Neuroprotection of N-benzylcinnamide on scopolamine-induced cholinergic dysfunction in human SH-SY5Y neuroblastoma cells

2017-10-11 02:21NichaPuangmalaiWipawanThangniponRungtipSoiampornkulNirutSuwannaPatoomratanaTuchindaSaksitNobsathian

Nicha Puangmalai, Wipawan Thangnipon,, Rungtip Soi-ampornkul, Nirut Suwanna, Patoomratana Tuchinda, Saksit Nobsathian

1 Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, Tailand

2 Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Tailand

3 Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Kampaeng Saen, Nakhonpathom,Tailand

4 Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Tailand

5 Nakhon Sawan Campus, Mahidol University, Phayuhakiri, Nakhon Sawan, Tailand

RESEARCH ARTICLE

Neuroprotection of N-benzylcinnamide on scopolamine-induced cholinergic dysfunction in human SH-SY5Y neuroblastoma cells

Nicha Puangmalai1, Wipawan Thangnipon1,*, Rungtip Soi-ampornkul2, Nirut Suwanna3, Patoomratana Tuchinda4, Saksit Nobsathian5

1 Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, Tailand

2 Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Tailand

3 Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Kampaeng Saen, Nakhonpathom,Tailand

4 Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Tailand

5 Nakhon Sawan Campus, Mahidol University, Phayuhakiri, Nakhon Sawan, Tailand

Abstract

Alzheimer’s disease, a progressive neurodegenerative disease, affects learning and memory resulting from cholinergic dysfunction. Scopolamine has been employed to induce Alzheimer’s disease-like pathologyin vivoandin vitrothrough alteration of cholinergic system.N-benzylcinnamide (PT-3), purified fromPiper submultinerve, has been shown to exhibit neuroprotective properties against amyloid-β-induced neuronal toxicity in rat cortical primary cell culture and to improve spatial learning and memory of aged rats through alleviating oxidative stress. We proposed a hypothesis that PT3 has a neuroprotective effect against scopolamine-induced cholinergic dysfunction. PT-3 (125–200 nM) pretreatment was performed in human neuroblastoma SH-SY5Y cell line following scopolamine induction. PT-3 (125–200 nM) inhibited scopolamine (2 mM)-induced generation of reactive oxygen species, cellular apoptosis, upregulation of acetylcholinesterase activity, downregulation of choline acetyltransferase level, and activation of p38 and JNK signalling pathways. Tese findings revealed the underlying mechanisms of scopolamine-induced Alzheimer’s disease-like cellular dysfunctions, which provide evidence for developing drugs for the treatment of this debilitating disease.

Key Words:Alzheimer’s disease; acetylcholine; apoptosis; acetylcholinesterase inhibitor; oxidative stress;N-benzylcinnamide; natural product; scopolamine; neuronal regeneration

Introduction

Alzheimer’s disease (AD), an age-related neurodegenerative disease, is the most common cause of dementia (Kumar et al., 2015). Hyperphosphorylation of tau protein generates neuritic plaques comprising amyloid-β and neurofibrillary tangles, which are characteristics of AD (Anand et al., 2014).A cholinergic deficit, particularly in basal forebrain, together with a decrease in acetylcholine promotes cognitive impairment (Schliebs and Arendt, 2011). In addition, the presence of acetylcholinesterase (AChE), results in a decline in cholinergic transmission (Mesulam, 2013). Currently, primary treatment for AD is a cholinergic replacement therapy based on AChE inhibitors, such as donepezil, galantamine, and rivastigmine (Zemek et al., 2014).

Scopolamine, a muscarinic acetylcholine receptor antagonist used in animal models and in humans (Bajo et al., 2015;Ghumatkar et al., 2015; Alvarez-Jimenez et al., 2016), induces cognitive impairment, which is associated with an attenuation of cholinergic neurotransmission as well as an increase in oxidant stress and inflammation (Kwon et al., 2010; Min et al., 2015; Balaban et al., 2017). Scopolamine has been employed inin vitrotesting of new compounds, especially those from natural products, for their potential to restore cognitive impairment. For example, scopolamine causes cytotoxicity and downregulation of neuronal and glial cell markers in neuronal (IMR32) and C6 glioma cells respectively, properties that are reversed upon treatment of an alcoholic extract of Ashwagandha leaves (Konar et al., 2011). Pandareesh and Anand (2013) demonstrated that pre-treatment withBacopa monnieraextract protects against scopolamine-induced damage in rat phochromocytoma PC12 cell line associated with upregulation of AChE and downregulation of brain-derived neurotrophic factor. Lee et al. (2014) reported that sulforaphane (10 or 20 μM), an organosulfur compound present in cruciferous vegetables, attenuates acetylcholine and choline acetyltransferase (ChAT) expression in scopolamine-treated primary mouse cortical neurons.

