NP-1250, an ABCG2 inhibitor, induces apoptotic cell death in mitoxantrone-resistant breast carcinoma MCF7 cells via a caspase-independent pathway

  • Authors:
    • Masumi Ito
    • Kazunori Kajino
    • Masaaki Abe
    • Tsutomu Fujimura
    • Reiko Mineki
    • Takako Ikegami
    • Toshihisa Ishikawa
    • Okio Hino
  • View Affiliations

  • Published online on: January 24, 2013     https://doi.org/10.3892/or.2013.2249
  • Pages: 1492-1500
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chemoresistance is one of the main obstacles to successful cancer therapy and is frequently associated with multidrug resistance (MDR). One of the most studied mechanisms of MDR is the high expression of ATP-binding cassette (ABC) transporters. Here, we demonstrated that NP-1250, an ABCG2 inhibitor, induced apoptotic cell death in ABCG2-overexpressing multidrug-resistant MCF7/mitoxantrone-resistant (MX) human breast carcinoma cells via a caspase-independent pathway. Incubation of MCF7/MX cells with NP-1250 significantly reduced cell viability, while NP-1250 had little effect on the viability of drug-sensitive MCF7/wild-type cells. Although the target molecules of NP-1250 in cell death remain unknown, investigation of NP-1250 will aid in the elucidation of the molecular mechanism of drug resistance and NP-1250 may become a new therapy for MDR cancers.

Introduction

A frequent problem in cancer chemotherapy is the development of multidrug resistance (MDR) which renders tumors unresponsive to a diverse array of compounds. There are a number of mechanisms by which a cell can acquire MDR, one of the most common being overexpression of members of the ATP-binding cassette (ABC) transporter family. This is a group of plasma membrane proteins that actively extrude a broad range of substrates from cells. They are predominantly found in areas such as the epithelial cells of the intestine, hepatocytes, capillary epithelial cells of the blood-brain barrier, and in various stem cells where the physiological role of these ATPase efflux ‘pumps’ is to protect cells from damage by rapidly extruding xenobiotics (1). Unfortunately, their unwanted expression in tumor cells can lead to chemotherapy resistance in these cells (2).

Most research concerning ABC transporter-overexpressing MDR cancer cells have focused on molecules or signaling pathways that regulate expression of these transporters (3,4), and a few potential therapeutic target molecules have been identified. In recent years, it has been reported that suppression of ABC transporters inhibits cancer cell proliferation, particularly in transporter-overexpressing MDR cancer cells (5,6). However, there are no reports concerning the relationship between inhibition of ABC transporters and cell death.

In the present study, we demonstrated that treatment with NP-1250, an ABCG2 inhibitor, alone induced apoptotic cell death via a caspase-independent pathway in MDR MCF7 breast cancer cells (MCF7/MX). MCF7/MX cells (1) have the MDR phenotype characterized by high levels of the ABCG2 transporter. We speculated that NP-1250 would have an effect on molecules involved in growth and survival, whose expression levels have been elevated in the process of acquiring resistance to anticancer drugs. Using a proteomics approach, we identified three candidate molecules: peroxiredoxin-2, protein disulfide-isomerase A4 and prohibitin-2. These molecules may be potential candidates for the treatment of MDR cancers.

Materials and methods

Materials

Pan-caspase inhibitor (Z-VAD-FMK) and caspase-3/7-specific inhibitor (Ac-DEVD-CHO) were purchased from Promega (Madison, WI, USA). Doxorubicin was obtained from Kyowa Hakko Kirin (Tokyo, Japan) and mitoxantrone hydrochloride was obtained from Wyeth Lederle Japan, Ltd. (Tokyo, Japan). NP-1250 was synthesized at Nippon Chemiphar Co., Ltd. (Saitama, Japan). It is still under development, therefore, disclosure of the structure of this compound is prohibited.

