Oroxylum indicum (L.) extract protects human neuroblastoma SH‑SY5Y cells against β‑amyloid‑induced cell injury

  • Authors:
    • Nootchanat Mairuae
    • James R. Connor
    • Benjaporn Buranrat
    • Sang Y. Lee
  • View Affiliations

  • Published online on: June 21, 2019     https://doi.org/10.3892/mmr.2019.10411
  • Pages: 1933-1942
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It has been reported that amyloid β peptide, the major component of senile plaques, serves a critical role in the development and progression of Alzheimer's disease (AD) by generating reactive oxygen species (ROS), leading to oxidative stress. The aim of the present study was to investigate the protective effect of Oroxylum indicum (L.) extract against Aβ25‑35‑induced oxidative stress and cell injury using SH‑SY5Y cells as a model, and at exploring the underlying mechanisms. The results revealed that the exposure of cells to 20 µM Aβ25‑35 significantly increased cellular oxidative stress, as evidenced by the increased ROS levels. Aβ25‑35 treatment also increased caspase‑3/7 activity and lactate dehydrogenase (LDH) release, and caused viability loss. Oroxylum indicum treatment not only attenuated the generation of ROS and suppressed caspase‑3/7 activity but also reduced the neurotoxicity of Aβ25‑35 in a concentration‑dependent manner, as evidenced by the increased cell viability and decreased LDH release. Treatment with Oroxylum indicum also increased superoxide dismutase (SOD) and catalase (CAT) activity, increased the phosphorylation of Akt and cAMP‑responsive element binding protein (CREB), and contributed to the upregulation of Bcl‑2 protein. In combination, these results indicated that Oroxylum indicum extract could protect SH‑SY5Y cells against Aβ25‑35‑induced cell injury, at least partly, by inhibiting oxidative stress, increasing SOD and CAT activity, attenuating caspase 3/7 activity and promoting the cell survival pathway, Akt/CREB/Bcl‑2. The approach used in the present study may also be useful for preventing the neurotoxicity induced by Aβ in AD and related neurodegenerative diseases. Further studies investigating the activity of Oroxylum indicum extract in vivo are now required.

Introduction

Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder that mostly affects the elderly. Prevalence studies have revealed that >35 million people are suffering from AD worldwide, and this number is predicted to reach >100 million by the year 2050, if new preventive or neuroprotective therapies do not emerge (1). The neuropathology of AD is characterized by extracellular deposition of amyloid β (Aβ) plaques, and intracellular neurofibrillary tangles and loss of neurons in the brain (2). Although the mechanisms of neuronal cell death in AD still remain unknown, the deposition of Aβ has been reported to be neurotoxic both in vitro and in vivo, involving reactive oxygen species (ROS) generation, inflammation, and an increase in intracellular Ca2+ (38). A large number of studies have confirmed that the excessive production of Aβ itself leads to Aβ-induced free radical generation and elevated oxidative stress, leading to cell death (9). Thus, one promising preventive or therapeutic intervention in AD may be to attenuate or suppress oxidative stress-dependent, Aβ-mediated cytotoxicity.

Plants are a major component of diets that possess neuroprotective effects, including antioxidant and anti-inflammatory effects, and can improve memory and cognitive functions (1013). The most important advantages of the medicinal use of plants are their minimal side effects and their relatively low cost, as compared to synthetic medicines. According to the World Health Organization, ~80% of the world's population currently uses medicinal plants for their primary healthcare (14), and the current trend is to conduct investigations into plant-based medicines. Therefore, searching for plants that can attenuate oxidative stress might be a useful strategy for preventing and/or treating Aβ-induced neurotoxicity. Oroxylum indicum (L.), also known as ‘broken bones plant’, ‘Indian trumpet flower’, ‘Shyonaka’ and ‘Midnight horror’, belongs to the Bignoniaceae family, which is widely distributed in tropical countries, such as India, Taiwan, Cambodia, Laos, Myanmar, Indonesia, Malaysia, Vietnam, Nepal, China, the Philippines and Thailand (15). Oroxylum indicum has been used in traditional herbal medicine in Asian countries for the treatment of various diseases over several centuries (16). Studies have indicated that almost all parts of the plant possesses medicinal properties (15), mainly antioxidant, anti-inflammatory, anticancer and immunomodulatory properties. Other effects, such as antibacterial and gastro-protective, have also been reported. The principal active components of this plant are the flavonoids chrysin, oroxylene A and baicalein (15,17). Other secondary metabolites, such as triterpene, carboxylic acid, ursolic acid, glycosides, tannins, alkaloids and terpenoids, have also been identified. Although many medicinal properties of Oroxylum indicum have been demonstrated, the effects of Oroxylum indicum extract on Aβ-induced oxidative stress have not, to our knowledge, been investigated. The aim of the present study was to investigate the protective effect of Oroxylum indicum (L.) fruit pod extract against Aβ25-35-induced oxidative stress and injury in SH-SY5Y cells. The mechanisms underlying its neuroprotection were also investigated. Phenolic compounds and flavonoids have been shown to be very good antioxidants (18), thus their concentration in Oroxylum indicum extract was also determined. The fruit pod of Oroxylum indicum was selected for the present study, since this part is more readily edible than other parts (17). Aβ25–35 was chosen because this fragment is an active toxic fragment of Aβ 1–42 peptides (19), and it has been reported that Aβ25–35 and Aβ 1–42 have similar effects in inducing neuronal cell death and neuritic atrophy (20,21). SH-SY5Y cells were selected as a model, since they are commonly used in AD research and differentiated SH-SY5Y cells have displayed properties similar to those of mature neurons (22).

