Tat‑aldose reductase prevents dopaminergic neuronal cell death by inhibiting oxidative stress and MAPK activation

  • Authors:
    • Su Bin Cho
    • Won Sik Eum
    • Min Jea Shin
    • Hyeon Ji Yeo
    • Eun Ji Yeo
    • Yeon Joo Choi
    • Hyun Jung Kwon
    • Sung‑Woo Cho
    • Jinseu Park
    • Kyu Hyung Han
    • Keun Wook Lee
    • Jong Kook Park
    • Duk‑Soo Kim
    • Dae Won Kim
    • Soo Young Choi
  • View Affiliations

  • Published online on: December 8, 2020     https://doi.org/10.3892/ijmm.2020.4812
  • Pages: 751-760
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aldose reductase (AR) is known to detoxify aldehydes and prevent oxidative stress. Although AR exerts antioxidant effects, the role of AR in Parkinson's disease (PD) remains unclear. The objective of the present study was to investigate the protective effects of AR protein against 1‑methyl‑4‑phenylpyridinium (MPP+)‑induced SH‑SY5Y cell death and 1‑methyl‑4‑phenyl‑1,2,3,6‑tetrahydropyridine (MPTP)‑induced PD in a mouse model using the cell permeable Tat‑AR fusion protein. The results revealed that when Tat‑AR protein was transduced into SH‑SY5Y cells, it markedly protected the cells against MPP+‑induced death and DNA fragmentation. It also reduced the activation of mitogen-activated protein kinase (MAPKs) and regulated the expression levels of Bcl‑2, Bax and caspase‑3. Immunohistochemical analysis revealed that when Tat‑AR protein was transduced into the substantia nigra (SN) of mice with PD, it markedly inhibited dopaminergic neuronal cell death. Therefore, Tat‑AR may be useful as a therapeutic protein for PD.

Introduction

Parkinson's disease (PD) is a common neurodegenerative disorder. It is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta with Lewy bodies in the brain (1,2). Patients with PD are clinically characterized by tremors, rigidity, bradykinesia and postural instability (3,4). Several studies have indicated that oxidative stress and environmental factors are highly associated with PD (5-7). It has been reported that oxidative stress can induce a high level of toxic aldehydes, such as 3,4-Dihydroxyphenylacetaldehyde (DOPAL) in brains of PD patients. Such aldehydes can then trigger dopaminergic neuronal cell death (5,6,8). Neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydro pyridine (MPTP) can cause PD-like symptoms. It is metabolized to 1-methyl-4-phenylpyridinium (MPP+) which can then lead to dopaminergic neuronal cell death (9,10).

Cytosolic enzyme aldose reductase (AR) is widely expressed in human tissues. It catalyzes the reduction of toxic aldehydes, including 4-hydroxy-2-nonenal (4HNE) produced by lipid peroxidation. 4HNE is known as a biomarker of oxidative stress (11-13). AR can also reduce toxic aldehyde DOPAL and induce dopaminergic neuronal cell death when this aldehyde is increased in the brain under conditions of oxidative stress (14,15). Several studies have demonstrated that AR can protect cells against oxidative stress by decreasing reactive oxygen species (ROS) levels in various cells, such as smooth muscle and lens epithelial cells (16-18).

It is known that macromolecule protein fused with protein transduction domains (PTDs) consisting of 10-16 amino acids can be transduced into cells (19). A number of studies have reported that PTD fused protein can be efficiently transduced into cells as a tool for protein therapy (20-26). In the presents study, a Tat-AR protein expression vector was constructed based on the pET-15b vector. The Tat-AR expression vector contained a cDNA sequence encoding human AR, Tat PTD and an amino-terminal tag consisting of 6 histidine residues. Tat PTD was cloned into the NdeI and XhoI sites, and human AR cDNA was cloned into the XhoI and BamHI sites of pET-15b vector. Tat-AR protein or control AR expression vector consist of a His tag consisting of 6 histidine residues. A recent study demonstrated that a deficiency of AR can lead to dopaminergic neuronal loss (27). Thus, the objective of the present study was to investigate whether transduced Tat-AR protein can protect dopaminergic neuron cells against oxidative stress both in vitro and in vivo.

Materials and methods

Cell lines and reagents

Human neuroblastoma SH-SY5Y cells were obtained from the American Type Culture Collection (ATCC) and maintained in Dulbecco's modified Eagle's medium (Lonza/BioWhittaker) containing 15% FBS, 4 mM glutamine, 100 U/ml penicillin, and 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a 5% CO2 incubator.

Dichlorofluorescein diacetate (DCF-DA), methyl-4-phenyl-pyridinium (MPP+) and 1-methyl-4-phenyl-1,2,3,6-tetrahydro pyridine (MPTP) were obtained from Sigma-Aldrich; Merck KGaA. Tat peptide was synthesized from Peptron, Inc. Enhanced chemiluminescence agent was purchased from Amersham; Cytiva. Antibodies were obtained from Cell Signaling Technology, Inc. Tyrosine Hydroxylase (TH) was purchased from Santa Cruz Biotechnology, Inc. All other chemicals and reagents, unless otherwise stated, were of the highest analytical grade available.