Cell cultures and treatment

The MCF7 human breast cancer cell line, the HL-60 human promyelocytic leukemia cell line, the multidrug-resistant HL-60/MX1 cell line, the MES-SA human uterus sarcoma cell line, and the multidrug-resistant MES-SA/Mx2 and MES-SA/Dx5 cancer cell lines were obtained from American Type Culture Collection (ATCC, Rockville, MD, USA) and cultured in the recommended medium containing 10% heat-inactivated fetal bovine serum (FBS) at 37°C in a 95% air/5% CO2 atmosphere. Mitoxantrone-resistant MCF7 (MCF7/MX) cells (7) were a gift from Dr Masayuki Nakagawa (Department of Urology, Kagoshima University, Kagoshima, Japan) and doxorubicin-resistant MCF7 (MCF7/DOX) cells were a gift from Dr Takao Yamori (Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan). Prior to any experiments, MCF7/MX cells and MCF7/DOX cells were maintained in drug-free medium for one passage. Other resistant cells were cultured in the absence of anticancer drugs. ABCG2-overexpressing Flp-In-293 (Flp-In-293/ABCG2) cells and mock-transfected Flp-In-293 (Flp-In-293/Mock) cells were established and cultured as previously described (8).

Evaluation of the inhibitory effect on the ABCG2 transporter

To evaluate ABCG2-NP-1250 interaction, plasma membrane vesicles prepared from ABCG2-expressing Sf9 cells were incubated with [3H]methotrexate (MTX). Transport into the vesicles was measured by counting the radioactivity remaining on the filter of MultiScreen plates, as previously described (9).

Cell viability assay

To evaluate the effectiveness of NP-1250 in overcoming ABCG2-mediated drug resistance, Flp-In-293/ABCG2 and MCF7/MX cells were incubated with anticancer drug SN-38 and NP-1250, and the cell viabilities were measured.

Tumor cells were seeded in 96-well plates. After 24 h, the medium was replaced with medium containing different concentrations of NP-1250 in triplicate. After a 72-h incubation at 37°C, cell viability was determined at the indicated times, as previously described (10).

TUNEL staining

MCF7/WT and MCF7/MX cells were seeded in 35-mm glass-bottom dishes. After 24 h, the medium was replaced with medium containing 10, 20 or 40 μM of NP-1250. After a 48-h incubation at 37°C, TUNEL staining was performed as previously described (10). The frequency (%) of TUNEL-positive cells per DAPI-positive cells was determined in five different fields in duplicate. Two replicate experiments were performed.

Annexin V/PI staining

The Annexin V-FITC fluorescence microscopy kit (Pharmingen, San Diego, CA, USA) was used to detect apoptosis. MCF7/MX cells were seeded in 35-mm glass-bottom dishes. After 6, 12, 24 or 48 h of incubation with 20 μM of NP-1250 at 37°C, Annexin V/propidium iodide (PI) staining was performed according to the manufacturer's protocol. Stained cells were visualized using a confocal microscope (Leica).

RNA isolation and cDNA synthesis

Total RNA was prepared using the FastPure® RNA kit (9190; Takara, Shiga, Japan) according to the manufacturer's protocol, and RNA concentrations were measured at an absorbance A260. Reverse transcription (RT) was carried out with 1 μg RNA using a ReverTraAce® qPCR RT kit (FSQ-101; Toyobo, Osaka, Japan).

Real-time PCR

Real-time PCR was carried out using Fast SYBR-Green Master Mix (4385612, Applied Biosystems, Life Technologies Corp., Carlsbad, CA, USA) using a 7500 Fast Real-Time PCR system (Applied Biosystems). All primers used for real-time PCR are described in Table I. PCR was carried out with an initial 20-sec denaturation at 95°C followed by 40 cycles of PCR (3 sec at 95°C and 30 sec at 60°C). Reactions were performed in triplicate for three biological replicates, and the amount of each cDNA was normalized to GAPDH gene expression. Melt-curve analysis was performed to ensure that the mRNA-specific fragments were amplified and data were analyzed using the standard curve method.

Table I

Sequence of primers for real-time PCR.

Table I

Sequence of primers for real-time PCR.

