Open Access

Isobaric tags for relative and absolute quantitation‑based proteomics analysis of the effect of ginger oil on bisphenol A‑induced breast cancer cell proliferation

  • Authors:
    • Dan Lei
    • Tao Hong
    • Longxue Li
    • Lai Chen
    • Xiaoquan Luo
    • Qinghua Wu
    • Zhiyong Liu
  • View Affiliations

  • Published online on: December 8, 2020     https://doi.org/10.3892/ol.2020.12362
  • Article Number: 101
  • Copyright: © Lei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Several chemicals in the environment, particularly those with estrogenic activity and small amounts (micromolar or lower) of environmental estrogen can cause changes in cell function and interfere with endocrine functions of animals and humans. These compounds enter the human body and increase the load of estrogen in the body, leading to an increasing incidence of estrogen‑related tumors in breast cancer, ovarian cancer and endometrial cancer. Previous studies have demonstrated that ginger can inhibit the expression of estrogen receptors, while the bioactive ingredients of ginger sig‑nificantly inhibit proliferation and promote the apoptosis of breast cancer cells. In the present study, a quantitative proteomics technique based on relative and absolute quanti‑tative isobaric labeling was used to determine the effect of ginger essential oil (GEO) and BPA combined treatment on the proteomic characteristics of MCF‑7 cells. In total, 5,084 proteins were detected. Proteins that were upregulated >1.2‑fold and downregu‑lated by >0.8‑fold were differentially expressed. Overall, 528 differentially expressed proteins were identified. Compared with the control group, MCF‑7 cells treated with GEO, BPA and GEO‑BPA resulted in 45 (14 up‑ and 31 downregulated), 481 (141 up‑ and 340 downregulated) and 34 (13 up‑ and 21 downregulated) differentially ex‑pressed proteins, respectively. Compared with the BPA group, MCF- 7 cells treated with GEO-BPA resulted in 210 (117 up- and 93 downregulated ) differentially expressed proteins, among the 210 differentially expressed proteins in the GEO-BPA group, 10 proteins were associated with oxidative phosphorylation pathways, while succinate dehydrogenase (ubiquinone) iron‑sulfur subunit (SDHB), succinate dehydrogenase cytochrome b560 subunit, mitochondrial (SDHC), cytochrome c oxidase subunit 2 and superoxide dismutase (Mn), mitochondrial (SOD2) expression was decreased with GEO‑BPA combined treatment. Through the analysis of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, the cellular localization, func‑tional annotation and biological pathways of differentially expressed proteins were ex‑amined. The results indicated that GEO‑BPA may act through the oxidative phosphory‑lation pathway, decreased the expression of SDHB and SDHC, affected the tricarbox‑ylic acid cycle and decreased the expression of SOD2. This may have led to oxidative stress and the death of breast cancer cells, and the SDH signaling pathway may be an important mediator of the inhibitory effects of GEO in MCF‑7 breast cancer cells. GEO can inhibit the proliferation of breast cancer MCF‑7 cells induced by BPA, and the underlying mechanism may be associated with oxidative phosphorylation. These results may aid the development of future treatment strategies for breast cancer caused by environmental estrogen exposure.

Introduction

Breast cancer is one of the most common malignant tumors among females. According to statistics in 2018, ~9.57 million individuals died of cancer worldwide, of which ~627,000 died from breast cancer (1). However, the current prevention and treatment methods need to be improved. Clinical, epidemiological and biological studies have indicated that exogenous estrogen is associated with the occurrence and development of breast cancer (2). Bisphenol A (BPA) is an exogenous estrogen, which is one of the most widely used industrial compounds in human daily life. BPA is similar to estrogen in structure and has stable chemical properties. It is widely used in plastic products, food containers, beverage bottles and dental fillings materials (3). When heated in mi-crowave ovens, plastic residues, such as bisphenol A may penetrate into the food (4). BPA is difficult to metabolise and excrete, thus interfering with the endocrine system of the human body (5). In previous years, a large number of experiments have confirmed that BPA can interfere with the normal functions of the human reproductive, nervous and immune systems and embryonic development. In addition, BPA promotes the occurrence and development of various tumors, such as breast and prostate cancer and children's reproductive system tumors (6,7). Liu et al (8) reported that bisphenol can regulate the expression of EMT-related protein markers by promoting the expression of Snail, thereby enhancing the migration ability of breast cancer MCF-7 cells.

Ginger (Zingiber officinale) is the fresh rhizome of ginger, a perennial herb of the Zingiberaceae family. Ginger is a traditional Chinese medicine that is used for both food and medicine (9). Previous studies have reported that ginger serves an antitumor role in various types of malignant tumors (1017). For example, 6-gingerol, the main active component of ginger, can induce the apoptosis of gastric cancer cells via different mechanisms (11). (6)-gingerol can effectively inhibit colon tumor growth in nude mice (12) and (6)-paradol has antitumor-promoting properties (13). Ginger extract can inhibit growth and induce apoptosis in prostate cancer models in vivo and in vitro (14). Volatile ginger oil has strong cytotoxicity in cervical cancer cells (15) and treatment of gastroin-testinal tumors (16). Karki has demonstrated that ginger oil inhibits hallmarks [cyclin D1, cyclin dependent kinase (Cdk)-2, Cdk-4 and Bcl-2] of breast cancer cells (17). The purpose of the present study was to investigate the function of ginger essential oil (GEO) on breast cancer cells induced by bisphenol A, and provide an experimental and theoretical basis of potential therapeutic targets and the development of novel drugs.

