Downregulation of miR‑21 suppresses 1‑methyl‑4‑phenylpyridinium‑induced neuronal damage in MES23.5 cells

  • Authors:
    • Huawu Mao
    • Lidong Ding
  • View Affiliations

  • Published online on: August 5, 2019     https://doi.org/10.3892/etm.2019.7853
  • Pages: 2467-2474
  • Copyright: © Mao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Accumulating evidence suggests that overproduction of oxidative stress, increases neuroinflammation and activates apoptosis. These two processes are associated with the development of Parkinson's disease (PD). The present study aimed to investigate the role of miR‑21 in the development of PD. 1‑Methyl‑4‑phenylpyridinium (MPP+) was used to induce a PD‑like model in MES23.5 cells. The results of the reverse transcription‑quantitative PCR assays indicated that miR‑21 levels were markedly increased in MES23.5 cells following MPP+ treatment. Furthermore, MES23.5 cells were transfected with miR‑21 inhibitor, mimics and/or relevant negative control, following MPP+ administration. The results of the functional assays revealed that downregulation of miR‑21 significantly attenuated the induction of cell apoptosis and reactive oxygen species (ROS) production, while it enhanced the survival of MPP+‑induced MES23.5 cells. Furthermore, downregulation of miR‑21 increased the expression levels of tyrosine hydroxylase, whereas suppression of miR‑21 inhibited the production of pro‑inflammatory cytokines [interleukin (IL)‑6, IL‑1β and tumor necrosis factor‑α] in MES23.5 cells. Western blot analysis further indicated that the Bcl‑2/Bax protein expression ratio was significantly increased and double luciferase assay analysis confirmed that Bcl‑2 was a direct target of miR‑21. Taken collectively, the data demonstrated that downregulation of miR‑21 protected cells from MPP+‑mediated cytotoxicity by the inhibition of apoptosis induction, the reduction of the inflammatory response and the suppression of ROS production. The present findings may provide novel approaches for PD clinical treatment.

Introduction

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by selective loss of dopaminergic neurons in the substantia nigra. This process leads to dopamine (DA) depletion in the striatum affecting several million patients worldwide, notably older adults (1). Although the biochemical and molecular pathogenesis of the loss of dopaminergic neurons in PD have not yet been fully characterized, a number of biochemical processes and molecular mechanisms have been identified as mediators of neuronal cell death in PD, including the overproduction of oxidative stress, the increase in neuroinflammation and the activation of the apoptotic cascade (2). Oxidative stress and apoptosis have been shown to play central roles in the degeneration of dopaminergic neurons in PD (24). Therefore, the exploration of novel molecular mechanisms involved in oxidative stress and apoptosis is of great significance for the treatment of PD.

MicroRNAs (miRNAs/miRs), are a class of endogenous highly conserved noncoding RNA molecules, which are ~22 nucleotides in length (5). By binding directly to the 3′-UTR (3′-untranslated region) of target mRNAs, miRNAs are involved in a series of physiological and pathological processes (6). Accumulating evidence suggests that the dysregulation of miRNA plays an important role in the pathogenesis of neurodegenerative disorders, including PD (7,8). Fu et al (9) demonstrated that docosahexaenoic acid upregulated the expression of Peroxisome Proliferator Activated Receptor α by inhibiting miR-21 in SH-Y5Y cells. Moreover, a recent study showed that the levels of miR-21 in PD models were significantly higher than those in normal controls in SH-SY5Y cells (10). This evidence suggests that miR-21 may play an important role in PD, while the underlying mechanism remains unclear.

In the present study, the classic neurotoxin 1-methyl-4-phenylpyridinium (MPP+) was used to produce a PD cell model in vitro. This compound is selectively transported into dopaminergic neurons via the DA transporter and localizes in the mitochondria (11). The aim of the current study was to investigate the role of miR-21 in MPP+-induced neurotoxicity of MES23.5 cells and to prove the mechanism of miR-21 in PD to provide novel approaches for clinical treatment.

Materials and methods

Cell culture and treatment

MES23.5 cells were cultured in DMEM/F12 (Sigma-Aldrich; Merck KGaA) supplemented with 5% fetal bovine serum (Thermo Fisher Scientific, Inc.), 2% of 50X of Sato's solution (Thermo Fisher Scientific, Inc.) at 37°C in a humidified (70–80%) atmosphere containing 5% CO2. The cells were seeded in 96-well plates at a density of 1×105 cells/well and treated with different concentrations of MPP+ (100, 200 or 300 µM) for 3, 6, 12 or 24 h to optimize the experimental conditions.

