potential, not only can reduce the occurrence of diseases, but also can play a role in protecting organisms. Therefore, the development and utilization of ingredients in natural plants with antioxidant properties has been a focus of research.
Flavonoid compounds have been widely used as antioxidants in living organisms, and are very effective in scavenging active oxygen radicals in the organism and synergistically acting with peroxy radicals to terminate radical chain reaction in autoxidation of unsaturated fatty acids[7]Thereby protecting the cell membrane from damage. Astragalin is a flavonoid substance, and high performance liquid chromatography ultraviolet detection (HPLC-UV) finds that astragalin and DNA are combined through interaction with G-C base pair, possibly through hydrogen bond to form stable intercalation[8]. At the same time astragalin pair CCl4Has anti-hemolytic and liver toxic effects and is resistant to diseases[9]. The oxidation hemolysis of normal human erythrocytes induced by the superoxide radical AAPH can be inhibited by astragalin in a time-and dose-dependent manner. Astragalin can also prevent depletion of glutathione, a vesicolysin type antioxidant in erythrocytes[3]. Protecting the depletion of the cytoplasmic antioxidant Glutathione (GSH) in erythrocytes[5]. To H2O2Induced active oxygen generation inhibition, and significantly reduced H2O2Induced reactive oxygen species production in HEK-293 cells[10]. Astragalin was shown to be effective against acute I/R injury in Sprague Dawley rats and to exert its effect by reducing intracellular oxidative stress and apoptosis. Expressed as a decrease in MDA, TNF-alpha, IL-6, ROS and Bax expression, and an increase in the GSH/GSSG ratio[11]
(2) Anti-inflammatory effect of astragalin
Inflammation is the body's direct response to tissue damage caused by pathogens and noxious stimuli (e.g., physical or chemical injury). Although the inflammatory response is a defense mechanism, if persisting, it can lead to a variety of pathological conditions, such as cancer, allergy, atherosclerosis, and autoimmune diseases[12]. Studies show that astragalin has anti-inflammatory activity, and can reduce inflammation induced by Lipopolysaccharide (LPS) in mastitis mouse model and lung injury model by reducing the activity of peroxidase and adiponectin. Can also alleviate the deterioration of Ik-B alpha and limit nuclear translocation of NF-kappa B by inhibiting NF-kappa B activation induced by LPS, thereby further playing the anti-inflammatory role of astragalin[13]. Another related study result shows that macrophage can inhibit activation of NF-kB signal channel through astragalin after being stimulated by LPS, thereby inhibiting expression of some related proinflammatory mediators in cells[4]. At the same time, astragalin can prevent MAPK and NF-kB pathways in mouse leptospira-induced inflammation of uterus and epithelium[14,15]. Has the ability to inhibit the production of prostaglandin E2(PGE2) in periodontal pathogen-induced periodontitis[16]. Can effectively lowerReducing eosinophil count in lung tissue and inhibiting ovalbumin-induced eosinophilia[17]. Can inhibit the activity of PGE2 in RAW 264.7 cells stimulated by LPS, and down-regulate the production of nitrite and IL-6 in cells[18]. Astragalin treatment inhibited alveolar destruction, allergic inflammation and airway thickening in an ovalbumin-induced inflammatory mouse model[17]
(3) Immune regulation effect of astragalin
Astragalin is an effective component for treating body diseases, and can enhance the immunity of organisms. The addition of astragalin (5-0.05 μ g/ml) with proper concentration to human peripheral blood NK cells has remarkable promoting effect on the activity thereof, and the action intensity of astragalin is related to the immunity level of human body[19]. But also can reduce the leucocytosis caused by resisting cyclophosphamide and 60CO gamma rays in peripheral blood, inhibit the peripheral blood lymphocytosis of mice and obviously improve the tolerance of the mice when being subjected to external stimulation such as high temperature and cold[20](ii) a And can clearly show that the proliferation of pulmonary artery smooth muscle cells and the production and release of collagen caused by hypoxia are inhibited[21](ii) a And can improve phagocytic ability of macrophage[22]
2. Antioxidant enzyme system
The crucial factor for the body to be able to exert its normal action is due to the steady state of ROS, and many types of antioxidant systems known to have a major impact are currently studied to exist in the body. One class is antioxidase, including SOD, CAT, GPx, glycomacropeptide reductase GR, and reduced coenzyme II NADPH, etc. Of these, the catalase CAT plays a key role in protecting cells from the toxic effects of hydrogen peroxide. NADPH cannot be separated from Glutathione (GSH) which is an important reducing agent synthesized by cells and Thioredoxin (TXN) which is thioredoxin[28]It has been found that interference with NADPH synthesis affects cell division and mitochondrial membrane permeability, increases cellular sensitivity to oxidative stress and can cause apoptosis[29]. Superoxide dismutase SOD is a ubiquitous antioxidant enzyme capable of disproportionating superoxide free radicalsIs hydrogen peroxide (H)2O2)[30]. GSH-Px is a decomposition enzyme existing in cells, belongs to one of antioxidant enzyme systems, and can accelerate H2O2Reaction with GSH to H2Rates of O and oxidative GSSG, followed by Glutathione (GR) conversion of GSSG back to GSH. glutathione-S transferase catalyzes electrophilic conjugation of GSH to various substrates through sulfhydryl, thereby reducing content of peroxide[31]. Exposure of cells, tissues and extracellular matrix to harmful reactive species leads to a cascade of reactions and induces activation of various internal defense mechanisms (enzymatic or non-enzymatic), thereby removing reactive species and their derivatives. Non-enzymatic antioxidants are represented by molecules characterized by their ability to rapidly deactivate free radicals and oxidants. These non-enzymatic antioxidants also play an extremely important role in maintaining the redox balance within the cell[32]. For example, endogenous non-enzymatic antioxidants include Metal Binding Proteins (MBPs), Glutathione (GSH), Uric Acid (UA), Bilirubin (BIL), and Polyamines (PAs). The antioxidant molecule GSH is the most abundant non-enzyme substance in the organism, and the existence of GSH is crucial to maintain the redox balance reaction of the organism[33]. Reduced GSH is the major low molecular weight thiol in cells, acting as a nucleophilic scavenger and an enzyme-catalyzed antioxidant in oxidative tissue damage. Thus, GSH plays an important role as a protector of biological structure and function[34]. A plurality of regulatory factors capable of regulating dynamic balance among redox reactions in cells exist in organisms, and a plurality of regulatory factors including Nrf2, p53 and the like can regulate the redox balance of the cells[35]. Of these regulators, Nrf2 is the most powerful of the oxidative stress responses[36]Nrf2 has a regulatory role in many important antioxidant pathways in the body.
