Dihydroartemisinin inhibits the Raf/ERK/MEK and PI3K/AKT pathways in glioma cells

Corrigendum in: /10.3892/ol.2021.12636

  • Authors:
    • Wei Du
    • Changhe Pang
    • Yake Xue
    • Qingjun Zhang
    • Xinting Wei
  • View Affiliations

  • Published online on: September 15, 2015     https://doi.org/10.3892/ol.2015.3699
  • Pages: 3266-3270
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It has previously been reported that dihydroartemisinin (DHA) is an effective novel anticancer compound in a number of types of tumor cells. Previous studies have demonstrated the anticancer activity of DHA in gioma cells. However, its underlining mechanism remains unclear. In the present study, the anticancer activity of DHA was examined in the glioma cell lines BT325 and C6. Western blot analysis was also employed to determine the signaling pathway changes. It was demonstrated that DHA effectively inhibited cell growth and induced apoptosis in glioma cells. Moreover, western blot analysis indicated that DHA‑induced apoptosis was accompanied by inactivation of the Raf/MEK/ERK and PI3K/AKT signaling pathways, in addition to the downregulation of anti‑apoptotic proteins Mcl‑1 and Bcl‑2 expression levels.

Introduction

Glioma represents the most prevalent and lethal primary brain tumor (1). A number of glioma cases have demonstrated low responses to conventional cytotoxic chemotherapy. Despite great efforts to improve therapeutics, the clinical outcome of glioma remains poor (1). Therefore, there is an urgent need to develop novel treatment to improve the prognosis of glioma (2).

Dysregulated signaling pathways have been considered to be involved in cancer progression; the Raf/mitogen-activated and extracellular signal-regulated kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) cascade and the phosphatidylinositol 3-kinase (PI3K)/Akt cascade are commonly activated pathways in a number of types of cancer (3).

The Raf/MEK/ERK pathway is one of the most common oncogenic pathways and is critical in driving cell proliferation, survival and preventing apoptosis. Upon activation by growth factors, serum, polypeptide hormones, neurotransmitters or chemokines, activated ERK phosphorylates and regulates multiple cytoplasmic signaling proteins and transcription factors (4). These events result in gene expression changes and alterations in cell survival, division and metabolism. A previous study demonstrated that dysregulation of this pathway is involved in the oncogenesis of various types of human cancer (5). Therefore, inhibition of the Raf/MEK/ERK pathway is a promising strategy for cancer therapy (6).

The PI3K/AKT pathway is another crucial signaling pathway for cancer development (7). Growth factors and cytokines activate the lipid kinase PI3K and then generate phosphatidylinositol 3,4,5-triphosphate (PIP3), which activates the recruitment of protein kinase AKT to cell membrane. Activated AKT phosphorylates Ser/Thr residues in proteins that are involved in cell survival, proliferation, and metabolic pathways. Abberrant AKT activation has been documented in a number of types of human cancer (8). Therefore, inhibition of the PI3K/AKT pathway is also a promising strategy for cancer therapy (9).

A previous study indicated that the Raf/MEK/ERK and PI3K/AKT signaling pathways cooperatively link with each other to enhance the proliferation and apoptotic resistance capacity in cancer cells (10). Both signaling pathways are activated by growth factor receptors and their complicated crosstalk at multiple points greatly increases the therapeutic effects of single signaling inhibition in cancer cells (11). In addition, these two pathways can exert complementary and redundant functions when only one single pathway is inhibited (12). Therefore, more clinical benefit may be obtained by simultaneously targeting both cascades (13).

Dihydroartemisinin (DHA) is a semi-synthetic derivative of the anti-malarial drug artemisinin. Previous studies have demonstrated that DHA may exert its anti-proliferation and apoptosis-inducing effects on a number of types of cancer cells, including leukemia, prostate cancer, ovarian cancer and colorectal cancer cells (1417). In addition, it has been reported that DHA induces apoptosis in C6 glioma cells by increasing the generation of reactive oxygen species (ROS) and inhibiting activation of hypoxia inducible factor-1α (HIF-1α) (18). However, the underlying molecular mechanisms by which DHA exerts its anti-glioma effects remain unclear. In the present study, the effects of DHA treatment of glioma cells and the potential molecular mechanisms were investigated, particularly focusing on the impact on Raf/MEK/ERK and PI3K/AKT pathways.

