Skip to main content
  • Research article
  • Open access
  • Published:

The effect of Malus doumeri leaf flavonoids on oxidative stress injury induced by hydrogen peroxide (H2O2) in human embryonic kidney 293 T cells

Abstract

Background

In this study, Malus doumeri leaf flavonoids (MDLF) were used as the research object to observe their in vitro antioxidant stress ability. Hydrogen peroxide (H2O2) was used to induce oxidative stress in 293 T cells.

Methods

MTT, flow cytometry, and qPCR were used to verify the effect of MDLF.

Results

In vitro cell experiments showed that at a concentration of 0–160 μg/mL, MDLF did not affect the normal proliferation of human embryonic kidney 293 T cells (HEK 293 T cells), and MDLF had no cytotoxic effect in this concentration range. It was found that MDLF could maintain the survival of HEK 293 T cells (82.6%) at a high concentration (160 μg/mL). Morphological observation also found that MDLF can inhibit the cell structure imperfection caused by H2O2. It was also observed that MDLF could significantly increase the levels of catalase (CAT), superoxide dismutase (SOD), glutathione (GSH), and glutathione peroxidase (GSH-Px) and reduce the level of malondialdehyde (MDA). The results of quantitative polymerase chain reaction (qPCR) showed that MDLF could significantly up-regulate the mRNA expression levels of CAT, SOD, GSH, GSH-Px, B-cell lymphoma-2 (Bcl-2) and downregulate the expression levels of B-cell lymphoma-2 associated x protein (Bax), tumor necrosis factor-alpha (TNF-α), and nuclear factor kappa-B (NF-κB) in oxidative stress-injured cells. The HPLC analysis showed that MDLF contained hyperin, isoquercetin, quercitrin, hesperidin, myricetin, baicalin and quercetin.

Conclusion

From the experimental results, it was observed that MDLF has a strong anti-oxidation ability in vitro, and it can interfere with the oxidative stress damage caused by H2O2 in 293 T cells. Therefore, MDLF is a type of natural substance with good anti-oxidant effect, and it has the potential to interfere with many diseases.

Peer Review reports

Background

The production of oxidative stress is due to the excessive production of free radicals and the release of a large number of reactive oxygen species (ROS) and other oxidizing substances after the body is injured by endogenous or exogenous factors. However, the ability of the body to resist oxidation is decreased and the accumulation of ROS cannot be inhibited, which leads to a imbalance between the ability of oxidation and anti-oxidation. The oxidation reaction of the body is far beyond the ability of antioxidant clearance. Excessive ROS caused by oxidative stress can stimulate cells to initiate lipid peroxidation reaction, which can damage cell function and structure [1, 2]. The concentration of intracellular Ca2+ is increased under oxidative stress. It increases the expression of the apoptotic gene, but it decreases the expression of the anti-apoptotic protein, leading to normal cell apoptosis and pathological changes in the body [3]. When infectious inflammation occurs, pathogens invade the body, activate neutrophils, increase oxygen consumption, and generate a large number of oxygen free radicals. At the same time, when pathogens invade and cause a stress reaction, they can promote the release of catecholamines and produce a large number of oxygen free radicals. The resulting oxidative stress reaction can lead to inflammatory infiltration of neutrophils and can produce a large number of pro-inflammatory mediators. In addition, cell death caused by oxidative stress can also activate the inflammatory cells [4]. Oxidative stress and inflammatory reaction can cause a variety of diseases, such as hypertension and atherosclerosis [5].

The body balance is disrupted after oxidative stress, and this causes many diseases. Under the stimulation by external adverse factors, the level of skin internal oxidation rises, resulting in the metabolic imbalance of the oxidation system, which directly or indirectly leads to the occurrence of some skin diseases, such as skin photoaging, skin tumors, pigmented diseases, and wound healing and repair [6]. Studies have shown that the involvement of oxidative stress in the pathogenesis of hypertension is mainly due to vascular damage. ROS accumulation caused by oxidative stress can lead to dysfunction of the vascular endothelium, dysfunction of vasoconstriction and relaxation, failure to regulate blood circulation, and increase in blood pressure [7]. Clinical studies have found that the more serious the incidence of type 2 diabetes, the lower the expression of glyoxal reductase 1 gene and blocked metabolism of fatty acids in patients, resulting in excessive ROS to promote the body’s oxidative stress response. Oxidative stress can disrupt the function of islet β cells and promote insulin resistance, which eventually leads to the occurrence of type 2 diabetes [8]. The accumulation of a large number of ROS caused by oxidative stress can affect the central nervous system. Oxidation of the central nervous system can lead to the occurrence of multicentric vascular diseases, such as atherosclerosis and heart failure [9]. Under oxidative stress, excessive ROS can activate hepatic stellate cells, increase the accumulation of extracellular matrix, damage the liver, further produce a large number of ROS, and aggravate liver fibrosis by regulating the signal transduction pathway [10]. Clinical research also noted that the oxidation level in cancer patients was higher than that in normal people [11]. Therefore, it is very important to regulate the level of oxidation and inhibit abnormal oxidative stress.

