Cancer Letters

Cancer Letters

Volume 435, 28 October 2018, Pages 66-79
Cancer Letters

Original Articles
β-catenin contributes to cordycepin-induced MGMT inhibition and reduction of temozolomide resistance in glioma cells by increasing intracellular reactive oxygen species

https://doi.org/10.1016/j.canlet.2018.07.040Get rights and content

Highlights

  • Cordycepin inhibited the viability of glioma cells in vitro and in vivo.

  • Cordycepin reduced the expression of MGMT and augmented TMZ-mediated chemotherapy in vitro and in vivo.

  • Cordycepin-induced reduction of TMZ resistance in glioma cells was MGMT-dependent.

  • β-catenin regulated cordycepin-induced MGMT inhibition and reduction of TMZ resistance in glioma cells.

  • ROS is required for cordycepin-induced MGMT inhibition and reduction of TMZ resistance in glioma cells.

Abstract

Glioblastoma multiforme (GBM) is one of the most aggressive human tumors, and it has a poor prognosis. Temozolomide (TMZ) is the primary alkylating agent used to treat GBM. Nevertheless, a number of GBM patients are resistant to TMZ. Therefore, there is an urgent need for more effective therapeutic options. Cordycepin (COR) is a natural chemical with anti-tumor effects, although its mechanism of action is poorly understood. Several lines of evidence suggest that O6-methylguanine DNA methyltransferase (MGMT) repairs damaged DNA and contributes to drug resistance to TMZ in gliomas. The Wnt/β-catenin pathway regulates MGMT gene expression. However, whether cordycepin inhibits MGMT expression by downregulating the β catenin pathway and augmenting chemosensitivity to TMZ in glioma cells remains unclear. In the present study, we found that cordycepin inhibited the viability of glioma cells and induced apoptosis, cell cycle arrest, overproduction of reactive oxygen species (ROS) and reduction of glutathione (GSH) in vitro. Moreover, cordycepin significantly reduced tumor volume and prolonged median survival of tumor-bearing rats in vivo. We also found that cordycepin inhibited MGMT expression and augmented chemosensitivity to TMZ in glioma cells in vitro and in vivo, accompanied by downregulation of p-GSK-3β and β-catenin. Moreover, overexpression of MGMT reversed the synergistic effect of cordycepin and TMZ. Pharmacological inhibition of GSK-3β with CHIR-99021 or overexpression of β-catenin reversed cordycepin-induced reduction of cell viability, downregulation of β-catenin and MGMT, increase of apoptosis and reduction of TMZ resistance. Furthermore, we found that β-catenin regulated cordycepin-induced overproduction of ROS by decreasing GSH. Inhibition of ROS production with N-acetyl-l-cysteine (NAC) not only rescued the reduction of cell viability but also eliminated β-catenin and MGMT inhibition, prevented glioma cells apoptosis and reversed the synergistic effect of cordycepin and TMZ. Taken together, we demonstrated that β-catenin contributed to cordycepin-induced MGMT inhibition and reduction of TMZ resistance in glioma cells via increasing intracellular ROS. These results indicate that cordycepin may be a novel agent to improve GBM treatment, especially in TMZ-resistant GBM with high MGMT expression.

Introduction

Glioblastoma multiforme (GBM) is the most common and aggressive type of intrinsic brain tumor, with a very poor prognosis and high recurrence and morbidity [1]. The current standard care for newly diagnosed GBM generally consists of surgical resection followed by radiotherapy in conjunction with the alkylating agent temozolomide (TMZ) [2]. Nevertheless, malignant gliomas are resistant to many kinds of treatments, including radiation, chemotherapy and other adjuvant therapies. Therefore, an alternative therapeutic strategy is needed to effectively treat GBM patients.

