Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Free Radical Biology and Medicine 110 (2017) 280–290

Contents lists available at ScienceDirect

Free Radical Biology and Medicine


journal homepage: www.elsevier.com/locate/freeradbiomed

Original article

Cold atmospheric plasma restores tamoxifen sensitivity in resistant MCF-7 MARK


breast cancer cell
Seungyeon Leea,1, Hyunkyung Leea,1, Dawoon Jeonga, Juyeon Hama, Sungbin Parka,

Eun Ha Choib, Sun Jung Kima,
a
Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea
b
Plasma Bioscience Research Center, Kwangwoon University, Seoul, Republic of Korea

A R T I C L E I N F O A B S T R A C T

Keywords: Cancer recurrence, which is frequently accompanied by chemotherapy, has been a challenge in cancer treat-
Apoptosis ment. This study was carried out to examine the potential applications of the reactive oxygen species (ROS)-
Cold atmospheric plasma producing cold atmospheric plasma (CAP) to overcome the cancer cells’ drug resistance, which has been
Breast cancer emerging as an alternative therapeutic tool for cancer. For this, we developed a tamoxifen (Tam)-resistant MCF-7
Genome-wide expression
(MCF-7/TamR) breast cancer cell model and examined the effect of CAP on the recovery of Tam sensitivity at the
Reactive oxygen species
cellular and molecular level. The ROS level was increased 1.9-fold in CAP-treated MCF-7/TamR cells compared
to the non-treated cell. CAP was proven to restore sensitivity by up to 50% for MCF-7/TamR cells against Tam
after CAP treatment. The comparison of genome-wide expression between the acquisition of Tam resistance and
CAP treatment identified 20 genes that commonly showed significant expression changes. Notably, all the genes
except two have been oppositely dysregulated in the two cellular statuses, and the majority of them are known to
contribute to the acquisition of Tam resistance. The protein expression of selected genes, MX1 and HOXC6, was
recovered to that of their parental cell by CAP. Furthermore, the dysregulation of MX1 and HOXC6 in MCF-7/
TamR alleviated the drug sensitivity recovery effect of CAP. Taken together, CAP inhibited the growth of Tam-
resistant MCF-7 cancer cells and reset it to the Tam-sensitive status by restoring the expression of drug re-
sistance–related genes. These findings may lend credence to CAP as an alternative or complementary tool in the
treatment or prevention of Tam-resistant cancer.

1. Introduction signaling pathway, Rac1-PAK1 pathway [7], EGFR/HER2 regulatory


loops [8], and PI3K-AKT-mTOR pathway [9] have been identified as
Drug resistance recurrence is a core clinical challenge in cancer playing key roles in Tam resistance, the alteration of which led to in-
treatment, because in the case of breast cancer, approximately a quarter creased migration and proliferation rates, hormone independent phe-
of cancer patients treated with tamoxifen for five years acquired re- notypes, and cell autophagy.
currence, limiting its capability as a cancer treatment [1]. A few me- A few studies have suggested experimental approaches to over-
chanisms have been explicated for the recurrence of tamoxifen re- coming TamR, including targeting ER with chemical compounds [10],
sistance (TamR), including the alteration of specific gene expressions modulating the components of the ER signaling pathway, or controlling
and signaling pathways. For example, the overexpression of transcrip- the components of other signaling pathways that crosstalk with the ER
tion factor YBX1 in sensitive cells conferred resistance against Tam, pathway [11]. Nonetheless, in the efforts to overcome TamR, the
which was associated with the accelerated degradation of estrogen re- identified molecular mechanisms are limited and the prognosis for
ceptor (ER) [2]. In another study, the Tam-specific activation of Oct-4 cancer patients remains poor.
recruited ER onto its preferential genomic binding sites, eventually Cold atmospheric plasma (CAP) is a specific type of partially ionized
enhancing tumor growth [3]. In addition, factors that interact with ER gas defined as a non-thermal non-equilibrium plasma with complex
such as FOXA1 [4] and constituents of ER transcriptome such as IL-8, chemical composition such as NO-, NO2-, OH-, and O- [12]. By virtue of
NRF-1, and HOTAIR contribute to resistance [5,6]. As instances of the its efficacy at inducing cell death in multiple cancer cell lines including


Corresponding author.
E-mail address: sunjungk@dongguk.edu (S.J. Kim).
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.freeradbiomed.2017.06.017
Received 17 March 2017; Received in revised form 9 June 2017; Accepted 26 June 2017
Available online 27 June 2017
0891-5849/ © 2017 Elsevier Inc. All rights reserved.
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 1. CAP increases ROS in the MCF-7/TamR cell.


Fluorescence microscopic images were taken for the
MCF-7 and MCF-7/TamR cells using a fluorochrome
DCF-DA after treating the cells with CAP for 0 s or 10
times for 30 s. NAC was treated to scavenge the in-
tracellular ROS. Black and white-colored images are
from bright-field microscopy. (B) The bar graph
shows the quantification of fluorescence intensity
(mean ± SE of three replicates).