Materials and methods

Cells and reagents

The BT325 cell line was obtained from Beijing Neurosurgical Institute Collection (Beijing, China) and the C6 cell line was purchased from American Type Culture Collection (Manassas, VA, USA). All cells were cultured at 37°C in Dulbecco's modified Eagle medium (DMEM) (Gibco Life Technologies, Carlsbad, CA, USA) containing 10% fetal bovine serum (FBS) (Gibco), and 100 U/ml penicillin-streptomycin (Gibco). Dihydroartemisinin (DHA) was purchased from Sigma-Aldrich (St. Louis, MO, USA) and dissolved in dimethylsulfoxide (DMSO) with cell medium resulting in a final DMSO concentration of 1%.

Cell viability assay

Cell viability was measured by 3-[4,5-dimethylthiazol-2-thiazolyl]-2,5-diphenyl-tetrazolium bromide (MTT) assay. Logarithmically growing glioma cells were seeded in 96-well culture clusters (Costar, Cambridge, MA, USA) at a density of 5,000–6,000 cells/well with culture medium and incubated for 24 h. On the following day, cells were treated with desired concentrations of DHA. Four hours before the desired time points, 10 µl of 10 mg/ml MTT was added, and incubated for 4 h, the medium was removed and 200 µl DMSO was added to each well. The optical density (OD) value at 570 nm was measured using MRX II absorbance reader (DYNEX Technologies, Chantilly, VA, USA). The cell viability was expressed as the percentage of absorbance in cells with DHA treatment versus the control group. The experiment was replicated in triplicate. Results were expressed as the mean ± standard deviation (SD) values.

Apoptosis analysis by flow cytometry

Apoptotic cells were measured with Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) kit (BD Biosciences, Sparks, MD, USA). Briefly, cells were cultured in 6-well plates at 3×105 cells per well. After DHA-treatment for 10 h, a cell suspension was prepared by trypsinization, and was centrifuged at 800 × g for 5 min at 4°C, and then the cells were resuspended in 500 µl of binding buffer. The cells were incubated with 10 µl Annexin V-FITC solution and 5 µl PI at 37°C for 30 min away from the light. FACs analysis was performed by flow cytometry (BD Biosciences, Franklin Lakes, NJ, USA). For each sample, 10,000 cells were analyzed. All of the experiments were performed in triplicate.

Western blot analysis

Cells were plated in tissue culture dishes overnight and treated with DHA at the given concentrations for 24 h. Following harvest, adherent cells were washed with cold PBS and resuspended in lysis buffer (150 mM NaCl, 50 mM Tris-HCl, pH 7.4, 2 mM ethylenediaminetetra-acetic acid (EDTA), 1% NP-40) containing protease inhibitor cocktail (Amresco Inc., Framingham, MA, USA). Protein levels in the extracts were quantified using BCA assay (Pierce Biotechnology, Inc., Rockford, IL, USA). Equal amounts of total protein extracts were resolved by 10% standard sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto a polyvinylidene fluoride (PVDF) membrane (0.45 mm; EMD Millipore, Billerica, MA, USA). Membranes were blocked with 5% fat-free dry milk/Tris-buffered saline (TBS)-Tween 20 (TBST; cat. no. 9997, Cell Signaling Technology Inc., Danvers, MA, USA) at room temperature for 1 h, then incubated with the following anti-human primary antibodies to MEK (monoclonal rabbit; 1:1,000; cat. no. 8727), ERK (monoclonal rabbit; 1:1,000; cat. no. 4348), AKT (monoclonal mouse; 1:1,000; cat. no. 12694), phospho-MEK (monoclonal rabbit; 1:1,000; cat. no. 2338), phospho-ERK (monoclonal rabbit; 1:1,000; cat. no. 4377), phospho-AKT (polyclonal rabbit; 1:1,000; cat. no. 9272), Bcl-2 (monoclonal mouse; 1:500; cat. no. 15071), Mcl-1 (monoclonal rabbit; 1:500; cat. no. 5453), Bcl-xL (monoclonal rabbit; 1:500; cat. no. 13835), Bax (monoclonal rabbit; 1:500; cat. no. 5023) and GAPDH (monoclonal rabbit; 1:2,000; cat. no. 2118; Cell Signaling Technology Inc.) overnight at 4°C. The membranes were then washed three times with TBST for 5 min at room temperature. Secondary horseradish peroxidase-linked mouse anti-sheep (1:1,000; cat. no. 7076) or rabbit anti-sheep IgG (1:1,000; cat. no. 5127) antibodies (Cell Signaling Technology Inc.) were incubated for 1 h at room temperature. The membranes were then washed three times with TBST for 5 min at room temperature, and the immunoblots were visualized by enhanced chemiluminescence reagent (GE Healthcare Life Sciences, Logan, UT, USA).

