Adenosine decreases oxidative stress and protects H2O2‑treated neural stem cells against apoptosis through decreasing Mst1 expression

  • Authors:
    • Masoumeh Gholinejad
    • Iraj Jafari Anarkooli
    • Amirhossein Taromchi
    • Alireza Abdanipour
  • View Affiliations

  • Published online on: March 27, 2018     https://doi.org/10.3892/br.2018.1083
  • Pages: 439-446
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Overproduction of free radicals during oxidative stress induces damage to key biomolecules and activates programed cell death pathways. Neuronal cell death in the nervous system leads to a number of neurodegenerative diseases. The aim of the present study was to evaluate the neuroprotective effect of adenosine on inhibition of apoptosis induced by hydrogen peroxide (H2O2) in bone marrow‑derived neural stem cells (B‑dNSCs), with focus on its regulatory effect on the expression of mammalian sterile 20‑like kinase 1 (Mst1), as a novel proapoptotic kinase. B‑dNSCs were exposed to adenosine at different doses (2, 4, 6, 8 and 10 µM) for 48 h followed by 125 µM H2O2 for 30 min. Using MTT, terminal deoxynucleotidyl transferase dUTP nick‑end labeling and real‑time reverse transcription polymerase chain reaction assays, the effects of adenosine on cell survival, apoptosis and Mst1, nuclear factor (erythroid‑derived 2)‑like 2 and B‑cell lymphoma 2 and adenosine A1 receptor expression were evaluated in pretreated B‑dNSCs compared with controls (cells treated with H2O2 only). Firstly, results of the MTT assay indicated 6 µM adenosine to be the most protective dose in terms of promotion of cell viability. Subsequent assays using this dosage indicated that apoptosis rate and Mst1 expression in B‑dNSCs pretreated with 6 µM adenosine were significantly decreased compared with the control group. These findings suggest that adenosine protects B‑dNSCs against oxidative stress‑induced cell death, and therefore, that it may be used to promote the survival rate of B‑dNSCs and as a candidate for the treatment of oxidative stress‑mediated neurological diseases.

Introduction

Numerous studies have described that oxidative stress, induced by reactive oxygen species (ROS) and mitochondrial dysfunction, is implicated in the etiology of neurodegenerative disorders (13). Adenosine is a naturally occurring nucleoside that is used to control heart arrhythmia (4). Adenosine is a neuromodulator that suppresses neuronal excitability through activation of the inhibitory adenosine A1 receptor (AA1R) (5). Therefore, AA1R has therapeutic potential as a neuroprotective candidate.

Apoptosis, a form of programmed cell death, serves an important role in neurodegenerative disorders (6). Studies on apoptosis and its role in neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease and ischemia have elucidated the possible apoptotic mechanisms underlying these disorders (79). Apoptosis is regulated by the expression or activation of proapoptotic genes and proteins including mammalian sterile 20-like kinase 1 (Mst1) (10). Mst1 is a stress-activated, proapoptotic kinase that, following caspase-mediated cleavage, enters the nucleus and induces chromatin condensation followed by DNA fragmentation (11,12). Mst1 protein has been reported to induce the mitochondrial-dependent pathway of apoptosis as well as promote apoptosis via caspase-dependent and -independent pathways (13,14).

In the present study, the in vitro neuroprotective effects of adenosine in inhibiting apoptosis and promoting cell survival were investigated in bone marrow-derived neural stem cells (B-dNSCs) preexposed to hydrogen peroxide (H2O2). The effects of adenosine on expression of proapoptotic Mst1 and antiapoptotic nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and B-cell lymphoma 2 (Bcl-2) genes in the H2O2-induced B-dNSCs were evaluated. Recent studies have demonstrated that Nrf1, Nrf2 and Bcl-2 family coactivators control mitochondrial transcription specificity factors (15,16). Furthermore, Bcl-2 and Nrf2 are established as critical factors in protecting cells against H2O2-induced apoptosis (17). The results from this study indicated that apoptosis rate and Mst1 expression in the adenosine-treated B-dNSCs were significantly decreased while Nrf2 and AAIR mRNA levels were increased. The present study aimed to develop understanding of the neuroprotective effects of adenosin in neurological disorders.