breast, cervical, glioblastoma, melanoma, and lung cancers, and in inflammation-related genes in keratinocytes [17], epigenetic changes in
xenograft model animals, CAP has received attention for its clinical breast cancer cells [18], and the enhanced expression of cell cycle–re-
application as an alternative cancer treatment option [13]. One re- lated γH2A.X [19].
markable advantage of CAP resides in its preference for acting on In this study, we hypothesized that CAP can reverse the drug-re-
cancer cells rather than normal cells. Cancer cells are known to have a sistance status of cancer cells provoked by continued exposure to Tam.
higher ROS level than normal cells, and CAP accumulates more ROS, This was explored by developing MCF-7/TamR breast cancer cells and
eventually inducing the specific death of cancer cells [14]. Recently, the then illuminating the cells with CAP. The appropriate recovery of
hampering effect of cholesterol on the permeation of ROS through the sensitivity against Tam was observed in the CAP-treated cells with cell
phospholipids bilayer was suggested to explain the increased ingress of death. The molecular mechanism of sensitivity recovery was in-
ROS in cancer cells, due to the decreased cholesterol fraction in their vestigated, revealing that many of the genes that had been dysregulated
cell membranes [15]. One of the major phenomena caused by the sig- by Tam had been set back to their original sensitive status. The current
nificant rise of intracellular ROS is the intense DNA double-strand study is expected to provide insight into how to overcome the drug
break, which results in the mitochondrial apoptosis pathway [14]. resistance of cancer cells using CAP.
Several of CAP's alterations to the gene expression and signaling
pathways other than the apoptotic pathway have been identified: EGFR
dysfunction in oral squamous cell carcinoma [16], the induction of

281
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 2. CAP inhibits the proliferation of MCF-7/TamR cells. (A) The reduced tumorigenicity of CAP-treated MCF-7/TamR. The effect of CAP on the tumorigenicity of MCF-7/TamR was
examined by colony formation assay. CAP was treated 10 times for 30 s each, and the colonies were counted using ImageJ after culturing for 14 days. (B) Suppressed proliferation of CAP-
treated MCF-7/TamR. The effect of CAP on proliferation was analyzed by crystal violet dye–based assay. CAP was treated for 600 s or 10 times for 30 s each, and the proliferation was
chased for six days. All assays were performed in triplicate, and the results are depicted as mean ± SE. (C) Increased apoptosis of CAP-treated MCF-7/TamR. The effect of CAP on the
apoptosis of MCF-7/TamR cells was analyzed on a flow cytometer after treating cells 10 times with CAP for 30 s each. All assays were performed in triplicate, and the results are depicted
as mean ± SE.

282
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 3. CAP recovers the drug sensitivity of the Tamoxifen-resistant MCF-7. (A) Expression of TamR marker genes in the MCF-7/TamR. The acquisition of resistance and recovery of
sensitivity against Tam was examined by the RT-PCR of previously known marker genes. The effect of CAP on the sensitivity of MCF-7 and MCF-7/TamR against Tam was examined by
colony formation (B) and dye-based proliferation assay (C). Cells were treated with CAP 10 times for 30 s each, followed by treatment with 0.1 and 0.5 μM of Tam. The number of colonies
was counted using ImageJ and indicated as a bar graph. All assays were performed in triplicate, and the results are depicted as mean ± SE.

2. Materials and methods 1640 medium supplemented with 10% FBS and 1% penicillin/strepto-
mycin under humidified 5% CO2 condition. The MCF-7/TamR cell line
2.1. Cell culture and treatment with CAP was generated by sequential exposure to increasing doses of 4-hydro-
xytamoxifen (Sigma-Aldrich, St. Louis, MO, USA) up to 0.1 µM. A mesh-
Breast cancer cell line MCF-7 was purchased from American Type type Dielectric Barrier Discharge (DBD) CAP device was manufactured
Culture Collection (ATCC; Manassas, VA, USA), and cultured in RPMI- at the Plasma Bioscience Research Center (Kwangwoon University,

283
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Table 1
Genes of which expression was restored to the level of MCF-7 after CAP treatment in MCF7/TamR.

Gene symbol Accession No. Description Fold change (MCF-7 vs. MCF- Fold change (MCF-7/TamR vs. MCF-7/
7/TamR) TamR + CAP 600 S)

FKBP14 NM_017946.2 FK506 Binding Protein 14 3.231 −1.657


ZNF483 NM_001007169.1 Zinc Finger Protein 483 2.766 −1.602
DEM1 NM_022774.1 Exonuclease 5, Defects In Morphology Protein 1 Homolog 2.506 −1.518
PLIN5 NM_001013706.2 Perilipin 5 2.442 −1.828
XRCC2 NM_005431.1 X-Ray Repair Complementing Defective Repair In Chinese 2.405 −1.534
Hamster Cells 2
DST NM_001723.4 Dystonin 2.401 −1.640
SRPK2 NM_182691.1 SRSF Protein Kinase 2 2.290 −1.554
SULT1A1 NM_177530.1 Sulfotransferase Family, Cytosolic, 1A, Phenol-Preferring, 2.056 −1.579
Member 1
LOC440353 NR_002603.1 Nuclear Pore Complex Interacting Protein Family, Member 2.017 −1.523
A1 Pseudogene
HOXC6 NM_004503.3 Homeobox C6 1.944 −1.533
TNFSF15 NM_005118.2 Tumor Necrosis Factor (Ligand) Superfamily, Member 15 1.863 −1.512
CDK5RAP3 NM_176095.1 CDK5 Regulatory Subunit Associated Protein 3 1.640 −1.712
FLAD1 NM_025207.3 Flavin Adenine Dinucleotide Synthetase 1 1.593 −1.643
SOX9 NM_000346.2 SRY (Sex Determining Region Y)-Box 9 −1.556 1.594
CPM NM_001005502.1 Carboxypeptidase M −2.475 1.516
IFI6 NM_022872.2 Interferon, Alpha-Inducible Protein 6 −4.692 1.615
IFI6 NM_022873.2 Interferon, Alpha-Inducible Protein 6 −4.692 1.526
MX1 NM_002462.2 MX Dynamin-Like GTPase 1 −5.731 1.598

