Original Contribution
Resistance of neuroblastoma GI-ME-N cell line to glutathione depletion involves Nrf2 and heme oxygenase-1

https://doi.org/10.1016/j.freeradbiomed.2011.11.007Get rights and content

Abstract

Cancer cell survival is known to be related to the ability to counteract oxidative stress, and glutathione (GSH) depletion has been proposed as a mechanism to sensitize cells to anticancer therapy. However, we observed that GI-ME-N cells, a neuroblastoma cell line without MYCN amplification, are able to survive even if GSH-depleted by l-buthionine-(S,R)-sulfoximine (BSO). Here, we show that in GI-ME-N cells, BSO activates Nrf2 and up-regulates heme oxygenase-1 (HO-1). Silencing of Nrf2 restrained HO-1 induction by BSO. Inhibition of HO-1 and silencing of Nrf2 or HO-1 sensitized GI-ME-N cells to BSO, leading to reactive oxygen/nitrogen species overproduction and decreasing viability. Moreover, targeting the Nrf2/HO-1 axis sensitized GI-ME-N cells to etoposide more than GSH depletion. Therefore, we have provided evidence that in GI-ME-N cells, the Nrf2/HO-1 axis plays a crucial role as a protective factor against cellular stress, and we suggest that the inhibition of Nfr2/HO-1 signaling should be considered as a central target in the clinical battle against neuroblastoma.

Graphical abstract

Highlights

► GI-ME-N neuroblastoma cell survival is independent from GSH depletion by BSO. ► BSO activates Nrf2 and up-regulates heme oxygenase-1 (HO-1) in GI-ME-N cells. ► Inhibition of HO-1 and silencing of Nrf2 or HO-1 sensitizes GI-ME-N cells to BSO. ► Inhibition of Nrf2/HO-1 axis sensitizes GI-ME-N cells to etoposide more than BSO. ► Inhibition Nfr2/HO-1 should be a potential target in neuroblastoma therapy.

Section snippets

Cell culture and treatments

GI-ME-N neuroblastoma cells were maintained in RPMI 1640 medium (Euroclone, Milan, Italy) supplemented with 10% fetal bovine serum (FBS; Euroclone), 2 mM glutamine (Sigma–Aldrich, Milan, Italy), 1% penicillin/streptomycin (Sigma–Aldrich), and 1% amphotericin B (Sigma–Aldrich), at 37 °C in a 5% CO2 humid atmosphere and subcultured every 5 days at 1:5.

Cell treatments included 1 mM BSO (Sigma–Aldrich), 2.5 μM zinc(II) protoporphyrin IX (ZnPP; Sigma–Aldrich), a combined treatment of BSO + ZnPP at the

BSO treatment induces severe GSH depletion in GI-ME-N cells

Cell exposure to 1 mM BSO was able to induce deep depletion of tGSH content. As shown in Fig. 1, after 9 and 12 h of BSO treatment, tGSH levels decreased by about 50 and 75%, respectively, in comparison to the untreated cells. After 24 h, tGSH decreased to very low levels (< 5% vs CTR) and these levels were stable after 48 and 72 h. After BSO treatment (24, 48, 72 h) MTT assay showed only a slight reduction in cell viability in comparison to the untreated samples (data not shown).

BSO treatment reduces cell proliferation rate

Cell counts

Discussion

In this study, we investigated the molecular mechanisms involved in GI-ME-N cell resistance to GSH depletion induced by BSO. Our findings provide evidence that (1) BSO activates Nrf2 and up-regulates HO-1 in GI-ME-N cells; (2) Nrf2 is crucial in HO-1 up-regulation by BSO; (3) HO-1 inhibition, as well as Nrf2 or HO-1 silencing, sensitizes GI-ME-N cells to BSO; and (4) BSO is able to sensitize GI-ME-N cells to etoposide but inhibition of the Nrf2/HO-1 axis is by far more effective in this

Acknowledgments

This work was supported by grants from the Italian Ministry of University (PRIN 2009M8FKBB_002 and 2008N9N9KL_002) and Genova University (Progetto Ricerca Ateneo 2010). We thank Mr. Giuseppe Catalano for technical assistance. We thank Ms. Suzanne Patten for reviewing the use of English in the manuscript.

References (71)

  • J. Alam et al.

    Transcriptional activation of the heme oxygenase gene by heme and cadmium in mouse hepatoma cells

    J. Biol. Chem.

    (1989)
  • R. Foresti et al.

    Thiol compounds interact with nitric oxide in regulating heme oxygenase-1 induction in endothelial cells: involvement of superoxide and peroxynitrite anions

    J. Biol. Chem.

    (1997)
  • R.R. Crichton et al.

    Molecular and cellular mechanisms of iron homeostasis and toxicity in mammalian cells

    J. Inorg. Biochem.

    (2002)
  • M.D. Maines et al.

    Expression of heme oxygenase-1 (HSP32) in human prostate: normal, hyperplastic, and tumor tissue distribution

    Urology

    (1996)
  • C.P. Anderson et al.