GeneDirectionPrimer sequence (5′-3′)Product size (bp)
GAPDHForward GCCATCAATGACCCCTTC114
Reverse GATGACAAGCTTCCCGTTC
ABCB1Forward AACTTCCGAACCGTTGTTTC110
Reverse CCAAAGATGTGTGCTTTCCTC
ABCC1Forward CTACCTCCTGTGGCTGAATC150
Reverse ATCAGCTTGATCCGATTGTC
ABCG2Forward ACAGGTGGAGGCAAATCTTC94
Reverse GCGGTGCTCCATTTATCAG
PRDX1Forward CTGTCATCTAGCATGGGTCAAT106
Reverse CCCATAATCCTGAGCAATG
PRDX2Forward GTCCGTGCGTCTAGCCTTTG128
Reverse CAGCTTCACCTCTTTGAAGG
LGULForward CCAAGGATTTTCTATTGCAG97
Reverse TGGATTAGCGTCATTCCAAGA
GLTPForward CCCTTCTTCGATTGCCTTG107
Reverse AACTTGGCTGGGTTGGTGTC
TAGL2Forward CCAAACTGTGGACCTCTGG143
Reverse GATTCTCCTTGGATTTCTTAGGG
PSB5Forward CCACCCTGGCCTTCAAGTTC132
Reverse CCATGGTGCCTAGCAGGTATG
PEBP1Forward GCCCACCCAGGTTAAGAATAG145
Reverse GACCACCAGGAAATGATGC
GNA1Forward CCTGTGCTAAGAGAGGAAGAGT143
Reverse TGGTAGACATTCAAGGGTAATC
DCTP1Forward CTTTCAGTGGAAAACCGATG144
Reverse ACTGCTAGCGGCAGATCCAC
VDAC2Forward GATTTGGTTTTGGGTTGGTG115
Reverse TCCAAGGTCCCAGTAACTTT
P5CR1Forward TGGACCTGGCCACAGTTT103
Reverse TTCACAGCCAGGAAGAGCACAT
SFXN1Forward TTGGCTTCTGTTTGGTGTTT113
Reverse GCTCTCTTGGATCTTAGCTTGC
PDIA4Forward CCGCTAACAACCTGAGAGAAG105
Reverse CAGGCTGCATTACAACCAAC
HNRPMForward AAGGGTGAAGGAGAACGACCTG111
Reverse GGCTCTGTATCTTTTAGTTGGA
ECHAForward AACTCTCCCAATTCAAAGGT117
Reverse TGATGAGATAAGGACGGCACT
GPDMForward TCACCAGAGGACTAAAAACAGC91
Reverse ACACCACCATGGATCAATTT
IPYRForward AATGGAGATTGCTACAAAGGAC142
Reverse TGGGTCTTCCCAAGTCTG
OTUB1Forward CAGCGGTTCAAGGCTGTGT133
Reverse CGACAGAGGTCTGCTTCTCC
PHB2Forward CTCCAAAGACCTACAGATGGTG130
Reverse GTTGACAATGGACGGCAACA
GGCTForward CCGCCTGCAGGATTTTAAG98
Reverse CGCCAGGACTCTGAAAAATG
LDHAForward CTGTCATGGGTGGGTCCTT106
Reverse CCCTAAATCTGGGTGCAGAGTC
Immunofluorescence staining of ABCG2

Cells were fixed in 4% paraformaldehyde, permeabilized with 0.2% Triton X-100, and blocked with 1% bovine serum albumin and 10% normal goat serum in phosphate-buffered saline (PBS). Incubation with primary monoclonal antibody for ABCG2 (ALX-801-029; Wako, Osaka, Japan) at a dilution of 1:200 was carried out overnight at 4°C, followed by incubation with the Alexa Fluor-conjugated secondary antibody (A11001; Molecular Probes, Inc., Carlsbad, CA, USA) for 1 h at room temperature. Immunofluorescence was detected and analyzed using a Leica TCS SP5 Laser Scanning Confocal Microscope system.

Western blot analysis

Cells were washed two times with PBS, harvested by scraping from the culture dishes in lysis buffer and collected. Protein concentrations were determined using the DC protein assay kit according to the manufacturer's instructions (500-0116; Bio-Rad, Hercules, CA, USA). Equal amounts of proteins were denatured by boiling and subjected to SDS-polyacrylamide gel electrophoresis. The separated proteins were transferred to polyvinylidene difluoride membranes. The membranes were incubated overnight at 4°C with primary polyclonal antibody at a dilution of 1:1000 for caspase-3, cleaved-PARP, cleaved-caspase-7 or cleaved-caspase-9 (9665, 9541, 9491, 9501; Cell Signaling Technology, Beverly, MA, USA). Membranes were then incubated with goat anti-rabbit IgG-HRP secondary antibody for 1 h at room temperature and visualized using an enhanced chemiluminescence detection system.