Korea). Cells with 50% confluence in a 60 mm plate were irradiated 10 2.4. Tam sensitivity assay
times for 30 s each hour, with at a distance of 5 mm from the DBD disc
to the media surface. The discharged voltage was measured at 0.46 kV The sensitivity against Tam was evaluated by seeding 5×103 cells
when the argon gas flow rate was set to 1.5 L/min with 0.12 kV supply per 60 mm dish, or 2×103 cells per well of a 96-well plate and cul-
voltage. turing for 12 h. Then, transient transfection of siRNA or recombinant
plasmid vectors was conducted, followed by CAP treatment. The cells
were exposed to either 100 or 500 nM of 4-hydroxytamoxifen or
2.2. Cell transfection ethanol (Sigma-Aldrich) as a vehicle control. Cell viability was mea-
sured using CCK-8 reagent or colony counting through the aforemen-
siRNAs were synthesized by Bioneer (Korea), and transiently tioned procedures.
transfected into the MCF-7 and MCF-7/TamR cells at approximately
50% confluence and a final concentration of 30 nM using Lipofectamine 2.5. Genome-wide expression assay
RNAiMAX (Invitrogen, Carlsbad, CA, USA). cDNA was overexpressed by
transiently transfecting 1 µg of recombinant pEZ-M02 plasmid vector The total RNA was isolated from MCF-7 and MCF-7/TamR, and
(GeneCopoeia, Rockville, MD, USA) using Lipofectamine 3000 and CAP-treated MCF-7/TamR using the ZR-Duet DNA/RNA MiniPrep kit
PLUS reagents (Invitrogen), and the cells were then cultured for 24 h (Zymo Research, Irvine, CA, USA). For the experiment, 750 ng of
for further analysis. Information about the siRNAs and plasmid clones is Biotin-labeled cRNA was synthesized from the RNA and was hybridized
indicated in Supplementary Table S1. on the Illumina HumanHT-12 v4 Expression BeadChip (Illumina, San
Diego, CA, USA) by following the supplier's protocol. All arrays were
quantile normalized, and data processing was performed using Illumina
2.3. Cell proliferation and apoptosis analysis GenomeStudio v2011.1. All raw data were deposited in the Gene
Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) with series
The cell growth rate was measured once 1×103 cells were seeded in entry number GSE 95208.
each well of 96-well plates, incubated for 12 h, and then exposed to
CAP 10 times for 30 s each. After 24 h, Tam was added to the culture 2.6. Pathway and clustering analysis
media at a final concentration of 0, 0.1, and 0.5 µM. At day two, four,
and six after Tam treatment, 10 µl of CCK reagent from a cell counting Relevant networks were constructed through the use of Ingenuity
kit-8 assay kit (Dojindo Laboratories, Japan) was added to each well, Pathway Analysis (IPA, Qiagen, Redwood City, CA, USA) (http://www.
and absorbance at 450 nm was measured after 1 h using a microplate qiagen.com/ingenuity) using differentially expressed genes (|fold
reader. In the case of colony formation assay, 4×103 cells were seeded change| ≥ 1.5, P-value < 0.05) from the microarray analysis.
in a 60 mm culture dish and cultured for two weeks. The colonies were Clustering analysis was conducted using Cluster 3.0 software (http://
then fixed with acetic acid/methanol (1:7), stained with 0.5% crystal bonsai.hgc.jp/~mdehoon/software/cluster/) and visualized through a
violet (Sigma-Aldrich), and counted with Image J software (National TreeView program (http://jtreeview.sourceforge.net/) with genes dif-
Institutes of Health, Bethesda, MD, USA). Apoptosis analysis was car- ferentially expressed in both comparisons, MCF-7 vs. MCF-7/TamR and
ried out once cells had been harvested with trypsin, washed twice with MCF-7/TamR vs. CAP-treated MCF-7/TamR.
PBS, and then resuspended in 1x binding buffer from FITC Annexin V
Apoptosis Detection Kit I (BD Biosciences, Franklin Lakes, NJ, USA) at a 2.7. Real-time RT-PCR
concentration of 1×106 cells/ml. The cell suspension was incubated as
200 µl samples with 5 µl of Annexin V FITC for 15 min, and then with Total RNA was reverse transcribed using ReverTra Ace qPCR RT
2 µl of propidium iodide (PI) at room temperature in the dark for 3 min. Master Mix with gDNA remover (Toyobo, Japan) following the sup-
All samples were analyzed with a Becton Dickinson Accuri C6 flow plier's instruction. An ABI 7300 system (Applied Biosystems, Foster
cytometer (BD Biosciences) within 1 h. City, CA, USA) was utilized to perform quantitative real-time PCR using

284
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 4. Clustering of genes affected by Tam and CAP in MCF-7. (A) Genome-wide expression analysis identified 20 genes that commonly showed significant expression changes (> 1.5
fold) during the acquisition of Tam resistance and CAP treatment. The heatmap was constructed with genes from MCF-7/Tam vs. MCF-7 and CAP-treated MCF-7/Tam vs. MCF-7/Tam. (B)
The RT-PCR analysis of genes affected by Tam and CAP. RT-PCR was carried out for 10 genes that appeared in (A). Black bar, MCF-7 parent cell; red bar, Tam-resistant MCF-7; cyan bar,
CAP-treated MCF-7/Tam. All assays were performed in triplicate, and the results are depicted as mean ± SE.