Cordycepin (Fig. 1A), isolated from cultured Cordyceps militaris, possesses a variety of clinical effects such as regulation of immunity [3], inflammation [4], protein synthesis [5], platelet aggregation [6] and steroidogenesis [7]. Moreover, cordycepin has been demonstrated to induce apoptosis not only in glioma cells [8,9] but also in other types of cancer cells such as breast cancer cells [10], leukemia cells [11] and gallbladder cancer cells [12]. Whether cordycepin can enhance the sensitivity to TMZ remains unclear.

TMZ is the primary alkylating agent used to treat GBM, however, overall responses are poor, amounting to approximately 26.5% [2]. TMZ induces DNA methylation of guanine at the O6 position, leading to double-strand breakage in the genome, resulting in cell cycle arrest and induction of apoptosis. O6-metG is quickly repaired by the DNA repair protein O6-methylguanine DNA methyltransferase (MGMT) [13]. MGMT expression is considered to be a major determinant of TMZ resistance in GBM, and this is supported by large amounts of data from both clinical and basic studies [[14], [15], [16]]. Several lines of evidence indicate that the prognosis of patients with high MGMT expression is much poorer than of those with low expression [17]. Therefore, other therapeutic agents that suppress MGMT expression and increase sensitivity to TMZ are highly desired.

β-catenin, the primary downstream effector of the canonical Wnt pathway, participates in many cellular processes, including cell proliferation and differentiation, cell cycle, migration and invasion [18,19]. β-catenin stability is controlled by a protein complex composed of several proteins including: Axin, CK, APC and GSK-3β. Complex-associated β-catenin is phosphorylated by GSK-3β and is consequently degraded via the proteasome-dependent pathway [20]. Previous studies have shown that Wnt/β-catenin pathway regulates MGMT gene expression in cancer [21]. Cordycepin inhibited the Wnt/β-catenin pathway in leukemia, and this suppressive effect was mediated by GSK-3β regulation [22]. We speculated that cordycepin may inhibit MGMT expression by downregulating the β-catenin pathway and augmenting chemosensitivity to TMZ in glioma cells. Therefore, we used glioma cell lines and xenograft glioma models to examine whether cordycepin enhanced the efficacy of TMZ and to investigate the underlying mechanisms.

Section snippets

Reagents

Cordycepin, NAC (N-acetyl-l-cysteine) and temozolomide were all purchased from Sigma (St. Louis, MO, USA). CHIR-99021 was purchased from Selleckchem Company (Houston, TX, USA). Primary antibodies for specific detection of PARP-1, cleaved caspase-3, Bax, Bcl-2, c-Myc, cyclin D1, cyclin B1, p-GSK-3β (Ser9), GSK-3β and β-catenin were purchased from Cell Signaling Technology (Beverly, MA, USA). Anti-MGMT antibody was purchased from Abcam (Cambridge, MA, USA). Anti-β-actin antibody was purchased

Cordycepin inhibited the viability of glioma cells in vitro

To investigate the effect of cordycepin on glioma cell growth, LN18, T98G, LN229 and SHG-44 cells were treated with various concentrations of cordycepin for 24, 48 and 72 h. The MTT assay results (Fig. 1B) showed that cell viability decreased with increasing concentrations of cordycepin and with increased duration of treatment. The 50% growth-inhibitory concentrations (IC50) after treatment with cordycepin for 48 h were 254.4 μmol/L in LN18 cells, 301.2 μmol/L in T98G cells, 145.4 μmol/L in

Discussion

GBM is the most common intracranial tumor and it has an extremely poor prognosis. TMZ is widely recognized as the first-line drug for patients with GBM, however, the emergence of drug resistance in majority of GBM patients represents a major setback that requires urgent attention [28]. Because of the challenges associated with treating GBM, many efforts have been directed towards increasing sensitivity to TMZ to improve overall survival of GBM patients. Sulforaphane combined with TMZ

Conflicts of interest

The authors declare no conflict of interest.

Acknowledgments

This study was jointly supported by the Young Scientists Fund of the National Natural Science Foundation of China (Nos. 21401072) and the National Natural Science Foundation of China (Nos. 81772684).

