Advances in Clinical and Experimental Medicine

Title abbreviation: Adv Clin Exp Med
JCR Impact Factor (IF) – 2.1 (5-Year IF – 2.0)
Journal Citation Indicator (JCI) (2023) – 0.4
Scopus CiteScore – 3.7 (CiteScore Tracker 3.8)
Index Copernicus  – 171.00; MNiSW – 70 pts

ISSN 1899–5276 (print)
ISSN 2451-2680 (online)
Periodicity – monthly

Download original text (EN)

Advances in Clinical and Experimental Medicine

2021, vol. 30, nr 11, November, p. 1195–1203

doi: 10.17219/acem/139130

Publication type: original article

Language: English

License: Creative Commons Attribution 3.0 Unported (CC BY 3.0)

Download citation:

  • BIBTEX (JabRef, Mendeley)
  • RIS (Papers, Reference Manager, RefWorks, Zotero)

Cite as:


Ran H, Liu H, Wu P. Echinatin mitigates H2O2-induced oxidative damage and apoptosis in lens epithelial cells via the Nrf2/HO-1 pathway. Adv Clin Exp Med. 2021;30(11):1195–1203. doi:10.17219/acem/139130

Echinatin mitigates H2O2-induced oxidative damage and apoptosis in lens epithelial cells via the Nrf2/HO-1 pathway

Haijun Ran1,B,D, Han Liu2,B,C, Ping Wu3,A,E,F

1 Nanchong Aier Mega Eye Hospital, China

2 Department of Ophtalmology, Jiangjin Central Hospital of Chongqing, China

3 Chongqing Aier Eye Hospital, China

Abstract

Background. Oxidative stress has been reported to be an early factor in the development of cataracts. Echinatin (Ech) is an active ingredient of licorice that exhibits antioxidant effects.

Objectives. To investigate the effects of Ech on oxidative stress-induced lens epithelial cell (LEC) damage.

Materials and methods. Human lens epithelial B3 cells (HLECs) were exposed to hydrogen peroxide (H2O2) and were pretreated with or without Ech. For rescue experiments, ML385, an inhibitor of the Nrf2 pathway, was added into the medium.

Results. Echinatin reversed the H2O2-induced reduction of cell viability in B3 cells. Additionally, H2O2 induced oxidative stress, evidenced by an increase of reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and a decrease in superoxide dismutase (SOD) and catalase (CAT) levels, which could be abolished by Ech. Echinatin treatment also reduced HLEC apoptosis induced by H2O2. In addition, Ech pretreatment promoted Bcl-2 expression, and suppressed Bax and caspase-3 expression levels, in H2O2-treated B3 cells. Moreover, H2O2 significantly reduced Nrf2 nuclear localization, as well as HO-1 and NQO1 expression, which could be reversed by Ech. Inhibition of Nrf2 by ML385 aggravated H2O2-induced oxidative damage and apoptosis in HLECs, and the protective effects of Ech on H2O2-induced oxidative damage and apoptosis could be restored by ML385.

Conclusions. Echinatin mitigates H2O2-induced oxidative damage and apoptosis in HLECs via the Nrf2/HO-1 pathway, suggesting that Ech may be a potential drug for the treatment of cataracts.

Key words: cataract, lens epithelial cells, apoptosis, oxidative stress, echinatin

Background

Cataracts have become the main cause of loss of useful vision worldwide.1 It is currently believed that, with the exception of congenital cataracts, the apoptosis of lens epithelial cells (LECs) is the cytological basis for the formation of various types of cataracts.2 Due to long-term exposure to light, the lens is continuously damaged by reactive oxygen species (ROS), which is considered to be a key factor in the development of cataracts.3, 4, 5 Studies have confirmed that cataracts are directly related to the apoptosis of LECs caused by oxygen free radicals.6, 7 Thus, it is important to explore antioxidant drugs that can prevent the formation and development of cataracts.

The process of oxidative stress mainly involves a variety of stress-sensitive signaling pathways.8, 9 As one of the main cellular defense mechanisms against oxidative stress, Nrf2 is crucial in resisting cell damage caused by endogenous and exogenous stress.10 As the main regulator of the antioxidant response, Nrf2 can induce the expression of target genes, such as NAD(P)H quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO-1) and catalase (CAT). The dysfunction of Nrf2 is inseparable from the development of cataract. Studies have shown that the protein and gene expression of Nrf2 in the lens significantly decreases with age.11, 12 The decreased activity of Nrf2 limits the transcription of its downstream antioxidant enzymes and causes the antioxidant system to fail, which ultimately leads to age-related cataracts.12 Targeted activation of Nrf2 signaling can protect LECs from damage induced by oxidative stress.13, 14

Echinatin (Ech; 4,4’-dihydroxy-2-methoxychalcone; Figure 1A), a retrochalcone, is an active ingredient of licorice and the main active form with pharmacokinetic function.15 Studies have shown that Ech has a wide range of biological properties, including anti-inflammatory and anti-tumor effects.16 Importantly, Liang et al.17 confirmed that Ech may undergo electron transfer and proton transfer to cause antioxidant effects. However, to date, the effects of Ech on oxidative stress-induced LEC damage have not been reported, and its molecular mechanism is largely unclear. Hence, the present study attempted to investigate the potential role of Ech as an agent for controlling cataract progression against H2O2-induced oxidative stress and apoptosis in human B3 cells.

