Skip to main content

Neuroprotective effects of Paeonia Lactiflora extract against cell death of dopaminergic SH-SY5Y cells is mediated by epigenetic modulation

Abstract

Background

The Paeonia lactiflora extract (PLE) has been reported to have neuroprotective effect against neurodegeneration that are induced by cellular stress such as oxidative stress. Its underlying mechanisms, however, remain unclear. In latest decades, emerging evidence has suggested that epigenetic mechanisms play a key role in gene regulation in response to the cellular stress. We investigated whether epigenetic modulation was involved in neuronal cell death by the neurotoxicant, 1-Methyl-4-phenylpyridinium (MPP+), and the neuroprotective effect of PLE.

Methods

Differentiated SH-SY5Y, which is a well-established dopaminergic cell line model, was treated with 0 ~ 200 μg/ml PLE for 4 h prior to MPP+ treatment. The effect of PLE on cell viability was determined by MTT assays. Gene expression levels of oxidative stress responsive genes, such as Heme oxygenase 1 (HMOX1), and histone modifiers, such as histone acetyltransferases (HATs) and deacetylases (HDACs) were measured by quantitative RT PCR. In order to investigate the changes in epigenetic modifications, the acetylated lysine 9 (H3K9ac) and lysine 27 (H3K27ac) of Histone H3 were measured by western blot using histones extracted from the cells.

Results

MPP+-induced cell death in SH-SY5Y cells was significantly reduced by PLE pretreatment in a dose-dependent manner, indicating the potent neuroprotective effects of PLE. It was accompanied by induced expression of HMOX1. MPP+ treatment increased the expression of HATs and consistently increased H3K9ac and H3K27ac of Histone H3. PLE pretreatment impeded the changes in H3K9ac and H3K27ac, coincided with increased expression of HDAC5 without changes in HAT expression.

Conclusions

The results suggested that MPP+-induced cell death in the dopaminergic SH-SY5Y cells was related with transcriptional induction of HATs and increased histone H3 acetylation and that PLE might prevent the cells from MPP+-induced cell death via tempering histone H3 acetylation.

Peer Review reports

Background

Parkinson’s disease, the second most common neurodegenerative disorder following Alzheimer’s disease, is mainly caused by progressive degeneration of dopaminergic neurons [1]. One major risk factor leading to the degeneration is cellular oxidative stress [2]. Oxidative stress is a cellular oxidative imbalanced condition due to impaired mitochondrial function and elevated reactive oxygen species (ROS) in the cell. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its metabolite, MPP+, has been used as oxidative stressors that induce cell death in dopaminergic neurons in vivo and in vitro, which mimic Parkinson’s disease [2]. MPP+ enters dopaminergic neuronal cells via dopamine transporters and induces mitochondria dysfunction. It produces ROS [3] and releases cytochrome C (Cyt-C), which activates caspase-3 and eventually, leads dopaminergic neuronal cells to apoptosis [4]. MPP+-induced cell system serves as a useful model to study the molecular mechanism underlying loss of dopaminergic neurons and therapeutic potentials.

Paeonia lactiflora Pall is one of the most commonly used medicinal herbs across different traditional Chinese medicine treatment for its anti-inflammatory and antioxidant effects [5]. The herbal extract of this plant, PLE, has been reported to have neuroprotective effects against oxidative stress both in vivo [6] and in vitro [7, 8], in addition to the main compounds such as albiflorin [9], paeoniflorin [1012], and paeonol [13, 14] that have been extracted from this plant. It indicates its potentials to protect dopaminergic neurons from cell death and to attenuate the process of Parkinson’s disease [6, 15, 16]. The underlying molecular mechanism of PLE, however, remains elusive.

Recently, the emerging evidence on disease-preventive and therapeutic effects of phytochemicals and herb extracts suggests that they mainly exert their functions, at least in part, via epigenetic modulation [17]. Epigenetic mechanisms such as DNA methylation and histone modifications regulate gene expression via changes in chromatin accessibility in response to environmental stimuli such as oxidative stress [18]. Disturbed patterns of histone acetylation as well as altered activity of HATs and HDACs have been connected to chronic diseases, including cancer and neurodegenerative diseases [18]. Cellular stress alters histone acetylation at specific residues such as histone H3 lysine9 and histone H4 lysine12 [1921] and changes the activities of HDACs [22, 23], suggesting the potential development of therapeutic strategies for neurodegenerative conditions [2427]. However, the epigenetic mechanism has been investigated on limited phytochemicals, for examples, curcumin enriched in turmeric and sulforaphane enriched in broccoli, which have effects on cell death and proliferation by inhibiting HATs and HDACs, respectively [2830]. Consequently, to identify potential modulators of histone acetylation in neuronal cells may provide novel therapeutic avenues in neurodegenerative disorders.