Materials and methods

Bone marrow-derived stromal cell (BMSC) isolation and culture

In the present study, 5 male Wistar rats (weighing 150–200 g, aged 8 weeks old), purchased from Razi Vaccine and Serum Research Institute (Karaj, Iran), were sacrificed under anesthesia with 100 mg/kg ketamine and 10 mg/kg xylazine. All experimental protocols were approved by the Zanjan University of Medical Sciences (Zanjan, Iran) Ethics Committee. All rats were housed in a temperature (25–27°C) and humidity (~50%) controlled environment under a 12-h light/dark cycle. The rat tibia and femoral bone marrow were aseptically aspirated with 2 ml Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and then centrifuged for 5 min at 1,500 × g and 4°C. The supernatant was removed, 1 ml fresh media was added and repeated pipetting was performed to create a single cell suspension. Subsequently, cells were transferred to a 25-cm2 flask for tissue culture with 5 ml low-glucose DMEM containing 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 100 U/ml penicillin and 100 mg/ml streptomycin (Sigma-Aldrich; Merck KGaA). The isolated cells were incubated at 37°C in 5% CO2 for 2 days. The adherent cells were collected and subcultured, and the culture medium was replaced every 2–3 days until cells became 70–80% confluent.

The cells were harvested with trypsin-EDTA (0.25%; Sigma-Aldrich; Merck KGaA) and passaged up to three times (P3). For identifying BMSCs, immunocytochemical evaluation was performed for cluster of differentiation (CD)-90 as a mesenchymal stem cell marker (18). Briefly, BMSCs were cultured on cover slides and fixed in 3% paraformaldehyde for 15 min at room temperature (RT), then permeabilized with 0.4% Triton X-100. Blocking was performed in 10% FBS in phosphate-buffered saline (PBS) for 30 min at RT. The cells were then incubated with anti-CD90 monoclonal antibodies (ab225; 1:200; Abcam, Cambridge, UK) overnight at 4°C, followed by incubation with a fluorescein isothiocyanate (FITC)-conjugated rabbit anti-rat antibody (ab6730; 1:300; Abcam) for 4 h at RT. Nuclei were counterstained with ethidium bromide for 30 sec at RT.

Neurosphere formation and expansion

To form neurosphere-like structures, isolated BMSCs at P3 were seeded (1×105 cells/ml) in 25-cm2 non-adherent plastic flasks and incubated with NSC expansion medium containing DMEM/F12 supplemented with 2% B27 (Gibco; Thermo Fisher Scientific, Inc.), 20 ng/ml basic fibroblast growth factor (bFGF; Invitrogen; Thermo Fisher Scientific, Inc.), 20 ng/ml epidermal growth factor (EGF; Invitrogen; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin. The medium and growth factors were added every 3 days for a week. For the preparation of a single cells from the neurospheres, the floating structures were isolated by centrifuging for 5 min at 300 × g and 4°C.

They were then dissociated enzymatically using Trypsin-EDTA (0.25%) and mechanically (by pipetting) to single cells, and subsequently expanded on 6-well adherent plates coated with poly-l-lysine (Sigma-Aldrich; Merck KGaA). The cells (105 cells/well) were then suspended in DMEM/F12 supplemented with 2% B27, 20 ng/ml bFGF and 20 ng/ml EGF and 5% FBS and passaged up to three times (19). For identifying B-dNSCs, immunocytochemical evaluation was performed for nestin as an NSC/progenitor cell marker (20) with corresponding antibody (ab6142; 1:300; Abcam) as above. The FITC-conjugated rabbit anti-rat antibody (ab6730; 1:300; Abcam) was used for detection (4 h at RT in the dark). Ethidium bromide (20 sec) was used for nuclei counterstaining at RT. Images were captured with an Olympus BX51 fluorescence microscope (Olympus Corporation, Tokyo, Japan).

Adenosine dose response

B-dNSC viability and the protective effects of adenosine were evaluated using the 3-(4, 5-dimethythiazol-2-yl)-2, 5-diphenyl-tetrazolium (MTT) bromide assay (21). B-dNSCs were cultured in 96-well plates (105 cells/well) in NSC expansion medium. The cells were then incubated with different concentrations of adenosine (0, 2, 4, 6, 8 and 10 µM) at 37°C for 48 h. To induce oxidative stress, H2O2 was prepared from a 30% stock solution. The pretreated NSCs were incubated with 125 µM H2O2 for 30 min at 37°C. Subsequently, the cells were incubated with 1 mg/ml MTT at 37°C for 4 h. The culture medium was removed and 100 µl dimethyl sulfoxide was added to each well to dissolve the formazan crystals. The amount of formazan dissolved was quantified at absorbance (A)570 nm using a microplate ELISA reader. The relative cell viability in percentage was calculated as (A570 of treated samples/A570 of untreated samples) × 100 (22).