Purification and transduction of Tat-AR proteins into SH-SY5Y cells

Tat-AR protein was prepared as described in a previous study (28). To develop a therapeutic protein, a human AR gene fused with a Tat PTD to produce a cell permeable Tat-AR expression vector which contains 6X His, Tat PTD (RKKRRQRRR) and the AR gene. Bovine serum albumin was used as a standard and the purified Tat-AR protein concentration was measured by Bradford assay (29).

To examine the Tat-AR protein transduction efficiency, SH-SY5Y cells were exposed to various concentrations of Tat-AR and AR protein (0.5-3 µM) for 1 h. The SH-SY5Y cells were exposed of Tat-AR and AR protein (3 µM) for various periods of time (15-60 min). The cells were then washed with PBS and treated with trypsin-EDTA (Gibco; Thermo Fisher Scientific, Inc.). The intracellular stability of Tat-AR protein was also determined. To confirm the stability of Tat-AR protein, the cells were further cultured (1-30 h) following transduction. The levels of transduced proteins were measured by western blot analysis using anti-histidine antibody.

Western blot analysis

Cell lysates were prepared with RIPA lysis buffer containing a cocktail of protease inhibitors (Elpis-Biotech, Inc.). The protein concentration was measured using the Bradford method. Equal amounts of each protein (30 µg) were loaded into 12% SDS-PAGE and electrotransferred to a polyvinylidene difluoride (PVDF) membrane (Thermo Fisher Scientific, Inc.). The membrane was blocked with TBS-T (25 mM Tris-HCl, 140 mM NaCl, 0.1% Tween-20, pH 7.5) buffer containing 5% non-fat dry milk for 1 h at room temperature. After washing with TBST, the membrane was incubated with the indicated primary antibodies overnight at 4°C and followed by incubation with horseradish peroxidase-conjugated secondary antibodies for 1 h at 37°C. His (1:5,000; sc-804; Santa Cruz Biotechnology, Inc.), JNK (1:1,000; #9258), p-JNK (1:1,000; #9251), ERK (1:2,000; #9102), p-ERK (1:2,000; #4376), p38 (1:2,000; #9212), p-p38 (1:2,000; #4631), Bcl-2 (1:1,000; #2876), Bax (1:1,000; #2772), caspase-3 (1:1,000; #9662), cleaved caspase-3 (1:1,000; #9661), β-actin (1:5,000; #4967), and appropriate secondary antibodies (1:10,000; #7074). All of the above-mentioned antibodies were purchased from the Cell Signaling Technology, Inc. The membrane was then washed with TBST buffer 3 times and the protein bands were identified using chemiluminescent reagents as recommended by the manufacturer (Amersham; Cytiva). Bands were quantified using ImageJ software (version 1.48; National Institutes of Health) (25,30).

Confocal fluorescence microscopy analysis

To determine the intracellular distribution of transduced Tat-AR protein in SH-SY5Y cells, confocal fluorescence microscopy was performed, as previously described (24). SH-SY5Y cells were placed on coverslips and treated with 3 µM Tat-AR protein 1 h. The cells were washed with PBS twice and fixed with 4% paraformaldehyde for 5 min. The cells were treated in PBS containing 3% bovine serum albumin, 0.1% Triton X-100 (PBS-BT) at room temperature for 30 min and washed with PBS-BT. Histidine primary antibody (sc-804; Santa Cruz Biotechnology, Inc.) was diluted 1:1,500 and incubated at room temperature for 3 h. Alexa Fluor 488-conjugated secondary antibody (#32723; Invitrogen; Thermo Fisher Scientific, Inc.) was diluted 1:1,500 and incubated in the dark for 1 h at room temperature. Nuclei were stained with 1 µg/ml DAPI (Roche Applied Science, Mannheim, Germany) for 2 min at room temperature. The stained cells were analyzed by confocal fluorescence microscopy using a confocal laser-scanning system (Bio-Rad MRC-1024ES, Bio-Rad Laboratories, Inc.).

MTT assay

Cell viability was measured by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, as previously described (24). The SH-SY5Y cells were pre-treated with Tat-AR (0.5-3 µM), AR (0.5-3 µM) and Tat peptide (0.5-3 µM) for 1 h and MPP+ (5 mM) was then added to the culture medium for 13 h. The absorbance was measured at 570 nm using an ELISA microplate reader (Labsystems Multiskan MCC/340; Thermo Fisher Scientific, Inc.) and the cell viability was defined as the percentage of untreated control cells.