Statistical analysis

Data are expressed as the mean ± SD, and evaluated for significant differences using one-way analysis of variance (ANOVA) via SPSS software, version 13.0 (SPSS Inc., Chicago, IL, USA). A value of P<0.05 was considered to indicate a statistically significant difference.

Results

DHA inhibited glioma cell proliferation

The effects of DHA on inhibition of proliferation of BT325 and C6 cells at dosages between 25–200 µM, were examined. MTT assay demonstrated that the effects of DHA on the viability of the cell lines were dose- and time-dependent (Fig. 1). Treatment of BT325 and C6 cells with 100 µM DHA for 48 h caused 62.5±3.6% and 77.3±6.4% reduction in cell viability, respectively (Fig. 1B; P<0.05).

DHA results in apoptotic death in glioma cells

The proapoptotic effects of DHA were examined in BT325 and C6 cells. As presented in Fig. 2, DHA promoted apoptosis in both cell lines in a dose-dependent manner (P<0.05; Fig 2C and D).

DHA inhibits Raf/MEK/ERK signaling pathway in glioma cells

A previous study reported that DHA inhibits Raf/MEK/ERK pathway in a number of types of cancer (14). Therefore the impact on the phosphorylation of these signaling molecules was examined in glioma cells. Western blot analysis demonstrated that after 24 h of DHA exposure, the expression levels of Raf/MEK/ERK signaling pathway members were reduced in both cell lines in a dose-dependent manner (Fig. 3).

DHA inhibits AKT activation in glioma cells

Dysregulation of the PI3K/AKT pathway is commonly observed in glioma cells (19). The effects of DHA on the activation of PI3K/AKT signaling were examined in glioma cells. Western blot analysis demonstrated that after 24 h of DHA exposure, DHA treatment markedly inhibited the phophorylation of AKT in a dose-dependent manner (Fig. 4).

Involvement of Bcl-2 family proteins in DHA-induced glioma cell apoptosis

The Bcl-2 family proteins serve important roles in the regulation of cell apoptosis (20). The effects of DHA treatment on Bcl-2 protein expression levels were examined. Western blot analysis demonstrated that DHA treatment markedly inhibited the expression of Bcl-2 and Mcl-1, two antiapoptotic Bcl-2 proteins (Fig. 5). However, DHA treatment had no impact on the protein expression levels of Bcl-xL or Bax.

Discussion

As important anti-apoptotic signaling pathways, Raf/MEK/ERK and PI3K/AKT signaling cascades have attracted considerable attention (3). These signaling pathways have been demonstrated to serve critical roles in the transduction of growth factor signals, which regulate gene expression to control cell survival, proliferation, motility, and metabolism. Mutations may occur in upstream receptors such as EGFR and Flt-3, or downstream members such as Ras, Raf, PI3K, PTEN and AKT (13). Accumulating evidence has indicated that the crosstalk between Ras/Raf/MEK/ERK and the PI3K/AKT pathways serves important roles in promoting cell proliferation and inhibiting apoptosis of cancer, and pathway cross-inhibition reduces the effectiveness of single agents (12). A number of studies have demonstrated synergistic anticancer effects of simultaneous inhibition of both pathways. Treatment of basal-like cell lines with a selective MEK inhibitor resulted in enhanced PI3K pathway signaling, whereas supplementation of PI3K inhibitor induces apoptosis both in vitro and in vivo (18,21). These evidence indicates that dual inhibition of Raf/MEK/ERK and PI3K/AKT signaling may result in more clinical benefit and therefore may provide certain advantages over single pathway inhibitors (22).

Although the anti-cancer activity of DHA has been demonstrated in a number of types of cancer (2325), the underlying mechanisms remain largely unknown. To optimize the therapeutic effect of DHA against glioma cells and to explore its molecular mechanisms, the BT325 and C6 cell lines were used in the present study. Indeed, using cell viability and apoptosis analysis, it was indicated that DHA is capable of inducing the apoptosis of human glioma cells in a concentration- and time-dependent manner. For further illuminating the possible mechanisms, the present study was extended to investigate the effect of DHA on MEK/ERK and PI3K/AKT signaling factors. The results demonstrated that DHA suppressed MEK, ERK and AKT phosphorylation in the dose-dependent manner, indicating that DHA may induce apoptosis through a process that involves Raf/MEK/ERK and PI3K/AKT pathways inactivation. The Raf/MEK/ERK and PI3K/AKT signal pathways have been implicated in a wide variety of processes in cancer cells including the regulation of cell proliferation, survival and apoptosis. Therefore, the present study hypothesized that the combination of Raf/MEK/ERK and PI3K/AKT pathway inhibition and DHA treatment would greatly suppress the proliferation of glioma cells and markedly induce apoptosis.