Materials and methods

Chemicals and antibodies

SH-SY5Y cell line was purchased from the American Type Culture Collection (Manassas, VA, USA; catalog number CRL-2266). Cell culture reagents, including penicillin/streptomycin were from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). DMEM/F12 medium was from Thermo Fisher Scientific, Inc. and fetal bovine serum (FBS) from Gemini Bio-Products (West Sacramento, CA, USA). Non-essential amino acids and all-trans retinoic acid (RA) were from Merck KGaA (Darmstadt, Germany). The ROS detection kit was also obtained from Merck KGaA. The catalase activity kit was from Biovision Inc. (Milpitas, CA, USA), and the superoxide dismutase (SOD) activity kit from Cayman Chemical Company (Ann Arbor, MI, USA). The lactate dehydrogenase (LDH) kit, together with Aβ25–35 and 3-(3,4-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) were from Merck KGaA. The LDH assay kit was from Thermo Fisher Scientific, Inc., and the caspase 3/7 activity kit was from Promega Corporation (Madison, WI, USA). Antibodies against total- and phosphor(p)-Akt, and p-cAMP-responsive element binding protein (CREB) were from Cell Signaling Technology, Inc., (Danvers, MA, USA). Secondary anti-rabbit or anti-mouse antibodies and ECL detection kits were from GE Healthcare (Chicago, IL, USA). RIPA buffer, protease and phosphatase inhibitor cocktail, calcein-AM and quercetin were obtained from Merck KGaA. The BCA protein assay was from Thermo Fisher Scientific, Inc.

Plant material and extraction

Fruits of Oroxylum indicum were collected from Maha Sarakham Province, Thailand, in July 2016. Species identification was performed by members of the Applied Thai Traditional Medicine Department, Faculty of Medicine, Mahasarakham University. A specimen was deposited at the herbarium at the Faculty of Science, Mahasarakham University (specimen no. MSUT_7234). An ethanolic extract of Oroxylum indicum was prepared by drying the fruits, then weighing and chopping them, and macerating them in 95% (v/v) ethanol for 7 days at room temperature (RT). The extract was then filtered, concentrated using a rotary evaporator and lyophilized. The % yield of extract was 12.89% per dry weight of Oroxylum indicum fruits.

Determination of total flavonoid content

The total flavonoid content of Oroxylum indicum crude extract was determined by the aluminum chloride colorimetric method. In brief, 100 µl of 1 mg/ml Oroxylum indicum crude extract was mixed with 0.9 ml flavonoid mixture (10% aluminum hydroxide, 1 M potassium acetate; dilution, 0.1, 0.1 and 4.3 ml). The mixture was incubated for 30 min at RT in the dark, and the absorbance intensity was measured at 450 nm. The total flavonoid content was calculated from a calibration curve and the result was expressed as mg rutin equivalent per g dry weight.

Determination of total phenol content

The total phenolic content of the Oroxylum indicum extract was determined using the Folin-Ciocalteu method. In brief, 100 µl of 1 mg/ml Oroxylum indicum crude extract was mixed thoroughly with 0.45 ml Folin-Ciocalteu reagent for 5 min, followed by the addition of 0.45 ml of 60 g/l sodium bicarbonate. The mixture was kept in the dark for a further 1 h at RT, and the absorbance intensity was measured at 650 nm. The total phenolic content was calculated from the calibration curve, and the results were expressed as mg of gallic acid equivalent per g dry weight.

Cell culture

Human neuroblastoma SH-SY5Y cells were maintained in DMEM/F12 medium supplemented with 10% FBS, 1% penicillin-streptomycin and 1% non-essential amino acids at 37°C in a humidified atmosphere containing 5% CO2. The culture medium was changed every 3 days. Cells were plated at an appropriate density according to each experiment. Cells were differentiated with 10 µM all-trans retinoic acid over 6 days prior to treatments. At the beginning of each experiment, the culture medium in each well was completely removed and replaced with fresh medium containing Aβ with or without Oroxylum extract.

Preparation of Aβ25–35 stock solution

Aβ25–35 peptide was dissolved in deionized distilled water as a 1 mM stock solution and incubated at 37°C for 3 days. The solution was aliquoted into 1-ml tubes, kept at −20°C and thawed for subsequent use.

Cell viability assay

The in vitro cytotoxicity of Aβ25–35 and Oroxylum indicum were determined using the MTT assay. SH-SY5Y cells were plated in 96-well plates at a density of 1×104 cells/well and cultured as described above. Cells were treated with various concentrations of Aβ25–35 (10–30 µM) and Oroxylum indicum (0–100 µg/ml) in 1% FBS for 24 h. To determine whether Oroxylum indicum protects against Aβ-induced neurotoxicity, cells were treated with Aβ25–35 with or without of Oroxylum indicum extract in 1% FBS for 24 h. Following 24 h of treatment, the medium was removed and replaced with MTT reagent at a final concentration of 0.5 mg/ml. Cells were then incubated for 4 h at 37°C in 5% CO2 in an incubator. Following incubation, MTT reagent was aspirated and 100 µl dimethyl sulfoxide was added to dissolve the insoluble purple formazan product. Absorbance was determined at 570 nm using a Synergy-4 plate reader (BioTek Instruments, Inc Winooski, VT, USA). Results were expressed as a percentage of the control.

Intracellular ROS assay

Cells were seeded at a density of 1×104 cells/well in 96-well plates and cultured as described above. After 24 h, intracellular ROS levels were measured using the fluorescent probe 2′,7′-dichlorofluorescein, as previously described (13). Data were expressed as the percentage of ROS relative to untreated controls.

