Basic Study Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Feb 21, 2017; 23(7): 1189-1202
Published online Feb 21, 2017. doi: 10.3748/wjg.v23.i7.1189
Dysregulation of mRNA profile in cisplatin-resistant gastric cancer cell line SGC7901
Xiao-Que Xie, Hua Wang, Kang-Sheng Gu, Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui Province, China
Qi-Hong Zhao, Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei 230032, Anhui Province, China
Author contributions: Xie XQ performed experiments, prepared the manuscript and analyzed data; Zhao QH performed select experiments; Zhao QH and Wang H read the manuscript and gave important intellectual suggestion; Wang H and Gu KS designed and supervised the project.
Supported by Projects of Foreign Science and Technology Cooperation of Anhui Province, No. 1604b0602027; New Century Excellent Talents in University, Ministry of Education of China, No. NCET-13-0644; and Wanjiang Scholars Program of Anhui Province of China.
Institutional review board statement: The study was reviewed and approved by the First Affiliated Hospital of Anhui Medical University Institutional Review Board.
Informed consent statement: All study participants, or their legal guardian, provided informed written consent prior to study enrollment.
Conflict-of-interest statement: The authors declared no conflict of interest.
Data sharing statement: Technical appendix and dataset available from the corresponding author at 13805692145@163.com. Participants gave informed consent for data sharing.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Kang-Sheng Gu, PhD, Professor, Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230032, Anhui Province, China. 13805692145@163.com
Telephone: +86-551-62923504
Received: October 9, 2016
Peer-review started: October 10, 2016
First decision: November 9, 2016
Revised: November 24, 2016
Accepted: December 16, 2016
Article in press: December 19, 2016
Published online: February 21, 2017

Abstract
AIM

To explore novel therapeutic target of cisplatin resistance in human gastric cancer.

METHODS

The sensitivity of SGC7901 cells and cisplatin-resistant SGC7901 cells (SGC7901/DDP) for cisplatin were detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay. High-quality total RNA which isolated from SGC7901/DDP cells and SGC7901 cells were used for mRNA microarray analysis. Results were analyzed bioinformatically to predict their roles in the development of cisplatin resistance and the expression of 13 dysregulated mRNAs we selected were validated by quantitative real-time polymerase chain reaction (qRT-PCR).

RESULTS

SGC7901/DDP cells highly resistant to cisplatin demonstrated by MTT assay. A total of 1308 mRNAs (578 upregulated and 730 downregulated) were differentially expressed (fold change ≥ 2 and P-value < 0.05) in the SGC7901/DDP cells compared with SGC7901 cells. The expression of mRNAs detected by qRT-PCR were consistent with the microarray results. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway and protein-protein interaction analysis demonstrated that the differentially expressed mRNAs were enriched in PI3K-Akt, Notch, MAPK, ErbB, Jak-STAT, NF-kappaB signaling pathways which may be involved in cisplatin resistance. Several genes such as PDE3B, VEGFC, IGFBP3, TLR4, HIPK2 and EGF may associated with drug resistance of gastric cancer cells to cisplatin.

CONCLUSION

Exploration of those altered mRNAs may provide more promising strategy in diagnosis and therapy for gastric cancer with cisplatin resistance.

Key Words: Gastric cancer, Dysregulate, Cisplatin resistance, Microarray, Biology

Core tip: We tested the sensitivity of human gastric cancer cells SGC7901/DDP and SGC7901 for cisplatin and compared their mRNA expression profile using a human mRNA microarray, and then performed bioinformatics analysis to depict comprehensively the properties of the differentially expressed mRNAs. Results demonstrated that the dysregulated mRNA were enriched in functions and pathways that may be involved in cisplatin resistance. Exploration of the dysregulated genes could suggest a promising strategy in diagnosis and therapy of gastric cancer with cisplatin resistance.



INTRODUCTION

Gastric cancer is the fourth most common cancer and the second leading cause of cancer death globally[1], and more than two thirds of patients when diagnosed with unresectable disease[2]. The 5-year overall survival rate of patients with advanced gastric cancer approximately 25%[3]. Currently, platinum-based chemotherapy regimen is the standout chemotherapy frequently used for advanced gastric cancer[4,5], and median overall survival and progression free survival was significantly longer in cisplatin-containing combination therapy compared to non-cisplatin containing regimens[6,7]. However, cisplatin-based chemotherapeutic agents are often limited in chemotherapy due to drug resistance[8,9].

Cisplatin resistance of gastric cancer is multifactorial, accumulating evidence have suggested that the aberrant expression of proteins which associated with decreased cellular accumulation, increased DNA repair capacity, increased drug inactivation[10] play important role in the acquisition of cisplatin resistance. Previous researches have shown that abnormal expression of copper transporter 1 (CTR1) and MRP2 lead to cisplatin resistance by reducing the concentration of cisplatin in cells[11-13]. Moreover, the upregulation of excision repair cross complementing 1 (ERCC1)[14], X-ray repair cross complementing 1 (XRCC1)[15] and breast cancer 1 (BRCA1)[16] have shown to be involved in cisplatin resistance by removal of Pt-DNA adducts[17,18]. Other studies have shown that downregulation of the human epidermal growth factor receptor II (ErbB2) can significantly enhanced the apoptosis-inducing effects of cisplatin in gastric cancer[19,20].

The mechanisms of cisplatin resistance are quite complex and have not been fully revealed till now, so investigation of the molecular mechanisms and biomarkers is urgently needed. This study aims to analyze mRNA expression profiles in SGC7901/DDP cells to explore more chemotherapeutic molecular targets and to guide appropriate chemotherapy for gastric cancer with cisplatin resistance.

MATERIALS AND METHODS
Cell lines and culture

The human cisplatin-resistant gastric cancer cell line SGC7901/DDP and its parental cells SGC7901 were purchased from KeyGEN Biotechnology Company (Nanjing, Jiangsu, China). Cells were cultured in RPMI-1640 medium (Gibco, Grand Island, NY, United States) containing 10% fetal calf serum (Gibco, NY, United States) supplemented with 100 U/mL penicillin and 100 μg/mL streptomycin. Cells were cultured in a humidified atmosphere with 5% CO2 at 37 °C. Cisplatin (Sigma, CA, United States) with final concentration of 800 ng/mL was added to the culture media for SGC7901/DDP cells to maintain the cisplatin-resistant phenotype.