Proteomics
SDS-PAGE and detection by MS compatible silver staining

MCF7/WT and MCF7/MX cells were fractionated using the ProteoExtract® Subcellular Proteome Extraction kit (Merck KGaA, Darmstadt, Germany). Protein samples (5 μg) were separated by SDS-PAGE. Upon completion, the gel was transferred to a clean and dry glass chamber for silver staining using the PlusOne Silver Staining kit, Protein (GE Healthcare, Hino, Japan).

Identification of protein

In-gel digestion using trypsin was performed as previously described (11). The tryptic peptides were used for identification of proteins by mass spectrometry. The digested peptides were analyzed using a nanoflow LC-MS/MS system with a direct nanoflow LC system (DiNa; KYA Technologies, Tokyo, Japan) and a LTQ Orbitrap XL-ETD mass spectrometer (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Data were searched against NCBI human sequence database using Mascot (Matrix Science, Ltd., London, UK).

Statistical analysis

The results are expressed as means ± SD. Each value represents the mean ± SD of two or three independent experiments. One-way analysis of variance (ANOVA) with Dunnett multiple comparison test and t-test were performed, and P<0.01 was considered to indicate a statistically significant difference.

Results and Discussion

NP-1250 is an ABCG2 inhibitor and treatment alone reduces the cell viability of ABCG2-overexpressing MCF7/MX cells

We measured ATP-dependent [3H]MTX transport in plasma membrane vesicles, prepared from ABCG2-expressing Sf9 cells, in the presence of a potassium salt of NP-1250. Potassium salt of NP-1250 inhibited ABCG2-mediated MTX transport in a dose-dependent manner (Fig. 1A). This degree of inhibition was comparable to gefitinib (9), suggesting that NP-1250 is a powerful ABCG2 inhibitor. To examine whether NP-1250 overcomes ABCG2-mediated resistance, the cytotoxic effect of ABCG2-substrate SN-38 in the presence of NP-1250 on Flp-In-293/ABCG2 cells was measured. NP-1250 dose-dependently reversed SN-38 resistance in Flp-In-293/ABCG2 cells (Fig. 1B). We then evaluated NP-1250 on MCF7/MX cells, a mitoxantrone-resistant cell line in which ABCG2 was amplified and overexpressed (Fig. 2) by chronic exposure to mitoxantrone. Differing from our expectation, NP-1250 exhibited no reversing effect on MCF7/MX cells at concentrations <20 μM (Fig. 1C). Notably, 20 μM of NP-1250 inhibited cell viability with a very low dose of SN-38. These results suggest that ABCG2-inhibitor NP-1250 itself may have a cytotoxic effect.

NP-1250 induces apoptotic cell death in MCF7/MX cells

We compared the chemosensitivity to NP-1250 of drug-sensitive MCF7/WT cells and multidrug-resistant MCF7/MX cells by examining the cell viability and cell morphological changes. We discovered that NP-1250 alone reduced the cell viability of MCF7/MX cells, notably, more than that of MCF7/WT cells (Fig. 3A). For the cell morphological study, the number of MCF7/MX cells was significantly less than the number of cells in the control and the adherent cells displayed large vacuoles in the cytoplasm, exhibited atrophic changes or were almost dying (Fig. 3B). In contrast, little change was observed in MCF7/WT cells after a 72-h treatment (Fig. 3B). To confirm these initial observations, we analyzed apoptosis by TUNEL assay and phosphatidylserine (PS) externalization. After a 48-h treatment with NP-1250, in MCF7/MX cells, the number of TUNEL-positive cells was elevated (Fig. 3C) and the TUNEL-positive rate was significantly increased in a dose-dependent manner (Fig. 3D). Apoptotic cell death observed in the MCF7/MX cells upon treatment with NP-1250 was further confirmed by determining Annexin V binding to PS translocated from the inner to the outer membrane surface of the plasma membrane, a hallmark of early stage apoptosis (Fig. 3E).