Mechanism of action of Nrf2 activity in regulation of redox balance
Nuclear factor E2-related factor 2(Nrf2) is a powerful nuclear transcription factor that coordinates the antioxidant cytoprotective system complex stimulated by increased OS. Nrf2 can promote GCLM (modified subbunit) and GCLC (catalytic subbunit) to be in cellsThe reason why Nrf2 can directly control GSH synthesis is that GCL is the rate-limiting enzyme catalyzing the reaction of glutamic acid and cysteine to produce GSH[37]. Meanwhile, Nrf2 can indirectly control the synthesis of GSH in cells, GSH is catalyzed and synthesized into GSSG by antioxidant enzyme and reduces the level of ROS in cells, but GSSG can be catalyzed and synthesized into GSH by NADPH. Therefore, Nrf2 can indirectly regulate GSH production by interfering with NADPH production levels. "Fenton reaction" is an important chemical reaction in organisms that eliminates ROS, Fe2+Participating in the important "fenton reaction" in the cell: fe2++H2O2→Fe3++ -OH + HO. The presence of Nrf2 can increase the rate of HMOX1 gene expression[38]Therefore, Nrf2 can not only control the decomposition of heme to produce Fe2+And can indirectly promote the 'Fenton reaction' of cells and can convert Fe3+Accumulate in large amounts and thus have a higher efficiency in scavenging excess intracellular ROS. Nrf2 is the most important regulator in the collective redox reaction equilibrium.
Therefore, the invention takes the exhaustion exercise mouse as a research object, discusses the repair effect of astragalin on muscle injury of the exhaustion exercise mouse, analyzes the action mechanism of astragalin and lays a foundation for the development and application of astragalin.
Exhaustive exercise can cause tissue damage or inflammation, produce excessive free radicals, and cause toxic effects, and the attack of free radicals can cause continuous damage to cells, thereby causing the body to begin to produce oxidative stress. Research shows that astragalin has multiple medicinal functions and can treat some diseases generated by the body, such as the oxidation resistance and the regulation of the cellular immunity function of the body by the cooperation of an oxidative stress signal path in the body when the body generates oxidative stress reaction. The invention researches the repair effect of astragalin on oxidative damage in a mouse body after exhaustion exercise.
The technical content of the present invention will be further described with reference to examples. The following examples are illustrative and not intended to be limiting, and are not intended to limit the scope of the invention. The experimental procedures used in the following examples are all conventional procedures unless otherwise specified. Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
Example 1:
experimental Material
1. Laboratory animal
SPF grade c57BL/6J (9 week old) male mice were purchased from Changchun Biotechnology Ltd.
2. The experimental reagent fetal bovine serum is purchased from Biological Industries, the DMEM dry powder is purchased from GIBCO, the vitamin E is purchased from Beijing Soilebao technology, the SOD, GSH, GR, GSH-Px, NCR-cytochrome c reductase, hydroxyl radical and superoxide anion detection kit is purchased from Nanjing to build a bioengineering institute, the MTT method cell viability detection kit is purchased from Biyunnan biotechnology institute, and the astrin is purchased from Shanghai-sourced leaf biotechnology.
Preparation of astragalin mother liquor
10 mu L of DMSO was dissolved in 10mL of physiological saline, and then 500 mu L of 0.1% DMSO was dissolved in 50mg of astragalin to prepare a suspension with a concentration of 5mg/mL, which was used for gavage mice.
3. Laboratory apparatus
Spark 10M microplate reader (tiken, switzerland), inverted fluorescence microscope (OLYMPUS, japan), carbon dioxide incubator (Thermo Electron, usa), fluorescent quantitative PCR instrument (Eppendorf, usa), Odyssey dual infrared laser imaging system (LI-COR, usa).
4. Experimental methods
Experimental group mouse movement scheme
Six male SPF grade c57BL/6J (9 week old) mice were housed in a cage (27 x 17 x 13cm) and the experimental procedure followed the ethical review guide for animal experimental welfare of the scientific and ethical committee of zizahal university. Subsequently, the mice were randomly divided into 4 groups: DMSO group, Exercise (EX), EX + AG 50mg/kg group and EX + AG 100mg/kg (n ═ 6). All mice were familiar with swimming training one week prior to the experiment. Mice from the AG and EX + AG groups were euthanized with AG (100mg/kg and 50mg/kg body weight) after exhaustive exercise and sacrificed 1h after the effect to allow sufficient time for the potential effect to appear before the drug half-life was over. Mice in DMSO and EX groups were fed with water at the same time as other groups, mice in EX and EX + AG groups were subjected to blood sampling in abdominal artery after swimming until exhaustion of muscle and fatigue and supplementing with AG or water for 1h, centrifuged at 4 deg.C and 5000r/min for 5min to obtain serum, and simultaneously liver and gastrocnemius muscles were rapidly excised and frozen in liquid nitrogen. All tissue and plasma samples were stored at-80 ℃ until analysis.
Example 2:
effect of astragalin on liver function of mice
1. Measurement of alanine Aminotransferase (ALT)
(1) 1h after gavage, mice were sacrificed, and blood was taken from the abdominal artery and placed on ice;
(2) the experimental system was set up as in table 1.
TABLE 1 ALT Experimental System
Figure BDA0003368957900000071
And (3) uniformly mixing the cells in a 96-well plate, standing and culturing for 15min at room temperature, measuring the OD value of each well at the position with the wavelength of 510nm by using a microplate reader, determining the OD value of each well as the OD value of each measurement well to the OD value of a control well, and obtaining the AST unit activity of the sample according to a standard curve.