MCF-7, a widely studied epithelial cancer cell line derived from breast adenocarcinoma, has characteristics of differentiated mammary epithelium (18). In the present study, relative and absolute quantitative isobaric labeling (iTRAQ) technology was for comprehensive proteomic analysis of MCF-7 cells, which were treated with single or a therapeutic trace estrogen combination (BSA). In addition, bioinformatics and function-al analysis, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), cluster analysis and protein-protein interaction (PPI) network analysis, were used. The present study may provide an experimental basis to improve our under-standing of the underlying molecular mechanisms of GEO-induced apoptosis in breast cancer cells.

Materials and methods

Cell culture

The human breast cancer cell line MCF-7 was obtained from Fuheng Bio-logical Company (https://www.fudancell.com). Cell culture was completed at the Laboratory Animal Science and Technology Center of Jiangxi University of Traditional Chinese Medicine. MCF-7 cells were cultured in MEM medium (Solibao, China, http://www.solarbio.com) with 10% fetal bovine serum (FBS; Serana Europe) and 1% penicillin-streptomycin (Solibao, China) and incubated in a humid CO2 incubator at 37°C. MCF-7 is an estrogen receptor breast cancer cell line (18).

Preparation of GEO

In total, 500 g fresh ginger was crushed, put into a 5,000-ml round-bottom flask, mixed with 3,000 ml distilled water, heated and refluxed in a heating hood for 6 h. The volatile oil was extracted using n-hexane and dried over anhydrous sodium sulfate. Weight the ginger oil and stored at 4°C. Additionally, 200 mg/l was determined as the optimal concentration of ginger essential oil to treat MCF-7 cells through cytotoxicity tests (19).

Cell treatment

BPA was purchased from Macklin Inc., and dissolved with DMSO (Solibao, China, http://www.solarbio.com) solution. MCF-7 cells were plated at 1×106 cells per well in a 6-well plate. Then MCF-7 cells were incubated for 24 h at 37°C to allow adhesion, and then the minimum Eagle's medium (MEM; Hyclone, Cytiva) was removed and new media with GEO (25, 50, 100, 150, 200 and 250 mg/l) or BPA (10-5, 10-6, 10-7, 10-8 and 10-9 mol/l) or GEO-BPA (GEO 200 mg/l, BPA 10-5, 10-6, 10-7, 10-8 and 10-9 mol/l) at the different concentrations were added, MCF-7 cells were again incubated at 37°C in a 5% CO2 incubator. After 48 h, the cells were collected for further analysis. All experiments were performed in triplicate.

Cell viability analysis

Cell viability was determined using an MTT assay. MCF-7 cells were inoculated in a 96-well plate with 1×104 cells per well. The cells were incubated at 37°C for 24 h to allow them to adhere to the bottom of the plate, then MEM was removed and new media of GEO (25, 50, 100, 150, 200 and 250 mg/l) or BPA (10-5, 10-6, 10-7, 10-8 and 10-9 mol/l) or GEO-BPA (GEO 200 mg/l, BPA 10-5, 10-6, 10-7, 10-8, and 10-9 mol/l) at the different concentrations were added. After reaching the treatment time (24, 48 or 72 h), 20 µl of 5 mg/ml MTT was added to each well. The cells were incubated in a 5% CO2 incubator at 37°C for 4 h. The medium was then removed and MTT was dissolved with 150 µl DMSO solvent per well. The cells were stirred in the dark for 15 min on a shaking table at 75 rpm, and then the absorbance was measured at 490 nm using spectrophotometry.

Protein extraction

An appropriate amount (10–15 µl) of cell sample was treated with liquid nitrogen, then protein lysate (8 M urea + 1% SDS, including protease inhibitor cocktail (Thermo Fisher Scientific, Inc.) at a ratio of 1:5 was added. Cells were centri-fuged at 16,000 × g at 4°C for 30 min to collect the supernatant and 5× volume of pre-cooled acetone was added and precipitated overnight at −20°C. The next day the supernatant was centrifuged at 12,000 × g at 4°C for 30 min and the precipitate was collected. The precipitate was washed with 90% pre-cooled acetone and air dried. The precipitate was then dissolved it with 200 µl of protein lysate (8M urea + 1% SDS, with cocktail) and finally centrifuged at 12,000 × g at 4°C for 30 min to collect the protein supernatant.

SDS-PAGE

The protein sample (20 µg) was mixed with a 5X loading buffer and boiled for 5 min. Then the SDS-PAGE was performed on a 12.5% (v/w) polyacrylamide gel for quantification. Biologically repeated in triplicate.