Inducing apoptosis is an attractive strategy for cancer therapy, which is finally determined by the balance between pro- and anti-apoptotic mechanisms (20). The expression of the Bcl-2 protein family was also examined; this family of proteins serves important roles in the regulation of mitochondria-dependent apoptosis. Bcl-2 protein is the prototype of this family, which inhibits cell apoptosis through multiple mechanisms (26). Mcl-1 is another highly expressed anti-apoptotic protein expressed in a number of types of malignancies and has been demonstrated to mediate resistance to chemotherapy, whereas Bax is a pro-apoptotic member. In the present study, no significant differences in the expression of Bax protein were observed after DHA treatment in glioma cells. However, the expression of Bcl-2 and Mcl-1 proteins was significantly suppressed by DHA in a dose-dependent manner. These results indicated that DHA may exerts its anti-glioma effects through the inhibition of pro-apoptotic proteins Bcl-2 and Mcl-1.

In summary, the data in the present study indicates that DHA suppresses the Raf/MEK/ERK and PI3K/AKT pathways in the glioma cells, which provides valuable information on the molecular mechanism of its anticancer activity. Therefore, DHA may be a promising molecule for the treatment of glioma.

Glossary

Abbreviations

Abbreviations:

DHA

dihydroartemisinin

MTT

3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide

MEK

mitogen-activated and extracellular signal-regulated kinase kinase)

ERK

extracellular signal-regulated kinase

MAPK

mitogen-activated protein kinase

PI3K

phosphatidylinositol 3-kinase

PIP3

phosphatidylinositol 3,4,5 triphosphate

References

1 

Sathornsumetee S, Reardon DA, Desjardins A, et al: Molecularly targeted therapy for malignant glioma. Cancer. 110:13–24. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Wang Y and Jiang T: Understanding high grade glioma: Molecular mechanism, therapy and comprehensive management. Cancer Lett. 331:139–146. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Saini KS, Loi S, de Azambuja E, et al: Targeting the PI3K/AKT/mTOR and Raf/MEK/ERK pathways in the treatment of breast cancer. Cancer Treat Rev. 39:935–946. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Redman EK, Brookes PS and Karcz MK: Role of p90(RSK) in regulating the crabtree effect: implications for cancer. Biochem Soc Trans. 41:124–126. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Hasskarl J: Sorafenib: Targeting multiple tyrosine kinases in cancer. Recent Results Cancer Res. 201:145–164. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Han L, Yang Y, Yue X, et al: Inactivation of PI3K/AKT signaling inhibits glioma cell growth through modulation of beta-catenin-mediated transcription. Brain Res. 1366:9–17. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Chin YR and Toker A: Function of Akt/PKB signaling to cell motility, invasion and the tumor stroma in cancer. Cell Signal. 21:470–476. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Ghayad SE and Cohen PA: Inhibitors of the PI3K/Akt/mTOR pathway: New hope for breast cancer patients. Recent Pat Anticancer Drug Discov. 5:29–57. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Li Q, Wu J, Zheng H, et al: Discovery of 3-(2-aminoethyl)-5-(3-phenyl-propylidene)-thiazolidine-2,4-dione as a dual inhibitor of the Raf/MEK/ERK and the PI3K/Akt signaling pathways. Bioorg Med Chem Lett. 20:4526–4530. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Steelman LS, Abrams SL, Whelan J, et al: Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to leukemia. Leukemia. 22:686–707. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Mendoza MC, Er EE and Blenis J: The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends Biochem Sci. 36:320–328. 2011. View Article : Google Scholar : PubMed/NCBI

13 

McCubrey JA, Steelman LS, Kempf CR, et al: Therapeutic resistance resulting from mutations in Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR signaling pathways. J Cell Physiol. 226:2762–2781. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Lee J, Zhang G, Wu X, Xu F, Zhou J and Zhang X: Growth inhibitory effect of dihydroartemisinin on Bcr/Abl+ chronic myeloid leukemia K562 cells involve, AKT, ERK and NF-kappaB modulation. J Cancer Res Clin Oncol. 138:2095–2102. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Wang XM, Zhang L, Ding GF and Wang QZ: Inhibitory effect of dihydroartemisinin on the growth of human prostate cancer PC-3 M cells and its mechanism. Zhonghua Nan Ke Xue. 18:590–594. 2012.(In Chinese). PubMed/NCBI