Experimental groups

Based on results of the MTT assay, cells were assigned to different experimental groups as follows: N (untreated B-dNSCs), NA (B-dNSCs treated with 6 µM adenosine), NH (B-dNSCs treated with 125 µM H2O2) and NAH (B-dNSCs pretreated with 6 µM adenosine followed by 125 µM H2O2).

Assessment of apoptosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining. Cells were fixed with 4% paraformaldehyde in PBS for 30 min at room temperature (RT). DNA fragmentation was assessed with an In-Situ Cell Death Detection kit (Roche Diagnostics GmbH, Mannheim, Germany) according to the manufacturer's instructions. TUNEL-positive cells were colored using diaminobenzidine as the chromogen for 5 min at RT, and counterstained with hematoxylin for 1 min at RT. The percentage of TUNEL-positive cells was assessed in five randomly selected microscopic fields of the Olympus BX51 fluorescence microscope per culture well.

Immunofluorescence staining

Cells were cultured on cover slides at a density of 1×105 cells/ml in NSC growth media as described above and fixed in 4% paraformaldehyde for 15 min at RT, followed by permeabilization in PBS-0.1% Triton X-100 for 30 min at RT. Blocking was performed in 10% FBS in PBS for 30 min at RT. For immunofluorescence staining, cells were incubated with primary polyclonal antibodies against rat CD90 (ab225; 1:200), nestin (ab6142; 1:300; both from Abcam), Nrf2 (sc-722; 1:1,200) and Mst1 (sc-100449; 1:300; both from Santa Cruz Biotechnology, Inc., Dallas, TX, USA) individually overnight at 4°C, then incubated with a fluorescein isothiocyanate-conjugated rabbit anti-rat antibody (ab6730; 1:300; Abcam) for 4 h at RT in the dark. DAPI (5 min) and ethidium bromide (20 sec) were used for nuclei counterstaining at RT. Images were captured with the Olympus BX51 fluorescence microscope.

Real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Real-time RT-qPCR was performed with cDNA from the experimental groups following treatments. In all groups, 1,000 ng purified RNA extracted using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) from cultured cells was used to synthesize 20 µl cDNA, using a RevertAid™ First Strand cDNA Synthesis kit (Fermentas; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. cDNA (25 ng of RNA samples) was used to quantify Bcl-2, Nrf2, Mst1 and AA1R mRNA levels. As an internal control, primers for GAPDH were used. The primer sequences of all primers used are listed in Table I. The PCR reaction was performed in a 12.5-µl final volume containing forward and reverse primers (200 nM each), cDNA (0.5 µl), SYBR®-Green I (6.5 µl; Fermentas; Thermo Fisher Scientific, Inc.) and nuclease-free water up to final volume for 40 cycles (Applied Biosystems 7500; Thermo Fisher Scientific, Inc.) at 95°C for 15 sec followed by 60°C for 1 min. For analyzing relative changes in mRNA levels, the 2−ΔΔCq method was employed (23).

Table I.

PCR primer sequences.

Table I.

PCR primer sequences.

GeneGenBank accession no.Forward, 5′-3′Reverse, 5′-3′Product size, bp
Mst1NM_001107800.1 GCTAAAGTGAAGTGGACGGATACC GGAACAGTTGCTACCAGAGTGTCAG173
Nrf2NM_031789.2 CACCAGTGGATCTGTCAGCTACTC GTGGTGAAGACTGAGCTCTCAACG168
Bcl-2NM_016993.1 GTGGCCTTCTTTGAGTTCGGTG ATCCCAGCCTCCGTTATCCTG147
GAPDHNM_017008 AACCCATCACCATCTTCCAG GTGGTTCACACCCATCACAA197
AA1RXM_006249861 CCACAGACCTACTTCCACACC CTGTCTTGTACCGGAGAGGGA136

[i] Primers were designed with Gene Runner 3.05 software (http://generunner.net; product by info@genfanavaran.com). Mst1, mammalian sterile 20-like kinase 1; Nrf2, nuclear factor (erythroid-derived 2)-like 2; Bcl-2, B-cell lymphoma 2; AA1R, adenosine A1 receptor.

Statistical analysis

Statistical analysis of data was performed using SPSS 15.0 software (SPSS, Inc., Chicago, IL, USA) based on a minimum of three independent experiments. The results were normalized with those of the experimental controls, and were presented as the mean percentage ± standard error of the mean and analyzed by one-way analysis of variance followed by the Tukey's post hoc multiple group comparison test. A difference between groups was considered statistically significant when P<0.05.