Reductive alkylation and enzymatic hydrolysis

Triethylammonium bicarbonate buffer (TEAB) (1 mol/l) was added to a 100-µg protein sample of different treatment groups to make a TEAB final concentration 100 mM in tubes. Then tris (2-carboxyethyl) phosphine (TCEP) was added to each tube to make a final TCEP concentration of 100 mM, and reacted at 37°C for 60 min. After, iodoacetamide was added to each tube to make a final concentration of 40 mM. The reaction solution was incubated at room temperature in the dark for 40 min, then pre-cooled acetone (acetone: sample v/v =6:1) was added to each tube. The solution was precipitated at −20°C for 4 h, then centrifuged at 10,000 × g for 20 min at 4°C. The precipitate was collected and fully dissolved with 100 µl of 100 mM TEAB. Trypsin was then added according to the enzyme: protein (m/m) = 1:50, and incubated at 37°C overnight.

iTRAQ labeling

The samples were digested with trypsin, the peptides were dried using a vacuum pump, and reconstituted with 0.5 M TEAB. According to the manufacturer's instructions, peptides (~100 µg) in each group were labeled using the iTRAQ Labeling kit (cat. no. 4390812; Shanghai AB SCIEX Analytical Instrument Trading Co.).

High pH ultra high-performance liquid chromatography (UPLC) first dimension separation

The peptide samples (100 mM/l) of MCF-7 cells in different treatment groups were reconstituted with UPLC loading buffer [2% acetonitrile (adjusted to pH 10.0 with ammonia)], and a high pH liquid phase separation was performed with a reverse phase ACQUITY UPLC BEH C18 column (1.7 µm, 2.1 mm X 150; Waters Coporation) using the Vanquish Flex Binary UHPLC system (Thermo Fisher Scientific, Inc.). Mobile phase A: 2% Acetonitrile (adjusted to pH 10.0 with ammonia), Mobile phase B: 80% Acetonitrile (adjusted to pH 10.0 with ammonia). The ultraviolet detection wavelength was 214 nm. The flow rate was 200 µl/min and the gradient was 47 min.

High-performance liquid chromatography-mass spectrometry (HPLC-MS)

Samples were analyzed using HPLC-MS in positive and negative ion mode, a reverse phase C18 column (75 µm × 25 cm) and an Easy-1200 chromatography instrument (both Thermo Fisher Scientific, Inc.). The MS instrument was Q_Exactive HF-X (Thermo Fisher Scientific, Inc.) and chromatographic separation time was 120 min. Mobile phase A: 2% Acetonitrile 0.1% formic acid and B: 80% acetonitrile 0.1% formic acid. The flow rate was 300 nl/min, and the MS scanning range (m/z) was 350-1,300. Nitrogen gas tem-perature was 35°C and nebulizer pressure was 5500 psi.

Proteomics data analysis

The original MS data were extracted from the original files. These files were submitted to the Proteome discoverer server (https://www.thermofisher.com/order/catalog/product/OPTON-30810#/OPTON-30810) and the NCBInr database (https://www.ncbi.nlm.nih.gov) was searched.

Bioinformatics analysis

For proteins obtained by MS, proteins that were upregulated by >1.2-fold, downregulated by >0.8-fold and P<0.05 were considered to be differentially expressed (20,21). Cluster version 3.0 was used for hierarchical cluster analysis, and the cellular components and molecules of the different proteins were analyzed by BLAST2GO version 2.5.0 (https://www.blast2go.com), KOBAS version 2.1.1 (http://kobas.cbi.pku.edu.cn) and Goa tools version 0.6.5 (https://www.ebi.ac.uk/GOA). The functional and biological processes were classified by clustering, and the differences in protein functions were discussed based on annotation information. The gene symbols obtained from the target protein sequence database were used to study the target protein and experimental evidence using Cytoscape version 3.7.2 and the STRING database (https://string-db.org/) for network analysis of protein-protein interactions (PPIs) (22).

Statistical analysis

For one-variable experiments, P-values were calculated using the paired Student's t-test, while P-values from two-variable experiments were calculated using the two-way ANOVA and Dunnett's multiple comparisons test. Data are expressed as mean ± standard deviation and mean ± SEs of three independent biological replicates. P<0.05 was considered to indicate a statistically significant difference.

Results

Cell viability

As shown in Fig. 1, when the treatment time was constant, the viability MCF-7 cells decreased with increasing GEO concentration. This was most notable when the treatment concentration was 250 mg/l at all treatment durations. When the GEO concentration was constant, the viability of MCF-7 cells decreased with the prolongation of the treatment time, and reached the lowest at 72 h. Compared with the control group, BPA improved cell viability and the cell viability reached its maximum when the BPA concentration was 10-7 mol/l. Meanwhile, following GEO-BPA treatment, the cell viability decreased compared with the BPA group. In short, GEO inhibited the proliferation of breast cancer MCF-7 cells, while BPA promoted this; however, GEO overcame BPA-induced promotion of proliferation.

LC-MS/MS

The present study used iTRAQ technology to analyze the proteomic characteristics of MCF-7 cells, which were treated with different concentrations of GEO or BPA alone or GEO mixed with BPA. The experimental procedure is shown in Fig. 2. Overall, a total of 5,084 proteins were detected (Table SI). The quality control of protein data showed that the molecular mass of the protein was in the range of 0–100 kDa (Fig. 3A), and the length of most peptides were between 7 and 17 amino acids (Fig. 3B), which is similar to the properties of known trypsin peptides (23).