Analysis of cell injury by the LDH assay

Cells were plated at a density 1×104 cells/well in 96-well plates. Cells were then cultured and treated as described above. Following 24 h of treatment, Aβ-induced cell injury was measured by determining how much of the intracellular enzyme LDH had been released into the culture medium. Culture medium (100 µl) was collected from each well and transferred to a new 96 well plate, and 100 µl reaction mixture was added to each well and incubated for 30 min at 37°C. Absorbance was measured at 492 nm using a microplate reader. The quantity of LDH released was then expressed as a percentage of the untreated control.

Determination of catalase (CAT) activity assay

Cells were plated at a density 1×105 cells/well in 6-well plates. Overnight cultured cells were treated as described above. Following 24 h of treatment, cells were harvested with a rubber policeman and collected by centrifugation (2,000 × g for 10 min at 4°C). The cell pellets were homogenized in cold assay buffer and centrifuged at 10,000 × g for 15 min at 4°C. The supernatant was also collected for the assay. Catalase activity was determined using a commercially available assay kit (Biovision Inc.), according to the manufacturer's instructions.

SOD activity assay

Cells (1×105 cells/well) were plated in 6-well plates and were then cultured and treated as described above. Following 24 h of treatment, cells were harvested as described for the CAT activity assay. Cell pellets were then homogenized in 20 mM cold HEPES buffer, and centrifuged at 1,500 × g for 5 min at 4°C. The supernatant was also collected for the assay. Superoxide dismutase activity was measured using an assay kit from Cayman Chemical, according to the manufacturer's instructions. Results were expressed as a percentage of the untreated control.

Western blotting

Cells (1×105 cells/well) were plated in 6-well plates, and then cultured and treated as described above. Following treatment, cells were collected and total protein concentration was determined using a BCA kit. Equal amounts of proteins were separated by 4–20% SDS-polyacrylamide gel and transferred onto nitrocellulose membranes. The membranes were then incubated with primary antibodies against Bcl-2 (1:1,000), p-Akt, total Akt (1:1,000), p-CREB (1:1,000) and actin (1:5,000) overnight at 4°C. The membranes were then washed with TBST (Tris-buffered saline, 0.1% Tween 20), and probed with secondary antibody conjugated with HRP for 1 h at RT. Protein bands were detected using an enhanced chemiluminescence detection kit, and results were expressed as a fold change of the untreated control.

Detection of caspase-3/7 activity in cell culture

Caspase-3/7 activity was measured using Caspase-Glo® 3/7 kits from Promega Corporation, according to the manufacturer's instructions. Briefly, the caspase-GloR 3/7 buffer and lyophilized caspase-GloR 3/7 substrate were equilibrated at RT. The contents of the caspase-GloR 3/7 buffer were transferred into the bottle containing caspase-GloR3/7 substrate. Equal volumes of reaction mixture were added to samples and incubated for 30 min-2 h prior to the luminescence measurement. Results were expressed as a percentage of the untreated control.

Statistical analysis

All data are expressed as the mean ± SEM from at least three independent experiments performed in triplicate. Multiple comparisons of data were evaluated by one-way ANOVA followed by Bonferroni post-hoc test. A P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of Aβ25–35 on the viability of differentiated and undifferentiated SH-SY5Y cells

Since the SH-SY5Y cell line shares only a few properties with mature neurons (23), it is important to differentiate these cells with retinoic acid, so that they are comparable to in vivo models. Undifferentiated SH-SY5Y cells have been used as a model of cytotoxicity in several studies (2426), but the comparative cytotoxic effects of Aβ on cell survival of RA-differentiated and undifferentiated SH-SY5Y cells has not yet been reported. Therefore, the purpose of this study was to compare the in vitro cytotoxicity of Aβ25–35 between differentiated and undifferentiated SH-SY5Y cells. Both differentiated and undifferentiated cells were treated with various concentrations of Aβ25–35 (0–30 µM). As shown in Fig. 1A, Aβ25–35 treatments were toxic to both cell groups, beginning at 10 µM for undifferentiated cells and 20 µM for differentiated cells. Although undifferentiated SH-SY5Y cells were more susceptible to Aβ25–35 than differentiated cells, the differentiated cells possess more neuron-like properties, including neurite outgrowth and morphological changes of neurons in the brain. Therefore, RA-differentiated SH-SY5Y cells were selected for subsequent assays, and Aβ25–35 was used at a concentration of 20 µM.

Effects of Oroxylum indicum on the viability of SH-SY5Y cells

To determine whether Oroxylum indicum has any effect on the viability of SH-SY5Y cells, the cells were treated with various concentrations of the extract (0–100 µg/ml). The results indicated that Oroxylum indicum extract at concentrations of 25 and 50 µg/ml increased cell viability (139.45±7.89 and 130.61±5.83% of control value, respectively) and that concentrations of up to 100 µg/ml were non-toxic to SH-SY5Y cells (Fig. 1B). Therefore, the highest non-toxic concentrations of Oroxylum indicum (50 and 100 µg/ml) were used in subsequent assays.

Oroxylum indicum protected SH-SY5Y cells against Aβ25-35-induced cytotoxicity

To determine the effect of Oroxylum indicum on Aβ25-35-induced cytotoxicity, SH-SY5Y cells were challenged with 20 µM Aβ25–35 in the presence or absence of 50 and 100 µg/ml Oroxylum indicum extract for 24 h. As shown in Fig. 2A, treatment of SH-SY5Y cells with 20 µM Aβ25–35 for 24 h induced cytotoxicity, as demonstrated by a cell viability reduction to 76.83±0.67%, when compared with the control group. When the cells were treated with Oroxylum indicum extract at concentrations of 50 and 100 µg/ml, cell viability was restored to 94.14±2.79 and 98.35±3.74%, respectively, indicating a concentration-dependent cytoprotective effect.