2. Measurement of aspartate Aminotransferase (AST)
(1) 1h after gavage, mice were sacrificed, and blood was taken from the abdominal artery and placed on ice;
(2) the experimental system was set up as in table 2.
TABLE 2 AST Experimental systems
Figure BDA0003368957900000072
Figure BDA0003368957900000081
3. Determination of alkaline phosphatase (AKP)
(1) 1h after gavage, mice were sacrificed, and blood was taken from the abdominal artery and placed on ice;
(2) the experimental system was set up as in table 3.
TABLE 3 AKP Experimental System
Figure BDA0003368957900000082
And (3) uniformly mixing 96-well plates to draw a splay pattern, measuring the absorbance OD value of each well by using an enzyme-labeling instrument at the wavelength of 520 nm.
Example 3:
influence of astragalin on mouse antioxidant function
1. Determination of hydroxyl radical (. OH)
(1) Preparation of 10% homogenate:
according to the weight and volume of 1: 9, accurately weighing the tissue, adding 9 times of physiological saline, preparing tissue homogenate, centrifuging at 2500r/min for 10min, sucking supernatant, discarding precipitate, and testing.
(2) Set up the experimental system as in Table 4
TABLE 4 OH Experimental System
Figure BDA0003368957900000083
Mixing, standing at room temperature for 20min with wavelength of 550nm and optical path of 1cm, adjusting to zero with distilled water, and measuring absorbance value of each tube.
(3) Calculating the formula:
Figure BDA0003368957900000091
2. superoxide anion (O)2-) Measurement of (2)
(1) Preparation of 10% homogenate as determined by OH of 1;
(2) the experimental system was set up as in table 5.
TABLE 5O2-Experimental System
Figure BDA0003368957900000092
After mixing, standing at room temperature for 10min with wavelength of 550nm and optical path of 1cm, adjusting to zero with distilled water, and measuring absorbance value of each tube.
(3) Calculating the formula:
Figure BDA0003368957900000093
3. determination of superoxide dismutase (SOD)
(1) Preparation of 10% homogenate as determined by OH of 1;
(2) the experimental system was set up as in table 6.
TABLE 6 SOD test System
Figure BDA0003368957900000094
Transferring 240 μ L of the solution to a 96-well plate at 37 deg.C for 20min, measuring OD with an enzyme-labeling instrument at 450 nm.
(3) Calculating the formula:
Figure BDA0003368957900000101
Figure BDA0003368957900000102
4. assay for glutathione peroxidase (GSH-Px)
(1) Preparation of 10% homogenate as determined by OH of 1;
(2) the experimental system was set up as in table 7.
TABLE 7 GSH-Px Experimental systems
Figure BDA0003368957900000103
After mixing, centrifuging for 10min at 4000r/min, and after centrifuging, taking 1mL of supernatant for subsequent color reaction.
(3) Set up the color reaction system as shown in Table 8
TABLE 8 GSH-Px color reaction System
Figure BDA0003368957900000104
After mixing, the total volume of 2.3mL per well was transferred to a 6-well plate at a wavelength of 412nm, and the OD value of each well was measured with a microplate reader.
(4) Calculating the formula:
Figure BDA0003368957900000105
5. determination of Glutathione (GSH) content
(1) Preparation of 10% homogenate as determined by OH of 1;
(2) the experimental system was set up as in table 9.
TABLE 9 GSH Experimental systems
Figure BDA0003368957900000111
Standing for 5min, with the wavelength of 405nm, and measuring the OD value of each well by using an enzyme-labeling instrument.
(3) Calculating the formula:
Figure BDA0003368957900000112
6. determination of Glutathione Reductase (GR)
(1) Preparation of 10% homogenate as determined by OH of 1;
(2) the experimental system was set up as in table 10.
TABLE 10 GR test System
Figure BDA0003368957900000113
(3) UV spectrophotometer, reading absorbance value A1 at 30s at 340 nm. Placing in an accurate water bath at 37 ℃ for 2min, and reading the absorbance value A2 at 150 s.
(4) Calculating the formula:
Figure BDA0003368957900000114
6.22 is the extinction coefficient of 1mM NADPH at 340nm wavelength 1cm optical path.
Example 4:
effect of astragalin on mRNA expression of Nrf2 and downstream genes in mouse muscle
1. Total RNA extraction
(1) Muscle tissue of mice in DMSO group, sport (EX), EX + AG 50mg/kg group and EX + AG 100mg/kg group was taken out from-80 ℃, put into a DEPC-treated mortar, ground into fine powder with liquid nitrogen, and then added with 1mL of TRIzol Reagent (invitrogen, USA), mixed well, and lysed on ice for 30 min.
(2) Adding 200 μ L chloroform, incubating on ice for 15min, 12000r/min, and centrifuging at 4 deg.C for 15 min.
(3) Add the supernatant to a new 1.5mL centrifuge tube, add 500. mu.L isopropanol, mix well, incubate for 15min on ice.
(4) Centrifuging at 12000r/min at 4 deg.C for 10 min.
(5) The supernatant was discarded, washed with 75% ethanol 2 times at 4 ℃ and 12000r/min, and centrifuged for 5 min.
(6) After the ethanol is completely volatilized, 20 mu L of DEPC-ddH is added2And O, measuring the RNA concentration.
2. Reverse transcription
Taking RNA of muscle tissues of mice of an extracted DMSO group, a sports (EX), an EX + AG 50mg/kg group and an EX + AG 100mg/kg group as templates, carrying out reverse transcription to obtain cDNA, and carrying out the detailed steps as follows:
the extracted RNA was heat-denatured at 65 ℃ for 5min, and immediately cooled on ice. For the first use, 8.8. mu.L of gDNA Remover was added to 4 XDN Master Mix (440. mu.L), and then a reaction solution was prepared on ice, including 2. mu.L of 4 XDN Master Mix, 0.5. mu.g of RNA template, and nucleic-free Water was added to make up the total volume to 8. mu.L, after mixing, incubation was carried out at 37 ℃ for 5min, after completion, immediately placed on ice, 2. mu.L of 5 XRT Master Mix II was added, reaction was carried out at 37 ℃ for 15min, then reaction was carried out at 98 ℃ for 5min, and finally storage was carried out at 4 ℃.
qRT-PCR detection:
taking cRNA of mouse muscle tissues of a DMSO group, a sports (EX), an EX + AG 50mg/kg group and an EX + AG 100mg/kg group obtained by reverse transcription as a template, carrying out PCR amplification, and detecting the expression of Nrf2 and mRNA of a downstream antioxidation related gene thereof, wherein the detailed steps are as follows:
the reverse transcription product cDNA was prepared according to 1: 2 plus ddH2O dilution and then qRT-PCR as per Table 11.
TABLE 11 qRT-PCR reaction System
Figure BDA0003368957900000121
