Guanosine exerts neuroprotective effects by reversing mitochondrial dysfunction in a cellular model of Parkinson's disease

  • Authors:
    • Da-Wei Li
    • Min Yao
    • Yan-Hua Dong
    • Min-Na Tang
    • Wei Chen
    • Guang-Ren Li
    • Bi-Quan Sun
  • View Affiliations

  • Published online on: August 19, 2014     https://doi.org/10.3892/ijmm.2014.1904
  • Pages: 1358-1364
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The mitochondria are the most important cytoplasmic organelles in determining cell survival and death. Mitochondrial dysfunction leads to a wide range of disorders, including neurodegenerative diseases. The central events in the mitochondrial‑dependent cell death pathway are the activation of the mitochodrial permeability transition pore (mPTP) and the disruption of mitochondrial membrane potential, which cause the release of apoptogenic molecules and finally lead to cell death. This is thought to be at least partly responsible for the loss of dopaminergic neurons in Parkinson's disease (PD); thus, the attenuation of mitochondrial dysfunction may contribute to alleviating the severity and progression of this disease. Guanosine is a pleiotropic molecule affecting multiple cellular processes, including cellular growth, differentiation and survival. Its protective effects on the central nervous system and and on several cell types by inhibiting apoptosis have been shown in a number of pathological conditions. This study aimed to analyze the ability of guanosine to protect neuronal PC12 cells from the toxicity induced by 1-methyl-4-phenylpyridinium (MPP+), the active metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which mediates selective damage to dopaminergic neurons and causes irreversible Parkinson-like symptoms in humans and primates. Our results demonstrated that the apoptosis of PC12 cells induced by MPP+ was significantly prevented by pre-treatment for 3 h with guanosine. In addition, guanosine attenuated the MPP+-induced collapse of mitochondrial transmembrane potential and prevented the sebsequent activation of caspase-3, thereby protecting dopaminergic neurons against mitochondrial stress-induced damage.

Introduction

Parkinson’s disease (PD) is a common neurodegenerative disorder characterized by the gradually progressive and selective loss of dopaminergic neurons in the substantia nigra (1). The progressive loss of dopaminergic neurons is a complex process, and multiple pathological events are involved in this process (26). While the underlying mechanisms of nigrostriatal dopaminergic neuron degeneration are not yet completely understood, accumulating evidence indicates that mitochondrial dysfunction may be a central event in neurodegenerative diseases (79). The mitochondria are multifunctional organelles that are important for living cells. Mitochondrial dysfuntion has a multitude of consequences for cells, including apoptosis (10). The activation of the mitochondrial permeability transition pore (mPTP) and the collapse of the mitochondrial membrane potential may be major contributors to mitochondrial-dependent cell death and at least partly reponsible for the pathogenesis of PD and several other neurodegenerative disorders (79).

PC12 cells treated with 1-methyl-4-phenylpyridinium (MPP+) provide a reliable in vitro model for investigating the pathogenesis of PD. MPP+ is an active metobolite of the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which is known to selectively kill dopaminergic neurons and cause irreversible Parkinson-like symptoms in humans and primates (1113). MPTP is a lipophilic molecule that can rapidly cross the blood-brain barrier; it is subsequently oxidized in the brain to its toxic metabolite, MPP+, by type B monoamine oxidase (14). The neurotoxic action of MPP+ is related to the activation of the mPTP and the collapse of mitochondrial membrane potential through oxidative damage, which together initiate the downstream apoptotic pathway, including the release of cytochome c and the activation of caspases, finally leading to neuronal cell death (79). Damage to the mitochondria is considered as an initial and irreversible step towards apoptosis; thus, mitochondrial-targeted therapeutic strategies may be a promising treatment for PD. Guanosine, a non-adenine-based purine, is an intercellular signaling molecule affecting multiple cellular processes, including cellular growth, differentiation and survival (1517). In multiple cell types, it exerts protective effects against apoptosis induced by a number of agents, such as staurosporine (18), β-amyloid (19) and MPTP (20). The neuroprotective effects of guanosine in the central nervous system have also been recognized (15). The present study was designed to investigate the effects of guanosine on MPP+-induced apoptosis in PC12 cells and the underlying mechanisms for these actions. Our results demonstrated that guanosine effectively prevented MPP+-induced PC12 cell apoptosis by stabilizing the mitochondrial membrane potential and attenuating the subsequent activation of caspases. In addition, guanosine inhibited the production of reactive oxygen species (ROS) and increased the expression levels of glutathione (GSH), further supporting the protective role of guanosine in oxidative conditions. Overall, these findings indicate the protective role of guanosine in mitochondrial stress-induced dopaminergic neuronal damage, thus providing potential effective strategies for the treatment of PD.

