Uptake transporter organic anion transporting polypeptide 1B3 contributes to the growth of estrogen-dependent breast cancer

https://doi.org/10.1016/j.jsbmb.2010.06.014Get rights and content

Abstract

Estrone-3-sulfate is one of the most abundant estrogen precursors in postmenopausal women. We previously showed that estrone-3-sulfate transporters are present in human breast cancer-derived MCF-7 cells (J. Pharmacol. Exp. Ther. 311 (2004) 1032–1037) and that inhibition of estrone-3-sulfate uptake resulted in the suppression of cell growth (Pharm. Res. 22 (2005) 1634–1641); therefore, estrone-3-sulfate transporter should be a novel target for therapy of hormone-dependent breast cancers. The purpose of the present study is to identify the transporter(s) responsible for the uptake of estrone-3-sulfate in breast cancer cells. We obtained two subclones of MCF-7 cells with different estrone-3-sulfate uptake activities and searched for differentially expressed transporter genes by means of DNA microarray analysis. Among several candidate transporters identified, OATP1B3 was further evaluated, since the uptake characteristics of estrone-3-sulfate by MCF-7 cells seemed consistent with the transport properties of OATP1B3. The contribution of OATP1B3 to estrone-3-sulfate uptake by MCF-7 cells was examined by the relative activity factor (RAF) method, and was calculated to amount to 6%. This result suggests that OATP1B3 is one of the transporters contributing to the supply of the estrogen precursor estrone-3-sulfate to estrogen-dependent breast cancer cells.

Introduction

Breast cancer is one of the major causes of cancer death in women. Since two-thirds of breast cancers are estrogen-dependent, estrogen is an important risk factor for progression of breast tumors [1]. However, breast cancers frequently occur during the postmenopausal period, when the circulating estrogen level is low because of loss of ovarian function [2], [3]. Although the circulating level of estrogen is low, tissue concentrations of estrogen in breast cancer are significantly higher than those found in the plasma or in adjacent normal breast tissues, suggesting a specific mechanism of local biosynthesis from precursors [2]. The biologically active form of estrogen is estradiol, which is synthesized from the precursors through two main pathways; one is the aromatase pathway, in which aromatase converts androgens to estrogens, and the other is the sulfatase pathway, in which sulfatase converts estrone-3-sulfate to estrone. Both of them are currently molecular targets of endocrine therapy of breast cancer. Although estrone-3-sulfate is a biologically weak ligand of estrogen receptor, it is one of the most important forms of circulating estrogen. The plasma concentration of estrone-3-sulfate is about 5–10 times higher than that of other conjugated estrogens, and its half-life is longer than that of estradiol in postmenopausal women [4], [5], [6]. Moreover, sulfatase activity is 50–200 times higher than aromatase activity in breast cancer cells [6], [7], [8]. In addition, sulfatase activity in breast cancer cells is higher than that of normal breast cells [9]. Therefore, estrone-3-sulfate is thought to play an important role in the progression of breast cancer as a precursor of active estrogen.

Estrone-3-sulfate is highly hydrophilic, so that it cannot readily cross the plasma membrane, whereas unconjugated estrogens, such as estrone and estradiol, are lipophilic and can enter cells via simple diffusion. Thus, estrone-3-sulfate import into cells across the plasma membrane is expected to involve active transport. Indeed, we previously reported that the uptake of estrone-3-sulfate across the plasma membrane in T-47D cells and MCF-7 cells, which are estrogen-dependent breast cancer cell lines, is mediated by a specific transport mechanism [10], [11].

Accordingly, we hypothesized that the transporter responsible for this transport process could be a novel target for endocrine therapy of estrogen-dependent breast cancers. To develop this concept further, it is necessary to identify the estrone-3-sulfate uptake transporter(s) in breast cancer cells. In our previous study, mRNA expression of some organic anion transporting peptide (OATP) and organic anion transporter (OAT) molecules was detected in MCF-7 cells by RT-PCR [11]. In addition, the uptake of estrone-3-sulfate by MCF-7 cells and T-47D cells was inhibited by anionic compounds, such as bromosulfophthalein, taurocholate, cholate, and probenecid, which are substrates of OATPs or OATs. Some of these transporters may be involved in the uptake of estrone-3-sulfate in MCF-7 cells, but their contribution remains to be examined.

The purpose of this study is to identify the responsible transporter molecules among previously identified estrone-3-sulfate transporters, based on the correlation between mRNA expression levels and the uptake activity of estrone-3-sulfate in subclones of breast cancer-derived MCF-7 cells with differential transport activity.

