Open Access

Involvement of the ERK pathway in the protective effects of glycyrrhizic acid against the MPP+-induced apoptosis of dopaminergic neuronal cells

  • Authors:
    • Lesheng Teng
    • Chunjia Kou
    • Chengyu Lu
    • Jiaming Xu
    • Jing  Xie
    • Jiahui  Lu
    • Yan Liu
    • Zhenzuo Wang
    • Di Wang
  • View Affiliations

  • Published online on: July 2, 2014     https://doi.org/10.3892/ijmm.2014.1830
  • Pages: 742-748
  • Copyright: © Teng et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

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


Abstract

Glycyrrhizic acid (GA), a major compound separated from Radix Glycyrrhizae, has been shwon to exert various biochemical effects, including neuroprotective effects. In the present study, we investigated the protective effects of GA against 1-methyl-4-phenylpyridinium (MPP+)‑induced damage to differentiated PC12 (DPC12) cells. Compared with the MPP+-treated cells, GA markedly improved cell viability, restored mitochondrial dysfunction, suppressed the overexpression of cleaved poly(ADP-ribose) polymerase (PARP), and suppressed the overproduction of lactate dehydrogenase (LDH) and intracellular Ca2+ overload. The protective effects of GA on cell survival were further confirmed in primary cortical neurons. GA markedly increased the expression of phosphorylated extracellular signal-regulated kinase (p-ERK), as well as its migration from the cytoplasm to nucleus. PD98059, an inhibitor of ERK, blocked GA-enhanced ERK activation and reduced cell viability. However, pre-treatment with GA had no effects on the expression of phosphorylated AKT (p-AKT) and total AKT (t-AKT). These results indicate that the GA-mediated neuroprotective effects are associated with its modulation of multiple anti-apoptotic and pro-apoptotic factors, particularly the ERK signaling pathway. This study provides evidence supporting the use of GA as a potential therapeutic agent for the treatment of neurodegenerative diseases and neuronal injury.

Introduction

Parkinson’s disease (PD) is a recognized progressive neurodegenerative disorder which is caused by a decrease in the number of dopaminergic neurons located in the substantia nigra (1,2). Although the etiology of the disease remains unknown, postmortem studies have indicated that dying cells not only bear signs of necrosis, but also of apoptosis, in particular chromatin condensation, DNA fragmentation, oxidative damage, mitochondrial dysfunction and caspase activation (3,4). 1-Methyl-4-phenylpyridinium (MPP+) is commonly used in vitro and in vivo to produce models of PD (5,6). Similar to other dopaminergic toxins, MPP+ causes oxidative stress and the selective death of dopaminerigic neuronal cells, such as PC12 (7) and SH-SY5Y cells (8).

A number of studies have reported that herbal preparations and their natural compounds display broad protective effects against neurotoxicity in various neurodegenerative diseases (9,10). Glycyrrhizic acid (GA), a major active ingredient separated from Radix Glycyrrhizae, possesses anti-inflammatory and anti-viral effects (11,12). It has been well documented that GA exerts marked neuroprotective effects against 6-hydroxydopamine- or glutamate-induced damage to neuronal cells (13,14). However, the contribution of GA toward MPP+-induced cell damage and the underlying mechanisms have not yet been fully elucidated.

It is well known that extracellular signal-regulated kinase (ERK) plays a key role in cell proliferation, differentiation, survival and apoptosis (15). The phosphorylation of ERK has been shown to be critical for mediating the neuroprotectives effects of leptin (16). Combined with the activation of ERK, mitochondrial depolarization is associated with apoptotic cell death (17). Another pathway involved in this process is the PI3K/AKT signaling pathway, which is essential for rescuing neuronal cells from oxidative stress (18).

We therefore hypothesized that GA exerts neuroprotective effects against MPP+-induced cell damage. To examine this hypothesis, in this study, we investigated the inhibitory effects of GA on MPP+ cytotoxicity and the underlying mechanisms. Our data revealed that GA attenuated MPP+-induced cell death, the high apoptotic rate, the intracellular Ca2+ overload, the overproduction of lactate dehydrogenase (LDH), as well as mitochondrial dysfunction. Further experiments indicated that the activation of ERK contributes to the GA-mediated neuroprotection of dopaminergic neuronal cells.