Experimental groups were as follows: Control group, MES23.5 cells with no treatment and the model group, MES23.5 cells with 200 µM MPP+ treatment for 24 h.

Transfection

miR-21 inhibitor (100 nM) and negative control (NC, 100 nM) sequences were obtained from GenePharma and were transfected into MES23.5 cells using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The sequences of the inhibitor and the NC samples were as follows: miR-21 inhibitor (5′-UCAACAUCAGUCUGAUAAGCUA-3′), and NC (5′-CAGUACUUUUGUGUAGUACAA-3′). At 24-h following transfection, the cells were treated with 200 µM of MPP+ for an additional 24 h at 37°C. Subsequently, the cells were harvested for further experiments. Experimental groups were as follows: Control group, MES23.5 cells with 200 µM MPP+ treatment and no transfection; NC group, MES23.5 cells with 200 µM MPP+ treatment and inhibitor NC and miR-21 inhibitor group, MES23.5 cells with 200 µM MPP+ treatment and miR-21 inhibitor.

Cell Counting Kit-8 (CCK-8) assay

The CCK-8 assay was used to assess cell viability. The cells were seeded in 96-well plates (2×104 cells/well) and incubated at 37°C for 24 h. At 3, 6, 12 and 24-h following MPP+ treatment (100, 200 and 300 µM) or MPP+ treatment (200 µM) for 24, 48 and 72-h, the cell proliferation indices were measured using a CCK-8 kit (cat. no. C0038, Beyotime Institute of Biotechnology), according to the manufacturer's protocol. The optical density was measured at 450 nm.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted using TRIzol reagent (Invitrogen Life Technologies; Thermo Fisher Scientific, Inc.). Single-stranded cDNA was synthesized using the TaqMan MicroRNA Reverse Transcription Kit (Takara) with the special stem-loop primers. The reverse transcription conditions were following 25°C for 10 min, 45°C for 30 min and 5 min at 95°C. RT-qPCR was performed using a Perfect Real Time SYBR Premix Ex Taq Kit (Takara Bio) with an ABI 7500 thermocycler (Thermo Fisher Scientific, Inc.). All procedures were performed according to the manufacturer's protocol. U6 was used as control for the expression levels of miR-21. The reaction conditions for PCR were as follows: Pre-denaturation at 95°C for 3 min and 40 cycles of denaturation at 95°C for 30 sec and annealing at 60°C for 30 sec. The relative expression levels of each gene were calculated by the 2−ΔΔCq method (12). miRNA-specific reverse transcription primers and quantitative PCR primers were obtained from RiboBio Co. Ltd. The following primer sequences were used: miR-21 stem-loop primer, 5′-GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACTCAACA-3′ forward, 5′-CACGCACGCATAGCTTATCAGACT-3′ and reverse, 5′-CCAGTGCAGGGTCCGAGGTA-3′. U6, stem-loop primer, 5′-GTCGTATCCAGTGCAGGGTCCGAGGTGCACTGGATACGACAAAATATGG-3′, forward, 5′-TGCGGGTGCTCGCTTCGGCAGC-3′ and reverse, 5′-CCAGTGCAGGGTCCGAGGTA-3′.

Immunocytochemistry

MES23.5 cells (1×105 cells/well) were fixed with 4% paraformaldehyde for 30 min at 4°C and permeabilized with 0.5% Triton X-100 for 20 min at room temperature. Following blocking with 5% goat serum (Gibco; Thermo Fisher Scientific, Inc.) for 1-h at room temperature and incubation with mouse anti-tyrosine hydroxylase (TH; cat. no. T2928; 1:500; Sigma-Aldrich; Merck KGaA) at 4°C overnight, the cells were further incubated with biotinylated goat anti-mouse secondary antibody (cat. no. A0286; 1,1000; Beyotime Institute of Biotechnology) for 15 min at 37°C. Subsequently, the samples were washed with PBS three times for 15 min at 37°C and visualized by diaminobenzidine (DAB). TH positive cells were counted in three randomly selected images (magnification, ×200) using an Olympus Fluoview FV5000 microscope (Thermo Fisher Scientific, Inc.).