Compared with the control group, MCF-7 cells treated with GEO, BPA and GEO-BPA showed 45 (14 up- and 31 downregulated) and 481 (141 up- and 340 downregulated) differentially expressed proteins. Compared with the BPA group, MCF-7 cells treated with GEO-BPA showed 210 (117 up- and 93 downregulated) differentially expressed proteins, respectively (Tables I and SIIIV). According to the optimal selection criterion for differentially expressed proteins, compared with cells treated with BPA alone, some proteins in cells treated with GEO and BPA were significantly upregulated or downregulated (P<0.05). According to the aforementioned criteria, a total of 34 differentially expressed proteins were further processed (Table II), of which 13 were significantly upregulated and 21 were significantly downregulated (P<0.05).

Table I.

Protein quantification in MCF-7 cells treated with GEO or BPA alone and GEO-BPA combined.

Table I.

Protein quantification in MCF-7 cells treated with GEO or BPA alone and GEO-BPA combined.

Comparison between groupsUpregulated proteins, nDownregulated pro-teins, nProtein count, n
GEO vs. C  14  31  45
BPA vs. C141340481
GEO-BPA vs. C  13  21  34
GEO-BPA vs. BPA11793210

[i] GEO, ginger essential oil; BPA, bisphenol A; C, control.

Clustering analysis

The results of hierarchical clustering are displayed in the form of a heat map, in which red indicates upregulation and blue indicates downregulation. The observed difference in protein expression between the groups are shown in Fig. 4. It was observed the overall expression pattern of genes in the GEO, BPA and GEO-BPA groups was different compared with those in the control group. In the control group, most genes showed a highly upregulated expression patterns (red bands), while in the GEO and GEO-BPA groups, most genes were downregulated.

GO function annotation and analysis

GO is a type of functional classification system. The GO database provides a standardized description of gene products from the per-spectives of function, participating in biological pathways and localization in cells. The GO database showed 53, 59 and 57 functional annotations for differentially expressed proteins in cells treated with GEO (n=45), BPA (n=481) and GEO-BPA (n=210) (Tables SVSVII). In addition, the GO functional annotations of differentially expressed proteins in each group were analyzed. The results showed that compared with the con-trol group, 45 differentially expressed proteins were more likely to be located at the cell parts and macromolecular complex in GEO alone group, and were closely associated with catalytic activity and protein binding activity. These proteins are involved in cellu-lar and metabolic processes and responses to stimuli (Fig. 5). Only 111 diffeentially expressed proteins in the BPA alone group were reported in the organelles and membrane, which were associated with structural and molecular activity. These proteins were involved in various biological processes, such as metabolic processes, organization of cellular component or biogenesis and localization (Fig. 6). The 192 differentially expressed proteins in the GEO-BPA combination group were primarily located in the cell part, single-organism process, organelle part and were involved in catalytic activity and structural molecule activity, which could affect signaling, cellular component organ-ization or biogenesis and negative regulation of biological processes (Fig. 7). Changes in biological processes indicated that GEO affects BPA-treated breast cancer cells

KEGG pathway analysis

The KEGG database can be used to associate the gene catalogue in the whole genomes with higher levels of system function at the cell, species and ecosystem levels (24). Through KEGG pathway analysis, the key signaling path-ways and related regulatory processes of each group were obtained (Tables SVIIISX). The KEGG secondary category of each group is presented in Figs. 810. KEGG analy-sis showed that the signaling pathways identified in the GEO group were primarily associated with energy metabolism, transportation and catabolism, immune system and cell motility (Fig. 8). KEGG signaling pathways in the BPA group were mainly associated with the oncogenes signaling pathway, cellular community-eukaryotes and the endocrine system (Fig. 9). The KEGG signaling pathway in the GEO-BPA combination group was associated with signal transduction, immune system, energy metabolism, cancer and endocrine and metabolic diseases (Fig. 10), and from the description of the pathway, it can be seen that the main pathways of these three groups are associated with oxidative phosphorylation, endocytosis, focal adhesions and ribosomes (Tables SVIIISX; Fig. 10).

PPI network analysis

Differentially expressed proteins between direct interaction mod-els may help in gaining important information about the target protein (25). Analysis of PPI network is shown in Fig. 11, the yellow represents the protein with highest degree value, while other colors represent other proteins that interact with these differentially expressed target proteins. The PPI analysis revealed the connection degree of the pro-tein interactions. A higher degree of connectivity may show more protein complexes. SNRPE, SDHB, SDHC, MRPL13, SOD2, MT-CYB, MT-CO2, HNRNPL, SFPQ and FGB showed a high degree of connectivity by comparing the differentially expressed proteins between GEO-BPA and BPA treatments and these targets marked as yellow. The middle-degree targets such as NDUFA 8, NDUFA 4, ND 2 and IGF 2 are marked in light green, and blue marked targets with low degree values.