Results

Isolation and culture of BMSCs

The results demonstrated that, after a 24-h culture, BMSCs successfully attached to the dish surface. When adhered, the BMSCs exhibited fibroblast-like or triangular shape (Fig. 1A). Following replacement of the medium, the majority of non-adherent cells were eliminated and adherent cells gradually proliferated. After 3–5 days, adherent cells formed clones (Fig. 1B). At 5 days the clones enlarged and fused with other clones. After 10 days, the adherent cells became confluent and could be subcultured. At 2 h after subculture the majority of BMSCs adhered, and after 3–4 days cells were confluent. The BMSCs were identified to express CD90, which served to identify cells as MSCs (Fig. 1C and D), at a positive expression rate of 97.30±0.68% (data not shown).

Induction of NSCs

Following culture in neural stem cell expansion medium, a number of the BMSCs proliferated into floating spherical aggregates, deemed as neurospheres with multipolar processes (Fig. 1E). On assessment of spheroid diameter (Fig. 1F-H), diameters at day 7 (325.61±9.09 µm) were significantly increased compared with those at 3 h (32.92±1.47 µm), 24 h (115.59±4.88 µm) and 4 days (134.69±4.21 µm; P<0.05; Fig. 2A). The neurospheres were passaged and induced to differentiate by neural stem cell expansion medium containing 5% FBS following plating on poly-L-lysine-coated adherent plates. To confirm the induced NSCs, the P3 cells were subjected to immunostaining analysis. The B-dNSCs from neurospheres expressed a high level of nestin (Fig. 2C) at a positive immunostaining rate of 98.00±0.35% (data not shown).

Dose response and cell viability

To evaluate the protective effect of adenosine against H2O2-induced cytotoxicity, B-dNSCs were treated with various concentrations of adenosine (2, 4, 6, 8 and 10 µM) for 48 h prior to H2O2 exposure. Cell viability was then evaluated by MTT assay (Fig. 2B). The pretreatment of B-dNSCs with adenosine increased cell viability in an apparent dose dependent manner, with the exception of 6 µM adenosine, which induced the greatest cell viability (90.02±0.24%) relative to the other concentrations tested, to a significant extent compared with 2, 4 and 8 µM adenosine (P<0.05). Therefore, 6 µM adenosine was selected as a suitable dose for further studies.

TUNEL assay

A TUNEL assay was performed to determine the antiapoptotic effect of adenosine in B-dNSCs treated with H2O2. As depicted in Fig. 3A-D, TUNEL-positive nuclei staining demonstrated that H2O2 induced apoptotic cell death. The percentage of TUNEL-positive cells was significantly decreased in the NAH group compared with that in the NH group (P<0.05; Fig. 3E). The mean percentages of TUNEL positive cells in the N, NA, NH and NAH groups were 5.47±0.48, 9.43±0.45, 53.19±1.12 and 30.07±1.16%, respectively.

Gene expression

The changes in expression of Bcl-2, Nrf2, Mst1 and AA1R in the experimental groups were examined using real-time RT-qPCR. The results were presented relative to the NH and N groups individually (Fig. 3F-G). In the groups treated with adenosine (relative to the NH group), all genes exhibited increased expression except Mst1 and AAR1. The mean fold-changes relative to the NH group were as follows: For Mst1, NA: 0.20±0.04, NAH: 0.03±0.00; for Bcl-2, NA: 0.85±0.04, NAH: 1.08±0.05; for Nrf2, NA: 2.37±0.37.4, NAH: 1.35±0.15; and for AA1R, NA: 0.20±0.04, NAH: 8.50±0.57 (Fig. 3F).

The mean values of fold-change in the Bcl-2, Nrf2, Mst1 and AA1R genes relative to levels in the N group are presented in Fig. 3G. mRNA expression of proapoptotic Mst1 in the NH group was significantly upregulated compared with that in the NAH and NA groups (P<0.05). Additionally, the relative expression ratio of AA1R was significantly increased in the NAH and NA groups compared with that in the NH group (P<0.05). The mean fold-changes relative to the N group were as follows: For Mst1, NH: 1.91±0.09, NA: 0.60±0.09, NAH: 0.06±0.01; for Bcl-2, NH: 0.92±0.02, NA: 0.75±0.03, NAH: 0.95±0.05; for Nrf2, NH: 0.12±0.00, NA: 0.20±0.02, NAH: 0.13±0.01; and for AA1R, NH: 1.36±0.05, NA: 3.27±0.17, NAH: 3.27±0.27 (Fig. 3G).