N-benzylcinnamide (PT-3), purified fromPiper submultinerve, has been shown to exhibit neuroprotective properties against amyloid-β-induced neuronal toxicity in rat corticalprimary cell culture (Thangnipon et al., 2013). PT-3 treatment also improves spatial learning and memory of aged rats through impacting on parameters associated with oxidant stress, inflammation, apoptosis, and AChE activity (Tangnipon et al., 2015). In addition, PT-3, in combination with bone morphogenetic protein 9, induces neuronal differentiation of human amniotic fluid mesenchymal stem cells by enhancing β-III tubulin-containing cell numbers and ChAT content (Tangnipon et al., 2016).

Hence, in this study, we investigated the neuroprotective mechanisms of PT-3 against scopolamine-induced cholinergic dysfunction in human neuroblastoma SH-SY5Y cell line.

Materials and Methods

Chemicals and reagents

Unless otherwise indicated, media and supplements used for cell culture were obtained from Gibco (Carlsbad, CA,USA). Scopolamine hydrobromide, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT),2,7-dichlorodihydrofluorescein diacetate (DCFH-DA),5,5′-dithio-bis-(2-nitrobenzoic acid) (DTNB), acetylthiocholine iodine and Bradford reagent were from Sigma (St. Louis,MO, USA). Rabbit anti-ChAT antibody was from Abcam(Cambridge, MA, USA) and other primary and secondary antibodies were from Cell Signalling Technology (Cell Signalling Technology Inc., MA, USA).

Cell culture and treatment

Human neuroblastoma SH-SY5Y cell line was a kind gift from Dr. Martin Broadstock (Wolfson Centre for Age Related Diseases, King’s College London, UK). Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin, and 100 μg/mL streptomycin at 37°C under a humidified atmosphere of 5% CO2and 95% air. At 80–90% confluence, cells were treated with 0.25% trypsin-ethylenediaminetetraacetic acid (EDTA)and plated onto a 96- or 24-well plate at a density of 2 × 105cells/mL. Culture medium was changed to serum-free DMEM(Huang et al., 2014) supplemented with 50–200 nM of PT-3 for 1 hour (Tangnipon et al., 2013) prior to incubation with 0.1–3 mM (usually 2 mM) scopolamine hydrobromide for 24 hours (Pandareesh and Anand, 2013). All experiments were performed using cells from passages 18 to 20.

Cell viability assay

Cells were incubated with 0.5 mg/mL MTT at 37°C for 3 hours, then supernatant was removed andA570nmof the blue formazan crystals dissolved in 100 μL of dimethyl sulfoxide(DMSO) was measured with a microplate reader (Spectra-Max®180, Sunnyvale, CA, USA) as described previously(Ivins et al., 1999). Cell viability was presented as the percentage of A570 nm of each sample relative to control.

Reactive oxygen species (ROS) assay

A DCFH-DAdye assay was used to determine levels of intracellular ROS levels (Lin et al., 2000). In brief, following sequential treatment with PT-3 and scopolamine, cells were incubated with 50 μM DCFH-DA in absolute ethanol for 1 hour in the dark at 37°C. Dichlorofluorescein (DCF) fluorescence (485 nm excitation, 530 nm emission) was quantified using a multimode reader (DTX880, Beckman Coulter, Wals, Austria).