NP-1250-induced cell death is ‘caspase-independent apoptosis’

After 48 h of treatment with NP-1250, cleavage of PARP was detected using an antibody recognizing the 89-kDa fragment (Fig. 4A). Although PARP is a well-known substrate of caspases, particularly caspase-3 and caspase-7, MCF7 cells do not express caspase-3 due to the deletion of 125 nucleotides in the CASP-3 mRNA (12). We confirmed the lack of caspase-3 protein in these MCF7/WT and MCF7/MX cells (data not shown). We, therefore, examined whether caspase-7 and caspase-9 were cleaved prior to cleavage of PARP, but we did not detect either one (Fig. 4A). To further ascertain whether NP-1250-induced apoptosis of MCF7/MX cells was caspase-dependent or -independent, MCF7/MX cells were pretreated with a caspase inhibitor prior to NP-1250 treatment, and cleavage of PARP was subsequently examined. Pretreatment with the pan-specific inhibitor completely abolished the cleavage of PARP, and the pharmacological inhibitor specific for caspase-3/7 also showed a significant inhibition of the cleavage of PARP (Fig. 4B). Under the same experimental conditions, pretreatment of MCF7/MX cells with these inhibitors did not block the production of apoptotic cells (Fig. 4C). These results suggest that caspases, which cleave PARP, are cleaved and activated upon exposure of MCF7/MX cells to NP-1250 but that this is not the main apoptotic cascade in the death of MCF7/MX cells.

To evaluate the effects of NP-1250 on other MDR cell lines that exhibit increased expression of ABC transporters, we treated several MDR cell lines and their drug-sensitive parental cell lines with NP-1250. We did not observe growth inhibition by NP-1250 in any of the cell lines (data not shown). We also treated several other cancer cell lines, specifically human hepatocellular carcinoma cell line HuH7, human cervical adenocarcinoma cell line HeLa and human mesothelioma cell lines H226 and MESO4, with NP-1250, but we did not confirm any effect (data not shown). Taken together, these results suggest that the antiproliferative/apoptotic effect of NP-1250 may be limited to MCF7/MX cells.

Search for target proteins by proteomics

There are no reports relating inhibition of ABC transporters to cell death, therefore, we speculated that NP-1250 may have other targets in MCF7/MX cells.

We excised several bands that were observed in the protein electrophoretograms of MCF7/MX cells. As a control, we also excised the band that appeared to be ABCG2, ~74 kDa, as observed in the membrane/organelle fraction of MCF7/MX cells. ABCG2 was identified in this band with a Mascot score of 192, thus we excluded proteins whose Mascot scores were <100. We also excluded proteins observed at positions >15 kDa different from their original molecular weight. Candidate proteins that passed our identification criteria are listed in Table II. We selected molecules preferentially involved in cell growth and survival from these proteins and performed real-time PCR to confirm whether the expression of genes encoding these proteins was increased in the MCF7/MX cells when compared to the MCF7/WT cells. Among the 46 proteins, we analyzed the gene expression of 22 proteins, including ABCG2. Three of these proteins, peroxiredoxin-2 (PRDX2), protein disulfide-isomerase A4 (PDIA4) and prohibitin-2 (PHB2), showed significantly elevated mRNA expression in MCF7/MX cells compared to the expression levels in MCF7/WT cells (Fig. 5). PDIA4 is a protein disulfide isomerase (PDI), an enzyme that catalyzes disulfide formation and isomerization and a chaperone that inhibits aggregation (13) and a member of a large family of dithiol/disulfide oxidoreductases, the thioredoxin superfamily. While it has been reported that PDI shows an inverse correlation with resistance (14), Liu et al reported that increased expression of PDI was found in multidrug-resistant MCF7/AdVp3000 cells when compared to MCF7 cells (15). PHB2, known as a repressor of estrogen receptor (ER) activity, has been shown to interact with and inhibit the transcriptional activity of the ER (16). While PHB2 has been implicated in a previously uncharacterized pathway of multidrug resistance in Caenorhabditis elegans(17), Keenan et al found that PHB2 was inversely correlated with resistance in a squamous lung cancer cell line (14). PRDX2 has been previously known as a natural killer-enhancing factor B (18) and is induced by various oxidative stimuli. It plays an important protective role against oxidative radical damage by reactive oxygen and nitrogen species (19). Its expression is correlated with resistance to apoptosis induced by radiation therapy or anticancer drugs (20), highlighting the potential clinical importance of PRDX2 in chemotherapy resistance in cancer. However, the association of PRDX2 and MCF7/MX cells has not been previously reported. Thus further studies, such as knockdown experiments, are warranted.

