Cancer Letters

Cancer Letters

Volume 388, 1 March 2017, Pages 149-157
Cancer Letters

Original Article
AG311, a small molecule inhibitor of complex I and hypoxia-induced HIF-1α stabilization

https://doi.org/10.1016/j.canlet.2016.11.040Get rights and content

Highlights

  • AG311 competitively inhibits ubiquinone-binding to complex I and prevents electron transfer and mitochondrial respiration.

  • Cellular response to hypoxia is reduced by AG311, including HIF-1α protein levels, target genes, and oxygen tension.

  • Combination of AG311 and dichloroacetate augments cytotoxicity and tumor growth reduction.

Abstract

Cancer cells have a unique metabolic profile and mitochondria have been shown to play an important role in chemoresistance, tumor progression and metastases. This unique profile can be exploited by mitochondrial-targeted anticancer therapies. A small anticancer molecule, AG311, was previously shown to possess anticancer and antimetastatic activity in two cancer mouse models and to induce mitochondrial depolarization. This study defines the molecular effects of AG311 on the mitochondria to elucidate its observed efficacy. AG311 was found to competitively inhibit complex I activity at the ubiquinone-binding site. Complex I as a target for AG311 was further established by measuring oxygen consumption rate in tumor tissue isolated from AG311-treated mice. Cotreatment of cells and animals with AG311 and dichloroacetate, a pyruvate dehydrogenase kinase inhibitor that increases oxidative metabolism, resulted in synergistic cell kill and reduced tumor growth. The inhibition of mitochondrial oxygen consumption by AG311 was found to reduce HIF-1α stabilization by increasing oxygen tension in hypoxic conditions. Taken together, these results suggest that AG311 at least partially mediates its antitumor effect through inhibition of complex I, which could be exploited in its use as an anticancer agent.

Introduction

In addition to genetic and epigenetic heterogeneity, the concept of metabolic heterogeneity has increased recently. Considerable heterogeneity exists between different tumors, but also within the same tumor. This intratumoral heterogeneity is predominately mediated by genetic diversity, energy demand, and the proximity to vasculature, dictating glucose and oxygen availability. Until recently, cancer cells were thought to rely only on glycolysis for ATP production, even in the presence of sufficient oxygen supply [1], but metabolic heterogeneity demonstrates that there are multiple metabolic phenotypes, going beyond the upregulated glycolysis as observed by Warburg. One of these metabolic phenotypes is mitochondrial respiration, as cancer cells have been shown to have functional mitochondria capable of producing ATP [2], [3], [4].

Anticancer agents acting through mitochondrial inhibition have been described. For example, the anti-diabetic biguanides metformin and phenformin have been shown to exert their anticancer effect by inhibiting complex I, the first enzyme in the mitochondrial electron transport chain (ETC) [5], [6], [7], [8]. BAY-87-2243, another complex I inhibitor, has shown efficacy in a preclinical model of resistant melanoma [9]. The small molecule, IACS-10759, was specifically designed as a complex I inhibitor to target chemoresistant dormant tumors [9], [10]. Complex I, NADH-ubiquinone oxidoreductase, is a crucial player in mitochondrial respiration. It transfers electrons from NADH to reduce ubiquinone to ubiquinol resulting in proton translocation into the mitochondrial intermembrane space, which establishes an electrochemical gradient that is used for ATP synthesis. In order for this process to continue, molecular oxygen is required as the final electron acceptor. Regions of solid tumors often exhibit low oxygen tension (hypoxia), which has been shown to be involved in tumor development, chemo- and radioresistance, and metastasis in addition to tumor bioenergetics [11], [12]. These alterations in bioenergetic processes are mediated in part by increasing gene expression involved in glycolysis and by lowering the activity of the electron transport chain [13], [14], [15]. The major cellular adaptive response to hypoxia is tightly regulated by hypoxia-inducible transcription factor-1α, HIF-1α, which is stabilized in low oxygen tensions, thus an inhibition of complex I can prevent electron transfer and decrease oxygen consumption, which in turn could decrease HIF-1α stabilization [16], [17]. HIF-1α is capable of inducing a broad range of cellular responses including angiogenesis, resistance to apoptosis and tumor energetics/metabolism. Due to the importance of mitochondrial oxidative phosphorylation in hypoxia and in supporting tumor growth, progression and metastasis [18], [19], the development of mitochondrial inhibitors seems well justified.

Herein we investigate the molecular mechanism of a small molecule antitumor agent, AG311. We have previously shown that AG311 significantly reduced primary tumor growth and lung metastases in two breast cancer mouse models, by 81–85% [20]. Upon further investigation, a distinct metabolic mechanism for AG311 emerged. We reported that AG311 rapidly induced necrotic cell death, depolarized the mitochondrial membrane and severely reduced intracellular ATP levels [20]. In the current study, we further define the molecular and antitumor mechanisms and identify complex I of the electron transport chain (ETC) as a likely molecular target of AG311 in isolated cells and in tumors. We show that as a downstream consequence of complex I inhibition, AG311 reduced hypoxia-induced HIF-1α stabilization. Additionally, we show that AG311 synergizes with dichloroacetate (DCA) to increase cell death in cancer cells and increase tumor volume in a xenograft tumor mouse model.