Detection of ROS levels

The ROS levels were assessed using the fluorescent dye 2′,7′-dichlorofluorescin diacetate (DCFDA; D6883, Sigma-Aldrich; Merck KGaA). MES23.5 cells were seeded at a density of 2×104 cells/well in 96-well plates. The cells were incubated with 2.5 µM of DCFDA for 15 min at 37°C. DCFDA fluorescence was acquired by confocal microscopy at excitation and emission wavelengths of 485 and 535 nm, respectively (Carl Zeiss LSM 700 Meta confocal microscope; Zeiss). The data were analyzed by the Carl Zeiss confocal laser scanning microscopes with the ZEN 2008 software.

ELISA

The culture medium of MES23.5 cells was collected. The levels of IL-6, IL-1β and tumor necrosis factor-α (TNF-α) were measured by the Interleukin-1β ELISA kit (cat. no. ab100562; Abcam) according to the manufacturer's protocol.

Cell apoptosis analysis

An Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (NanJing KeyGen Biotech Co., Ltd) was used to measure cell apoptosis according to the manufacturer's protocol. The samples were analyzed by flow cytometry. A total of 100 nM miR-21 inhibitor and NC sequences were transfected with MES23.5 cells for 24 h at 37°C then the cells were treated with MPP+ for an additional 24 h. The cells were subsequently harvested, washed twice with ice-cold PBS and suspended using binding buffer (500 µl; Thermo Fisher Scientific, Inc.). Subsequently, the cells were stained using (Annexin-V-FITC) reagent and propidium iodide (PI). The cells were analyzed by a FACSCalibur flow cytometer with the Cell Quest software (version 3.1; Becton Dickinson).

Western blot analysis

Total protein extraction was performed using RIPA lysis buffer (Beyotime Institute of Biotechnology). The protein concentration was measured using the bicinchoninic acid kit (Bio-Rad Laboratories, Inc.). The protein samples (20 µg/lane) were separated by SDS-PAGE (12% resolving gels, 4% stacking gels) and blotted onto polyvinylidene difluoride membranes (EMD Millipore). The membranes were blocked using 5% non-fat milk for 1-h at room temperature, followed by overnight incubation at 4°C with the indicated antibodies against Bax (cat. no. MAB4601, 1:200, EMD Millipore) and Bcl-2 (cat. no. 05-826, 1:500, EMD Millipore). Subsequently, the membranes were incubated with secondary rabbit anti-mouse IgG-horseradish peroxidase antibodies (cat. no. sc-358914, 1:5,000, Santa Cruz Biotechnology, Inc.) at room temperature for a further 2-h. Then, membranes were washed with Tris-buffered saline and Polysorbate 20 seven times (3 min per wash). Chemiluminescent signals were visualized using the enhanced chemiluminescence detection reagent (EMD Millipore). The results were analyzed using ImageJ software 1.4 (National Institutes of Health).

Dual-luciferase reporter assay

TargetScanHuman (www.targetscan.org) was used to predict the putative target site of Bcl-2. The mutant type of the Bcl-2 3′UTR sequence was constructed using a QuickChange Multi Site-Directed Mutagenesis kit (Agilent Technologies, Inc.), according to the manufacturer's protocol. The wild type or mutant types of the Bcl-2 3′UTR sequences were cloned into the firefly luciferase reporter pGL3-promoter vector (Promega Corp.) to generate the recombinant, wild type (3′UTR-WT) or mutant type (3′UTR-MUT) pGL3-Bcl-2-3′UTR luciferase plasmids. MES23.5 cells (1×105 cells/well) were cultured in 24-well plates for 24 h at room temperature, and the cells were co-transfected with 50 ng of 3′UTR-WT (or 3′UTR-MUT) vector and 20 µM of miR-21 (or NC) mimics (Shanghai GenePharma Co., Ltd.) using Lipofectamine™ 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. At 24 h following transfection, the Dual-Luciferase Reporter Assay System (Promega Corp.) was used to determine the luciferase activity, which was normalized to Renilla luciferase activity.