Materials and methods

Drugs and chemicals

All reagents and chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA) unless stated otherwise.

PC12 cell cultures

The PC12 cells were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China) and maintained in high glucose Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% heat-inactivated fetal bovine serum, 4.00 mM L-glutamine, 100 U/ml of penicillin and 100 μg/ml of streptomycin (Gibco, Grand Island, NY, USA). The cultures were maintained in a humidified 5% CO2 atmosphere at 37°C. The culture medium was changed every 3–4 days and the cells were seeded at a density of 30,000 cells/cm2.

Cell viability assay

Cell viability was measured using the modified 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), a mitochondrial dye which is converted into a blue formazan product by mitochondrial dehydrogenases in metabolically active cells. The PC12 cells were plated at a density of 30,000 cells/cm2 in 96-well plates and incubated for 24 h. To assess the neuroprotective effects of guanosine on MPP+-induced toxicity in PC12 cells, the cells were pre-treated with various concentrations of guanosine (0.01–1,000 μM) for 3 h and were then exposed to 500 μM MPP+ for 72 h, under optimal conditions for the assessment of the neuroprotective effects as previously described (21). MTT (5 mg/ml) solution was added to the wells and the cells were incubated for 4 h. Subsequently, the culture medium was removed, and dimethyl sulfoxide was added to each well to solubilize the formazan into a colored solution. The absorbance of colored solution was measured at 570 nm using a microplate reader (Epoch; BioTek, Winooski, VT, USA). The results were expressed as the percentage of the absorbance of the control culture wells. Based on these results, we used guanosine at a dose of 10 μM in all the subsequent experiments.

Nuclear staining assay

The morphological signs of apoptosis induced by MPP+ were detected using acridine orange (AO)/ethidium bromide (EB) staining of the PC12 cells. The cells were plated in 6-well plates at a density of 30,000 cells/cm2 and were incubated in DMEM medium at 37°C. After 3 h of pre-treatment with guanosine (10 μM), MPP+ (500 μM) was added to the medium for 72 h. The cells were washed and resuspended in phosphate-buffered saline (PBS) and AO/EB was added at a final concentration of 1 μg/ml. Subsequently, the number of apoptotic cells was randomly counted under a fluorescence microscope (IX71; Olympus, Tokyo, Japan). Viable cells with intact structures stained with AO only showed bright green nuclear staining; the early apoptotic cells were bright green and later apoptotic cells were red-orange with condensed chromatin. The number of apoptotic cells is expressed as a percentage of the total cells counted.

Measurement of apoptosis in cells

Apoptosis was assessed by measuring DNA fragmentation with single-stranded DNA (ssDNA) apoptosis enzyme-linked immunosorbent assay (ELISA) kits (Chemicon International, Temecula, CA, USA) according to the manufacture’s instructions. The cells plated at a concentration of 30,000 cells/cm2 were cultured for 24 h, followed by treatment with 10 μM guanosine prior to the addition of 500 μM MPP+ for 3 h. The cells were washed 3 times with PBS and formamide was then added which selectively denaturates DNA in apoptotic cells. Anti-ssDNA monoclonal antibody and peroxidase-conjugated secondary antibody were then added to the cells; the ssDNA was then measured at 450 nm using a microplate reader (Epoch; BioTek).

Measurement of mitochondrial transmembrane potential

Mitochondrial membrane potential is a key indicator of mitochondrial function and cell death or injury, which can be detected using the mitochondrial dye, 3,3-dihexyloxacarbocyanine iodide [DiOC6(3)]. This dye is a lipophilic fluorescent stain and becomes highly fluorescent when incorporated into membranes. The cells at a concentration of 30,000 cells/cm2 were cultured in 24-well plates for 24 h, followed by treatment with 10 μM guanosine prior to the addition of 500 μM MPP+ for 3 h. Following 72 h of incubation, 1 ml of serum-free culture medium containing DiOC6(3) was added to each well with the final concentration of 1 μM, and the cells were cultured in a humidified incubator for 15 min. The cells were collected and centrifuged at 1,000 × g for 5 min, and the cell pellets were resuspended in PBS containing 0.5 mM EDTA. The intensity of DiOC6(3) fluorescence was recorded using a flow cytometer (Becton-Dickinson, San Diego, CA, USA).

