Open Access

Neuroprotective effect of emodin against Alzheimer's disease via Nrf2 signaling in U251 cells and APP/PS1 mice

Corrigendum in: /10.3892/mmr.2023.12994

  • Authors:
    • Zhiping Li
    • Hui Bi
    • Hongbo Jiang
    • Jingjing Song
    • Qingfan Meng
    • Yizhi Zhang
    • Xiaofang Fei
  • View Affiliations

  • Published online on: December 2, 2020     https://doi.org/10.3892/mmr.2020.11747
  • Article Number: 108
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Emodin is a naturally‑occurring medicinal herbal ingredient that possesses numerous pharmacological properties, including anti‑inflammatory and antioxidant effects. In the present study, potential neuroprotective effects associated with the antioxidant activity of emodin were assessed in U251 cells that were subjected to β‑amyloid peptide (Aβ)‑induced apoptosis and in amyloid precursor protein (APP)/presenilin‑1 (PS1) double‑transgenic mice. U251 is a type of human astroglioma cell line (cat. no. BNCC337874; BeNa Culture Collection). In apoptotic U251 cells, 3‑h emodin pre‑treatment prior to 24‑h Aβ co‑exposure improved cell viability, suppressed lactate dehydrogenase leakage and caspase‑3, ‑8 and ‑9 activation to inhibit apoptosis. Compared with those after Aβ exposure alone, emodin ameliorated the dissipation of the mitochondrial membrane potential, inhibited the over‑accumulation of reactive oxygen species, enhanced the expression levels of nuclear factor‑erythroid‑2‑related factor 2 (Nrf2), haemeoxygenase‑1, superoxide dismutase 1, Bcl‑2 and catalase in addition to decreasing the expression levels of Bax. In APP/PS1 mice, an 8‑week course of emodin administration improved spatial memory and learning ability and decreased anxiety. Emodin was also found to regulate key components in the Nrf2 pathway and decreased the deposition of Aβ, phosphorylated‑τ and 4‑hydroxy‑2‑nonenal in APP/PS1 mice. Taken together, the present data suggest that emodin may serve as a promising candidate for the treatment of Alzheimer's disease.

Introduction

Alzheimer's disease (AD) is characterised by behavioural impairment and cognitive dysfunction (1) and is associated with age, where the prevalence may be as high as 50% among humans aged >95 years (2). The pathological cause of AD has not been fully elucidated. Previous research has indicated that the accumulation of β-amyloid peptide (Aβ) in the brain may be an important contributor to the development of AD (3). In patients with AD, Aβ aggregates that form around neurons not only induce neuronal apoptosis but also trigger a cascade of cellular damage, including τ hyperphosphorylation and mitochondrial reactive oxygen species (ROS) production (4). Previous reports have indicated that symptoms of AD are attributable to the occurrence of oxidative stress (2,5,6). In particular, oxidative stress induces mitochondrial dysfunction and excessive ROS accumulation, ultimately leading to neuronal cell death (7).

Mitochondria are the primary source of intracellular ROS and are key targets of Aβ toxicity during the AD pathological process (8). In addition to increasing the levels of Aβ in the brain, overaccumulation of ROS and subsequent collapse of mitochondrial membrane potential (MMP) lead to reductions in ATP levels (9). Therefore, the function and integrity of the mitochondria must be maintained to protect nerve cells. During the occurrence and development of oxidative stress, the transcription factor nuclear factor E2-related factor 2 (Nrf2) dissociates from Kelch-like ECH-associated protein 1 (Keap-1) to activate numerous downstream proteins and detoxification enzymes, including superoxide dismutase 1 (SOD 1) and haemeoxygenase-1 (HO-1) (10,11). Since activation of the Nrf2 pathway has been documented to ameliorate oxidative stress and decreases accumulation of Aβ (12,13), rendering Nrf2 to be a potential therapeutic target for AD (14).

Emodin, which is structurally known as 1,3,8-trihydroxy-6-methylanthraquinone (Fig. 1), is a naturally occurring anthraquinone derivative. This compound can be isolated from medicinal herbs, including Rheum palmatum (15), Cassia obtusifolia (16) and Polygonum multiflorum (17), and has been used as a laxative in eastern Asia for 2,000 years (18). Previous studies have indicated that emodin is multifunctional. For example, evidence suggests that emodin enhances nerve cell survival by upregulating Bcl-2 expression levels and blocking Aβ-induced autophagy (19). In addition, emodin exhibits antioxidative effects: In viral myocarditis, emodin alleviates oxidative stress by increasing myocardial SOD expression levels whilst decreasing malondialdehyde expression levels (20). Emodin has also been shown to decrease collagen overproduction and inflammation by stimulating the Nrf2-antioxidant signalling pathway in pulmonary fibrosis (21), and to serve important roles in neurological disorders (22). For example, emodin protects neurons from Aβ25-35-induced neurotoxicity and inhibits excess τ accumulation in cortical neurons (19,23). To the best of our knowledge, however, no reports have yet described the systematic protective effects of emodin against AD or Aβ deposition in either in vitro or in vivo mouse models.

In the present study, amyloid precursor protein (APP)/presenilin 1 (PS1) double-transgenic mice and U251 cells subjected to Aβ1-42-induced apoptosis were used to determine the neuroprotective effects of emodin against AD. Emodin exerted protective effects against Aβ toxicity in U251 cells and ameliorated behavioural effects in the double-transgenic mouse model. The experimental data indicated that emodin protected neurons against neurodegeneration in AD.