Terminal deoxynucleotidyl transferase-mediated biotinylated dUTP nick-end labelling (TUNEL) staining

To examine whether transduced Tat-AR proteins protect against MPP+-induced DNA damage in cells, the SH-SY5Y cells were pre-treated with Tat-AR (3 µM), AR (3 µM) and Tat peptide (3 µM) for 1 h and MPP+ (5 mM) was added to the culture medium for 16 h. TUNEL staining was performed using a Cell Death Detection kit (Roche Applied Science). Nuclei were stained with DAPI (1 µg/ml) for 2 min at room temperature. Fluorescent images were obtained using a fluorescence microscope (Nikon eclipse 80i, Nikon Corporation) and cells exhibiting fluorescence were counted under a phase contrast microscope (×200 magnification; Nikon Corporation), as previously described (24,25).

Measurement of ROS production

Intracellular ROS levels were determined by 2′,7′-dichlorofluorescein diacetate (DCF-DA) staining as previously described (28,31). To deter-mine the effects of Tat-AR protein against MPP+-induced intracellular ROS production in SH-SY5Y cells, the cells were placed on coverslips in 24-well plates, incubated for 12 h, and washed twice with PBS. After pretreated with Tat-AR, AR, and Tat peptide (3 µM) for 1 h, MPP+ (5 mM) was added to the culture medium for 1 h. The cells were washed with PBS and incubated for 30 min with DCF-DA (10 µM). Protein was separately processed as mentioned above to obtain the fluorescent image and fluorescence intensity. One was used to obtain a fluorescent image. To obtain fluorescent images for each well, the cells were washed with PBS, mounted, and the cell images were obtained using a fluorescence microscope (Nikon eclipse 80i, Nikon Corporation).

The other was used to obtain the fluorescence intensity. To detect the fluorescence intensity for each well, the cells were collected and washed with PBS. After added 300 µl of PBS buffer and resuspended the cells, the cells (100 µl) were transferred into 96-well plate reader. The fluorescence intensity of the samples was measured using a Fluoroskan ELISA plate reader (Labsystems Diagnostics Oy) at an excitation wavelength of 485 nm and an emission wavelength of 538 nm.

Animal experiments and immunohistochemistry

Male C57BL/6 mice (total, n=35), 8 weeks old (weighing 16-20 g), were acquired from the Hallym University Experimental Animal Center. They were housed at 23°C and a humidity of 60%. They were exposed to regulated 12 h cycles of light and dark and were provided with ad libitum access to food and water. The procedures for the care of animals conformed to the Guide for the Care and Use of Laboratory Animals of the National Veterinary Research and Quarantine Service of Korea and approved by the Institutional Animal Care and Use Committee of Soonchunhyang University (SCH16-0051).

To determine whether transduced Tat-AR protein protects against PD, the mice were divided into 5 groups (n=7/each group) as follows: The normal control, MPTP-treated, Tat-AR-treated, control AR-treated and Tat peptide-treated groups. The mice received 4 injections of MPTP (20 mg/kg) at 2-h intervals. Mice were intraperitoneally (i.p.) injected with Tat-AR (2 mg/kg) 12 h prior to MPTP treatment. At 1 week after the final injection, the animals were deeply anesthetized with a mixture of 3% isoflurane (Baxter Healthcare Corporation) in 33% oxygen and 67% nitrous oxide. The brains of these animals were then harvested for immunohistochemistry.

Immunohistochemistry was performed as described in a previous study (31). The frozen and sectioned midbrains were prepared and fixed with 4% paraformaldehyde for 10 min. For removal of non-specific immunoreactivity, free-floating sections were first incubated with 0.3% Triton X-100 and 10% normal goat serum in PBS for 1 h at room temperature. They were then incubated with a rabbit anti-tyrosine hydroxylase (TH) monoclonal antibody (diluted 1:200; sc-14007; Santa Cruz Biotechnology, Inc.) for 48 h at 4°C and sequentially incubated with a biotinylated goat anti-rabbit IgG (diluted 1:250; BA-1000; Vector Laboratories, Inc.) for 2 h at room temperature. The sections were then visualized with 3,3-diaminobenzidine (DAB) (40 mg DAB, 0.045% H2O2 in 100 ml PBS) mounted on gelatin-coated slides. To detect viable cells, cresyl violet (0.1%, Sigma-Aldrich; Merck KGaA) counterstaining for Nissl bodies was conducted for 20 min at room temperature following TH immunostaining. The sections were visualized with 3,3′-diaminobenzidine in 0.1 M Tris buffer and mounted on gelatin-coated slides. Images were captured and analyzed using an Olympus DP72 digital camera and a DP2-BSW microscope digital camera software. Figures were prepared using Adobe Photoshop 7.0. The manipulation of images was restricted to threshold and brightness adjustments applied to the entire image. The images shown are representatives from each group and the sections were processed and analyzed by a blinded observer. To establish the specificity of the immunostaining, a negative control test was carried out with pre-immune serum instead of the primary antibody. The negative control resulted in the absence of immunoreactivity in any structures.

For the quantification of TH immunostaining, a cell count was performed. TH immunostaining images (10 sections/mouse) were captured in the same region. Images were sampled from at least 5 different points within each SN section. Thereafter, the number of TH-positive cells was actu-ally counted within the sampled images. All immunoreactive cells were counted regardless the intensity of labeling. Cell counts were performed by 2 different investigators who were blind to the classification of the tissues.