Western blot analysis

Following treatment, the PC12 cells were collected and lysed with cell lysis solution containing 4% sodium dodecyl sulfate (SDS), 2 mM EDTA and 50 mM Tris-HCl, pH 6.8. Equal amounts of protein were loaded onto a 12% SDS-polyacrylamide gel. Following electrophoretic separation, the polyacrylamide gels were transferred onto PVDF transfer membranes (Amersham Biosciences, Uppsala, Sweden). The membranes were incubated in Tris-buffered saline/Tween-20 (TBST) supplemented with 5% fat-free milk for 1 h to block non-specific binding. The blots were incubated using rabbit anti-Bax, anti-B-cell lymphoma 2 (Bcl-2) antibodies. Horseradish peroxidase (HRP)-conjugated anti-rabbit antibodies were used as the secondary antidodies.

Measurement of ROS production

Intracellular ROS produced during the inhibition of mitochondrial complex I was detected using 2′–7′-dichlorofluorescein diacetate (DCFH-DA). This is a non-fluorescent cell-permeating compound that can easily diffuse into cells and be converted into dichlorofluorescin (DCFH) by intracellular esterase. DCFH is then trapped within the cell and oxidized into fluorescent dichlorofluorescein (DCF) by intracellular ROS. Following treatment, the cells were incubated in BSA-free DMEM with DCFH-DA at a final concentration of 20 μM for 30 min at 37°C. Thereafter, 10,000 cells of each group were analyzed by flow cytometry using the FL1 flow cytometer detection channels. The excitation wavelength was 485 nm and the reading was performed at 530 nm.

Measurement of GSH levels

GSH levels were measured using GSH reductase, as previously described (22). Briefly, following centrifugation and washing with PBS, the cells were dissovled with 2% 5-sulfosalicylic acid and incubated in 100 μl of the reaction mixture containing 20 mM sodium EDTA, 600 μM nicotinamide adenine dinucleotide phosphate (NADPH), 12 mM 5,5′-dithiobis(2-nitrobenzoic acid) and 105 mM NaH2PO4. GSH reductase was added to each well, and the cells were cultured in a humidified incubator for 10 min. Absorbance was measured at 450 nM, and the calibration curve was performed with standard GSH solutions. The results are expressed as percentages of the control condition.

Evaluation of caspase-3 activity

Caspase-3 activity was measured using an ApoAlert caspase-3 assay kit according to the manufacturer’s instructions. Briefly, the cells were lysed and centrifuged at 1,000 × g for 10 min, then the supernatant was added to the reaction mixture containing dithiothreitol and caspase-3 substrate (N-acetyl-Asp-Glu-Val-Asp p-nitroanilide). The cells were incubated for 1 h at 37°C, and the absorbance of the chromophore p-nitroanilide produced was measured at 450 nm. The standard curves were obtained from the absorbance of p-nitroanilide standard reagent diluted with cell lysis buffer. One unit of the enzyme was defined as the activity producing 1 nmol of p-nitroanilide.

Statistical analysis

Data are expressed as the means ± standard error of the mean (SEM). Statistical analysis was performed by one-way analysis of variance, followed by Dunnett’s multiple-comparisons test. Differences between mean values were considered statistically different at p<0.05.

Results

Guanosine reduces the MPP+-induced loss of cell viability

The ability of guanosine to reverse the cytotoxicity to PC12 cells induced by MPP+ was investigated using MTT, which is a mitochondrial dye and can be converted into a blue formazan product by mitochondrial dehydrogenases; therefore, it can partially detect the levels of metabolically active cells. The measurements revealed a significant decrease in the viability of the PC12 cells following exposure to 500 μM MPP+ for 72 h; however, the cells treated with guanosine alone did not show a decrease in cell viability. Pre-treatment with 10 μM guanosine significantly decreased the MPP+-induced cytotoxicity (Fig. 1).

Guanosine attenuates MPP+-induced apoptosis in PC12 cells

To determine whether guanosine prevents MPP+-induced apoptosis in PC12 cells, AO/EB and DNA fragmentation assays were performed. Apoptosis is a process of programmed cell death characterized by a series of distinct nuclear morphological changes. These changes can be detected by AO/EB staining. This assay identified 3 types of cells under a fluorescence microscope: live cells (green), early apoptotic cells (bright green with condensed chromation) and later apoptotic cells (red-orange with condensed chromation). The administration of guanosine alone did not induce changes in the number of apoptotic cells, while the administration of MPP+ significantly increased the number of apoptotic cells compared to the control group (p<0.01). Pre-treatment with 10 μM guanosine significantly decreased the number of apoptotic cells induced by exposure to MPP+ (p<0.01; Fig. 2), indicating that guanosine plays an anti-apoptotic role. To clarify the neuroprotective role of guanosine on MPP+-induced toxicity in PC12 cells, DNA fragmentation, a marker of late apoptosis, was further investigated by ssDNA assay. The results revealed that the increase in DNA fragmentation induced by exposure to MPP+ was markedly attenuated by pre-treatment with guanosine (Fig. 3), supporting the protective role of guanosine in conditions of oxidative stress.