Acetylcholinesterase (AChE) activity assay

SH-SY5Y cells (5 × 105/mL) were cultured in complete DMEM medium in a 96-well plate for 24 hours, then incubated with 50–200 nM PT-3 for 1 hour, followed by 2 mM scopolamine in serum-free DMEM for further 24 hours.Ten cells were washed twice with 100 μL of phosphate-buf fered saline (PBS) and incubated at room temperature with 50 μL of 1.3 mM 5,5′-dithiobis(2-nitrobenzoic acid) (DTNB)and 50 μL of 1.9 mM acetylthiocholine iodide, both dissolved in PBS (Ellman et al., 1961; Gustafsson et al., 2010).A420nmof TNB2–was measured at 1, 15, and 30 minutes using a multimode reader (DTX880, Beckman Coulter, Fullerton,CA, USA). Absorbance without substrate or cells was subtracted as background.

Western blot analysis

Cell lysate proteins (30 μg; determined using Bradford method) were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under sulfhydryl-reducing condition and transferred onto polyvinylidene fluoride membrane. Membranes were incubated with the following primary antibodies in TBS-T overnight at 4°C at the specified dilution: rabbit anti-ChAT (1:1,000 dilution),anti-Bcl-2 (1:1,000 dilution), anti-Bax (1:1,000 dilution),anti-activated caspase-3 (1:500 dilution), anti-phospho-p38(1:1,000 dilution), anti-p38 (1:1,000 dilution), anti-phospho-ERK1/2 (1:1,000 dilution), anti-ERK1/2 (1:1,000),anti-phospho-JNK (1:1,000), anti-JNK (1:1,000), anti-phospho-Akt (1:1,000 dilution), anti-Akt (1:1,000 dilution) or anti-β-actin (1:2,500 dilution) (latter as internal control)antibodies, and then incubated with horseradish peroxidase(HRP)-conjugated secondary anti-rabbit IgG (1:1,000 dilution) antibodies at room temperature for 1 hour. Immunoreactive protein bands were detected using an ECL western blotting substrate (Bio-Rad, Hercules, CA, USA) and recorded on HyperfilmTM(Amersham Pharmacia Biotech). Band density was determined as scanning units using imageJ soft-ware (National Institutes of Health, MA, USA) and expression levels were quantified relative to that of β-actin.

Statistical analysis

One-way analysis of variance (ANOVA) was performed using Prism 5.0a (GraphPad Software, Inc., San Diego, CA, USA)to measure statistical significance of differences, followed by Student-Newman-Keuls test. Data are presented as the mean± SEM of three independent experiments in triplicate. Results are considered statistically significant atP-value < 0.05.

Results

PT-3 attenuated scopolamine-induced cytotoxicity, ROS generation and apoptosis of SH-SY5Y cells

Figure 1 N-benzylcinnamide (PT-3) pretreatment prevented scopolamine (Sco)-induced cytotoxicity and intracellular reactive oxygen species (ROS) generation in SH-SY5Y cells.

Figure 2 N-benzylcinnamide (PT-3) pretreatment prevented scopolamine (Sco)-induced reduction of choline acetyltransferase(ChAT) activity and induction of pro-apoptosis events in SH-SY5Y cells.

Figure 3 N-benzylcinnamide (PT-3) pretreatment reversed scopolamine (Sco)-induced elevation of acetylcholinesterase (AChE)activity in SH-SY5Y cells.

Figure 4 N-benzylcinnamide (PT-3) pretreatment reversed scopolamine (Sco)-induced upregulation of phosphorylated p38 (p-p38) and downregulation of phosphorylated JNK (p-JNK), but did not affect phosphorylation of ERK1/2 and Akt in SH-SY5Y cells (western blot analysis).

Exposure to scopolamine (1–3 mM) for 24 hours signifi-cantly decreased human neuroblastoma SH-SY5Y cell viability in a dose-dependent manner (Figure 1A), which is in accordance with findings from a previous study (Konar et al., 2011). In all subsequent experiments, 2 M scopolamine resulted in 50.6 ± 2.2% loss in cell viability, which was inhibited by pretreatment with 125–200 nM PT-3 for 1 hour in a concentration-dependent manner (Figure 1B). In addition,PT-3 pretreatment ameliorated scopolamine-induced ROS formation over the same concentration range in a dose-dependent manner (Figure 1C). Treatment with PT-3 alone had no apparent effect on cell viability or endogenous ROS level.

An inverted phase-contrast microscopy revealed that live cells presented shrinkage and cell death (apoptosis) of SHSY5Y cells following scopolamine exposure, which were prevented by PT-3 pretreatment (Figure 1D–F). Cell apoptosis was confirmed by the presence of activated caspase-3 (Figure 2B) and elevated Bax/Bcl-2 ratio (Figure 2C), which again were diminished by PT-3 (200 nM) pretreatment.