At present, many plants have been found to have antioxidant effects, which mainly arise from flavonoids, saponins, polysaccharides, and organic acids [12, 13]. Antioxidant plants restore the balance of the oxidation antioxidant system by scavenging excessive free radicals [14]. Most of the plants rich in flavonoids have a certain antioxidant effect, and most of the flavonoids have a certain antioxidant effect. The 2,3-double bond, 4-carbonyl, and 3 or 5-hydroxy in the structure of flavonoids make an important contribution to the antioxidant effect of flavonoids [15]. Flavonoids contained in plants can not only clear the free radicals in the chain initiation stage, but also directly capture the free radicals in the reaction chain, block the free radical chain reaction, play a dual role of prevention and chain breakage, and exert an antioxidant effect [16]. Malus doumeri is a Rosaceae family plant, whose fruit is consumed. The leaves are also used to a certain extent. Firstly, they are processed into drinks such as tea; secondly, they are used as folk drugs to prevent diarrhea; thirdly, they have antiseptic and bacteriostatic effects, and they are often mixed with other foods and drugs as natural preservatives [17]. Studies have shown that there are many chemical components in the leaves of Malus that are beneficial to human health; among them, the vitamin C content reaches 177.75 mg/g [18]; in addition, there are 12 free amino acids in the healthy tea made from Malus; among them, the methionine content is the highest [19]. The content of polyphenols in the leaves of Malus doumeri is about 5%, and the content of flavonoids in general tea is only 1–2% of the total polyphenols, while that in the leaves of Malus doumeri is more than 60%. After rough processing, the yield of total flavonoids in the dry leaves of Malus doumeri can be more than 3%, which shows that the content of flavonoids in the leaves of Malus doumeri is high [20].

Because of the limited area of application and cognition, there are few researches on Malus doumeri. However, with an increasing number of drinks and health products processed from the leaves of Malus doumeri, its biological activity and mechanism need to be studied further. In this study, the components of flavonoids in the leaves of Malus doumeri leaf (MDLF) were analyzed, and the effect of MDLF on oxidative stress injury induced by hydrogen peroxide (H2O2) in human embryonic kidney 293 T cells was studied for the first time, which is the theoretical basis for further utilization of MDLF accumulation.

Methods

Extraction of MDLF

After the dried Malus doumeri leaves (identification number: SC11442282300212, Enshi Xihaitang Biotechnology Co., Ltd., Enshi, Hubei, China) were crushed and sieved. A voucher specimen is deposited at the herbarium of the Chongqing University of Education under the number CCICFF-201902. Professor Xin Zhao authenticated the sample. Then 200 g of these leaves were weighed and placed in a beaker precisely and 70% ethanol was added according to the liquid to material ratio of 20:1; then they were placed in a 60 °C water bath for 3 h and the liquid was extracted for standby, passing through FL-3 macroporous resin. The liquid passing through the resin was evaporated to dryness through a rotary evaporator, and the extracted MDLF was obtained [21].

Cytotoxicity of MDLF in HEK 293 T cells

After being resuscitated, HEK 293 T cells (Cell Bank of the Chinese Academy of Science, Shanghai, China) were inoculated in the deme medium containing 10% fetal bovine serum and 1% penicillin streptomycin double antibody. Then the cells were cultured in a carbon dioxide incubator (37 °C, 5% carbon dioxide, MCO-18 AC, Panasonic, Osaka, Japan) for 48 h and digested with 0.25% trypsin. Further, 293 T cells were counted and made into a 1 × 104 cells/mL suspension; then 200 μL cell suspension was inoculated into a 96 well cell culture plate and cultured at 37 °C for 24 h until cells were attached to the wall. Then the culture medium was discarded, and 200 μL new des medium containing 0–200 μg/mL MDLF was added for 48 h. Then the medium in the 96 well culture plate was discarded, and the deme medium containing 5 mg/mL MTT (200 μL, Solarbio Life Sciences, Beijing, China) was added for continuous culture for 4 h. The medium was discarded again, dimethyl sulfoxide (DMSO, Solarbio Life Sciences) of 200 μL was added, and the reaction was carried out for 30 min without light and vibration. Finally, the optical density (OD) value of each hole was measured at 494 nm (Evolution™ 350, Thermo Fisher Scientific, New York, USA), and the cell survival rate (cell survival rate/% = (Ar) after the MDLF treatment was calculated / As) × 100%, where Ar: OD value of MDLF treated cells; As: OD value of normal cells) to observe the toxic effect of MDLF in 293 T cells [22].

Effect of MDLF on H2O2-induced oxidative damage in HEK 293 T cells

Cells were resuscitated and cultured according to the experimental operation in the cytotoxicity experiment of MDLF on HEK 293 T cells. Cells were made into a cell suspension and cultured in 96 well plates. Cells were attached to the wall and then discarded. Further, 200 μL of deme medium containing 0.3 mmol/L H2O2 was added and cultured for 4 h to induce a cell oxidative damage model. After that, the medium was discarded again, the deme medium with concentration of 0–200 μg/mL MDLF was added to culture for 48 h, and then the OD value of each cell was measured at 494 nm according to the cytotoxicity experiment of MDLF on HEK 293 T cells and the inhibition effect of MDLF on oxidative damage induced by H2O2 in HEK 293 T cells was observed by calculating the survival rate of cells after oxidative damage prevention by each concentration of MDLF [22].