Objectives

Oxidative stress has been shown to be an early factor in the development of cataracts. Echinatin is the active ingredient of licorice, and its pharmacological effects are closely related to antioxidants. Thus, the aim of the current study is to investigate the effects of Ech on oxidative stress-induced LEC damage.

Materials and methods

All in vitro experiments in the current study were carried out using human LECs (HLECs). This study does not contain any experiments using human participants or animal subjects.

Cell culture and treatment

The human lens epithelial cell (HLEC) line B3 cells were purchased from American Type Culture Collection (CRL-11421; ATCC, Manassas, USA). B3 cells were cultured in Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, USA) containing 10% fetal bovine serum (FBS; Invitrogen) and 100 mg/mL streptomycin in a humidified atmosphere with 5% CO2 at 37°C. B3 cells at 80% confluence were treated with different concentrations of H2O2 (Sigma–Aldrich, Seelze, Germany; 0, 50, 100, 200, 400, and 800 µM) for 24 h. To investigate the role of Ech, B3 cells were pretreated with different concentrations of Ech (0, 5, 10, 20, 50, and 100 µM) for 12 h before the H2O2 treatment. For rescue experiments, B3 cells were pretreated with ML385 (5 µM; Sigma–Aldrich), a specific inhibitor of Nrf2, for 12 h in the presence or absence of Ech (50 µM), followed by exposure to H2O2. Echinatin (C16H14O4; CAS No. 34221-41-5, M.W. 270.2, purity 99%) was purchased from Chengdu Alfa Biotechnology Co. Ltd. (Chengdu, China).

MTT assay

The cells (2 × 104) were seeded in a 96-well culture plate and incubated with H2O2 (0, 50, 100, 200, 400, and 800 µM) for 24 h alone or after pretreatment with Ech (0, 5, 10, 20, 50, and 100 µM) for 12 h. The cells were then incubated with 5 mg/mL MTT solution (Beyotime, Shanghai, China) for 4 h at 37°C. The supernatant was aspirated and dimethyl sulfoxide (DMSO) was added to the cells. The optical density at 490 nm was observed using a microplate reader (SpectraMax Id3; Molecular Devices, San Jose, USA).

Determination of intracellular ROS levels

The ROS level was gauged using 6-carboxy-2’, 7’-dichlorodihydrofluorescein diacetate (DCHFDA; Beyotime) according to the manufacturer’s protocol. Briefly, after exposure to corresponding treatment, the culture medium was removed and B3 cells were incubated with 25 µM of DCHFDA mix for 45 min at 37°C. The absorbance was detected at a wavelength of 488 nm.

Measurement of MDA, SOD and CAT

After centrifugation of cell lysates, the supernatants were collected. The protein concentration was measured according to the BCA Assay Kit instructions (Beyotime). Malondialdehyde (MDA) was measured using a lipid peroxidation MDA assay kit (Beyotime). The test working solution was added to the sample and mixed. The mixture was heated at 100°C for 15 min. After centrifugation, the supernatants were collected. The absorbance was detected at a wavelength of 532 nm and the results were expressed as nmol/mg protein.

Total superoxide dismutase (SOD) content was determined using a SOD assay kit (Beyotime), and the results were expressed as U/mg protein. A Cu/Zn-SOD inhibitor was added to the samples to inhibit the activity of the Cu/Zn-SOD enzyme, followed by incubation with the WST-8/enzyme working solution at 37°C for 30 min. The absorbance was detected at a wavelength of 450 nm. One SOD enzymatic activity unit (U) was defined as the amount of sample needed to achieve a 50% inhibition rate of WST-8 formazan dye.

Catalase (CAT) content was tested using a CAT assay kit (Beyotime). Catalase detection buffer and hydrogen peroxide solution were added to the sample, and then incubated at 25°C for 5 min. The reaction stop solution was added to the mixture, then it was inverted and mixed to stop the reaction. After adding the detection buffer and chromogenic working buffer, the mixture was incubated at 25°C for 15 min. The absorbance was detected at a wavelength of 520 nm, and the results were expressed as U/mg protein. All operations were performed in accordance with the manufacturer’s instructions.

Measurement of apoptosis

Cells (1 × 106) were seeded and pretreated with or without Ech for 12 h, followed by exposure to 100 µM H2O2 for 24 h. Cells were fixed in 4% paraformaldehyde for 20 min at room temperature. The apoptosis of B3 cells was measured using the terminal deoxyribonucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay (Cell Death Detection Kit; Beyotime), according to the instructions of the manufacturer. The cells were visualized under a fluorescent microscope (Olympus Corp., Tokyo, Japan). Data were expressed as the ratio of TUNEL-positive cells to total cells.

Western blot assay

The protein expression in whole cell lysates or nuclear extracts was analyzed using western blot analysis. The radioimmunoprecipitation assay (RIPA) lysis buffer (Beyotime) was used to extract total protein from B3 cells. Next, a BCA protein assay kit (Beyotime) was used to quantify the proteins. Then, 30-µg protein samples were separated using 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride (PVDF) membranes (Merck Millipore, Darmstadt, Germany). The membranes were then blocked using 5% skimmed milk for 1 h at room temperature, followed by incubation with primary antibodies against Bcl-2 (Abcam, Cambridge, USA), Bax (Abcam), Nrf2 (Cell Signaling Technology, Beverly, USA), HO-1 (Abcam), NQO1 (Cell Signaling Technology), and GAPDH (Cell Signaling Technology) at 4°C overnight. Subsequently, the membranes were incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit secondary antibody (Abcam) for 1 h at room temperature, followed by exposure to the enhanced chemiluminescent reagent (Pierce, Rockford, USA). The intensity of proteins signals was quantified using Quantity One software v. 4.1.1 (Bio-Rad Laboratories, Hercules, USA).