Materials and methods

Cell culture

U251 cells (cat. no. KCB200965Y), a human glioma cell line that was purchased from the Cell bank of Chinese Academy of Sciences, were cultured in high glucose DMEM containing 10% fetal bovine serum, 1% 100 U/ml penicillin and 100 µg/ml streptomycin at 37°C in a 5% CO2 incubator (Thermo Fisher Scientific, Inc.) to provide a humidified atmosphere. All reagents were purchased from Invitrogen, Thermo Fisher Scientific, Inc.

Measurement of cell viability

U251 cells (5×103 cells/well) cultured in plates (96-well) were pre-treated with emodin (purity, ≥90%; CAS no. 518-82-1; cat. no. E7881; Sigma-Aldrich; Merck KGaA) at doses of 25 and 50 µM at 37°C for 3 h and then co-incubated with 15 µM Aβ1-42 [cat. no. 87233; Gill Biochem (Shanghai) Co., Ltd.] or culture medium at 37°C for a further 24 h. Non-treated cells served as the control. Cell viability was analysed using MTT assay as described previously (24). Briefly, cells were exposed to 5 mg/ml (final concentration) MTT at 37°C for 4 h in the dark before the formazan precipitates were dissolved in 100 µl DMSO (Sigma-Aldrich; Merck KGaA). Absorbance was analysed using a Synergy™ 4 Microplate Reader at 490 nm (BioTek Instruments, Inc.).

Measurement of lactate dehydrogenase (LDH) levels and activity of caspases-3, −8 and −9

U251 cells (2×105 cells/well) were cultured in plates (6-well) and treated with emodin at doses of 25 and 50 µM at 37°C for 3 h, then co-incubated with 15 µM Aβ1-42 or culture medium at 37°C for another 24 h. Culture medium-only-treated U251 cells served as the control group. The intracellular levels of LDH and the activities of caspase-3, −8 and −9 were detected using LDH (cat. no. C0016), caspase-3 (cat. no. C1168M), −8 (cat. no. C1151) and −9 (cat. no. C1157) Activity Assay kits (Beyotime Institute of Biotechnology) according to manufacturer's protocols.

Flow cytometry assay

U251 cells (2×105 cells/well) were cultured in 6-well plates with emodin at doses of 25 and 50 µM at 37°C for 3 h before being co-incubated with 15 µM Aβ1-42 or culture medium for a further 24 h. Non-treated cells served as the control. Cells (1×105) were collected and incubated with Annexin V and propidium iodide (cat. no. APT750; EMD Millipore) for 15 min at 37°C in the dark, and detected using a Muse Cell Analyzer (EMD Millipore) and analyzed with FlowJo v10 (FlowJo LLC).

Measurement of MMP and ROS levels

U251 cells (2×105 cells/well) cultured in plates (6-well) with emodin at doses of 25 and 50 µM at 37°C for 3 h and then co-incubated with 15 µM Aβ1-42 or culture medium at 37°C for a further 24 h. Culture medium-only-treated U251 cells served as the control group. MMP was analysed using a MMP Detection kit (cat. no. M8650; Beijing Solarbio Science & Technology Co., Ltd.) according to the manufacturer's instructions. The red fluorescence represented the higher membrane potential, and the green fluorescence represented the damaged or lower membrane potential. Intracellular ROS was analysed using a Reactive Oxygen Detection kit (cat. no. S0033; Beyotime Institute of Biotechnology) according to the manufacturer's instructions. After the cells were washed, the fluorescence intensities were detected using a fluorescence microscope (magnification, ×400; Eclipse TE 2000-S; Nikon Corporation). Quantitative data analysis was performed using ImageJ software version 1.46 (National Institutes of Health), where the data were presented as the fluorescence intensity.

Animals and treatment

For the animal model, 8-month old male (42–49 g) B6C3-Tg (genotype APPswe, PSEN1De9/Nju) APP/PS1 double-transgenic mice (n=30) and wild-type (WT) littermates (n=10) were purchased from Nanjing Biomedical Research Institute of Nanjing University [SCXK (Su) 2015-0001; Nanjing, China]. All mice were housed in a temperature-controlled environment with a standard 12-h light/dark cycle at 23±1°C and 40–60% humidity, with access to food and water ad libitum.

The present study was approved by the Animal Ethics Committee of School of Life Sciences, Jilin University (approval no. SY20171208; Changchun, China). APP/PS1 mice were randomly divided into model (n=10; orally received 10 ml/Kg normal saline) and emodin-treated groups [orally received 10 (n=10) or 20 mg/kg (n=10) emodin once per day for 8 continuous weeks]. WT mice (n=10) orally received normal saline once per day for 8 continuous weeks. On the last five days of agent administration, behavioural tests were performed on each mouse, and blood was collected from the caudal vein of the mice under anaesthesia with isoflurane inhalation with an initial level of 4% and a maintenance level of 1.2%. Mice were then euthanized using a small animal euthanasia system (32×25×20 cm; cat. no. CL-1000-S2; Shanghai Yuyan Scientific Instrument Co., Ltd.). Briefly, mice were placed in the carbon dioxide tank for 2 min at a CO2 concentration of 30%. The whole brain tissue was collected from each mouse for further investigation.

Animal behaviour detection

For experimental mice, learning and memory ability were analysed using Morris water maze test (MWM), whilst anxiety was evaluated using the open field test as previously described (25). For the MWM test, water was mixed with titanium dioxide to form a white liquid to hide the platform and the mice were placed in the water maze (cat. no. MT-200; Techman Software Co., Ltd.) to locate a platform that was hidden in white water. The route taken by mice was then recorded using the watermaze software (version 2.0; Techman Software Co., Ltd.). In MWM, mice were trained for 7 days starting at week 7 to allow them to learn or remember the position of platform. Open field test was used to evaluate anxiety and performed on the day after the MWM test. An open field experimental video analyser was used to record the path taken by each mouse (cat. no. 1056306; Zhong Shi Di Chuang). The box was washed after each experiment to clear any remaining scents or traces that could affect the results of the next test.