Materials and methods

Cell culture

PC12 cells (rat adrenal gland pheochromocytoma cells; obtained from ATCC, Manassas, VA, USA; CRL-1721 passages <10) were maintained as monolayer cultures in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% horse serum (HS), 5% fetal bovine serum (FBS) and penicillin (100 IU/ml), and streptomycin (100 μg/ml) (all from Invitrogen, Carlsbad, CA, USA), under a humidified atmosphere containing 5/95% CO2/air at 37 °C. The culture medium was changed every 3 days. The PC12 cells were treated with 20 ng/ml nerve growth factor (NGF; Sigma-Aldrich, St. Louis, MO, USA) in DMEM supplemented with 1% FBS and 1% HS and incubated for 72 h to induce differentiation.

Primary cultures of neurons were prepared from fetal cortices of pregnant Sprague-Dawley rats [embryonic days (E) 17–18] as previously described (19). Briefly, the neurons were dissociated from the cerebral cortex of embryonic rats and were plated in 96-well culture plates which had been previously coated with poly-D-lysine (Invitrogen). The cells were maintained in neurobasal medium supplemented with 2% B27 and 1% GlutaMAX (both from Invitrogen). The purity of the primary cortical neuronal cells was 88.1±5.6% (Fig. 2A) which was determined by β3-tubulin (red fluorescence) staining using ImageJ software.

Analysis of cell viability and cellular morphology

Cell viability was measured by a quantitative colorimetric assay with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; Sigma-Aldrich) as described in a previous study (20). Briefly, the differentiated PC12 (DPC12) cells and primary neurons were seeded into 96-well plates at 5×104 cells per well. The cells were pre-treated with 5–100 μM GA for 3 h, co-treated with 0.5 or 4 mM MPP+ for 24 h, and then incubated with MTT solution (5 mg/ml) for a further 4 h. A total of 100 μl dimethyl sulfoxide (DMSO) was added to each well and then the absorbance was measured using a microplate reader (Bio-Rad, Berkeley, CA, USA) at 540 nm. Cell viability was expressed as a percentage of the value in the control group (untreated cells). Prior to MTT assay, the morphology of the primary neurons was detected by normal photography (10′, Axio Observer Z1; Carl Zeiss, Inc., Oberkochen, Germany).

Measurement of LDH release

The release of LDH into the culture medium was measured using an In Vitro Toxicology Assay kit (Sigma-Aldrich). Briefly, the PC12 cells were seeded into 96-well plates at 2×104 cells per well. Following differentiation, the cells were pre-treated with 25 μM GA for 3 h and co-treated with 4 mM MPP+ for 24 h. A total of 60 μl mixed assay solution was added into 30 μl culture medium collected from each group. Following 30 min of incubation at room temperature in the dark, 10 μl 1 N HCl were added to terminate the reaction. The absorbance was measured at a wavelength of 490 nm using a microplate reader (Bio-Rad). The values of the treated cells were expressed as a percentage of the corresponding untreated cells.

Mitochondrial membrane potential (MMP) analysis

5,5′,6,6′-Tetrachloro-1,1′,3,3′ tetraethylbenzimidazolylcarbocyanine iodide (JC-1; Sigma-Aldrich) was used to measure changes in MMP. The PC12 cells (1×105) were seeded in 12-well plates and allowed to differentiate. The cells were pre-treated with 25 μM GA for 3 h and co-treated with 4 mM MPP+ for 24 h. The treated DPC12 cells were incubated with 2 μM JC-1 at 37°C for 10 min. After 3 washes with phosphate-buffered saline (PBS), the changes in MMP of the DPC12 cells were analyzed using a fluorescence microscope (×20 magnification; Axio Observer Z1, CCD camera; Carl Zeiss, Inc.).