To further confirm the cytoprotective effect of Oroxylum indicum, LDH, another indicator of cell toxicity, was also examined. The results were similar to those determined by the MTT assay. The exposure of SH-SY5Y cells to 20 µM Aβ25–35 resulted in a 1.45-fold increase in LDH release in the medium, when compared to that in the control group. Treatment of cells with 50 and 100 µg/ml Oroxylum indicum extract reduced Aβ25-35-induced LDH release in a concentration-dependent manner (Fig. 2B).

Oroxylum indicum inhibited Aβ25-35-induced intracellular accumulation of ROS

ROS play a critical role in Aβ-dependent cell death. Therefore, in the present study, the effect of Oroxylum indicum on Aβ25-35-induced intracellular ROS production was examined. SH-SY5Y cells exposed to Aβ25–35 for 24 h exhibited elevated ROS levels (152.4±3.53%, as compared to the control group) (Fig. 3A). When the cells were treated with Oroxylum indicum extract at concentrations of 50 and 100 µg/ml, there was a significant and concentration-dependent inhibition of Aβ25-35-induced intracellular ROS levels (89.17±2.46 and 64.25±6.21%, respectively).

Oroxylum indicum increased the activity of anti-oxidative enzymes challenged by Aβ25-35

To determine whether the cytoprotective effect of Oroxylum indicum is associated with the activity of anti-oxidative enzymes, SOD and CAT activity was determined. Following exposure to Aβ25–35 for 24 h, SOD activity was significantly decreased to 76.48±0.6% of the control value. Treatment of cells with 50 and 100 µg/ml Oroxylum indicum extract for 24 h restored SOD activity to 87.30±0.81 and 100.76±1.19%, respectively (Fig. 3B). Following exposure to Aβ25-35, CAT activity was significantly increased to 122.45±0.029% of the control value. CAT activity following treatment with Oroxylum indicum extract was significantly increased beyond that of the Aβ treatment and control groups (144.15 and 148.4%, respectively) (Fig. 3C).

Oroxylum indicum reduced Aβ25-35-induced caspase-3/7 activity

It has been reported that Aβ-induced neuronal cell death through the activation of the caspase pathway (2731). To study the protective mechanism of Oroxylum indicum, caspase-3/7 activity was measured. Following Aβ25–35 treatment, caspase-3/7 activity significantly increased to levels that were 1.4-fold higher than that of the control group. However, the induction of caspase-3/7 activity was blocked in the presence of Oroxylum indicum (Fig. 3D)

Oroxylum indicum enhanced the phosphorylation of Akt

The activation of Akt has been associated with the inhibition of the apoptotic cleavage of caspases, as well as the promotion of neuronal survival (3235). We therefore hypothesized that Oroxylum indicum can modulate the signaling of Akt. The results showed that treatment with Aβ25–35 for 15 min significantly decreased p-Akt, as compared to the untreated control (Fig. 4A). Oroxylum indicum treatments significantly increased the phosphorylation of Akt, as compared to Aβ25–35 treatment.

Oroxylum indicum enhanced the phosphorylation of CREB

It has been reported that one element of the downstream signaling of Akt is CREB the transcription factor that regulates neuronal survival (35). We therefore examined whether CREB phosphorylation increased in SH-SY5Y cells following treatment with Aβ25–35 in the presence of Oroxylum indicum. When cells were treated with Aβ25–35 for 1 h, CREB phosphorylation was significantly decreased, when compared with the untreated control. The phosphorylation of CREB was significantly increased following treatment with Oroxylum indicum, when compared to both the Aβ25–35 treatment and control groups (Fig. 4B).

Oroxylum indicum enhanced Bcl-2 expression

The transcription factor CREB has been identified as a positive regulator of Bcl-2 (an apoptosis suppressor gene) expression (36,37). Therefore, these results demonstrated that Aβ25–35 ameliorated Bcl-2 expression, as compared with the control group. Treatment with Oroxylum indicum extract for 24 h caused a significant increase in Bcl-2 expression, as compared to the Aβ25–35 group (Fig. 4C).

Total phenolic and flavonoid content of extracts from fruit pot of Oroxylum indicum

The Oroxylum indicum fruit extract was standardized using colorimetric methods to quantify the total phenolic content using gallic acid as a standard, and the total flavonoid content using rutin as a standard. The values obtained were 10.50±0.68 and 17.08±0.85 mg/g of dried extract, respectively (Table I).

Table I.

Total phenolic and flavonoid content of Oroxylum indicum fruit extract.

Table I.

Total phenolic and flavonoid content of Oroxylum indicum fruit extract.

Total phenolic/flavonoid contentConcentrations (mg/g)
Total phenolic contenta30.50±0.68
Total flavonoid contentb17.08±0.85

a mg gallic acid equivalent/g dry weight.

b mg rutin equivalent/g dry weight. Values are presented as the mean of three biological replicates.