MTT method assay for SGC7901/DDP and SGC7901 cells viability

SGC7901/DDP and SGC7901 cells were suspended at a density of 1 × 105 cells/mL and planted into 96-well culture plate. After 24 hours, the cells were treated with freshly prepared DDP. The final concentrations were 133.34 μmol /L, 66.67 μmol/L, 6.67 μmol/L, 0.67 μmol/L and 0.067 μmol/L, because the human peak plasma concentration for DDP has been reported as 6.67 μmol/L[21]. Cell viability was examined after 48 h and was determined by adding 20 μL MTT (5 mg/mL) to each well and incubated for a further 4 h. The resulting formazan crystal was dissolved by addition of 150 μL dimethyl sulfoxide (DMSO) (sigma, Germany) each well, and then plates were shaken for 10 minutes. The absorbance at 490 nm was measured by spectrophotometer (ELx 800; BioTek; Winooski, VT, United States). The inhibition of growth (IC50) for DDP was calculated by the cells relative viability. Each experiment was performed in triplicate.

Total RNA extraction and mRNA microarray

Cells were harvested when they had grown to 80%-90% confluency and were still in logarithmic phase. Total RNA was extracted from the three matched pairs of SGC7901/DDP and SGC7901 cells using TRIzol reagent (Invitrogen, Carlsbad, CA, United States) according to the manufacturer’s instructions. The quality of total RNA was measured by NanoDrop ND-2000 spectrophotometer (Thermo Scientific, Waltham, MA, United States). Total RNA from three paired samples were amplified and transcribed into fluorescent cDNA, and then the fluorescent labeled samples were hybridized to the Agilent LncRNA-mRNA Human Gene Expression Microarray V4.0 (Capital Bio Corp, Beijing, China) which contains 25069 human mRNA according to the manufacturer’s recommendations. The microarray was scanned by an Agilent Microarray Scanner. Image processing was conducted using Agilent Feature Extraction software and raw microarray signals normalized using Agilent Gene-Spring software. The normalized mRNA expression profiles data output was received in Excel spreadsheets. The two group of samples data were analyzed by t-test to get the P-values. FC values representing the differently expressed mRNAs between SGC7901/DDP and their parental cells. Cluster 3.0 software was performed to show differential expression patterns of mRNAs.

Bioinformatics analysis

Bioinformatics analysis were generated using KOBAS software and STRING 9.1 software. KOBAS software was used to analyze Ontology, Disease and pathways of the dysregulated mRNAs. KOBAS associated with 1 ontology database (Gene Ontology), 5 disease databases (OMIM, KEGG DISEASE, PID Reactome, FunDO, GAD, NHGRI) and 7 pathway databases (KEGG PATHWAY, PID Curated, PID BioCarta, BioCyc, eactome, Panther). The entire analysis process includes two steps: first, bring the input gene ID map to the gene in the databases, and then annotate pathways, disease and function of these genes involved in. Second step, compare the first step results with background (usually the entire genome of the gene, or the entire probe on the chip), and unearth statistically significant enrichment pathways, disease or function. Fisher’s exact test and χ2 test were used as statistical tests and the FDR was performed to correct the P-value[22]. Additionally, we used STRING 9.1 software to decipher the protein-protein interaction (PPI) network of the differentially expressed proteins. The PPI network may help in understanding the molecular mechanism of cisplatin resistance. All mRNA microarray data were given by Capital Bio Corp.

Quantitative real-time PCR validation of microarray results

To validate the reliability of microarray analysis, we performed quantitative real-time PCR (qRT-PCR). The reverse transcription production cDNA was synthesized using oligo-dT primers and Superscript II reverse transcriptase. PCR was performed with SYBR® Premix Ex TaqTM (TaKaRa Bio; Japan) by a Light Cycler PCR system (Agilent Technologies, Palo Alto, CA, United States) according to the manufacturer’s instructions. After amplification, melting curves were analyzed. Beta-actin snRNA used as endogenous control, each sample was done in triplicate. The relative expression levels of target mRNAs were calculated using the 2-∆∆Ct method (where ∆∆Ct is the difference in threshold cycles for the ∆Ct of SGC7901/DDP sample and SGC7901 sample, and ∆Ct is the difference between the target gene and endogenous control beta-actin). Sequences of primers for qRT-PCR are provided in supporting Table 1.

Table 1 Primer of quantitative real-time polymerase chain reaction.
IDPrimerSequence (5'to3')Base (bp)Tm (°C)GC
HIPK2ForwardCCCCGTGTACGAAGGTATGG2059.9060%
ReverseGGGATGTTCTTGCTCTGGCT2060.0355%
PDE3BForwardTGAGAGTTATGGCTGCCTGT2058.7250%
ReverseCTGAGGGGCATTTGTAGCCA2060.3055%
FGF2ForwardTCCACCTATAATTGGTCAAAGTGGT2559.9940%
ReverseCATCAGTTACCAGCTCCCCC2059.8260%
TWIST1ForwardATTCAAAGAAACAGGGCGTGG2159.3947.6%
ReverseCAGAGGTGTGAGGATGGTGC2060.3940%
ZEB2ForwardGCCTCTGTAGATGGTCCAGTGA2261.2154.6%
ReverseATCGCGTTCCTCCAGTTTTCT2160.0047.6%
VEGFCForwardCCCGCCTCTCCAAAAAGCTA2060.0455%
ReverseCGGGTGTCAGGTAAAAGCCT2059.9655%
SPHK1ForwardGCTGCGAAGTTGAGCGAAAA2060.0450%
ReverseCGTTCCCTACAGTGGCCTG1960.0863.2%
BAXForwardGCCCTTTTGCTTCAGGGTTT2059.2450%
ReverseCATCCTCTGCAGCTCCATGT2059.8255%
PTENForwardCAGGATACGCGCTCGGC1760.7370.6%
ReverseACAGCGGCTCAACTCTCAAA2057.8950%
HTRA1ForwardAGCCAAAGAGCTGAAGGACC2059.9655%
ReverseGACATCATTGGCGGAGACCA2060.1155%
CCL5ForwardTGCTGCTTTGCCTACATTGC2059.7650%
ReverseCTTGTTCAGCCGGGAGTCAT2060.0455%
TGM2ForwardCCTCTGTCTCTCCGGGAACC2061.3265%
ReverseTGGCAACCAGGGGTCCTAT1960.2357.9%
TLR4ForwardCTCGGTCAGACGGTGATAGC2059.9760%
ReverseTTTAGGGCCAAGTCTCCACG2059.6855%
ACTBForwardCTCACCATGGATGATGATATCGC2359.1347.8%
ReverseAGGAATCCTTCTGACCCATGC2159.7952.4%
Statistical analysis

MTT test and qRT-PCR statistical analysis was performed using GraphPad Prism software (v. 5.0a; GraphPad Software, La Jolla, CA, United States). We used one-way analysis of variance (ANOVA) followed by Student’s t-test to assess the statistical significance of differences between different cell groups. The threshold for statistical significance was P-values < 0.05. Fold changes of mRNAs validated by qRT-PCR in SGC7901/DDP cells compared with SGC7901 cells are shown as mean ± SD.