Therefore, we investigated whether the neuroprotective effects of PLE against oxidative stress in dopaminergic neuron cells are mediated by epigenetic modulation. Using the MPP+-induced SY5Y cell model, gene expression involving the antioxidant pathway as well as histone acetylation histone acetylation levels at histone H3 lysine 9 (H3K9) and lysine 27 (H3K27) were analyzed with and without PLE treatment.

Methods

Cell culture

SH-SY5Y cell line was obtained from the Division of Epigenomics and Cancer Risk Factors, German Cancer Research Cancer (DKFZ, Heidelberg, Germany). The cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Welgene, Daegu, South Korea) supplemented with 10 % fetal bovine serum (FBS) (Gibco, Gaithersberg, MD, USA), 100 units/ml penicillin, and 100 μg/ml streptomycin (Gibco, Gaithersberg, MD, USA) with incubation at 37 °C in humidified 5 % CO2 and 95 % air. Upon cell passage, treatment of cells with 0.25 % trypsin-EDTA (Gibco, Gaithersberg, MD, USA), was followed by quenching in DMEM whereupon the cells were collected by centrifugation and rinsed with phosphate buffered saline (PBS) (Sigma Aldrich, St. Louis, MO, USA). The number of viable cells was counted in a hemocytometer under a microscope after excluding dead cells dyed with 0.4 % trypan blue solution (Gibco, Gaithersberg, MD, USA).

Cell differentiation and treatment

To obtain dopaminergic neuronal cells, SY5Y cells were differentiated using DMEM with 10 μM retinoic acid (Sigma Aldrich, St. Louis, MO, USA), 1 % FBS and100 units/ml penicillin, and 100 μg/ml streptomycin for 4 days. The medium was changed after 2 days during differentiation. The differentiation was confirmed by the increase in mRNA level of Tyrosine hydroxylase gene (TH). Differentiated cells were treated with 0-2 mM MPP+ (Sigma Aldrich, St. Louis, MO, USA) for 24 h to optimize the concentration of MPP+ treatment. To investigate PLE effects, the differentiated cells were pretreated with 0-200 μg/ml PLE for 4 h before 1 mM MPP+ treatment. Lyophilized PLE extract was prepared by Hanpoong Pham & Foods Co., Ltd (Jeonju, South Korea). Briefly, 300 g of Paeonia lactiflora was refluxed for 3 h in three liters of 30 % ethanol, passed through 0.1 μm filter and vaporized. PLE, obtained with 201.3 % yield, was dissolved in dimethyl sulfoxide (DMSO) (Sigma Aldrich, St. Louis, MO, USA) in 80 mg/ml PLE as a stock solution. The control groups were supplemented with the same amount of DMSO in the final maximum concentration less than 0.25 %.

MTT assays

3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazoliumbromide (MTT) (Sigma Aldrich, St. Louis, MO, USA) was dissolved in PBS at the concentration 5 mg/ml. The MTT solution was added to one-tenth the original culture volume and the cells were incubated at 37 °C for 2 h. Then, DMSO was added in cells to convert soluble MTT to insoluble purple formazan in mitochondria of living cells. Viable cells forming formazan were measured at 562 nm using a microplate reader (Lab tech international, Uckfield, UK).

RNA isolation and reverse transcription

Total RNA was isolated from cells by Trizol reagent (Tel-test Inc, Friendswood, TX, USA). Reverse transcription of 500 ng or 1 μg total RNA was performed for cDNA synthesis. In brief, RNA samples were treated with DNase I (Thermo Fisher Scientific, Waltham, MA, USA) to remove genomic DNA, after which RNA samples were incubated with dNTP (Thermo Fisher Scientific, Waltham, MA, USA), oligo dT (Thermo Fisher Scientific, Waltham, MA, USA), and reverse transcriptase (Thermo Fisher Scientific, Waltham, MA, USA) to synthesize cDNA, following the manufacturer’s protocol.