Guanosine modulates Bax and Bcl-2 protein expression

Bax and Bcl-2 are key members of the Bcl-2 family of proteins that contribute to the opening of mPTP, leading to the induction of apoptosis. To investigate the changes in Bax and Bcl-2 protein expression levels, western blot analysis was performed on the untreated cells and the cells treated with 500 μM MPP+ alone or 500 μM MPP+ in the presence of 10 μM of guanosine. The administration of MPP+ significantly increased the levels of Bax expression and decreased Bcl-2 expression. These changes were be markedly reversed by pre-treatment with guanosine. Treatment with guanosine alone did not induce changes in the expression levels of these proteins (Fig. 4), thus further demonstrating the protective role of guanosine in mitochondrial-stress induced cell damage.

Guanosine prevents the MPP+-induced collapse of mitochondrial transmembrane potential

Mitochondrial membrane potential maintenance is essential for living cells, and its collapse is a key event in the activation of the mitochondrial-dependent pathway. The collapse of mitochondrial transmembrane potential was assessed by measuring the response to the mitochondrial dye, DiOC6(3), which is converted into a highly green fluorescent dye following incorporation into mitochondrial membranes, thereby allowing the qualitative assessment of mitochondrial membrane potential. The administration of MPP+ in comparison with the control cells induced a significant decrease in fluorescence intensity, indicating the increasing percentage of the cells with collapse of mitochondrial membrane potential. The results also revealed a marked reduction in the number of cells with the collapse of mitochondrial membrane potential, when guanosine was administered prior to exposure to MPP+; no significant change was observed following treatment with guanosine alone (Fig. 5). These results suggest that the mitochondrial dysfunction induced by MPP+ can be partly restored by the administration of guanosine.

Guanosine inhitis the MPP+-induced production of ROS

The levels of ROS production were evaluated by flow cytometry with DCFH-DA. DCFH-DA is a stable compound that can easily diffuse into cells, where it is converted into DCFH by intracellular esterase. DCFH is then trapped within cells and oxidized to highly fluorescent DCF by intracellular ROS; thus, the intensity of fluorescence produced by DCF may reflect an intracellular oxidative state.

The administration of guanosine alone, compared with the control group, did not elicit changes in the levels of DCFH oxidation. The administration of MPP+ induced a significant increase in DCFH oxidation in the PC12 cells, which was markedly reversed by pre-treatment with guanosine (Fig. 6A), thus indicating that guanosine may play an antioxidant role.

Guanosine reverses the MPP+-induced reduction in GSH levels

GSH protein is a major non-enzymatic antioxidant that plays a crucial role in protecting neurons from oxidative damage in the central nervous system (23). To assess the protective role of guanosine in MPP+-induced oxidative damage, the levels of GSH were measured in the PC12 cells. The administration of MPP+ in comparison with the control cells induced a significant decrease in GSH levels; this effect was markedly reversed by pre-treatment with guanosine. The administration of guanosine alone did not elicit any changes in the levels of GSH (Fig. 6B).

Guanosine reduces caspase-3 activity

Caspase-3 is an effector caspase that cleaves a wide range of signal transduction proteins in the apoptotic process (24). To determine whether guanosine protects neuronal PC12 cells against MPP+-induced cell death, the activity of caspase-3 was measured by ELISA with an ApoAlert caspase-3 assay kit. The PC12 cells exposed to 500 μM MPP+ showed a significant increase in caspase-3 activity; however, treatment with guanosine alone did not induce any changes in caspase-3 activity. Pre-treatment with guanosine markedly inhibited the MPP+-induced the increment in caspase-3 activity (Fig. 7), illustrating the protective role of guanosine against MPP+-induced toxicity in PC12 cells.

Discussion

The non-adenine-based purine, guanosine, is a multifaceted intercellular signaling molecule affecting multiple cellular processes, including cellular growth, differentiation and survival (15). Its protective roles have been reported in previous studies. It protects several cell types against apoptosis induced by a number of agents, such as staurosporine, β-amyloid and MPP+ through its interactions with several steps of the biochemical and cellular cascade (18,19). The protective role of guanosine has also been reported in neurodegenerative diseases (20). The present study demonstrates that guanosine exerts protective effects against apoptotic cell death elicited by MPP+ by alleviating mitochondrial dysfunction, inhibiting the activation of caspase-3 and, subsequently, attenuating cytotoxic cell damage in a reliable cellular model of PD.