Immunohistochemical procedures

Brain tissue was fixed with 4% formalin solution at 25°C for 24 h, and then dehydrated with 30, 50, 70, 80, 95 and 100% ethanol, washed in xylene, embedded in paraffin and cut into 5-µm thick sections. All slides were gradient hydrated with 100, 95, 80, 70 and 50% ethanol and distilled water respectively (26). Following dewaxing and hydration, the brain slides were boiled in 10 mM sodium citrate buffer (pH 6.0) for 10 min. After cooling, the slides were incubated with 3% hydrogen peroxide for 10 min and blocked using 10% goat serum for 30 min at 25°C. The slides were then incubated with primary antibodies against phosphorylated (p)-τ (1:100; cat. no. SC12414; Santa Cruz Biotechnology, Inc.), Aβ1-42 (1: 500; cat. no. ab32136; Abcam) and 4-hydroxy-2-nonenal (4-HNE; 1:200; cat. no. ab46545; Abcam) overnight at 4°C, followed by 1-h incubation at 25°C with biotinylated horseradish peroxidase-conjugated secondary antibody (anti-rabbit; 1:500; cat. no. sc-3836; Santa Cruz Biotechnology, Inc.). After visualization using 3,3′-diaminobenzidine and Mayer's hematoxylin and immunoperoxidase staining of Aβ1-42, p-τ and 4-HNE at 25°C for 5 min, images were captured using light microscopy (magnification, ×200; cat. no. IX73; Olympus Corporation).

Western blot analysis

U251 cells (2×105 cells/well) were cultured at 37°C in 6-well plates with emodin at doses of 25 and 50 µM for 3 h, and co-incubated with 15 µM Aβ1-42 or culture medium at 37°C for a further 24 h. Culture medium only-treated U251 cells served as the control group. Treated cells and brain tissues were lysed using radio immunoprecipitation assay buffer (cat. no. 89900, Thermo Fisher, Inc.) containing 1% protease inhibitor cocktail (Sigma Aldrich; Merck KGaA). After detecting the protein concentration using a bicinchoninic acid protein assay kit, 40 µg protein/lane was separated by 12% SDS-PAGE and transferred onto 0.45 µm nitrocellulose membranes (Bio Basic, Inc.). The membranes were then blocked with 5% skimmed milk at room temperature for 1 h and incubated with primary antibodies against anti-SOD 1 (cat. no. sc-17767), anti-HO-1 (cat. no. sc-136960), anti-CAT (cat. no. sc-271803), anti-Nrf2 (cat. no. sc-365949), anti-Bax (cat. no. sc-7480), anti-Bcl-2 (cat. no. sc-7382) and GAPDH (cat. no. sc-47724; all from Santa Cruz Biotechnology, Inc.) at dilutions of 1:5,000 overnight at 4°C. The membranes were subsequently incubated with horseradish peroxidase-conjugated mouse anti rabbit secondary antibodies at 4°C for 4 h (dilution 1:5,000; cat. no. bs-0295G; Bioss). ECL Detection reagent (cat. no. PE0010; Beijing Solarbio Science & Technology Co., Ltd.) was then used to visualise the specific bands, where the intensity was quantified by ImageJ software (v1.46r National Institutes of Health).

Statistical analysis

Data are expressed as the mean ± SEM. Experimental repeats were n=5 in the slide staining experiments and n=6 in the other experiments, including apoptosis, ELISA and western blotting. One-way ANOVA followed by Tukey's post hoc test was used to determine statistical significance using SPSS 16.0 software (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Emodin protects U251 cells against Aβ1-42-induced cell apoptosis

Exposure to cytotoxic Aβ1-42 (15 µM) for 24 h led to a 50.66% decrease in the viability of U251 cells (P<0.001). However, co-treatment with emodin at 25 and 50 µM led to 16.4 and 18.9% increases in viability, respectively (both P<0.05; Fig. 2A). By contrast, incubation with emodin alone for 24 h had no effects on cell viability (Fig. 2A).

The cytotoxicity of Aβ1-42 treatment was next evaluated by measuring the release of LDH. Notably, 3-h pre-treatment with 50 µM emodin resulted in a 39.9% decrease of LDH in cells exposed to Aβ1-42 for 24 h (P<0.01; Fig. 2B). Furthermore, co-treatment with emodin at 25 µM and 50 µM suppressed Aβ1-42-induced apoptosis of U251 cells by 11.5% (P<0.05) and 17.3% separately (P<0.01 Fig. 2F and G). The activation of caspase −3, −8 and −9, which are classical markers of apoptosis (27), was also subsequently measured. It was found that 50 µM emodin treatment led to decreases in the activity of caspase-3, −8 and −9 by 39.9% (P<0.01; Fig. 2C), 25.9% (P<0.05; Fig. 2D) and 15.9% (P<0.05; Fig. 2E), respectively, in Aβ1-42-exposed U251 cells.

Emodin ameliorates Aβ1-42-induced mitochondrial dysfunction and activates the Nrf2 pathway

Mitochondrial injury one of the early events of cell apoptosis (28). MMP was observed to be significantly dissipated in Aβ1-42-exposed U251 cells. However, co-incubation with emodin, particularly at 50 µM, restored the MMP, as indicated by decreased green and increased red fluorescence (Fig. 3A). The mitochondria are the major site of ROS production in cells, such that accumulation of ROS may cause further mitochondrial dysfunction (29,30). In U251 cells exposed to Aβ1-42 for 24 h, 3-h pre-treatment with 50 µM emodin successfully suppressed the overaccumulation of intracellular ROS, as indicated by the decreased green fluorescence intensity (Fig. 3B).