RESULTS
Sensitivity of SGC7901/DDP and SGC7901 cells to DDP

To determine the chemotherapy sensitivity of SGC7901/DDP and SGC7901 cell line to cisplatin, varying concentrations of cisplatin were added into the 96-well plates and incubated for 48 h. From these data, half maximal inhibitory concentration (IC50) cisplatin dose was calculated. IC50 cisplatin doses for SGC7901/DDP and SGC7901 (after 48 h in DDP-containing media) were 43.47 ± 0.21 μmol/L and 1.24 ± 0.02 μmol/L, respectively, and the resistance index for SGC7901/DDP cell lines was 35.12, confirming that these cells are refractory to cisplatin. Cell viability was checked by MTT assay (Figure 1).

Figure 1
Figure 1 Cell viability treated with different concentrations of cisplatin for 48 h. MTT assay for SGC7901 cells and SGC7901/DDP cells treated with cisplatin (133.34, 66.67, 6.67, 0.67 and 0.067 μmol/L, respectively).
Expression profile of mRNAs in SGC7901/DDP cells

To show mRNA expression profile in cisplatin-resistant SGC7901/DDP cells, we used a stringency cutoff to identify significantly differently mRNAs (P < 0.05, FC ≥ 2) and two-dimensional hierarchical clustering 3.0 to represent expression profiles between samples (Figure 2). The results indicated that 1308 mRNAs were significantly differentially expressed in SGC7901/DDP cells compared with SGC7901 cells. Among these transcripts, 578 mRNAs were upregulated, and 730 mRNAs were downregulated.

Figure 2
Figure 2 mRNA expression levels from microarray. A: The volcano plot image showed the mRNA expression levels of microarray in SGC7901/DDP cells compared with SGC7901 cells. Black dots: equally expressed mRNAs between SGC7901/DDP cells and SGC7901 cells (FC ≤ 2); red dots: mRNAs were over-expressed in SGC7901/DDP cells compared with SGC7901 cells (FC ≥ 2); green dots: mRNAs in SGC7901/DDP cells were down-expressed compared to SGC7901 cells (P-values < 0.05, FC ≥ 2). Fold changes of these mRNAs in SGC7901/DDP cells compared with SGC7901 cells are shown as mean ± SD; B: Two-dimensional hierarchical clustering image of the 1308 dysregulated mRNAs in the SGC7901/DDP cells compared with the SGC7901 cells, each row represents an mRNA, each column represents a sample. 7901-1, 7901-2 and 7901-3 represent the three samples of SGC7901 cells, DDP-1, DDP-2 and DDP-3 represent the three samples of SGC7901/DDP cells. Red: Higher expression levels; green: Lower expression levels.
Validation of microarray results by qRT-PCR of 13 mRNAs

First, we concentrated on validating the microarray results. From the abnormally expressed (P < 0.05) mRNAs obtained from the microarray analyses, we selected 8 upregulated mRNAs (HIPK2, PDE3B, FGF2, TWIST1, ZEB2, VEGFC, SPHK1, BAX) and 5 downregulated (PTEN, HTRA1, CCL5, TGM2, TLR4) mRNAs for qRT-PCR validation. The relative fold-changes (SGC7901/DDP vs SGC7901) detected by qRT-PCR were consistent with the microarray results (Figure 3), indicating the dependability of our microarray platform.

Figure 3
Figure 3 Quantitative real-time polymerase chain reaction validation of the microarray results of the 13 mRNAs. Relative fold changes in expression between SGC7901/DDP cells and SGC7901 cells were in agreement with microarray.
Statistical analysis

To depict comprehensively the properties of the differentially expressed mRNA in SGC7901/DDP cells, GO annotation and enrichment analysis was performed to evaluate which cellular components, molecular functions and biological processes may be are affected by this dysregulation. The GO enrichment analysis showed that the differentially expressed genes were involved in a variety of functions, including locomotion, chemotaxis, cell adhesion, regulation of cell migration, extracellular matrix disassembly, response to xenobiotic chemotaxis, localization of cell adhesion and blood vessel morphogenesis (Figure 4A).

Figure 4
Figure 4 Bioinformatic analysis of differentially expressed mRNAs. Gene ontology analysis of mRNAs dysregulated in SGC7901/DDP cells compared with SGC7901 cells. A: Top 30 molecular functions of the dysregulated mRNAs may associated with. Gene ontology analysis include biological processes, cellular components and molecular function; B: Gene ontology enriched diseases. Top 30 diseases annotations of dysregulated mRNAs may involve in. The disease enrich system include 5 disease databases: OMIM, KEGG disease, FunDO, GAD and NHGRI GWAS Catalog.

Additionally, 59 human diseases were significant enriched (P < 0.05) in five human disease databases (KEGG DISEASE, FunDO, GAD, NHGRI GWAS Catalog and OMIM) (Figure 4B, Table 2). Furthermore, it is worth noting that in KEGG disease database, gastric cancer is the most highly enriched disease, and the input genes include DCC, CD44, CDH1, VEGFC, EGF, TGFA.