PD is a common neurodegenerative disease clinically characterized by rigidity, resting tremor, bradykinesia and postural instability caused by the degeneration and death of dopaminergic neurons in the pars compacta of the substantia nigra (25). Althought the cellular and molecular events underlying the loss of dopaminergic neurons remain unclear, accumulating evidence indicates that mitochondrial dysfunction may be a central event in the pathogenesis of PD (2,8). The mitochondria are the most important cytoplasmic organelles responsible for the life and death of cells (26). The maintenance of membrane potential and the low-conductance of the mPTP are the major properties of mitochondria in living cells, and any changes related to these properties are considered to be critical factors associated with mitochondrial dysfunction and cell death in neurodegenerative diseases (7,8). Multiple proteins are involved in intrinsic apoptotic events associated with mitochondrial dysfunction. The Bcl-2 family of proteins are recognized as key messengers for delivering the apoptotic signal to the mitochondria in response to various insults (27). Pro-apoptotic Bax and anti-apoptotic Bcl-2 are key members of the Bcl-2 family in apoptosis mediated by mitochondrial stress. Under pathogenic conditions, Bax is upregulated and translocates from the cytoplasm to the mitochondria. Once located in the mitochondrial membrane, this protein causes mitochondrial membrane disruption by sequestering Bcl-2 and oligomerizing within the mitochondrial membrane, leading to the opening of the mPTP, the collapse of mitochondrial membrane potential and the subsequent release of pro-apoptotic molecules into the cytoplasm (28,29). Compared to Bax, Bcl-2 is a key protein that preserves mitochondrial integrity, thereby preventing stress-induced mitochondrial damage in cells (30). Bcl-2 proteins are crucial effectors in the opening of the mPTP and the collapse of mitochondrial potential, thus determining the induction of downstream events in the mitochondrial-dependent cell death pathway, including the release of pro-apoptotic molecules and the activation of caspases (28).

Our results revealed that treatment with MPP+ induced the adverse expression levels of two Bcl-2 proteins and the disruption of the mitochondrial membrane potential, supporting the involvement of mitochondrial dysfunction in dopaminergic neuronal degeneration. These changes were reversed by the administration of guanosine prior to exposure to MPP+, demonstrating the protective role of guanosine in mitochondrial-stress induced cell damage, which was partly mediated through the regulation of the expression of proteins involved in the mitochondrial stage of the apoptotic cascade. However, the underlying mechanism responsible for this effect of guanosine is unclear.

A number of studies have indicated that the anti-apoptotic effects of guanosine are mediated by modulating the phosphatidylinositol 3-kinase (PI3K)/Akt/protein kinase B (PKB) and the mitogen-activated protein kinase (MAPK) cell survival pathways (18,19,31,32). PI3K is an upstream signal of glycogen synthase kinase 3 (GSK-3) that plays a central role in the mitochondrial-dependent cell death pathway through the regulation of anti-apoptotic and pro-apoptotic Bcl-2 family proteins, including Bcl-2 and Bax (3335). GSK-3β can directly phosphorylate Bax on serine 163, which results in the activation of Bax, and, subsequently, in its translocation from the cytoplasm to the mitochondria. The inhibition of GSK-3β suppresses the levels of Bax expression, but increases Bcl-2 expression, thereby promoting cell survival by alleviating the mitochondrial dysruption under multiple pathological conditions (34,36,37). Thus, the neuroprotective effects of guanosine may be mediated through the activation of PI3K, which inactivates the downstream signal protein, GSK-3β, leading to the attenuation of the opening of the mPTP through the regulation of Bcl-2 family proteins. This hypothesis is surported by our results that guanosine reversed the collapse of mitochondrial membrane potential, the downstream event of the opening of the mPTP in mitochondrial-mediated cell death. The opening of the mPTP causes the collapse of mitochondrial membrane potential and, subsequently, the release of apoptotic proteins from the mitochondria into the cytoplasm. Perhaps the most intriguing pro-apoptotic protein that is released is cytochrome c, which triggers the activation of the caspase cascade (28). Caspase-3 is a key effector in the mitochondrial-stress-induced apoptotic pathway, and its activation leads to the irreversible process toward apoptosis (38,39). Our results also demonstrated that guanosine, when administered to MPP+-treated neuronal PC12 cells, effectively prevented the collapse of mitochondrial potential and inhibited caspase-3 activity, further supporting its protective role in mitichondrial stress-induced neuronal cell damage.