Statistical analysis

The data are presented as the mean ± standard error of the mean. GraphPad Prism version 6 (GraphPad Software, Inc.) was used to perform the statistical analyses. The differences between two groups were carried out by a two-tailed Student's t-test. One-way analysis of variance followed by Bonferroni's multiple comparison tests was used to compare differences between means in more than two groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Downregulation of miR-21 enhances cell survival in MPP+-treated MES23.5 cells

The optimal concentration of MPP+ was determined based on neuronal viability, which was monitored by the CCK-8 assay. MPP+ treatment demonstrated dose- and time-dependent cytotoxicity of MES23.5 cells. These effects were noted at the concentrations of 100, 200 and 300 µM for 0, 3, 6, 12 and/or 24-h time points (Fig. 1A). The optimal concentration (200 µM) and time point (24 h) of incubation exhibited approximately 50% cell viability following MPP+ treatment (Fig. 1A). The expression levels of miR-21 in MPP+-treated MES23.5 cells were assessed by RT-qPCR. The results indicated that the expression levels of miR-21 were significantly higher in MES23.5 cells following MPP+ treatment compared with those of the control group (Fig. 1B). To further investigate the role of miR-21 in the development of PD, MES23.5 cells were transfected with miR-21 inhibitor and NC sequences, followed by treatment with 200 µM of MPP+ for 24 h. The expression levels of miR-21 were significantly decreased following transfection with miR-21 inhibitor in MES23.5 cells compared with those of the NC group (Fig. 1C). The CCK-8 assay indicated that the transfection of the miR-21 inhibitor significantly increased the cell survival of the MES23.5 cells following MPP+ treatment at the 24, 48 and 72-h time points compared with that noted in the NC group (Fig. 1D). Moreover, the characteristics of active neurons were classified according to the expression of TH (13). The results of the immunohistochemical assay demonstrated that downregulation of miR-21 significantly increased the number of TH positive cells (Fig. 2).

Downregulation of miR-21 causes inhibition of the inflammatory response and ROS production in MPP+-treated MES23.5 cells

To investigate the role of miR-21 in MPP+-mediated cell damage, the production of ROS was determined in MPP+-treated MES23.5 cells. The results suggested that downregulation of miR-21 attenuated ROS production compared with that of the NC group (Fig. 3A). Subsequently, the levels of neuroinflammation were assessed by ELISA analysis. The expression levels of the inflammatory markers IL-6, IL-1β and TNF-α were significantly reduced following transfection of the cells with the miR-21 inhibitor (Fig. 3).

Downregulation of miR-21 suppresses cell apoptosis and Bcl-2 is its direct target

The effects of miR-21 in the induction of apoptosis were further evaluated. The data demonstrated that downregulation of miR-21 resulted in a significant inhibition of apoptosis in MPP+-treated MES23.5 cells compared with that of the NC group (Fig. 4). In addition, the effects of miR-21 on the expression levels of the apoptotic proteins were also investigated. Inhibition of miR-21 resulted in increased Bcl-2 and decreased Bax levels compared with those of the NC group (Fig. 5). Moreover, MES23.5 cells were transfected with miR-NC and miR-21 mimic, and the miR-21 mimic significantly increased the levels of miR-21 (Fig. 6A). Furthermore, the results of Dual-luciferase reporter assay showed that cells co-transfected with miR-21 mimics and Bcl-2 3′UTR-WT, exhibited lower luciferase activity compared with those co-transfected with miR-21 mimics and Bcl-2 3′MUT (Fig. 6C). However, no significant difference was noted in the luciferase activity of the NC groups (Fig. 6C). These results confirmed that miR-21 could directly bind to the 3′UTR of the Bcl-2 protein.

Discussion

PD is characterized by progressive and irreversible loss of dopaminergic neurons in the substantia nigra. This disease is the second most common neurodegenerative disorder following Alzheimer disease (1). Current evidence suggests that miRs are involved in diverse biological processes including pathogenesis of neurodegenerative disorders and abnormal brain function (8,14). With regard to PD, several miRs have been reported to participate and play important roles in PD progression, via the regulation of various processes, such as apoptosis, autophagy, inflammation, mitochondrial dysfunction, and ROS activity (15,16). These molecules can be targeted for the development of future diagnostic tools and treatment strategies. For example, patients with PD and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced animals exhibit a significant decrease in the levels of miR-7 in brain tissue areas associated with dopaminergic neurodegeneration (17). Moreover, miR-7 has recently been shown to target α-synuclein, a protein involved in the pathological process of PD, and can therefore be used for the treatment of this disease (18).