SYBR FAST qPCR Kit (Kapa Biosystems, Wilmington, MA, USA) in tri- The blots were then incubated with anti-MX1 (1:500, Santa Cruz,
plicate. The expression of genes was normalized using GAPDH and Dallas, Texas, USA) and anti-HOXC6 (1:500, Novus, St. Charles, MO,
calculated according to 2−ΔΔCt method. The primers used for RT-PCR USA) antibodies, followed by incubation with HRP-conjugated goat
are detailed in Supplementary Table S2. anti-rabbit secondary antibodies (1:1000, Genetex, Pennsylvania, USA)
for 1 h at room temperature. Rabbit polyclonal beta actin antibody
(1:800, Novus) was used to normalize the quantification of target
2.8. Western blot analysis proteins. The bands were detected with West Save ECL reagents
(Abfrontier, Korea) and quantified with Image Lab software (Bio-Rad,
Total protein was collected using an ice-cold RIPA lysis buffer that Hercules, CA, USA).
contained a protease inhibitor cocktail (Thermo Fisher Scientific,
Waltham, MA, USA). After concentration quantification by BCA assay
(Thermo Fisher Scientific), 50 µg of protein from each sample was 2.9. ROS and NO detection assay
subjected to SDS-PAGE. The separated proteins were transferred to a
PVDF membrane (Whatman, UK), and blocked with 5% non-fat milk To detect ROS, CAP exposure was performed on 5×105 cells per
solution diluted in 0.1% Tween-20 TBS for 1 h at room temperature. well of 6-well plates 10 times for 30 s every hour, and these were

285
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 5. CAP regulates genes related with cell death and cancer.
The highest confidence IPA networks of genes displaying altered
expression as a consequence of CAP treatment of MCF-7/TamR.
The top networks were “Cell death and survival, Neurological
Disease, and Cancer.” Upregulated and downregulated genes are
respectively shaded in red and green, with the color intensity
indicating the magnitude of the expression change. Solid and
dashed lines respectively represent the direct and indirect inter-
actions.

treated with 20 µM of DCFH-DA (Sigma-Aldrich) for 30 min at 37 °C. obtained resistance against Tam. The detailed characteristics of the
Intracellular ROS was quantified by measuring the level of the fluor- developed MCF-7/TamR are beyond the scope of the current study and
escent DCF which was converted from the cell permeable DCFH-DA. To shall be described in a separate manuscript. When CAP was used to
detect NO, cells were loaded with 5 µM of DAF-FM diacetate illuminate the MCF-7/TamR cell, it appropriately increased the level of
(Invitrogen) and incubated for 20 min at 37 °C in the dark. Two mM of ROS (Fig. 1) and RNS (Supplementary Fig. S1) at a similar rate as the
N-Acetyl-L-Cysteine (NAC) (Sigma-Aldrich) and 2 mM of N(G)-Nitro-L- parental MCF-7, although MCF-7/Tam showed a higher level of ROS
Arginine Methyl Ester (L-NAME) (Sigma-Aldrich) were used to inhibit than MCF-7 before CAP treatment. The effect of CAP on the prolifera-
the generation of intracellular ROS and NO, respectively. After in- tion of MCF-7/TamR was chased by colony formation and dye-based
cubation for 6 h, the cells were exposed to CAP and the intracellular growth assay. The MCF-7 cell became more tumorigenic during the
ROS or NO levels were measured with aforementioned procedure. acquisition of drug resistance, as judged by the colony formation assay
Fluorescence was detected by a fluorescence microscope (Leica (Fig. 2A). CAP retarded the growth rate of MCF-7/TamR approximately
Microsystems, Germany) and quantified using an Infinite 200 Pro to the level of the parental MCF-7, as shown by the colony formation
fluorescence plate reader (Tecan, Switzerland). assay and proliferation assay (Fig. 2A and B). FACS analysis revealed an
increase in both early (32% increase) and late (33% increase) apoptosis
of MCF-7/TamR when CAP was treated (Fig. 2C).
2.10. Statistical analysis

Student's t-test was carried out to compare expressions between


3.2. CAP resensitized MCF-7/TamR cells to Tam
CAP-treated TamR and control cells using SPSS for Windows, release
17.0 (SPSS, Chicago, IL, USA). Experimental graphs were plotted with
During the acquisition of Tam resistance in cancer cells, specific
Microsoft Excel as mean ± standard error, and the expression differ-
metabolic and/or signaling pathways were known to be affected,
ences of selected genes among three groups, MCF-7, MCF-7/TamR, and
especially those of drug transportation and metabolism [20–22]. We
CAP-treated MCF-7/TamR, were determined by ANOVA comparisons. A
hypothesized that CAP could re-sensitize TamR cells to the drug at least
value of P < 0.05 was considered statistically significant.
in part by reversing the expression of affected genes. We first measured
the expression of four genes that had been previously identified as
3. Results being upregulated (BAG1, CD24) or downregulated (HDAC4, and
NGX6) during TamR acquisition to examine this, and the results con-
3.1. CAP inhibited the proliferation and promoted the apoptosis of MCF-7/ firmed the appropriate deregulation (Fig. 3A). All genes except NGX6
TamR showed expression recovery after CAP treatment, suggesting that CAP
could reverse the expression of TamR-related genes even though the
Interrogating the impact of cold atmospheric plasma on cancer cells recovery level was not the same as that in the parental MCF-7 (Fig. 3A).
for its potential application in the treatment of drug-resistant cancer Next, the MCF-7/TamR cell was examined for the restoration of Tam
required that we first developed a modified MCF-7 cell line that had sensitivity by CAP through colony formation assay. The results

