Open Access

Salidroside protects PC-12 cells against amyloid β-induced apoptosis by activation of the ERK1/2 and AKT signaling pathways

  • Authors:
    • Zheng‑Luan Liao
    • Heng Su
    • Yun‑Fei Tan
    • Ya‑Ju Qiu
    • Jun‑Peng Zhu
    • Yan Chen
    • Si‑Si Lin
    • Ming‑Hao Wu
    • Yan‑Ping Mao
    • Jiao‑Jiao Hu
    • En‑Yan Yu
  • View Affiliations

  • Published online on: February 1, 2019     https://doi.org/10.3892/ijmm.2019.4088
  • Pages: 1769-1777
  • Copyright: © Liao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Alzheimer's disease (AD) is one of the most frequent diseases in elderly people and causes high mortality. Its incidence is increasing annually and no effective therapeutic treatment currently exists. In the present study, salidroside, a major active ingredient of Rhodiola rosea, was able to protect PC‑12 cells from the toxicity and apoptosis induced by AD inducer amyloid (A)β1‑42. Salidroside significantly protected PC‑12 cells by inhibiting Aβ1‑42‑induced cytotoxicity and mitochondria‑mediated endogenous caspase apoptotic pathways. Mechanistic studies demonstrated that salidroside significantly activated the extracellular signal regulated kinase (ERK)1/2 and protein kinase B (AKT) signaling pathways. This observation was further confirmed using the ERK1/2 inhibitor PD98059 and the AKT inhibitor LY294002, which demonstrated that salidroside promoted PC‑12 cell survival and proliferation by activating the ERK1/2 and AKT signaling pathways. Salidroside is a therapeutic candidate for the treatment of AD and provides a basis for further drug development.

Introduction

Alzheimer’s disease (AD) is a major neurodegenerative disease characterized by significant memory impairment in addition to other cognitive impairments and maybe associated with mental symptoms and behavioral abnormalities (1-3). AD is prevalent in the elderly of >65 years of age, with 1 newly diagnosed case every 3 sec worldwide (4). The number of patients with AD is estimated to increase to 135.46 million by 2050, thereby leading to serious social issues in addition to the health, economic and social burden (5).

The accumulation of amyloid β (Aβ) and mutation of gene polymorphisms of clusterin (CLU) can be attributed to AD (6,7). Aβ is cleaved from the amyloid precursor protein and a mutation in CLU causes the brain to lose its Aβ-scavenging function. Of note, an excessive accumulation of Aβ has been detected in the brains of patients with AD (8-10). Accumulation of Aβ can eventually cause the cytotoxic death of nerve cells due to oxidative stress, decreased mitochondrial membrane potential and nuclear pyknosis (11-13). At present, modulation of Aβ-induced neurotoxicity is an an effective therapeutic approach for the treatment of AD; however, there are no safe and effective therapeutics for AD at present (14-16). Therefore, the identification and evaluation of potential protective candidates for AD treatment are necessary.

Salidroside is a major active ingredient extracted from Rhodiola rosea (17); it is widely employed in traditional Chinese medicine and has been reported to exert anti-inflammatory, anti-oxidative and anti-autophagic effects (18-20). Based on these functions, it has been speculated that salidroside may be able to treat AD. However, the molecular mechanisms underlying these effects of salidroside are currently not well understood and further research is required to clarify them. The present study aimed to confirm the anti-AD effects of salidroside and unravel its mechanism of action. The toxic effects of Aβ in the PC-12 cell line were established for use as an in vitro AD model for drug evaluation (21). The results revealed that salidroside could effectively inhibit the toxicity and apoptosis of PC-12 cells that was induced by Aβ. Furthermore, the protective effect of salidroside against Aβ-induced damage in PC-12 cells was mediated by activation of the extracellular signal regulated kinase (ERK)1/2 and protein kinase B (AKT) signaling pathways. By promoting cell survival and proliferation, the toxic effects of Aβ were effectively inhibited by salidroside, thereby further demonstrating that salidroside is a potential candidate for AD treatment.