Statistical analysis

Data are expressed as the means ± SEM of 3 experiments. Differences between groups were analyzed by ANOVA followed by a Bonferroni's post-hoc test (using GraphPad Prism 8; GraphPad Software, Inc.). Statistical significance was considered at P<0.05.

Results

Transduction of Tat-AR protein into SH-SY5Y cells

To produce cell-permeable Tat-AR protein, the Tat-AR protein expression vector was constructed by subcloning the cDNA encoding human AR into a pET-15b plasmid containing a Tat PTD. Tat-AR protein expression vector contained a continuous cDNA sequence encoding human AR, a Tat PTD and 6 histidines. In addition, a control AR expression vector containing no Tat PTD was constructed (Fig. 1A). The purified Tat-AR and AR proteins are presented in Fig. 1B. To investigate the transduction efficiency of Tat-AR protein, SH-SY5Y neuro-blastoma cells were treated with Tat-AR protein (0.5-3 µM) for 1 h or with Tat-AR protein at 3 µM for 15-60 min. Tat-AR protein was transduced into the cells in a concentration- and time-dependent manner (Fig. 2A and B). Tat-AR protein was also transduced into the cytosol and nucleus of the cells (Fig. 2C). Since stability is one of the major factors in protein therapy, the stability of the transduced Tat-AR protein was examined. Transduced Tat-AR protein persisted until 12 h in the cells (Fig. 2D). These results indicate that Tat-AR protein can be efficiently transduced into the SH-SY5Y cells and can exist for at least 12 h in the cells.

Effects of Tat-AR protein against MPP+-induced SH-SY5Y cells

It is known that MPP+ can induce ROS production in dopaminergic neuronal cells, and can cause DNA damage and cell death (32). Thus, the protective effects of transduced Tat-AR protein against MPP+-induced cell death were examined. Cell viability was 53% in the cells exposed only to MPP+. However, cell viability was markedly increased up to 84% in the cells treated with Tat-AR protein following exposure to MPP+. By contrast, AR protein and Tat peptide failed to prevent cell death under the same experimental conditions (Fig. 3A).

The protective effects of Tat-AR protein against MPP+-induced DNA damage and intracellular ROS production were also determined by TUNEL and DCF-DA staining, respectively (Fig. 3B and C). DNA damage and intracellular ROS production levels were increased in the cells exposed only to MPP+. However, DNA damage and intracellular ROS production levels were markedly inhibited in the Tat-AR protein-treated cells following MPP+ exposure. However, AR protein and Tat peptide failed to inhibit DNA damage and intracellular ROS production. These results indicate that Tat-AR protein can inhibit SH-SY5Y cell death by decreasing DNA damage and intra-cellular ROS production, functioning as an antioxidant in the cells.

Effects of Tat-AR protein on MPP+-induced signaling pathways

MPP+ can trigger mitogen-activated protein kinase (MAPK) signaling pathway activation (9,33,34). Thus, the effects of Tat-AR proteins on MPP+ induced MAPKs signaling pathways were determined in the present study. In the SH-SY5Y cells exposed to MPP+, the expression levels of phosphorylated MAPKs were higher than those in the control cells. By contrast, Tat-AR protein significantly decreased expression the levels of phosphorylated MAPKs in the cells exposed to MPP+. However, the expression levels of phosphorylated MAPKs in the cells treated with AR protein or Tat peptide were similar to those in the untreated cells exposed to MPP+ (Fig. 4).

It is well known that the neurotoxin, MPP+, can cause the overproduction of ROS in cells and activate apoptosis signaling pathways (32). Thus, effects of Tat-AR proteins on the MPP+-induced expression levels of Bax, Bcl-2 and caspase 3 were investigated. As shown in Fig. 5A, the Bcl-2 expression levels were decreased in the MPP+-exposed SH-SY5Y cells. By contrast, Tat-AR protein significantly increased the Bcl-2 expression levels in cells the exposed to MPP+. However, the expression levels of Bax exhibited opposite results from those of Bcl-2. Tat-AR protein also markedly increased the expression of caspase-3 in the MPP+-exposed cells. The cleaved caspase-3 expression levels were significantly decreased in the cells exposed only to MPP+. In addition, Tat-AR protein reduced the cleaved Caspase-3/Caspase-3 ratio in MPP+ treated cells. AR protein and Tat peptide failed to affect the expression levels of caspase-3 and cleaved caspase-3 proteins induced by MPP+ (Fig. 5B). These results indicate that transduced Tat-AR protein can prevent SH-SY5Y cell death from MPP+ by regulating phosphorylation levels of MAPKs and apoptosis-related protein expression.