Discussion

Accumulation of amyloid plaques in the brains of AD patients has been reported to induce cytotoxicity mediated through the generation of ROS and elevated oxidative stress (9). Therefore, the attenuation or suppression of this oxidative stress-dependent, Aβ-mediated cytotoxicity may be a promising strategy for preventive or therapeutic intervention in AD. Recently, natural antioxidants from medicinal and edible plants have attracted considerable attention as promising agents for reducing the risk of oxidative stress-induced neurological diseases. In the present study, RA-differentiated SH-SY5Y cells were selected as a model, since they displayed properties similar to those of mature neurons. It was demonstrated that Aβ25-35-treated cells exhibited increased ROS production, which was consistent with previous reports (24,38,39). Treatment with Oroxylum indicum extract inhibited Aβ-induced ROS production in a concentration-dependent manner, indicating an antioxidant effect of Oroxylum indicum. Phenolic and flavonoid compounds in plants are reported to be very good antioxidants. Phenolics are effective hydrogen donors, while flavonoids act as scavengers of various oxidizing species, such as hydroxyl radicals, peroxy radicals and the superoxide anion (40). Therefore, it was reasonable to determine the quantities of these phytochemical classes in Oroxylum indicum. In the present study, it was demonstrated that the total phenolic and flavonoid content of Oroxylum indicum was 10.50±0.68 and 17.08±0.85 mg/g, respectively. Although Oroxylum indicum extract contained only low levels of phenolics and flavonoids, a marked decrease in ROS production was observed following treatment with the extract, suggesting that there is another antioxidative defense mechanism that can eliminate ROS. Under normal physiological conditions, ROS production is balanced by endogenous cellular antioxidant systems, including the cooperative action of SOD and CAT (41). SOD is the first line of defense against free radicals, its ROS-metabolizing activity occurring due to catalytic dismutation of the superoxide anion radical (O2•−) into O2 and H2O2. H2O2 is then converted into O2 and H2O by CAT, another major primary antioxidant defense component (41). It was demonstrated herein that treatment with Aβ25–35 decreases SOD activity, which was consistent with a previous report (42). A Aβ25–35 treatment-induced SOD activity reduction in cultured cells is possibly due to it metabolites or a direct toxic effect of Aβ25-35; however, unknown factors other than severe cell damage may also cause this effect. Of note, an increase in CAT activity was observed following exposure of cells to Aβ25-35, which might have been either a direct induction or a compensatory mechanism against Aβ insult. When cells are treated with Oroxylum indicum, SOD and CAT activity is increased, suggesting that Oroxylum indicum may stimulate cells to increase SOD and CAT expression, leading to an increase in their enzymatic activity in order to cope with Aβ-induced ROS production or may result from it metabolites. Therefore, the mechanism(s) through which Oroxylum indicum attenuates ROS production may be due to its phenolic and flavonoid content and/or its ability to increase SOD and CAT enzyme activity.

It has been reported that Aβ-induced neurotoxicity is related to ROS generation and caspase activation (2731), so we next investigated the effects of Oroxylum indicum on the activation of caspases-3/7, which are known to be effector caspases. The protective effect of Oroxylum indicum against Aβ-induced cytotoxicity was also determined. Treatment with Oroxylum indicum not only attenuated Aβ-induced caspase-3/7 activity, but also protected SH-SY5Y cells against Aβ-induced cytotoxicity, as determined by the MTT assay. The MTT assay was selected, since it has repeatedly been shown to be a very sensitive indicator of Aβ-induced cell death (43). The protective effect of Oroxylum indicum was further confirmed by LDH assay, another indicator of cell toxicity. The results reported above demonstrated that treatment with Oroxylum indicum reduced Aβ-induced LDH release. This finding indicated that Oroxylum indicum extract can protect SH-SY5Y cells against Aβ-induced cell injury.

The PI3K/Akt pathway is a key signal transduction pathway that mediates cell growth and promotes cell survival (44). The activation of Akt can phosphorylate CREB (45). Phosphorylated CREB is then translocated to the nucleus, where it upregulates the anti-apoptotic protein Bcl-2 (45). As Akt is an upstream signal that regulates p-CREB, and the phosphorylation process is rapid during protein translation, which occurs several hours following treatment, p-Akt and p-CREB were detected at 15 min and 1 h, respectively, and Bcl-2 at 24 h following treatment. Exposure of SH-SY5Y cells to Aβ decreased p-Akt. This finding was consistent with a previous report, which showed that intraneuronal Aβ accumulation leads to a decrease in the levels of phosphor-Akt (46). Exposure of SH-SY5Y cells to Aβ also decreased CREB phosphorylation, as well as the protein expression of Bcl-2. However, treatment with Oroxylum indicum increased the phosphorylation of Akt and CREB, as well as the expression of Bcl-2 protein. These results suggested that the activation of Akt/CREB/Bcl-2 pathway also plays a critical role in Oroxylum indicum-induced neuronal protective effects against Aβ insult.

To the best of our knowledge, there is no published study regarding the effects of Oroxylum indicum on Aβ exposure in neuronal cells. The present study was the first to show that Oroxylum indicum extract protects SH-SY5Y cells against Aβ25-35-induced cell injury. Although a cell line was used in this study, several studies have demonstrated the effects of Oroxylum indicum extract in vivo, including its protective effects against paracetamol-induced liver damage in experimental rats and cisplatin-induced renal injury in Wistar male albino rats, its hepatoprotective activity against CCl4-induced liver damage in rats, as well as its anti-central nervous system-depressant activity in an animal model (4751). We hope that this finding will encourage further investigations into the activity of Oroxylum indicum in AD and other neurodegenerative diseases.