Materials and methods

Cell viability assay

Cell viability was evaluated using cytotoxicity assays. Briefly, PC-12 cells were seeded into 96-well plates with 5,000 cells per well and incubated with drugs or inhibitors at the indicated concentrations for 48 h. The salidroside was added at the concentrations of 12.5, 25, 50, 100 or 200 µM; Aβ1-42 was added at concentrations of 0.01 to 1 µM; while the inhibitors were added at concentrations of 5 to 20 µM. A volume of 25 µl MTT solution (5 mg/ml) was added to each well and incubated for additional 4 h. Then, dimethyl sulfoxide (DMSO) was added to dissolve the MTT formazan product and the absorbance was determined at 570 nm using a SpectraMax M5 device (Molecular Devices, LLC, Sunnyvale, CA, USA). The relative cell viability rates of the test group were calculated vs. the untreated controls.

Lactate dehydrogenase (LDH) assay

The quantity of LDH that is released into the incubation medium when the cell membrane is destroyed was determined for evaluation of Aβ-induced cytotoxicity. A total of 5×103 PC-12 cells were seeded into 96-well plates. After incubation with 50 µM salidroside for 1 h, followed by incubation with Aβ for additional 24 h, the activity of the LDH that was released into the medium was determined according to the protocol of CytoTox-ONE™ Homogeneous Membrane Integrity Assay (Promega Corporation, Madison, WI, USA). The fluorescent intensity was determined using a microplate reader (Multiskan™ FC; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at an excitation wavelength of 560 nm and an emission wavelength of 590 nm. The percentage values of the released LDH were normalized to the control group.

Nuclear staining

PC-002D12 cells were fixed with 3.7% formaldehyde in PBS for 5 min at room temperature and blocked with 5% bovine serum albumin (Sangon Biotech Co., Ltd., Shanghai, China) containing 0.1% Triton X-100 (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for 30 min at room temperature. The prepared specimens were stained with DAPI (SouthernBiotech, Birmingham, AL, USA) at a concentration of 10 µg/ml at room temperature for 2 min and then observed under a confocal microscope (Nikon Corporation, Tokyo, Japan). Apoptotic cells exhibited nuclear condensation with intensely stained nuclei, while the normal cells did not exhibit nuclear condensation (22).

Determination of intracellular reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) levels

Cells were seeded in 6-well plates at a density of 5×104 cells/cm2. After 24 h of incubation, cells were exposed to 50 µM salidroside for 1 h and to 0.3 µM Aβ for 24 h. Then, some of the cells were incubated with 5 µM fluorescent probe of H2DCF-DA for 20 min and the fluorescence emission was visualized using a fluorescence microscope (Nikon Corporation). The other cells were analysed using the MDA and SOD detection kits (KeyGen Biotech Co., Ltd., Nanjing, China).

Determination of mitochondrial membrane potential

PC-12 cells were seeded into a black 96-well plate with 5,000 cells per well, then incubated with 50 µM salidroside for 1 h, followed by incubation with Aβ. Following 24 h, cells were incubated with a fluorescent probe of JC-1 (Beyotime Institute of Biotechnology, Haimen, China) at a concentration of 5 mg/ml for 15 min at 37°C and then washed twice with PBS. The intensity of red and green fluorescence was determined using an Infinite M200 PRO Multimode Microplate (Tecan Group, Ltd., Mannedorf, Switzerland) and a fluorescence microscope (Nikon Corporation). The mitochondrial membrane potential was calculated using the ratio of JC-1 red/green fluorescence intensity, the color changed from red to green when apoptosis occurred and the value was normalized to the control group.

Caspase-3/7 activity assay

Once PC-12 cells had been incubated with salidroside for 1 h and with Aβ for 24 h, the cells were lysed with lysis buffer of radioimmuno-precipitation assay (RIPA; KeyGen Biotech Co., Ltd.) and centrifuged at 12,500 x g for 5 min at 4°C. Then, the cell lysate was incubated with 2X substrate working solution at room temperature for 30 min in 96-well plates. The activity of caspase-3/7 was determined using the commercially available Caspase-Glo 3/7 Assay (Invitrogen; Thermo Fisher Scientific, Inc.). The fluorescence intensity of each sample was normalized to the protein concentration of the sample. All the percentage values of caspase-3/7 activities were normalized to the control group.

Flow cytometry assay

PC-12 cell apoptosis was evaluated with the Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) Apoptosis Detection kit (KeyGen Biotech Co., Ltd.). Upon treatment, the adherent and non-adherent cells were harvested. The cells were then stained with Annexin V-FITC and PI in binding buffer for 15 min. Flow cytometric analysis was performed on a fluorescence-activated cell sorting flow cytometer (BD Biosciences; Franklin Lakes, NJ, USA) and the data were analyzed with Cell Quest software (version 3.4, BD Biosciences).