It is important to note that the miR-21 levels of a SH-SY5Y cell PD model were significantly higher than those noted in normal cells as reported by recent studies (9,10). Su et al (10) demonstrated that miR-21 upregulated the expression levels of α-synuclein by directly targeting lysosome-associated membrane protein 2 in SH-SY5Y cells pretreated with MPP+. However, the effects and the underlying molecular mechanism of miR-21 in neural cells are still unclear. In the present study, the role and underlying molecular mechanism of miR-21 in PD was investigated using the neurotoxin MPP+, a well-established compound in PD cell models (13). It was demonstrated that treatment of MPP+ resulted in a dose- and time-dependent cytotoxicity in MES23.5 cells. The present study demonstrated that miR-21 levels were considerably increased in MPP+-treated dopaminergic neuronal MES23.5 cells, which is consistent with a previous study (10). To further investigate the role of miR-21, MES23.5 cells were transfected with miR-21 inhibitor sequences. The results suggested that downregulation of miR-21 considerably enhanced cell survival and inhibited cell apoptosis induced by MPP+ treatment. Moreover, inhibition of miR-21 significantly increased the TH positive cell number in MPP+-treated MES23.5 cells. The levels of the anti-apoptotic protein Bcl-2 and the pro-apoptotic protein Bax were also examined. These two proteins are considered important regulators of apoptosis and the disruption of the balance of the Bcl-2/Bax ratio could lead to the release of pro-apoptotic proteins from the mitochondria to the cytoplasm (19). Western blot analysis was performed to detect the expression levels of the aforementioned proteins, and the results indicated that downregulation of miR-21 increased the Bcl-2/Bax ratio in the MPP+-treated MES23.5 cells. Moreover, dual-luciferase reporter assay demonstrated that Bcl-2 was a direct target gene of miR-21, which may indicate that miR-21 participated in the induction of cell apoptosis in MES23.5 cells via the regulation of Bcl-2. In addition, MPP+ has been shown to cause neurotoxicity by the induction of ROS and the secretion of inflammatory factors (20). Devathasan et al (21) demonstrated that patients with PD exhibited a significant increase in the production of ROS in specific brain regions. The overproduction of ROS and the induction of oxidative stress have been shown to be involved in PD and to lead to dopaminergic neuronal cell death and apoptosis (22). The data reported in the present study demonstrated that downregulation of miR-21 resulted in a significant inhibition of ROS production in MPP+-induced MES23.5 cells. The results indicated that downregulation of miR-21 may exert a neuroprotective effect in MPP+-treated MES23.5 cells by the inhibition of cell apoptosis and the induction of inflammation and ROS production.

In conclusion, the present study demonstrated that miR-21 expression was increased in MPP+-treated MES23.5 cells, whereas inhibition of miR-21 alleviated MPP+-induced MES23.5 cell damage by suppressing intracellular ROS and inflammatory factor production, and by inhibiting apoptosis. Moreover, the present study reported that Bcl-2 was a direct target gene of miR-21 and that the downregulation of miR-21 could significantly increase TH expression in MPP+-treated MES23.5 cells. Although further studies should be conducted, miR-21 may be considered a potential target for PD diagnosis and treatment.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

HWM and LDD participated in experiment design, data collection and paper writing.

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 

Kalia LV and Lang AE: Parkinson's disease. Lancet. 386:896–912. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Andican G, Konukoglu D, Bozluolcay M, Bayulkem K, Firtiina S and Burcak G: Plasma oxidative and inflammatory markers in patients with idiopathic Parkinson's disease. Acta Neurol Belg. 112:155–159. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Mullin S and Schapira AH: Pathogenic mechanisms of neurodegeneration in Parkinson disease. Neurol Clin. 33:1–17. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Maiti P, Manna J and Dunbar GL: Current understanding of the molecular mechanisms in Parkinson's disease: Targets for potential treatments. Transl Neurodegener. 6:282017. View Article : Google Scholar : PubMed/NCBI

5 

Mohr AM and Mott JL: Overview of microRNA biology. Semin Liver Dis. 35:3–11. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Moss EG: MicroRNAs: Hidden in the genome. Curr Biol. 12:R138–R140. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Ma L, Wei L, Wu F, Hu Z, Liu Z and Yuan W: Advances with microRNAs in Parkinson's disease research. Drug Des Devel Ther. 7:1103–1113. 2013.PubMed/NCBI