Table II

The candidate target proteins of NP-1250 identified by mass spectrometry.

Table II

The candidate target proteins of NP-1250 identified by mass spectrometry.

No.Prot_accaIdentified proteinaProt_pIbProt_massbProt_scorec
1PRDX2_HUMAN Peroxiredoxin-25.6622,091533
2RL9_HUMAN60S ribosomal protein L99.9621,992274
3RL18_HUMAN60S ribosomal protein L1811.7321,763232
4RAB7A_HUMANRas-related protein Rab-7a6.4023,830191
5RS5_HUMAN40S ribosomal protein S59.7323,075256
6LGUL_HUMANLactoylglutathione lyase5.1221,048145
7RHOA_HUMANTransforming protein RhoA5.8322,180184
8COMD3_HUMANCOMM domain-containing protein 35.6322,421118
9RAB10_HUMANRas-related protein Rab-108.5922,811150
10PRDX1_HUMAN Peroxiredoxin-18.2722,380146
11GLTP_HUMANGlycolipid transfer protein6.9024,048112
12TAGL2_HUMANTransgelin-28.4122,590648
13PSB5_HUMANProteasome subunit β type-56.4328,675420
14PEBP1_HUMAN Phosphatidylethanolamine-binding protein 17.0121,186366
15RS7_HUMAN40S ribosomal protein S710.0922,113248
16RS9_HUMAN40S ribosomal protein S910.6622,649262
17GGCT_HUMAN γ-glutamylcyclotransferase5.0721,278185
18GNA1_HUMANGlucosamine 6-phosphate N-acetyltransferase8.1721,162147
19DCTP1_HUMANdCTP pyrophosphatase 14.9318,811105
20RCL_HUMANDeoxyribonucleoside 5′-monophosphate N-glycosidase4.9719,239110
21DCD_HUMANDermcidin6.0811,419100
22LDHA_HUMANL-lactate dehydrogenase A chain8.4437,021566
23PCNA_HUMANProliferating cell nuclear antigen4.5729,177346
24RL5_HUMAN60S ribosomal protein L59.7334,625333
25IPYR_HUMANInorganic pyrophosphatase5.5433,207284
26RL6_HUMAN60S ribosomal protein L610.5932,779289
27RA1L3_HUMANPutative heterogeneous nuclear ribonucleoprotein A1-like 39.2334,415180
28OTUB1_HUMANUbiquitin thioesterase OTUB14.8531,549176
29CHIP_HUMANE3 ubiquitin-protein ligase CHIP5.6135,403110
30AN32E_HUMANAcidic leucine-rich nuclear phosphoprotein 32 family member E3.7730,958106
31PP4C_HUMAN Serine/threonine-protein phosphatase 4 catalytic subunit4.9135,768115
32NACA_HUMANNascent polypeptide-associated complex subunit α4.5223,370117
33PHB2_HUMANProhibitin-29.8333,276489
34VDAC2_HUMANVoltage-dependent anion-selective channel protein 27.4932,186319
35ROA1_HUMANHeterogeneous nuclear ribonucleoprotein A19.1738,865238
36P5CR1_HUMAN Pyrroline-5-carboxylate reductase 1, mitochondrial7.1833,624356
37EFTS_HUMANElongation factor Ts, mitochondrial8.6235,794157
38ELAV1_HUMANELAV-like protein 19.2336,282143
39SFXN1_HUMANSideroflexin-19.2235,952108
40PDIA4_HUMANProtein disulfide-isomerase A44.9673,3131,623
41HNRPM_HUMANHeterogeneous nuclear ribonucleoprotein M8.8477,819566
42ECHA_HUMANTrifunctional enzyme subunit α, mitochondrial9.1683,870331
43ABCG2_HUMANATP-binding cassette sub-family G member 28.9173,120192
44GPDM_HUMAN Glycerol-3-phosphate dehydrogenase, mitochondrial7.5781,441262
45TRAP1_HUMANHeat shock protein 75 kDa, mitochondrial8.3080,415117
46GRP78_HUMANGlucose-regulated protein 78 kDa5.0772,431248

a Proteins were derived from Swiss-Prot and the National Center for Biotechnology Information non-redundant databases.