Measurement of caspase-3 activity

As described previously,18 caspase-3 activity was measured using a caspase-3 activity assay kit (Beyotime) according to the manufacturer’s protocol. For each sample, an equal amount of protein (200 µg) was mixed with reaction buffer (50 µL) and caspase-3 substrate (5 µL) in the dark at 37°C for 30 min. The absorbance at 485 nm was measured using a microplate reader.

Statistical analyses

Data are expressed as mean ± standard deviation (SD) from at least 3 independent experiments, and the results were analyzed using GraphPad Prism v. 8.0 (GraphPad Software, San Diego, USA). Data from individual groups were confirmed to follow a normal distribution using the Kolmogorov–Smirnov test. Comparisons between 2 groups were analyzed using Student’s t-tests (Welch’s correction was used in cases of unequal variance), and comparisons among multiple groups were analyzed using one-way analysis of variance (ANOVA), followed by Tukey’s post hoc tests. A p-value of less than 0.05 was considered statistically significant.

Results

Echinatin attenuates the effects
of H2O2 on viability of HLECs

We performed a MTT assay to evaluate the effects of H2O2 and Ech on the viability of B3 cells. The B3 cells were exposed to H2O2 (0–800 µM) for 24 h and the results showed that higher concentrations of H2O2 (100–800 µM) restrained cell viability in a dose-dependent manner (Figure 1B). In view of the results that the treatment with 100 µM H2O2 for 24 h could reduce cell viability to approx. 50% compared to the control group, this concentration was chosen for subsequent experiments. Additionally, Ech (0–50 µM) did not affect the viability of HLECs (Figure 1C). Moreover, the pretreatment with Ech (10–50 µM) illustrated a protective effect against H2O2-induced damage in a dose-dependent manner (Figure 1D).

Echinatin reduces H2O2-induced
oxidative damage in HLECs

Oxidative stress is considered to be an early factor in the development of cataracts.19 B3 cells were treated with H2O2 for 24 h and exhibited the onset of oxidative stress manifested by the enhanced levels of ROS (Figure 2A) and MDA (Figure 2B), and suppressed levels of SOD (Figure 2C) and CAT (Figure 2D), compared to control cells. The cells pretreated with Ech restored levels similar to those of control cells in a dose-dependent manner (Figure 2A–D), indicating that Ech had a protective effect on oxidative damage in HLECs.

Ech reduces cell apoptosis
induced by H2O2 in HLECs

Apoptosis of LECs is the main cytological basis for the formation of various types cataract.2 Here, the TUNEL assay was used to measure HLEC apoptosis after exposure to Ech and/or H2O2. The results illustrated that H2O2 exposure markedly induced apoptosis, compared to the control group (Figure 3A,B). Additionally, Ech treatment could reduce HLEC apoptosis induced by H2O2 in a dose-dependent manner (Figure 3A,B). Moreover, western blot analysis showed that the expression of the anti-apoptotic protein Bcl-2 was markedly suppressed, while the expression of the pro-apoptotic protein Bax was significantly enhanced after the exposure to H2O2, which could be reversed by Ech pretreatment (Figure 3C,D). Furthermore, Ech pretreatment eliminated the increase in caspase-3 activity induced by H2O2 (Figure 3E).

Ech activates the Nrf2/HO-1 pathway in H2O2-treated HLECs

To determine the effects of Nrf2 on H2O2-induced HLECs, the expression levels of Nrf2 and its downstream targets (HO-1 and NQO1) were analyzed using the western blot assay. Since Nrf2 nuclear translocation is an essential step for activation of the Nrf2 pathway, the nuclear localization of Nrf2 in B3 cells was also analyzed using the western blot assay. The results showed that H2O2 significantly reduced Nrf2 nuclear localization, which could be reversed with Ech pretreatment (Figure 4A,B). Moreover, compared to the control group, the expression levels of HO-1 and NQO1 were decreased after the exposure to H2O2, which could be reversed with Ech pretreatment (Figure 4C,D). These data demonstrate that Ech may activate the Nrf2/HO-1 pathway in H2O2-treated HLECs.

Inhibition of Nrf2 by ML385 aggravates H2O2-induced oxidative damage
and apoptosis in HLECs

We assessed the effect of ML385 (5 µM) alone on the viability of B3 cells, and the results confirmed that ML385 had no effect on the viability of B3 cells, but aggravated the inhibitory effect of H2O2 on cell viability (Figure 5A). Compared with the control group, ML385 increased ROS and MDA levels, and decreased SOD and CAT levels in B3 cells. Simultaneously, ML385 enhanced the oxidative stress damage caused by H2O2 (Figure 5B–E). There was no change in the apoptosis rate between the ML385 group and the control group. Importantly, inhibition of Nrf2 by ML385 also exacerbated H2O2-induced cell apoptosis in HLECs (Figure 5F).