Morphological observation of cells

According to the effect of MDLF on the oxidative damage of HEK 293 T cells induced by hydrogen peroxide, the cells were cultured, induced oxidative damage and treated with MDLF for 48 h. The morphology of cells in each group was observed under the microscope (EVOS M7000, Thermo Fisher Scientific, New York, USA).

Observation of apoptosis by flow cytometry

According to the experimental operation of MDLF on oxidative damage induced by H2O2 in HEK 293 T cells, the cells were cultured and treated with MDLF for 48 h; then the cells were fixed, and apoptosis was detected by flow cytometry after staining (AccuriC6, BD Biosciences, San Jose, CA, USA) [23].

Determination of MDA, SOD, GSH, GSH PX, and CAT levels in HEK 293 T cells

According to the experimental operation of MDLF on oxidative damage induced by H2O2 in HEK 293 T cells, the cells were cultured and treated with MDLF for 48 h. Then the MDA, SOD, GSH, GSH-Px, and CAT levels in cells of each group were measured according to the kit instructions (Nanjing Jiancheng Bioengineering Institute, Nanjing City, China) [24].

qPCR method

In the experiment of oxidative damage induced by H2O2 in HEK 293 T cells by MDLF, cells in each group were used to extract total RNA of cells by Trizol reagent (Solarbio Life Sciences). Further, 1 μL of oligo (DT) 18 primer (500 ng/μL, Solarbio Life Sciences) and 1.0 μL of total RNA (1.0 μg/μL) were added to 10.0 μL of nuclease free water and heated in PCR for 5 min (65 °C). Then, 4.0 μL of 5 × reaction buffer, 1.0 μL of ribolock RNase inhibitor (20 U), 2.0 μL of 10 mM dNTP mix, and 1.0 μL of reversible reverse transcription (200 U/μL) were added to the above reaction system solution for cDNA transcription (60 min at 42 °C, 5 min at 70 °C). Then, 1 μL 10.0 μM upstream primer and 1 μL 10.0 μM downstream primer (Table 1, Thermo Fisher Scientific) and 7.0 μL sterile double distilled water were added to the 1.0 μL reaction fluid for amplification. The reaction conditions were 95 °C denaturation for 3 min, 60 °C annealing for 30 s, and 95 °C extension for 1 s. The relative expression intensity of each gene to be tested and the control cell (hydrogen peroxide induced oxidative damage cell) was calculated by the 2-∆∆Ct method through the measured CT value [24].

Table 1 The primer seque quantitative polymerase chain reaction

Main components of MDLF in high-performance liquid chromatography (HPLC)

MDLF was dissolved in DMSO to prepare a sample solution with a concentration of 10 mg/ml; then it was diluted with 50% methanol to prepare a solution to be tested with a concentration of 2 mg/mL and the solution to be tested was filtered by a 0.22 μm organic filter membrane and tested on the machine. The detection conditions were chromatographic column: Thermo Scientific Accucore C18 (4.6 mm × 150 mm, 2.6 μm), gradient elution mobile phase C was acetonitrile; mobile phase B was 0.5% glacial acetic acid aqueous solution; flow rate was 0.5 mL/min, column temperature was 35 °C; detection wavelength was 360 nm; and injection volume was 10 μL. Then the content of MDLF was calculated by the external standard peak area method according to the HPLC method (UltiMate3000 HPLC System, Thermo Fisher Scientific), and the formula was as follows: \( {M}_x={C}_{r\times}\frac{A_x}{A_r}\times C \) MC=C1 × A1 × C÷AR.

Where: Mc is the content of flavonoids in the sample, mg/g; C1 is the concentration of the standard, mg/ml; Ar is the peak area of the standard; A1 is the peak area of the sample; C is the concentration of the original solution of the sample, 0.0025 g/mL.

Data processing

All experiments were carried out three times in parallel. The mean ± standard deviation (SD) was used to express the results. SAS9.1 statistical software was used to analyze the significant differences between groups of data at the level of P < 0.05. The analysis method was Duncan’s multiple range test single factor analysis of variance (ANOVA).

Results

Toxic effect of MDLF in HEK 293 T cells

When HEK 293 T cells were treated with MDLF at concentrations of 20, 40, 60, 80, 100, 120, 140, 160, 180, and 200 μg/mL, compared with HEK 293 T cells that were not treated with MDLF, MDLF at concentrations of 20–160 μg/mL did not significantly affect the proliferation of HEK 293 T cells, and the survival rate of HEK 293 T cells in this concentration range was more than 95% (Fig. 1). Therefore, 40, 80, and 160 μg/mL MDLF were used as sample concentrations for further tests.

Fig. 1
figure 1

Toxic effect of Malus doumeri leaf flavonoids on human embryonic kidney 293 T cells. Values presented are the mean ± standard deviation

Effect of MDLF on oxidative stress injury induced by H2O2 in HEK 293 T cells

The survival rate of HEK 293 T cells treated with H2O2 was significantly lower than that of HEK 293 T cells treated without H2O2, and the survival rate was only 23.7% (Fig. 2). The survival rate of HEK 293 T cells increased with an increase in the concentration of MDLF. When the concentration of MDLF was 160 μg/mL, the survival rate of HEK 293 T cells increased to 82.6%.