16 

Feng X, Li L, Jiang H, Jiang K, Jin Y and Zheng J: Dihydroartemisinin potentiates the anticancer effect of cisplatin via mTOR inhibition in cisplatin-resistant ovarian cancer cells: involvement of apoptosis and autophagy. Biochem Biophys Res Commun. 444:376–381. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Lu M, Sun L, Zhou J and Yang J: Dihydroartemisinin induces apoptosis in colorectal cancer cells through the mitochondria-dependent pathway. Tumour Bio. 35:5307–5314. 2014. View Article : Google Scholar

18 

Hoeflich KP, O'Brien C, Boyd Z, et al: In vivo antitumor activity of MEK and phosphatidylinositol 3-kinase inhibitors in basal-like breast cancer models. Clin Cancer Res. 15:4649–4664. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Kubiatowski T, Jang T, Lachyankar MB, Salmonsen R, Nabi RR, Quesenberry PJ, Litofsky NS, Ross AH and Recht LD: Association of increased phosphatidylinositol 3-kinase signaling with increased invasiveness and gelatinase activity in malignant gliomas. J Neurosurg. 95:480–488. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Yip KW and Reed JC: Bcl-2 family proteins and cancer. Oncogene. 27:6398–6406. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Mirzoeva OK, Das D, Heiser LM, et al: Basal subtype and MAPK/ERK kinase (MEK)-phosphoinositide 3-kinase feedback signaling determine susceptibility of breast cancer cells to MEK inhibition. Cancer Res. 69:565–572. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Sheppard KE, Cullinane C, Hannan KM, et al: Synergistic inhibition of ovarian cancer cell growth by combining selective PI3K/mTOR and RAS/ERK pathway inhibitors. Eur J Cancer. 49:3936–3944. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Huang XJ, Li CT, Zhang WP, Lu YB, Fang SH and Wei EQ: Dihydroartemisinin potentiates the cytotoxic effect of temozolomide in rat C6 glioma cells. Pharmacology. 82:1–9. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Lu YY, Chen TS, Qu JL, Pan WL, Sun L and Wei XB: Dihydroartemisinin (DHA) induces caspase-3-dependent apoptosis in human lung adenocarcinoma ASTC-a-1 cells. J Biomed Sci. 16:162009. View Article : Google Scholar : PubMed/NCBI

25 

Liu Y, Wang W, Xu J, et al: Dihydroartemisinin inhibits tumor growth of human osteosarcoma cells by suppressing Wnt/beta-catenin signaling. Oncol Rep. 30:1723–1730. 2013.PubMed/NCBI

26 

Heath-Engel HM, Chang NC and Shore GC: The endoplasmic reticulum in apoptosis and autophagy: Role of the BCL-2 protein family. Oncogene. 27:6419–6433. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 10 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Du W, Pang C, Xue Y, Zhang Q and Wei X: Dihydroartemisinin inhibits the Raf/ERK/MEK and PI3K/AKT pathways in glioma cells Corrigendum in /10.3892/ol.2021.12636. Oncol Lett 10: 3266-3270, 2015
APA
Du, W., Pang, C., Xue, Y., Zhang, Q., & Wei, X. (2015). Dihydroartemisinin inhibits the Raf/ERK/MEK and PI3K/AKT pathways in glioma cells Corrigendum in /10.3892/ol.2021.12636. Oncology Letters, 10, 3266-3270. https://doi.org/10.3892/ol.2015.3699
MLA
Du, W., Pang, C., Xue, Y., Zhang, Q., Wei, X."Dihydroartemisinin inhibits the Raf/ERK/MEK and PI3K/AKT pathways in glioma cells Corrigendum in /10.3892/ol.2021.12636". Oncology Letters 10.5 (2015): 3266-3270.
Chicago
Du, W., Pang, C., Xue, Y., Zhang, Q., Wei, X."Dihydroartemisinin inhibits the Raf/ERK/MEK and PI3K/AKT pathways in glioma cells Corrigendum in /10.3892/ol.2021.12636". Oncology Letters 10, no. 5 (2015): 3266-3270. https://doi.org/10.3892/ol.2015.3699