Elsevier

Biochemical Pharmacology

Volume 74, Issue 11, 3 December 2007, Pages 1587-1595
Biochemical Pharmacology

The antiproliferative effects of phenoxodiol are associated with inhibition of plasma membrane electron transport in tumour cell lines and primary immune cells

https://doi.org/10.1016/j.bcp.2007.08.019Get rights and content

Abstract

Although the redox-active synthetic isoflavene, phenoxodiol, is in Phase 3 clinical trials for drug-resistant ovarian cancer, and in early stage clinical trials for prostate and cervical cancer, its primary molecular target is unknown. Nevertheless, phenoxodiol inhibits proliferation of many cancer cell lines and induces apoptosis by disrupting FLICE-inhibitory protein, FLIP, expression and by caspase-dependent and -independent degradation of the X-linked inhibitor of apoptosis, XIAP. In addition, phenoxodiol sensitizes drug-resistant tumour cells to anticancer drugs including paclitaxel, carboplatin and gemcitabine. Here, we investigate the effects of phenoxodiol on plasma membrane electron transport (PMET) and cell proliferation in human leukemic HL60 cells and mitochondrial gene knockout HL60ρo cells that exhibit elevated PMET. Phenoxodiol inhibited PMET by both HL60 (IC50 32 μM) and HL60ρo (IC50 70 μM) cells, and this was associated with inhibition of cell proliferation (IC50 of 2.8 and 6.7 μM, respectively), pan-caspase activation and apoptosis. Unexpectedly, phenoxodiol also inhibited PMET by activated murine splenic T cells (IC50 of 29 μM) as well as T cell proliferation (IC50 of 2.5 μM). In contrast, proliferation of WI-38 cells and HUVECs was only weakly affected by phenoxodiol. These results indicate that PMET may be a primary target for phenoxodiol in tumour cells and in activated T cells.

Introduction

The synthetic isoflavene, phenoxodiol [1], is in advanced clinical trials for the treatment of drug-resistant ovarian cancer and is in early stage trials for prostate and cervical cancer, both as a single agent and in chemo-sensitization settings. However, despite progression into clinical trials, the primary cellular target or targets of phenoxodiol remain elusive. Rather, the drug is often referred to as a multiple signal transduction regulator [2], a term consistent with its redox activity and multiple targets of the parent compound, genistein. Mechanistic studies have shown that phenoxodiol inhibits DNA topoisomerase II [1], and induces apoptosis in ovarian carcinoma [3], [4] and melanoma cells [5]. Both caspase-dependent and -independent apoptosis have been described. With chemoresistant ovarian cancer cells, phenoxodiol induced early ubiquitination and degradation of the X-linked inhibitor of apoptosis, XIAP, via the proteosome [3], [4], and disrupted expression of the FLICE-inhibitory protein, FLIP, through the Akt signal transduction pathway [4]. With other human cancer cell lines including head and neck and colon carcinoma, phenoxodiol induced caspase-independent apoptosis following G1 cell cycle arrest and this was associated with p21WAF1 expression [6]. Induction of apoptosis by anticancer drugs is often a consequence of a primary site or sites of action physically distinct from the pro-apoptotic machinery rather than being a direct target. Therefore, understanding the primary molecular target and the linkage of this target to key components of apoptotic pathways is central to optimizing drug development and usage.

Fine control of the intracellular redox status of cells is critical for cell viability, growth and function. Perturbation of key redox couples such as the NADH/NAD+ ratio, glutathione balance and membrane ubiquinone redox status and content can have profound effects on cells resulting in apoptosis [7], [8]. Small redox-active amphipathic molecules such as phenoxodiol may disrupt essential redox cycling at many different levels within cells and their membranes. Because phenoxodiol is poorly water-soluble and requires the use of organic solvents, it will have a tendency to partition in membrane systems. This suggests that redox-active molecules in membranes may be primary targets for phenoxodiol action. Two main membrane systems orchestrate the bioenergetic status of cells. The inner mitochondrial membrane contains an electron transport system that is responsible for generating the membrane potential that drives most ATP production via oxidative phosphorylation. Ubiquinone is an essential redox-active mediator of mitochondrial electron transport (MET), shuttling electrons from Complexes I and II to Complex III. In the absence of mitochondrial electron transport, cells must rely on glycolysis to generate ATP. Increasing evidence indicates that the plasma membrane also contains an essential electron transport system that utilizes ubiquinone as a one electron acceptor and donor [9], [10], and that this system may support glycolytic ATP production by oxidizing cytosolic NADH [11], [12], a role usually attributed to lactate dehydrogenase (LDH). This suggests that plasma membrane electron transport (PMET) may be a primary site of action for the anticancer activity of redox-active lipophilic flavonoids such as phenoxodiol, catechins like epigallocatechin gallate, resveratrol and quinone-containing anticancer drugs such as doxorubicin.