In conclusion, the results of the present study suggested that Oroxylum indicum extract protects SH-SY5Y cells against Aβ25-35-induced injury, at least partly, by inhibiting oxidative stress, increasing SOD and CAT activity, attenuating caspase 3/7 activity and promoting cell survival pathways, such as Akt/CREB/Bcl-2 (Fig. 5). The present data suggested that Oroxylum indicum could be useful in the prevention of Aβ-induced neurotoxicty in AD and related neurodegenerative diseases. Further studies into the activity of Oroxylum indicum extract in vivo are now required.

Figure 5.

Hypothetical scheme illustrating signaling pathways through which Oroxylum indicum protects cells against Aβ-induced cell injury. It has been reported that Aβ causes ROS accumulation (52–53). ROS accumulation can induce the release of cytochrome c through several mechanisms. One mechanism is associated with the Bcl-2 family of proteins (Bcl-2, Bcl-Xl, Bax and Bid) (54). Once cytochrome c is released, it can bind to caspase-9 to form a complex, which subsequently activates caspases-3/7 (and other caspases) and eventually causes cell death (54). Oroxylum indicum also increases SOD and CAT activity, decreases ROS production and decreases caspases-3/7 activity. Increases in SOD and CAT activity might contribute to inhibition of ROS production. If ROS production decreases, the activation of caspases-3/7 may decrease. It has been reported that the Akt/CREB/Bcl-2 pathway is important in promoting cell survival (45). Oroxylum indicum also increases the phosphorylation of Akt and CREB, and increases expression of the anti-apoptotic protein Bcl-2. Increasing phosphorylation of Akt might contribute to an increase in the phosphorylation of CREB, resulting in an increase in Bcl-2. Therefore, activation of the Akt/CREB/Bcl-2 pathway might also play a critical role in Oroxylum indicum-induced neuronal protective effects against Aβ insult. Information in red front represents finding from the current study. Aβ, Amyloid-β; ROS, reactive oxygen species; SOD, superoxide dismutase; CAT, catalase; CREB, cAMP-responsive element binding protein.

Acknowledgements

Not applicable.

Funding

This study was financially supported by a grant from Mahasarakham University Faculty of Medicine and George M. Leader Family Funds.

Availability of data and materials

All the data generated and analyzed in the present study are available from the corresponding author on reasonable request.

Authors' contributions

NM was responsible for the conception and design of the study, as well as the acquisition of data and drafting of the manuscript. NM and BB performed the experiments. NM, JRC and SYL were responsible for data analysis and interpretation, and revising the manuscript. The final version of the manuscript has been read and approved by all authors.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Querfurth HW and LaFerla FM: Alzheimer's disease. N Engl J Med. 362:329–344. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Paulson JB, Ramsden M, Forster C, Sherman MA, McGowan E and Ashe KH: Amyloid plaque and neurofibrillary tangle pathology in a regulatable mouse model of Alzheimer's disease. Am J Pathol. 173:762–772. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Butterfield DA, Drake J, Pocernich C and Castegna A: Evidence of oxidative damage in Alzheimer's disease brain: Central role for amyloid beta-peptide. Trends Mol Med. 7:548–554. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Butterfield DA, Griffin S, Munch G and Pasinetti GM: Amyloid beta-peptide and amyloid pathology are central to the oxidative stress and inflammatory cascades under which Alzheimer's disease brain exists. J Alzheimers Dis. 4:193–201. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Butterfield DA, Swomley AM and Sultana R: Amyloid beta-peptide (1–42)-induced oxidative stress in Alzheimer disease: Importance in disease pathogenesis and progression. Antioxid Redox Signal. 19:823–835. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Bharadwaj PR, Dubey AK, Masters CL, Martins RN and Macreadie IG: Abeta aggregation and possible implications in Alzheimer's disease pathogenesis. J Cell Mol Med. 13:412–421. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Gray CW and Patel AJ: Neurodegeneration mediated by glutamate and beta-amyloid peptide: A comparison and possible interaction. Brain research. 691:169–179. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Ueda K, Shinohara S, Yagami T, Asakura K and Kawasaki K: Amyloid beta protein potentiates Ca2+ influx through L-type voltage-sensitive Ca2+ channels: A possible involvement of free radicals. J Neurochem. 68:265–271. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Muthaiyah B, Essa MM, Chauhan V and Chauhan A: Protective effects of walnut extract against amyloid beta peptide-induced cell death and oxidative stress in PC12 cells. Neurochem Res. 36:2096–2103. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Mohd Sairazi NS, Sirajudeen KN, Asari MA, Muzaimi M, Mummedy S and Sulaiman SA: Kainic acid-induced excitotoxicity experimental model: protective merits of natural products and plant extracts. Evid Based Complement Alternat Med. 2015:9726232015. View Article : Google Scholar : PubMed/NCBI

11 

Solanki I, Parihar P, Mansuri ML and Parihar MS: Flavonoid-based therapies in the early management of neurodegenerative diseases. Adv Nutr. 6:64–72. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Venkatesan R, Ji E and Kim SY: Phytochemicals that regulate neurodegenerative disease by targeting neurotrophins: A comprehensive review. Biomed Res Int. 2015:8140682015. View Article : Google Scholar : PubMed/NCBI

13 

Nootchanat MPC, Chalisa LC and Walaiporn T: Okra (Abelmoschus esculentus Linn) inhibits lipopolysaccharide-induced inflammatory mediators in BV2 microglial cells. Tropical J Pharmaceutical Res. 16:1285–1295. 2017. View Article : Google Scholar

14 

World Health Organization: General Guidelines for Methodologies on Research and Evaluation of Traditional Medicine. WHO; Geneva, Switzerland: 2000

15 

Lawania RD, Mishra A and Gupta R: Oroxylum indicum: A Review. Pharm J. 2:304–310. 2010.