    Depletion of glutathione by buthionine sulfoxine is cytotoxic for human neuroblastoma cell lines via apoptosis

    Exp. Cell Res.

    (1999)
  • B. Marengo et al.

    Mechanisms of BSO (L-buthionine-S, R-sulfoximine)-induced cytotoxic effects in neuroblastoma

    Free Radic. Biol. Med.

    (2008)
  • M.W. Fariss et al.

    High-performance liquid chromatography of thiols and disulfides: dinitrophenol derivatives

    Methods Enzymol.

    (1987)
  • H. Sies

    Glutathione and its role in cellular functions

    Free Radic. Biol. Med.

    (1999)
  • S. Urbani et al.

    Use of CFDA-SE for evaluating the in vitro proliferation pattern of human mesenchymal stem cells

    Cytotherapy

    (2006)
  • A. Negre-Salvayre et al.

    Detection of intracellular reactive oxygen species in cultured cells using fluorescent probes

    Methods Enzymol.

    (2002)
  • G. Wang et al.

    Lipid peroxidation-derived aldehyde–protein adducts contribute to trichloroethene-mediated autoimmunity via activation of CD4+ T cells

    Free Radic. Biol. Med.

    (2008)
  • J.D. Hayes et al.

    NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer

    Trends Biochem. Sci.

    (2009)
  • G.S. Shim et al.

    Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the NRF2 pathway

    Free Radic. Biol. Med.

    (2009)
  • K. Ito et al.

    Effects of vitamin E deficiency and glutathione depletion on stress protein heme oxygenase 1 mRNA expression in rat liver and kidney

    Biochem. Pharmacol.

    (1997)
  • P. Gong et al.

    Heme oxygenase-1 protects HepG2 cells against cytochrome P450 2E1-dependent toxicity

    Free Radic. Biol. Med.

    (2004)
  • K. Srisook et al.

    Super-induction of HO-1 in macrophages stimulated with lipopolysaccharide by prior depletion of glutathione decreases iNOS expression and NO production

    Nitric Oxide

    (2005)
  • L. Chi et al.

    Depletion of reduced glutathione enhances motor neuron degeneration in vitro and in vivo

    Neuroscience

    (2007)
  • B. Su et al.

    Chronic oxidative stress causes increased tau phosphorylation in M17 neuroblastoma cells

    Neurosci. Lett.

    (2010)
  • C.P. Anderson et al.

    Buthionine sulphoximine alone and in combination with melphalan (L-PAM) is highly cytotoxic for human neuroblastoma cell lines

    Eur. J. Cancer

    (1997)
  • G.K. Balendiran et al.

    The role of glutathione in cancer

    Cell Biochem. Funct.

    (2004)
  • B. Zucker et al.

    Glutathione depletion in fibroblasts is the basis for apoptosis-induction by endogenous reactive oxygen species

    Cell Death Differ.

    (1997)
  • R.R. Ratan et al.

    Oxidative stress induces apoptosis in embryonic cortical neurons

    J. Neurochem.

    (1994)
  • J.S. Lewis-Wambi et al.

    Buthionine sulfoximine sensitizes antihormone-resistant human breast cancer cells to estrogen-induced apoptosis

    Breast Cancer Res.

    (2008)
  • J.I. Chuang et al.

    Glutathione depletion-induced apoptosis of Ha-ras-transformed NIH3T3 cells can be prevented by melatonin

    Oncogene

    (2003)
  • T. Schnelldorfer et al.

    Glutathione depletion causes cell growth inhibition and enhanced apoptosis in pancreatic cancer cells

    Cancer

    (2000)
  • Cited by (35)

    • Heme oxygenase-1-derived bilirubin protects endothelial cells against high glucose-induced damage

      2015, Free Radical Biology and Medicine
      Citation Excerpt :

      Heme oxygenase is the first and rate-limiting enzyme in the degradation of heme into biliverdin, carbon monoxide (CO), and free iron [20]. Heme oxygenase-1 (HO-1) is a 32 kDa stress protein present at low levels in most mammalian tissues [21], and its expression is induced by a wide variety of stress stimuli, including ROS, nitric oxide, and inflammatory cytokines [22–24]. HO-1 has been described as a key regulator of adaptive cellular responses to oxidative stress and plays a role in cellular homeostasis through the strong antioxidant activity of bilirubin, derived from biliverdin, ferritin, induced by free iron, and the antiapoptotic effect of carbon monoxide [25–27].

    • HO-1 up-regulation: A key point in high-risk neuroblastoma resistance to bortezomib

      2014, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      High levels of HO-1 have been observed in various human solid tumors (e.g. renal, prostatic and pancreatic) [14–16]. Moreover, HO-1 expression in tumor cells can be further increased by anticancer treatments (chemo-, radio-, and photodynamic therapy) [16] and it has been hypothesized that HO-1 and its products may have an important role in the development of a resistant phenotype [17,18]. Among the new therapeutic strategies designed to overcome cancer cell resistance, the use of proteasome inhibitors has been proposed [19,20].

    View all citing articles on Scopus
    View full text