Phenoxodiol was recently shown to bind with high affinity to a purified recombinant NADH-oxidase, compromising its ability to oxidize both NADH and ubiquinol and to catalyze protein disulfide–thiol interchange activity [13]. This protein is a truncated form of a tumour-specific cell surface NADH-oxidase, tNOX, thought to be involved in the transfer of electrons from intracellular NADH to an extracellular acceptor via plasma membrane ubiquinone [14], [15]. These effects of phenoxodiol were shown to be associated with inhibition of HeLa cell enlargement and growth [13]. In contrast, activity of the constitutively-expressed cell surface NADH-oxidase, CNOX, present on both cancer and non-cancer cells was said to be unaffected by phenoxodiol.

In order to directly assess the effects of phenoxodiol on PMET, we have used mitochondrial gene knockout (ρ°) cells [11], [16] in which bioactivity can be attributed to interaction with redox-sensitive systems other than MET, greatly simplifying interpretation of results. Using this system, we investigated the effects of phenoxodiol on PMET and cell proliferation in human leukemic HL60 cells and HL60ρo cells, activated murine splenic T cells and primary human cells in culture. Phenoxodiol inhibited PMET and cell proliferation of both HL60 cells and activated primary splenic T cells to a similar extent, while HL60ρo cells were less sensitive. These results support a primary mechanism of action of phenoxodiol that includes PMET and indicate that inhibition of PMET compromises proliferation of both HL60 cells and activated primary immune cells.

Section snippets

Cells and cell culture

HL60 cells, originally from ATCC, were obtained from Dr. Graeme Findlay (University of Auckland, NZ); WI-38 at passage 7 were from Dr. David Brown (Novogen, Australia), and were expanded from passages 9–10 stocks; primary HUVEC cells at passages 3–4 were from Dr. Sara Gunningham (Christchurch School of Medicine and Health Science, University of Otago). All cell lines used were free of mycoplasma. The mitochondrial DNA-knockout cell line, HL60ρo, was derived from its parental cell line, HL60, by

Effects of phenoxodiol on PMET by HL60, HL60ρo and primary cells

The effects of phenoxodiol on PMET by human leukemic HL60 cells and HL60ρo cells were determined by measuring reduction of the cell-impermeable tetrazolium dye, WST-1 in real time, in the presence of its mandatory intermediate electron acceptor, 1 mPMS. Phenoxodiol inhibited PMET by HL60 cells with an IC50 of 32 μM and HL60ρo cells with an IC50 of 70 μM (Fig. 1A; Table 1). To determine whether the effects of phenoxodiol on PMET by HL60 cells were cancer-specific, WST-1 reduction was determined in

Discussion

The question of whether PMET is a target for the anticancer drug, phenoxodiol, was addressed, in this study, by directly measuring reduction of the extracellular tetrazolium dye, WST-1. Strong inhibition of dye reduction by phenoxodiol was observed with activated splenic T cells, HL60 cells, and to a lesser extent with HL60ρo cells. Although HL60ρo cells have elevated PMET compared with HL60 cells [11], they have a longer cell cycle time that could in part explain their reduced sensitivity to

Acknowledgements

This research was supported by Marshall Edwards Inc., the Cancer Society of New Zealand, by Genesis Oncology Trust and by the Department of Radiation Therapy, Wellington School of Medicine and Health Sciences, University of Otago.

References (21)

There are more references available in the full text version of this article.

Cited by (0)

View full text