PT-3 suppressed AChE activity but restored ChAT activity in scopolamine-treated SH-SY5Y cells

As scopolamine upregulated AChE activity in rat PC12 cells(Pandareesh and Anand, 2013), a similar phenomenon was observed when SH-SY5Y cells were treated with 2 mM scopolamine, resulting in 150.5 ± 3.5%, 131.7 ± 2.1%, and 113.4± 1.5% increase in AChE activity compared to untreated control when assayed for 1, 15 and 30 minutes, respectively(Figure 3), which, as expected, was prevented by PT-3 (50–200 nM) pretreatment. PT-3 restored ChAT activity, reduced by scopolamine, with 1.5 ± 0.16-fold increase in ChAT activity compared to control cells at the highest (200 nM) PT-3 concentration used (Figure 2A).

PT-3 modulated p38 and JNK pathways in SH-SY5Y cells exposed to scopolamine

Scopolamine activated redox-responsive cell signaling pathways, such as oxidative stress, inflammation, and cell death (Foyet et al., 2015). In order to identify the pathway(s)involved in the situation of SH-SY5Y cells, western blot analysis was employed to identify key signature factors of activated signaling pathways. Exposure to 2 mM scopolamine resulted in 1.6-fold increase in phospho-p38/p38 ratio over untreated control SH-SY5Y cells, and this was restored to near control value by prior incubation with 100–150 nM PT-3, but only to 50.2 ± 1.8% of control with 200 nM PT-3(Figure 4A). Whereas 2 mM scopolamine reduced phospho-JNK/JNK ratio to 75.4 ± 2.4% of control cells, which was further reduced by 100 and 150 nM PT-3 pretreatment(Figure 4A,B). However, phosphorylation of ERK1/2 and Akt was not affected (Figure 4C,D).

Discussion

SH-SY5Y cells were chosen for study as they are commonly used as models to investigatein vitroneuronal function(Forster et al., 2016) and to assess pharmacological and toxicological potential of AChE inhibitors (Kanhed et al., 2015;Santillo and Liu, 2015). In a previous study (Zhang et al.,2016), scopolamine-induced ROS formation and cellular apoptosis (viathe classical Bax/Bcl-2 pathway) in SH-SY5Y cells were ameliorated by pretreatment with PT-3. PT-3 manifests antioxidant property (Thangnipon et al., 2013).C6 glioma cells pre-treated with 0.5–2 μM lactucopicrin,a sesquiterpene lactone derived fromLactuca virosaandCichorium intybus, have reduced 2′,7′-dichlorofluorescein toxicity induced by 3 mM scopolamine (Venkatesan et al.,2016).

Interestingly, PT-3 pretreatment was capable of suppressing scopolamine-stimulated SH-SY5Y AChE activity. Xian et al. (2015) reported that Honokiol, a lignin from the bark ofMagnolia of ficinalis, decreases AChE activity in brain tissues of scopolamine-treated mice. Honokiol contains two phenolic groups, which can exhibit antioxidant property similar to PT-3 (Dikalov et al., 2008).

There is evidence that undifferentiated or differentiated SH-SY5Y cells contain AChR mRNA (Korecka et al., 2013).In this study, PT-3 restored ChAT activity in SH-SY5Y cells that had been reduced by scopolamine, probably by increasing ChAT expression and thereby restoring cholinergic cell function. Tis property is not confined to PT-3 as Lim et al.(2016) recently demonstrated that curcumin, a natural antioxidant, increases immunoreactive ChAT level in scopolamine-treated mouse hippocampus and improves learning impaired by scopolamine insult.