Table 2 Different expressed mRNAs enriched by KOBAS.
TermDatabaseP valueInput gene symbols
Gastric cancerKEGG DISEASE0.0016DCC, CD44, CDH1, VEGFC, EGF, TGFA
Skin diseasesKEGG DISEASE0.0078DSP, TGM1, CCL5, IL31RA, SPINK5, HLA, FERMT1, KRT14, CTSC, COL17A1, LAMA3, REEP1, RIN2, ALOXE3, ABCC6, WNT10A, FBLN5
Skin and soft tissue diseasesKEGG DISEASE0.0078DSP, TGM1, CCL5, IL31RA, SPINK5, HLA, FERMT1, KRT14, CTSC, COL17A1, LAMA3, REEP1, RIN2, ALOXE3, ABCC6, WNT10A, FBLN5
Macular degenerationKEGG DISEASE0.0140C3, FBLN5, CFH, TLR4
Cancers of the digestive systemKEGG DISEASE0.0439DCC, CD44, CDH1, VEGFC, EGF, TGFA
Familial thoracic aortic aneurysm and dissection (TAAD)KEGG DISEASE0.0459MYLK, TGFBR1
HypomagnesemiaKEGG DISEASE0.0459TRPM6, EGF
Multiple epiphyseal dysplasia (MED)KEGG DISEASE0.0459COL9A3, MATN3
Transient neonatal diabetes mellitus (TNDM)KEGG DISEASE0.0459PLAGL1, ZFP57
Non-syndromic autosomal dominant mental retardationKEGG DISEASE0.0461EPB41L1, DOCK8, PACS1, SMARCA4
Cardiac hypertrophyNHGRI GWAS Catalog0.0028PLXNA2, GRIK2, COL17A1, JAG1, SNAP25, BTBD3, SLX4IP
Response to fenofibrate (adiponectin levels)NHGRI GWAS Catalog0.0046OAS2, PMEPA1, SHANK2, SCUBE1, SLC30A4, PCK1
Complement C3 and C4 levelsNHGRI GWAS Catalog0.0094HLA, CFHR3, CFH, C3
Neutrophil countNHGRI GWAS Catalog0.0119PLCB4, TGFA, FGGY, PDGFD, PSD3
Nephropathy(idiopathic membranous)NHGRI GWAS Catalog0.0137HLA, ITGB6, PLA2R1
Sleep durationNHGRI GWAS Catalog0.0195PLLP, TMC5, ADAMTS14
Airflow obstructionNHGRI GWAS Catalog0.0259HYKK, LEF1, SERPINB8, GPR126, MAP3K13, PTPRD
Cystic fibrosis severityNHGRI GWAS Catalog0.0265HLA, EHF, AHRR
Metabolite levels (5-HIAA/ MHPG Ratio)NHGRI GWAS Catalog0.0265PIEZO2, ROBO2, ADAM12
Bronchopulmonary dysplasiaNHGRI GWAS Catalog0.0296PLXDC2, ZNF770, SPOCK1, TRPS1, RASGRF1, HIVEP3
Major depressive disorderNHGRI GWAS Catalog0.0346PCLO, SLC6A15, ENOX1, SYPL2, IGFBP1, IGFBP3, C12orf5, ATXN1, PIEZO2, TRPS1, RASGEF1B, FGF12, KCNH5
IgA nephropathyNHGRI GWAS Catalog0.0346HLA, ACOXL, TNFSF13
Pulmonary function declineNHGRI GWAS Catalog0.0368MUSK, CSMD1, RORA, FLRT2
Palmitic acid (16:0) plasma levelsNHGRI GWAS Catalog0.0368SCD, CNN3, GRIK2, PTPRD
Male-pattern baldnessNHGRI GWAS Catalog0.0439AUTS2, EDA2R, AR
Response to citalopram treatmentNHGRI GWAS Catalog0.0439LAMA1, RORA, EGFLAM
HyperlipidemiaFunDO0.0050IRS1, CCL5, C3, PAPPA, TXNIP, APOC1, F3, SCD
ThrombocytopeniaFunDO0.0068GATA1, CCL5, ITGB3, IL11, CXCL8, MPL
FibromyalgiaFunDO0.0126MAOB, CXCL8, BDNF, IGFBP3
CirrhosisFunDO0.0209RBP4, KRT18, IGFBP3, KRT8, EGF, F3, FGF2IGFBP1
Hepatitis CFunDO0.0321CD274, CCL5, RBP4, MKI67, CXCL8, KRT18, TLR4, KRT8, FGF2
ThalassemiaFunDO0.0345LCN2, CXCL8, ANK2, KIR3DL1, MUC1
Gingival overgrowthFunDO0.0417EDN1, IL15, FGF7
Pulmonary fibrosisFunDO0.0474CSF1, BDNF, MMP7, EDN1, CCL5, ERBB3
Ovary cancerFunDO0.0477LCN2, IL15, CXCL8, FGF7, CASP1
Esophageal tumorFunDO0.0477CD274, TSPAN8, FRAT1, PDCD1LG2, FGF7
HyperlipidemiaGAD0.0093CCL5, HLA, CXCL8, CD22, TNFRSF1B, CD19
ThrombocytopeniaGAD0.0114CSMD1, DOCK4, GALNTL6, SOBP, PLXDC2, SESN3, ADAMTS5, EHF, TMC5, LPL, CD109, FAM117B, PDE1C, TAGLN, PTN, FGD4, DYNC2H1, GNG4, MUSK, FBLN5, CCDC54, TTC9, PMEPA1, TLR4, ANK3, EDA2R, APOC1, BMP2, TOX3, NRG1, ITPK1, PTPRD, KLF6, PAM, PTPRU, LEPR, IKZF2, LHX5, MCTP2, ANKRD50, SEMA6D, PLXNA2, DPYD, GRIK2, SRGAP3, ACOXL, TDRKH, FAM135B, VEGFC, CHST2
FibromyalgiaGAD0.0136GLI3, CELF2, VWA3B, PLXDC2, EDNRA, EDN1, JUN, DOCK8, DCLK2, BTBD3, DCN, CD74, EGFLAM, TLL1, TLR4, BMP2, PTPRD, ANK2, PTPRU, JADE2, IGF2BP2, PAPPA, DOCK2, KLK4, FAM49A, RGS3, AATK, FN1, IGSF10, NCOA7, SCIN, TNS1, FAM135B, MUC16, ADAM19, ATXN1, MTUS2, NXNL2, KCNQ3, ANPEP, CDH2
CirrhosisGAD0.0204IRS1, CCL5, ITGB3, NPR1, NPR3, APOC1, LPL
Hepatitis CGAD0.0258DPYD, CELF4, CELF2, FAM117B, TDRKH, LPCAT4, FBLN5, SOBP, PMEPA1, CSMD1, STOX1, CACNB2, CADM1, VEGFC, SLC7A11, LPL, CD109, MCTP2, SLC24A2, PTPRD, ITPK1
ThalassemiaGAD0.0362MCTP2, PSD3, CCDC54, ROBO2, ELOVL6
Gingival overgrowthGAD0.0419PLXNA2, ATXN1, IGF2BP2, ABCA13, FN1
Pulmonary fibrosisGAD0.0420CREG2, GALNTL6, LINC01550, KIF16B, SH3BGR, TRPS1, PDE1C, NCKAP5, TNFRSF21, RYR3, MAGEC2, EDIL3, CXCL16, MCF2, DTD1, GPC5, KLF6, IKZF2, KCNH5, AJAP1, BTBD3, PHACTR2, ITPK1, IGSF10, SRGAP3, C12orf75, ABI3BP, FOS, SCUBE1
Ovary cancerGAD0.0426CELF4, TRPS1, TWIST1, PQLC2L, MAL2, PSD3, RCAN2, SUPT3H, TGFA, TMEM131L, HIVEP3, CSMD1, ROBO2, CCDC54, PRNP, APOC1, HRK, GPC5, AR, FN1, ABCA13, F2RL2, KLF6, IGF2BP2, LEPREL1, GNG4, SNAP25, MCTP2, FAM49A, ANKRD50, CACNA2D1, PLXNA2, ELOVL6, RUNX2, SCN8A, ATXN1, ID2, SLC24A2, CMTM7, LINGO2
Esophageal tumorGAD0.048CACNA2D1, SLC46A3, CHST2, PKDCC, PPID, CDH2

To determine which pathway might be involved in drug resistance formation, KEGG pathway analysis was used to authenticate pathways and understand biological functions of significantly differentially expressed genes. The result indicated that the differentially expressed mRNAs were enriched for 233 pathways, including the Rap1 signaling pathway, PI3K-Akt signaling pathway, ECM-receptor interaction, TNF signaling pathway, and pathways in cancer, among others (Figure 5, Table 3). Cluster 3.0 software were performed the heat-map.This finding identified many candidate pathways and input genes that may play an important role in resistance mechanism.