286
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 6. CAP sets the expression of TamR-related MX1 and HOXC6


genes back to their MCF-7 status. The expression of MX1 (A) and
HOXC6 (B) in CAP-treated MCF-7/TamR cells was examined by
Western blot analysis. MX1 and HOXC6 were selected because
they respectively showed downregulation and upregulation in the
MCF-7/Tam compared to MCF-7. Expression was compared
among MCF-7, MCF-7/Tam, and CAP-treated MCF-7/Tam. All
assays were performed in triplicate and a representative image is
shown. Results are depicted as mean ± SE. (C) The effect of CAP
on the expression of MX1 and HOXC6 in the presence of NAC, an
ROS inhibitor, was measured by qPCR. The assay was performed
in triplicate and the result is depicted as mean ± SE.

indicated that the Tam-treated parental MCF-7 showed a sharp decrease compared groups. Notably, of the 20 genes, 18 showed the opposite
in colony size, while the MCF-7/TamR cells showed a much lesser de- expression pattern, i.e., they were once upregulated in the MCF-7/
crease (Fig. 3B). Notably, when CAP was illuminated to the MCF-7/ TamR, but the gene had been downregulated by CAP and vice versa
TamR, sensitivity against the drug appeared again approximately at the (Table 1 and Fig. 4A). Ten of the 18 genes were randomly selected, and
level of MCF-7. The effect of CAP on drug sensitivity was also mon- their expressions were examined by quantitative RT-PCR to further
itored using a dye-based cell proliferation assay. The result supported confirm their involvement in drug resistance and sensitivity. Thus, all
the data of the colony formation assay by showing a retarded growth genes recovered their expression with regard to MCF-7 when CAP was
rate for CAP-treated MCF-7/TamR compared to non-treated MCF-7/ treated to MCF-7/TamR (Fig. 4B). This fact strongly implies sensitivity
TamR (Fig. 3C). restoration in the MCF-7/TamR cells for the drug through CAP resetting
many of the genes that had undergone expression changes during their
3.3. MX1 and HOXC6 mediated the restoration of sensitivity against Tam resistance acquisition. Ingenuity pathway analysis was carried out to
identify the highest confidence network for the CAP-treated MCF-7/
The molecular mechanism of how CAP recovers sensitivity against TamR, and this resulted in the “Cell death and survival, Neurological
Tam was interrogated using genome-wide expression analysis, which disease and Cancer” pathway (Fig. 5).
was carried out for MCF-7/TamR and CAP-treated MCF-7/TamR cells. Interrogating the role of genes mediated by CAP activity to recover
In total, 322 genes with expression changes higher than 1.5-fold from drug sensitivity revealed two genes, MX1 and HOXC6, that respectively
MCF-7 vs. MCF-7/TamR and 39 genes from MCF-7/TamR vs. CAP- showed up- and down-regulation at the RNA level by CAP; these were
treated MCF-7/TamR were identified, with 20 genes hitting both selected and their protein levels were analyzed by Western blot. As

287
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

Fig. 7. Recovery of CAP-mediated Tam sensitivity is hampered by the dysregulation of MX1 and HOXC6 in the MCF-7/TamR cell. (A) Effect of the downregulation of MX1 on the recovery
of Tam sensitivity. MX1 was downregulated with siRNA in MCF-7/TamR cells, and the sensitivity against Tam was examined by colony formation assay after treating cells with CAP. The
colonies were counted using ImageJ and denoted with a bar graph. siNC, negative-control siRNA. All assays were performed in triplicate, and the results are depicted as mean ± SE. (B)
The effect of the upregulation of HOXC6 on the Tam-sensitivity recovery. HOXC6 was upregulated with a HOXC6 cDNA-containing recombinant plasmid by transiently transfecting the
MCF-7/TamR cell, and the sensitivity against Tam was examined.

shown in Fig. 6A, MX1 was decreased in the MCF-7/TamR compared to Supplementary Fig. S2B). The growth rate of MCF-7/TamR was re-
MCF-7, but increased after CAP treatment. In the case of HOXC6, the versed when ORF of MX1 and siRNA of HOXC6 were used
expression trend was opposite to that of MX1 (Fig. 6B). The regulation (Supplementary Figs. S2 and S3). These observations were also found
of the two genes by CAP was abrogated when NAC, an ROS inhibitor, with a similar pattern in the parental MCF-7 cell (Supplementary Figs.
was treated to the cell before CAP exposure judged by qRT-PCR, in- S4 and S5). To see whether MX1 or HOXC6 affects the ROS production,
dicating the role of ROS relaying the CAP activity (Fig. 6C). Next, the the genes were deregulated in the MCF-7/TamR cell, and ROS level was
effect of CAP on drug sensitivity was monitored in the genes’ dysre- measured after CAP treatment. This resulted in no significant change of
gulated conditions through colony formation assay. When MX1 was ROS level, suggesting that the genes were regulated downstream of the
induced to be downregulated by the siRNA (Supplementary Fig. S2A), ROS (Supplementary Fig. S6). Cumulatively, these results suggest that
MCF-7/TamR grew at a higher rate than in control siRNA-treated cells, MX1 and HOXC6 are strongly involved in mediating the effect of CAP at
implying a lower sensitivity against Tam (Fig. 7A). Meanwhile, higher least in part through ROS on recovering Tam sensitivity.
cell growth was observed when HOXC6 was induced to be upregulated
with an ORF-containing recombinant plasmid (Fig. 7B and

