Elsevier

Life Sciences

Volume 145, 15 January 2016, Pages 57-65
Life Sciences

Overexpression of NAD(P)H:quinone oxidoreductase 1 (NQO1) and genomic gain of the NQO1 locus modulates breast cancer cell sensitivity to quinones

https://doi.org/10.1016/j.lfs.2015.12.017Get rights and content

Abstract

Aims

Alterations in the expression of antioxidant enzymes are associated with changes in cancer cell sensitivity to chemotherapeutic drugs (menadione and β-lapachone). Mechanisms of acquisition of resistance to pro-oxidant drugs were investigated using a model of oxidative stress-resistant MCF-7 breast cancer cells (Resox cells).

Main methods

FISH experiments were performed in tumor biopsy and breast cancer cells to characterize the pattern of the NQO1 gene. SNP-arrays were conducted to detect chromosomal imbalances. Finally, the importance of NQO1 overexpression in the putative acquisition of either drug resistance or an increased sensitivity to quinones by cancer cells was investigated by immunoblotting and cytotoxicity assays.

Key findings

Genomic gain of the chromosomal band 16q22 was detected in Resox cells compared to parental breast cancer MCF-7 cells and normal human mammary epithelial 250MK cells. This genomic gain was associated with amplification of the NQO1 gene in one tumor biopsy as well as in breast cancer cell lines. Using different breast cell models, we found that NQO1 overexpression was a main determinant for a potential chemotherapy resistance or an increased sensitivity to quinone-bearing compounds.

Significance

Because NQO1 is frequently modified in tumors at genomic and transcriptomic levels, the impact of NQO1 modulation on breast cancer cell sensitivity places NQO1 as a potential link between cancer redox alterations and resistance to chemotherapy. Thus, the NQO1 gene copy number and NQO1 activity should be considered when quinone-bearing molecules are being utilized as potential drugs against breast tumors.

Section snippets

Chemical compounds

β-Lapachone (PubChem CID:3885)

Menadione (PubChem CID: 23665888)

Dicumarol (PubChem CID: 54676038)

Chemicals

β-Lapachone (3,4-Dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione), menadione (2-methyl-1,4-naphthoquinone) and dicumarol (3,3′-methylene-bis[4-hydroxycoumarin]) were purchased from Sigma (St Louis, MO, USA). All other chemicals were of ACS reagent grade.

Cell lines, cell culture

MCF-7 breast cancer cell line was purchased from ATCC (Manassas, USA). The Resox cell line was derived from MCF-7 cells [20], which were rendered resistant by chronic exposure to an H2O2-generating system made by mixing a redox cycler

NQO1 protein levels and NQO1 gene copy number in human breast tumors

Because of the general consensus that NQO1 is overexpressed in tumor cells, NQO1 protein levels were measured in 10 human mammary cancer biopsies. Indeed, high levels of NQO1 gene expression have been found in liver, lung, colon and breast tumors compared to normal tissues of the same origin [34]. The NQO1 protein level was increased in 6 out of 10 tumor samples compared to levels in tissue surrounding the tumor (Fig. 1A–B). NQO1 expression was enhanced about 2-fold in tumors compared to in

Discussion

NQO1, the enzyme that reduces quinone to hydroquinone [9], is often increased in tumors compared to healthy tissues [14], [15], [16]. In this study, the NQO1 protein levels were increased in 6 out of 10 invasive mammary ductal carcinoma samples compared to levels in tissue surrounding the tumor (Fig. 1A–B) and decreased in 3 samples. The increase or decrease of NQO1 protein levels in cancer cells could be dependent on different potentially cooperative mechanisms: corresponding genomic

Conclusions

NQO1 contributes to either drug cytotoxicity or drug elimination making the cellular response to quinone-bearing compounds a complex process. The data suggest that, in addition to the expression of NQO1 per se, NQO1 activity has a major influence on the sensitivity of cancer cells to quinone-based molecules as depicted in Fig. 5. Thus, it is tempting to suggest that determining NQO1 activity and the number of copies of the chromosomal band 16q22, more specifically of NQO1 gene copies, may be

Conflict of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgments

This project was funded by FNRS-Télévie Grant (Grant no. 7.4575.12F). The authors are grateful to Pr. David Ross and Dr. Jadwiga Kepa for providing the plasmid construct containing human NQO1 cDNA and Dr. Julie Stockis for plasmid sequencing. The authors are grateful to Sandrine Nonckreman, Khadija Bahloula, Elisabeth Wyns and their colleagues at the human genetic center of Saint-Luc hospital in Brussels for their help in establishing karyotypes, SNP arrays and FISH analyses. We thank also Pr.