Fig. 2
figure 2

Effect of Malus doumeri leaf flavonoids on hydrogen peroxide induced oxidative damage of human embryonic kidney 293 T cells. Values presented are the mean ± standard deviation. Different letters indicate significant differences from each other at the level of p < 0.05 according to Tukey’s honestly significant difference. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Effect of MDLF on the change in morphology induced by H2O2 in HEK 293 T cells

After cell culture, the normal HEK 293 T cells were found to be abundant in quantity, complete in cell structure, abundant in cytoplasm and orderly in cell arrangement in a microscopic field (Fig. 3); after being treated with H2O2, the number of HEK 293 T cells was decreased compared with the number of cells without H2O2 treatment. The cell morphology was severely atrophied, the cell structure was changed, and the cell damage was severe. MDLF could obviously improve the abnormal cell structure caused by H2O2 and could restore the cell structure and quantity. With an increase in MDLF concentration, the recovery of cells was better.

Fig. 3
figure 3

Morphological observation of hydrogen peroxide damaged human embryonic kidney 293 T cells. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Effect of MDLF on apoptosis induced by H2O2 in HEK 293 T cells

HEK 293 T cells exposed to H2O2 showed a high level of apoptosis compared with those not exposed to H2O2 (Fig. 4). After oxidative stress injury, HEK 293 T cells exposed to MDLF at concentrations of 40, 80, and 160 μg/mL showed decreased apoptosis and improved growth of cells. The higher the concentration of MDLF, the better the cell status and the lesser the number of apoptotic cells.

Fig. 4
figure 4

Observation of apoptosis on hydrogen peroxide damaged human embryonic kidney 293 T cells by flow cytometry. Values presented are the mean ± standard deviation. Different letters indicate significant differences from each other at the level of p < 0.05 according to Tukey’s honestly significant difference. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Effects of MDLF on the levels of MDA, SOD, GSH, GSH-Px, and CAT in HEK 293 T cells treated with H2O2

The MDA level in HEK 293 T cells treated with H2O2 was significantly higher than that in cells not treated with H2O2 (P < 0.05), while the SOD, GSH, GSH-Px, and CAT levels were significantly lower (P < 0.05, Fig. 5). The concentration of MDLF at 40, 80, and 160 μg/ml decreased the MDA level and increased the levels of SOD, GSH, GSH PX, and CAT in the cells treated with H2O2. Among them, the concentration of MDLF at 160 μg/mL restored the MDA, SOD, GSH, GSH-Px, and CAT levels in the cells closest to those in normal HEK 293 T cells.

Fig. 5
figure 5

MDA, SOD, GSH, GSH-Px and CAT levels of hydrogen peroxide damaged human embryonic kidney 293 T cells. Values presented are the mean ± standard deviation. Different letters indicate significant differences from each other at the level of p < 0.05 according to Tukey’s honestly significant difference. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Effect of MDLF on the expression levels of SOD, CAT, GSH, and GSH-Px mRNA induced by H2O2 in HEK 293 T cells

The levels of SOD, CAT, GSH, and GSH-Px mRNA expression in HEK 293 T cells treated with H2O2 were significantly lower than those in cells not treated with H2O2 (P < 0.05, Fig. 6). Different concentrations of MDLF could up-regulate the expression levels of SOD, CAT, GSH, and GSH-Px mRNA in H2O2 treated cells. With an increase in the concentration of MDLF, the expression levels of SOD, CAT, GSH, and GSH-Px in oxidatively damaged HEK 293 T cells were up-regulated. The results of the qPCR test were consistent with those of the kit test.

Fig. 6
figure 6

SOD, CAT, GSH and GSH-Px mRNA expression level of hydrogen peroxide damaged human embryonic kidney 293 T cells. Different letters indicate significant differences from each other at the level of p < 0.05 according to Tukey’s honestly significant difference. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Effect of MDLF on the expression levels of apoptosis-related Bax and Bcl-2 mRNA in HEK 293 T cells

The expression level of Bax mRNA in HEK 293 T cells was significantly increased (P < 0.05, Fig. 7) and the expression level of Bcl-2 was significantly decreased. MDLF could interfere with the abnormal expression of Bax and Bcl-2 mRNA caused by H2O2 in HEK 293 T cells; it up- regulated the expression of Bcl-2 and downregulated the expression of Bax.

Fig. 7
figure 7

Bax and Bcl-2 mRNA expression level of hydrogen peroxide damaged human embryonic kidney 293 T cells. Different letters indicate significant differences from each other at the level of p < 0.05 according to Tukey’s honestly significant difference. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Effect of MDLF on the expression levels of TNF-α and NF-κB mRNA induced by H2O2 in HEK 293 T cells

The expression levels of TNF-α and NF-κB mRNA in HEK 293 T cells treated with H2O2 were up-regulated compared to those in cells treated without H2O2 (P < 0.05, Fig. 8). MDLF could significantly inhibit the expression of TNF-α and NF-κB induced by H2O2 in HEK 293 T cells. The higher the concentration of MDLF, the stronger the effect.