Both anti- and pro-apoptotic members of the Bcl-2 protein family serve key roles in modulating mitochondrial cell apoptosis (31). Compared with U251 cells exposed to Aβ1-42 alone, those co-treated with emodin exhibited significantly increased Bcl-2 (P<0.001) and decreased Bax expression levels (P<0.01; Fig. 3C).

Nrf2 is activated to regulate the functions of mitochondria during the initiation and progression of oxidative stress (32). Compared with Aβ1-42-exposed U251 cells, emodin-treated cells exhibited increased expression levels of Nrf2 (P<0.05), SOD 1 (P<0.01), HO-1 (P<0.001) and the antioxidant enzyme CAT (P<0.01; Fig. 3C). Taken together, these results suggest that emodin restored MMP and decreased ROS in cells whilst activating the Nrf2 pathway.

Emodin improves AD-like behaviour and suppresses Aβ1-42, p-τ and 4-HNE deposition in APP/PS1 mice

Cognitive impairment and anxiety are the primary clinical manifestations of AD (33). Therefore, MWM and open field tests were used to evaluate these manifestations in APP/PS1 mice. Compared with untreated mice, those treated with emodin at 20 mg/kg exhibited a 14.3% decrease in time required to locate the platform during the MWM test, indicating that emodin enhanced the spatial memory and learning ability of the mice (P<0.05; Fig. 4A). In the open field test, mice treated with emodin at 20 mg/kg spent significantly less time in the centre of the field, which indicated that this agent ameliorated anxiety in the mouse model (P<0.01; Fig. 4B).

H&E staining was used to detect neuronal damage in brain tissue. However, there was no obvious pathological reaction in the brain (Fig. 4C). The presence of extracellular Aβ and intracellular neurofibrillary tangles in the brain are two defining aetiological characteristics of AD (34). Compared with untreated APP/PS1 mice, emodin-treated mice, particularly those that received a dose of 20 mg/kg, exhibited lower expression levels of Aβ1-42 (Fig. 4D) and p-τ (Fig. 4E) in the brain. Tissue was also stained for 4-HNE, a biomarker of oxidative stress, to further elucidate the function of emodin in this process. Notably, emodin decreased expression levels of 4-HNE in the brain compared with those in untreated APP/PS1 mice (Fig. 4F).

Emodin regulates Nrf2 signalling in the brain of mice. To further investigate the possible mechanism by which emodin exerts its effects in APP/PS1 mice, the expression levels of proteins associated with the Bcl-2 family and the Nrf2 pathway, in addition to the antioxidant enzyme CAT, were evaluated. Compared with those in untreated APP/PS1 mice, 20 mg/kg emodin exhibited significantly increased expression levels of Bcl-2 (P<0.01) and significantly decreased expression levels of Bax in the brain tissue of mice (P<0.001; Fig. 5). Furthermore, an 8-week course of 20 mg/kg emodin administration led to significant increases in Nrf2 (P<0.01), SOD 1 (P<0.01) and HO-1 expression (P<0.001) and a significant decrease in CAT expression in the brain tissue, even at an emodin dose of 10 mg/kg (P<0.01; Fig. 5). These results demonstrated the ability of emodin to regulate Nrf2 pathway activity in APP/PS1 mice and its potential as an antioxidant.

Discussion

The majority of symptoms of AD can be attributed to oxidative stress (35,36). Therefore, the elimination of excess ROS or induction of endogenous antioxidant activity may be a potential strategy for the treatment of AD (37). Emodin is a biologically active compound that can be found in a number of herbal laxatives and exhibits pharmacological activity due to its strong antioxidative effects (38). The present study confirmed the neuroprotective effects of emodin against AD by using U251 cells subjected to Aβ1-42-induced apoptosis and in APP/PS1 double-transgenic mice.

The U251 human astrocyte cell line has previously been used to investigate AD (39). In the present study, U251 cells were treated with Aβ1-42 to induce cell injury. Aβ, a peptide containing 39–43 amino acids, has been shown to exert numerous toxic effects both in vitro and in vivo (40). For example, Aβ accumulation is neurotoxic and can depolarise the cell membrane, decrease mitochondrial potential and increase the production of ROS, induce synaptic dysfunction and cause oxidative stress to mediate mitochondrial damage (41). The present study demonstrated that emodin improved cell viability and suppressed apoptosis in cells exposed to Aβ1-42. Apoptotic neuronal death causes fatal injury to the brain tissue (42). Caspases-3, −8 and −9 are major components of the classical apoptosis pathway (43). Caspase-8 is located primarily in the mitochondria and is activated to form the apoptosome, which recruits pro-caspase-9 and induces pro-caspase-3 cleavage, ultimately leading to mitochondrial apoptosis (44).

The mitochondria are energy-producing organelles that serve as the primary source of ROS (45). Aβ accumulation directly exhibits negative effects on mitochondrial energy metabolism, specifically on α-ketoglutarate dehydrogenase and pyruvate dehydrogenase activity, which ultimately results in cell apoptosis (46). The Bcl-2 family member of proteins, which include the pro-apoptotic protein Bax and the anti-apoptotic protein Bcl-2, have also been reported to be involved in mitochondrial injury (47). The pro-apoptotic protein Bax is activated in response to apoptotic stimuli (48), which leads to mitochondrial oxidative respiratory chain damage and decreased MMP (49).