Mst1 and Nrf2 protein expression

To determine the neuroprotective effect of adenosine, Mst1 and Nrf2 protein expression was detected by immunocytochemistry (Figs. 4 and 5). The percentages of Nrf2 and Mst1 positive cells were calculated in five microscopic fields (Fig. 4A and B). The control group (N) exhibited Mst1 and Nrf1 positive cells at rates of 9.75±1.08 and 23.82±1.31%, respectively. By contrast, a significantly increased percentage of Mst1 positive cells (26.4±1.56%) was identified in the NH group compared with the NAH group (P<0.05). The percentage of Mst1 positive cells was significantly decreased in the NAH group (18.46±1.17%) compared with NH group (P<0.05). Additionally, the percentage of Nrf2 positive cells was significantly increased in the NA group (45.56±1.67) compared with the NH group (31.1±1.92; P<0.05; Fig. 5).

Discussion

The results of the present study indicate that adenosine upregulates antiapoptotic Bcl-2 and Nrf2 and downregulates Mst1 in B-dNSCs, and thus protects the cells against H2O2-induced apoptosis. Additionally, adenosine may increase the mRNA expressions of AA1R, as one of the adenosine receptors activated in NSCs. The results further demonstrated that adenosine attenuated the apoptotic and necrotic effects of H2O2 at the cellular level. While adenosine is currently only used to treat cardiac arrhythmias, research increasingly suggests various combinations of novel clinical applications of this drug (24,25).

The current study reports a novel cytoprotective mechanism of adenosine involving Nrf2 and Bcl-2-mediated induction of oxidative stress-related proteins. Oxidative damage is among the causes of neurodegenerative diseases (26). Increased production of cellular oxidants and defective protective mechanisms against oxidants are two major factors in the development of oxidative damage (21). However, physiological concentrations of free radicals in cells are essential for normal function and have key importance in the regulation of signaling pathways (14) including those associated with antiapoptotic transcription of genes and DNA repair proteins (17). The main targets of ROS during oxidative stress are considered to be DNA, RNA, proteins and lipids (22). In apoptosis, a series of events occurs systematically and repeatedly. The process is regulated by interventional genes including caspases and Fas-associated protein with death domain, and also molecular systems including Bcl-2/Bcl-2-associated X protein and Fas/Fas ligand (27). Therefore, interference in antiapoptotic and proapoptotic gene expression may be used to establish alterations in apoptosis (23).

Studies have indicated that adenosine may protect stem cells against oxidative damage and thus reduce apoptosis in these cells (28,29). The results of the current study confirmed previous findings regarding the protective effect of adenosine (30). The concept that purines, including ATP, ADP and adenosine, serve as extracellular signaling molecules was first suggested in 1929 by DeMaagd and Philip (31). Depending on the specific receptor activated, extracellular purines mediate biological functions including transmission of nerve impulses, muscle contraction, secretion from endocrine and exocrine glands and inflammation (31). Zhai et al (32) demonstrated that the protein level of AA1R was significantly increased in an intracerebral hemorrhage condition, while there was no significant change in the protein levels of the other adenosine receptors. Furthermore, activation of AA1R attenuates neuronal apoptosis in cortical neurons (7). The present results demonstrated that treatment with adenosine during oxidative damage caused overexpression of the antiapoptotic Bcl-2 and Nrf2 genes, as well as increased expression of AA1R and decreased expression of proapoptotic Mst1.

Mst1, also known as serine/threonine kinase 4, is a stress-activated, proapoptotic kinase that is involved in the Hippo pathway (33). Despite extensive studies, the mechanisms underlying the regulation of apoptosis by Mst1 are not well understood, though Mst1-induced caspase-3 cleavage may be involved (34). It has also been suggested that increased expression of Mst1 initiates apoptosis through activation of p53, but the molecular mechanisms are not fully understood (35). The Nrf2 gene affects cell proliferation, cell growth and regulation of cell metabolism via the phosphatidylinositol-3-kinase/Akt pathway (36). Sarkar et al demonstrated that Nrf2 enhanced the expression of anti-apoptotic Bcl-2 (37). Pingle et al reported that AA1R, protects against human immunodeficiency virus type 1 toxicity by inhibiting nu clear factor-κB and thereby reducing the expression of inducible nitric oxide (NO) synthase and NO radicals and neuronal apoptosis (38).