Western blotting

The cells were lysed with RIPA buffer, the concentration of proteins were determined by Bicinchoninic Acid detection kit (KeyGen Biotech Co., Ltd.). Immunoblots were performed with samples that contained total protein (30 µg), which had been separated by 12% SDS-PAGE gel and then transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were incubated with primary antibodies against phosphorylated (p)-ERK1/2 (1:1,000; AF1015; Affinity Biosciences, Cincinnati, OH, USA), total (t)-ERK (1:1,000; AF6240; Affinity Biosciences), p-AKT (1:1,000; AF0016; Affinity Biosciences), t-AKT (1:1,000; AF4718; Affinity Biosciences), B-cell lymphoma (BCL)-2 (1:1,000; AF6139; Affinity Biosciences), BCL2 associated X (Bax; AF0120; 1:1,000; Affinity Biosciences) and GAPDH (1:1,000; AF7021; Affinity Biosciences), followed by incubation with a secondary antibody of goat anti-rabbit IgG-horseradish peroxidase (1:5,000; sc-2004; Santa Cruz Biotechnology, Inc., Dallas, TX, USA). Blots were developed using an enhanced chemiluminescence detection reagent (EMD Millipore). GAPDH was used as an internal loading control.

Statistical analysis

All experiments were repeated at least in triplicate, all data were evaluated using SPSS v.19.0 (IBM Corp., Armonk, NY, USA). Data are presented as the mean ± standard deviation. One-way analysis of variance and Bonferroni’s test were used to compare different treatment samples. P<0.05 was considered to indicate a statistically significant difference.

Results

Salidroside suppresses Aβ1-42-induced cytotoxicity in PC-12 cells in a concentration-dependent manner

The biological activity of salidroside (Fig. 1A) was assessed by cell viability assays. The cytotoxicity of Aβ1-42 on PC-12 cells was first examined by MTT assay. As presented in Fig. 1B, exposure of PC-12 cells to Aβ1-42 (0.001-1 µM) for 24 h induced a dose-dependent decrease in cell viability, thereby demonstrating that Aβ1-42 could induce toxicity in PC-12 cells. The viability of PC-12 cells was only increased or slightly decreased and the differences compared with the control group were not significant, following treatment with salidroside (12.5-200 µM) for 24 h, thereby indicating that salidroside was not toxic to PC-12 cells under these treatment conditions (Fig. 1C). Compared with the Aβ1-42 group, pre-treatment with salidroside at concentrations of 25, 50 and 100 µM significantly improved cell viability (P<0.05; Fig. 1D). The multi-combination test results further demonstrated that salidroside protected and rescued PC-12 cells from Aβ1-42-induced cell death (Fig. 1E).

Salidroside suppresses Aβ1-42-induced LDH release and apoptosis in PC-12 cells

LDH assay was used for evaluation of the protective activity of salidroside. When pre-treated with 50 µM salidroside for 1 h, PC-12 cells exhibited a significantly reduced Aβ1-42-induced LDH leakage (from 135-110% relative to the control group; P<0.01; Fig. 2A). Nuclei condensation was detected in PC-12 cells upon exposure to Aβ1-42 by DAPI staining (Fig. 2B). Pre-treatment with 50 µM salidroside significantly inhibited Aβ1-42-induced apoptosis compared with Aβ1-42 alone (from 35-23%; P<0.05 Fig. 2C). To further evaluate the influence of salidroside in response to oxidative stress in PC-12 cells, the cells were treated with Aβ1-42 for 24 h following being incubated with salidroside for 1 h. Cell apoptosis rates were detected by flow cytometry (Fig. 2D). The apoptosis rate of PC-12 cells increased following 24 h of Aβ1-42 treatment, while salidroside treatment could protect PC-12 cells from Aβ1-42-induced apoptosis.