In the present study, emodin also enhanced the activity of the antioxidant enzyme CAT and activated the transcription factor Nrf2. The latter is known to regulate a number of cytoprotective and detoxification genes. For example, Nrf2 binds with Keap-1 in the cytoplasm, which represses Nrf2 nuclear translocation (50). However, this interaction is disrupted by oxidative stress, which allows Nrf2 to translocate into the nucleus t interact with the antioxidant response element, thus activating an effector cascade involving HO-1 and SOD1 (10). Both HO-1 and SOD1 are powerful antioxidative reagents that can neutralize toxic superoxide radicals produced in AD (51,52). In addition to oxidative stress, Nrf2 controls the expression of nuclear genes that encode mitochondrial proteins, which affect mitochondrial biological function. For example, Nrf2 deficiency affects mitochondrial electron transport chain activity, fatty acid oxidation and availability of substrates (NADH and FADH2/succinate) for respiration, and ATP synthesis (53), furthermore, it can also exacerbate APP and τ pathology (54). In the present study, emodin strongly decreased activity of casapase-3, −8 and −9, improved mitochondrial function, decreased ROS accumulation, enhanced the Bcl-2/Bax ratio and activated the Nrf2 pathway in both U251 cells subjected to Aβ1-42-induced apoptosis and APP/PS1 mice, suggesting that emodin exerts both antioxidant and neuroprotective effects.

In the present study, double transgenic APP/PS1 mice were used to generate a model of AD with severe pathology. In this transgenic model, overexpression of the gene encoding APP and a mutant form of PS1 have previously been demonstrated to impair the processing of amyloid proteins and increase levels of Aβ (55). Accumulation of Aβ exacerbates cognitive impairment and induces anxiety in humans with AD (56). Therefore, the manifestations in APP/PS1 mice were similar to AD-like symptoms (57). The present study used a MWM test to evaluate the spatial learning ability (58) of mice and the open field test to evaluate anxiety (59). In neurons, aggregation of Aβ affects the activity of kinases and phosphatases, leading to the hyperphosphorylation of τ protein and formation of neurofibrillary tangles (60). An increase in the number of neuroinflammatory plaques caused by excess p-τ is another pathological feature of AD that has previously been observed in the brain of APP/PS1 mice (61). In turn, excess p-τ inhibits kinesin-dependent transportation and blocks APP transport into axons and dendrites, which increases accumulation of Aβ in the neurons (62). In the present study, emodin treatment led to significant improvements in AD-like behaviour of APP/PS1 mice and decreased the levels of aggregated Aβ, pathogenic τ and the peroxidation product 4-HNE.

Increases in the production of H2O2 and other oxidative products lead to accumulation of Aβ and thus contribute to the initiation and progression of AD (35,63,64). An effective antioxidant therapy is necessary to relieve the symptoms of AD. The present study demonstrated that emodin improved AD-associated behaviour in APP/PS1 mice, ameliorated severe oxidative stress and activated the Nrf2 pathway both in vivo and in vitro.

In conclusion, emodin was demonstrated to exert protective effects against Aβ1-42-induced cell apoptosis in vitro and Aβ deposition in vivo in APP/PS1 double-transgenic mice. These effects are likely due to the Nrf2-mediated anti-oxidative activity of emodin and suggest a potential role for this agent in AD protection.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Special Projects of the Cooperation between Jilin University and Jilin Province (grant no. SXGJXX2017-1), the Projects from the Science and Technology Department of Jilin Province in China (grant nos. 20200708091YY and 20200708068YY) and the Department of Finance of Jilin Province, China (grant no. JLSCZD2019-012).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YZ and XF designed the experiments, drafted and revised the manuscript. ZL, HB and HJ performed the experiments and analyzed the data. JS and QM revised the manuscript critically for important intellectual content. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The experiments were approved by Animal Ethics Committee of School of Life Sciences, Jilin University (approval no. SY20171208).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

APP

amyloid precursor protein

PS1

presenilin-1

LDH

lactate dehydrogenase

ROS

reactive oxygen species

AD

Alzheimer's disease

MMP

mitochondrial membrane potential

Keap-1

Kelch-like ECH-associated protein-1

4-HNE

4-hydroxy-2-nonenal

SOD

superoxide dismutase

CAT

catalase

HO-1

heme oxygenase-1

MWM

Morris water maze

References

1 

Rygiel K and Rygiel K: Novel strategies for Alzheimer's disease treatment: An overview of anti-amyloid beta monoclonal antibodies. Indian J Pharmacol. 48:629–636. 2016. View Article : Google Scholar

2 

Viña J, Lloret A, Giraldo E, Badia MC and Alonso MD: Antioxidant pathways in Alzheimers disease: Possibilities of intervention. Curr Pharm Des. 17:3861–3864. 2011. View Article : Google Scholar

3 

Evans DA, Beckett LA, Field TS, Feng L, Albert MS, Bennett DA, Tycko B and Mayeux R: Apolipoprotein E epsilon4 and incidence of Alzheimer disease in a community population of older persons. JAMA. 277:822–824. 1997. View Article : Google Scholar

4 

Faizi M, Seydi E, Abarghuyi S, Salimi A, Nasoohi S and Pourahmad J: A Search for Mitochondrial Damage in Alzheimer's Disease Using Isolated Rat Brain Mitochondria. Iran J Pharm Res. 15 (Suppl):185–195. 2016.