Discussion

iTRAQ is one of the most advanced technologies in modern quantitative proteomics (26). It combines stable isotope labeling and tandem mass spectrometry, and can compare the relative content of protein in normal and diseased samples in one experiment (27). The present study systematically identified and analyzed the differences of proteome expression in breast cancer cells treated with GEO-BPA and BPA alone. The re-sults showed that GEO effectively inhibited the viability of breast cancer cells, which is in accordance with the findings of Karkihave, GEO inhibits CD44/ALDH1, the hall-marks of breast cancer cells (17), and BPA promoted the proliferation of MCF-7 cells, which is consistent with previous research (28). The GEO-BPA, BPA and GEO treat-ment groups showed 34, 481 and 210 differentially expressed proteins, respectively, compared with the control group. These differentially expressed proteins could be used as biomarkers to evaluate the effect of GEO on breast cancer induced by BPA, and to guide future treatment strategies for breast cancer.

Through GO annotation and KEGG pathway analysis, the specific regulation function and signal transduction pathways during GEO-BPA treatment were determined, which provided new insights into breast cancer development and proposed potential treatment strategies. GO functional annotations showed that, compared with the control group, 210 proteins were differentially expressed in the GEO-BPA combined treatment group. These differentially expressed proteins were mainly located in organelles and membranes, which can affect signal transduction, tissue or biogenesis of cellular com-ponents and negative regulation of biological processes. KEGG signal pathway analysis showed that oxidative phosphorylation, cAMP signaling pathway, focal adhesions and ribosomes in the GEO-BPA combination treatment group was richer compared with other pathways. The oxidative phosphorylation pathway contains the largest number of differential proteins, which is related to energy metabolism and is the key to regulating the content of ATP and oxygen in cells (29).

Tumor cells obtain energy through the glycolysis and oxidative phosphorylation pathways. Under normoxic conditions, normally differentiated cells obtain energy primarily through oxidative phosphorylation, which is generally ≥40% of the total ATP. However, cancer cells tend to rely on aerobic glycolysis (a phenomenon of converting glucose into lactic acid under aerobic conditions) to generate energy, and use the inter-mediate products of glycolysis as building blocks of cell proliferation (30). The dysregulation of mitochondrial metabolism is a characteristic of the metabolic reprogramming of cancer cells (31). However, aerobic glycolysis is not enough to completely replace the energy produced by oxidative phosphorylation (32). Even in an anoxic environment, continuous mitochondrial oxidative phosphorylation is still indispensable for the biological activities of tumor cells. In recent years, research on the antitumor effect of energy metabolism pathways has become increasingly popular (33,34). Sharma and Singh reported that dichloroacetate can stimulate oxidative phosphorylation by changing mitochondrial morphology for glycolysis, inducing apoptosis and inhibiting proliferation of breast cancer cells (33). Fan et al (34) reported that Jar-TA, a natural diterpenoid derivative, induces apoptosis of esophageal cancer cells by double inhibition of glycolysis/oxidative phosphorylation.

KEGG analysis showed that there were 10 differentially expressed proteins in the GEO-BPA combined treatment group, which regulated the oxidative phosphorylation pathway and possibly regulated intracellular energy metabolism and oxygen transport in breast cells. Meanwhile, the top five degrees of PPI network interaction results were SNRPE, SDHB, SDHC, MRPL13 and SOD2, among which SDHB, SDHC and SOD2 were associated with energy metabolism (35). The present study showed that down-regulation of the protein of SDHB, SDHC, SOD2 and COX-2 may be an important factor related to oxidative damage that promotes GEO to treat BPA-induced breast cancer cells. SDH is a key enzyme in the tricarboxylic acid cycle and oxidative phosphorylation of the respiratory chain. The lack of SDH inhibits the tricarboxylic acid cycle and the transmission of electrons in the respiratory chain, resulting in the production of ROS and oxidative damage to cells. Therefore, whether the functions of the SDHB and SDHC genes are normal or not will affect the function of SDH, which will eventually lead to excessive production of ROS in mitochondria and induce apoptosis (35). Functional studies of yeast models have shown that deletion of the SDHA or SDHB genes and point mutations of SDHB, SDHC and SDHD are associated with SDH dysfunction and increased production of ROS (36). In human cell lines, SDHB-silencing, drug inhibition or RNA interference also eliminates the activity of complex II, and increases production of ROS and nuclear stability of HIF1α under normoxic conditions (37). Compared with parent cells, SDHC mutations also increase the level of ROS production, genomic instability and tumorigenesis of mutant cells (38). The present study also demonstrated that the expression of SOD2 protein was downregulated. SOD2 is a type of superoxide dismutase, which catalyzes the conversion of superoxide to oxygen and hydrogen peroxide through a disproportionate reaction, thus removing superoxide and protecting cells from oxidative damage (39). SOD2-downregulation in the present study indicated that there may have been an oxidative stress reaction, releasing peroxide and inducing cell death.