Section snippets

Materials & methods

Additional information is described in the supplemental methods

Preferential cytotoxicity in glucose-deprived cancer cells and mitochondrial depolarization by AG311

Because cancer cells have an increased energy demand, induction of metabolic stress with glucose-depleted media can sensitize them to mitochondrial inhibitors. AG311 showed enhanced cytotoxicity in cancer cells (MDA-MB-435) treated in glucose-depleted medium, whereas docetaxel did not have an effect (Fig. 1A). AG311 has been shown to cause rapid mitochondrial membrane depolarization in cancer cells [20]. The mitochondrial depolarization was more profound in cancerous cells (MDA-MB-435),

Discussion

As a continuation of our previous findings showing that AG311 depolarizes mitochondrial membrane potential and induces necrotic cell death, here we explored the effect of AG311 on mitochondria function. We showed that the AG311-induced mitochondrial membrane depolarization was more pronounced in cancerous cells as compared to normal cells (Fig. 1B). In an endeavor to identify the molecular target of AG311, ETC complexes were tested. It was found that AG311 potently inhibited complex I of the

Acknowledgement

This work was supported, in part, by the National Institutes of Health, National Cancer Institute [Grant CA136944 (A.G.)], by College of Pharmacy startup funds (M.A.I.), and by a Grant-In-Aid of Research, Sigma Xi (A.B.). The authors acknowledge support from the Presbyterian Health Foundation Equipment grant (College of Pharmacy) and the Institutional Development Award (IDeA) from the National Institute of General Medical Sciences of the National Institutes of Health [Grant P20-GM103639]

References (64)

  • Y.W. Choi et al.

    Sensitization of metformin-cytotoxicity by dichloroacetate via reprogramming glucose metabolism in cancer cells

    Cancer Lett.

    (2014)
  • D.B. Shackelford et al.

    LKB1 inactivation dictates therapeutic response of non-small cell lung cancer to the metabolism drug phenformin

    Cancer Cell.

    (2013)
  • R. Vatrinet et al.

    Targeting respiratory complex I to prevent the Warburg effect

    Int. J. Biochem. Cell Biol.

    (2015)
  • E.D. Segal et al.

    Relevance of the OCT1 transporter to the antineoplastic effect of biguanides

    Biochem. Biophys. Res. Commun.

    (2011)
  • R.A. Gatenby et al.

    Why do cancers have high aerobic glycolysis?

    Nat. Rev. Cancer

    (2004)
  • A. Viale et al.

    Tumors and mitochondrial respiration: a neglected connection

    Cancer Res.

    (2015)
  • W.H. Koppenol et al.

    Otto Warburg's contributions to current concepts of cancer metabolism

    Nat. Rev. Cancer

    (2011)
  • R. Moreno-Sánchez et al.

    The bioenergetics of cancer: is glycolysis the main ATP supplier in all tumor cells?

    BioFactors

    (2009)
  • M.R. Owen et al.

    Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain

    Biochem. J.

    (2000)
  • H.R. Bridges et al.

    Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria

    Biochem. J.

    (2014)
  • W.W. Wheaton et al.

    Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis

    eLife

    (2014)
  • S. Matsuzaki et al.

    Selective inhibition of deactivated mitochondrial complex I by biguanides

    Biochemistry

    (2015)
  • L. Schöckel et al.

    Targeting mitochondrial complex I using BAY 87-2243 reduces melanoma tumor growth

    Cancer Metab.

    (2015)
  • M. Protopopova et al.

    Abstract 4380: IACS-10759: a novel OXPHOS inhibitor which selectively kill tumors with metabolic vulnerabilities

    Cancer Res.

    (2015)
  • B. Muz et al.

    The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy

    Hypoxia

    (2015)
  • A.R. Diers et al.

    Mitochondrial bioenergetics of metastatic breast cancer cells in response to dynamic changes in oxygen tension: effects of HIF-1α

    PLoS ONE

    (2013)
  • W.W. Wheaton et al.

    Hypoxia. 2. Hypoxia regulates cellular metabolism

    Am. J. Physiol. Cell Physiol.

    (2011)
  • P. Ellinghaus et al.

    BAY 87-2243, a highly potent and selective inhibitor of hypoxia-induced gene activation has antitumor activities by inhibition of mitochondrial complex I

    Cancer Med.

    (2013)
  • A. Viale et al.

    Tumors and mitochondrial respiration: a neglected connection,

    Cancer Res.

    (2015)
  • A. Bastian et al.

    A small molecule with anticancer and antimetastatic activities induces rapid mitochondrial-associated necrosis in breast cancer,

    J. Pharmacol. Exp. Ther.

    (2015)
  • A. Nerlich

    Density-dependent lineage instability of MDA-MB-435 breast cancer cells

    Oncol. Lett.

    (2013)
  • Q. Zhang et al.

    Human breast cancer cell lines Co-Express neuronal, epithelial, and melanocytic differentiation markers in vitro and in vivo

    PLoS ONE

    (2010)
  • Cited by (39)

    • Metabolic tricks of cancer cells

      2022, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      Many mitochondria-targeting drugs with potential for cancer therapy target electron chain transport complexes. These drugs include complex I inhibitors, such as BAY 87–2243 [199], fenofibrate [200], AG311 [201], and drugs used for type 2 diabetes treatment, such as metformin [202] and canagliflozin [203]. Another complex I inhibitor, IACS-010759, is currently under investigation in phase I clinical trials [204,205].

    View all citing articles on Scopus
    View full text