5 

Chen Z and Zhong C: Oxidative stress in Alzheimer's disease. Neurosci Bull. 30:271–281. 2014. View Article : Google Scholar

6 

Ahmad W, Ijaz B, Shabbiri K, Ahmed F and Rehman S: Oxidative toxicity in diabetes and Alzheimer's disease: Mechanisms behind ROS/ RNS generation. J Biomed Sci. 24:762017. View Article : Google Scholar

7 

Angelova PR and Abramov AY: Role of mitochondrial ROS in the brain: From physiology to neurodegeneration. FEBS Lett. 592:692–702. 2018. View Article : Google Scholar

8 

Nesi G, Sestito S, Digiacomo M and Rapposelli S: Oxidative Stress, Mitochondrial Abnormalities and Proteins Deposition: Multitarget Approaches in Alzheimer's Disease. Curr Top Med Chem. 17:3062–3079. 2017.

9 

Hardy J: Alzheimer's disease: The amyloid cascade hypothesis: an update and reappraisal. J Alzheimers Dis. 9 (Suppl. 3):151–153. 2006. View Article : Google Scholar

10 

Kang MI, Kobayashi A, Wakabayashi N, Kim SG and Yamamoto M: Scaffolding of Keap1 to the actin cytoskeleton controls the function of Nrf2 as key regulator of cytoprotective phase 2 genes. Proc Natl Acad Sci USA. 101:2046–2051. 2004. View Article : Google Scholar

11 

Kaur SJ, McKeown SR and Rashid S: Mutant SOD1 mediated pathogenesis of Amyotrophic Lateral Sclerosis. Gene. 577:109–118. 2016. View Article : Google Scholar

12 

Kensler TW, Wakabayashi N and Biswal S: Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 47:89–116. 2007. View Article : Google Scholar

13 

Kärkkäinen V, Pomeshchik Y, Savchenko E, Dhungana H, Kurronen A, Lehtonen S, Naumenko N, Tavi P, Levonen AL, Yamamoto M, et al: Nrf2 regulates neurogenesis and protects neural progenitor cells against Aβ toxicity. Stem Cells. 32:1904–1916. 2014. View Article : Google Scholar

14 

Calkins MJ, Johnson DA, Townsend JA, Vargas MR, Dowell JA, Williamson TP, Kraft AD, Lee JM, Li J and Johnson JA: The Nrf2/ARE pathway as a potential therapeutic target in neurodegenerative disease. Antioxid Redox Signal. 11:497–508. 2009. View Article : Google Scholar

15 

Wang JB, Zhao HP, Zhao YL, Jin C, Liu DJ, Kong WJ, Fang F, Zhang L, Wang HJ and Xiao XH: Hepatotoxicity or hepatoprotection? Pattern recognition for the paradoxical effect of the Chinese herb Rheum palmatum L. in treating rat liver injury. PLoS One. 6:e244982011. View Article : Google Scholar

16 

Yang YC, Lim M-Y and Lee H-S: Emodin isolated from Cassia obtusifolia (Leguminosae) seed shows larvicidal activity against three mosquito species. J Agric Food Chem. 51:7629–7631. 2003. View Article : Google Scholar

17 

Lee MH, Kao L and Lin C-C: Comparison of the antioxidant and transmembrane permeative activities of the different Polygonum cuspidatum extracts in phospholipid-based microemulsions. J Agric Food Chem. 59:9135–9141. 2011. View Article : Google Scholar

18 

Dong X, Fu J, Yin X, Cao S, Li X, Lin L, Ni J and Emodin: A Review of its Pharmacology, Toxicity and Pharmacokinetics. Phytother Res. 30:1207–1218. 2016. View Article : Google Scholar

19 

Liu T, Jin H, Sun QR, Xu JH and Hu HT: Neuroprotective effects of emodin in rat cortical neurons against beta-amyloid-induced neurotoxicity. Brain Res. 1347:149–160. 2010. View Article : Google Scholar

20 

Lin J, Ma C and Lin HH: Emodin Alleviates Viral Myocarditis in BALB/c Mice and Underlying Mechanisms. Lat Am J Pharm. 38:1979–1984. 2019.

21 

Park SY, Jin ML, Ko MJ, Park G and Choi YW: Anti-neuroinflammatory Effect of Emodin in LPS-Stimulated Microglia: Involvement of AMPK/Nrf2 Activation. Neurochem Res. 41:2981–2992. 2016. View Article : Google Scholar

22 

Tian SL, Yang Y, Liu XL and Xu QB: Emodin Attenuates Bleomycin-Induced Pulmonary Fibrosis via Anti-Inflammatory and Anti-Oxidative Activities in Rats. Med Sci Monit. 24:1–10. 2018. View Article : Google Scholar

23 

Mizuno M, Kawamura H, Takei N and Nawa H: The anthraquinone derivative Emodin ameliorates neurobehavioral deficits of a rodent model for schizophrenia. J Neural Transm (Vienna). 115:521–530. 2008. View Article : Google Scholar

24 

Li Z, Chen X, Zhang Y, Liu X, Wang C, Teng L and Wang D: Protective roles of Amanita caesarea polysaccharides against Alzheimer's disease via Nrf2 pathway. Int J Biol Macromol. 121:29–37. 2019. View Article : Google Scholar

25 

Han Y, Nan S, Fan J, Chen Q and Zhang Y: Inonotus obliquus polysaccharides protect against Alzheimer's disease by regulating Nrf2 signaling and exerting antioxidative and antiapoptotic effects. Int J Biol Macromol. 131:769–778. 2019. View Article : Google Scholar

26 

Zhang Y, Wang J, Wang C, Li Z, Liu X, Zhang J, Lu J and Wang D: Pharmacological Basis for the Use of Evodiamine in Alzheimer's Disease: Antioxidation and Antiapoptosis. Int J Mol Sci. 19:192018.