16 

Narisa K, Wilkinson JM and Cavanagh H: Cytotoxic effect of four thai edible plants on mammalian cell proliferation. Thai Pharm Health Sci J. 1:189–195. 2006.

17 

Jiwajinda S, Santisopasri V, Murakami A, Kim OK, Kim HW and Ohigashi H: Suppressive effects of edible thai plants on superoxide and nitric oxide generation. Asian Pac J Cancer Prev. 3:215–223. 2002.PubMed/NCBI

18 

Diaz P, Jeong SC, Lee S, Khoo C and Koyyalamudi SR: Antioxidant and anti-inflammatory activities of selected medicinal plants and fungi containing phenolic and flavonoid compounds. Chin Med. 7:262012. View Article : Google Scholar : PubMed/NCBI

19 

Yan SD, Fu J, Soto C, Chen X, Zhu H, Al-Mohanna F, Collison K, Zhu A, Stern E, Saido T, et al: An intracellular protein that binds amyloid-beta peptide and mediates neurotoxicity in Alzheimer's disease. Nature. 389:689–695. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Kaminsky YG, Tikhonova LA and Kosenko EA: Critical analysis of Alzheimer's amyloid-beta toxicity to mitochondria. Front Biosci (Landmark Ed). 20:173–197. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Hughes E, Burke RM and Doig AJ: Inhibition of toxicity in the beta-amyloid peptide fragment beta-(25–35) using N-methylated derivatives: A general strategy to prevent amyloid formation. J Biol Chem. 275:25109–25115. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Liu YQ, Jia MQ, Xie ZH, Liu XF, Hui Y and Zheng XL: Arrestins contribute to amyloid beta-induced cell death via modulation of autophagy and the alpha7nAch receptor in SH-SY5Y cells. Sci Rep. 7:34462017. View Article : Google Scholar : PubMed/NCBI

23 

Shipley MM, Mangold CA and Szpara ML: Differentiation of the SH-SY5Y human neuroblastoma cell line. J Vis Exp. 531932016.PubMed/NCBI

24 

Wang Y, Miao Y, Mir AZ, Cheng L, Wang L, Zhao L, Cui Q, Zhao W and Wang H: Inhibition of beta-amyloid-induced neurotoxicity by pinocembrin through Nrf2/HO-1 pathway in SH-SY5Y cells. J Neurol Sci. 368:223–230. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Liu XY, Wang LX, Chen Z and Liu LB: Liraglutide prevents beta-amyloid-induced neurotoxicity in SH-SY5Y cells via a PI3K-dependent signaling pathway. Neurol Res. 38:313–319. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Sarkar B, Dhiman M, Mittal S and Mantha AK: Curcumin revitalizes Amyloid beta (25–35)-induced and organophosphate pesticides pestered neurotoxicity in SH-SY5Y and IMR-32 cells via activation of APE1 and Nrf2. Metab Brain Dis. 32:2045–2061. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Deshpande A, Mina E, Glabe C and Busciglio J: Different conformations of amyloid beta induce neurotoxicity by distinct mechanisms in human cortical neurons. J Neurosci. 26:6011–6018. 2006. View Article : Google Scholar : PubMed/NCBI

28 

He Y, Cui J, Lee JC, Ding S, Chalimoniuk M, Simonyi A, Sun AY, Gu Z, Weisman GA, Wood WG and Sun GY: Prolonged exposure of cortical neurons to oligomeric amyloid-β impairs NMDA receptor function via NADPH oxidase-mediated ROS production: Protective effect of green tea (−)-epigallocatechin-3-gallate. ASN Neuro. 3:e000502011.PubMed/NCBI

29 

Sponne I, Fifre A, Koziel V, Oster T, Olivier JL and Pillot T: Membrane cholesterol interferes with neuronal apoptosis induced by soluble oligomers but not fibrils of amyloid-beta peptide. FASEB J. 18:836–838. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Shelat PB, Chalimoniuk M, Wang JH, Strosznajder JB, Lee JC, Sun AY, Simonyi A and Sun GY: Amyloid beta peptide and NMDA induce ROS from NADPH oxidase and AA release from cytosolic phospholipase A2 in cortical neurons. J Neurochem. 106:45–55. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Pate KM, Rogers M, Reed JW, van der Munnik N, Vance SZ and Moss MA: Anthoxanthin polyphenols Attenuate Aβ oligomer-induced neuronal responses associated with alzheimer's disease. CNS Neurosci Ther. 23:135–144. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Allan LA, Morrice N, Brady S, Magee G, Pathak S and Clarke PR: Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nat Cell Biol. 5:647–654. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Hermann C, Assmus B, Urbich C, Zeiher AM and Dimmeler S: Insulin-mediated stimulation of protein kinase Akt: A potent survival signaling cascade for endothelial cells. Arterioscler Thromb Vasc Biol. 20:402–409. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Kitazumi I and Tsukahara M: Regulation of DNA fragmentation: The role of caspases and phosphorylation. FEBS J. 278:427–441. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Bonni A, Brunet A, West AE, Datta SR, Takasu MA and Greenberg ME: Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 286:1358–1362. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Pugazhenthi S, Miller E, Sable C, Young P, Heidenreich KA, Boxer LM and Reusch JE: Insulin-like growth factor-I induces bcl-2 promoter through the transcription factor cAMP-response element-binding protein. J Biol Chem. 274:27529–27535. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Wilson BE, Mochon E and Boxer LM: Induction of bcl-2 expression by phosphorylated CREB proteins during B-cell activation and rescue from apoptosis. Mol Cell Biol. 16:5546–5556. 1996. View Article : Google Scholar : PubMed/NCBI