Quantitative reverse transcriptase (RT)-PCR

Quantitative RT-PCR was performed using SYBR green mix (QIAGEN, Hilden, Germany) and Rotor-Gene Q (QIAGEN, Hilden, Germany). RT PCR primer sequences were the followings: TBP, 5’-AGCCAAGAGTGAAGAACAGTCC-3’ (forward) and 5’-CACAGCTCCCCACCATATTC-3’ (reverse); HMOX1, 5’-AACTTTCAGAAGGGCCAGGT-3’ (forward) and 5’-GTAGACAGGGGCGAAGACTG-3’ (reverse); Superoxide dismutase 2 (SOD2), 5’-TCAATAAGGAACGGGGACAC-3’ (forward) and 5’-ACACATCAATCCCCAGCAGT-3’ (reverse); E1A binding protein p30 (EP300),5’-AGGGGCAACAAGAAGAAACC-3’ (forward) and 5’-GAGGCGGATCACAAAGAAGA-3’ (reverse); CREB Binding Protein (CREBBP), 5’-CACTGCAGCCAGTGAAACC-3’ (forward) and 5’-CATGCTGGGCTTCTTCTTGT-3’ (reverse); HDAC5, 5’-AAGGTCCTCATCGTGGACTG-3’ (forward) and 5’-GCACAGAGGGGTCATTGTAGA-3’ (reverse). Amplification procedure was 95 °C for 5 min, followed by 40 cycles at 95 °C for 5 s, 60 °C for 10 s. Primers for TBP mRNA were used to normalize mRNA expression. All experiments were done in biological triplicate.

Histone extraction

Histones were extracted from SH-SY5Y cells by acid extraction method in accordance with Shechter, et al.’s protocol [31]. Cells were washed with 1X PBS and pelleted by centrifugation at 1000 g for 3 min. Cell pellets were incubated in hypotonic lysis buffer (10 mM Tris, 1 mM KCl, 1 mM DTT, 1.5 mM MgCl2, 10 mM Sodium butyrate, 1 mM PMSF, and 7X protease inhibitor cocktail (Roche, Robert Rohrberg, Germany)) on a rotator at 4 °C for 30 min. The nuclei were collected by centrifugation at 10,000 x g at 4 °C for 10 min, and the pelleted nuclei were suspended in 0.4 N H2SO4 and placed on a rotator at 4 °C overnight. Next day, the nuclear lysate was collected from the supernatants after centrifugation at 14,620 x g at 4 °C for 10 min to remove nuclear debris. Histones were precipitated by TCA and washed with acetone-HCl and acetone. Precipitated histones were collected by centrifugation at 14,620 x g at 4 °C for 5 min, air-dried at room temperature for 15 min and then dissolved in distilled water. Histones were stored at -80 °C. All steps were carried out by keeping samples on ice whenever possible.

Western blotting analysis

Isolated histones (2 or 3 μg per sample) were run on a 15 % PAGE. Histones were transferred to Nitrocellulose (BioRad Laboratories, Hercules, CA, USA) or PVDF (Millipore, Billerica, MA, USA) membranes. Membranes were blocked in TBS-T with 5 % skim milk (DifcoLaboratories, Detroit, Mich, USA) for 1 h at room temperature and incubated overnight at 4 °C with primary antibodies from Cell Signaling Technology, Danvers, MA, USA. The following antibodies were used: rabbit antibodies towards Histone H3 (1:1500), H3K9ac (1:1000) and H3K27ac (1:1000) and mouse antibodies to Histone H4 (1:1000). After washing, rabbit or mouse IgG-HRP secondary antibodies (Santa Cruz Biotechnology, Santa Cruz, CA, USA) were incubated for 1 h at room temperature and then developed at Pierce Enhanced chemiluminescence (ECL) kit (Thermo Scientific, Rockford, IL, USA). The level of acetylated histone was quantified by Image J. All experiments were done in biological triplicate.

Statistical analysis

Mean ± S.E.M. of cell viability and mRNA expression levels were calculated by using Microsoft Excel 2010 (Microsoft, Redmond, WA, USA). One-way ANOVA analysis, followed by Duncan post hoc multiple comparisons, was performed using SPSS (ver. 20.0, SPSS Inc., Chicago, IL, USA). Statistical significance was set to p ≤ 0.05.

Results

MPP+-mediated changes of cell viability and SOD2 and HMOX1 expression in differentiated SH-SY5Y cells

We analyzed cell viability of SH-SY5Y cells upon MPP+ treatment using MTT assays. Treatment with MPP+ concentrations ranging from 0 to 2 mM decreased cell viability in a dose-dependent manner, down to about 52 % (Fig. 1a). To investigate whether MPP+ treatment changed the expression of genes related with the cellular anti-oxidative pathway in response to oxidative stress, mRNA expression levels of SOD2 and HMOX1 were analyzed (Fig. 1b and c). Gene expression level of HMOX1 significantly increased at in 1 to 2 mM MPP+ treatment and SOD2 significantly decreased at the 2 mM concentration. The results indicated that MPP+ indeed acted as a neurotoxicant in dopaminergic neuronal cells by inducing oxidative stress-mediated cell death. While the dose at 1 mM MPP+ is relatively low, it was sufficiently toxic to significantly elevate HMOX1 gene expression and reduce cell viability. Consequently, for subsequent experiment we used the 1 mM dose to analyze the neuroprotective effects provided by PLE.