b Theoretical pI and molecular weight obtained from Swiss-Prot.

c MASCOT score for the identified proteins based on the peptide ion score.

In the present study, NP-1250 showed no cytostatic effect on other MDR cancer cell lines. However, it may be effective against specific tumors that are clinically similar to MCF7/MX cells and there is still a possibility that it may lead to the identification of new therapeutic molecules for MDR cancers.

Acknowledgements

We thank Dr Takaichi Hamano (Nippon Chemiphar Co., Ltd.) for the helpful discussions. This study was supported by a Grant-in-Aid for Cancer Research and Grants-in Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan, and the Ministry of Health, Labor and Welfare of Japan.

Abbreviations:

ABC

ATP-binding cassette

Flp-In-293/ABCG2 cells

ABCG2-overexpressing Flp-In-293 cells

Flp-In-293/Mock cells

mock-transfected Flp-In-293 cells

MCF7/MX cells

mitoxantrone-resistant MCF7 cells

MCF7/WT cells

wild-type MCF7 cells

MDR

multidrug resistance

MTX

methotrexate

PARP

poly(ADP-ribose) polymerase

PDIA4

protein disulfide-isomerase A4

PHB2

prohibitin-2

PRDX2

peroxiredoxin-2

References

1 

Scheffer GL, Maliepaard M, Pijnenborg AC, et al: Breast cancer resistance protein is localized at the plasma membrane in mitoxantrone- and topotecan-resistant cell lines. Cancer Res. 60:2589–2593. 2000.PubMed/NCBI

2 

Kuo MT: Roles of multidrug resistance genes in breast cancer chemoresistance. Adv Exp Med Biol. 608:23–30. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Guo X, Ma N, Wang J, Song J, Bu X, Cheng Y, et al: Increased p38-MAPK is responsible for chemotherapy resistance in human gastric cancer cells. BMC Cancer. 8:3752008. View Article : Google Scholar : PubMed/NCBI

4 

Honma K, Iwao-Koizumi K, Takeshita F, Yamamoto Y, Yoshida T, Nishio K, et al: RPN2 gene confers docetaxel resistance in breast cancer. Nat Med. 14:939–948. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Katoh SY, Ueno M and Takakura N: Involvement of MDR1 function in proliferation of tumour cells. J Biochem. 143:517–524. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Chen Z, Liu F, Ren Q, Zhao Q, Ren H, Lu S, et al: Suppression of ABCG2 inhibits cancer cell proliferation. Int J Cancer. 126:841–851. 2010.PubMed/NCBI

7 

Nakagawa M, Schneider E, Dixon KH, Horton J, Kelley K, Morrow C and Cowan KH: Reduced intracellular drug accumulation in the absence of P-Glycoprotein (mdr1) overexpression in mitoxantrone-resistant human MCF-7 breast cancer cells. Cancer Res. 52:6175–6181. 1992.PubMed/NCBI

8 

Wakabayashi K, Nakagawa H, Adachi T, Kii I, Kobatake E, Kudo A and Ishikawa T: Identification of cysteine residues critically involved in homodimer formation and protein expression of human ATP-binding cassette transporter ABCG2: a new approach using the flp recombinase system. J Exp Ther Oncol. 5:205–222. 2006.