Reaction conditions are as follows: at 95 ℃ for 2 min; 40 × (95 ℃, 15 s; 60 ℃, 40s), 95 ℃, 15 s; 60 ℃ for 1 min; the dissolution profile was added after the cycle was complete.
TABLE 12 real-time fluorescent quantitation primer sequences
Figure BDA0003368957900000122
Figure BDA0003368957900000131
Example 5:
influence of astragalin on expression of Nrf2 and downstream gene proteins in mouse muscle
1. Tissue whole protein extraction
(1) The tissue sample was ground thoroughly with liquid nitrogen and added to the protein lysate. Placing on a shaking bed, cracking at 4 deg.C for 30min, 13000r/min, centrifuging for 10min, collecting supernatant, and storing at-80 deg.C for use.
2. Protein concentration determination:
(1) protein concentration assay standards were prepared as shown in table 13.
(2) A clean 1.5mL EP tube was added with 10. mu.L and 40. mu.L of ddH2O, respectively, and mixed well.
(3) A 96-well plate was used, and 25 μ L of the mixed sample and 200 μ L of the mixed solution were added to each well (solution a: solution B: 50: 1).
(4) Incubating in an incubator at 37 ℃ for 30min, and measuring the OD value at 562nm by using an enzyme-labeling instrument.
(5) And drawing a standard curve, and leveling the protein concentration according to the standard curve.
TABLE 13 protein concentration measurement Standard configuration
Figure BDA0003368957900000132
SDS-PAGE electrophoresis
(1) Preparing separation gel, and sealing the layer with isopropanol.
(2) After the gel is formed, the isopropanol on the upper layer is discarded, the concentrated gel is prepared, and the gel is inserted into a gel hole comb.
(3) After waiting for gelation, the electrophoresis solution was poured in, and then the inserted comb was pulled out, followed by spotting of the protein sample at 30. mu.L/well.
(4) And (5) switching on a power supply, regulating the voltage to 80V, and running the glue at constant voltage. When the bromophenol blue reaches the bottom end, a stop switch is pressed, and the glue running is stopped.
TABLE 14 Release glue formulations
Figure BDA0003368957900000133
TABLE 15 concentrated gum formulations
Figure BDA0003368957900000134
Figure BDA0003368957900000141
4. Rotary film
(1) And cutting the gel according to the size of the detection antibody band.
(2) The sponge, the filter paper (4 layers), the glue, the PVDF membrane, the filter paper and the sponge are placed in a rotating membrane clamp in sequence.
(3) And (3) mounting the assembled film transferring clamp in a transfer printing groove according to the sequence of red-to-red and black-to-black, and pouring the film transferring liquid.
(4) Electrifying, and crossly flowing 300mA for 90 min.
Western hybridization
(1) The blocking solution (containing 5% skimmed milk powder) was incubated for 1 h.
(2) Primary antibody was added and incubated overnight at 4 ℃.
(3) 0.2% TBS-Tween was washed 3 times for 15 min/time.
(4) The secondary antibody was incubated at 4 ℃ for 1 h.
(5) 0.2% TBS-Tween was washed 3 times for 15min each.
(6) The strips were examined with an Odyssey infrared fluorescence scanner and gray-scale scanned.
Statistical analysis
The experimental data are expressed as Mean ± standard deviation (Mean ± SD), analyzed with SPSS Statistics 22.0 software, and differences were considered statistically significant with p <0.05, plotted with Graphpad Prism 5.0 software.
Results of the experiment
1. Detection of astragalin on mouse liver injury
In order to detect whether astragalin damages a mouse, the activities of ALT, AST and AKP in the serum of the mouse are detected by an enzyme chemical method after different concentrations of astragalin are perfused, and the results are shown in figure 1, compared with a 50% ethanol control group, the astragalin with different concentrations of perfused stomach of 50mg/kg, 100mg/kg and 200mg/kg does not damage the liver of the mouse, and the astragalin has no toxic effect on the body of the mouse.
2. Preparation of mouse sports injury model
The preparation of the acute exhaustion exercise injury model of the mouse, the enzyme chemistry detection of the influence of different exercise time on the production of superoxide anions in the serum of the mouse is shown in figure 2, and the result shows that the content of superoxide anions produced in the serum of the mouse is increased (p is less than 0.01) along with the extension of the exercise time of the mouse in the exercise time range of 2-4h and the superoxide anions are produced in a time-dependent manner.
3. Morphological changes before and after movement of mice
The shape change of the mice before and after exercise is shown in figure 3, and before the exercise, the diet and the drinking water of each group of mice show a normal phenomenon; during the experiment, all groups of mice normally move, and the phenomena of anorexia, dysphoria and the like do not occur; after the movement, the mouse feels tired and weak on the back and does not move any more; after the body of a mouse is dissected, no abnormal change is observed in organs such as heart, lung, liver, kidney and the like, and the astragalin is proved to be safe to use.
4. Astragalin can reduce the influence of exhaustive exercise on the liver injury of mice
The influence of astragalin on liver injury of mice after exercise is shown in fig. 4, and compared with a control group, ALT and AST activities in mice after exhaustion exercise are remarkably increased (p is less than 0.01); ALT and AST activity was significantly reduced after gavage astragalin compared to the exhaustion exercise treatment group (p < 0.01). The results show that the liver of the mouse is damaged by the exhaustive exercise, and astragalin can reduce the liver damage caused by the exhaustive exercise.
5. Influence of astragalin on mouse antioxidant function
5.1 Effect of astragalin on mouse hydroxyl radical (. OH) scavenging ability
The influence of astragalin on the scavenging capacity of the muscle hydroxyl free radicals after the exhaustion exercise of the mice is shown in figure 5, and compared with a DMSO group, the scavenging capacity of the muscle hydroxyl free radicals of the exhaustion exercise group is obviously reduced (p is less than 0.01); compared with the control group of exhaustion exercise, the astragalin can obviously enhance the scavenging capacity of the hydroxyl free radicals of the muscle of the mice after exhaustion exercise (p is less than 0.01).
5.2 Astragaloside superoxide anion (O) of mouse muscle2-) Influence of scavenging ability