Te activation of p38 and JNK signalling pathways is associated with cellular stress and pro-inflammatory responses(Davis, 2000). Scopolamine induces a number of pathological phenomena associated with neurodegenerative diseases linked to memory decline, including increased oxidative stress, impaired antioxidative defence system and mitochondrial dysfunction (Wong-Guerra et al., 2017). Additionally,JNK and p38 are stress-activated MAP kinases preferentially activated by cell stress-inducing signals, including oxidative stress, environmental stress and toxic chemical insults. Sustained activation of JNK or p38 is implicated in the induction of many forms of neuronal apoptosis in response to a variety of cellular injuries (Wong-Guerra et al., 2017). In scopolamine-treated SH-SY5Y cells, PT-3 inhibits phosphorylation of p38 and JNK, as have been observed in rat primary cultures and human mesenchymal stem cells (Tangnipon et al., 2013, 2016). Although low doses of PT-3 (100–150 nM)inhibited scopolamine-induced cytotoxicity via inactivation of JNK, surprisingly high dose of PT-3 (200 nM) enhanced JNK activation (but lower than that in control cells) as has been reported by Shi et al. (2009). In that study, low concentrations (50–100 μg/mL) of EGb761, an extract fromGinkgobiloba leaves, inhibits H2O2-induced cell apoptosisviasuppression of JNK and caspase-3 activation while high concentrations (250–500 μg/mL) enhance JNK phosphorylation. It is worth noting that pre-treatment with a number of natural phenolic compounds show similar effects on p38 and JNK pathways in oxidant stressed SH-SY5Y cells (Dhanalakshmi et al., 2015; Zhu et al., 2015).

ROS plays a critical role in cell signalling, particularly inredox mechanisms involved in apoptosis, such as the mitochondria-to-cytosol release of cytochrome c, a central event in apoptosis initiation (Circu and Aw, 2010). Interestingly,aLiriope platyphyllaextract exerts neuroprotective effects against H2O2by modulating only p38 and not ERK in SHSY5Y cells (Park et al., 2015). In addition, p38 inhibitor SB203580 blocks cell loss in H2O2-treated SH-SY5Y cells(Park et al., 2015). Thus, in SH-SY5Y cells, H2O2requires p38 activation for cytotoxicity.

Lee et al. (2005) suggested that baicalein, a flavonoid fromScutellariaroot, induces a significant reduction in phospho-JNK level of 6-hydroxydopamine-induced neuronal cell death. Tey also found that ERK and Akt pathways are not altered by scopolamine treatment. These pathways are responsible for cell proliferation, cell differentiation and cell survival (Junttila et al., 2008). Similarly, Voleti et al. (2013)found a low dose of scopolamine treatment (25 μg/kg) does not disturb activated ERK level and has a minimal effect on phospho-Akt in cultured rat pyramidal neurons. Gunjima et al. (2014) reported a similar observation of protection by 3,4-dihydroxybenzalacetone, a catechol-containing compound isolated fromInonotus obliquus(persoon) Pilat, against 6-hydroxydopamine toxicity in SH-SY5Y cells that is not affected by ERK inhibitor PD98059. Similarly, Wang et al. (2014)demonstrated that glycyrrhizic acid, a major compound ofGlycyrrhiza radix, alone or in combination with 20 mM glutamate, has no effect on phospho-Akt level in PC12 cells.

Acknowledgments:Te authors are grateful to Professor Prapon Wilairat,Mahidol University, Tailand for critical reading of the manuscript.

Author contributions:WT and NP designed and evaluated the study.All authors participated in performing the experiments and approved the?nal version of this paper.

Conflicts of interest:Te authors declare that there are no conflicts of interest.

Data sharing statement:Te datasets analyzed during the current study are available from the corresponding author on reasonable request.

Plagiarism check:Checked twice by iTenticate.

Peer review:Externally peer reviewed.

Open access statement:Tis is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

Open peer review reports:

Reviewer 1:Aysegul Yildiz-Unal, Mugla Sitki Kocman University, Turkey.

Comments to authors:Tere are a number of studies with PT-3 showing its protective effect on degenerating primary neuron cultures. In one of them, Thangnipon W. et al. showed that N-benzylcinnamide protects rat cultured cortical neurons from β-amyloid peptide-induced neurotoxicity. After 2 years, the same research group showed the potential role of N-benzylcinnamide in inducing cholinergic neuronal differentiation.Since scopalamine is also an Alzheimer’s disease-like pathology inducer,it makes sense to analyze the protective role of PT-3 on scopalamine -induced Alzheimer’s disease-like pathology in a different cell type, SH-SY5Y neuroblastoma cells. See additional ?le for more details.

Additional file:Open peer review report 1.

Reviewer 2:Siyu Zhang, University of California Berkeley, USA.