Oxidative stress is another pathological event associated with cell death mechanisms in PD. Studies using postmortem samples of PD have demonstrated that oxidative markers, including soluble protein carbonyl modifications, lipid peroxidation and DNA oxidative damage are selectively observed in the dopaminergic neurons in the pars compacta of the substantia nigra, indicating the correlation of oxidative damage with striatal dopaminergic neurodegeneration (4,5,40). The inhibition of mitochondrial complex I with MPP+ and rotenone, well-established inducers of Parkinson-like symptoms in humans and primates, can lead to an increase in ROS production and selective dopaminergic neuronal loss in the substantia nigra, supporting the involvement of oxidative stress in the pathogenesis of PD (41). ROS are mainly produced as by-products of oxidative phosphorylation in the mitochondria, and many mitochondrial proteins, which possess iron-sulfur clusters for oxidation-reduction reactions and lack protective histones, are particularly vulnerable to ROS attack (42). Generally, cells develop complex antioxidant systems to scavenge ROS. The GSH protein is recognized as the major non-enzymatic antioxidant in the central nervous system (23). A number of studies have indicated that increased levels of GSH exert protective effects in various neurodegenerative diseases, such as Alzheimer’s disease and PD (22,43,44). The reduction of GSH levels contributes to the dysfunction of the mitochondria and increases the sensitivity of neurons to toxic insults (45). The neurotoxin, MPP+, is an inhibitor of the mitochondrial respiratory chain and an inducer of ROS in the mitochondria. Our results revealed that pre-treatment with guanosine reduced the MPP+-induced increase in the production of ROS, the crucial contributors to mitochondrial dysfunction through oxidative damage and the opening of the mPTP. Moreover, guanosine alleviated the decreased levels of GSH induced by the administration of MPP+, reinforcing its protective role in oxidative conditions and its role as a potential neuroprotectant in mitochondrial-mediated neurodegenerative diseases.

In conclusion, this study clearly demonstrates that the neuroprotective effects of guanosine promote dopaminergic neuronal survival by alleviating mitochontrial dysfunction in a cellular model of PD. These neuroprotective effects are partly mediated through the stabilization of mitochondrial membrane potential via the modulation of the expression levels of intrinsic apoptotic proteins involved in the mitochondrial apoptotic pathway. Further studies are required to fully elucidate the mechanisms responsible for the protective effects of guanosine in neurodegenerative diseases, which may promote the development of potentially effective treatments for neurodegenerative diseases by targeting mitochondrias-mediated neuronal damage.

Abbreviations:

PD

Parkinson’s disease

MPTP

1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

MPP+

1-methyl-4-phenylpyridinium

mPTP

mitochodrial permeability transition pore

DCF

2′7′-dichlorodihydrofluorescein

Bcl-2

B-cell lymphoma 2

ROS

reactive oxygen species

References

1 

Forno LS: Neuropathology of Parkinson’s disease. J Neuropathol Exp Neurol. 55:259–272. 1996.

2 

Abou-Sleiman PM, Muqit MM and Wood NW: Expanding insights of mitochondrial dysfunction in Parkinson’s disease. Nat Rev Neurosci. 7:207–219. 2006.

3 

McGeer PL and McGeer EG: Glial reactions in Parkinson’s disease. Mov Disord. 23:474–483. 2008.

4 

Olanow CW: The pathogenesis of cell death in Parkinson’s disease - 2007. Mov Disord. 22(Suppl 17): S335–S342. 2007.

5 

Zhou C, Huang Y and Przedborski S: Oxidative stress in Parkinson’s disease: a mechanism of pathogenic and therapeutic significance. Ann NY Acad Sci. 1147:93–104. 2008.

6 

Li DW, Liu ZQ, Chen W, Yao M and Li GR: Association of glycogen synthase kinase-3β with Parkinson’s disease (Review). Mol Med Rep. 9:2043–2050. 2014.