ML385 attenuates the protective effect of Ech on H2O2-induced oxidative damage and apoptosis in HLECs

To confirm that Ech could resist H2O2-induced cell damage by activating the Nrf2 pathway, B3 cells were pretreated with Ech (50 µM) and ML385 (5 µM) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h. The results showed that Ech restrained the enhancement of ROS and MDA, and weakened SOD and CAT induced by H2O2, whereas ML385 abolished the effects of Ech on oxidative stress (Figure 6A–D). Similarly, the anti-apoptotic effects of Ech on H2O2-induced B3 cells also could be blocked by ML385 (Figure 6E–G). These results reveal that Ech protected B3 cells against H2O2-induced oxidative injury and apoptosis via activation of the Nrf2 pathway (Figure 7).

Discussion

Oxidative stress has been proven to be an early factor in the development of cataracts,19 and drugs that prevent this event are needed to resist adverse cellular reactions.20 Oxidative stress damage refers to the comprehensive effect of exogenous or endogenous ROS on the cell signal transduction system, or to damage to nucleic acids, proteins and lipid molecules.21 Hydrogen peroxide can induce the production of ROS in cells, leading to oxidative damage.22 Early studies confirmed that high concentrations of H2O2 in the lens and aqueous humor can cause cataracts.23 Exogenous H2O2 treatment is a simple and feasible cell model for studying LEC oxidative damage, which can effectively simulate the process of oxidative damage in LECs that results in cataracts.24 Therefore, in the present study, 100 µM H2O2 was selected as an inducer of oxidative damage in B3 cells.

Previous work has confirmed that the use of antioxidants and certain metabolic receptor agonists can delay the occurrence of cataracts.20 For instance, glycyrrhizin, a substance extracted from licorice, prevents sodium iodate-induced retinal pigment epithelium and retinal injury via the inhibition of ROS.25 Echinatin has been shown to prevent or treat cardiovascular disease, tumors and diabetic nephropathy.26 Recently, the antioxidant properties of Ech have been confirmed.17 Kwak et al.28 reported that Ech can exert anti-cancer effects by inducing ROS/endoplasmic reticulum stress (ERS)-dependent apoptosis. Tian et al.26 proposed that Ech improves myocardial injury caused by ischemia and reperfusion by reducing oxidative stress and apoptosis of cardiomyocytes. However, the effects of Ech on the development of cataracts remain unclear. In the present study, Ech significantly improved the viability of H2O2-treated LECs. Simultaneously, Ech pretreatment prevented the production of ROS and MDA, and enhanced the activity of SOD and CAT in LECs treated with H2O2. These data suggest that Ech can effectively prevent oxygen free radicals from entering the lens, and has a protective effect on the oxidative damage of LECs induced by H2O2 by enhancing cell viability.

Studies on the prevention and treatment of primary or subsequent cataracts mainly focus on how to control the apoptosis of LECs.29, 30 Apoptosis is a type of programmed cell death different from cell necrosis,31 where the mitochondrial apoptotic pathway plays a key role. The Bcl-2 family and the caspase family play important roles in the mitochondrial pathway.32, 33 Bcl-2 family members play a key role in maintaining the integrity of the outer mitochondrial membrane and regulate the release of cytochrome C, which determines the direction of apoptosis regulation in the apoptosis pathway.34 Caspase is a protease system that can lead to the disintegration of apoptotic cells, among which caspase-3 is the main executor of apoptosis.35, 36 In addition to congenital cataracts, the apoptosis of LECs plays a vital role in the formation of other types of cataracts. Studies have shown that p53-dependent LECs can be induced to undergo apoptosis by ultraviolet radiation, which further leads to the formation of cataracts.37, 38 Tamada et al.39 found that apoptosis in selenite cataracts may be an early event, which is reflected in the increase of caspase-3 enzyme activity. Similar studies have found that high glucose can also induce the apoptosis of LECs, which is essential for the formation of cataracts.40, 41 Moreover, cataracts are directly related to the apoptosis of LECs caused by oxygen free radicals.42, 43

It has been reported that antioxidant genes and drugs can inhibit the oxidative damage caused by H2O2 by reducing the activity of ROS.44, 45 Lens epithelial cells initiate apoptosis-related signal transduction pathways under oxidative stress, which mediates the apoptosis of LECs and promote the development of cataracts. Reactive oxygen species promote the entry of cytochrome C into the cytoplasm by oxidizing the thiol group on the adenine nucleotide transporter, and induce the caspase cascade, which ultimately leads to irreversible cell apoptosis.46 Wu et al.47 reported that the ERS inhibitor, salubrinal, reduces the H2O2-induced oxidative stress damage in HepG2 cells by inhibiting cell apoptosis. Here, we analyzed the effect of Ech on the apoptosis of LECs treated with H2O2. Similar to previous reports,48 this study showed that H2O2 induced the apoptosis of HLECs, which may be an important mechanism for the development of cataracts caused by oxidative stress. Additionally, Ech reduced the apoptosis of HLECs induced by H2O2 treatment. Echinatin also inhibited the levels of Bax and caspase-3, and promoted the expression of Bcl-2. These results indicate that Ech can protect HLECs from apoptosis caused by oxidative stress.