Measurement of intracellular Ca2+ concentration

Fluo-4 AM (Invitrogen) staining was used to measure the intracellular Ca2+ concentration. The DPC12 cells were pre-treated with 25 μM GA for 3 h and then co-treated with 4 mM MPP+ for a further 3 h. The cells were incubated with Fluo-4 AM (final concentration, 5 μM) at 37°C for 30 min, and washed 3 times to remove the excess probe. The fluorescence intensity was determined using a fluorescence microscope (×40 magnification; Axio Observer Z1, CCD camera; Carl Zeiss). The experiment was repeated 3 times and the average fluorescence intensity for each cell was calculated using ImageJ software.

Flow cytometric analysis of apoptosis

Flow cytometric analysis was used to assess the membrane and nuclear events during apoptosis, as previously described (7). The assay was performed with a two-color analysis of fluorescein isothiocyanate (FITC)-labeled Annexin V binding propidium iodide (PI) (Becton-Dickinson Co., Miami, FL, USA). The DPC12 cells were pre-treated with 25 μM GA for 3 h and then co-treated with 4 mM MPP+ for a further 12 h. The cells were suspended (1×106/ml) in binding buffer and incubated for 10 min with 5 μl Anexin V-FITC (20 μg/ml) and 10 μl PI (50 μg/ml) at room temperature in the dark. The level of fluorescence was analyzed using a flow cytometer (Cytomics™ FC 500; Beckman Coulter, Inc., Brea, CA, USA). The experiment was repeated 3 times.

Western blot analysis

The treated cells were lysed with RIPA buffer containing 1% protease inhibitor cocktails and 2% phenylmethanesulfonyl fluoride (PMSF) (all from Sigma-Aldrich). For the detection of ERK translocation, the preparation of cytoplasmic and nuclear extracts was carried out as described in a previous study by Yang et al (21). After the supernatant collection, the protein concentration was determined using the Bradford method. Proteins were separated on a 10% SDS-PAGE gel and transferred electrophoretically onto nitrocellulose membranes (Bio Basic, Inc., Markham, Ontario, Canada). The transferred membranes were then blotted with primary antibodies (dilution of 1:1,000) to: phosphorylated ERK (p-ERKs), total ERK (t-ERKs), phosphorylated AKT (p-AKT), total AKT (t-AKT), cleaved poly(ADP-ribose) polymerase (PARP), glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and lamin B (Cell Signaling Technology, Inc., Danvers, MA, USA) at 4°C overnight, followed by treatment with horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology, Inc., Dallas, TX, USA). Chemiluminescence was detected using ECL detection kits (GE Healthcare, Buckinghamshire, UK). The intensity of the bands was quantified by scanning densitometry using Quantity One 4.5.0 software (Bio Basic, Inc.).

Statistical analysis

Data are expressed as the means ± SD. One-way ANOVA was applied to determine the statistical significance, followed by post-hoc multiple comparisons (Dunn’s test). A value of P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of GA on cell viability, LDH release and apoptosis of DPC12 cells or primary cortical neurons

Exposure to 4 mM MPP+ for 24 h resulted in 41.9±3.3% cell death compared with the control cells; however, pre-treatment with 25 μM GA significantly attenuated the MPP+-induced cytotoxicity and increased cell viability (58.1±3.3 vs. 69.2±1.3%; P<0.01; Fig. 1A). The protective effects of GA (25 μM) on cell viability were further confirmed in primary neurons (48.2±0.8 vs. 65.3±1.1%; P<0.01; Fig. 2B). Pre-treatment with 25 μM GA for 3 h following exposure to 4 mM MPP+ for 24 h markedly suppressed the overproduction of LDH induced by MPP+ in the DPC12 cells (108.4±2.9 vs. 142.7±12.2%; P<0.01; Fig. 1B). Moreover, double-staining with Annexin V-FITC and PI was used to detect the apoptotic rate of DPC12 cells following treatment. The increase in the apoptotic rate of the MPP+-exposed cells was reversed by pre-treatment with 25 μM GA (7.2 vs. 19.1%; Fig. 1C). The morphological changes in the primary cortical neurons were visualized by phase-contrast imaging. Compared with the untreated cells, shrinkage and detachment were observed following exposure to MPP+. Pre-treatment with 25 μM GA markedly reversed the morphological damage caused by MPP+ (Fig. 2C).