288
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

4. Discussion 5. Conclusion

This study aimed to examine CAP for its potential applications for CAP has successfully demonstrated its capacity to return the TamR
overcoming drug resistance in breast cancer. When CAP was applied to of MCF-7 breast cancer cells to the original drug-sensitive status by
Tam-resistant MCF-7 cells, it appropriately set the expression of many setting back the expression of cell death and survival pathway–related
genes back to the status of the original MCF-7 cells, which had been genes such as MX1 and HOXC6. The involvement of MX1 and HOXC6 in
dysregulated by Tam. A previous study reported the restoration of re- the recovery of drug sensitivity was proven by showing the deteriora-
sponsiveness in temozolomide-resistant glioma cells against the drug by tion of the recovery effect upon dysregulation. The identified genes
CAP [23]. However, the molecular mechanism of how CAP relieves could be developed as diagnostic markers, and could be molecular
drug-resistance is relatively unexplored. targets for the clinical treatment of Tam-resistant breast cancer.
It is speculated that two main streams of CAP activities exist that Furthermore, the successful restoration of sensitivity against Tam pro-
induce cell death of TamR cells. First, CAP might induce cell death in vides insights into the potential use of CAP for the treatment of TamR
TamR cells via the typical apoptotic pathway. In detail, most apoptosis cancer.
pathways observed in CAP-treated cancer cells are based on the mi-
tochondrial pathway triggered by DNA damage and mitochondrial da- Conflict of interest
mage [12,24]. Accordingly, a double-strand break marker, γ-H2AX, has
been commonly observed after CAP treatment [25]. The tumor necrosis The authors declare no conflict of interest.
factor receptor (TNFR)-based apoptosis pathway can also be activated
by the rise of intracellular ROS in a few cancers such as melanoma [26] Acknowledgments
and head and neck cancer cells [27]. In the current study, this was
supported by the results that the TamR cells showed a similar apoptotic This work was supported by the Science Research Center Program
rate as the parental cells, and that many apoptosis-related genes were (NRF-2010–0027963) and the Basic Science Research Program (NRF-
dysregulated by CAP. In this sense, CAP seems to have a similar effect 2016R1D1A1B01009235) of the National Research Foundation of
on proliferation inhibition, regardless of the drug sensitivity status. Korea funded by the Ministry of Education, Science, and Technology.
TamR cells showed more cancerous and stem-cell like characteristics
[28]; these alterations may increase the ROS level and decrease the Appendix A. Supplementary material
amount of membrane cholesterol, possibly making TamR cells more
vulnerable to CAP. Supplementary data associated with this article can be found in the
Second, CAP might have notably contributed to the proliferation online version at http://dx.doi.org/10.1016/j.freeradbiomed.2017.06.
inhibition of TamR cells by restoring the expression of genes that were 017.
involved in cancer cell proliferation, such as MX1 and HOXC6, which
had been deregulated during the acquisition of drug resistance. MX1 is References
a dynamin family GTPase that strongly interacts with tubulin, sug-
gesting its involvement in mitosis. The protein is known to have tumor- [1] C. Davies, H. Pan, J. Godwin, R. Gray, R. Arriagada, V. Raina, M. Abraham,
suppressive activity for several cancers, and the loss of its expression V.H. Medeiros Alencar, A. Badran, X. Bonfill, J. Bradbury, M. Clarke, R. Collins,
S.R. Davis, A. Delmestri, J.F. Forbes, P. Haddad, M.F. Hou, M. Inbar, H. Khaled,
results in increased metastasis and decreased sensitivity to Docetaxel J. Kielanowska, W.H. Kwan, B.S. Mathew, I. Mittra, B. Muller, A. Nicolucci,
[29]. HOXC6 is a homeobox-containing gene associated with mammary O. Peralta, F. Pernas, L. Petruzelka, T. Pienkowski, R. Radhika, B. Rajan,
gland development and is overexpressed in a variety of cancers in- M.T. Rubach, S. Tort, G. Urrutia, M. Valentini, Y. Wang, R. Peto, G. Adjuvant
Tamoxifen: Longer Against Shorter Collaborative, Long-term effects of continuing