Comments to authors: Strengths: the authors tested the changes of the scopolamine induced in SH-SY5Y cells. Weakness:To establish a sound model in SH-SY5Y by scopolamine, the authors did not provide a positive control group which can ameliorate those changes induced by scopolamine. See additional ?le for more details.

Additional file:Open peer review report 2.

Alvarez-Jimenez R, Groeneveld GJ, van Gerven JM, Goulooze SC,Baakman AC, Hay JL, Stevens J (2016) Model-based exposure-response analysis to quantify age related differences in the response to scopolamine in healthy subjects. Br J Clin Pharmacol 82:1011-1021.

Anand R, Gill KD, Mahdi AA (2014) Terapeutics of Alzheimer’s disease: Past, present and future. Neuropharmacology76:27-50.

Bajo R, Pusil S, Lopez ME, Canuet L, Pereda E, Osipova D, Maestu F,Pekkonen E (2015) Scopolamine effects on functional brain connectivity: a pharmacological model of Alzheimer’s disease. Sci Rep 5:9748.

Balaban H, Naziroglu M, Demirci K, Ovey IS (2017) Te protective role of selenium on scopolamine-induced memory impairment, oxidative stress, and apoptosis in aged rats: the involvement of TRPM2 and TRPV1 channels. Mol Neurobiol 54:2852-2868.

Circu ML, Aw TY (2010) Reactive oxygen species, cellular redox systems, and apoptosis. Free Radic Biol Med 48:749-762.

Davis RJ (2000) Signal transduction by the JNK group of MAP kinases.Cell 103:239-252.

Dhanalakshmi C, Manivasagam T, Nataraj J, Tenmozhi A, Essa MM(2015) Neurosupportive role of Vanillin, a natural phenolic compound, on rotenone induced neurotoxicity in SH-SY5Y neuroblastoma cells. Evid Based Complement Alternat Med 2015:626028.

Dikalov S, Losik T, Arbiser JL (2008) Honokiol is a potent scavenger of superoxide and peroxyl radicals. Biochem Pharmacol 76:589-596.

Ellman GL, Courtney KD, Andres V, Feather-Stone RM (1961) A new and rapid colorimetric determination of acetylcholinesterase activity.Biochem Pharmaco l7:88-95.

Forster JI, Koglsberger S, Trefois C, Boyd O, Baumuratov AS, Buck L,Balling R, Antony PM (2016) Characterization of differentiated SHSY5Y as neuronal screening model reveals increased oxidative vulnerability. J Biomol Screen 21:496-509.

Foyet HS, Abaissou HH, Wado E, Acha EA, Alin C (2015) Emilia coccinae (SIMS) G extract improves memory impairment, cholinergic dysfunction, and oxidative stress damage in scopolamine-treated rats. BMC Complement Altern Med 15:333.

Ghumatkar PJ, Patil SP, Jain PD, Tambe RM, Sathaye S (2015) Nootropic, neuroprotective and neurotrophic effects of phloretin in scopolamine induced amnesia in mice. Pharmacol Biochem Behav135:182-191.

Gunjima K, Tomiyama R, Takakura K, Yamada T, Hashida K, Nakamura Y, Konishi T, Matsugo S, Hori O (2014) 3,4-dihydroxybenzalacetone protects against Parkinson’s disease-related neurotoxin 6-OHDA through Akt/Nrf2/glutathione pathway. J Cell Biochem1 15:151-160.

Gustafsson H, Runesson J, Lundqvist J, Lindegren H, Axelsson V,Forsby A (2010) Neurofunctional endpoints assessed in human neuroblastoma SH-SY5Y cells for estimation of acute systemic toxicity.Toxicol Appl Pharmacol 245:191-202.

Huang HC, Tang D, Xu K, Jiang ZF (2014) Curcumin attenuates amyloid-β-induced tau hyperphosphorylation in human neuroblastoma SH-SY5Y cells involving PTEN/Akt/GSK-3β signaling pathway. J Recept Signal Transduct Res 34:26-37.

Ivins KJ, Ivins JK, Sharp JP, Cotman CW (1999) Multiple pathways of apoptosis in PC12 cells. CrmA inhibits apoptosis induced by β-amyloid. J Biol Chem 274:2107-2112.