Nrf2 signal transduction is a pivotal mechanism to maintain oxidation and antioxidant homeostasis, and to reduce oxidative stress damage.49 Normally, Nrf2 is anchored in the cytoplasm through Keap1. Nrf2 is dissociated from Keap1 and transferred to the nucleus under the stimulation of oxidative stress, phosphorylation or electrophiles.50 HO-1 and NQO1 are the key downstream factors of Nrf2 signal transduction, which are very important in protecting cells from the oxidative damage.51, 52 Recent studies have shown that trimetazidine can delay the formation of age-related cataracts by regulating the expression of Nrf2 and reducing the production of ROS.53 Whitson et al.54 found that LECs lacking glutathione (GSH) depend on the activation of the Nrf2 signaling pathway to trigger oxidative stress. Moreover, Nrf2 inhibitors may increase the oxidative stress of the lens, and Nrf2 inducers can prevent cataract formation by reducing oxidative stress.55 Therefore, Nrf2 pathway activation can be used as a target for the prevention and treatment of age-related cataracts induced by oxidative stress. In the present study, we found that Ech abolished the inhibitory effect of H2O2 on Nrf2 nuclear translocation in B3 cells, as well as the expression of HO-1 and NQO1. Furthermore, administration of the Nrf2 inhibitor ML385 could reverse the protective effect of Ech, suggesting that the potential antioxidant mechanism of Ech may include Nrf2 signal transduction. Importantly, it has been reported that Ech can inhibit activation of the NF-κB pathway56 and the AKT/mTOR pathway.57 Therefore, the protective effect of Ech may also involve other signal pathways, a hypothesis that needs further study. In addition, because the potential toxicity and side effects of Ech and its derivatives are still unclear, there is still a lot of research to be performed before this drug can be applied in the clinic.

Limitations

The therapeutic effect of Ech should be further identified in an animal model of cataracts. In addition, more work needs to be done to elucidate its underlying molecular mechanisms.

Conclusions

As far as we know, the present study is the first demonstration that Ech can protect HLECs from the oxidative stress damage caused by the exposure to H2O2. More importantly, Ech pretreatment reduced cell apoptosis induced by H2O2, providing new directions in the search for novel drugs to prevent and treat cataracts.

Figures


Fig. 1. Echinatin (Ech) attenuates the effects of H2O2 on the viability of human lens epithelial B3 cells (HLECs)
A. Structure of Ech; B. B3 cells were exposed to H2O2 (0–800 µM) for 24 h and the viability of B3 cells was analyzed using the MTT assay; C. B3 cells were treated with Ech (0–100 µM) for 12 h and the viability of B3 cells was analyzed using the MTT assay; D. B3 cells were pretreated with Ech (0–100 µM) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h, and the viability of B3 cells was analyzed using the MTT assay; *p < 0.05, ****p < 0.0001, #p < 0.05, ##p < 0.01, ###p < 0.001.
Fig. 2. Echinatin (Ech) alleviates H2O2-induced oxidative damage in human lens epithelial B3 cells (HLECs)
B3 cells were pretreated with Ech (10 μM, 20 μM and 50 μM) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h. A. The levels of intracellular reactive oxygen species (ROS) were determined using the DCFHDA method; B. The content of malondialdehyde (MDA) was measured using a Lipid Peroxidation MDA Assay Kit; C. The levels of superoxide dismutase (SOD) were analyzed using a Total Superoxide Dismutase Assay Kit; D. The levels of catalase (CAT) were determined using a Catalase Assay Kit. ****p < 0.0001, #p < 0.05, ##p < 0.01, ###p < 0.001, ####p < 0.0001.
Fig. 4. Echinatin (Ech) activates the Nrf2/HO-1 pathway in H2O2-treated human lens epithelial B3 cells (HLECs)
B3 cells were pretreated with Ech (10 μM, 20 μM and 50 μM) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h. A. The expression of nuclear and cytosolic Nrf2 was analyzed using the western blot assay; B. Quantitative results of Nrf2 levels; C. The expression of HO-1 and NQO1 was analyzed using the western blot assay; D. Quantitative results of HO-1 and NQO1 levels; ****p < 0.0001, #p < 0.05, ##p < 0.01, ###p < 0.001, ####p < 0.0001.
Fig. 3. Echinatin (Ech) reduces cell apoptosis induced by H2O2 in human lens epithelial B3 cells (HLECs)
B3 cells were pretreated with Ech (10 μM, 20 μM and 50 μM) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h. A. The TUNEL assay was used to analyze the changes in apoptosis; B. Quantitative results of TUNEL-positive cells; C. The expression of Bcl-2 and Bax was analyzed using the western blot assay; D – Quantitative results of Bcl-2 and Bax levels; E. The activity of caspase-3 was analyzed using a corresponding kit; ****p < 0.0001, #p < 0.05, ##p < 0.01, ###p < 0.001, ####p < 0.0001.
Fig. 5. ML385 aggravates H2O2-induced oxidative damage and cell apoptosis in human lens epithelial B3 cells (HLECs)
B3 cells were pretreated with ML385 (5 µM) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h; the viability of B3 cells was analyzed using the MTT assay (A); reactive oxygen species (ROS) (B), malondialdehyde (MDA) (C), superoxide dismutase (SOD) (D), and catalase (CAT) (E) levels were determined using corresponding kits; F. The TUNEL assay was used to analyze cell apoptosis; *p < 0.05, ****p < 0.0001, #p < 0.05, ##p < 0.01, ###p < 0.001, ####p < 0.0001.
Fig. 6. ML385 attenuates the protective effect of echinatin (Ech) on H2O2-induced oxidative damage and apoptosis in human lens epithelial B3 cells (HLECs)
B3 cells were pretreated with Ech (50 µM) and ML385 (5 µM; an inhibitor of Nrf2) for 12 h, followed by exposure to H2O2 (100 µM) for 24 h; reactive oxygen species (ROS) (A), malondialdehyde (MDA) (B), superoxide dismutase (SOD) (C), and catalase (CAT) (D) levels were determined using corresponding kits; E. The TUNEL assay was used to analyze the changes in apoptosis; F. The expression of Bcl-2 and Bax was analyzed using the western blot assay; G. The activity of caspase-3 was analyzed using a corresponding kit; ***p < 0.001, ****p < 0.0001, ##p < 0.01, ###p < 0.001, ####p < 0.0001, &p < 0.05, &&p < 0.01.
Fig. 7. Schematic diagram of the protective effect of echinatin (Ech) on H2O2-induced oxidative damage and apoptosis in human lens epithelial B3 cells (HLECs)