Fig. 1
figure 1

Changes of cell viability and mRNA expressions of SOD2 and HMOX1 in MPP+ treated cells. a MPP+ treatment decreased cell viability depending on concentrations of MPP+. b MPP+ treatment decreased mRNA expressions of SOD2 related with clearing mitochondrial ROS. The results confirmed MPP+ induces apoptosis by way of inhibition of mitochondrial complex. c MPP+ treatment increased mRNA expressions of HMOX1 related with the antioxidant pathway. The results consent HMOX1 is induced by oxidative stress.; The data was indicated as the mean ± S.E.M. P < 0.05 was defined statistical significance. One-way ANOVA followed by Duncan post hoc test

Effects of PLE on MPP+-mediated cell death and HMOX1 expression

To investigate the effect of PLE against MPP+-induced cell death, we pretreated SH-SY5Y cells with PLE in the range of 0 to 200 μg/ml for 4 h prior to a 24 h 1 mM MPP+ treatment. PLE pretreatment attenuated MPP+-induced cell death in a dose-dependent manner (Fig. 2a). Significantly, 200 μg/ml PLE recovered cell viability up to 93 % compared with MPP+ nontreated group. Interestingly, treatment with PLE did not alter SOD2 gene expression levels, a key component of the antioxidant pathway (Fig. 2b), while HMOX1 gene expression levels in 20-200 μg/ml PLE samples were significantly increased 1.5 to 1.9 fold, respectively, when compared to 0 μg/ml PLE group (Fig. 2c).

Fig. 2
figure 2

Cell viability and mRNA expressions of SOD2 and HMOX1 in PLE and MPP+ treated cells. a PLE treatment recovered 1 mM MPP+ treated cell viability depending on concentrations of PLE. The results suggested PLE has neuroprotective effects against MPP+-induced apoptosis. b PLE treatment didn’t alter mRNA expressions of SOD2 in 1 mM MPP+ treated SH-SY5Y cells. c PLE treatment further activated mRNA expressions of HMOX1 in 1 mM MPP+ treated SH-SY5Y cells. The results suggested PLE has neuroprotective effects on antioxidant function.; The data was indicated as the mean ± S.E.M. P < 0.05 was defined statistical significance. One-way ANOVA followed by Duncan post hoc test

Changes in acetylated levels of histones in MPP+ treated, differentiated SH-SY5Y cells

To determine whether MPP+-induced cell death was a result of epigenetic changes, we investigated alteration in acetylation levels of histones and HAT gene expression levels. The acetylation levels of H3K9 and H3K27 (relative to total histones H3 and H4) were analyzed using western blot analysis. When treating cells with MPP+ ranging from 0.5 to 2 mM, we observed a linear correlation with increased acetylation levels of H3K9 and H3K27 (Fig. 3a). In addition, gene expression levels of the major HAT proteins, EP300 and CREBBP, were also increased in the MPP+ treated samples (Fig. 3b). The results suggested that increased HAT expression under neurotoxic condition led to increased acetylation levels of H3K9 and H3K27.

Fig. 3
figure 3

Acetylation of H3K9 and H3K27 and mRNA expressions of HATs in MPP+ treated cells. a MPP+ treatment increased acetylation levels of H3K9 and H3K27. b MPP+ treatment increased mRNA expressions of HATs such as EP300 and CREBBP. Figure 3 suggested HATs increase acetylation levels of H3K9 and H3K27.; The data was indicated as the mean ± S.E.M. P < 0.05 was defined statistical significance. One-way ANOVA followed by Duncan post hoc test

H3K9ac and H3K27ac and HDAC5 and HATs mRNA expressions in and MPP+ treated cells

Next, we determined whether pretreatment of PLE had neuroprotective effect against MPP+-induced cell death by means of epigenetic modulation. We investigated alteration of acetylation levels of histones and HATs and HDACs gene expression levels. We measured acetylation levels of H3K9 and H3K27 in MPP+ treated cells with and without PLE using western blotting analysis. In the PLE pretreated group, we observed that acetylation level of H3K9 decreased, but not H3K27 acetylation (Fig. 4a). The decreased acetylated H3K9 seemed to be due to induction of the HDAC5 gene expression (Fig. 4b), rather than altered EP300 and CREBBP expression (Fig. 4c), as HDAC5 robustly increased with elevated PLE concentration. Our data suggest that HDAC5 decreased acetylation levels of H3K9 in the 200 μg/ml PLE group, overriding the effect from HATs, potentiation PLE as an anti-neurotoxicant by tempering histone acetylation.