Table 3 Cisplatin resistance pathway and input gene (P < 0.05, FC ≥ 2.0).
PathwayInput geneFold changeRegulationGenomic coordinatesCyto band
PI3K-Akt signaling pathwayLAMA12.60826UpChr18:6958512-6956742hs|18p11.31
LAMA12.75269UpChr18:6942035-6941976hs|18p11.31
GNG42.09356UpChr1:235714443-235714384hs|1q42.3
ITGB32.96629UpChr17:45389027-45389086hs|17q21.32
ITGB67.72783UpChr2:160964233-160958330hs|2q24.2
VEGFC2.92538UpChr4:177604882-177604823hs|4q34.3
PDGFD2.42861UpChr11:103778445-103778386hs|11q22.3
IRS12.00967UpChr2:227596677-227596618hs|2q36.3
GNGT12.04779UpChr7:93536149-93540155hs|7q21.3
CSF12.25620UpChr1:110466137-110466196hs|1p13.3
EGF4.76437UpChr4:110932689-110932748hs|4q25
FGF23.02437UpChr4:123819331-123819390hs|4q28.1
FGF22.99240UpChr4:123819317-123819376hs|4q28.1
FN12.31254UpChr2:216288895-216288217hs|2q35
COL4A62.08497UpChrx:107399109-107399050hs|Xq22.3
FGF1210.99211UpChr3:191860574-191860515hs|3q28
GNG112.01984UpChr7:93555764-93555823hs|7q21.3
FGF72.19252UpChr15:49776810-49776869hs|15q21.2
LAMA32.56116DownChr18:21534735-21534794hs|18q11.2
IFNA212.30808DownChr9:21166331-21166272hs|9p21.3
CREB3L32.40183DownChr19:4172219-4172278hs|19p13.3
TLR42.13271DownChr9:120476856-120476915hs|9q33.1
COL6A22.89458DownChr21:47546086-47546145hs|21q22.3
CD192.09302DownChr16:28950600-28950659hs|16p11.2
LPAR53.83177DownChr12:6728794-6728735hs|12p13.31
COL4A42.11177DownChr2:227867523-227867464hs|2q36.3
PCK14.49558DownChr20:56141030-56141089hs|20q13.31
VTN3.82587DownChr17:26694806-26694747hs|17q11.2
GNGT216.48365DownChr17:47284034-47283975hs|17q21.32
IL2RG2.87954DownChrx:70328539-70328480hs|Xq13.1
COL5A37.53410DownChr19:10070602-10070543hs|19p13.2
FGF1317.08866DownChrx:137713947-137713888hs|Xq26.3
MAPK signaling pathwayFLNC4.57879DownChr7:128498538-128498597hs|7q32.1
FLNC4.81302DownChr7:128498476-128498535hs|7q32.1
CACNB27.83293DownChr10:18787305-18787364hs|10p12.31
RASGRF14.87152DownChr15:79254554-79254495hs|15q25.1
FOS2.17501DownChr14:75748214-75748273hs|14q24.3
JUN2.04000DownChr1:59246570-59246511hs|1p32.1
RASGRP23.10358DownChr11:64508971-64508912hs|11q13.1
FGF1317.08866DownChrx:137713947-137713888hs|Xq26.3
TGFBR12.93035UpChr9:101916322-101916381hs|9q22.33
TGFBR14.76437UpChr4:110932689-110932748hs|4q25
EGF4.76437UpChr4:110932689-110932748hs|4q25
FGF1210.99211UpChr3:191860574-191860515hs|3q28
MAP3K132.25019UpChr3:185161379-185165590hs|3q27.2
FGF23.02437UpChr4:123819331-123819390hs|4q28.1
FGF22.99240UpChr4:123819317-123819376hs|4q28.1
MAP2K72.08267UpChr19:7979302-7979361hs|19p13.2
FGF72.19252UpChr15:49776810-49776869hs|15q21.2
CACNG48.83585UpChr17:65028139-65028198hs|17q24.2
CACNG42.94145UpChr17:65029115-65029174hs|17q24.2
CACNB42.14311UpChr2:152694239-152694180hs|2q23.3
GADD45A2.56659UpChr1:68153371-68153430hs|1p31.3
BDNF2.32411UpChr11:27679959-27679900hs|11p14.1
BDNF2.30323UpChr11:27677072-27677013hs|11p14.1
CACNA2D12.09452UpChr7:81579504-81579445hs|7q21.11
Notch signaling pathwayMAML22.03379UpChr11:95712434-95712375hs|11q21
JAG13.20086UpChr20:10619120-10619061hs|20p12.2
MAML32.57919UpChr4:140810806-140810747hs|4q31.1
DTX49.99859DownChr11:58975615-58975674hs|11q12.1
ErbB signaling pathwayEGF4.76437UpChr4:110932689-110932748hs|4q25
NRG12.77996UpChr8:32474390-32585512hs|8p12
MAP2K72.08267UpChr19:7979302-7979361hs|19p13.2
JUN2.04000DownChr1:59246570-59246511hs|1p32.1
ERBB35.29571DownChr12:56482380-56482439hs|12q13.2
ERBB38.12050DownChr12:56496160-56496219hs|12q13.2
TGFA2.37427DownChr2:70675378-70675319hs|2p13.3
Jak-STAT signaling pathwayIL114.21849UpChr19:55875847-55875788hs|19q13.42
IL152.92970UpChr4:142654431-142654490hs|4q31.21
SOCS22.00180UpChr12:93969799-93969858hs|12q22
SPRY15.95682UpChr4:124324494-124324553hs|4q28.1
LEPR2.90187UpChr1:66102129-66102188hs|1p31.3
IL2RG2.87954DownChrx:70328539-70328480hs|Xq13.1
MPL3.41581DownChr1:43819826-43819885hs|1p34.2
NF-kappaB signaling pathwayCXCL22.03846UpChr4:74963044-74962985hs|4q13.3
CXCL89.97781UpChr4:74609265-74609324hs|4q13.3
TLR42.13271DownChr9:120476856-120476915hs|9q33.1
HIF-1 signaling pathwayEDN12.39081UpChr6:12296672-12296731hs|6p24.1
EDN12.46437UpChr6:12296218-12296277hs|6p24.1
EGF4.76437UpChr4:110932689-110932748hs|4q25
TLR42.13271DownChr9:120476856-120476915hs|9q33.1
MicroRNAs in cancerIRS12.00967UpChr2:227596677-227596618hs|2q36.3
ZEB23.32563UpChr2:145146320-145146261hs|2q22.3
ZEB22.70558UpChr2:145182422-145182363hs|2q22.3
CD442.02409UpChr11:35253812-35253871hs|11p13
RECK2.25018UpChr9:36124319-36124378hs|9p13.3
ITGB32.96629UpChr17:45389027-45389086hs|17q21.32
SERPINB52.61864UpChr18:61172218-61172277hs|18q21.33
GLS2.36144UpChr2:191829716-191829775hs|2q32.2
GLS2.07371UpChr2:191827822-191827881hs|2q32.2
ERBB35.29571DownChr12:56482380-56482439hs|12q13.2
ERBB38.12050DownChr12:56496160-56496219hs|12q13.2
Figure 5
Figure 5 Heat-map of gene ontology enriched cisplatin resistance pathways and input mRNAs which significantly altered in SGC7901/DDP cells compared with SGC7901 cells. A: PI3K-Akt signaling pathway and input genes; B: MAPK signaling pathway and input genes; C: Notch signaling pathway and input genes; D: ErbB signaling pathway and input genes; E: Jak-STAT signaling pathway and input genes; F: NF-kappa B signaling pathway and input genes; G: HIF-1 signaling pathway and input genes; H: MicroRNAs in cancer and input genes. Each row represents an mRNA, and each column represents a sample. The intensity of the color indicates the relative levels of mRNAs. Red: Higher expression levels; green: Lower expression levels. The name of the input mRNAs which significantly altered (P < 0.05, FC ≥ 2) is present at the right of the figure.
Interaction network analysis