In addition, the use of AA1R agonist has been reported to lead to upregulation of Bcl-2 and Nrf2 gene expression (39). Nrf2 expression also leads to increased expression of mitochondrial transcription factor A, as a key regulator of antioxidant signaling (40). The current results indicated that Nrf2-mediated upregulation of antiapoptotic protein Bcl-2 may lead to decreased apoptotic cell death and increased cell survival in response to H2O2 exposure. Nrf2 is a transcription factor that controls the expression of a variety of antioxidant genes (41). A previous study demonstrated that Nrf2 was retained in the cytoplasm by the inhibitor kelch-like ECH-associated protein 1, which functions as an adapter for cullin 3/ring-box 1-mediated degradation of Nrf2 (42). The expression of the mitochondrial protein uncoupling protein 3 is driven by the Nrf2 transcription factor, which decreases ROS production and prevents cell death (43). Additionally, Nrf2 may increase the expression of certain antioxidant enzymes, including heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 and manganese superoxide dismutase (44). Taken together, the Nrf2 transcription factor may control the expression of a number of protective genes in response to oxidative stress (45). Thus, according to studies on adenosine, manipulating the adenosine system as a novel strategy for the management of brain disorders may have promise (31). However, to achieve successful clinical application in the treatment of brain disorders, further studies are required to evaluate the use of adenosine and its receptor agonists as neuroprotective drugs.

In conclusion, the results of the current study demonstrated that adenosine, as a receptor-specific agonist of AA1R, protected NSCs derived from BMSCs against oxidative damage. Adenosine can cross the blood-brain barrier and therefore may be considered as a suitable drug for the treatment of diseases of the nervous system caused by oxidative stress.

Acknowledgements

The authors are grateful to Dr Soghrat Faghihzadeh at the Department of Biostatistics and Epidemiology of Zanjan University of Medical Sciences (Zanjan, Iran), who provided valuable feedback on the manuscript.

Funding

The current study was funded by Zanjan University of Medical Sciences (Zanjan, Iran; grant no. A-12-973-2).

Availability of data and materials

All primary data are archived at the Zanjan University of Medical Sciences and available on request.

Authors' contributions

MG performed the experiments. IJA acted as study advisor and was responsible for technical aspects in the immunocytochemistry. AT is the co-corresponding author, analyzed data regarding gene expression, was responsible for primer design and supervised the project. AA designed and directed the project, supervised the project, monitored experiments, is the co-corresponding author and wrote the manuscript. All authors discussed the results and contributed to the final manuscript.

Ethics approval and consent to participate

All experimental protocols were approved by the Zanjan University of Medical Sciences Ethics Committee.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yeh LK, Liu CY, Chien CL, Converse RL, Kao WW, Chen MS, Hu FR, Hsieh FJ and Wang IJ: Molecular analysis and characterization of zebrafish keratocan (zKera) gene. J Biol Chem. 283:506–517. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Chen X, Guo C and Kong J: Oxidative stress in neurodegenerative diseases. Neural Regen Res. 7:376–385. 2012.PubMed/NCBI

3 

Uttara B, Singh AV, Zamboni P and Mahajan RT: Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol. 7:65–74. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Liu Z, Liu Y, Gao R, Li H, Dunn T, Wu P, Smith RG, Sarkar PS and Fang X: Ethanol suppresses PGC-1α expression by interfering with the cAMP-CREB pathway in neuronal cells. PLoS One. 9:e1042472014. View Article : Google Scholar : PubMed/NCBI

5 

Cunha RA: Adenosine as a neuromodulator and as a homeostatic regulator in the nervous system: Different roles, different sources and different receptors. Neurochem Int. 38:107–125. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Xu L, Xu H, Cao Y, Yang P, Feng Y, Tang Y, Yuan S and Ming J: Validation of reference genes for quantitative real-time PCR during bicolor tepal development in asiatic hybrid lilies (Lilium spp.). Front Plant Sci. 8:6692017. View Article : Google Scholar : PubMed/NCBI

7 

Gorman AM: Neuronal cell death in neurodegenerative diseases: Recurring themes around protein handling. J Cell Mol Med. 12(6A): 2263–2280. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Gibson RM: Does apoptosis have a role in neurodegeneration? BMJ. 322:1539–1540. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Mattson MP: Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol. 1:120–129. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Abdanipour A, Tiraihi T and Mirnajafi-Zadeh J: Improvement of the pilocarpine epilepsy model in rat using bone marrow stromal cell therapy. Neurol Res. 33:625–632. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Racine RJ: Modification of seizure activity by electrical stimulation. II. Motor seizure. Electroencephalogr Clin Neurophysiol. 32:281–294. 1972. View Article : Google Scholar : PubMed/NCBI