Fig. 4
figure 4

H3K9ac and H3K27ac and HDAC5 and HATs mRNA expressions in PLE and MPP+ treated cells. a 200 μg/ml PLE treatment decreased acetylation levels of H3K9 in 1 mM MPP+ treated cells. b PLE treatment did not significantly alter mRNA expressions of HATs in 1 mM MPP+ treated cells. c 200 μg/ml PLE treatment increased mRNA expressions of HDAC5 in 1 mM MPP+ treated cells. Figure 4 suggested HDAC5 rather than HATs decreases acetylation levels of H3K9. The data was indicated as the mean ± S.E.M. P < 0.05 was defined statistical significance. One-way ANOVA followed by Duncan post hoc test

Discussion

In this study, we investigated whether MPP+ induced cell death in the dopaminergic neuronal SH-SY5Y cells through epigenetic modulation, and whether PLE had a neuroprotective effect against cell death by reversing epigenetic changes. We demonstrated that MPP+-induced cell death decreased SOD2 gene expression and increased the HMOX1 gene expression levels. The results are consistent with previous reports showing downregulation of SOD2 and upregulation of HMOX1 under oxidative stress [15, 32, 33]. PLE was recently reported to have protective effect on neuronal cell death in studies both in vivo [6] and in vitro [7]. The positive influence of PLE on dopaminergic neuronal cells potentiates its use in attenuating the process of Parkinson’s disease [15, 16, 34]. However, the molecular mechanism of this protective effect remains unclear.

One possibility is that the PLE effect involves the antioxidant pathway. Our results demonstrated that PLE upregulated HMOX1 gene expression, while it did not alter SOD2 gene expression. This indicates that the PLE effect may be restricted to the HMOX1 pathway. HMOX1 gene encodes heme oxygenase 1 (HMOX1), which catalyzes heme catabolism and plays a protective role as an antioxidant. HMOX1 gene is activated in response to oxidative stress, mainly via a major transcriptional factor, Nuclear factor erythroid 2 related factor 2 (NRF2), which exerts as a ROS scavenger [35]. A recent study showed that Bromodomain protein 4 (BRD4) also transcriptionally activates HMOX1 expression by targeting of SP1 binding sites in the HMOX1 promoter [36]. BRD4, an acetylated histone-binding protein, is a key factor for regulation of gene expression [37, 38]. siRNA-mediated silencing of Bromodomain protein 4 (BRD4), in the absence of stress, resulted in downregulation of HMOX1 expression [36]. However, under oxidative stress, siRNA-mediated knockdown of BRD4 upregulates HMOX1 expression further via downregulation of Kelch-like ECH-associated protein 1 (KEAP1), which inhibits the degradation of NRF2 [36]. The obvious connection between hypoacetylated histone residues, especially H3K9, is reflected by the silencing of BRD4 expression, since BRD4 binds to particularly acetylated histones.

The neurprotective effect of PLE on MPP+-induced cell death of the dopaminergic cells seems mediated by epigenetic modulation, supported by our observation of changes in acetylation of histone. Treatment of MPP+ increased gene expression levels of HATs such as EP300 and CREBBP, resulting in hyperacetylation of H3K9 and H3K27. This is consistent with a previous report demonstrating that neurotoxicity induced hyperacetylation and transcriptional activation of pro-apoptotic genes, such as PKC gamma and IL-8 [39]. Conversely, the pretreatment of PLE increased gene expression of HDAC5, resulting in hypoacetylation of H3K9. HDAC5 has been reported to involve neuronal cell survival, proliferation, and differentiation possibly via regulation of Myocyte enhancer factor-2 (MEF2) [4042]. The resulting hypoacetylation upon PLE treatment may mimic the effect of BRD4 silencing under stress, leading to upregulated HMOX1 expression.

Our study of the neuroprotective properties of PLE suggest that epigenetic modulation is a key component of neurotoxic stress, and that therapeutic intervention should consider this avenue of treatment in diseases such as Parkinson’s disease.