The STRING 9.1 software (Search Tool for the Retrieval of Interacting Genes) was used to perceive functional relations and generate networks of differential expression of proteins (Figure 6). For all of the 1002 differentially expressed proteins, we extracted a network containing 443 upregulated and 559 downregulated proteins which functionally associated with each other. We found that interacting proteins which participate in angiogenesis, toll-like receptor signaling pathway and cell adhesion had a high level of co-expression.

Figure 6
Figure 6 Interaction network analyses of differentially express proteins. In the network, nodes represents proteins, lines as functional associations between the abnormal expressed proteins and the thickness of the lines indicates the level of confidence in association reported.
DISCUSSION

Cisplatin is widely used against a variety of solid neoplasms, including testicular, ovarian, colorectal, bladder, head and neck cancers and gastric cancer[23]. However, the repeated clinical expose to cisplatin often results in the tumor cells evading the apoptosis program initiated by cisplatin. Therefore, there is a need to explore the molecular mechanisms of cisplatin resistance, in order to overcome drug resistance in tumor therapy. Recently, several studies have indicated that many proteins are involved in the recognition of Pt-DNA adducts and cisplatin-induced apoptosis program[24,25]. In this study, we used microarray, GO, KEGG pathway and protein-protein interaction (PPI) analysis to explore the roles of differentially expressed mRNAs in cisplatin resistance and to support other studies.

Many genes which shown differentially expression in the microarray analysis have been demonstrated to be associated with cisplatin resistance in human cancer (Table 4), such as PDE3B, which was substantially upregulated (P value = 0.00029, Fold Chang (FC) = 10.45) in SGC7901/DDP cells. Treatment with a combination of a PDE3B inhibitor and DDP can significantly increase the number of apoptotic and cell growth-suppressive cancer cells in cisplatin resistant squamous cell carcinoma (SCC) and Hela cells[26]. Research shows that VEGFC, which is upregulated in our data (P value = 0.00013 FC = 2.93), enhanced cell invasion and cisplatin resistance in gastric cancer[27]. In non-small cell lung cancer, loss of IGFBP-3 expression may activate the PI3K/AKT pathway and induce resistance to cisplatin[28]. In support of this association, our results showed that this mRNA is downregulated (P = 0.00007, FC = 2.93) in SGC7901/DDP cells.

Table 4 Dysregulated mRNAs (P < 0.05, FC ≥ 2.0) associated with cisplatin resistance.
Gene symbolP valueFC (abs)RegulationGenenameRef.
FGF70.000352.19252UpFibroblast growth factor 7PMID: 22990650
HIPK22.63E-064.06213UpHomeodomain interacting protein kinase 2PMID: 24846322
EDN19.94E-052.46437UpEndothelin 1PMID: 21220476
CBS0.001082.29340UpCystathionine-beta-synthasePMID: 24236104
PDE3B0.0002910.44998UpPhosphodiesterase 3B, cgmp-inhibitedPMID: 24133626
E2F50.000412.42888UpE2F transcription factor 5, p130-bindingPMID: 22193543
PIN10.001042.13293UpPeptidylprolyl cis/trans isomerase, NIMA-interacting 1PMID: 26820938
EGF0.003464.76437UpEpidermal growth factorPMID: 27086487
CSF10.000252.25620UpColony stimulating factor 1 (macrophage)PMID: 22005523
PCNA0.001032.17028UpProliferating cell nuclear antigenPMID: 24474685
HIPK22.63E-064.06213UpHomeodomain interacting protein kinase 2PMID: 24846322
ENTPD60.000112.43726UpEctonucleoside triphosphate diphosphohydrolase 6 (putative)PMID: 21519793
AKR1C10.000972.29646UpAldo-keto reductase family 1, member C1PMID: 23165153, PMID: 17266043
ASNS0.001722.19491UpAsparagine synthetase (glutamine-hydrolyzing)PMID: 23956056, PMID: 17409444
BDNF0.000622.32411UpBrain-derived neurotrophic factorPMID: 22276165, PMID: 17044982
CABYR0.010892.55664UpCalcium binding tyrosine-(Y)-phosphorylation regulatedPMID: 24362251
FGF22.15E-062.99240UpFibroblast growth factor 2 (basic)PMID: 12894531
SLC7A111.95E-052.93256UpSolute carrier family 7 member 11PMID: 24516043
TUBB30.000462.00213UpTubulin, beta 3 class IIIPMID: 25107571
TWIST10.001802.96340UpTwist family bhlh transcription factor 1PMID: 22673193, PMID: 22245869
JAG19.41E-053.20086UpJagged 1PMID: 24659709
ANXA110.000312.36619DownAnnexin A11PMID: 19484149, PMID: 17982121
CCL52.67E-055.05630DownChemokine (C-C motif) ligand 5PMID: 26983899
FGF130.0004417.08866DownFibroblast growth factor 13PMID: 24113164
IGFBP37.48E-052.92508DownInsulin-like growth factor binding protein 3PMID: 20023704
KLK60.000662.24596DownKallikrein-related peptidase 6PMID: 23307575
SLC7A84.50E-055.36735DownSolute carrier family 7 member 8PMID: 23462296
TGM22.88E-056.24520DownTransglutaminase 2PMID: 21424127, PMID: 24828664
TLR40.001142.13271DownToll-like receptor 4PMID: 21616060, PMID: 22583829
XAF10.024053.20613DownXIAP associated factor 1PMID: 25824780, PMID: 25240826
TCEA20.000613.65969DownTranscription elongation factor A (SII), 2PMID: 16142353