27 

Sharifi AM, Eslami H, Larijani B and Davoodi J: Involvement of caspase-8, −9, and −3 in high glucose-induced apoptosis in PC12 cells. Neurosci Lett. 459:47–51. 2009. View Article : Google Scholar

28 

Berry BJ, Trewin AJ, Amitrano AM, Kim M and Wojtovich AP: Use the Protonmotive Force: Mitochondrial Uncoupling and Reactive Oxygen Species. J Mol Biol. 430:3873–3891. 2018. View Article : Google Scholar

29 

Tan S, Sagara Y, Liu Y, Maher P and Schubert D: The regulation of reactive oxygen species production during programmed cell death. J Cell Biol. 141:1423–1432. 1998. View Article : Google Scholar

30 

Grivennikova VG and Vinogradov AD: Generation of superoxide by the mitochondrial Complex I. Biochim Biophys Acta. 1757:553–561. 2006. View Article : Google Scholar

31 

Reed JC, Jurgensmeier JM and Matsuyama S: Bcl-2 family proteins and mitochondria. Biochim Biophys Acta. 1366:127–137. 1998. View Article : Google Scholar

32 

McMahon M, Itoh K, Yamamoto M, Chanas SA, Henderson CJ, McLellan LI, Wolf CR, Cavin C and Hayes JD: The Cap'n'Collar basic leucine zipper transcription factor Nrf2 (NF-E2 p45-related factor 2) controls both constitutive and inducible expression of intestinal detoxification and glutathione biosynthetic enzymes. Cancer Res. 61:3299–3307. 2001.

33 

Benedict C: Candidate mechanisms underlying the association between poor sleep and obesity. Endocrine Abstracts. 49:pp. S282017, simplehttps://doi.org/10.1530/endoabs.49.S28.1

34 

Choi ML and Gandhi S: Crucial role of protein oligomerization in the pathogenesis of Alzheimer's and Parkinson's diseases. FEBS J. 285:3631–3644. 2018. View Article : Google Scholar

35 

Ansari MA and Scheff SW: Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J Neuropathol Exp Neurol. 69:155–167. 2010. View Article : Google Scholar

36 

Selkoe DJ: Alzheimer's disease: Genes, proteins, and therapy. Physiol Rev. 81:741–766. 2001. View Article : Google Scholar

37 

Saxena G, Singh SP, Agrawal R and Nath C: Effect of donepezil and tacrine on oxidative stress in intracerebral streptozotocin-induced model of dementia in mice. Eur J Pharmacol. 581:283–289. 2008. View Article : Google Scholar

38 

Monisha BA, Kumar N and Tiku AB: Emodin and Its Role in Chronic Diseases. Adv Exp Med Biol. 928:47–73. 2016. View Article : Google Scholar

39 

Handattu SP, Monroe CE, Nayyar G, Palgunachari MN, Kadish I, van Groen T, Anantharamaiah GM and Garber DW: In vivo and in vitro effects of an apolipoprotein E mimetic peptide on amyloid-β pathology. J Alzheimers Dis. 36:335–347. 2013. View Article : Google Scholar

40 

Carrillo-Mora P, Luna R and Colín-Barenque L: Amyloid beta: Multiple mechanisms of toxicity and only some protective effects? Oxid Med Cell Longev. 2014:7953752014. View Article : Google Scholar

41 

Anandatheerthavarada HK, Biswas G, Robin M-A and Avadhani NG: Mitochondrial targeting and a novel transmembrane arrest of Alzheimer's amyloid precursor protein impairs mitochondrial function in neuronal cells. J Cell Biol. 161:41–54. 2003. View Article : Google Scholar

42 

Pluta R, Ułamek-Kozioł M and Czuczwar SJ: Neuroprotective and Neurological/Cognitive Enhancement Effects of Curcumin after Brain Ischemia Injury with Alzheimer's Disease Phenotype. Int J Mol Sci. 19:40022018. View Article : Google Scholar

43 

Viswanath V, Wu Y, Boonplueang R, Chen S, Stevenson FF, Yantiri F, Yang L, Beal MF and Andersen JK: Caspase-9 activation results in downstream caspase-8 activation and bid cleavage in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease. J Neurosci. 21:9519–9528. 2001. View Article : Google Scholar

44 

Cain K, Bratton SB, Langlais C, Walker G, Brown DG, Sun XM and Cohen GM: Apaf-1 oligomerizes into biologically active approximately 700-kDa and inactive approximately 1.4-MDa apoptosome complexes. J Biol Chem. 275:6067–6070. 2000. View Article : Google Scholar

45 

Luca M, Luca A and Calandra C: The Role of Oxidative Damage in the Pathogenesis and Progression of Alzheimer's Disease and Vascular Dementia. Oxid Med Cell Longev. 2015:5046782015. View Article : Google Scholar

46 

Casley CS, Canevari L, Land JM, Clark JB and Sharpe MA: Beta-amyloid inhibits integrated mitochondrial respiration and key enzyme activities. J Neurochem. 80:91–100. 2002. View Article : Google Scholar

47 

Gross A, McDonnell JM and Korsmeyer SJ: BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 13:1899–1911. 1999. View Article : Google Scholar

48 

Zhu W, Cowie A, Wasfy GW, Penn LZ, Leber B and Andrews DW: Bcl-2 mutants with restricted subcellular location reveal spatially distinct pathways for apoptosis in different cell types. EMBO J. 15:4130–4141. 1996. View Article : Google Scholar

49 

Hsu YT and Youle RJ: Nonionic detergents induce dimerization among members of the Bcl-2 family. J Biol Chem. 272:13829–13834. 1997. View Article : Google Scholar

50 

Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD and Yamamoto M: Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 13:76–86. 1999. View Article : Google Scholar