Conclusions

MPP+-induced cell death in differentiated SH-SY5Y cells was significantly reduced by PLE pretreatment in a dose-dependent manner, accompanied by induced expression of HMOX1. MPP+-treatment increased the expression of HATs and consistently increased acetylation of H3K9 and H3K27, while PLE pretreatment attenuated the hyperacetylation in H3K9, coincided with increased expression of HDAC5 without changes in HAT expression. Our study suggested that MPP+ treatment induced cell death in dopaminergic SH-SY5Y cells via upregulation of HATs and hyperacetylation of H3K9 and H3K27 and PLE pretreatment may attenuate the cell death, in part, by upregulation of HMOX1 expression through hypoacetylation of H3K9, possibly mediated by HDAC5 upregulation. Further study is needed to identify direct targets for HDAC5-mediated hypoacetylation of H3K9 in the neuroprotective effects and to investigate whether it is recapitulated in vivo.

Abbreviations

BRD4, Bromodomain protein 4; DMEM, Dulbecco’s modified Eagle’s medium; DMSO, dimethyl sulfoxide; EP300, E1A binding protein p30; FBS, fetal bovine serum; H3K27, histone H3 lysine 27; H3K27ac, acetylated lysine 27 of histone H3; H3K9, histone H3 lysine 9; H3K9ac, acetylated lysine 9 of histone H3; HAT, histone acetyltransferases; HDAC, histone deacetylases; HMOX1, heme oxygenase 1; MPP+, 1-methyl-4-phenylpyridinium; NRF2, nuclear factor erythroid 2 related factor 2; PBS, phosphate buffered saline; PLE, Paeonia lactiflora extract; ROS, reactive oxygen species; SOD2, superoxide dismutase 2

References

  1. Bertram L, Tanzi RE. The genetic epidemiology of neurodegenerative disease. J Clin Invest. 2005;115(6):1449–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Blesa J, Trigo-Damas I, Quiroga-Varela A, Jackson-Lewis VR. Oxidative stress and Parkinson’s disease. Front Neuroanat. 2015;9:91.

    PubMed  PubMed Central  Google Scholar 

  3. Mizuno Y, Sone N, Saitoh T. Effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and 1-methyl-4-phenylpyridinium ion on activities of the enzymes in the electron transport system in mouse brain. J Neurochem. 1987;48(6):1787–93.

    Article  CAS  PubMed  Google Scholar 

  4. Petrozzi L, Ricci G, Giglioli N, Siciliano G, Mancuso M. Mitochondria and neurodegeneration. Biosci Rep. 2007;27(1-3):87–104.

    Article  CAS  PubMed  Google Scholar 

  5. He DY, Dai SM. Anti-inflammatory and immunomodulatory effects of paeonia lactiflora pall., a traditional chinese herbal medicine. Front Pharmacol. 2011;2:10.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Liu HQ, Zhang WY, Luo XT, Ye Y, Zhu XZ. Paeoniflorin attenuates neuroinflammation and dopaminergic neurodegeneration in the MPTP model of Parkinson’s disease by activation of adenosine A1 receptor. Br J Pharmacol. 2006;148(3):314–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lee S-M, Yoon M-Y, Park H-R. Protective effects of Paeonia lactiflora pall on hydrogen peroxide-induced apoptosis in PC12 cells. Biosci Biotechnol Biochem. 2008;72(5):1272–7.

    Article  CAS  PubMed  Google Scholar 

  8. Kim SH, Lee MK, Lee KY, Sung SH, Kim J, Kim YC. Chemical constituents isolated from Paeonia lactiflora roots and their neuroprotective activity against oxidative stress in vitro. J Enzyme Inhib Med Chem. 2009;24(5):1138–40.

    Article  CAS  PubMed  Google Scholar 

  9. Suh KS, Choi EM, Lee YS, Kim YS. Protective effect of albiflorin against oxidative-stress-mediated toxicity in osteoblast-like MC3T3-E1 cells. Fitoterapia. 2013;89:33–41.

    Article  CAS  PubMed  Google Scholar 

  10. Liu DZ, Xie KQ, Ji XQ, Ye Y, Jiang CL, Zhu XZ. Neuroprotective effect of paeoniflorin on cerebral ischemic rat by activating adenosine A1 receptor in a manner different from its classical agonists. Br J Pharmacol. 2005;146(4):604–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu D-Z, Zhu J, Jin D-Z, Zhang L-M, Ji X-Q, Ye Y, et al. Behavioral recovery following sub-chronic paeoniflorin administration in the striatal 6-OHDA lesion rodent model of Parkinson’s disease. J Ethnopharmacol. 2007;112(2):327–32.