The effect of astragalin on the superoxide anion scavenging ability of the muscle of mice after exhaustion exercise is shown in figure 6, and compared with DMSO control group, the muscle O of exhaustion exercise group2-The clearance capacity is significantly reduced (p)<0.01); compared with the control group of exhaustion exercise, the astragalin can remarkably enhance the mice after exhaustion exerciseMuscle O2-Clearing ability (p)<0.01)。
5.3 Effect of astragalin on mouse muscle GSH-Px, GR Activity
The influence of astragalin on the activity of GSH-Px enzyme of muscle after exhaustion exercise of mice is shown in figure 7, and compared with a DMSO control group, the activity of GSH-Px in the muscle of mice of an exhaustion exercise group is obviously reduced (p is less than 0.01); compared with the exhaustive exercise treatment group, the periplocoside can obviously increase the mouse muscle GSH-Px activity expression (p is less than 0.01).
The effect of astragalin on GR activity of the post-exercise muscles of the mice is shown in fig. 8, and compared to the DMSO control group, GR activity was significantly reduced in the mice of the exhaustive exercise treated group (p < 0.05); the periplocin can increase GR enzyme activity in the mice muscle after exercise (p <0.01) compared with the exhaustion exercise treatment group.
5.4 Effect of astragalin on mouse muscle GSH content
The influence of astragalin on the muscle GSH content of mice after exhaustion exercise is shown in figure 9, and compared with a DMSO control group, the muscle GSH content of mice in an exhaustion exercise treatment group is obviously reduced (p is less than 0.05); compared with the exhausted exercise treatment group, the periplocin can obviously increase the muscle GSH content of the mice after exercise (p is less than 0.05).
5.5 Effect of astragalin on mouse muscle SOD Activity
The effect of astragalin on mouse post-exercise muscle SOD enzyme activity is shown in figure 10, compared with DMSO control group, SOD activity in mouse muscle tissue of exhaustive exercise treatment group is reduced, compared with exhaustive exercise treatment group, gastric perfusion of 50mg/kg astragalin can increase mouse muscle SOD enzyme activity (p < 0.05).
6. Effect of astragalin on mouse muscle Nrf2 and downstream gene mRNA expression
The effect of astragalin on the mouse muscle antioxidant gene Nrf2, GCLC, GCLM, HO-1, NQO1, SOD1 and SOD2mRNA expression is shown in figure 11, and it can be seen from the figure that the antioxidant gene Nrf2, GCLC, GCLM, HO-1, NQO1, SOD1 and SOD2mRNA expression in muscle after exhaustive exercise is significantly reduced (p <0.05) compared with DMSO control group; compared with the exhaustion exercise treatment group, the expression of Nrf2 and the downstream gene mRNA thereof is obviously increased after the astragalin is perfused (p is less than 0.01).
7. Influence of astragalin on expression of mouse muscle Nrf2 and downstream gene protein
The influence of astragalin on the expression of mouse muscle antioxidant gene Nrf2 and downstream gene protein is shown in figure 12, and the graph shows that compared with a control group, the expression of the muscle antioxidant Nrf2, GCLC, GCLM, HO-1, NQO1, SOD1 and SOD2 gene protein is obviously reduced (p is less than 0.05) after exhaustion exercise; compared with the exhaustion exercise treatment group, the protein expression of Nrf2, GCLC, GCLM, HO-1, NQO1, SOD1 and SOD2 is obviously increased after the gastric lavage astragalin injection (p is less than 0.01).
In conclusion, a mouse movement injury model is prepared, and O generated in mouse serum at different movement time is detected by an enzyme chemistry method2-Content, exercise injury time 4h, O in mouse serum2-The highest content, and the time of sports injury is determined to be 4 h. Before exercise, all groups of mice normally move without the phenomena of appetite reduction, dysphoria and the like, and after the exercise is exhausted, the mice feel tired and weak in back and do not do any exercise any more. After the astragalin with different concentrations is perfused into the stomach, the influence of the astragalin on the activity of ALT, AST and AKP in the serum of the mouse is detected, compared with a positive control group of 50% ethanol, 50mg/kg, 100mg/kg and 200mg/kg of astragalin do not cause damage to the liver of the mouse, and the fact that the astragalin has no toxic effect on the body of the mouse is proved; detection of muscle tissue OH and O after exhaustive exercise by enzymatic chemistry2-The scavenging ability is reduced, the GSH content is reduced, and the enzyme activities of GSH-Px, SOD and GR are reduced; gavage after exhaustion exercise 50mg/kg and astragalin 100 mg/kg-OH and O2-The scavenging ability is enhanced, the GSH content is increased, and the enzyme activities of GSH-Px, SOD and GR are increased; compared with the exhaustion exercise treatment group, after gastric lavage, qRT-PCR and Western Blot results show that the mRNA and protein expression levels of Nrf2 dependent antioxidant genes NQO1, HO-1, GCLC, GCLM, SOD1 and SOD2 genes in liver and muscle are averagely up-regulated.
In summary, as shown in FIG. 13, H2O2After the organism is damaged, astragalin is added to promote Nrf2 to regulate and control the expression of GCLM, GCLC, NQO-1, HO-1, SOD1 and SOD2 genes so as to up-regulate the oxidation resistance of the organism.
Reference documents:
[1] chapter force. extraction process of effective components in thesium chinense dynamics simulation and structural identification [ D ]. Wuhan, Wuhan engineering university, 2019.
[2] Zhang Xiao Hu, Wang Chao, Shanlo Lily fruit astragalin extraction method research [ J ]. Shanxi agricultural science, 2018, 64 (05): 11-15.
[3]Choi J.,Kang H.J.,Kim S.Z.et al..Antioxidant effect of astragalin isolated from the leaves of Morus alba L.against free radical-induced oxidative hemolysis of human red blood cells[J].Archives of pharmacal research,2013,36(7):912-917.
[4]Soromou L.W.,Chen N.,Jiang L.et al..Astragalin attenuates lipopolysaccharide-induced inflammatory responses by down-regulating NF-kappa B signaling pathway[J].Biochemical and biophysical research communications,2012,419(2):256-261.
[5]Kotani M.,Matsumoto M.,Fujita A.et al..Persimmon leaf extract and astragalin inhibit development of dermatitis and Ig E elevation in NC/Nga mice[J].The Journal of allergy and clinical immunology,2000,106(1Pt 1):159-166.
[6]Sarfraz I.,Rasul A.,Jabeen F.et al..Fraxinus:a plant with versatile pharmacological and biological activities[J].Evidence-Based Complementary and Alternative Medicine,2017.4269868.