Salidroside affects Aβ1-42 induced ROS, MDA and SOD productions in PC-12 cells

Since the toxicity of Aβ1-42 may be affected by the generation of ROS, MDA and SOD (23), the inhibitory/promotional effect of salidroside on ROS, MDA, and SOD production was examined. PC-12 cells were pre-treated with or without 50 µM salidroside for 1 h and then treated with Aβ1-42 for 24 h. Differences in fluorescence intensity were observed among the different groups (Fig. 3A). The results revealed that Aβ1-42 could effectively induce intracellular ROS generation, whereas salidroside could significantly inhibit the generation of ROS induced by Aβ1-42 (from 145 to 125% relative to control group; P<0.05; Fig. 3B). While the MDA detection results demonstrated that the MDA levels were significantly increased due to oxidative stress injury but significantly decreased by salidroside pre-treatment (P<0.01; Fig. 3C). By contrast, the SOD activity in salidroside-pretreated cells increased compared with Aβ-treated cells (Fig. 3D).

Salidroside improves Aβ1-42-induced alterations in the mitochondrial membrane potential and caspase-3/7 activity

Previous studies reported that the loss of the mitochondrial membrane potential was involved in the progression of neuron apoptosis caused by Aβ1-42 during AD (24,25). In the present study, mitochondrial membrane permeability was detected using the JC-1 probe in order to evaluate the anti-apoptotic effects of salidroside. Red and green fluorescence represented high mitochondrial membrane permeability in viable cells and low mitochondrial membrane permeability in apoptotic cells, respectively (Fig. 4A). When incubated with Aβ1-42 for 24 h, the mitochondrial membrane permeability was significantly depolarized in Aβ1-42-treated PC-12 cells compared with the control (P<0.01), whereas pre-treatment with salidroside effectively prevented the loss of mitochondrial membrane permeability (Fig. 4B). Treatment of PC-12 cells with 0.3 µM Aβ1-42 for 24 h triggered a significant increase in caspase-3/7 activity compared with the control (P<0.01), whereas pre-treatment with 50 µM salidroside for 1 h significantly inhibited caspase-3/7 activity (P<0.05; Fig. 4C).

Salidroside stimulates the phosphorylation of ERK1/2 and AKT in a time and concentration-dependent manner in PC-12 cells

Since the ERK1/2 and AKT signaling pathways are classical apoptosis-associated pathways, the present study evaluated whether the anti-apoptotic effect of salidroside was mediated by the ERK1/2 and AKT signaling pathways. The present study examined the phosphorylated and total expression levels of ERK1/2 and AKT in PC-12 cells treated with salidroside by western blotting. As presented in Fig. 5A-C, the phosphorylation levels of ERK1/2 and AKT gradually increased upon the addition of salidroside in a time and dose-dependent manner.

Activation of the ERK1/2 and AKT signaling pathways mediates the protective effects of salidroside in Aβ1-42-induced PC-12 cells

To confirm the roles of the ERK1/2 and AKT signaling pathways in the inhibitory effect of salidroside on Aβ1-42-induced apoptosis in PC-12 cells, the specific inhibitors of the ERK1/2 and AKT signaling pathways, PD98059 and LY294002, respectively, were used. The two pathway inhibitors blocked the protective effects of salidroside in cells treated with Aβ1-42 (Fig. 6A). PC-12 cells were pre-treated with PD98059 or LY294002 (5, 10 and 20 µM) for 30 min and then treated with 50 µM salidroside for 1 h, and the viability of cells was determined by MTT assay 24 h later. The protective effect of salidroside was blocked in the presence of an all concentrations of PD98059 and LY294002 (Fig. 6B and C). Upon staining with DAPI, pyknosis was detected in treated PC-12 cells (Fig. 6D). Pre-treatment with 50 µM salidroside reversed the effect of Aβ1-42 on PC-12 cells, whereas incubation with PD98059 or LY294002 abolished the protective effect of salidroside (Fig. 6E). The western blot results further demonstrated that upon treatment of cells with Aβ1-42, the incubation with PD98059 or LY294002 for 24 h inhibited the phosphorylation of ERK1/2 and AKT, respectively. Salidroside reversed the decrease in the phosphorylation levels of ERK1/2 and AKT induced by Aβ1-42, whereas PD98059 and LY294002 pre-treatment blocked the reversing effects of salidroside (Fig. 6F). The anti Aβ-induced apoptotic effect of salidroside may be mediated by other signaling pathways, such as BCL-2/Bax (Fig. S1); however, further investigation is required.