Section snippets

Materials

[3H]Estrone-3-sulfate, ammonium salt (1702.0 Gbq/mmol) and [3H]cholecystokinin octapeptide (CCK8) (3590 Gbq/mmol) were purchased from Perkin Elmer Life and Analytical Sciences (Boston, MA) and GE Healthcare UK Ltd. (Buckinghamshire, England), respectively. MCF-7 cells were purchased from American Type Culture Collection (ATCC, Manassas, VA). Fetal bovine serum (FBS) was obtained from Invitrogen Life Technologies (Carlsbad, CA). All other reagents were purchased from Sigma–Aldrich (St. Louis, MO)

Microarray analysis

To identify transporters involved in the uptake of estrone-3-sulfate into MCF-7 cells, MCF-7 cells were subcloned and the uptake activity in each clone was measured. Those clones showed different uptake activities for estrone-3-sulfate (data not shown). Among them, two subclones were selected for further experiments, i.e., the one that exhibited the highest activity of estrone-3-sulfate uptake (MCF-7-High cells), and the one that exhibited the lowest activity (MCF-7-Low cells) (Fig. 1). The

Discussion

Estrone-3-sulfate is a major circulating estrogen and also participates in the progression of breast cancer cells [19]. Although estrone-3-sulfate itself is a weak ligand of the estrogen receptor, it is converted to the active estrogen, estradiol, by cytosolic sulfatase and 17β-hydroxysteroid dehydrogenase type 1. Since estrone-3-sulfate is hydrophilic, it was thought that a specific transporter would be involved in import of estrone-3-sulfate across the plasma membrane of breast cancer cells.

Acknowledgment

This study was partly supported by Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology, Japan.

References (28)

  • T. Abe et al.

    LST-2, a human liver-specific organic anion transporter, determines methotrexate sensitivity in gastrointestinal cancers

    Gastroenterology

    (2001)
  • I.C. Henderson et al.

    Cancer of the breast: the past decade (second of two parts)

    N. Engl. J. Med.

    (1980)
  • J.R. Pasqualini et al.

    Recent insight on the control of enzymes involved in estrogen formation and transformation in human breast cancer

    J. Steroid Biochem. Mol. Biol.

    (2005)
  • T. Suzuki et al.

    In situ production of estrogens in human breast carcinoma

    Breast Cancer

    (2002)
  • Cited by (26)

    • Systematic Identification of Pharmacological Targets from Small-Molecule Phenotypic Screens

      2016, Cell Chemical Biology
      Citation Excerpt :

      For example, DePick predicted that several members of the solute carrier organic anion transporter family (SLCO1A2, SLCO1B3, SLCO1C1, SLCO4C1) are related to the “E-Cadherin Synthetic Lethal” assay. These proteins increase the transport of the metabolite estrone 3-sulfate (Geyer et al., 2004; Maeda et al., 2010; Pizzagalli et al., 2002; Yamaguchi et al., 2010), a precursor of estradiol. Estradiol, in turn, has been shown to decrease the expression of E-cadherin (Oesterreich et al., 2003).

    • Organic anion transporting polypeptide 2B1 expression correlates with uptake of estrone-3-sulfate and cell proliferation in estrogen receptor-positive breast cancer cells

      2015, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      Although some of their transcripts were detected in MCF-7 cells and breast tissues, the expression profiles of OATPs were different between the cell line and breast tissues in the present study. In addition, Maeda et al. [32] obtained two subclones of MCF-7 cells with different E1S uptake activities and examined the contribution of OATP1B3 to E1S uptake by MCF-7 cells. From these, the expression of OATPs may be heterogeneous or depend on the condition of each breast cancer cell.

    • Steroid derivatives as inhibitors of steroid sulfatase

      2013, Journal of Steroid Biochemistry and Molecular Biology
      Citation Excerpt :

      Instead of local production of estrogens in tumors, it has been hypothesized that tumor growth is stimulated in part by estrogens derived from their sulfated precursors which are produced in peripheral tissues. These soluble sulfated precursors are transported into the cancer cells by specific membrane transporters where they are then desulfated by STS [9]. Several lines of evidence suggest that STS plays an important role in the progression of steroid-dependent breast cancer.

    • Enhanced expression of organic anion transporting polypeptides (OATPs) in androgen receptor-positive prostate cancer cells: Possible role of OATP1A2 in adaptive cell growth under androgen-depleted conditions

      2012, Biochemical Pharmacology
      Citation Excerpt :

      We have previously shown that transport of estrone 3-sulfate via OATPs sustains the growth of hormone-dependent human breast cancer cells [7,8], because estrone 3-sulfate can be hydrolyzed by steroid sulfatase (STS) to estrone and eventually estradiol. We recently showed that OATP1B3 contributes to estrone 3-sulfate uptake in estrogen receptor-positive human breast cancer MCF-7 cells [9]. In addition to estrone 3-sulfate, these OATPs translocate dehydroepiandrosterone sulfate (DHEAS) [10,11].

    • Identification of a new organic anion transporting polypeptide 1B3 mRNA isoform primarily expressed in human cancerous tissues and cells

      2012, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Other studies have shown an association between genetic variations in the SLCO1B3 gene and the clinical outcomes in prostate cancer patients [6–8]. Based on these reports, several attempts have been made to identify mechanisms underlying its mode of action using cancer cells [9,10]. However, the results obtained so far cannot fully explain the clinical results, and therefore the precise role of OATP1B3 in cancer tissues and cells has yet to be elucidated.

    View all citing articles on Scopus
    View full text