Cancer Letters

Cancer Letters

Volume 307, Issue 2, 28 August 2011, Pages 141-148
Cancer Letters

Autophagy inhibition promotes paclitaxel-induced apoptosis in cancer cells

https://doi.org/10.1016/j.canlet.2011.03.026Get rights and content

Abstract

Paclitaxel has been demonstrated to be an effective mitotic inhibitor and apoptosis inducer to treat aggressive malignancies. In this paper, we have provided a line of evidence that promotion of apoptotic cell death by paclitaxel was accompanied with induction of autophagy in A549 cells. Paclitaxel treatment could lead to the formation of acidic vesicular organelles (AVOs), the induction of Atg5, Beclin 1 and microtubule-associated protein 1 light chain 3 (LC3) expressions, and the increase of punctate fluorescent signals in A549 cells pre-transfected with green fluorescent protein (GFP)-tagged LC3. Interestingly, paclitaxel-mediated apoptotic cell death was further potentiated by pretreatment with autophagy inhibitor 3-methyladenine (3-MA) or small interfering RNA against the autophagic gene beclin 1. These findings suggest that paclitaxel-elicited autophagic response plays a protective role that impedes the eventual cell death, and inhibition of autophagy could be an adjunctive strategy for enhancing chemotherapeutic effect of paclitaxel as an antitumor agent.

Introduction

Chemotherapy is an important option in curing or controlling various cancers including lung cancer. Paclitaxel, which stabilizes microtubule and causes apoptosis, offers both symptomatic and survival benefits for lung adenocarcinoma. The paclitaxel-based combination therapies are standard treatments for nearly all patients diagnosed with non-small cell lung carcinoma (NSCLC) [1]. However, clinical treatment with paclitaxel often encounters a number of undesirable side effects as occurred using other anticancer agents. The dose increment of systemic administration of paclitaxel would generate unacceptable levels of toxicity to normal cells, especially of bone marrow, gastrointestinal tract, and the hair follicles [2]. Therefore, many attempts have been made to enhance its therapeutic effectiveness, simultaneously reducing its toxicity. In an effort to search for strategies that could enhance cancer cell killing mediated by paclitaxel, we have investigated possible pro-survival pathways that are activated in response to paclitaxel. Herein, we report the induction of autophagy by paclitaxel.

Autophagy is an evolutionary conserved process in which cell engulfs cytoplasmic constituents within a double-membrane vacuole (named autophagosome) and delivers them to the lysosome for degradation [3]. Autophagy contributes to maintaining cellular homeostasis as a result of quality control of both proteins and organelles. In addition to its basic role in the turnover of proteins and organelles, autophagy has multiple physiological and pathophysiological functions [4], [5]. When cells encounter environmental stressors such as nutrient starvation and pathogen infection, autophagy is induced to provide nutrients and energy required for cell survival. So autophagy is recognized as a cytoprotective process against environmental stress [6], [7]. Meanwhile, autophagy is also an alternative route of programmed cell death, called type-2 programmed cell death or autophagic cell death [8]. In tumor cells, the role of autophagy may depend on the type of tumor, the stage of tumorigenesis, and the nature and extent of the insult. Appropriate modification of autophagy, that is, inhibition of cytoprotective autophagy or promotion of cyto-killing autophagy could augment cytotoxicity caused by anticancer therapy in tumor cells [9], [10], [11]. Thus, in addition to apoptotic response, it would be very useful to determine if an antitumor agent can induce autophagy and what type of autophagies it is. In this study, we found that paclitaxel-induced autophagy in cancer cells, and inhibition of autophagy could lead to enhancement of paclitaxel-mediated cytotoxicity through increasing apoptosis.

Section snippets

Cell lines and cell culture

A549, PC-3, and HT-29 cell lines were obtained from The Cell Bank of Chinese Academy of Sciences (Shanghai). Human glioma cancer cell line U87 which stably expresses GFP-LC3 protein was kindly gifted by Yan bing (Shan Dong university). A549 cells were cultured with F12 medium supplemented with 10% foetal bovine serum and antibiotics (100Ā U/mL penicillin and 100Ā Ī¼g/mL streptomycin). PC-3, HT-29 and GFP-LC3 transfected U87 cells were maintained in DMEM medium supplemented with 10% foetal bovine

Paclitaxel induces apoptosis in A549 cells

In order to determine whether the observed cell death caused by paclitaxel was due to apoptosis, apoptosis parameters were analyzed by DAPI staining and Western-blot assay. As shown in Fig. 1A, nuclear morphological changes of apoptosis were observed in paclitaxel-treated cells. Paclitaxel caused significant increases in apoptotic cell number with condensed and fragmented DNA (as indicated by a strong blue fluorescence). In addition, alternations in the expression levels of Bax and Bcl-2

Discussion

Despite having diverse mechanisms of action, many frontline anticancer agents would be predicted to stimulate autophagy including arsenic trioxide, 5-FU, histone deacetylase (HDAC) inhibitors, tamoxifen, imatinib, and ionizing radiation [18], [19], [20], [21], [22], [23]. Paclitaxel is a widely used agent that is effective in the treatment of a variety of human cancers. Despite its capability to stabilize microtubules and impair mitosis, paclitaxel also exhibits induction of apoptosis, and

Conflicts of interest

None of the authors has any financial or other interest with regard to the submitted manuscript that might be constructed as a conflict of interest.

Acknowledgments

The authors thank Xuejun Jiang (Institute Microbiology Chinese Academy of Sciences) for providing the GFP-LC3 expressing vector.

This work was supported by the National Natural Science Foundation of China (30973551), and Shandong Scientific Technology Program (2008GG10002042).

References (34)

  • M. Katayama et al.

    DNA damaging agent-induced autophagy produces a cytoprotective adenosine triphosphate surge in malignant glioma cells

    Cell Death Differ.

    (2007)
  • M.C. Maiuri et al.

    Self-eating and self-killing: crosstalk between autophagy and apoptosis

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • T. Kanzawa et al.

    Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells

    Cell Death Differ.

    (2004)
  • H. Takeuchi et al.

    Synergistic augmentation of rapamycin-induced autophagy in malignant glioma cells by phosphatidylinositol 3-kinase/protein kinase B inhibitors

    Cancer Res.

    (2005)
  • T. Shingu et al.

    Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells

    Int. J. Cancer

    (2009)
  • Y. Kabeya et al.

    LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing

    EMBO J.

    (2000)
  • A. Bommareddy et al.

    Atg5 regulates phenethyl isothiocyanate-induced autophagic and apoptotic cell death in human prostate cancer cells

    Cancer Res.

    (2009)
  • Cited by (0)

    View full text