Discussion

AD is a multigenetic neurodegenerative disease caused by genetic and environmental factors (26). It is associated with a series of physiological and pathological mechanisms, including Aβ accumulation, abnormal phosphorylation of Tau protein, lipid metabolism, inflammatory reaction and oxidative stress (6,10,11,27). Although hypotheses have been proposed for its pathogenesis, no theory has been completely validated thus far (28). The above physiological and pathological causes eventually lead to a neuroinflammatory reaction at the disease site, thereby resulting in nerve cell damage or apoptosis (29). Therefore, compounds that can effectively inhibit the nerve cell apoptosis caused by cytotoxicity-like oxidative stress may be potential therapeutic candidates for AD.

Salidroside has been reported to have therapeutic effects on various diseases including cancer, pulmonary fibrosis and cerebrovascular disease, mainly due to its pharmacological activities, including anti-fatigue, anti-aging, anti-apoptosis, immune regulation and free-radical scavenging (19,30,31). Based on these characteristics, salidroside was selected in the present study to detect its effects on AD treatment. The toxic effects of Aβ on the PC-12 cell line were used in the present study for the in vitro screening model. Aβ is a small peptide that consists of 42 amino acids and is cleaved from its precursor protein. The full length and fragments of Aβ include Aβ1-42, Aβ1-40 and Aβ25-35, which can be used as an inducer (32). Among these fragments, Aβ1-42 has the best induction effect of cell apoptosis (33). Therefore, Aβ1-42 was used in the present study to establish an in vitro AD model and to conduct pharmacodynamic tests. Salidroside effectively improved cell apoptosis induced by cell pyknosis, oxidative stress and mitochondrial membrane potential decrease in Aβ-induced PC-12 cells. Therefore, salidroside was also most likely to exhibit activity for treating AD in vivo systems, which needs further evaluation.

Apoptosis involves multiple signaling pathways, including ERK1/2 and AKT (34,35). Therefore, upon confirmation of the anti-apoptotic effect of salidroside, the effect of salidroside on these two signaling pathways was examined. Salidroside significantly activated the ERK1/2 and AKT signaling pathways. To further confirm the effect exerted by the ERK1/2 and AKT signaling pathways, the ERK1/2 inhibitor PD98059 and the AKT inhibitor LY294002 were used (36,37). The results were consistent with those from previous experiments.

In conclusion, salidroside effectively inhibited the apoptosis of Aβ-induced PC-12 cells by activating the ERK1/2 and AKT signaling pathways, thereby indicating that salidroside is a potential candidate for the treatment of AD. The present study provides a basis for further drug development.

Supplementary Materials

Funding

The present study was supported by the Natural Science Foundation of China (grant no. 81771158), Science Foundation from Health Commission of Zhejiang Province (grant no. ZKJ-ZJ-1503, 2018278601 and 2019321345).

Availability of data and materials

The data used and analyzed in this study are available from the corresponding author on reasonable request.

Authors’ contributions

EYY and ZLL made substantial contributions to the design of the present study. HS, YFT, YJQ, and JPZ performed the cell viability and apoptosis-associated experiments. YC, SSL and MHW performed all other experiments. ZLL, YPM, and JJH analyzed data. EYY and ZLL wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

No human trials were involved in this study.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Jarrett JT and Lansbury PT Jr: Seeding ‘one-dimensional crystallization’ of amyloid: A pathogenic mechanism in Alzheimer’s disease and scrapie? Cell. 73:1055–1058. 1993. View Article : Google Scholar : PubMed/NCBI

2 

Winblad B, Palmer K, Kivipelto M, Jelic V, Fratiglioni L, Wahlund LO, Nordberg A, Bäckman L, Albert M, Almkvist O, et al: Mild cognitive impairment-beyond controversies, towards a consensus: Report of the international working group on mild cognitive impairment. J Intern Med. 256:240–246. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Coyle JT, Price DL and DeLong MR: Alzheimer’s disease: A disorder of cortical cholinergic innervation. Science. 219:1184–1190. 1983. View Article : Google Scholar : PubMed/NCBI

4 

Alzheimer’s A: 2011 Alzheimer’s disease facts and figures. W V Med J. 107:82–83. 2011.