The current results demonstrated that the expression of COX-2 protein was de-creased in the GEO-BPA treatment group, which is consistent with the results of previous studies. For example, Han et al showed that downregulation of COX-2 can make survivin and Bcl-2 mRNA and protein expression levels decrease, Bax mRNA and pro-tein expression levels increase, thus affect proliferation and apoptosis of human breast cancer MCF-7 cells (40). Meanwhile, malonate and 3-nitropropionic acid are com-pounds that specifically inhibit SDH activity and then induce ROS production and apoptosis (41). However, as anticancer drugs, both of these compounds have secondary toxicity (neurological diseases), so their value for in vivo treatment is limited (42). Therefore, the active ingredients of traditional Chinese medicine that are used for medi-cine and food are worthy of in-depth study. Besides, decreased expression of enzymes involved in oxidative phosphorylation, including NADH dehydrogenase (ubiquinone) 1 β subcomplex subunit 8 (NDUFA8), NADH dehydrogenase (ubiquinone) iron-sulfur protein 4 (NDUFS4) and NADH dehydrogenase 2 (ND2), may cause mitochondrial defects and affect the energy metabolism of breast cancer cells (43).

Overall, the iTRAQ-based proteomics data presented in the current study may be useful to clarify the proteomics profile of MCF-7 cells treated with GEO and BPA, and these data may help improve our understanding of the molecular mechanisms underlying the effects of these treatments. It was reported that, in MCF-7 cells treated with GEO-BPA, seven differentially expressed proteins were associated with oxidative phosphorylation (COX-2, NDUFA8, NDUFS4, ND2, SDHB, SDHC and SOD2) were significantly reduced (P<0.05). KEGG analysis highlighted that oxidative phos-phorylation may be one of the mechanisms by which GEO inhibited BPA-induced MCF-7 breast cancer cell proliferation. PPI analysis showed that SNRPE, SDHB, SDHC, MRPL13 and SOD2 had high connectivity and were in the center of the network. These differentially expressed proteins may be the key to the inhibitory effect of GEO on BPA-induced MCF-7 cells and showed that the molecular function about energy metabolism underlying this effect. The limitation of the present study lies in screening differentially expressed proteins through in vitro experiments. In future research, in vivo and in vitro experiments must be used to further analyze the role and mechanisms of ginger against breast cancer.

Supplementary Material

Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by The Double-First Class Discipline Construction Project (grant nos. JXSYLXK-ZHYAO115 and ZHYI025) and The Program of Health Commission Jiangxi Province (grant nos. 20195635 and 2019A262).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

DL, TH and ZL carried out the experiments and drafted the manuscript. DL and LL participated in the statistical analyses. LC, XL and QW conceived and designed the study and helped to draft the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

iTRAQ

isobaric tags for relative and absolute quantitation

GEO

ginger essential oil

BPA

bisphenol A

GEO-BPA

ginger essential oil and bisphenol A

COX2

cytochrome c oxidase subunit 2

TEAB

triethylammonium bicarbonate buffer

TCEP

tris (2-carboxyethyl) phosphine

LC

liquid chromatography

MS

mass spectrometry

DMSO

dimethylsulfoxide

TAM

tamoxifen

NDUFA8

NADH dehydrogenase (ubiquinone) 1 alpha subcomplex subunit 8

NDUFS4

NADH dehydrogenase (ubiquinone) iron-sulfur protein 4, mitochondrial

ND2

NADH-ubiquinone oxidoreductase chain 2

SDHB

succinate dehydrogenase (ubiquinone) iron-sulfur subunit, mitochondrial

SDHC

succinate dehydrogenase cytochrome b560 subunit, mitochondrial

SOD2

superoxide dismutase (Mn), mitochondrial

SNRPE

small nuclear ribonucleoprotein E

MRPL13

39S ribosomal protein L13, mitochondrial

ROS

39S ribosomal protein L13, mitochondrial

OXPHOS

oxidative phosphorylation

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Derouiche S, Warnier M, Mariot P, Gosset P, Mauroy B, Bonnal JL, Slomianny C, Delcourt P, Prevarskaya N and Roudbaraki M: Bisphenol A stimulates human prostate cancer cell migration via remodelling of calcium signalling. Springerplus. 2:542013. View Article : Google Scholar : PubMed/NCBI

3 

Kloukos D, Pandis N and Eliades T: In vivo bisphenol - a release from dental pit and fissure sealants: A systematic review. J Dent. 41:659–667. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Krishnan AV, Stathis P, Permuth SF, Tokes L and Feldman D: Bisphenol-A: An estrogenic substance is released from polycarbonate flasks during autoclaving. Endocrinology. 132:2279–2286. 1993. View Article : Google Scholar : PubMed/NCBI

5 

Huang YQ, Wong CK, Zheng JS, Bouwman H, Barra R, Wahlström B, Neretin L and Wong MH: Bisphenol A (BPA) in China: A review of sources, environmental levels, and potential human health impacts. Environ Int. 42:91–99. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Rogers JA, Metz L and Yong VW: Review: Endocrine disrupting chemicals and immune responses: a focus on bisphenol-A and its potential mechanisms. Mol Immunol. 53:421–430. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Vom Saal FS, Nagel SC, Coe BL, Angle BM and Taylor JA: The estrogenic endocrine disrupting chemical bisphenol A (BPA) and obesity. Mol Cell Endocrinol. 354:74–84. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Lui YZ, Zeng GQ, Ge LC, Liu H, Du J and Wang HS: Bisphenol A induces epithelial mesenchymalization of human breast cancer MCF-7 cells. Acta Scientiae Circumstantiae. 35:608–612. 2015.(In Chinese).