7 

Vila M and Przedborski S: Targeting programmed cell death in neurodegenerative diseases. Nat Rev Neurosci. 4:365–375. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Perier C, Tieu K, Guégan C, et al: Complex I deficiency primes Bax-dependent neuronal apoptosis through mitochondrial oxidative damage. Proc Natl Acad Sci USA. 102:19126–19131. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Roucou X and Martinou JC: Conformational change of Bax: a question of life or death. Cell Death Differ. 8:875–877. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Shiba-Fukushima K, Imai Y, Yoshida S, et al: PINK1-mediated phosphorylation of the Parkin ubiquitin-like domain primes mitochondrial translocation of Parkin and regulates mitophagy. Sci Rep. 2:10022012. View Article : Google Scholar : PubMed/NCBI

11 

Kopin IJ and Markey SP: MPTP toxicity: implications for research in Parkinson’s disease. Annu Rev Neurosci. 11:81–96. 1988.

12 

Heikkila RE, Sieber BA, Manzino L and Sonsalla PK: Some features of the nigrostriatal dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the mouse. Mol Chem Neuropathol. 10:171–183. 1989. View Article : Google Scholar : PubMed/NCBI

13 

Calon F, Lavertu N, Lemieux AM, et al: Effect of MPTP-induced denervation on basal ganglia GABA(B) receptors: correlation with dopamine concentrations and dopamine transporter. Synapse. 40:225–234. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Chiba K, Trevor A and Castagnoli N Jr: Metabolism of the neurotoxic tertiary amine, MPTP, by brain monoamine oxidase. Biochem Biophys Res Commun. 120:574–578. 1984. View Article : Google Scholar : PubMed/NCBI

15 

Rathbone MP, Middlemiss PJ, Gysbers JW, et al: Trophic effects of purines in neurons and glial cells. Prog Neurobiol. 59:663–690. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Dal-Cim T, Martins WC, Santos AR and Tasca CI: Guanosine is neuroprotective against oxygen/glucose deprivation in hippocampal slices via large conductance Ca2+-activated K+ channels, phosphatidilinositol-3 kinase/protein kinase B pathway activation and glutamate uptake. Neuroscience. 183:212–220. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Chang R, Algird A, Bau C, Rathbone MP and Jiang S: Neuro-protective effects of guanosine on stroke models in vitro and in vivo. Neurosci Lett. 431:101–105. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Di Iorio P, Ballerini P, Traversa U, et al: The antiapoptotic effect of guanosine is mediated by the activation of the PI 3-kinase/AKT/PKB pathway in cultured rat astrocytes. Glia. 46:356–368. 2004.PubMed/NCBI

19 

Pettifer KM, Kleywegt S, Bau CJ, et al: Guanosine protects SH-SY5Y cells against beta-amyloid-induced apoptosis. Neuroreport. 15:833–836. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Pettifer KM, Jiang S, Bau C, et al: MPP(+)-induced cytotoxicity in neuroblastoma cells: Antagonism and reversal by guanosine. Purinergic Signal. 3:399–409. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Li DW, Li GR, Lu Y, et al: α-lipoic acid protects dopaminergic neurons against MPP+-induced apoptosis by attenuating reactive oxygen species formation. Int J Mol Med. 32:108–114. 2013.

22 

Yim SB, Park SE and Lee CS: Protective effect of glycyrrhizin on 1-methyl-4-phenylpyridinium-induced mitochondrial damage and cell death in differentiated PC12 cells. J Pharmacol Exp Ther. 321:816–822. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Banerjee R, Vitvitsky V and Garg SK: The undertow of sulfur metabolism on glutamatergic neurotransmission. Trends Biochem Sci. 33:413–419. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Fischer U, Jänicke RU and Schulze-Osthoff K: Many cuts to ruin: a comprehensive update of caspase substrates. Cell Death Differ. 10:76–100. 2003. View Article : Google Scholar : PubMed/NCBI

25 

de Lau LM and Breteler MM: Epidemiology of Parkinson’s disease. Lancet Neurol. 5:525–535. 2006.

26 

Budd SL and Nicholls DG: Mitochondria in the life and death of neurons. Essays Biochem. 33:43–52. 1998.PubMed/NCBI

27 

Akhtar RS, Ness JM and Roth KA: Bcl-2 family regulation of neuronal development and neurodegeneration. Biochim Biophys Acta. 1644:189–203. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Armstrong JS: Mitochondrial membrane permeabilization: the sine qua non for cell death. Bioessays. 28:253–260. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Martinou JC and Green DR: Breaking the mitochondrial barrier. Nat Rev Mol Cell Biol. 2:63–67. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Gollapudi S, McCormick MJ and Gupta S: Changes in mitochondrial membrane potential and mitochondrial mass occur independent of the activation of caspase-8 and caspase-3 during CD95-mediated apoptosis in peripheral blood T cells. Int J Oncol. 22:597–600. 2003.PubMed/NCBI