Effects of GA on mitochondrial dysfunction, intracellular Ca2+ overload and the expression of cleaved PARP

MMP related to mitochondrial permeability plays an important role in the cell apoptotic pathway. Compared with the MPP+-exposed cells, after 12 h of co-treatment, 25 μM GA restored the dissipation of MMP indicated by an increase in the emission of red fluorescence (Fig. 3A).

The results from Fluo-4 AM staining showed that after 3 h of incubation of the PC12 cells, GA mitigated the calcium overload caused by MPP+, indicated by a reduction in the emission of green fluorescence (Fig. 3B).

Furthermore, the level of cleaved PARP, a hallmark of apoptosis, was determined by western blot analysis. A significant enhancement in the expression of cleaved PARP was observed in the DPC12 cells exposed to 4 mM MPP+ for 24 h. Conversely, pre-treatment with 25 μM GA reduced the overexpression of cleaved PARP (P<0.05; Fig. 4).

The ERK, but not the AKT pathway contributes to the GA-mediated neuroprotective effects

Exposure to MPP+ significantly suppressed the expression of p-ERK, but not that of t-ERK (Fig. 5A). Treatment with GA (25 μM) alone increased the phosphorylation of ERK after 30 min of incubation (P<0.01; Fig. 5B).Pre-treatment with GA (25 μM) markedly reversed the decrease in the expression of p-ERK caused by MPP+ at 10 and 30 min of treatment (Fig. 5C). Furthermore, the translocation of activated ERK from the cytoplasm to the nucleus was determined. Following exposure to 4 mM MPP+ for 60 min, the expression of p-ERK in the nucleus was reduced; by contrast, pre-treatment with GA for 3 h significantly reversed the MPP+-induced suppression of the translocation of ERK to the nucleus (Fig. 5E).

Additionally, treatment with MPP+ alone, GA alone and MPP+ plus GA had no effects on the expression of p-AKT and t-AKT (Fig. 6A–D).

Further experiments revealed that the GA-induced increase in the phosphorylation of ERK and the improvement in cell viability were markedly abrogated by pre-treatment with 10 μM PD98059, an ERK inhibitor (Fig. 7A and B). Collectively, our data suggest that the ERK, but not the AKT signaling pathway contributes to the GA-mediated neuroprotective effects against MPP+-induced DPC12 cell damage.

Discussion

PC12 cells, which possess a dopamine synthesis, metabolism and transporting system (22), can shift their phenotype, changing from proliferating, undifferentiated cells into post-mitotic, differentiated, neurite-bearing neurons following incubation with NGF (23). Studies have demonstrated that DPC12 cells are more sensitive to neurotoxins (24). The present study clearly confirms that GA exerts marked neuroprotective effects, as evidenced by our results: GA greatly ameliorated the MPP+-induced reduction in cell viability, the increased apoptotic rate, the intracellular Ca2+ overload and the dissipation in MMP in the DPC12 cells. GA also exerted suppressive effects on the overproduction of intracellular LDH caused by MPP+. Consistent with the results of previous studies (25,26), this suppressive effect on the LDH level is a consequence of the protective effects of GA against neurotoxicity. PARP is a family of proteins involved in a number of cellular processes, including DNA repair and programmed cell death (27). GA markedly suppressed the enhanced expression of cleaved PARP caused by MPP+.

The pro-survival activation of ERK is well known in dopaminergic neurons (16,28). The phosphorylation of ERK in SH-SY5Y cells has been shown to be suppressed after 4 h of exposure to MPP+ (29). Our study confirmed that MPP+ reduced the phosporylation of ERK; by contrast, GA reversed the MPP+-mediated inhibition of ERK activation. Additionally, the presence of PD98059 eradicated the effects of GA on ERK activation and protection of cell viability. Compared with the MPP+-treated cells, treatment with GA resulted in an increase in p-ERK nuclear migration. It has previously been demonstrated that activated ERK migrates to the nucleus where it regulates transcription factors, leading to changes in gene expression and cell proliferation (30). Collectively, our data indicate that the ERK signaling pathway contributes to the GA-mediated protective effects against MPP+-induced DPC12 cell damage.