GO enrichment analysis exhibits many functions which the differently expressed mRNAs are involved in, including locomotion, chemotaxis, cell adhesion, regulation of cell migration, extracellular matrix disassembly, response to xenobiotic chemotaxis, localization of cell adhesion and blood vessel morphogenesis. Functional annotation showed that the differently expressed mRNAs mainly regulate cellular biological behaviors in the progress of regulation of transcription. How the underlying targets of each GO term are implicated in the cisplatin resistance needs further investigation in the future.

Our KEGG pathway analysis showed that the differently expressed mRNAs are enriched in pathways of ECM-receptor interaction, PI3K-Akt, Rap1, MAPK, Notch1, ErbB, ABC transporters, Jak-STAT, NF-κB, HIF-1 and TGF-β. All of those pathways have been confirmed to be involved in cisplatin resistance in different experiments described previously. For example, the inhibition of PI3K-Akt signaling pathway may increase the sensitivity of gastric cancer cells to cisplatin chemotherapy[29]. Another study found that Janus kinase 2 (JAK2) signal transducer and activator of transcription 3 (STAT3) signaling pathways were activated by overexpressed AKT in cisplatin resistant human gastric cancer cells[30]. A study revealed that the canonical NF-κB signaling pathway was involved in APRIL-mediated cisplatin resistance in gastric cancer[31]. Our data are consistent with these previous studies, and these pathways and input genes deserve our attention in gastric cancer cisplatin resistance.

Although protein expression is generally stable when organs mature, under various pathological and physiological conditions, gene expression may change and ultimately result in aberrant protein levels. Therefore, research on proteomics is helpful to illustrate some biological mechanisms, including cisplatin resistance. Protein-protein interaction network analysis might uncover previously unknown molecular mechanisms of cisplatin resistance. Hub proteins of subnetworks which interact with many partners might associate with drug resistance. For example, studies have shown that dysregulation of the genes PDE3B, TLR4, and HIPK2 is associated with cisplatin resistance in human SCC cells, ovarian granulosa tumor cells and bladder cancer cells, respectively[26,32,33]. Moreover, hub proteins and their partners may have similar biological functions. Since downregulation of EGF has been shown to substantially overcome resistance to cisplatin in ovarian cancer[34], we predict that the proteins EDN1 and DCN, whose hub protein is EGF, may contribute to cisplatin resistance in a similar fashion. We also found that ZEB2, which over-expressed in SGC7901/DDP compared with SGC7901 has a similar expression profile to TWIST1, suggesting that ZEB2 may play an important role in cisplatin resistance by regulating the expression of TWIST1. Nevertheless, more evidence and research is needed.

In conclusion, our study identified mRNAs differentially expressed between gastric cancer cell lines SGC7901/DDP and SGC7901. These results provide a global view of the function of the differentially expressed mRNAs. Several molecular and pathway abnormalities detected in our study have previously been reported to be associated with drug resistance in gastric cancer. The dysregulated mRNAs identified participate in cisplatin resistance through diverse mechanisms, and further investigation is required to confirm the role in drug resistance of these transcripts, pathways and the interaction networks of the proteins they code for.

COMMENTS
Background

Cisplatin-contained chemotherapy is one of the most frequently used for advanced gastric cancer; however, this chemotherapeutic agent is often limited due to drug resistance and result unsatisfactory prognosis. Research increasingly suggests that abnormal expression of biological pathway and proteins associated with cisplatin resistance. This demonstrated that more bioinformatics study is needed to predict targets for gastric cancer with cisplatin.

Research frontiers

Bioinformatics analysis demonstrated that some mRNAs which related to the biological behavior abnormal expression in SGC7901/DDP cells. These mRNAs have already been shown to play important roles in the process of cisplatin resistance of various cancers, including gastric cancer.

Innovations and breakthroughs

The authors performed bioinformatics analysis of mRNA expression profile in SGC7901/DDP cells compared with SGC7901 cells, and found that many mRNAs and pathways in SGC7901/DDP cells expressed abnormally, these may participate in and predict cisplatin resistance in gastric cancer.

Applications

These results suggest that targeting the differently expression mRNA may provide more selective approaches to reverse cisplatin resistance of therapeutic targets.

Terminology

The definition of cisplatin resistance: in the clinic, if a patient who have disease recurrence within the first months after the recent cisplatin dose, the patient is considered cisplatin resistance; in cells, generally, resistance index > 20 exhibited high resistance, resistance index 5-15 is moderate resistance, resistance index < 5 represent low or no resistance. Correct P: Using Benjamini Hochberg FDR method for correction of p values. Fold change (FC): gene expression in SGC7901 / DDP cells compared with SGC7901 cells.