    Article  CAS  PubMed  Google Scholar 

  12. Wang D, Wong HK, Feng Y-B, Zhang Z-J. Paeoniflorin, a natural neuroprotective agent, modulates multiple anti-apoptotic and pro-apoptotic pathways in differentiated PC12 cells. Cell Mol Neurobiol. 2013;33(4):521–9.

    Article  CAS  PubMed  Google Scholar 

  13. Hsieh C-L, Cheng C-Y, Tsai T-H, Lin I-h, Liu C-H, Chiang S-Y, et al. Paeonol reduced cerebral infarction involving the superoxide anion and microglia activation in ischemia-reperfusion injured rats. J Ethnopharmacol. 2006;106(2):208–15.

    Article  CAS  PubMed  Google Scholar 

  14. Zhong S-Z, Ge Q-H, Qu R, Li Q, Ma S-P. Paeonol attenuates neurotoxicity and ameliorates cognitive impairment induced by d-galactose in ICR mice. J Neurol Sci. 2009;277(1):58–64.

    Article  CAS  PubMed  Google Scholar 

  15. Ye Q, Huang B, Zhang X, Zhu Y, Chen X. Astaxanthin protects against MPP + -induced oxidative stress in PC12 cells via the HO-1/NOX2 axis. BMC Neurosci. 2012;13:156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cao B-Y, Yang Y-P, Luo W-F, Mao C-J, Han R, Sun X, et al. Paeoniflorin, a potent natural compound, protects PC12 cells from MPP+ and acidic damage via autophagic pathway. J Ethnopharmacol. 2010;131(1):122–9.

    Article  CAS  PubMed  Google Scholar 

  17. Anderson OS, Sant KE, Dolinoy DC. Nutrition and epigenetics: an interplay of dietary methyl donors, one-carbon metabolism and DNA methylation. J Nutr Biochem. 2012;23(8):853–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tsankova N, Renthal W, Kumar A, Nestler EJ. Epigenetic regulation in psychiatric disorders. Nat Rev Neurosci. 2007;8(5):355–67.

    Article  CAS  PubMed  Google Scholar 

  19. Ferland CL, Harris EP, Lam M, Schrader LA. Facilitation of the HPA Axis to a Novel Acute Stress Following Chronic Stress Exposure Modulates Histone Acetylation and the ERK/MAPK Pathway in the Dentate Gyrus of Male Rats. Endocrinology. 2014;155(8):2942–52.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chandramohan Y, Droste SK, Arthur JSC, Reul JM. The forced swimming‐induced behavioural immobility response involves histone H3 phospho‐acetylation and c‐Fos induction in dentate gyrus granule neurons via activation of the N‐methyl‐d‐aspartate/extracellular signal‐regulated kinase/mitogen‐and stress‐activated kinase signalling pathway. Eur J Neurosci. 2008;27(10):2701–13.

    Article  PubMed  Google Scholar 

  21. Ferland CL, Schrader LA. Regulation of histone acetylation in the hippocampus of chronically stressed rats: a potential role of sirtuins. Neuroscience. 2011;174:104–14.

    Article  CAS  PubMed  Google Scholar 

  22. Lee JB, Wei J, Liu W, Cheng J, Feng J, Yan Z. Histone deacetylase 6 gates the synaptic action of acute stress in prefrontal cortex. J Physiol. 2012;590(7):1535–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sando R, Gounko N, Pieraut S, Liao L, Yates J, Maximov A. HDAC4 governs a transcriptional program essential for synaptic plasticity and memory. Cell. 2012;151(4):821–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Krishnan V, Nestler EJ. Linking molecules to mood: new insight into the biology of depression. Am J Psychiatry. 2010;167(11):1305–20.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Menke A, Klengel T,B, Binder E. Epigenetics, depression and antidepressant treatment. Curr Pharm Des. 2012;18(36):5879–89.

    Article  CAS  PubMed  Google Scholar 

  26. Dietz KC, Casaccia P. HDAC inhibitors and neurodegeneration: at the edge between protection and damage. Pharmacol Res. 2010;62(1):11–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Archer T, Oscar-Berman M, Blum K, Gold M. Epigenetic modulation of mood disorders. J genetic Syndrome Gene Ther 2013;4(120):1000120.

  28. Balasubramanyam K, Varier RA, Altaf M, Swaminathan V, Siddappa NB, Ranga U, et al. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. J Biol Chem. 2004;279(49):51163–71.

    Article  CAS  PubMed  Google Scholar 

  29. Kang SK, Cha SH, Jeon HG. Curcumin-induced histone hypoacetylation enhances caspase-3-dependent glioma cell death and neurogenesis of neural progenitor cells. Stem Cells Dev. 2006;15(2):165–74.