References (57)

  1. Lee CM, Afshari NA. The global state of cataract blindness. Curr Opin Ophthalmol. 2017;28(1):98–103. doi:10.1097/icu.0000000000000340
  2. Yan Q, Liu JP, Li DW. Apoptosis in lens development and pathology. Differentiation. 2006;74(5):195–211. doi:10.1111/j.1432-0436.2006.00068.x
  3. Shahinfar J, Keshavarzi Z, Ahmadi M, et al. Serum oxidative stress markers in patients with senile cataract and healthy controls. J Coll Physicians Surg Pak. 2018;28(6):448–451. doi:10.29271/jcpsp.2018.06.448
  4. Babizhayev MA, Yegorov YE. Biomarkers of oxidative stress and cataract. Novel drug delivery therapeutic strategies targeting telomere reduction and the expression of telomerase activity in the lens epithelial cells with N-acetylcarnosine lubricant eye drops: Anti-cataract which helps to prevent and treat cataracts in the eyes of dogs and other animals. Curr Drug Deliv. 2014;11(1):24–61. doi:10.2174/15672018113106660062.
  5. Spector A. Oxidative stress-induced cataract: Mechanism of action. FASEB J. 1995;9(12):1173–1182. PMID:7672510
  6. Lu B, Christensen IT. SUMOylation evoked by oxidative stress reduced lens epithelial cell antioxidant functions by increasing the stability and transcription of TP53INP1 in age-related cataracts. Oxid Med Cell Longev. 2019;2019:7898069. doi:10.1155/2019/7898069
  7. Zou Y, Cui B, Liang P, et al. Inhibition of NLRP3 protects human lens epithelial cells against oxidative stress-induced apoptosis by NF-κB signaling. Ophthalmic Res. 2020;63(2):174–181. doi:10.1159/000504303
  8. Tóbon-Velasco JC, Cuevas E, Torres-Ramos MA. Receptor for AGEs (RAGE) as mediator of NF-kB pathway activation in neuroinflammation and oxidative stress. CNS Neurol Disord Drug Targets. 2014;13(9):1615–1626. doi:10.2174/1871527313666140806144831
  9. Beyfuss K, Hood DA. A systematic review of p53 regulation of oxidative stress in skeletal muscle. Redox Rep. 2018;23(1):100–117. doi:10.1080/13510002.2017.1416773
  10. Hybertson BM, Gao B, Bose SK, McCord JM. Oxidative stress in health and disease: The therapeutic potential of Nrf2 activation. Mol Aspects Med. 2011;32(4–6):234–246. doi:10.1016/j.mam.2011.10.006
  11. Periyasamy P, Shinohara T. Age-related cataracts: Role of unfolded protein response, Ca(2+) mobilization, epigenetic DNA modifications, and loss of Nrf2/Keap1 dependent cytoprotection. Prog Retin Eye Res. 2017;60:1–19. doi:10.1016/j.preteyeres.2017.08.003
  12. Gao Y, Yan Y, Huang T. Human age-related cataracts: Epigenetic suppression of the nuclear factor erythroid 2-related factor 2-mediated antioxidant system. Mol Med Rep. 2015;11(2):1442–1447. doi:10.3892/mmr.2014.2849
  13. Ma TJ, Lan DH, He SZ, et al. Nrf2 protects human lens epithelial cells against H(2)O(2)-induced oxidative and ER stress: The ATF4 may be involved. Exp Eye Res. 2018;169:28–37. doi:10.1016/j.exer.2018.01.018
  14. Elanchezhian R, Palsamy P, Madson CJ, Lynch DW, Shinohara T. Age-related cataracts: Homocysteine coupled endoplasmic reticulum stress and suppression of Nrf2-dependent antioxidant protection. Chem Biol Interact. 2012;200(1):1–10. doi:10.1016/j.cbi.2012.08.017
  15. Li T, Ye W, Huang B, et al. Determination and pharmacokinetic study of echinatin by UPLC-MS/MS in rat plasma. J Pharm Biomed Anal. 2019;168:133–137. doi:10.1016/j.jpba.2019.02.023.
  16. Oh HN, Lee MH, Kim E, et al. Dual inhibition of EGFR and MET by echinatin retards cell growth and induces apoptosis of lung cancer cells sensitive or resistant to gefitinib. Phytother Res. 2020;34(2):388–400. doi:10.1002/ptr.6530
  17. Liang M, Li X. Antioxidant mechanisms of echinatin and licochalcone A. Molecules. 2018;24(1):3. doi:10.3390/molecules24010003
  18. Jung ES, Jang HJ, Hong EM, et al. The protective effect of 5-aminosalicylic acid against non-steroidal anti-inflammatory drug-induced injury through free radical scavenging in small intestinal epithelial cells. Medicina (Kaunas). 2020;56(10):515. doi:10.3390/medicina56100515