9 

Saito H, Hirano H, Nakagawa H, Fukami T, Oosumi K, Murakami K, et al: A new strategy of high-speed screening and quantitative structure-activity relationship analysis to evaluate human ATP-binding cassette transporter ABCG2-drug interactions. J Pharmacol Exp Ther. 317:1114–1124. 2006. View Article : Google Scholar

10 

Wang T, Kajino K, Abe M, Tan K, Maruo M, Sun G, et al: Suppression of cell death by the secretory form of N-terminal ERC/mesothelin. Int J Mol Med. 26:185–191. 2010.PubMed/NCBI

11 

Fujimura T, Shinohara Y, Tissot B, Pang PC, Kurogochi M, Saito S, et al: Glycosylation status of haptoglobin in sera of patients with prostate cancer vs. benign prostate disease of normal subjects. Int J Cancer. 122:39–49. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Janicke RU, Sprengart ML, Wati MR and Porter AG: Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. J Biol Chem. 273:9357–9360. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Wilkinson B and Gilbert HF: Protein disulfide isomerase. Biochim Biophys Acta. 1699:35–44. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Keenan J, Murphy L, Henry M, Meleady P and Clynes M: Proteomic analysis of multidrug-resistance mechanisms in adriamycin-resistant variants of DLKP, a squamous lung cancer cell line. Proteomics. 9:1556–1566. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Liu Y, Liu H, Han B and Zhang JT: Identification of 14-3-3sigma as a contributor to drug resistance in human breast cancer cells using functional proteomic analysis. Cancer Res. 66:3248–3255. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Montano MM, Ekena K, Delage-Mourroux R, Chang W, Martini P and Katzenellenbogen BS: An estrogen receptor-selective coregulator that potentiates the effectiveness of antiestrogens and represses the activity of estrogens. Proc Natl Acad Sci USA. 96:6947–6952. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Zubovych I, Doundoulakis T, Harran PG and Roth MG: A missense mutation in Caenorhabditis elegans prohibitin 2 confers an atypical multidrug resistance. Proc Natl Acad Sci USA. 103:15523–15528. 2006.PubMed/NCBI

18 

Shau H and Kim A: Identification of natural killer enhancing factor as a major antioxidant in human red blood cells. Biochem Biophys Res Commun. 199:83–88. 1994. View Article : Google Scholar : PubMed/NCBI

19 

Kowaltowski AJ, Netto LE and Vercesi AE: The thiol-specific antioxidant enzyme prevents mitochondrial permeability transition evidence for the participation of reactive oxygen species in this mechanism. J Biol Chem. 273:12766–12769. 1998. View Article : Google Scholar

20 

Chung YM, Yoo YD, Park JK, Kim YT and Kim HJ: Increased expression of peroxiredoxin II confers resistance to cisplatin. Anticancer Res. 21:1129–1133. 2001.PubMed/NCBI

Related Articles

Journal Cover

April 2013
Volume 29 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ito M, Kajino K, Abe M, Fujimura T, Mineki R, Ikegami T, Ishikawa T and Hino O: NP-1250, an ABCG2 inhibitor, induces apoptotic cell death in mitoxantrone-resistant breast carcinoma MCF7 cells via a caspase-independent pathway. Oncol Rep 29: 1492-1500, 2013
APA
Ito, M., Kajino, K., Abe, M., Fujimura, T., Mineki, R., Ikegami, T. ... Hino, O. (2013). NP-1250, an ABCG2 inhibitor, induces apoptotic cell death in mitoxantrone-resistant breast carcinoma MCF7 cells via a caspase-independent pathway. Oncology Reports, 29, 1492-1500. https://doi.org/10.3892/or.2013.2249
MLA
Ito, M., Kajino, K., Abe, M., Fujimura, T., Mineki, R., Ikegami, T., Ishikawa, T., Hino, O."NP-1250, an ABCG2 inhibitor, induces apoptotic cell death in mitoxantrone-resistant breast carcinoma MCF7 cells via a caspase-independent pathway". Oncology Reports 29.4 (2013): 1492-1500.
Chicago
Ito, M., Kajino, K., Abe, M., Fujimura, T., Mineki, R., Ikegami, T., Ishikawa, T., Hino, O."NP-1250, an ABCG2 inhibitor, induces apoptotic cell death in mitoxantrone-resistant breast carcinoma MCF7 cells via a caspase-independent pathway". Oncology Reports 29, no. 4 (2013): 1492-1500. https://doi.org/10.3892/or.2013.2249