Fig. 8
figure 8

TNF-α and NF-κB mRNA expression level of hydrogen peroxide damaged human embryonic kidney 293 T cells. Different letters indicate significant differences from each other at the level of p < 0.05 according to Tukey’s honestly significant difference. Normal: untreated human embryonic kidney 293 T cells; model: hydrogen peroxide induced oxidative damage human embryonic kidney 293 T cells

Analysis of the main components of MDLF

HPLC analysis showed that MDLF mainly contained 7 compounds, namely hypericin, isoquercetin, quercitrin, hesperidin, myricetin, baicalin and quercetin (Fig. 9). Their contents in MDLF were 67.193, 58.063, 381.858, 12.447, 5.828, 20.135, and 17.391 mg/g, respectively.

Fig. 9
figure 9

Composition analysis of Malus doumeri leaf flavonoids. (a) HPLC chromatogram of standards; (b) HPLC chromatogram of Malus doumeri leaf flavonoids; (c) content of components

Discussion

As a traditional Chinese medicine or tea substitute, plant leaves have the characteristics of little or no side effects, and play an important role in human health. Malus doumeri Leaf is also a plant with traditional medicinal effects, but its mechanism of action has been less studied [18]. In this study, the antioxidant effect and mechanism of Malus doumeri leaf flavonoids in vitro were preliminarily studied.

After the occurrence of oxidative stress, normal metabolism and increment of tissue cells are affected. In serious cases, a large number of cells die, and proliferation slows down or even stagnates [25]. At the same time, abnormal oxidative stress leads to the destruction of normal cell structure, apoptosis of cells, and then abnormal cell death. Serious decline affects human health, and even threatens life. Reducing the degree of apoptosis caused by oxidative stress can protect the tissues and cells, and resist the attack of diseases on the body [26]. In this study, MDLF has been proved to inhibit abnormal cell death and apoptosis caused by oxidative stress in vitro; thus, it protect the cells.

MDA is recognized as a marker of oxidative stress. MDA released from the cell membrane can react with protein and nucleic acid, causing cross-linking polymerization and making normal synthesis of the protein impossible. MDA can also cause abnormal structure and function of the cell membrane, thus causing an abnormal physiological state of the body [27, 28]. SOD, GSH Px, and CAT are antioxidant enzymes in vivo. MDA is highly cytotoxic and can inhibit antioxidant enzymes in vivo. Oxidative stress caused by abnormal expression of these indicators is involved in the pathogenesis of many clinical diseases [29]. GSH is also an active peptide with good antioxidant effect, which can effectively regulate the oxidation balance of the body and inhibit the damage caused by oxidative stress [30]. The results of this study also showed that MDLF can interfere with the abnormality of oxidation-related indexes after H2O2 treatment of cells, and thus, it reduces the cell damage caused by oxidative stress and plays a role in protecting the normal cells.

Oxidative stress is closely related to cell apoptosis, which is mainly related to the mitochondrial pathway, the mitogen activated protein kinase (MAPK) pathway, endoplasmic reticulum stress, NF-κB, and Bcl-2 family [31]. In the Bcl-2 family, there are inhibitory factors, such as Bcl-2, and pro-apoptotic factors, such as Bax, and both of them have a synergistic effect. Bcl-2 is an antioxidant present in the body, which has an antioxidant effect and can protect the normal cells [32]. The results showed that the expression of Bcl-2 was increased significantly, while Bax expression was decreased under the action of antioxidants. Apoptosis of renal tubular epithelial cells after ureteral obstruction could be reduced by regulating Bax and bcl-2. This study also showed a similar effect. MDLF can intervene in the oxidative stress of HEK 293 T cells by enhancing the expression of Bcl-2 and reducing the expression of Bax.

Oxidative stress and inflammation promote each other. Research shows that TNF-α can enhance the expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and inhibit the expression of SOD, which leads to an oxidative stress reaction. However, inhibiting the expression of TNF-α can effectively inhibit the oxidative stress reaction [33]. NF-κB is also an important inflammatory factor. NF-κB plays a key role in many inflammatory reactions and immune responses. The incorrect regulation of NF-κB can lead to autoimmune diseases, chronic inflammation, and a variety of cancers [34]. In addition, oxidative stress can also activate the NF-κB signaling pathway. Activated NF-κB enters the nucleus and combines with c-myc and other apoptosis-related genes to promote gene transcription and cell apoptosis [35]. In this study, MDLF could also downregulate the inflammatory expression of HEK 293 T cells, so as to intervene in the relationship between oxidative stress and inflammation and protect the cells.