  19. Nita M, Grzybowski A. The role of the reactive oxygen species and oxidative stress in the pathomechanism of the age-related ocular diseases and other pathologies of the anterior and posterior eye segments in adults. Oxid Med Cell Longev. 2016;2016:3164734. doi:10.1155/2016/3164734
  20. Liang B, Wei W, Wang J, et al. Protective effects of Semiaquilegia adoxoides n-butanol extract against hydrogen peroxide-induced oxidative stress in human lens epithelial cells. Pharm Biol. 2016;54(9):1656–1663. doi:10.3109/13880209.2015.1113993
  21. Sies H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015;4:180–183. doi:10.1016/j.redox.2015.01.002
  22. Sies H. Role of metabolic H2O2 generation: Redox signaling and oxidative stress. J Biol Chem. 2014;289(13):8735–8741. doi:10.1074/jbc.R113.544635
  23. Spector A, Garner WH. Hydrogen peroxide and human cataract. Exp Eye Res. 1981;33(6):673–681. doi:10.1016/s0014-4835(81)80107-8
  24. Li X, Meng F, Li H, et al. L-carnitine alleviates oxidative stress-related damage via MAPK signaling in human lens epithelial cells exposed to H2O2. Int J Mol Med. 2019;44(4):1515–1522. doi:10.3892/ijmm.2019.4283
  25. He H, Wei D, Liu H, et al. Glycyrrhizin protects against sodium iodate-induced RPE and retinal injury though activation of AKT and Nrf2/HO-1 pathway. 2019;23(5):3495–3504. doi:10.1111/jcmm.14246
  26. Tian XH, Liu CL, Jiang HL, et al. Cardioprotection provided by echinatin against ischemia/reperfusion in isolated rat hearts. BMC Cardiovasc Disord. 2016;16:119. doi:10.1186/s12872-016-0294-3
  27. Ke Z, Su Z, Zhang X, et al. Discovery of a potent angiotensin converting enzyme inhibitor via virtual screening. Bioorg Med Chem Lett. 2017;27(16):3688–3692. doi:10.1016/j.bmcl.2017.07.016
  28. Kwak AW, Choi JS, Lee MH, Oh HN, Cho SS. Retrochalcone echinatin triggers apoptosis of esophageal squamous cell carcinoma via ROS- and ER stress-mediated signaling pathways. Molecules. 2019;24(22):4055. doi:10.3390/molecules24224055
  29. Xiang J, Kang L, Gao H, et al. BLM can regulate cataract progression by influencing cell vitality and apoptosis. Exp Eye Res. 2019;178:99–107. doi:10.1016/j.exer.2018.08.022
  30. Wang HM, Li GX, Zheng HS, Wu XZ. Protective effect of resveratrol on lens epithelial cell apoptosis in diabetic cataract rat. Asian Pac J Trop Med. 2015;8(2):153–156. doi:10.1016/s1995-7645(14)60307-2
  31. Fleisher TA. Apoptosis. Ann Allergy Asthma Immunol. 1997;78(3):245–249, quiz 249–250. doi:10.1016/s1081-1206(10)63176-6
  32. Piro LD. Apoptosis, Bcl-2 antisense, and cancer therapy. Oncology (Williston Park). 2004;18(13 Suppl 10):5–10. PMID:15651171
  33. Fan TJ, Han LH, Cong RS, Liang J. Caspase family proteases and apoptosis. Acta Biochim Biophys Sin (Shanghai). 2005;37(11):719–727. doi:10.1111/j.1745-7270.2005.00108.x
  34. Bruckheimer EM, Cho SH, Sarkiss M, Herrmann J, McDonnell TJ. The Bcl-2 gene family and apoptosis. Adv Biochem Eng Biotechnol. 1998;62:75–105. doi:10.1007/BFb0102306
  35. Boatright KM, Salvesen GS. Caspase activation. Biochem Soc Symp. 2003;70(70):233–242. doi:10.1042/bss0700233
  36. Fulda S. Therapeutic opportunities based on caspase modulation. Semin Cell Dev Biol. 2018;82:150–157. doi:10.1016/j.semcdb.2017.12.008
  37. Galichanin K, Löfgren S, Bergmanson J, Söderberg P. Evolution of damage in the lens after in vivo close to threshold exposure to UV-B radiation: Cytomorphological study of apoptosis. Exp Eye Res. 2010;91(3):369–377. doi:10.1016/j.exer.2010.06.009
  38. Ayala M, Strid H, Jacobsson U, Söderberg PG. p53 expression and apoptosis in the lens after ultraviolet radiation exposure. Invest Ophthalmol Vis Sci. 2007;48(9):4187–4191. doi:10.1167/iovs.06-0660
  39. Tamada Y, Fukiage C, Nakamura Y, et al. Evidence for apoptosis in the selenite rat model of cataract. Biochem Biophys Res Commun. 2000;275(2):300–306. doi:10.1006/bbrc.2000.3298