References (55)

  • J.J. Lee et al.

    Cytogenetic methods for the mouse: preparation of chromosomes, karyotyping, and in situ hybridization

    Anal. Biochem.

    (1990)
  • C. Glorieux et al.

    Catalase overexpression in mammary cancer cells leads to a less aggressive phenotype and an altered response to chemotherapy

    Biochem. Pharmacol.

    (2011)
  • T. Mosmann

    Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays

    J. Immunol. Methods

    (1983)
  • T.E. Downing et al.

    Prognostic and predictive value of 16p12.1 and 16q22.1 copy number changes in human breast cancer

    Cancer Genet. Cytogenet.

    (2010)
  • S.A. Shaposhnikov et al.

    A map of nuclear matrix attachment regions within the breast cancer loss-of-heterozygosity region on human chromosome 16q22.1

    Genomics

    (2007)
  • E. Blanco et al.

    Beta-lapachone-containing PEG-PLA polymer micelles as novel nanotherapeutics against NQO1-overexpressing tumor cells

    J. Control. Release

    (2007)
  • S. Knuutila et al.

    DNA copy number losses in human neoplasms

    Am. J. Pathol.

    (1999)
  • D. Di Monte et al.

    Alterations in intracellular thiol homeostasis during the metabolism of menadione by isolated rat hepatocytes

    Arch. Biochem. Biophys.

    (1984)
  • T.W. Gant et al.

    Redox cycling and sulphydryl arylation; their relative importance in the mechanism of quinone cytotoxicity to isolated hepatocytes

    Chem. Biol. Interact.

    (1988)
  • A.C. Collier et al.

    The mitochondrial uncoupler dicumarol disrupts the MTT assay

    Biochem. Pharmacol.

    (2003)
  • K. Maruyama et al.

    Syntheses of alpha- and beta-lapachones and their homologues by way of photochemical side chain introduction to quinone

    Chem. Lett.

    (1977)
  • R.H. Thompson

    Naturally occurring quinones III, recent advances

    (1987)
  • P.L. Chesis et al.

    Mutagenicity of quinones: pathways of metabolic activation and detoxification

    Proc. Natl. Acad. Sci. U. S. A.

    (1984)
  • H.G. Williams-Ashman et al.

    Oxydation of reduced pyridine nucleotides in mammary gland and adipose tissue following treatment with polynuclear hydrocarbons

    Med. Exp. Int. J. Exp. Med.

    (1961)
  • J. Verrax et al.

    Enhancement of quinone redox cycling by ascorbate induces a caspase-3 independent cell death in human leukaemia cells. An in vitro comparative study

    Free Radic. Res.

    (2005)
  • A. Strassburg et al.

    Differential gene expression of NAD(P)H:quinone oxidoreductase and NRH:quinone oxidoreductase in human hepatocellular and biliary tissue

    Mol. Pharmacol.

    (2002)
  • J.J. Schlager et al.

    Cytosolic NAD(P)H:(quinone-acceptor)oxidoreductase in human normal and tumor tissue: effects of cigarette smoking and alcohol

    Int. J. Cancer

    (1990)
  • Cited by (27)

    • Profiling the chemical nature of anti-oxytotic/ferroptotic compounds with phenotypic screening

      2021, Free Radical Biology and Medicine
      Citation Excerpt :

      However, there are also reports showing that NQO1 is necessary to activate quinone toxicity. This property has been investigated as a potential anti-cancer approach using quinones, including β-lapachone and tanshinone, given that NQO1 is frequently overexpressed in a variety of tumors [36,82–85]. Based on these observations and studies suggesting that the antioxidant role of the QH2 form of the endogenous ubiquinone is specifically involved in preventing lipid peroxidation versus other types of ROS, it is possible that the QH2 forms generated by NQO1 and FPS1 are responsible for the anti-oxytotic/ferroptotic activity of the quinones analyzed here via prevention of lipid peroxidation while still leading to an increase in ROS [41,86–88].