9 

White B: Ginger: An overview. Am Fam Physician. 75:1689–1691. 2007.PubMed/NCBI

10 

Ali BH, Blunden G, Tanira MO and Nemmar A: Some phytochemical, pharmacological and toxicological properties of ginger (Zingiber officinale Roscoe): A review of recent research. Food Chem Toxicol. 46:409–420. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Ishiguro K, Ando T, Maeda O, Ohmiya N, Niwa Y, Kadomatsu K and Goto H: Ginger ingredients reduce viability of gastric cancer cells via distinct mechanisms. Biochem Biophys Res Commun. 362:218–223. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Jeong C-H, Bode AM, Pugliese A, Cho YY, Kim HG, Shim JH, Jeon YJ, Li H, Jiang H and Dong Z: [6]-Gingerol suppresses colon cancer growth by targeting leukotriene A4 hydrolase. Cancer Res. 69:5584–5591. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Surh YJ, Park KK, Chun KS, Lee LJ, Lee E and Lee SS: Anti-tumor-promoting activities of selected pungent phenolic substances present in ginger. J Environ Pathol Toxicol Oncol. 18:131–139. 1999.PubMed/NCBI

14 

Brahmbhatt M, Gundala SR, Asif G, Shamsi SA and Aneja R: Ginger phytochemicals exhibit synergy to inhibit prostate cancer cell proliferation. Nutr Cancer. 65:263–272. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Santos PA SR, Avanço GB, Nerilo SB, Marcelino RIA, V. Janeiro V, Valadares MC and Machinski M: Assessment of Cytotoxic Activity of Rosemary (Rosmarinus officinalis L.), Turmeric (Curcuma longa L.), and Ginger (Zingiber officinale R.) Essential Oils in Cervical Cancer Cells (HeLa). ScientificWorldJournal. 2016:92730782016. View Article : Google Scholar : PubMed/NCBI

16 

Prasad S and Tyagi AK: Ginger and its constituents: role in prevention and treatment of gastrointestinal cancer. Gastroenterol Res Pract. 2015:1429792015. View Article : Google Scholar : PubMed/NCBI

17 

Karki N: Inhibitory activity of ginger oil against breast cancer cells (2011). LSU Master's Theses. 1714. simplehttps://digitalcommons.lsu.edu/gradschool_theses/1714

18 

Brooks SC, Locke ER and Soule HD: Estrogen receptor in a human cell line (MCF-7) from breast carcinoma. J Biol Chem. 248:6251–6253. 1973.PubMed/NCBI

19 

Ninth Chinese Pharmacopoeia Commission: Pharmacopoeia of the People's Republic of China (1). China Medical Science and Technology Press; China: pp. 932010

20 

Kan Y, Lyu Q, Jiang N, Han S, Li J, Burdman S and Luo L: iTRAQ-based proteomic analyses of the plant-pathogenic bacterium Acidovorax citrulli during entrance into and resuscitation from the viable but nonculturable state. J Proteomics. 211:1035472020. View Article : Google Scholar : PubMed/NCBI

21 

Gao Z, Wang J, Bai Y, Bao J and Dal E: Identification and Verification of the Main Differentially Expressed Proteins in Gastric Cancer via iTRAQ Combined with Liquid Chromatography-Mass Spectrometry. Anal Cell Pathol. 2019:53106842019. View Article : Google Scholar

22 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47D:D607–D613. 2019. View Article : Google Scholar

23 

Swaney DL, Wenger CD and Coon JJ: Value of using multiple proteases for large-scale mass spectrometry-based proteomics. J Proteome Res. 9:1323–1329. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Yu LR, Zeng R, Shao XX, Wang N, Xu YH and Xia QC: Identification of differentially expressed proteins between human hepatoma and normal liver cell lines by two-dimensional electrophoresis and liquid chromatography-ion trap mass spectrometry. Electrophoresis. 21:3058–3068. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Unwin RD, Griffiths JR and Whetton AD: Simultaneous analysis of relative protein expression levels across multiple samples using iTRAQ isobaric tags with 2D nano LC-MS/MS. Nat Protoc. 5:1574–1582. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Jamaluddin MFB, Nagendra PB, Nahar P, Oldmeadow C and Tanwar PS: Proteomic Analysis Identifies Tenascin-C Expression Is Upregulated in Uterine Fibroids. Reprod Sci. 26:476–486. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Feifei S and Yinyin W: Research status and progress of relationship between environmental bisphenol A and reproductive system malignant tumors. Health Res. 38:532–537. 2018.

29 

Lui Q, Hu LG, Zhou QF and Jiang GB: Tetrabromobisphenol A bis(2-hydroxyethylether) induced dysfunction of respiratory chain oxidative phosphorylation and energy metabolism rat pheochromocytoma cells by high performance liquid chromatography-electrospray ionization-tandem mass spectrometry. Chinese J Chromatography. 35:1–7. 2017. View Article : Google Scholar

30 

Li QX, Zhang P and Liu H: Characteristics and progress of energy metabolism of tumor cells. Chinese Pharmacological Bulletin. 33:1499–1502. 2017.

