Elsevier

Biochemical Pharmacology

Volume 77, Issue 9, 1 May 2009, Pages 1480-1486
Biochemical Pharmacology

PPARα signaling mediates the synergistic cytotoxicity of clioquinol and docosahexaenoic acid in human cancer cells

https://doi.org/10.1016/j.bcp.2009.02.002Get rights and content

Abstract

This study investigated the involvement of PPARγ and PPARα signaling in the synergistic anticancer activity of clioquinol (5-chloro-7-iodo-8-hydroxyquinoline) and docosahexaenoic acid (DHA) in human cancer cells. The synergistic cytotoxicity of DHA and clioquinol was demonstrated in nine human cancer cell lines representing different tissues of origin. A2780, a well-established ovarian cancer model system, was chosen for further characterization because of its sensitivity to DHA and clioquinol. Both PPARα and PPARγ were expressed in A2780 cells when analyzed with western blotting and reporter gene technique. Treatment of the cells with clofibrate (a PPARα agonist) and clioquinol for three days mimicked the synergy of DHA and clioquinol, whereas this synergy could not be seen with the use of troglitazone (a PPARγ agonist) and clioquinol, suggesting that PPARα signaling is involved in the synergistic action. When used alone, the IC50 of clofibrate was 513 μM in A2780 cells. However, the addition of 5 μM clioquinol to clofibrate-treated cells led to a dramatic reduction of its IC50 value (148 μM). The combination effects index (CI) analysis confirmed the synergy of clioquinol and clofibrate on inhibiting cancer cell viability. Using inhibitors to block PPARα signaling diminished the synergistic cytotoxicity of clioquinol and DHA. These results provide pharmacological evidence that the synergistic anticancer action of clioquinol and DHA is mediated by PPARα signaling in human cancer cells.

Introduction

The biological differences between normal and cancer cells are too subtle to easily achieve selective cancer cell targeting [1]. Recently, drug combinations have been utilized in an attempt to more effectively kill cancer cells. Combinations of drugs with different modes of action may enhance individual drugs’ anticancer action yet protect the host from side effects. Such synergistic interactions of anticancer drugs have been reported in studies of cultured tumor cell lines, animal models, and cancer patients [2], [3], [4], [5], [6], [7], [8].

The mechanisms by which a combination of drugs may selectively target tumor cells have not been well understood, although different modes of action are proposed to be responsible for the observed synergy [1]. Several cellular signaling molecules or pathways are proposed to be involved in the drug synergy on cancer cells, including Akt, NF-κB, apoptosis-related proteins, and the COX-2 pathway [9]. It is apparent that each drug combination may have distinct mechanisms of action because they may target different molecules or signaling pathways [1]. Since anticancer drugs are often toxic to normal cells, drug combinations, in some cases, may also be synergistically toxic to the host, parallel to their enhanced anticancer effects [1]. Therefore, more insight into the cellular mechanisms responsible for the synergistic action is critical to the development of an effective drug combination against cancer.

DHA is a long-chain n-3 polyunsaturated fatty acid, and it has been recognized to have anticancer activity in cultured tumor cells and various animal models [10], [11], [12], [13], [14], [15]. Its synergy with chemotherapeutics has also been reported [16], [17], [18], [19]. Since DHA is an essential fatty acid and protects neuronal cells from apoptosis [20], [21], the combination of DHA and chemotherapeutics may achieve enhanced toxic effects selectively towards cancer cells. Indeed, DHA has been shown to enhance the cytotoxicity of paclitaxel in experimental models and in human clinical trials without seeing enhanced side effects in patients [3], [4]. These observations justify the use of DHA as combination therapy with chemotherapeutics. We have recently demonstrated that DHA acts synergistically with clioquinol, a metal binding compound, to kill tumor cells [22]. Clioquinol has been recently tested for the treatment of Alzheimer's disease in clinical trials [23], [24] and is a newly discovered anticancer agent [25], [26], [27], [28]. Understanding the mechanisms of the synergistic action is essential for further development of DHA and clioquinol as a novel drug combination for chemotherapy. Because DHA is a well-established peroxisome proliferator activated receptor (PPAR) ligand [29], the present study examined the involvement of PPAR signaling in the synergistic anticancer action. We report here that PPARα, but not PPARγ, mediates the synergistic anticancer action of DHA and clioquinol in human cancer cells.