    • Activatable imaging probes for cancer-linked NAD(P)H:quinone oxidoreductase-1 (NQO1): Advances and future prospects

      2020, TrAC - Trends in Analytical Chemistry
      Citation Excerpt :

      Studies have also found that the consumption of tumor-NQO1 significantly reduces the proportion of cancer stem cells with high aldehyde dehydrogenase (ALDH) activity, which is an indicator of lung cancer progression, metastasis and chemoresistance [18]. Overexpression of NQO1 is also considered to be one of the main factors of potential chemoresistance [19]. For instance, the cooperation of overexpressed NQO1 and γ-glutamylcysteine synthetase is involved in the resistance of hypoxic cells to cisplatin and mitomycin C [20].

    • NADPH: Quinone oxidoreductase 1 (NQO1) mediated anti-cancer effects of plumbagin in endocrine resistant MCF7 breast cancer cells

      2020, Phytomedicine
      Citation Excerpt :

      Quinones are widely found from natural products and serve as scaffold for cancer drugs. Quinone compounds are reduced by NQO1 enzyme that uses nicotinamide adenine dinucleotide phosphate (NADH) and its reduced form, NADPH as its cofactor (Glorieux et al., 2016). The NQO1 enzyme reduces quinone compounds to either a stable form or unstable form.

    • Phosphorylation of nuclear factor erythroid 2-like 2 (NFE2L2) in mammary tissue of Holstein cows during the periparturient period is associated with mRNA abundance of antioxidant gene networks

      2018, Journal of Dairy Science
      Citation Excerpt :

      By using NADPH as the hydrogen donor, NQO1 catalyzes the reduction of a broad array of quinones to their corresponding hydroquinones, hence this protein prevents the 1-electron reduction of quinones that results in ROS production (Dinkova-Kostova et al., 2004; Dinkova-Kostova and Talalay, 2010). In addition to its catalytic role as an antioxidant enzyme in the reduction of quinines (Glorieux et al., 2016), NQO1 has been reported to protect against estrogen quinone-mediated DNA and protein damage in human mammary epithelial cells (Yang et al., 2013). Similar to the observed postpartal upregulation of NQO1 in mammary tissue, it was reported that NQO1 was upregulated in the liver of dairy cows during the transition period (Gessner et al., 2013).

    • Prodrug strategy for cancer cell-specific targeting: A recent overview

      2017, European Journal of Medicinal Chemistry
      Citation Excerpt :

      In addition, oxidoreductases are important therapeutic targets due to their primary role in oxidative stress. NAD(P) H:quinone oxidoreductase 1 (NQO1), formerly known as DT-diaphorase, is an oxidoreductase with high levels of expression in a variety of cancers, including colon, breast, pancreas and lung cancers [68]. Anticancer prodrug molecules possessing quinone pharmacophore will undergo bioreductive activation by NQO1 to generate hydroquinones, which will exert activity through a number of pathways [69].

    • Chromatin remodeling regulates catalase expression during cancer cells adaptation to chronic oxidative stress

      2016, Free Radical Biology and Medicine
      Citation Excerpt :

      Protein probabilities were assigned by the Protein Prophet algorithm [43]. Cell metabolic status was assessed by following the reduction of MTT (3-(4,5-dimethylthia-zolyl-2)-2,5-diphenyltetrazolium bromide) to blue formazan as previously described [44,45]. Briefly, 1×104 cells/well were plated onto 96-well plates and, after confluence, they were preincubated 48 h with inhibitors, then exposed to the respective treatments for 24 h. Afterwards, cells were washed twice with PBS and incubated for 2 h with MTT (0.5 mg/ml).

    View all citing articles on Scopus
    1

    Current address: Université Bordeaux-Segalen, Inserm U1053, Bordeaux, France.

    View full text