Protective effects of Tat-AR protein against MPTP-induced cell death in an animal model of PD

After Tat-AR protein (2 mg/kg) was i.p. injected into the mice, immunohistochemistry was performed using histidine antibody to determine whether Tat-AR protein was transduced into the mouse brains (Fig. 6A). Tat-AR protein was highly expressed in the SN region of the midbrain. However, AR protein was not detected in the SN region of the midbrain. These results indicate that Tat-AR protein can be transduced into mouse brains by crossing the blood-brain barrier (BBB).

Dopaminergic neuronal cell death is major indicator of PD. Thus, the protective effects of Tat-AR protein against MPTP-induced dopaminergic neuronal cell death were determined by immunohistochemistry using a TH antibody and cresyl violet staining. As shown in Fig. 6B and C, the TH-positive cell numbers were increased in the SN of the Tat-AR protein-treated group. In addition, neuronal cell survival was markedly increased in the Tat-AR protein-treated group. By contrast, the AR protein- and Tat peptide-treated groups did not exhibit such protective effects. These results indicate that Tat-AR protein can markedly prevent dopaminergic neuronal cell death.

Discussion

PD is a progressive neurodegenerative disorder, characterized by the loss of dopamine neurons in the SN pars compacta and the presence of Lewy bodies in the brain (1-3). Since oxidative stress is highly associated with the pathogenesis of various diseases, including PD, the regulation of oxidative stress is crucial to preventing these diseases (5-7,35-39). The definitive cause of dopamine neuron cell death in PD and precise etiological mechanism remain unclear.

AR can reduce levels of toxic aldehydes, including 4HNE. It can increase cell survival by decreasing oxidative stress and toxic aldehydes (11-15). Although several studies have suggested that AR plays a crucial role in cellular responses to oxidative stress by detoxifying ROS and decreasing reactive aldehydes in keratinocytes (40) and by neutralizing the toxicity of lipid peroxidation in arterial wall injuries (41), the precise protective role of AR in dopaminergic cells against oxidative stress remains poorly understood. Therefore, the present study investigated whether Tat-AR protein can protect SH-SY5Y cells against MPP+-induced dopaminergic neuronal cell death whether it can protect animals against MPTP-induced PD. The results revealed that purified Tat-AR protein could be transduced into SH-SY5Y cells in a time- and dose-dependent manner, and that such a transduced protein could significantly enhance cell survival and inhibit DNA damage in MPP+-exposed SH-SY5Y cells.

It is well known that neurotoxin MPP+ can induce oxidative stress and lead to dopaminergic neuronal death via mitochondrial damage (9,10,32). Other studies have demonstrated that the overexpression of AR protein can protect cells against methylglyoxal (MG)-induced aortic smooth muscle cell damage (42) and prevent aldehyde-induced human lens epithelial cell death (18). AR is also involved in cell survival as a detoxification enzyme (43). The results of the present study demonstrated the same protective pattern. Further studies are required to confirm such results.

MPP+ can trigger cellular signaling pathways, including MAPKs and apoptosis signaling pathways (32). Thus, the effects of Tat-AR protein on MAPK signaling pathways, and the expression levels of Bax, Bcl-2 and caspase-3 were determined in the present study. In the MPP+-exposed SH-SY5Y cells, Tat-AR protein markedly inhibited Bax and cleaved caspase-3 expression levels, but significantly increased the Bcl-2 expression levels. Other studies have also demonstrated that the overexpression of AR can increase the Bcl-2 level, but can decrease Bax expression levels and the phosphorylation level of JNK and p38 in aldehyde-induced human lens epithelial cells (18).

In addition, AR can reduce ultraviolet-B (UVB)-induced JNK and p38 phosphorylation in HaCaT cells (40). The phosphorylation levels of EKR1/2 have been shown to be significantly increased in MPTP-exposed AR−/− mouse brains (27). These reports suggest that dopaminergic neuronal loss is associated with the phosphorylation of EKR1/2 and that AR plays an important role in protecting dopaminergic neurons in PD (27,40). The results of the present study also demonstrated that Tat-AR protein reduced the activation of MAPKs in MPP+-exposed SH-SY5Y cells, suggesting that Tat-AR protein plays a protective role against MPP+-induced dopaminergic neuronal cell death via the modulation of MAPKs and apoptosis signaling pathways.

The MPTP-induced mouse model of PD has been generally used to examine the pathological mechanisms of PD as this model exhibits a similar pathophysiology to human PD (44,45). In animal models of MPTP-induced PD, Tat-AR protein was shown to significantly protect dopaminergic neuronal cell death in the present study. Goldstein et al previously demonstrated that the overexpression of AR protein significantly reduced cell death in patients with PD (8). In addition, a number of studies have reported that the levels of toxic aldehyde DOPAL are markedly increased in patients with PD and that AR can decrease DOPAL levels (5,6,8), suggesting that AR plays a pivotal role in detoxifying DOPAL in PD (14,15,46). A recent study demonstrated that AR deficiency in MPTP-exposed mice led to increases in the characteristics of PD and that AR protected dopaminergic neurons against neurotoxic metabolites (27).