12 

Collak FK, Yagiz K, Luthringer DJ, Erkaya B and Cinar B: Threonine-120 phosphorylation regulated by phosphoinositide-3-kinase/Akt and mammalian target of rapamycin pathway signaling limits the antitumor activity of mammalian sterile 20-like kinase 1. J Biol Chem. 287:23698–23709. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Ardestani A, Paroni F, Azizi Z, Kaur S, Khobragade V, Yuan T, Frogne T, Tao W, Oberholzer J, Pattou F, et al: MST1 is a key regulator of beta cell apoptosis and dysfunction in diabetes. Nat Med. 20:385–397. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Ura S, Masuyama N, Graves JD and Gotoh Y: MST1-JNK promotes apoptosis via caspase-dependent and independent pathways. Genes Cells. 6:519–530. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Strom J, Xu B, Tian X and Chen QM: Nrf2 protects mitochondrial decay by oxidative stress. FASEB J. 30:66–80. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Murata H, Takamatsu H, Liu S, Kataoka K, Huh NH and Sakaguchi M: NRF2 regulates PINK1 expression under oxidative stress conditions. PLoS One. 10:e01424382015. View Article : Google Scholar : PubMed/NCBI

17 

Zhong J and Li L: Skin-derived precursors against UVB-induced apoptosis via Bcl-2 and Nrf2 upregulation. Biomed Res Int. 2016:68947432016. View Article : Google Scholar : PubMed/NCBI

18 

Maleki M, Ghanbarvand F, Behvarz Reza M, Ejtemaei M and Ghadirkhomi E: Comparison of mesenchymal stem cell markers in multiple human adult stem cells. Int J Stem Cells. 7:118–126. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Yang Q, Mu J, Li Q, Li A, Zeng Z, Yang J, Zhang X, Tang J and Xie P: A simple and efficient method for deriving neurospheres from bone marrow stromal cells. Biochem Biophys Res Commun. 372:520–524. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Suzuki S, Namiki J, Shibata S, Mastuzaki Y and Okano H: The neural stem/progenitor cell marker nestin is expressed in proliferative endothelial cells, but not in mature vasculature. J Histochem Cytochem. 58:721–730. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Albrecht-Küpper BE, Leineweber K and Nell PG: Partial adenosine A1 receptor agonists for cardiovascular therapies. Purinergic Signal. 8 Suppl 1:91–99. 2012. View Article : Google Scholar

22 

Abbasnia K, Ghanbari A, Abedian M, Ghanbari A, Sharififar S and Azari H: The effects of repetitive transcranial magnetic stimulation on proliferation and differentiation of neural stem cells. Anat Cell Biol. 48:104–113. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Lee JH, Cheon YH, Woo RS, Song DY, Moon C and Baik TK: Evidence of early involvement of apoptosis inducing factor-induced neuronal death in Alzheimer brain. Anat Cell Biol. 45:26–37. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Mori M, Nishizaki T and Okada Y: Protective effect of adenosine on the anoxic damage of hippocampal slice. Neuroscience. 46:301–307. 1992. View Article : Google Scholar : PubMed/NCBI

25 

Cunha RA: Neuroprotection by adenosine in the brain: From A(1) receptor activation to A (2A) receptor blockade. Purinergic Signal. 1:111–134. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Migita H, Kominami K, Higashida M, Maruyama R, Tuchida N, McDonald F, Shimada F and Sakurada K: Activation of adenosine A1 receptor-induced neural stem cell proliferation via MEK/ERK and Akt signaling pathways. J Neurosci Res. 86:2820–2828. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Elmore S: Apoptosis: A review of programmed cell death. Toxicol Pathol. 35:495–516. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Huang NK: Adenosine A2A receptors regulate oxidative stress formation in rat pheochromocytoma PC12 cells during serum deprivation. Neurosci Lett. 350:127–131. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Ramkumar V, Hallam DM and Nie Z: Adenosine, oxidative stress and cytoprotection. Jpn J Pharmacol. 86:265–274. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Vitolo OV, Ciotti MT, Galli C, Borsello T and Calissano P: Adenosine and ADP prevent apoptosis in cultured rat cerebellar granule cells. Brain Res. 809:297–301. 1998. View Article : Google Scholar : PubMed/NCBI

31 

DeMaagd G and Philip A: Parkinson's disease and its management: Part 1: Disease entity, risk factors, pathophysiology, clinical presentation, and diagnosis. P T. 40:504–532. 2015.PubMed/NCBI

32 

Zhai W, Chen D, Shen H, Chen Z, Li H, Yu Z and Chen G: A1 adenosine receptor attenuates intracerebral hemorrhage-induced secondary brain injury in rats by activating the P38-MAPKAP2-Hsp27 pathway. Mol Brain. 9:662016. View Article : Google Scholar : PubMed/NCBI