31 

Liberti MV and Locasale JW: The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem Sci. 41:2872016. View Article : Google Scholar : PubMed/NCBI

32 

Jose C, Bellance N and Rossignol R: Choosing between glycolysis and oxidative phosphorylation: A tumor's dilemma? Biochim Biophys Acta. 1807:552–561. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Sharma P and Singh S: Combinatorial Effect of DCA and Let-7a on Triple-Negative MDA-MB-231 Cells: A Metabolic Approach of Treatment. Integr Cancer Ther. 19:15347354209114372020. View Article : Google Scholar : PubMed/NCBI

34 

Fan XX, Su N, Huang XJ, Li YF, Jia A, Fan JP, Wang AF, Zhao NM and Ma YC: Jar-TTA, a natural diterpenoid derivatives, induces apoptosis in human esophageal cancer cells through dual inhibition of glycolysis and oxidative phosphorylation. Chinese Pharmacological Bulletin. 35:950–957. 2019.

35 

Guzy RD, Sharma B, Bell E, Chandel NS and Schumacker PT: Loss of the SdhB, but Not the SdhA, subunit of complex II triggers reactive oxygen species-dependent hypoxia-inducible factor activation and tumorigenesis. Mol Cell Biol. 28:718–731. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Goffrini P, Ercolino T, Panizza E, Giachè V, Cavone L, Chiarugi A, Dima V, Ferrero I and Mannelli M: Functional study in a yeast model of a novel succinate dehydrogenase subunit B gene germline missense mutation (C191Y) diagnosed in a patient af-fected by a glomus tumor. Hum Mol Genet. 18:1860–1868. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Guzy RD, Sharma B, Bell E, Chandel NS and Schumacker PT: Loss of the SdhB, but Not the SdhA, subunit of complex II triggers reactive oxygen species-dependent hypoxia-inducible factor activation and tumorigenesis. Mol Cell Biol. 28:718–731. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Slane BG, Aykin-Burns N, Smith BJ, Kalen AL, Goswami PC, Domann FE and Spitz DR: Mutation of succinate dehydrogenase subunit C results in increased O2.-, oxidative stress, and genomic instability. Cancer Res. 66:7615–7620. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Zelko IN, Mariani TJ and Folz RJ: Superoxide dismutase multigene family: A comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med. 33:337–349. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Han H, Yang S, Lin SG, Xu CS and Han ZH: Effects and mechanism of downregulation of COX 2 expression by RNA interference on proliferation and apoptosis of hu-man breast cancer MCF 7 cells. Mol Med Rep. 10:3092–3098. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Gomez-Lazaro M, Galindo MF, Melero-Fernandez de Mera RM, Fernandez-Gómez FJ, Concannon CG, Segura MF, Comella JX, Prehn JH and Jordan J: Reactive oxygen species and p38 mitogen-activated protein kinase activate Bax to induce mito-chondrial cytochrome c release and apoptosis in response to malonate. Mol Pharmacol. 71:736–743. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Liot G, Bossy B, Lubitz S, Kushnareva Y, Sejbuk N and Bossy-Wetzel E: Com plex II inhibition by 3-NP causes mitochondrial fragmentation and neuronal cell death via an NMDA- and ROS-dependent pathway. Cell Death Differ. 16:899–909. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Balsa E, Marco R, Perales-Clemente E, Szklarczyk R, Calvo E, Landázuri MO and Enríquez JA: NDUFA4 is a subunit of complex IV of the mammalian electron transport chain. Cell Metab. 16:378–386. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lei D, Hong T, Li L, Chen L, Luo X, Wu Q and Liu Z: Isobaric tags for relative and absolute quantitation‑based proteomics analysis of the effect of ginger oil on bisphenol A‑induced breast cancer cell proliferation. Oncol Lett 21: 101, 2021
APA
Lei, D., Hong, T., Li, L., Chen, L., Luo, X., Wu, Q., & Liu, Z. (2021). Isobaric tags for relative and absolute quantitation‑based proteomics analysis of the effect of ginger oil on bisphenol A‑induced breast cancer cell proliferation. Oncology Letters, 21, 101. https://doi.org/10.3892/ol.2020.12362
MLA
Lei, D., Hong, T., Li, L., Chen, L., Luo, X., Wu, Q., Liu, Z."Isobaric tags for relative and absolute quantitation‑based proteomics analysis of the effect of ginger oil on bisphenol A‑induced breast cancer cell proliferation". Oncology Letters 21.2 (2021): 101.
Chicago
Lei, D., Hong, T., Li, L., Chen, L., Luo, X., Wu, Q., Liu, Z."Isobaric tags for relative and absolute quantitation‑based proteomics analysis of the effect of ginger oil on bisphenol A‑induced breast cancer cell proliferation". Oncology Letters 21, no. 2 (2021): 101. https://doi.org/10.3892/ol.2020.12362