51 

Zhang Y, Unnikrishnan A, Deepa SS, Liu Y, Li Y, Ikeno Y, Sosnowska D, Van Remmen H and Richardson A: A new role for oxidative stress in aging: The accelerated aging phenotype in Sod1−/− mice is correlated to increased cellular senescence. Redox Biol. 11:30–37. 2017. View Article : Google Scholar

52 

Kansanen E, Kuosmanen SM, Leinonen H and Levonen AL: The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 1:45–49. 2013. View Article : Google Scholar

53 

Dinkova-Kostova AT and Abramov AY: The emerging role of Nrf2 in mitochondrial function. Free Radic Biol Med. 88B:B179–B188. 2015. View Article : Google Scholar

54 

Rojo AI, Pajares M, Rada P, Nuñez A, Nevado-Holgado AJ, Killik R, Van Leuven F, Ribe E, Lovestone S, Yamamoto M, et al: NRF2 deficiency replicates transcriptomic changes in Alzheimer's patients and worsens APP and TAU pathology. Redox Biol. 13:444–451. 2017. View Article : Google Scholar

55 

Hu D, Serrano F, Oury TD, Klann E and Hu D: Aging-dependent alterations in synaptic plasticity and memory in mice that overexpress extracellular superoxide dismutase. J Neurosci. 26:3933–3941. 2006. View Article : Google Scholar

56 

Pietrzak RH, Lim YY, Neumeister A, Ames D, Ellis KA, Harrington K, Lautenschlager NT, Restrepo C, Martins RN, Masters CL, et al Australian Imaging, Biomarkers, Lifestyle Research Group, : Amyloid-β, anxiety, and cognitive decline in preclinical Alzheimer disease: A multicenter, prospective cohort study. JAMA Psychiatry. 72:284–291. 2015. View Article : Google Scholar

57 

Delatour B, Guégan M, Volk A and Dhenain M: In vivo MRI and histological evaluation of brain atrophy in APP/PS1 transgenic mice. Neurobiol Aging. 27:835–847. 2006. View Article : Google Scholar

58 

Mehta MA: Morris Water Maze. Encyclopedia of Psychopharmacology. Stolerman IP: Springer; Berlin, Heidelberg: 2010, simplehttps://doi.org/10.1007/978-3-540-68706-1_1638

59 

Prut L and Belzung C: The open field as a paradigm to measure the effects of drugs on anxiety-like behaviors: A review. Eur J Pharmacol. 463:3–33. 2003. View Article : Google Scholar

60 

Kamat PK, Kalani A, Rai S, Swarnkar S, Tota S, Nath C and Tyagi N: Mechanism of Oxidative Stress and Synapse Dysfunction in the Pathogenesis of Alzheimer's Disease: Understanding the Therapeutics Strategies. Mol Neurobiol. 53:648–661. 2016. View Article : Google Scholar

61 

Leroy K, Ando K, Laporte V, Dedecker R, Suain V, Authelet M, Héraud C, Pierrot N, Yilmaz Z, Octave JN, et al: Lack of tau proteins rescues neuronal cell death and decreases amyloidogenic processing of APP in APP/PS1 mice. Am J Pathol. 181:1928–1940. 2012. View Article : Google Scholar

62 

Stamer K, Vogel R, Thies E, Mandelkow E and Mandelkow EM: Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 156:1051–1063. 2002. View Article : Google Scholar

63 

Smith MA, Hirai K, Hsiao K, Pappolla MA, Harris PL, Siedlak SL, Tabaton M and Perry G: Amyloid-beta deposition in Alzheimer transgenic mice is associated with oxidative stress. J Neurochem. 70:2212–2215. 1998. View Article : Google Scholar

64 

Wirths O, Multhaup G, Czech C, Feldmann N, Blanchard V, Tremp G, Beyreuther K, Pradier L and Bayer TA: Intraneuronal APP/A beta trafficking and plaque formation in beta-amyloid precursor protein and presenilin-1 transgenic mice. Brain Pathol. 12:275–286. 2002. View Article : Google Scholar

Related Articles

Journal Cover

February-2021
Volume 23 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li Z, Bi H, Jiang H, Song J, Meng Q, Zhang Y and Fei X: Neuroprotective effect of emodin against Alzheimer's disease via Nrf2 signaling in U251 cells and APP/PS1 mice Corrigendum in /10.3892/mmr.2023.12994. Mol Med Rep 23: 108, 2021
APA
Li, Z., Bi, H., Jiang, H., Song, J., Meng, Q., Zhang, Y., & Fei, X. (2021). Neuroprotective effect of emodin against Alzheimer's disease via Nrf2 signaling in U251 cells and APP/PS1 mice Corrigendum in /10.3892/mmr.2023.12994. Molecular Medicine Reports, 23, 108. https://doi.org/10.3892/mmr.2020.11747
MLA
Li, Z., Bi, H., Jiang, H., Song, J., Meng, Q., Zhang, Y., Fei, X."Neuroprotective effect of emodin against Alzheimer's disease via Nrf2 signaling in U251 cells and APP/PS1 mice Corrigendum in /10.3892/mmr.2023.12994". Molecular Medicine Reports 23.2 (2021): 108.
Chicago
Li, Z., Bi, H., Jiang, H., Song, J., Meng, Q., Zhang, Y., Fei, X."Neuroprotective effect of emodin against Alzheimer's disease via Nrf2 signaling in U251 cells and APP/PS1 mice Corrigendum in /10.3892/mmr.2023.12994". Molecular Medicine Reports 23, no. 2 (2021): 108. https://doi.org/10.3892/mmr.2020.11747