38 

Hwang S, Lim JW and Kim H: Inhibitory effect of lycopene on amyloid-β-induced apoptosis in neuronal cells. Nutrients. 9(pii): E8832017. View Article : Google Scholar : PubMed/NCBI

39 

Wang H, Xu Y, Yan J, Zhao X, Sun X, Zhang Y, Guo J and Zhu C: Acteoside protects human neuroblastoma SH-SY5Y cells against beta-amyloid-induced cell injury. Brain Res. 1283:139–147. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Ratty AK and Das NP: Effects of flavonoids on nonenzymatic lipid peroxidation: Structure-activity relationship. Biochem Med Metab Biol. 39:69–79. 1988. View Article : Google Scholar : PubMed/NCBI

41 

Olsvik PA, Kristensen T, Waagbo R, Rosseland BO, Tollefsen KE, Baeverfjord G and Berntssen MH: mRNA expression of antioxidant enzymes (SOD, CAT and GSH-Px) and lipid peroxidative stress in liver of Atlantic salmon (Salmo salar) exposed to hyperoxic water during smoltification. Comp Biochem Physiol C Toxicol Pharmacol. 141:314–323. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Tong Y, Bai L, Gong R, Chuan J, Duan X and Zhu Y: Shikonin protects PC12 cells against β-amyloid peptide-induced cell injury through antioxidant and antiapoptotic activities. Sci Rep. 8:262018. View Article : Google Scholar : PubMed/NCBI

43 

Behl C, Davis JB, Lesley R and Schubert D: Hydrogen peroxide mediates amyloid beta protein toxicity. Cell. 77:817–827. 1994. View Article : Google Scholar : PubMed/NCBI

44 

Shaw RJ and Cantley LC: Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 441:424–430. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Pugazhenthi S, Nesterova A, Sable C, Heidenreich KA, Boxer LM, Heasley LE and Reusch JE: Akt/protein kinase B up-regulates Bcl-2 expression through cAMP-response element-binding protein. J Biol Chem. 275:10761–10766. 2000. View Article : Google Scholar : PubMed/NCBI

46 

Magrane J, Rosen KM, Smith RC, Walsh K, Gouras GK and Querfurth HW: Intraneuronal beta-amyloid expression downregulates the Akt survival pathway and blunts the stress response. J Neurosci. 25:10960–10969. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Sastry AVS, Girija Sastry V, Mallikarjun P and Srinivas K: Chemical and pharmacological evaluation of aqueous extract of root bark of ‘Oroxylum indicum’ vent. Int J Pharm Technol. 3:1796–1806. 2011.

48 

Sreedevi Adikay, Usha Rani U and Bharathi Koganti: Protective effect of ethanolic extract of Oroxylum indicum against cisplatin-induced acute renal failure. Int J Pharm Therap. 2:48–53. 2011.

49 

Tenpe R, Aman U, Burle S and Yeole YG: In vitro antioxidant and preliminary hepatoprotective activity of Oroxylum indicum Vent leaf extracts. Pharmacologyonline. 1:35–43. 2009.

50 

Bichitra Nanda Tripathy, Panda SK, Sahoo S, Mishra SK and Nayak L: Phytochemical analysis and hepatoprotective effect of stem bark of Oroxylum indicum (L) Vent. On carbon tetrachloride induced hepatotoxicity in rat. Int J Pharma Biol Arc. 2:1714–1717. 2011.

51 

Kevalkumar R, Rathod, Rashmi CA, Miloni JK and Tejas HG: Evaluation of effect of Oroxylum indicum leaves on central nervous system with special emphasis on epilepsy. J Chemical Pharm Res. 8:680–685. 2016.

52 

Varadarajan S, Yatin S, Aksenova M and Butterfield DA: Review: Alzheimer's amyloid beta-peptideassociated free radical oxidative stress and neurotoxicity. J Struct Biol. 130:184–208. 2000. View Article : Google Scholar : PubMed/NCBI

53 

Butterfield DA: Beta-Amyloid-associated free radical oxidative stress and neurotoxicity: Implications for Alzheimer's disease. Chem Res Toxicol. 10:495–506. 1997. View Article : Google Scholar : PubMed/NCBI

54 

Redza-Dutordoir M and Averill-Bates DA: Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta. 1863:2977–2992. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 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
Mairuae N, Connor JR, Buranrat B and Lee SY: Oroxylum indicum (L.) extract protects human neuroblastoma SH‑SY5Y cells against β‑amyloid‑induced cell injury. Mol Med Rep 20: 1933-1942, 2019
APA
Mairuae, N., Connor, J.R., Buranrat, B., & Lee, S.Y. (2019). Oroxylum indicum (L.) extract protects human neuroblastoma SH‑SY5Y cells against β‑amyloid‑induced cell injury. Molecular Medicine Reports, 20, 1933-1942. https://doi.org/10.3892/mmr.2019.10411
MLA
Mairuae, N., Connor, J. R., Buranrat, B., Lee, S. Y."Oroxylum indicum (L.) extract protects human neuroblastoma SH‑SY5Y cells against β‑amyloid‑induced cell injury". Molecular Medicine Reports 20.2 (2019): 1933-1942.
Chicago
Mairuae, N., Connor, J. R., Buranrat, B., Lee, S. Y."Oroxylum indicum (L.) extract protects human neuroblastoma SH‑SY5Y cells against β‑amyloid‑induced cell injury". Molecular Medicine Reports 20, no. 2 (2019): 1933-1942. https://doi.org/10.3892/mmr.2019.10411