Section snippets

Materials

3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) was purchased from Promega (Madison, WI). Antibodies for PPARα and PPARγ were obtained from Santa Cruz Biotechnology (Santa Cruz, CA). The glyceraldehyde 3-phosphate dehydrogenase (GAPDH) antibody was obtained from ProMab Biotechnologies, Inc. (Albany, CA). The PPRE-lu reporter construct was obtained from Dr. Bruce Spiegelman at the Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA [30]

Effects of DHA and clioquinol on cell viability in human cancer cells

We first determined the effects of clioquinol and DHA on the viability of nine different human cancer cell lines. Treatment of the cells with clioquinol and DHA for three days inhibited cell viability in a concentration-dependent manner. The IC50s of clioquinol and DHA used alone or in combination were calculated (Table 1). The combination of these two compounds significantly lowered the IC50 value in each of the cell lines examined but the magnitude of the change differed among cell lines,

Discussion

The present study demonstrates, through pharmacological characterization, that PPARα signaling mediates the synergistic anticancer action of clioquinol and DHA in human cancer cells. These observations provide novel information on the synergistic anticancer action of clioquinol and DHA and suggest that PPARα ligands could potentially be used to enhance anticancer activity of chemotherapeutics.

Both DHA and clioquinol have been recognized to have anticancer activity [25], [26], [27], [28], [47],

Acknowledgement

Financial support was obtained from the Oklahoma Center for the Advancement of Science and Technology, the American Cancer Society (Institutional Seed Grant), the China Scholarship Council, and the College of Medicine, University of Oklahoma Health Sciences Center.

References (51)

  • W.E. Hardman

    (n-3) fatty acids and cancer therapy

    J Nutr

    (2004)
  • G.D. Girnun et al.

    Synergy between PPARgamma ligands and platinum-based drugs in cancer

    Cancer Cell

    (2007)
  • R.C. Reddy et al.

    Chemotherapeutic drugs induce PPAR-gamma expression and show sequence-specific synergy with PPAR-gamma ligands in inhibition of non-small cell lung cancer

    Neoplasia

    (2008)
  • K.M. Leu et al.

    Laboratory and clinical evidence of synergistic cytotoxicity of sequential treatment with gemcitabine followed by docetaxel in the treatment of sarcoma

    J Clin Oncol

    (2004)
  • M.O. Bradley et al.

    Tumor targeting by covalent conjugation of a natural fatty acid to paclitaxel

    Clin Cancer Res

    (2001)
  • A.C. Wolff et al.

    Phase I study of docosahexaenoic acid-paclitaxel: a taxane-fatty acid conjugate with a unique pharmacology and toxicity profile

    Clin Cancer Res

    (2003)
  • S. Banerjee et al.

    Molecular evidence for increased antitumor activity of gemcitabine by genistein in vitro and in vivo using an orthotopic model of pancreatic cancer

    Cancer Res

    (2005)
  • Y. Li et al.

    Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells

    Cancer Res

    (2005)
  • R.G. Azrak et al.

    Therapeutic synergy between irinotecan and 5-fluorouracil against human tumor xenografts

    Clin Cancer Res

    (2004)
  • F.H. Sarkar et al.

    Using chemopreventive agents to enhance the efficacy of cancer therapy

    Cancer Res

    (2006)
  • B.S. Reddy et al.

    Effect of diets high in omega-3 and omega-6 fatty acids on initiation and postinitiation stages of colon carcinogenesis

    Cancer Res

    (1991)
  • M. Hirose et al.