Hypericin, isoquercetin, quercitrin, hesperidin, myricetin, baicalin, and quercetin are commonly found flavonoids in plants, and all of them have strong antioxidant activity [36,37,38,39,40,41,42]. Hyperoside can exert many physiological activities, such as anti-inflammatory, anti-hypertensive, cholesterol lowering, and central pain relieving activities, and protective effects on the heart and brain vessels through its antioxidant capacity [36]. Isoquercetin and quercitrin have antioxidant, antitumor, hypoglycemic, and hypolipidemic effects, and they especially have significant inhibitory effects on inflammatory factors [37, 38]. Hesperidin has anti-inflammatory and antiviral effects, as well as certain antibacterial effects [39]. Myricetin has many biological activities, such as anti-inflammatory, anti-tumor, and anti-mutation activities; caries prevention; anti-oxidation; and elimination of free radicals in vivo [40]. Baicalin has antibacterial, anti-inflammatory, and cholesterol reducing effects, especially in liver disease and kidney disease [41]. Quercetin is not only used as an antioxidant in the food industry, but it also has anti-inflammatory and anti-cough effects [42]. MDLF contains these seven compounds. Their direct interaction constitutes the biological activity of MDLF, which confers a good ability to resist oxidative stress and the potential to prevent and treat other diseases. These flavonoids produce stable semiquinone free radicals through the reaction of phenolic hydroxyl group with free radicals, thus terminating the chain reaction of free radicals, which is the main mechanism for flavonoids to scavenge free radicals [43]. Phenolic hydroxyl group is the main active group of flavonoids. To a certain extent, the increase of phenolic hydroxyl group will increase the antioxidant activity; B-ring is the main active part of flavonoids to scavenge free radicals. When there is o-hydroxy group in the ring, the antioxidant activity is greatly improved; the 2,3-position double bond is beneficial to the formation of more stable radicals after the p-ring is de electron; 4-position carbonyl group can form hydrogen bond with o-hydroxyl, which makes free radical intermediate more stable; 3,5-hydroxyl belongs to synergic phenolic hydroxyl [44]. At the same time, flavonoids combine with cell membrane in the form of hydrogen bond to protect the unsaturated bond of cell membrane from contacting with free radicals, thus playing the role of antioxidant protection [45]. In this study, the MDLF may also play a role in avoiding human embryonic kidney 293 T cells damage by hydrogen peroxide.

Conclusion

In this study, the results showed that MDLF could protect HEK 293 T cells from oxidative stress by regulating oxidation, apoptosis, and inflammation. MDLF contains active flavonoid substances, which make it play these roles. This study preliminarily verified the effect of MDLF, but its antioxidative effect and mechanism in vivo still need to be studied further. In the future, an in-depth study is needed to assess the role of MDLF in the intervention for various diseases through its antioxidant effect.

Abbreviations

MDLF:

Malus doumeri leaf flavonoids

H2O2 :

hydrogen peroxide

qPCR:

quantitative polymerase chain reaction

MTT:

3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide

HEK 293 T cells:

human embryonic kidney 293 T cells

CAT:

catalase

SOD:

superoxide dismutase

GSH:

glutathione

GSH-Px:

glutathione peroxidase

MDA:

malondialdehyde

Bcl-2:

B-cell lymphoma-2

Bax:

B-cell lymphoma-2 associated x protein

TNF-α:

tumor necrosis factor-alpha

NF-κB:

nuclear factor kappa-B

HPLC:

high performance liquid chromatography

References

  1. Griendling KK, FitzGerald GA. Oxidative stress and cardiovascular injury: part I: basic mechanisms and in vivo monitoring of ROS. Circulation. 2003;108(16):1912–6.

    PubMed  Google Scholar 

  2. Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol. 2014;24(10):453–62.

    Google Scholar 

  3. Ray SK, Fidan M, Nowak MW, Wilford GG, Hogan EL, Banik NL. Oxidative stress and Ca2+ influx upregulate calpain and induce apoptosis in PC12 cells. Brain Res. 2000;852(2):326–34.

    CAS  PubMed  Google Scholar 

  4. Elmarakby AA, Sullivan JC. Relationship between oxidative stress and inflammatory cytokines in diabetic nephropathy. Cardiovasc Ther. 2010;30(1):49–59.

    PubMed  Google Scholar 

  5. Alexander RW. Theodore Cooper memorial lecture. Hypertension and the pathogenesis of atherosclerosis. Oxidative stress and the mediation of arterial inflammatory response: a new perspective. Hypertension. 1995;25(2):155–61.

    CAS  PubMed  Google Scholar 

  6. Spector A. Review: oxidative stress and disease. J Ocul Pharmacol Ther. 2000;16(2):193–201.

    CAS  PubMed  Google Scholar 

  7. Ceriello A. Possible role of oxidative stress in the pathogenesis of hypertension. Diabetes Care. 2008;31(S2):181–4.

    Google Scholar 

  8. Heidari H, Kamalinejad M, Noubarani M, Rahmati M, Jafarian I, Adiban H, Eskandari MR. Protective mechanisms of Cucumis sativus in diabetes-related modelsof oxidative stress and carbonyl stress. Bioimpacts. 2016;6(1):33–9.

    PubMed  PubMed Central  Google Scholar 

  9. Jeremy JY, Yim AP, Wan S, Angelini GD. Oxidative stress, nitric oxide, and vascular disease. J Card Surg. 2002;17(4):324–7.

    PubMed  Google Scholar 

  10. Luangmonkong T, Suriguga S, Mutsaers HAM, Groothuis GMM, Olinga P, Boersema M. Targeting oxidative stress for the treatment of liver fibrosis. Rev Physiol Biochem Pharmacol. 2018;175:71–102.

    CAS  PubMed  Google Scholar 

  11. Erhola M, Nieminen MM, Kellokumpu-Lehtinen P, Huusari H, Alanko K, Metsä-Ketelä T, Alho H. Effects of surgical removal of lung cancer on total plasma antioxidant capacity in lung cancer patients. J Exp Clin Cancer Res. 1998;17(2):219–25.