  40. Higashi Y, Higashi K, Mori A, et al. Anti-cataract effect of resveratrol in high-glucose-treated streptozotocin-induced diabetic rats. Biol Pharm Bull. 2018;41(10):1586–1592. doi:10.1248/bpb.b18-00328
  41. Li D, Liu GQ, Lu PR. High glucose: Activating autophagy and affecting the biological behavior of human lens epithelial cells. Int J Ophthalmol. 2019;12(7):1061–1066. doi:10.18240/ijo.2019.07.02
  42. Gu XL. MicroRNA-124 prevents H2O2-induced apoptosis and oxidative stress in human lens epithelial cells via inhibition of the NF-κB signaling pathway. Pharmacology. 2018;102(3–4):213–222. doi:10.1159/000491433
  43. Zhang Y, Huang WR. Sanguinarine induces apoptosis of human lens epithelial cells by increasing reactive oxygen species via the MAPK signaling pathway. Mol Med Rep. 2019;19(5):4449–4456. doi:10.3892/mmr.2019.10087
  44. Chen S, Sun P, Zhao X, et al. Gardenia jasminoides has therapeutic effects on L-NNA-induced hypertension in vivo. Mol Med Rep. 2017;15(6):4360–4373. doi:10.3892/mmr.2017.6542
  45. Mitter SK, Song C, Qi X, et al. Dysregulated autophagy in the RPE is associated with increased susceptibility to oxidative stress and AMD. Autophagy. 2014;10(11):1989–2005. doi:10.4161/auto.36184
  46. Sharma KK, Santhoshkumar P. Lens aging: Effects of crystallins. Biochim Biophys Acta. 2009;1790(10):1095–1108. doi:10.1016/j.bbagen.2009.05.008
  47. Wu Z, Wang H, Fang S, Xu C. Roles of endoplasmic reticulum stress and autophagy on H2O2-induced oxidative stress injury in HepG2 cells. Mol Med Rep. 2018;18(5):4163–4174. doi:10.3892/mmr.2018.9443
  48. Ma T, Chen T, Li P, et al. Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Exp Eye Res. 2016;146:318–329. doi:10.1016/j.exer.2016.02.013
  49. Luo YH, Cheng HJ, Tsai FY, Tsou TC, Lin SY. Primary amine modified gold nanodots regulate macrophage function and antioxidant response: Potential therapeutics targeting of Nrf2. Int J Nanomedicine. 2020;15:8411–8426. doi:10.2147/ijn.s268203
  50. Tamatam CM, Reddy NM, Potteti HR, et al. Preconditioning the immature lung with enhanced Nrf2 activity protects against oxidant-induced hypoalveolarization in mice. Nature. 2020;10(1):19034. doi:10.1038/s41598-020-75834-8
  51. Waz S, Heeba GH, Hassanin SO, Abdel-Latif RG. Nephroprotective effect of exogenous hydrogen sulfide donor against cyclophospha­mide-induced toxicity is mediated by Nrf2/HO-1/NF-κB signaling pathway. Life Sci. 2020;264:118630. doi:10.1016/j.lfs.2020.118630
  52. Ma JQ, Zhang YJ, Tian ZK, Liu CM. Bixin attenuates carbon tetrachloride induced oxidative stress, inflammation and fibrosis in kidney by regulating the Nrf2/TLR4/MyD88 and PPAR-γ/TGF-β1/Smad3 pathway. Int Immunopharmacol. 2020;90:107117. doi:10.1016/j.intimp.2020.107117
  53. Fang W, Ye Q, Yao Y, et al. Protective effects of trimetazidine in retarding selenite-induced lens opacification. Curr Eye Res. 2019;44(12):1325–1336. doi:10.1080/02713683.2019.1633359
  54. Whitson JA, Wilmarth PA, Klimek J, et al. Proteomic analysis of the glutathione-deficient LEGSKO mouse lens reveals activation of EMT signaling, loss of lens specific markers, and changes in stress response proteins. Free Radic Biol Med. 2017;113:84–96. doi:10.1016/j.freeradbiomed.2017.09.019
  55. Liu XF, Hao JL, Xie T, et al. Nrf2 as a target for prevention of age-related and diabetic cataracts by against oxidative stress. Aging Cell. 2017;16(5):934–942. doi:10.1111/acel.12645
  56. Funakoshi-Tago M, Tanabe S, Tago K, et al. Licochalcone A potently inhibits tumor necrosis factor alpha-induced nuclear factor-kappaB activation through the direct inhibition of IkappaB kinase complex activation. Mol Pharmacol. 2009;76(4):745–753. doi:10.1124/mol.109.057448
  57. Hong P, Liu QW, Xie Y, et al. Echinatin suppresses esophageal cancer tumor growth and invasion through inducing AKT/mTOR-dependent autophagy and apoptosis. Cell Death Dis. 2020;11(7):524. doi:10.1038/s41419-020-2730-7