31 

Tarozzi A, Merlicco A, Morroni F, et al: Guanosine protects human neuroblastoma cells from oxidative stress and toxicity induced by Amyloid-beta peptide oligomers. J Biol Regul Homeost Agents. 24:297–306. 2010.PubMed/NCBI

32 

Molz S, Dal-Cim T, Budni J, et al: Neuroprotective effect of guanosine against glutamate-induced cell death in rat hippocampal slices is mediated by the phosphatidylinositol-3 kinase/Akt/glycogen synthase kinase 3β pathway activation and inducible nitric oxide synthase inhibition. J Neurosci Res. 89:1400–1408. 2011.PubMed/NCBI

33 

Chuang DM, Chen RW, Chalecka-Franaszek E, et al: Neuro-protective effects of lithium in cultured cells and animal models of diseases. Bipolar Disord. 4:129–136. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Chen RW and Chuang DM: Long term lithium treatment suppresses p53 and Bax expression but increases Bcl-2 expression. A prominent role in neuroprotection against excitotoxicity. J Biol Chem. 274:6039–6042. 1999. View Article : Google Scholar : PubMed/NCBI

35 

Manji HK and Chen G: PKC, MAP kinases and the bcl-2 family of proteins as long-term targets for mood stabilizers. Mol Psychiatry. 7(Suppl 1): S46–S56. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Chen G, Zeng WZ, Yuan PX, et al: The mood-stabilizing agents lithium and valproate robustly increase the levels of the neuroprotective protein bcl-2 in the CNS. J Neurochem. 72:879–882. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Kaga S, Zhan L, Altaf E and Maulik N: Glycogen synthase kinase-3beta/beta-catenin promotes angiogenic and anti-apoptotic signaling through the induction of VEGF, Bcl-2 and survivin expression in rat ischemic preconditioned myocardium. J Mol Cell Cardiol. 40:138–147. 2006. View Article : Google Scholar

38 

Boatright KM and Salvesen GS: Mechanisms of caspase activation. Curr Opin Cell Biol. 15:725–731. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Kumar S: Caspase function in programmed cell death. Cell Death Differ. 14:32–43. 2007. View Article : Google Scholar

40 

Navarro A, Boveris A, Bandez MJ, et al: Human brain cortex: mitochondrial oxidative damage and adaptive response in Parkinson disease and in dementia with Lewy bodies. Free Radic Biol Med. 46:1574–1580. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Cassarino DS, Fall CP, Swerdlow RH, et al: Elevated reactive oxygen species and antioxidant enzyme activities in animal and cellular models of Parkinson’s disease. Biochim Biophys Acta. 1362:77–86. 1997.PubMed/NCBI

42 

Wallace DC: A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet. 39:359–407. 2005. View Article : Google Scholar : PubMed/NCBI

43 

Halliwell B: Oxidative stress and neurodegeneration: where are we now? J Neurochem. 97:1634–1658. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Lee M, Cho T, Jantaratnotai N, Wang YT, McGeer E and McGeer PL: Depletion of GSH in glial cells induces neurotoxicity: relevance to aging and degenerative neurological diseases. FASEB J. 24:2533–2545. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Hall AG: Review: The role of glutathione in the regulation of apoptosis. Eur J Clin Invest. 29:238–245. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2014
Volume 34 Issue 5

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
Li D, Yao M, Dong Y, Tang M, Chen W, Li G and Sun B: Guanosine exerts neuroprotective effects by reversing mitochondrial dysfunction in a cellular model of Parkinson's disease. Int J Mol Med 34: 1358-1364, 2014
APA
Li, D., Yao, M., Dong, Y., Tang, M., Chen, W., Li, G., & Sun, B. (2014). Guanosine exerts neuroprotective effects by reversing mitochondrial dysfunction in a cellular model of Parkinson's disease. International Journal of Molecular Medicine, 34, 1358-1364. https://doi.org/10.3892/ijmm.2014.1904
MLA
Li, D., Yao, M., Dong, Y., Tang, M., Chen, W., Li, G., Sun, B."Guanosine exerts neuroprotective effects by reversing mitochondrial dysfunction in a cellular model of Parkinson's disease". International Journal of Molecular Medicine 34.5 (2014): 1358-1364.
Chicago
Li, D., Yao, M., Dong, Y., Tang, M., Chen, W., Li, G., Sun, B."Guanosine exerts neuroprotective effects by reversing mitochondrial dysfunction in a cellular model of Parkinson's disease". International Journal of Molecular Medicine 34, no. 5 (2014): 1358-1364. https://doi.org/10.3892/ijmm.2014.1904