cluding breast and prostate cancers [30]. HOXC6 expression was adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oes-
strongly detected in multidrug-resistance (MDR) cancer [31]. An siRNA trogen receptor-positive breast cancer: atlas, a randomised trial, Lancet 381 (2013)
of HOXC6 significantly reduced the intracellular level of MDR-1, and 805–816.
[2] T. Shibata, K. Watari, H. Izumi, A. Kawahara, S. Hattori, C. Fukumitsu,
inhibited MDR cancer cells in a xenograft tumor. Y. Murakami, R. Takahashi, U. Toh, K.I. Ito, S. Ohdo, M. Tanaka, M. Kage,
A remarkable observation during the acquisition of TamR in cancer M. Kuwano, M. Ono, Breast cancer resistance to antiestrogens is enhanced by in-
cell is the function of estrogen receptors and crosstalk with growth creased ER degradation and ERBB2 expression, Cancer Res. 77 (2017) 545–556.
[3] S. Bhatt, J.D. Stender, S. Joshi, G. Wu, B.S. Katzenellenbogen, OCT-4: a novel es-
factor–receptor pathways [32]. A few genes in our genome-wide ana-
trogen receptor-alpha collaborator that promotes tamoxifen resistance in breast
lysis such as FKBP14, FGF21, and TNFSF15 in those pathways re- cancer cells, Oncogene 35 (2016) 5722–5734.
presented a significant expression change. Furthermore, drug re- [4] X. Fu, R. Jeselsohn, R. Pereira, E.F. Hollingsworth, C.J. Creighton, F. Li, M. Shea,
sistance–related genes such as SULT1A1, XRCC, and SOX9 were found A. Nardone, C. De Angelis, L.M. Heiser, P. Anur, N. Wang, C.S. Grasso,
P.T. Spellman, O.L. Griffith, A. Tsimelzon, C. Gutierrez, S. Huang, D.P. Edwards,
in the network and in the list of deregulated genes, where their ex- M.V. Trivedi, M.F. Rimawi, D. Lopez-Terrada, S.G. Hilsenbeck, J.W. Gray,
pression had changed to the opposite of TamR. SULT1A1 is a phase II M. Brown, C.K. Osborne, R. Schiff, FOXA1 overexpression mediates endocrine re-
enzyme responsible for the sulfation of 4-hydroxytamoxifen in breast sistance by altering the ER transcriptome and IL-8 expression in ER-positive breast
cancer, Proc. Natl. Acad. Sci. USA 113 (2016) E6600–E6609.
tumors [33]. XRCC2- [34] and SOX9-deficient [35] cells were found to [5] B.N. Radde, M.M. Ivanova, H.X. Mai, N. Alizadeh-Rad, K. Piell, P. Van Hoose,
be hypersensitive to the alkylating anticancer drug, temozolomide. The M.P. Cole, P. Muluhngwi, T.S. Kalbfleisch, E.C. Rouchka, B.G. Hill, C.M. Klinge,
molecular mechanism of how these proteins are associated with re- Nuclear respiratory factor-1 and bioenergetics in tamoxifen-resistant breast cancer
cells, Exp. Cell Res. 347 (2016) 222–231.
sistance and sensitivity by Tam should be revealed in further studies. [6] X. Xue, Y.A. Yang, A. Zhang, K.W. Fong, J. Kim, B. Song, S. Li, J.C. Zhao, J. Yu,
It should be mentioned that only 20 genes appeared in common LncRNA HOTAIR enhances ER signaling and confers tamoxifen resistance in breast
between MCF-7/TamR and TamR/CAP, while 39 genes underwent cancer, Oncogene 35 (2016) 2746–2755.
[7] N. Gonzalez, G.A. Cardama, M.J. Comin, V.I. Segatori, M. Pifano, D.F. Alonso,
significant expression changes in TamR/CAP. Therefore, other path- D.E. Gomez, P.L. Menna, Pharmacological inhibition of Rac1-PAK1 axis restores
ways should be also responsible for the restoration of drug sensitivity, tamoxifen sensitivity in human resistant breast cancer cells, Cell. Signal. 30 (2017)
which have yet to be elucidated. Another limitation of this study is the 154–161.
[8] L. Yin, Z.Y. Wang, Roles of the ER-alpha36-EGFR/HER2 positive regulatory loops in
lack of various strength conditions in the CAP treatment, because dif-
tamoxifen resistance, Steroids 111 (2016) 95–99.
ferent cellular response, perhaps even opposite cellular phenomena, [9] X. Yu, R. Li, W. Shi, T. Jiang, Y. Wang, C. Li, X. Qu, Silencing of MicroRNA-21
would be expected for different strengths of CAP [36]. A precise stan- confers the sensitivity to tamoxifen and fulvestrant by enhancing autophagic cell
dardization of the CAP condition would help establish appropriate death through inhibition of the PI3K-AKT-mTOR pathway in breast cancer cells,
Biomed. Pharmacother. 77 (2016) 37–44.
option for the treatment of drug-resistant cancer cells. [10] Q. Jiang, S. Zheng, G. Wang, Development of new estrogen receptor-targeting