In conclusion, the presents study demonstrated that Tat-AR protein could be transduced into SH-SY5Y cells and SN in mouse brains, and that Tat-AR protein could significantly protect against MPP+- and MPTP-induced dopaminergic neuronal cell death both in vitro and in vivo. Although further studies are required to elucidate the precise protective mechanisms, the present results suggest that Tat-AR protein may be useful as a therapeutic agent for PD.

Funding

The present study was supported by the National Research Foundation of Korea (NRF) grant funded the Korea government (MIST) (NRF-2018R1A2B6001941) and in part by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2019R1A6A1A11036849).

Availability of data and materials

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

Authors' contributions

SBC, WSE, MJS, DWK and SYC were involved in the conceptualization of the study. SBC, WSE, MJS, HJK and DSK were involved in the study methodology. HJY, EJY and YJC were involved in data validation. SWC, JP, KHH, KWL, JKP and SYC were involved in data curation. DWK and SYC were involved in the writing and editing of the manuscript and provided final approval of the version to be published. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The procedures for the care of animals conformed to the Guide for the Care and Use of Laboratory Animals of the National Veterinary Research and Quarantine Service of Korea and approved by the Institutional Animal Care and Use Committee of Soonchunhyang University (SCH16-0051).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

de Rijk MC, Launer LJ, Berger K, Breteler MM, Dartigues JF, Baldereschi M, Fratiglioni L, Lobo A, Martinez-Lage J, Trenkwalder C and Hofman A: Prevalence of Parkinson's disease in Europe: A collaborative study of population-based cohorts. Neurology. 54(11 Suppl 5): S21–S23. 2000.PubMed/NCBI

2 

Braak H, Del Tredici K, Rüb U, de Vos RA, Jansen Steur EN and Braak E: Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging. 24:197–211. 2003. View Article : Google Scholar

3 

Calne D: A definition of Parkinson's disease. Parkinsonism Relat Disord. 11(Suppl 1): S39–S40. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Hughes AJ, Daniel SE and Lees AJ: Improved accuracy of clinical diagnosis of Lewy body Parkinson's disease. Neurology. 57:1497–1499. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Fitzmaurice AG, Rhodes SL, Lulla A, Murphy NP, Lam HA, O'Donnell KC, Barnhill L, Casida JE, Cockburn M, Sagasti A, et al: Aldehyde dehydrogenase inhibition as a pathogenic mechanism in Parkinson disease. Proc Natl Acad Sci USA. 110:636–641. 2013. View Article : Google Scholar

6 

Koutsilieri E, Scheller C, Grunblatt E, Nara K, Li J and Riederer P: Free radicals in Parkinson's disease. J Neurol. 249(Suppl 2): II1–II5. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Drechsel DA and Patel M: Role of reactive oxygen species in the neurotoxicity of environmental agents implicated in Parkinson's disease. Free Radic Biol Med. 44:1873–1886. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Goldstein DS, Sullivan P, Holmes C, Kopin IJ, Basile MJ and Mash DC: Catechols in post-mortem brain of patients with Parkinson disease. Eur J Neurol. 18:703–710. 2011. View Article : Google Scholar

9 

Singer TP and Ramsay RR: Mechanism of the neurotoxicity of MPTP: An update. FEBS Lett. 274:1–8. 1990. View Article : Google Scholar : PubMed/NCBI

10 

Goping G, Pollard HB, Adeyemo OM and Kuijpers GA: Effect of MPTP on dopaminergic neurons in the goldfish brain: A light and electron microscope study. Brain Res. 687:35–52. 1995. View Article : Google Scholar : PubMed/NCBI

11 

O'Connor T, Ireland LS, Harrison DJ and Hayes JD: Major differences exist in the function and tissue-specific expression of human B1 aldehyde reductase and the principal human aldoketo reductase AKR1 family members. Biochem J. 343:487–504. 1999. View Article : Google Scholar

12 

Esterbauer H, Schaur RJ and Zollner H: Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med. 11:81–128. 1991. View Article : Google Scholar : PubMed/NCBI

13 

Doorn JA and Petersen DR: 4-Hydroxynonenal-mediated inhibition of enzyme catalyzed oxidation of the reactive electrophile 3,4-dihydroxyphenylacetaldehyde. Toxicol Sci. 78:9802004.