    CAS  PubMed  Google Scholar 

  12. Kicel A, Olszewska MA. Evaluation of antioxidant activity, and quantitative estimation of flavonoids, saponins and phenols in crude extract and dry fractions of Medicago lupulina aerial parts. Nat Prod Commun. 2015;10(3):483–6.

    PubMed  Google Scholar 

  13. Jia S, Shen M, Zhang F, Xie J. Recent advances in momordica charantia: functional components and biological activities. Int J Mol Sci. 2017;18(12):2555.

    PubMed Central  Google Scholar 

  14. Kandaswami C, Middleton E Jr. Free radical scavenging and antioxidant activity of plant flavonoids. Adv Exp Med Biol. 1994;366:351–76.

    CAS  PubMed  Google Scholar 

  15. Awad HM, Abd-Alla HI, Mahmoud KH, El-Toumy SA. In vitro anti-nitrosative, antioxidant, and cytotoxicity activities of plant flavonoids: a comparative study. Med Chem Res. 2014;23:3298–307.

    CAS  Google Scholar 

  16. Tancred SJ, Zeppa AG, Graham GC. The use of the PCR-RAPD technique in improving the plant variety rights description of a new Queensland apple (Malus domestica) cultivar. Aust J Exp Agri. 1994;34(5):665–7.

    Google Scholar 

  17. Li HZ, Zhong HY, Wei YQ. The research development of utilization on Mallus doumeri leaf. Food Sci Technol. 2000;2000(3):67–9.

    Google Scholar 

  18. Zeng XX, Tan SY, Luo ZM. Analysis on main chemical components of Malus healthy tea. J Hunan Agri Univ (Nat Sci). 1993;1993(5):474–6.

    Google Scholar 

  19. Zhang MJ, Su XJ. Simultaneous determination of five flavonoids from Malus doumeri leaves by HPLC. Guangzhou Chem Ind. 2015;43(24):151–3.

    CAS  Google Scholar 

  20. Khan MA, Rahman MM, Tania M, Shoshee NF, Xu AH, Chen HC. Antioxidative potential of Duranta repens (Linn.) fruits against H2O2 induced cell death in vitro. Afr J Tradit Complement Altern Med. 2013;10(3):436–41.

    PubMed  PubMed Central  Google Scholar 

  21. Huang JS, Fu XP. Study on separation and purification of flavonoids from Alpinia galanga (L.) Willd by macroporous resin. Storage Process. 2017;17(4):77–82.

    Google Scholar 

  22. Liu J, Tan F, Liu XH, Yi RK, Zhao X. Grape skin fermentation by lactobacillus fermentum CQPC04 has anti-oxidative effects on human embryonic kidney cells and apoptosis-promoting effects on human hepatoma cells. RSC Adv. 2020;10(8):4607–20.

    CAS  Google Scholar 

  23. Liu BH, Ma RD, Zhang J, Sun P, Yi RK, Zhao X. Preventive effect of small-leaved Kuding tea (Ligustrum robustum (Roxb.) Bl.) polyphenols on D-galactose-induced oxidative stress and aging in mice. Evid Based Complement Alternat Med. 2019;2019;3152324.

  24. Zhu K, Peng P, Wu N, Zhou XR, Mu JF, Zhao X. Preventive effect of Liupao tea polyphenols on HCl/ethanol-induced gastric injury in mice. J Food Quality. 2020;2020:5462836.

    Google Scholar 

  25. Wang ZJ, Zhang FM, Wang LS, Yao YW, Zhao Q, Gao X. Lipopolysaccharides can protect mesenchymal stem cells (MSCs) from oxidative stress-induced apoptosis and enhance proliferation of MSCs via toll-like receptor (TLR)-4 and PI3K/Akt. Cell Biol Int. 2009;33(6):665–74.

    CAS  PubMed  Google Scholar 

  26. Kumar S, Sitasawad SL. N-acetylcysteine prevents glucose/glucose oxidase-induced oxidative stress, mitochondrial damage and apoptosis in H9c2 cells. Life Sci. 2009;84(11–12):328–36.

    CAS  PubMed  Google Scholar 

  27. Zhu K, Zeng XF, Tan F, Li WF, Li C, Song YR, Zhao X. Effect of insect tea on D-galactose-induced oxidation in mice and its mechanisms. Food Sci Nutr. 2019;7(12):4105–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu BH, Zhang CF, Zhang J, Zhao X. Wu Shan Shen Cha (Malus asiatica Nakai. leaves)-derived flavonoids alleviate alcohol-induced gastric injury in mice via an anti-oxidative mechanism. Biomolecules. 2019;9(5):169.

  29. Li F, Huang GB, Tan F, Yi RK, Zhou XR, Mu JF, Zhao X. Lactobacillus plantarum KSFY06 on D-galactose-induced oxidation and aging in Kunming mice. Food Sci Nutr. 2019;8(1):379–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Long XY, Song JJ, Zhao X, Zhang Y, Wang HW, Liu XQ, Suo HY. Silkworm pupa oil attenuates acetaminophen-induced acute liver injury by inhibiting oxidative stress-mediated NF-κB signaling. Food Sci Nutr. 2019;8(1):237–45.