289
S. Lee et al. Free Radical Biology and Medicine 110 (2017) 280–290

therapeutic agents for tamoxifen-resistant breast cancer, Future Med. Chem. 5 [25] S. Kalghatgi, C.M. Kelly, E. Cerchar, B. Torabi, O. Alekseev, A. Fridman,
(2013) 1023–1035. G. Friedman, J. Azizkhan-Clifford, Effects of non-thermal plasma on mammalian
[11] M. Droog, K. Beelen, S. Linn, W. Zwart, Tamoxifen resistance: from bench to bed- cells, PLoS One 6 (2011) e16270.
side, Eur. J. Pharmacol. 717 (2013) 47–57. [26] M. Ishaq, S. Kumar, H. Varinli, Z.J. Han, A.E. Rider, M.D. Evans, A.B. Murphy,
[12] T. Adachi, H. Tanaka, S. Nonomura, H. Hara, S. Kondo, M. Hori, Plasma-activated K. Ostrikov, Atmospheric gas plasma-induced ROS production activates TNF-ASK1
medium induces A549 cell injury via a spiral apoptotic cascade involving the mi- pathway for the induction of melanoma cancer cell apoptosis, Mol. Biol. Cell. 25
tochondrial-nuclear network, Free Radic. Biol. Med. 79 (2015) 28–44. (2014) 1523–1531.
[13] P. Babington, K. Rajjoub, J. Canady, A. Siu, M. Keidar, J.H. Sherman, Use of cold [27] S.U. Kang, J.H. Cho, J.W. Chang, Y.S. Shin, K.I. Kim, J.K. Park, S.S. Yang, J.S. Lee,
atmospheric plasma in the treatment of cancer, Biointerphases 10 (2015) 029403. E. Moon, K. Lee, C.H. Kim, Nonthermal plasma induces head and neck cancer cell
[14] S.J. Kim, T.H. Chung, Cold atmospheric plasma jet-generated RONS and their se- death: the potential involvement of mitogen-activated protein kinase-dependent
lective effects on normal and carcinoma cells, Sci. Rep. 6 (2016) 20332. mitochondrial reactive oxygen species, Cell Death Dis. 5 (2014) e1056.
[15] J. Van der Paal, C. Verheyen, E.C. Neyts, A. Bogaerts, Hampering effect of choles- [28] H. Liu, H.W. Zhang, X.F. Sun, X.H. Guo, Y.N. He, S.D. Cui, Q.X. Fan, Tamoxifen-
terol on the permeation of reactive oxygen species through phospholipids bilayer: resistant breast cancer cells possess cancer stem-like cell properties, Chin. Med. J.
possible explanation for plasma cancer selectivity, Sci. Rep. 7 (2017) 39526. (Engl.). 126 (2013) 3030–3034.
[16] J.H. Lee, J.Y. Om, Y.H. Kim, K.M. Kim, E.H. Choi, K.N. Kim, Selective killing effects [29] S.G. Brown, A.E. Knowell, A. Hunt, D. Patel, S. Bhosle, J. Chaudhary, Interferon
of cold atmospheric pressure plasma with NO induced dysfunction of epidermal inducible antiviral MxA is inversely associated with prostate cancer and regulates
growth factor receptor in oral squamous cell carcinoma, PLoS One 11 (2016) cell cycle, invasion and Docetaxel induced apoptosis, Prostate 75 (2015) 266–279.
e0150279. [30] I. Hussain, A. Bhan, K.I. Ansari, P. Deb, S.A. Bobzean, L.I. Perrotti, S.S. Mandal,
[17] S. Arndt, M. Landthaler, J.L. Zimmermann, P. Unger, E. Wacker, T. Shimizu, Y.F. Li, Bisphenol-A induces expression of HOXC6, an estrogen-regulated homeobox-con-
G.E. Morfill, A.K. Bosserhoff, S. Karrer, Effects of cold atmospheric plasma (CAP) on taining gene associated with breast cancer, Biochim. Biophys. Acta 1849 (2015)
ss-defensins, inflammatory cytokines, and apoptosis-related molecules in keratino- 697–708.
cytes in vitro and in vivo, PLoS One 10 (2015) e0120041. [31] S.A. Kim, M.R. Lee, J.H. Yoon, S.G. Ahn, HOXC6 regulates the antitumor effects of
[18] S. Lee, H. Lee, H. Bae, E.H. Choi, S.J. Kim, Epigenetic silencing of miR-19a-3p by pheophorbide a-based photodynamic therapy in multidrug-resistant oral cancer
cold atmospheric plasma contributes to proliferation inhibition of the MCF-7 breast cells, Int. J. Oncol. 49 (2016) 2421–2430.
cancer cell, Sci. Rep. 6 (2016) 30005. [32] C.C. Benz, G.K. Scott, J.C. Sarup, R.M. Johnson, D. Tripathy, E. Coronado,
[19] O. Volotskova, T.S. Hawley, M.A. Stepp, M. Keidar, Targeting the cancer cell cycle H.M. Shepard, C.K. Osborne, Estrogen-dependent, tamoxifen-resistant tumorigenic
by cold atmospheric plasma, Sci. Rep. 2 (2012) 636. growth of MCF-7 cells transfected with HER2/neu, Breast Cancer Res. Treat. 24
[20] B.C. Baguley, Multiple drug resistance mechanisms in cancer, Mol. Biotechnol. 46 (1992) 85–95.
(2010) 308–316. [33] A.A. Adjei, R.M. Weinshilboum, Catecholestrogen sulfation: possible role in carci-
[21] H. Zahreddine, K.L. Borden, Mechanisms and insights into drug resistance in cancer, nogenesis, Biochem. Biophys. Res. Commun. 292 (2002) 402–408.
Front. Pharmacol. 4 (2013) 28. [34] R. Tsaryk, K. Fabian, J. Thacker, B. Kaina, Xrcc2 deficiency sensitizes cells to
[22] A. Ring, M. Dowsett, Mechanisms of tamoxifen resistance, Endocr. Relat. Cancer 11 apoptosis by MNNG and the alkylating anticancer drugs temozolomide, fotemustine
(2004) 643–658. and mafosfamide, Cancer Lett. 239 (2006) 305–313.
[23] J. Koritzer, V. Boxhammer, A. Schafer, T. Shimizu, T.G. Klampfl, Y.F. Li, C. Welz, [35] X.L. Li, X.Q. Chen, M.N. Zhang, N. Chen, L. Nie, M. Xu, J. Gong, P.F. Shen, Z.Z. Su,
S. Schwenk-Zieger, G.E. Morfill, J.L. Zimmermann, J. Schlegel, Restoration of X. Weng, J.Y. Tan, T. Zhao, H. Zeng, Q. Zhou, SOX9 was involved in TKIs resistance
sensitivity in chemo-resistant glioma cells by cold atmospheric plasma, PLoS One 8 in renal cell carcinoma via Raf/MEK/ERK signaling pathway, Int. J. Clin. Exp.
(2013) e64498. Pathol. 8 (2015) 3871–3881.
[24] N. Kaushik, N. Uddin, G.B. Sim, Y.J. Hong, K.Y. Baik, C.H. Kim, S.J. Lee, [36] D. Xu, B. Wang, Y. Xu, Z. Chen, Q. Cui, Y. Yang, H. Chen, M.G. Kong, Intracellular
N.K. Kaushik, E.H. Choi, Responses of solid tumor cells in DMEM to reactive oxygen ROS mediates gas plasma-facilitated cellular transfection in 2D and 3D cultures, Sci.
species generated by non-thermal plasma and chemically induced ROS systems, Sci. Rep. 6 (2016) 27872.
Rep. 5 (2015) 8587.

290

You might also like