33 

Meng Z, Moroishi T and Guan KL: Mechanisms of Hippo pathway regulation. Genes Dev. 30:1–17. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Wrann CD, White JP, Salogiannnis J, Laznik-Bogoslavski D, Wu J, Ma D, Lin JD, Greenberg ME and Spiegelman BM: Exercise induces hippocampal BDNF through a PGC-1α/FNDC5 pathway. Cell Metab. 18:649–659. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Youdim MB: The path from anti Parkinson drug selegiline and rasagiline to multifunctional neuroprotective anti Alzheimer drugs ladostigil and m30. Curr Alzheimer Res. 3:541–550. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Alexander GE: Biology of Parkinson's disease: Pathogenesis and pathophysiology of a multisystem neurodegenerative disorder. Dialogues Clin Neurosci. 6:259–280. 2004.PubMed/NCBI

37 

Sarkar S, Raymick J and Imam S: Neuroprotective and therapeutic strategies against Parkinson's disease: Recent Perspectives. Int J Mol Sci. 17:172016. View Article : Google Scholar

38 

Pingle SC, Jajoo S, Mukherjea D, Sniderhan LF, Jhaveri KA, Marcuzzi A, Rybak LP, Maggirwar SB and Ramkumar V: Activation of the adenosine A1 receptor inhibits HIV-1 tat-induced apoptosis by reducing nuclear factor-kappaB activation and inducible nitric-oxide synthase. Mol Pharmacol. 72:856–867. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Han J, Talorete TP, Yamada P and Isoda H: Anti-proliferative and apoptotic effects of oleuropein and hydroxytyrosol on human breast cancer MCF-7 cells. Cytotechnology. 59:45–53. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Zhang Z, Zhou S, Jiang X, Wang YH, Li F, Wang YG, Zheng Y and Cai L: The role of the Nrf2/Keap1 pathway in obesity and metabolic syndrome. Rev Endocr Metab Disord. 16:35–45. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Ishikawa T: Genetic polymorphism in the NRF2 gene as a prognosis marker for cancer chemotherapy. Front Genet. 5:3832014. View Article : Google Scholar : PubMed/NCBI

42 

Nuydens R, Dispersyn G, Van Den Keiboom G, de Jong M, Connors R, Ramaekers F, Borgers M and Geerts H: Bcl-2 protects against apoptosis-related microtubule alterations in neuronal cells. Apoptosis. 5:43–51. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Anedda A, López-Bernardo E, Acosta-Iborra B, Suleiman Saadeh M, Landázuri MO and Cadenas S: The transcription factor Nrf2 promotes survival by enhancing the expression of uncoupling protein 3 under conditions of oxidative stress. Free Radic Biol Med. 61:395–407. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Jeong YH, Park JS, Kim DH and Kim HS: Lonchocarpine increases Nrf2/ARE-mediated antioxidant enzyme expression by modulating AMPK and MAPK signaling in brain astrocytes. Biomol Ther (Seoul). 24:581–588. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Birben E, Sahiner UM, Sackesen C, Erzurum S and Kalayci O: Oxidative stress and antioxidant defense. World Allergy Organ J. 5:9–19. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 8 Issue 5

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gholinejad M, Jafari Anarkooli I, Taromchi A and Abdanipour A: Adenosine decreases oxidative stress and protects H2O2‑treated neural stem cells against apoptosis through decreasing Mst1 expression. Biomed Rep 8: 439-446, 2018
APA
Gholinejad, M., Jafari Anarkooli, I., Taromchi, A., & Abdanipour, A. (2018). Adenosine decreases oxidative stress and protects H2O2‑treated neural stem cells against apoptosis through decreasing Mst1 expression. Biomedical Reports, 8, 439-446. https://doi.org/10.3892/br.2018.1083
MLA
Gholinejad, M., Jafari Anarkooli, I., Taromchi, A., Abdanipour, A."Adenosine decreases oxidative stress and protects H2O2‑treated neural stem cells against apoptosis through decreasing Mst1 expression". Biomedical Reports 8.5 (2018): 439-446.
Chicago
Gholinejad, M., Jafari Anarkooli, I., Taromchi, A., Abdanipour, A."Adenosine decreases oxidative stress and protects H2O2‑treated neural stem cells against apoptosis through decreasing Mst1 expression". Biomedical Reports 8, no. 5 (2018): 439-446. https://doi.org/10.3892/br.2018.1083