5 

Colucci L, Bosco M, Fasanaro AM, Gaeta GL, Ricci G and Amenta F: Alzheimer’s disease costs: What we know and what we should take into account. J Alzheimers Dis. 42:1311–1324. 2014. View Article : Google Scholar

6 

Ittner LM and Gotz J: Amyloid-β and tau - a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci. 12:65–72. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Harold D, Abraham R, Hollingworth P, Sims R, Gerrish A, Hamshere ML, Pahwa JS, Moskvina V, Dowzell K, Williams A, et al: Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet. 41:1088–1093. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Bertram L and Tanzi RE: Alzheimer disease: New light on an old CLU. Nat Rev Neurol. 6:11–13. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Selkoe DJ: The cell biology of beta-amyloid precursor protein and presenilin in Alzheimer’s disease. Trends Cell Biol. 8:447–453. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Love S and Kehoe PG: Clearance of Abeta from the brain in Alzheimer’s disease. Foreword. Brain Pathol. 18:2392008. View Article : Google Scholar

11 

Akwa Y, Allain H, Bentue-Ferrer D, Berr C, Bordet R, Geerts H, Nieoullon A, Onteniente B and Vercelletto M: Neuroprotection and neurodegenerative diseases: From biology to clinical practice. Alzheimer Dis Assoc Disord. 19:226–239. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT and Brinton RD: Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 106:14670–14675. 2009. View Article : Google Scholar

13 

Zeng Z, Xu J and Zheng W: Artemisinin protects PC12 cells against β-amyloid-induced apoptosis through activation of the ERK1/2 signaling pathway. Redox Biol. 12:625–633. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Godyn J, Jonczyk J, Panek D and Malawska B: Therapeutic strategies for Alzheimer’s disease in clinical trials. Pharmacol Rep. 68:127–138. 2016. View Article : Google Scholar

15 

Cummings JL, Morstorf T and Zhong K: Alzheimer’s disease drug-development pipeline: Few candidates, frequent failures. Alzheimers Res Ther. 6:372014. View Article : Google Scholar

16 

Selkoe DJ and Hardy J: The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 8:595–608. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Abidov M, Crendal F, Grachev S, Seifulla R and Ziegenfuss T: Effect of extracts from Rhodiola rosea and Rhodiola crenulata (Crassulaceae) roots on ATP content in mitochondria of skeletal muscles. Bull Exp Biol Med. 136:585–587. 2003. View Article : Google Scholar

18 

Zhang J, Zhen YF, Pu-Bu-Ci-Ren, Song LG, Kong WN, Shao TM, Li X and Chai XQ: Salidroside attenuates beta amyloid-induced cognitive deficits via modulating oxidative stress and inflammatory mediators in rat hippocampus. Behav Brain Res. 244:70–81. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Liu Z, Li X, Simoneau AR, Jafari M and Zi X: Rhodiola rosea extracts and salidroside decrease the growth of bladder cancer cell lines via inhibition of the mTOR pathway and induction of autophagy. Mol Carcinog. 51:257–267. 2012. View Article : Google Scholar

20 

Zhang L, Yu H, Zhao X, Lin X, Tan C, Cao G and Wang Z: Neuroprotective effects of salidroside against beta-amyloid-induced oxidative stress in SH-SY5Y human neuroblastoma cells. Neurochem Int. 57:547–555. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Hu M, Waring JF, Gopalakrishnan M and Li J: Role of GSK-3beta activation and alpha7 nAChRs in Abeta(1–42)-induced tau phosphorylation in PC12 cells. J Neurochem. 106:1371–1377. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Mahbub AA, Le Maitre CL, Haywood-Small SL, McDougall GJ, Cross NA and Jordan-Mahy N: Differential effects of poly-phenols on proliferation and apoptosis in human myeloid and lymphoid leukemia cell lines. Anticancer Agents Med Chem. 13:1601–1613. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Simon HU, Haj-Yehia A and Levi-Schaffer F: Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis. 5:415–418. 2000. View Article : Google Scholar

24 

Ly JD, Grubb DR and Lawen A: The mitochondrial membrane potential (deltapsi(m)) in apoptosis. An update Apoptosis. 8:115–128. 2003. View Article : Google Scholar

25 

Green DR and Reed JC: Mitochondria and apoptosis. Science. 281:1309–1312. 1998. View Article : Google Scholar : PubMed/NCBI

26 

Bufill E, Bartes A, Moral A, Casadevall T, Codinachs M, Zapater E, Carles Rovira J, Roura P, Oliva R and Blesa R: Genetic and environmental factors that may influence in the senile form of Alzheimer’s disease: Nested case control studies. Neurologia. 24:108–112. 2009.In Spanish. PubMed/NCBI