    Effects of dietary perilla oil, soybean oil and safflower oil on 7,12-dimethylbenz[a]anthracene (DMBA) and 1,2-dimethyl-hydrazine (DMH)-induced mammary gland and colon carcinogenesis in female SD rats

    Carcinogenesis

    (1990)
  • R.A. Karmali et al.

    Effect of omega-3 fatty acids on growth of a rat mammary tumor

    J Natl Cancer Inst

    (1984)
  • M.J. Gonzalez et al.

    Dietary fish oil inhibits human breast carcinoma growth: a function of increased lipid peroxidation

    Lipids

    (1993)
  • D.P. Rose et al.

    Effects of dietary omega-3 fatty acids on human breast cancer growth and metastases in nude mice

    J Natl Cancer Inst

    (1993)
  • Cited by (30)

    • Low-density lipoprotein docosahexaenoic acid nanoparticles induce ferroptotic cell death in hepatocellular carcinoma

      2017, Free Radical Biology and Medicine
      Citation Excerpt :

      However, erastin mediates this process through a different triggering mechanism-the inhibition of system xc- cystine-glutamate transport [10]. The anticancer actions of PUFA, like DHA, have often been attributed to PPAR activation [24,25]. In this study we also sought to determine if these nuclear receptors also mediate LDL-DHA's tumor cytotoxicity.

    • PPARδ signaling mediates the cytotoxicity of DHA in H9c2 cells

      2015, Toxicology Letters
      Citation Excerpt :

      The fundamental difference between immortalized and primary cardiac cells dictates the importance of obtaining a better understanding of DHA-mediated biological events, which is important for the development of DHA-based therapeutic strategies to treat various pathogenic conditions. DHA has been shown to initiate a cytotoxic response in several cancer cell lines through peroxisome proliferator-activated receptors (PPARs) (Tuller et al., 2009). PPARs are ligand-activated transcription factors and members of the nuclear hormone receptor superfamily (Issemann and Green, 1990; Kliewer et al., 1999).

    • Characterization of docosahexaenoic acid (DHA)-induced heme oxygenase-1 (HO-1) expression in human cancer cells: The importance of enhanced BTB and CNC homology 1 (Bach1) degradation

      2014, Journal of Nutritional Biochemistry
      Citation Excerpt :

      Second, DHA is an established PPAR ligand which can activate the PPAR signaling pathway, thereby regulating expression of many target genes [64]. We have previously shown that DHA activates PPARα signaling in A2780 cells and PPARα mediates the suppression of superoxide dismutase 1 expression [39] and the cytotoxicity of DHA [40]. Third, there are well-established AREs and PPREs in the human HO-1 gene promoter, and both DNA elements were previously described to regulate HO-1 gene transcription [14,35,37].

    • Activation of peroxisome proliferator-activated receptor α (PPARα) suppresses hypoxia-inducible factor-1α (HIF-1α) signaling in cancer cells

      2012, Journal of Biological Chemistry
      Citation Excerpt :

      Recent studies demonstrated that these two drugs have anticancer properties in various model systems (9–11), but the mechanisms behind their actions remain to be elucidated. We have reported previously that activation of PPARα mediates the anticancer action of docosahexaenoic acid, likely through down-regulation of hypoxia signaling (12, 13). We therefore hypothesized that the anticancer properties of PPARα agonists may similarly be due to suppression of HIF-1α signaling in human cancer cells.

    • A novel 2,6-diisopropylphenyl-docosahexaenoamide conjugate induces apoptosis in T cell acute lymphoblastic leukemia cell lines

      2011, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Docosahexaenoic acid (22:6, DHA), a long chain dietary polyunsaturated fatty acid has been most widely used as the fatty acid of choice for conjugating drugs. DHA itself possesses moderate anticancer activities against a variety of cancers and shows synergy in pre-clinical models when used in combination with certain drugs and nutritional compounds [4–9]. Although these studies demonstrate the utility of using a polyunsaturated fatty acid ester for prodrugs to enhance efficacy of cancer chemotherapy, examples are lacking of chemicals that utilize an enhanced DHA effect on cancer cells.

    View all citing articles on Scopus
    View full text