References (39)

  • R. Stupp et al.

    Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma

    N. Engl. J. Med.

    (2005)
  • X. Zhou et al.

    Cordycepin is an immunoregulatory active ingredient of Cordyceps sinensis

    Am. J. Chin. Med.

    (2008)
  • H.Y. Pao et al.

    Cordycepin stimulated steroidogenesis in MA-10 mouse Leydig tumor cells through the protein kinase C Pathway

    J. Agric. Food Chem.

    (2012)
  • J.S. Baik et al.

    Apoptotic effects of cordycepin through the extrinsic pathway and p38 MAPK activation in human glioblastoma U87MG cells

    J. Microbiol. Biotechnol.

    (2016)
  • Y. Liao et al.

    Cordycepin induces cell cycle arrest and apoptosis by inducing DNA damage and up-regulation of p53 in Leukemia cells

    Cell Cycle

    (2015)
  • X.A. Wang et al.

    Cordycepin induces S phase arrest and apoptosis in human gallbladder cancer cells

    Molecules

    (2014)
  • Z.K. Qiu et al.

    Enhanced MGMT expression contributes to temozolomide resistance in glioma stem-like cells

    Chin. J. Canc.

    (2014)
  • M. Hermisson et al.

    O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells

    J. Neurochem.

    (2006)
  • M.D. Blough et al.

    Sensitivity to temozolomide in brain tumor initiating cells

    Neuro Oncol.

    (2010)
  • Cited by (35)

    • Mannose inhibits proliferation and promotes apoptosis to enhance sensitivity of glioma cells to temozolomide through Wnt/β-catenin signaling pathway

      2022, Neurochemistry International
      Citation Excerpt :

      We provided evidence that mannose inhibited MGMT expression via Wnt/β-catenin pathway to augment the sensitivity of glioma cells to TMZ. Similar study has been reported previously that β-catenin contributes to the inhibited MGMT and the reduced TMZ resistance induced by cordycepin in glioma cells (Bi et al., 2018). Besides, 20(S)-ginsenoside-Rg3 effectively downregulates MGMT level through Wnt/β-catenin signaling and reverses TMZ resistance in glioma cell lines (Chen et al., 2019).

    • Tumor-derived exosomes reversing TMZ resistance by synergistic drug delivery for glioma-targeting treatment

      2022, Colloids and Surfaces B: Biointerfaces
      Citation Excerpt :

      Therefore, it is vital to develop an effective strategy both for controlling tumor progression and prolonging survival rate. As the second-generation oral alkylating agent, TMZ exhibits excellent antitumor activity by methylating DNA molecule and interfering their replication, finally inhibits tumor cell proliferation [9,10]. Since approved by FDA, TMZ has become the first-line chemotherapeutic drugs to cure the glioma.

    • Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance.

      2021, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      In previous studies, several pathways have been identified that play essential roles in MGMT expression regulation. The Wnt/β-catenin signaling regulates MGMT expression, and cordycepin-mediated downregulation of Wnt/β catenin signaling subsequently suppresses MGMT expression, thereby augmenting chemosensitivity of glioma cells towards TMZ [169]. BMX, an HDAC8 inhibitor overcomes TMZ-resistance in GBM cells that have wild type p53 by enhancing cell death via downregulation of β catenin/c-Myc/SOX2 cellular signaling and MGMT inhibition [170].

    • Chemoattractants driven and microglia based biomimetic nanoparticle treating TMZ-resistant glioblastoma multiforme

      2021, Journal of Controlled Release
      Citation Excerpt :

      Glioblastoma multiforme (GBM) is a commonly primary intracranial malignant tumor with high recurrence rate, high mortality and poor prognosis while being the most aggressive tumor [1]. The standard treatment for GBM is surgical resection combined with radiotherapy and temozolomide (TMZ) chemotherapy [2]. At present, the 5-year survival rate of patients with GBM is barely 4.7% [3].

    View all citing articles on Scopus
    View full text