    PubMed  PubMed Central  Google Scholar 

  31. Xiang Y, Ye W, Huang C, Lou B, Zhang J, Yu D, Huang X, Chen B, Zhou M. Brusatol inhibits growth and induces apoptosis in pancreatic cancer cells via JNK/p38 MAPK/NF-κb/Stat3/Bcl-2 signaling pathway. Biochem Biophys Res Commun. 2017;487(4):820–6.

    CAS  PubMed  Google Scholar 

  32. Marone M, Scambia G, Mozzetti S, Ferrandina G, Iacovella S, De Pasqua A, Benedetti-Panici P, Mancuso S. Bcl-2, bax, bcl-XL, and bcl-XS expression in normal and neoplastic ovarian tissues. Clin Cancer Res. 1998;4(2):517–24.

    CAS  PubMed  Google Scholar 

  33. White JE, Tsan MF. Differential induction of TNF-alpha and MnSOD by endotoxin: role of reactive oxygen species and NADPH oxidase. Am J Respir Cell Mol Biol. 2001;24(2):164–9.

    CAS  PubMed  Google Scholar 

  34. Sun SC, Chang JH, Jin J. Regulation of NF-κB in autoimmunity. Trends Immunol. 2013;34(6):282–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Hong J, Wang Y, Hu BC, Xu L, Liu JQ, Chen MH, Wang JZ, Han F, Zheng Y, Chen X, Li Q, Yang XH, Sun RH, Mo SJ. Transcriptional downregulation of microRNA-19a by ROS production and NF-κB deactivation governs resistance to oxidative stress-initiated apoptosis. Oncotarget. 2017;8(41):70967–81.

    PubMed  PubMed Central  Google Scholar 

  36. Piao MJ, Kang KA, Zhang R, Ko DO, Wang ZH, You HJ, Kim HS, Kim JS, Kang SS, Hyun JW. Hyperoside prevents oxidative damage induced by hydrogen peroxide in lung fibroblast cells via an antioxidant effect. Biochim Biophys Acta. 2008;1780(12):1448–57.

    CAS  PubMed  Google Scholar 

  37. Cruz-Zúñiga JM, Soto-Valdez H, Peralta E, Mendoza-Wilson AM, Robles-Burgueño MR, Auras R, Gámez-Meza N. Development of an antioxidant biomaterial by promoting the deglycosylation of rutin to isoquercetin and quercetin. Food Chem. 2016;204:420–6.

    PubMed  Google Scholar 

  38. Babujanarthanam R, Kavitha P, Mahadeva Rao US, Pandian MR. Quercitrin a bioflavonoid improves the antioxidant status in streptozotocin: induced diabetic rat tissues. Mol Cell Biochem. 2011;358(1–2):121–9.

    CAS  PubMed  Google Scholar 

  39. Wilmsen PK, Spada DS, Salvador M. Antioxidant activity of the flavonoid hesperidin in chemical and biological systems. J Agric Food Chem. 2005;53(12):4757–61.

    CAS  PubMed  Google Scholar 

  40. Jimenez M, Garcia-Carmona F. Myricetin, an antioxidant flavonol, is a substrate of polyphenol oxidase. J Sci Food Agri. 1999;79(14):1993–2000.

    CAS  Google Scholar 

  41. Waisundara VY, Siu SY, Hsu A, Huang D, Tan BK. Baicalin upregulates the genetic expression of antioxidant enzymes in Type-2 diabetic Goto-Kakizaki rats. Life Sci. 2011;88(23–24):1016–25.

    CAS  PubMed  Google Scholar 

  42. Boots AW, Haenen GR, Bast A. Health effects of quercetin: from antioxidant to nutraceutical. Eur J Pharmacol. 2008;585(2–3):325–37.

    CAS  PubMed  Google Scholar 

  43. Boveris A. Biochemistry of free radicals: from electrons to tissues. Medicina. 1998;58(4):350–6.

    CAS  PubMed  Google Scholar 

  44. Coiner H, Schröder G, Wehinger E, Liu CJ, Noel JP, Schwab W, Schröder J. Methylation of sulfhydryl groups: a new function for a family of small molecule plant O-methyltransferases. Plant J Cell Mol Biol. 2006;46(2):193–205.

    CAS  Google Scholar 

  45. Reitman S, Frankel S. A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. Am J Clin Pathol. 1957;28(1):56–63.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Availability of data and material

The datasets generated and/or analyzed during the current study are not publicly available due to decision of author’s team but are available from the corresponding author on reasonable request.

Funding

This study was supported by funded by 2019 Chongqing Natural Science Foundation (cstc2019jcyj-msxmX0555), China.

The funding bodies played no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization and study design: YYL, YYL, ZEF, DH, and JDW; Data interpretation: YYL, YYL, ZEF, DH, YLY, DJZ, MCH and JDW; Manuscript preparation: YYL and JDW; All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Junda Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Li, Y., Fang, Z. et al. The effect of Malus doumeri leaf flavonoids on oxidative stress injury induced by hydrogen peroxide (H2O2) in human embryonic kidney 293 T cells. BMC Complement Med Ther 20, 276 (2020). https://doi.org/10.1186/s12906-020-03072-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-020-03072-6

Keywords