Our data further indicated that pre-treatment with GA markedly blocked the calcium influx caused by MPP+. Excessive cytosolic calcium causes a wide range of subcellular pathological responses, in particular the dysfunction of mitochondrial membrane permeability (31). Growing experimental evidence suggests that the mitochondrial-dependent pathway plays a central role in cell apoptosis (17). A key feature of mitochondrial apoptosis is the disruption of the membrane potential, mainly caused by increased membrane permeability (32). The present study demonstrated that GA restored the dissipation of MMP and promoted the activation of ERK. As reported previously, the activation of ERK regulates mitochondrial function (33,34). Previous studies have demonstrated that ERK inhibitors downregulate the expression of B-cell lymphoma-2 (Bcl-2) and Bcl-extra large (Bcl-xL) (33), which are located in the outer membrane of the mitochondria and regulate mitochondrial function (35). However, the association between the activation of ERK and mitochondrial function and their involvement in the GA-mediated neuroprotective effects require further investigation.

It has been demonstrated that GA exerts neuroprotective effects against 6-hydroxydopamine-induced cytotoxicity in PC12 cells via PI3K/AKT pathway (13); however, in our study, we did not observe any significant effects on the activation of AKT following treatment with GA or MPP+. In another separate experiment, GA did not exert any effects on the glutamate-induced decrease in the level of p-AKT. The different results noted in our study may due to the different microenvironmental system. Furthermore, the accumulation of cells in the G1 phase is considered one of the factors responsible for MPP+-induced cell damage (36). In our study, MPP+-induced G1 phase arrest was observed in the DPC12 cells; however, pre-treatment with GA failed to reverse this effect (data not shown).

In conclusion, our data demonstrate that GA exerts significant protective effects on neuronal cells against MPP+ neurotoxicity, as indicated by the suppression of the intracellular Ca2+ overload, the restoration of mitochondrial dysfunction, and the increase in the expression and migration of p-ERK. The present findings provide pharmacological evidence to support the therapeutic application of GA in the treatment of neurodegenerative diseases.

Acknowledgements

This study was supported by the National Science and Technology support program of P.R. China (grant no. 2012BAL29B05) and the Major Scientific and Technological Double Ten Project in Jilin Province (20130201006ZY).

References

1 

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

2 

Surendran S and Rajasankar S: Parkinson’s disease: oxidative stress and therapeutic approaches. Neurol Sci. 31:531–540. 2010.

3 

Hisahara S and Shimohama S: Toxin-induced and genetic animal models of Parkinson’s disease. Parkinsons Dis. 2011:9517092010.

4 

Schulz JB and Falkenburger BH: Neuronal pathology in Parkinson’s disease. Cell Tissue Res. 318:135–147. 2004.

5 

Sonsalla PK, Zeevalk GD and German DC: Chronic intraventricular administration of 1-methyl-4-phenylpyridinium as a progressive model of Parkinson’s disease. Parkinsonism Relat Disord. 14(Suppl 2): S116–S118. 2008.

6 

Li X, Ye X, Sun X, et al: Salidroside protects against MPP(+)-induced apoptosis in PC12 cells by inhibiting the NO pathway. Brain Res. 1382:9–18. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Qin R, Li X, Li G, et al: Protection by tetrahydroxystilbene glucoside against neurotoxicity induced by MPP+: the involvement of PI3K/Akt pathway activation. Toxicol Lett. 202:1–7. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Halvorsen EM, Dennis J, Keeney P, Sturgill TW, Tuttle JB and Bennett JB Jr: Methylpyridinium (MPP(+))-and nerve growth factor-induced changes in pro- and anti-apoptotic signaling pathways in SH-SY5Y neuroblastoma cells. Brain Res. 952:98–110. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Kim EH, Jang MH, Shin MC, Shin MS and Kim CJ: Protective effect of aqueous extract of Ginseng radix against 1-methyl-4-phenylpyridinium-induced apoptosis in PC12 cells. Biol Pharm Bull. 26:1668–1673. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Choi JG, Kim HG, Kim MC, et al: Polygalae radix inhibits toxin-induced neuronal death in the Parkinson’s disease models. J Ethnopharmacol. 134:414–421. 2011.PubMed/NCBI