[7]Choi J.,Kang H.J.,Kim S.Z.et al..Antioxidant effect of astragalin isolated from the leaves of Morus alba L.against free radical-induced oxidative hemolysis of human red blood cells[J].Archives of Pharmacal Research,2013,36(7):912–917.
[8]Saito S.,Silva G.,Santos R.X.et al..Astragalin from Cassia alata Induces DNA Adducts in Vitro and Repairable DNA Damage in the Yeast Saccharomyces cerevisiae[J].International journal of molecular sciences,2012,13(3):2846-2862.
[9]Kumar S.,Pandey A.K.Chemistry and biological activities of flavonoids:an overview[J].ScientificWorldJournal,2013::162750.
[10] Zhang Ying, Shenjian Ping, Wang Xiaolin, et al, research on the action of astragaloside against biological oxidation [ J ] pharmacology and clinics of Chinese medicine, 1991, 7 (6): 135-136.
[11]Ivanova A.,Mikhova B.,Najdenski H.et al..Chemical composition and antimicrobial activity of wild garlic Allium ursinum of Bulgarian origin[J].Natural Product Communications,2009,4(8):1059–1062.
[12]Qu D.,Han J.,Ren H.et al..Cardioprotective effects of astragalin against myocardial ischemia/reperfusion injury in isolated rat heart[J].Oxidative Medicine and Cellular Longevity,2016,11pages.
[13]Audial S,.Bonnotte B.Inflammation[J].Nature,2015,65(3):403-408.
[14]F Li.,D Liang.,Z Yang.et al..Astragalin suppresses in-flammatory responses via down-regulation of NF-κB signaling pathway in lipopolysaccharide-induced mastitis in a murine model[J].International Immunopharmacology,2013,478–482,.
[15]Zhang W.,Lu X.,Wang W.et al..Inhibitory effects of emodin,thymol,and astragalin on leptospira interrogansinduced inflammatory response in the uterine and endometrium epithelial cells of mice[J].Inflammation,2017,40(2):666–675.
[16]Inaba H.,Tagashira M.,Honma D.et al..Identification of hop polyphenolic components which inhibit prostaglandin E2 production by gingival epithelial cells stimulated with periodontal pathogen[J].Biological and Pharmaceutical Bulletin,2008,31(3):527–530.
[17]Lee H.B.,Kim E.K.,Park S.J.et al..Isolation and anti-inflammatory effect of astragalin synthesized by enzymatic hydrolysis of tea seed extract[J].Journal of the Science of Food and Agriculture,2011,91(13):2315–2321.
[18]Kim Y.H.,Choi Y.J.,Kang M.K.et al..Astragalin inhibits allergic inflammation and airway thickening in ovalbuminchallenged mice[J].Journal of Agricultural and Food Chemistry,2017,65(4):836–845.
[19] Yuliefin, Zhouyou Yao xi, Zhang Ying, etc. research on the influence factors of astragaloside on the natural killer activity of human peripheral blood [ J ], journal of Chinese immunology, 1990, 6(1):60-63.
[20] Zhang Ying, Xuanwu, Liu Xiao Hao, etc. St.Astragaloside raises leukocyte and has anti-stress effect research [ J ]. proceedings of Nanjing medical college, 1992, (03): 244-.
[21] Influence of astragalin on proliferation and collagen secretion of hypoxia-cultured human pulmonary artery smooth muscle cells in vitro [ J ]. J.Med.Med.Med.Med.J., 1997 (S1):206 + 207+301.
[22]Testai L..Flavonoids and mitochondrial pharmacology:A new paradigm for cardioprotection[J].Life Sciences,2015,135:68–76.
[23]Suzuki K.Chronic inflammation as an immunological abnormality and effectiveness of exercise[J].Biomolecules,2019,9(6):223.
[24]
Figure BDA0003368957900000181
J.,Gimeno A.,Sastre J.et al..Mechanism of free radical production in exhaustive exercise in humans and rats;role of xanthine oxidase and protection by allopurinol[J].IUBMB Life,2000,49(6):539–544.
[25]Banerjee A.K.,Mandal A.,Chanda D.et al..Oxidant,antioxidant and physical exercise[J].Molecular and cellular biochemistry,2003,253(1-2):307–312.
[26]Powers S.K.,Jackson M.J.Exercise-induced oxidative stress:Cellular mechanisms and impact on muscle force production[J].hysiological reviews,2008,88(4):1243-1276.
[27]Bentley D.J.,Dank S.,Coupland R.et al..Acute antioxidant supplementation improvesendurance performance in trained athletes[J].Research in sports medicine,2012,20(1):1-12.
[28]Murphy M.P.Mitochondrial Thiols in Antioxidant Protection and Redox Signaling:Distinct Roles for Glutathionylation and Other Thiol Modifications[J].Antioxidants&Redox Signaling,2012,16(6):476-495.
[29]Vaughn A.E.,Deshmukh M.Glucose metabolism inhibits apoptosis in neurons and cancer cells by redox inactivation of cytochrome c[J].Nature cell biology,2008,10(12):1477-1483.
[30]Sakamoto T.,Imai H.Hydrogen peroxide produced by superoxide dismutase SOD-2activates sperm in Caenorhabditis elegans[J].The journal of biological chemistry,2017,292(36):14804-14813.
[31]Sagara Y.,Dargusch R.,Chambers D.et al..Cellular Mechanisms of Resistance to Chronic Oxidative Stress[J].Free Radic Biol Med,1998,24(9):1375-89.
[32]Valko M.,Leibfritz D.,Moncol J.et al..Free radicals and antioxidants in normal physiological functions and human disease[J].The international journal of biochemistry&cell biology,2007,39(1):44-84.
[33]Meister A..Glutathione Deficiency Produced by Inhibition of Its Synthesis,and Its Reversal-Applications in Research and Therapy[J].Pharmacol Therapeut,1991,51(2):155-194.
[34]Ruffmann R.,Wendel A.GSH rescue by N-acetylcysteine[J].Klin Wochenschr,1991,69(18):857-62.
[35]Beyfuss K.,Hood D.A..A systematic review of p53 regulation of oxidative stress in skeletal muscle[J].Redox report:communications in free radical research,2018,23(1):100-117.
[36]Nguyen T.,Nioi P.,Pickett CB..The Nrf2-Antioxidant Response Element Signaling Pathway and Its Activation by Oxidative Stress[J].Journal of Biological Chemistry,2009,284(20):13291-13295.
[37]Meister A.Selective modification of glutathione metabolism[J].Science,1983,220(4596):472-477.
[38]Alam J.,Stewart D.,Touchard C.et al..Nrf2,a Cap'n'Collar transcription factor,regulates induction of the heme oxygenase-1gene[J].Journal of Biological Chemistry,1999,274(37):26071-26078.
[39]Racanelli V.,Rehermann B.The liver as an immunological organ[J].Hepatology,2006,43(2Suppl 1):S54-62.
It will be evident to those skilled in the art that the invention is not limited to the details of the foregoing illustrative embodiments, and that the present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. The present embodiments are therefore to be considered in all respects as illustrative and not restrictive, the scope of the invention being indicated by the appended claims rather than by the foregoing description, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein. Those skilled in the art should appreciate that many modifications and variations are possible in light of the above teaching without departing from the scope of the invention.