Junttila MR, Li SP, Westermarck J (2008) Phosphatase-mediated crosstalk between MAPK signaling pathways in the regulation of cell survival. FASEB J 22:954-965.

Kanhed AM, Sinha A, Machhi J, Tripathi A, Parikh ZS, Pillai PP, Giridhar R, Yadav MR (2015) Discovery of isoalloxazine derivatives as a new class of potential anti-Alzheimer agents and their synthesis.Bioorg Chem 61:7-12.

Konar A, Shah N, Singh R, Saxena N, Kaul SC, Wadhwa R, Takur MK(2011) Protective role of Ashwagandha leaf extract and its component withanone on scopolamine-induced changes in the brain and brain-derived cells. PLoS One 6:e27265.

Korecka JA, van Kesteren RE, Blaas E, Spitzer SO, Kamstra JH, Smit AB, Swaab DF, Verhaagen J, Bossers K (2013) Phenotypic characterization of retinoic acid differentiated SH-SY5Y cells by transcriptional profiling. PLoS One 8:e63862.

Kumar A, Singh A, Ekavali E (2015) A review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep 67:195-203.

Kwon SH, Lee HK, Kim JA, Hong SI, Kim HC, Jo TH, Park YI, Lee CK,Kim YB, Lee SY, Jang CG (2010) Neuroprotective effects of chlorogenic acid on scopolamine-induced amnesia via anti-acetylcholinesterase and anti-oxidative activities in mice. Eur J Pharmacol 649:210-217.

Lee HJ, Noh YH, Lee DY, Kim YS, Kim KY, Chung YH, Lee WB, Kim SS (2005) Baicalein attenuates 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y cells. Eur J Cell Biol 84:897-905.

Lee S, Kim J, Seo SG, Choi BR, Han JS, Lee KW, Kim J (2014) Sulforaphane alleviates scopolamine-induced memory impairment in mice.Pharmacol Res 85:23-32.

Lim DW, Son HJ, Um MY, Kim IH, Han D, Cho S, Lee CH (2016) Enhanced cognitive effects of demethoxycurcumin, a natural derivative of curcumin on scopolamine-induced memory impairment in mice.Molecules 21:E1022.

Lin JK, Chen PC, Ho CT, Lin-Shiau SY (2000) Inhibition of xanthine oxidase and suppression of intracellular reactive oxygen species in HL-60 cells by theaflavin-3,3′-digallate, (-)-epigallocatechin-3-gallate,and propyl gallate. J Agric Food Chem 48:2736-2743.

Mesulam MM (2013) Cholinergic circuitry of the human nucleus basalis and its fate in Alzheimer’s disease. J Comp Neurol 521:4124-4144.

Min AY, Doo CN, Son EJ, Sung, NY, Lee KJ, Sok DE, Kim MR (2015)N-palmitoyl serotonin alleviates scopolamine-induced memory impairment via regulation of cholinergic and antioxidant systems,and expression of BDNF and p-CREB in mice. Chem Biol Interact 242:153-162.

Pandareesh MD, Anand T (2013) Neuromodulatory propensity of Bacopa monniera against scopolamine-induced cytotoxicity in PC12 cells via down-regulation of AChE and up-regulation of BDNF and muscarnic-1 receptor expression. Cell Mol Neurobiol 33:875-884.

Park HR, Lee H, Park H, Jeon JW, Cho WK, Ma JY (2015) Neuroprotective effects of Liriope platyphylla extract against hydrogen peroxide-induced cytotoxicity in human neuroblastoma SH-SY5Y cells.BMC Complement Altern Med 15:171.

Santillo MF, Liu Y (2015) A fluorescence assay for measuring acetylcholinesterase activity in rat blood and a human neuroblastoma cell line (SH-SY5Y). J Pharmacol Toxicol Methods 76:15-22.

Schliebs R, Arendt T (2011) Te cholinergic system in aging and neuronal degeneration. Behav Brain Res 221:555-563.

Shi C, Zhao L, Zhu B, Li Q, Yew DT, Yao Z, Xu J (2009) Dosage effects of EGb761 on hydrogen peroxide-induced cell death in SH-SY5Y cells. Chem Biol Interact 180:389-397.