11 

Matsui S, Matsumoto H, Sonoda Y, et al: Glycyrrhizin and related compounds down-regulate production of inflammatory chemokines IL-8 and eotaxin 1 in a human lung fibroblast cell line. Int Immunopharmacol. 4:1633–1644. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Cinatl J, Morgenstern B, Bauer G, Chandra P, Rabenau H and Doerr HW: Glycyrrhizin, an active component of liquorice roots, and replication of SARS-associated coronavirus. Lancet. 361:2045–2046. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Kao TC, Shyu MH and Yen GC: Neuroprotective effects of glycyrrhizic acid and 18beta-glycyrrhetinic acid in PC12 cells via modulation of the PI3K/Akt pathway. J Agric Food Chem. 57:754–761. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Cherng JM, Lin HJ, Hung MS, Lin YR, Chan MH and Lin JC: Inhibition of nuclear factor kappaB is associated with neuroprotective effects of glycyrrhizic acid on glutamate-induced excitotoxicity in primary neurons. Eur J Pharmacol. 547:10–21. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Lin YL, Wang GJ, Huang CL, et al: Ligusticum chuanxiong as a potential neuroprotectant for preventing serum deprivation-induced apoptosis in rat pheochromocytoma cells: functional roles of mitogen-activated protein kinases. J Ethnopharmacol. 122:417–423. 2009. View Article : Google Scholar

16 

Weng Z, Signore AP, Gao Y, et al: Leptin protects against 6-hydroxydopamine-induced dopaminergic cell death via mitogen-activated protein kinase signaling. J Biol Chem. 282:34479–34491. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Lee CS, Kim YJ, Lee MS, Han ES and Lee SJ: 18beta-Glycyrrhetinic acid induces apoptotic cell death in SiHa cells and exhibits a synergistic effect against antibiotic anti-cancer drug toxicity. Life Sci. 83:481–489. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Lu S, Lu C, Han Q, et al: Adipose-derived mesenchymal stem cells protect PC12 cells from glutamate excitotoxicity-induced apoptosis by upregulation of XIAP through PI3-K/Akt activation. Toxicology. 279:189–195. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Wang JY, Chi SI and Hwang CP: Effects of various nitric oxide synthase inhibitors on NMDA-induced neuronal injury in rat cortical neurons. Chin J Physiol. 39:227–233. 1996.PubMed/NCBI

20 

Ocazionez RE, Meneses R, Torres FA and Stashenko E: Virucidal activity of Colombian Lippia essential oils on dengue virus replication in vitro. Mem Inst Oswaldo Cruz. 105:304–309. 2010.PubMed/NCBI

21 

Yang CL, Chik SC, Li JC, Cheung BK and Lau AS: Identification of the bioactive constituent and its mechanisms of action in mediating the anti-inflammatory effects of black cohosh and related Cimicifuga species on human primary blood macrophages. J Med Chem. 52:6707–6715. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Tuler SM, Hazen AA and Bowen JM: Release and metabolism of dopamine in a clonal line of pheochromocytoma (PC12) cells exposed to fenthion. Fundam Appl Toxicol. 13:484–492. 1989. View Article : Google Scholar : PubMed/NCBI

23 

Greene LA and Tischler AS: Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci USA. 73:2424–2428. 1976. View Article : Google Scholar

24 

Tahir SK, Trogadis JE, Stevens JK and Zimmerman AM: Cytoskeletal organization following cannabinoid treatment in undifferentiated and differentiated PC12 cells. Biochem Cell Biol. 70:1159–73. 1992. View Article : Google Scholar : PubMed/NCBI