Claims (10)

1. Application of astragalin in repairing muscle oxidative damage is provided.
2. Use according to claim 1, characterized in that: 50-200mg/kg of astragalin does not cause damage to the liver, and can enhance the anti-oxidation function of the muscle tissue of the mouse after exhaustion exercise.
3. Application of astragalin in reducing liver injury caused by exhaustion exercise is provided.
4. Use according to claim 3, characterized in that: the astragalin reduces the activities of glutamic-pyruvic transaminase, glutamic-oxalacetic transaminase and alkaline phosphatase.
5. Application of astragalin in enhancing the anti-oxidation function of exhaustive exercise.
6. Use according to claim 5, characterized in that: the astragalin can enhance the scavenging ability of hydroxyl free radicals and the scavenging ability of superoxide anions.
7. Use according to claim 5, characterized in that: the astragalin can improve GSH-Px activity, GR enzyme activity, SOD enzyme activity and GSH enzyme activity.
8. Application of astragalin in up-regulating mRNA expression of Nrf2 and downstream genes thereof.
9. Application of astragalin in up-regulating expression of Nrf2 and downstream gene proteins thereof.
10. Use according to claim 8 or 9, characterized in that: the downstream gene is selected from more than one of GCLC, GCLM, HO-1, NQO-1, SOD1 and SOD 2.
CN202111391366.2A 2021-11-23 2021-11-23 Application of astragalin in repairing oxidative damage of muscles Pending CN113876797A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111391366.2A CN113876797A (en) 2021-11-23 2021-11-23 Application of astragalin in repairing oxidative damage of muscles