Thangnipon W, Puangmalai N, Chinchalongporn V, Jantrachotechatchawan C, Kitiyanant N, Soi-Ampornkul R, Tuchinda P,Nobsathian S (2013) N-benzylcinnamide protects rat cultured cortical neurons from β-amyloid peptide-induced neurotoxicity. Neurosci Lett 556:20-25.

Tangnipon W, Suwanna N, Jantrachotechatchawan C, Ngampramuan S, Tuchinda P, Nobsathian S (2015) Protective roles of N-benzylcinnamide on cortex and hippocampus of aged rat brains. Arch Pharm Res 38:1380-1388.

Tangnipon W, Puangmalai N, Suwanna N, Soi-Ampornkul R, Phonchai R, Kotchabhakdi N, Mukda S, Phermthai T, Julavijitphong S,Tuchinda P, Nobsathian S (2016) Potential role of N-benzylcinnamide in inducing neuronal differentiation from human amniotic fluid mesenchymal stem cells. Neurosci Lett 610:6-12.

Venkatesan R, Subedi L, Yeo EJ, Kim SY (2016) Lactucopicrin ameliorates oxidative stress mediated by scopolamine-induced neurotoxicity through activation of the NRF2 pathway. Neurochem Int 99:133-146.

Voleti B, Navarria A, Liu RJ, Banasr M, Li N, Terwilliger R, Sanacora G,George TE, Aghajanian G, Duman RS (2013) Scopolamine rapidly increases mammalian target of rapamycin complex 1 signaling, synaptogenesis, and antidepressant behavioral responses. Biol Psychiatry 74:742-749.

Wang D, Guo TQ, Wang ZY, Lu JH, Liu DP, Meng QF, Xie J, Zhang XL,Liu Y, Teng LS (2014) ERKs and mitochondria-related pathways are essential for glycyrrhizic acid-mediated neuroprotection against glutamate-induced toxicity in differentiated PC12 cells. Braz J Med Biol Res 47:773-779.

Wong-Guerra M, Jiménez-Martin J, Pardo-Andreu GL, Fonseca-Fonseca LA, Souza DO, de Assis AM, Ramirez-Sanchez J, Del Valle RM,Nuñez-Figueredo Y (2017) Mitochondrial involvement in memory impairment induced by scopolamine in rats. Neurol Res 39:649-659.

Xian YF, Ip SP, Mao QQ, Su ZR, Chen JN, Lai XP, Lin ZX (2015) Honokiol improves learning and memory impairments induced by scopolamine in mice. Eur J Pharmacol 760:88-95.

Zemek F, Drtinova L, Nepovimova E, Sepsova V, Korabecny J, Klimes J, Kuca K (2014) Outcomes of Alzheimer’s disease therapy with acetylcholinesterase inhibitors and memantine. Expert Opin Drug Saf 13:759-774.

Zhang J, Cai S, Li J, Xiong L, Tian, L, Liu J, Huang J, Liu Z (2016) Neuroprotective effects of Theaflavins against oxidative stress-induced apoptosis in PC12 cells. Neurochem Res 41:3364-3372.

Zhu A, Wu Z, Meng J, McGeer PL, Zhu Y, Nakanishi H, Wu S (2015)Te neuroprotective effects of Ratanasampil on oxidative stress-mediated neuronal damage in human neuronal SH-SY5Y cells. Oxid Med Cell Longev 2015:792342.

Copyedited by Li CH, Song LP, Zhao M

How to cite this article: Puangmalai N, Tangnipon W, Soi-ampornkul R, Suwanna N, Tuchinda P, Nobsathian S (2017) Neuroprotection of N-benzylcinnamide on scopolamine-induced cholinergic dysfunction in human SH-SY5Y neuroblastoma cells. Neural Regen Res 12(9):1492-1498.

Funding: Te study was ?nancially supported by a joint Mahidol University and Te Tailand Research Fund (TRF) grant (IRG5780009), TRF Royal Golden Jubilee Ph.D. Program (grant No. PHD/0175/2552) and the Of fice of the Higher Education Commission, Ministry of Education,Tailand.

*Correspondence to:Wipawan Tangnipon, Ph.D.,wipawan.tha@mahidol.ac.th.

orcid:0000-0001-7889-2801(Wipawan Tangnipon)

10.4103/1673-5374.215262

Accepted: 2017-07-25