    Article  CAS  PubMed  Google Scholar 

  30. Myzak MC, Karplus PA, Chung FL, Dashwood RH. A novel mechanism of chemoprotection by sulforaphane: inhibition of histone deacetylase. Cancer Res. 2004;64(16):5767–74.

    Article  CAS  PubMed  Google Scholar 

  31. Shechter D, Dormann HL, Allis CD, Hake SB. Extraction, purification and analysis of histones. Nat Protoc. 2007;2(6):1445–57.

    Article  CAS  PubMed  Google Scholar 

  32. Piao Y, Kim HG, Oh MS, Pak YK. Overexpression of TFAM, NRF-1 and myr-AKT protects the MPP + -induced mitochondrial dysfunctions in neuronal cells. Biochim Biophys Acta (BBA) Gen Subj. 2012;1820(5):577–85.

    Article  CAS  Google Scholar 

  33. Zhu J, Gusdon A, Cimen H, Van Houten B, Koc E, Chu C. Impaired mitochondrial biogenesis contributes to depletion of functional mitochondria in chronic MPP&plus; toxicity: dual roles for ERK1/2. Cell Death Dis. 2012;3(5):e312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lu J, Park C-S, Lee S-K, Shin DW, Kang J-H. Leptin inhibits 1-methyl-4-phenylpyridinium-induced cell death in SH-SY5Y cells. Neurosci Lett. 2006;407(3):240–3.

    Article  CAS  PubMed  Google Scholar 

  35. Hussong M, Börno S, Kerick M, Wunderlich A, Franz A, Sültmann H, et al. The bromodomain protein BRD4 regulates the KEAP1/NRF2-dependent oxidative stress response. Cell Death Dis. 2014;5(4):e1195.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chorley BN, Campbell MR, Wang X, Karaca M, Sambandan D, Bangura F, et al. Identification of novel NRF2-regulated genes by ChIP-Seq: influence on retinoid X receptor alpha. Nucleic Acids Res. 2012;40(15):7416–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Huang B, Yang X-D, Zhou M-M, Ozato K, Chen L-F. Brd4 coactivates transcriptional activation of NF-kB via specific binding to acetylated RelA. Mol Cell Biol. 2009;29(5):1375–87.

    Article  CAS  PubMed  Google Scholar 

  38. Timothy B, Bodén M, Buske F, Frith M, Grant E, Clementi L, et al. MEME SUITE: tools for motif discovery and searching. Nuc Acids Res. 2009;37:W202–8.

    Article  Google Scholar 

  39. Tomita K, Barnes P, Adcock I. The effect of oxidative stress on histone acetylation and IL-8 release. Biochem Biophys Res Commun. 2003;301(2):572–7.

    Article  CAS  PubMed  Google Scholar 

  40. Flavell SW, Cowan CW, Kim T-K, Greer PL, Lin Y, Paradis S, et al. Activity-dependent regulation of MEF2 transcription factors suppresses excitatory synapse number. Science. 2006;311(5763):1008–12.

    Article  CAS  PubMed  Google Scholar 

  41. Chawla S, Vanhoutte P, Arnold FJ, Huang CLH, Bading H. Neuronal activity‐dependent nucleocytoplasmic shuttling of HDAC4 and HDAC5. J Neurochem. 2003;85(1):151–9.

    Article  CAS  PubMed  Google Scholar 

  42. Heidenreich KA, Linseman DA. Myocyte enhancer factor-2 transcription factors in neuronal differentiation and survival. Mol Neurobiol. 2004;29(2):155–65.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MEST) (2012R1A1A1018819 & 2014R1A1A2058942) and the Ewha Womans University Research Grant of 2011. GL and BYC were supported by the Brain Korea 21 Plus Project in 2013 (22A20130012143).

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article.

Authors’ contributions

GL, SJP, JCJ, and YJP designed hypothesis and supervise experiments; SJP and JCJ provided the sample; GL and BYC conducted the experiments and GL, AML, and YJP analyzed the data; GL, BYC, AML, and YJP wrote the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Soo Jung Park or Yoon Jung Park.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, G., Joo, J.C., Choi, B.Y. et al. Neuroprotective effects of Paeonia Lactiflora extract against cell death of dopaminergic SH-SY5Y cells is mediated by epigenetic modulation. BMC Complement Altern Med 16, 208 (2016). https://doi.org/10.1186/s12906-016-1205-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-016-1205-y

Keywords