27 

Di Paolo G and Kim TW: Linking lipids to Alzheimer’s disease: Cholesterol and beyond. Nat Rev Neurosci. 12:284–296. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Butterfield DA and Boyd-Kimball D: Oxidative stress, amyloid-β peptide, and altered key molecular pathways in the pathogenesis and progression of Alzheimer’s disease. J Alzheimers Dis. 62:1345–1367. 2018. View Article : Google Scholar :

29 

Knezevic D and Mizrahi R: Molecular imaging of neuroinflammation in Alzheimer’s disease and mild cognitive impairment. Prog Neuropsychopharmacol Biol Psychiatry. 80:123–131. 2018. View Article : Google Scholar

30 

Ma YG, Wang JW, Zhang YB, Wang BF, Dai ZJ, Xie MJ and Kang HF: Salidroside improved cerebrovascular vasodilation in streptozotocin-induced diabetic rats through restoring the function of BKCa channel in smooth muscle cells. Cell Tissue Res. 370:365–377. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Liu X, Bai Y, Liang L, Feng J and Deng F: Salidroside improves pulmonary fibrosis by down-regulation of cathepsin B and NF-κBp65 in rats. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 42:128–133. 2017.In Chinese. PubMed/NCBI

32 

Richter L, Munter LM, Ness J, Hildebrand PW, Dasari M, Unterreitmeier S, Bulic B, Beyermann M, Gust R, Reif B, et al: Amyloid beta 42 peptide (Abeta42)-lowering compounds directly bind to Aβ and interfere with amyloid precursor protein (APP) transmembrane dimerization. Proc Natl Acad Sci USA. 107:14597–14602. 2010. View Article : Google Scholar

33 

Postu PA, Noumedem JAK, Cioanca O, Hancianu M, Mihasan M, Ciorpac M, Gorgan DL, Petre BA and Hritcu L: Lactuca capensis reverses memory deficits in Aβ1–42-induced an animal model of Alzheimer’s disease. J Cell Mol Med. 22:111–122. 2018. View Article : Google Scholar

34 

Lu Z and Xu S: ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life. 58:621–631. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Franke TF, Hornik CP, Segev L, Shostak GA and Sugimoto C: PI3K/Akt and apoptosis: Size matters. Oncogene. 22:8983–8998. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Imai Y, Yamagishi H, Ono Y and Ueda Y: Versatile inhibitory effects of the flavonoid-derived PI3K/Akt inhibitor, LY294002, on ATP-binding cassette transporters that characterize stem cells. Clin Transl Med. 1:242012. View Article : Google Scholar

37 

Zhu C, Qi X, Chen Y, Sun B, Dai Y and Gu Y: PI3K/Akt and MAPK/ERK1/2 signaling pathways are involved in IGF-1-induced VEGF-C upregulation in breast cancer. J Cancer Res Clin Oncol. 137:1587–1594. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2019
Volume 43 Issue 4

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
Liao ZL, Su H, Tan YF, Qiu YJ, Zhu JP, Chen Y, Lin SS, Wu MH, Mao YP, Hu JJ, Hu JJ, et al: Salidroside protects PC-12 cells against amyloid β-induced apoptosis by activation of the ERK1/2 and AKT signaling pathways. Int J Mol Med 43: 1769-1777, 2019
APA
Liao, Z., Su, H., Tan, Y., Qiu, Y., Zhu, J., Chen, Y. ... Yu, E. (2019). Salidroside protects PC-12 cells against amyloid β-induced apoptosis by activation of the ERK1/2 and AKT signaling pathways. International Journal of Molecular Medicine, 43, 1769-1777. https://doi.org/10.3892/ijmm.2019.4088
MLA
Liao, Z., Su, H., Tan, Y., Qiu, Y., Zhu, J., Chen, Y., Lin, S., Wu, M., Mao, Y., Hu, J., Yu, E."Salidroside protects PC-12 cells against amyloid β-induced apoptosis by activation of the ERK1/2 and AKT signaling pathways". International Journal of Molecular Medicine 43.4 (2019): 1769-1777.
Chicago
Liao, Z., Su, H., Tan, Y., Qiu, Y., Zhu, J., Chen, Y., Lin, S., Wu, M., Mao, Y., Hu, J., Yu, E."Salidroside protects PC-12 cells against amyloid β-induced apoptosis by activation of the ERK1/2 and AKT signaling pathways". International Journal of Molecular Medicine 43, no. 4 (2019): 1769-1777. https://doi.org/10.3892/ijmm.2019.4088