8 

Quinlan S, Kenny A, Medina M, Engel T and Jimenez-Mateos EM: MicroRNAs in neurodegenerative diseases. Int Rev Cell Mol Biol. 334:309–343. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Fu Y, Zhen J and Lu Z: Synergetic neuroprotective effect of docosahexaenoic acid and aspirin in SH-Y5Y by inhibiting miR-21 and activating RXRalpha and PPARalpha. DNA Cell Biol. 36:482–489. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Su C, Yang X and Lou J: Geniposide reduces alpha-synuclein by blocking microRNA-21/lysosome-associated membrane protein 2A interaction in Parkinson disease models. Brain Res 1644. 98–106. 2016. View Article : Google Scholar

11 

Singer TP, Ramsay RR, McKeown K, Trevor A and Castagnoli NE Jr: Mechanism of the neurotoxicity of 1-methyl-4-phenylpyridinium (MPP+), the toxic bioactivation product of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Toxicology. 49:17–23. 1988. View Article : Google Scholar : PubMed/NCBI

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Peng J, Liu Q, Rao MS and Zeng X: Using human pluripotent stem cell-derived dopaminergic neurons to evaluate candidate Parkinson's disease therapeutic agents in MPP+ and rotenone models. J Biomol Screen. 18:522–533. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Ge X, Li W, Huang S, Yin Z, Yang M, Han Z, Han Z, Chen F, Wang H, Lei P and Zhang J: Increased miR-21-3p in injured brain microvascular endothelial cells following traumatic brain injury aggravates blood-brain barrier damage by promoting cellular apoptosis and inflammation through targeting MAT2B. J Neurotrauma. 36:1291–1305. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Oh SE, Park HJ, He L, Skibiel C, Junn E and Mouradian MM: The Parkinson's disease gene product DJ-1 modulates miR-221 to promote neuronal survival against oxidative stress. Redox Biol. 19:62–73. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Li J, Sun Y and Chen J: Identification of critical genes and miRNAs associated with the development of parkinson's disease. J Mol Neurosci. 65:527–535. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Wong G and Nass R: miRNAs and their putative roles in the development and progression of Parkinson's disease. Front Genet. 3:3152013. View Article : Google Scholar : PubMed/NCBI

18 

Titze-de-Almeida R and Titze-de-Almeida SS: miR-7 replacement therapy in parkinson's disease. Curr Gene Ther. 18:143–153. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Oltvai ZN, Milliman CL and Korsmeyer SJ: Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell. 74:609–619. 1993. View Article : Google Scholar : PubMed/NCBI

20 

Lotharius J, Dugan LL and O'Malley KL: Distinct mechanisms underlie neurotoxin-mediated cell death in cultured dopaminergic neurons. J Neurosci. 19:1284–1293. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Devathasan G, Chong PN, Puvanendran K, Lun KC and Wong PK: Low-dose bromocriptine therapy in severe Parkinson's disease. Clin Neuropharmacol. 7:231–237. 1984. View Article : Google Scholar : PubMed/NCBI

22 

Yadava N and Nicholls DG: Spare respiratory capacity rather than oxidative stress regulates glutamate excitotoxicity after partial respiratory inhibition of mitochondrial complex I with rotenone. J Neurosci. 27:7310–7317. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 18 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mao H and Mao H: Downregulation of miR‑21 suppresses 1‑methyl‑4‑phenylpyridinium‑induced neuronal damage in MES23.5 cells. Exp Ther Med 18: 2467-2474, 2019
APA
Mao, H., & Mao, H. (2019). Downregulation of miR‑21 suppresses 1‑methyl‑4‑phenylpyridinium‑induced neuronal damage in MES23.5 cells. Experimental and Therapeutic Medicine, 18, 2467-2474. https://doi.org/10.3892/etm.2019.7853
MLA
Mao, H., Ding, L."Downregulation of miR‑21 suppresses 1‑methyl‑4‑phenylpyridinium‑induced neuronal damage in MES23.5 cells". Experimental and Therapeutic Medicine 18.4 (2019): 2467-2474.
Chicago
Mao, H., Ding, L."Downregulation of miR‑21 suppresses 1‑methyl‑4‑phenylpyridinium‑induced neuronal damage in MES23.5 cells". Experimental and Therapeutic Medicine 18, no. 4 (2019): 2467-2474. https://doi.org/10.3892/etm.2019.7853