14 

Burke WJ, Li SW, Chung HD, Ruggiero DA, Kristal BS, Johnson EM, Lampe P, Kumar VB, Franko M, Williams EA and Zahm DS: Neurotoxicity of MAO metabolites of catechol-amine neurotransmitters: Role in neurodegenerative diseases. Neurotoxicol. 25:101–115. 2004. View Article : Google Scholar

15 

Burke WJ, Li SW, Williams EA, Nonneman R and Zahm DS: 3,4-Dihydroxyphenyl-acetaldehyde is the toxic dopamine metabolite in vivo: Implications for Parkinson's disease pathogenesis. Brain Res. 989:205–213. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Srivastava SK, Yadav UC, Reddy AB, Saxena A, Tammali R, Shoeb M, Ansari NH, Bhatnagar A, Petrash MJ, Srivastava S and Ramana KV: Aldose reductase inhibition suppresses oxidative stress-induced inflammatory disorders. Chem Biol Interact. 191:330–338. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Lyon RC, Li D, McGarvie G and Ellis EM: Aldo-keto reductases mediate constitutive and inducible protection against aldehyde toxicity in human neuroblastoma SH-SY5Y cells. Neurochem Int. 62:113–121. 2013. View Article : Google Scholar

18 

Pladzyk A, Ramana KV, Ansari NH and Srivastava SK: Aldose reductase prevents aldehyde toxicity in cultured human lens epithelial cells. Exp Eye Res. 83:408–416. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Moon JI, Han MJ, Yu SH, Lee EH, Kim SM, Han K, Park CH and Kim CH: Enhanced delivery of protein fused to cell penetrating peptides to mammalian cells. BMB Rep. 52:324–329. 2019. View Article : Google Scholar :

20 

van den Berg A and Dowdy SF: Protein transduction domain delivery of therapeutic macromolecules. Curr Opin Biotechnol. 22:888–893. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Dietz GP: Cell-penetrating peptide technology to deliver chaper-ones and associated factors in diseases and basic research. Curr Pharm Biotechnol. 11:167–174. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Gump JM and Dowdy SF: TAT transduction: The molecular mechanism and therapeutic prospects. Trends Mol Med. 13:443–448. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Kim DW, Shin MJ, Choi YJ, Kwon HJ, Lee SH, Lee S, Park J, Han KH, Eum WS and Choi SY: Tat-ATOX1 inhibits inflamma-tory responses via regulation of MAPK and NF-κB pathways. BMB Rep. 51:654–659. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Shin MJ, Kim DW, Lee YP, Ahn EH, Jo HS, Kim DS, Kwon OS, Kang TC, Cho YJ, Park J, et al: Tat-glyoxalase protein inhibits against ischemic neuronal cell damage and ameliorates ischemic injury. Free Radic Biol Med. 67:195–210. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Shin MJ, Kim DW, Jo HS, Cho SB, Park JH, Lee CH, Yeo EJ, Choi YJ, Kim JA, Hwang JS, et al: Tat-PRAS40 prevent hippo-campal HT-22 cell death and oxidative stress induced animal brain ischemic insults. Free Radic Biol Med. 97:250–262. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Yeo HJ, Shin MJ, You JH, Kim JS, Kim MY, Kim DW, Kim DS, Eum WS and Choi SY: Transduced Tat-CIAPIN1 reduces the inflammatory response on LPS- and TPA-induced damages. BMB Rep. 52:695–699. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Yeung PKK, Lai AKW, Son HJ, Zhang X, Hwang O, Chung SSM and Chung SK: Aldose reductase deficiency leads to oxidative stress-induced dopaminergic neuronal loss and autophagic abnormality in an animal model of Parkinson's disease. Neurobiol Aging. 50:119–133. 2017. View Article : Google Scholar

28 

Cho SB, Eum WS, Shin MJ, Kwon HJ, Park JH, Choi YJ, Park J, Han KH, Kang JH, Kim DS, et al: Transduced Tat-aldose reductase protects hippocampal neuronal cells against oxidative stress-induced damage. Exp Neurobiol. 28:612–627. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Bradford M: A rapid and sensitive method for the quantification of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 72:248–254. 1976. View Article : Google Scholar : PubMed/NCBI

30 

Han AR, Yang JW, Na JM, Choi SY and Cho SW: Protective effects of N,4,5-trimethylthiazol-2-amine hydrochloride on hypoxia-induced β-amyloid production in SH-SY5Y cells. BMB Rep. 52:439–444. 2019. View Article : Google Scholar :

31 

Ahn EH, Kim DW, Shin MJ, Kim YN, Kim HR, Woo SJ, Kim SM, Kim DS, Kim J, Park J, et al: PEP-1-ribosomal protein S3 protects dopaminergic neurons in an MPTP-induced Parkinson's disease mouse model. Free Radic Biol Med. 55:36–45. 2013. View Article : Google Scholar

32 

Kalivendi SV, Kotamraju S, Cunningham S, Shang T, Hillard CJ and Kalyanaraman B: 1-methyl-4-phenylpyridinium (MPP+)-induced apoptosis and mitochondrial oxidant generation: Role of transferrin-receptor-dependent iron and hydrogen peroxide. Biochem J. 371:151–164. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Chongthammakun V, Sanvarinda Y and Chongthammakun S: Reactive oxygen species production and MAPK activation are implicated in tetrahydrobiopterin induced SH-SY5Y cell death. Neurosci Lett. 449:178–182. 2009. View Article : Google Scholar