25 

Cao BY, Yang YP, Luo WF, et al: Paeoniflorin, a potent natural compound, protects PC12 cells from MPP+and acidic damage via autophagic pathway. J Ethnopharmacol. 131:122–129. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Zandbergen EG, de Haan RJ and Hijdra A: Systematic review of prediction of poor outcome in anoxic-ischaemic coma with biochemical markers of brain damage. Intensive Care Med. 27:1661–1667. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Piskunova TS, Yurova MN, Ovsyannikov AI, et al: Deficiency in poly(ADP-ribose) polymerase-1 (PARP-1) accelerates aging and spontaneous carcinogenesis in mice. Curr Gerontol Geriatr Res. 2008:7541902008. View Article : Google Scholar : PubMed/NCBI

28 

Hetman M and Gozdz A: Role of extracellular signal regulated kinases 1 and 2 in neuronal survival. Eur J Biochem. 271:2050–2055. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Zhu JH, Horbinski C, Guo F, Watkins S, Uchiyama Y and Chu CT: Regulation of autophagy by extracellular signal-regulated protein kinases during 1-methyl-4-phenylpyridinium-induced cell death. Am J Pathol. 170:75–86. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Zuber J, Tchernitsa OI, Hinzmann B, et al: A genome-wide survey of RAS transformation targets. Nat Genet. 24:144–152. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Giorgi C, Baldassari F, Bononi A, et al: Mitochondrial Ca(2+) and apoptosis. Cell Calcium. 52:36–43. 2012. View Article : Google Scholar

32 

Mayer B and Oberbauer R: Mitochondrial regulation of apoptosis. News Physiol Sci. 18:89–94. 2003.

33 

Boucher MJ, Morisset J, Vachon PH, Reed JC, Laine J and Rivard N: MEK/ERK signaling pathway regulates the expression of Bcl-2, Bcl-X(L), and Mcl-1 and promotes survival of human pancreatic cancer cells. J Cell Biochem. 79:355–369. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Galante JM, Mortenson MM, Bowles TL, Virudachalam S and Bold RJ: ERK/BCL-2 pathway in the resistance of pancreatic cancer to anoikis. J Surg Res. 152:18–25. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Chan SL and Yu VC: Proteins of the bcl-2 family in apoptosis signalling: from mechanistic insights to therapeutic opportunities. Clin Exp Pharmacol Physiol. 31:119–128. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Bai J, Nakamura H, Ueda S, et al: Proteasome-dependent degradation of cyclin D1 in 1-methyl-4-phenylpyridinium ion (MPP+)-induced cell cycle arrest. J Biol Chem. 279:38710–38714. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2014
Volume 34 Issue 3

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
Teng L, Kou C, Lu C, Xu J, Xie J, Lu J, Liu Y, Wang Z and Wang D: Involvement of the ERK pathway in the protective effects of glycyrrhizic acid against the MPP+-induced apoptosis of dopaminergic neuronal cells. Int J Mol Med 34: 742-748, 2014
APA
Teng, L., Kou, C., Lu, C., Xu, J., Xie, J., Lu, J. ... Wang, D. (2014). Involvement of the ERK pathway in the protective effects of glycyrrhizic acid against the MPP+-induced apoptosis of dopaminergic neuronal cells. International Journal of Molecular Medicine, 34, 742-748. https://doi.org/10.3892/ijmm.2014.1830
MLA
Teng, L., Kou, C., Lu, C., Xu, J., Xie, J., Lu, J., Liu, Y., Wang, Z., Wang, D."Involvement of the ERK pathway in the protective effects of glycyrrhizic acid against the MPP+-induced apoptosis of dopaminergic neuronal cells". International Journal of Molecular Medicine 34.3 (2014): 742-748.
Chicago
Teng, L., Kou, C., Lu, C., Xu, J., Xie, J., Lu, J., Liu, Y., Wang, Z., Wang, D."Involvement of the ERK pathway in the protective effects of glycyrrhizic acid against the MPP+-induced apoptosis of dopaminergic neuronal cells". International Journal of Molecular Medicine 34, no. 3 (2014): 742-748. https://doi.org/10.3892/ijmm.2014.1830