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111391366.2A CN113876797A (en) 2021-11-23 2021-11-23 Application of astragalin in repairing oxidative damage of muscles

Publications (1)

Publication Number Publication Date
CN113876797A true CN113876797A (en) 2022-01-04

Family

ID=79015462

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111391366.2A Pending CN113876797A (en) 2021-11-23 2021-11-23 Application of astragalin in repairing oxidative damage of muscles

Country Status (1)

Country Link
CN (1) CN113876797A (en)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
夏伯候等: "UPLC测定桑叶中抗氧化活性成分异槲皮苷、芦丁和紫云英苷的含量", 《中药材》 *
聂龙等: "紫云英苷生物活性及其机制的研究进展", 《热带农业科学》 *

Similar Documents

Publication Publication Date Title
Bhattacharya et al. Antioxidant activity of tannoid principles of Emblica officinalis (amla) in chronic stress induced changes in rat brain.
Ramesh et al. Effect of fermented Panax ginseng extract (GINST) on oxidative stress and antioxidant activities in major organs of aged rats
CN103442594B (en) For improving composition and the method for mitochondrial function and treatment neurodegenerative disease and cognitive disorder
Li et al. Polysaccharide from Ostrea rivularis attenuates reproductive oxidative stress damage via activating Keap1-Nrf2/ARE pathway
Wang et al. Hawthorn fruit increases the antioxidant capacity and reduces lipid peroxidation in senescence-accelerated mice
Jing et al. Antioxidant activity, antitumor effect, and antiaging property of proanthocyanidins extracted from Kunlun Chrysanthemum flowers
Rejitha et al. Amelioration of alcohol-induced hepatotoxicity by the administration of ethanolic extract of Sida cordifolia Linn.
Chen et al. Cytoprotective effect of ligustrum robustum polyphenol extract against hydrogen peroxide-induced oxidative stress via Nrf2 signaling pathway in Caco-2 cells
Misra et al. Protection of swimming-induced oxidative stress in some vital organs by the treatment of composite extract of Withania somnifera, Ocimum sanctum and Zingiber officinalis in male rat
Mujawdiya et al. Mangiferin: A potential natural molecule for management of metabolic syndrome
Nwakuilite et al. Evaluation of some trace elements in streptozocin induced diabetic rats treated with Moringa oleifera leaf powder
Farokhi et al. Effects of Endurance Training and Tribulus terrestris Extract on Oxidative Stress and Apoptosis Markers in the Liver Tissues of Rats Exposed to Arsenic.
CN113876797A (en) Application of astragalin in repairing oxidative damage of muscles
Ariyanfar et al. The effect of endurance training and Purslane (portulaca oleracea) seed consumption on cytochrome-C and malondialdehyde in the heart tissue of rats poisoned with H2O2
Miao et al. Evaluation of anti-fatigue activity of flavonoids from tartary buckwheat in mice
Kong et al. Protective effects of Antrodia camphorata extract against hypoxic cell injury and ischemic stroke brain damage
Chaturvedi et al. Hepatoprotective potentials of water extract of Bauhinia purpurea bark against alcohol induced toxicity
CN113456660A (en) Application of epimedin C in preventing and treating acute alcoholism
Kang et al. Water chestnut (Trapa japonica Flerov.) exerts inhibitory effect on postprandial glycemic response in rats and free radical scavenging activity in vitro
Ojieh Evaluation of Antidiabetic and Antioxidant potential of Nephrolepis unduranta leaves extract in Streptozotocin Induced Diabetic Wistar Rats.
KR102621205B1 (en) An anti-oxidant and anti-inflammatory composition comprising extracts of lacquer tree, fluafomitella fraxinea and kudingcha
CN112107603A (en) Application of hericium erinaceus solid culture extract in preparation of composition for delaying aging and treating Alzheimer&#39;s disease
Khaki et al. Compartments of quercetin & Allium cepa (onion) on blood glucose in diabetic rats
CN114470150B (en) Application of chicken-derived small molecular peptide in preparation of product for preventing and improving liver injury and secondary symptoms thereof and product
KR100607033B1 (en) Dragonfly extracts having antioxidant activity

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

Application publication date: 20220104

RJ01 Rejection of invention patent application after publication