34 

Zhu JH, Kulich SM, Oury TD and Chu CT: Cytoplasmic aggre-gates of phosphorylated extracellular signal-regulated protein kinases in Lewy body diseases. Am J Pathol. 161:2087–2098. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Romuk EB, Szczurek W, Oles M, Gabrysiak A, Skowron M, Nowak P and Birkner E: The evaluation of the changes in enzy-matic antioxidant reserves and lipid peroxidation in chosen parts of the brain in an animal model of Parkinson disease. Adv Clin Exp Med. 26:953–959. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Wang YL, Ju B, Zhang YZ, Yin HL, Liu YJ, Wang SS, Zeng ZL, Yang XP, Wang HT and Li JF: Protective effect of curcumin against oxidative stress-induced injury in rat with Parkinson's disease through the Wnt/β-catenin signaling pathway. Cell Physiol Biochem. 43:2226–2241. 2017. View Article : Google Scholar

37 

Madamanchi NR, Vendrov A and Runge MS: Oxidative stress and vascular disease. Arterioscler Thromb Vasc Biol. 25:29–38. 2005. View Article : Google Scholar

38 

Andersen JK: Oxidative stress in neurodegeneration: Cause or consequence? Nat Med. 10:S18–S25. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Cooke MS, Evans MD, Dizdaroglu M and Lunec J: Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J. 17:1195–1214. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Kang ES, Iwata K, Ikami K, Ham SA, Kim HJ, Chang KC, Lee JH, Kim JH, Park SB, Km JH, et al: Aldose reductase in kera-tinocytes attenuates cellular apoptosis and senescence induced by UV radiation. Free Radic Biol Med. 50:680–688. 2011. View Article : Google Scholar

41 

Rittner HL, Hafner V, Klimiuk PA, Szweda LI, Goronzy JJ and Weyand CM: Aldose reductase functions as a detoxification system for lipid peroxidation products in vasculitis. J Clin Invest. 103:1007–1013. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Yabe-Nishimura C, Nishinaka T, Iwata K and Seo HG: Up-regulation of aldose reductase by the substrate, methyglyoxal. Chem Biol Interact. 143-144:317–323. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Kang ES, Woo IS, Kim HJ, Eun SY, Paek KS, Kim HJ, Chang KC, Lee JH, Lee HT, Kim JH, et al: Up-regulation of aldose reductase expression mediated by phosphatidylinositol 3-kinase/Akt and Nrf2 is involved in the protective effect of curcumin against oxidative stress. Free Radic Biol Med. 43:535–545. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Camicioli R, Grossmann SJ, Spencer PS, Hudnell K and Anger WK: Discriminating mild parkinsonism: Methods for epidemiological research. Mov Disord. 16:33–40. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Park SW, Kim SH, Park KH, Kim SD, Kim JY, Baek SY, Chung BS and Kang CD: Protective effect of antioxidants in MPTP-induced mouse model of Parkinson's disease. Neurosci Lett. 363:243–246. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Goldstein DS, Sullivan P, Cooney A, Jinsmaa Y, Sullivan R, Gross DJ, Holmes C, Kopin IJ and Sharabi Y: Vesicular uptake blockade generates the toxic dopamine metabolite 3,4- dihydroxyphenylacetaldehyde in PC12 cells: Relevance to the pathogenesis of Parkinson's disease. J Neurochem. 123:932–943. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 47 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cho SB, Eum WS, Shin MJ, Yeo HJ, Yeo EJ, Choi YJ, Kwon HJ, Cho SW, Park J, Han KH, Han KH, et al: Tat‑aldose reductase prevents dopaminergic neuronal cell death by inhibiting oxidative stress and MAPK activation. Int J Mol Med 47: 751-760, 2021
APA
Cho, S.B., Eum, W.S., Shin, M.J., Yeo, H.J., Yeo, E.J., Choi, Y.J. ... Choi, S.Y. (2021). Tat‑aldose reductase prevents dopaminergic neuronal cell death by inhibiting oxidative stress and MAPK activation. International Journal of Molecular Medicine, 47, 751-760. https://doi.org/10.3892/ijmm.2020.4812
MLA
Cho, S. B., Eum, W. S., Shin, M. J., Yeo, H. J., Yeo, E. J., Choi, Y. J., Kwon, H. J., Cho, S., Park, J., Han, K. H., Lee, K. W., Park, J. K., Kim, D., Kim, D. W., Choi, S. Y."Tat‑aldose reductase prevents dopaminergic neuronal cell death by inhibiting oxidative stress and MAPK activation". International Journal of Molecular Medicine 47.2 (2021): 751-760.
Chicago
Cho, S. B., Eum, W. S., Shin, M. J., Yeo, H. J., Yeo, E. J., Choi, Y. J., Kwon, H. J., Cho, S., Park, J., Han, K. H., Lee, K. W., Park, J. K., Kim, D., Kim, D. W., Choi, S. Y."Tat‑aldose reductase prevents dopaminergic neuronal cell death by inhibiting oxidative stress and MAPK activation". International Journal of Molecular Medicine 47, no. 2 (2021): 751-760. https://doi.org/10.3892/ijmm.2020.4812