Peer-review

The paper is a good study on mRNAs expression profile in SGC7901/DDP cells. The investigators shown that many mRNAs was abnormal expressed in SGC7901/DDP cells and these mRNAs enriched in many biological process which have already been shown to play important roles in the process of cisplatin resistance in human cancer.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Ghosh RD S- Editor: Qi Y L- Editor: A E- Editor: Wang CH

References
1.  Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C, Lunet N. Worldwide trends in gastric cancer mortality (1980-2011), with predictions to 2015, and incidence by subtype. Eur J Cancer. 2014;50:1330-1344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 474]  [Article Influence: 47.4]  [Reference Citation Analysis (0)]
2.  Wöhrer SS, Raderer M, Hejna M. Palliative chemotherapy for advanced gastric cancer. Ann Oncol. 2004;15:1585-1595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 133]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
3.  Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10002]  [Cited by in F6Publishing: 10353]  [Article Influence: 739.5]  [Reference Citation Analysis (0)]
4.  Al-Batran SE, Hartmann JT, Probst S, Schmalenberg H, Hollerbach S, Hofheinz R, Rethwisch V, Seipelt G, Homann N, Wilhelm G. Phase III trial in metastatic gastroesophageal adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. J Clin Oncol. 2008;26:1435-1442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 525]  [Cited by in F6Publishing: 540]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
5.  Kang YK, Kang WK, Shin DB, Chen J, Xiong J, Wang J, Lichinitser M, Guan Z, Khasanov R, Zheng L. Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: a randomised phase III noninferiority trial. Ann Oncol. 2009;20:666-673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 544]  [Cited by in F6Publishing: 591]  [Article Influence: 39.4]  [Reference Citation Analysis (0)]
6.  Koizumi W, Narahara H, Hara T, Takagane A, Akiya T, Takagi M, Miyashita K, Nishizaki T, Kobayashi O, Takiyama W. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol. 2008;9:215-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1320]  [Cited by in F6Publishing: 1365]  [Article Influence: 85.3]  [Reference Citation Analysis (0)]
7.  Wagner AD, Unverzagt S, Grothe W, Kleber G, Grothey A, Haerting J, Fleig WE. Chemotherapy for advanced gastric cancer. Cochrane Database Syst Rev. 2010;CD004064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 380]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
8.  Rabik CA, Dolan ME. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev. 2007;33:9-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1096]  [Cited by in F6Publishing: 1154]  [Article Influence: 64.1]  [Reference Citation Analysis (0)]
9.  Garcia JA, Dreicer R. Systemic chemotherapy for advanced bladder cancer: update and controversies. J Clin Oncol. 2006;24:5545-5551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
10.  Wilson TR, Longley DB, Johnston PG. Chemoresistance in solid tumours. Ann Oncol. 2006;17 Suppl 10:x315-x324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 128]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
11.  Borst P, Evers R, Kool M, Wijnholds J. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst. 2000;92:1295-1302.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Hioki M, Gotohda N, Konishi M, Nakagohri T, Takahashi S, Kinoshita T. Predictive factors improving survival after gastrectomy in gastric cancer patients with peritoneal carcinomatosis. World J Surg. 2010;34:555-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
13.  Yang T, Chen M, Chen T, Thakur A. Expression of the copper transporters hCtr1, ATP7A and ATP7B is associated with the response to chemotherapy and survival time in patients with resected non-small cell lung cancer. Oncol Lett. 2015;10:2584-2590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
14.  Metzger R, Leichman CG, Danenberg KD, Danenberg PV, Lenz HJ, Hayashi K, Groshen S, Salonga D, Cohen H, Laine L. ERCC1 mRNA levels complement thymidylate synthase mRNA levels in predicting response and survival for gastric cancer patients receiving combination cisplatin and fluorouracil chemotherapy. J Clin Oncol. 1998;16:309-316.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Xu W, Chen Q, Wang Q, Sun Y, Wang S, Li A, Xu S, Røe OD, Wang M, Zhang R. JWA reverses cisplatin resistance via the CK2-XRCC1 pathway in human gastric cancer cells. Cell Death Dis. 2014;5:e1551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 44]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
16.  Shim HJ, Yun JY, Hwang JE, Bae WK, Cho SH, Lee JH, Kim HN, Shin MH, Kweon SS, Lee JH. BRCA1 and XRCC1 polymorphisms associated with survival in advanced gastric cancer treated with taxane and cisplatin. Cancer Sci. 2010;101:1247-1254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 60]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
17.  Liu J, Deng N, Xu Q, Sun L, Tu H, Wang Z, Xing C, Yuan Y. Polymorphisms of multiple genes involved in NER pathway predict prognosis of gastric cancer. Oncotarget. 2016;7:48130-48142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
18.  Sargent RG, Meservy JL, Perkins BD, Kilburn AE, Intody Z, Adair GM, Nairn RS, Wilson JH. Role of the nucleotide excision repair gene ERCC1 in formation of recombination-dependent rearrangements in mammalian cells. Nucleic Acids Res. 2000;28:3771-3778.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Funato T, Kozawa K, Fujimaki S, Miura T, Kaku M. Increased sensitivity to cisplatin in gastric cancer by antisense inhibition of the her-2/neu (c-erbB-2) gene. Chemotherapy. 2001;47:297-303.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Zhou N, Qu Y, Xu C, Tang Y. Upregulation of microRNA-375 increases the cisplatin-sensitivity of human gastric cancer cells by regulating ERBB2. Exp Ther Med. 2016;11:625-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
21.  Sileni VC, Fosser V, Maggian P, Padula E, Beltrame M, Nicolini M, Arslan P. Pharmacokinetics and tumor concentration of intraarterial and intravenous cisplatin in patients with head and neck squamous cancer. Cancer Chemother Pharmacol. 1992;30:221-225.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Zhou C, Zhang T, Liu F, Zhou J, Ni X, Huo R, Shi Z. The differential expression of mRNAs and long noncoding RNAs between ectopic and eutopic endometria provides new insights into adenomyosis. Mol Biosyst. 2016;12:362-370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
23.  Muggia FM, Bonetti A, Hoeschele JD, Rozencweig M, Howell SB. Platinum Antitumor Complexes: 50 Years Since Barnett Rosenberg’s Discovery. J Clin Oncol. 2015;33:4219-4226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 119]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
24.  Ge X, Liu X, Lin F, Li P, Liu K, Geng R, Dai C, Lin Y, Tang W, Wu Z. MicroRNA-421 regulated by HIF-1α promotes metastasis, inhibits apoptosis, and induces cisplatin resistance by targeting E-cadherin and caspase-3 in gastric cancer. Oncotarget. 2016;7:24466-24482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 90]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
25.  Sakai W, Swisher EM, Karlan BY, Agarwal MK, Higgins J, Friedman C, Villegas E, Jacquemont C, Farrugia DJ, Couch FJ. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature. 2008;451:1116-1120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 739]  [Cited by in F6Publishing: 796]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
26.  Uzawa K, Kasamatsu A, Baba T, Usukura K, Saito Y, Sakuma K, Iyoda M, Sakamoto Y, Ogawara K, Shiiba M. Targeting phosphodiesterase 3B enhances cisplatin sensitivity in human cancer cells. Cancer Med. 2013;2:40-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Cho HJ, Kim IK, Park SM, Baek KE, Nam IK, Park SH, Ryu KJ, Choi J, Ryu J, Hong SC. VEGF-C mediates RhoGDI2-induced gastric cancer cell metastasis and cisplatin resistance. Int J Cancer. 2014;135:1553-1563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 40]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
28.  Cortés-Sempere M, de Miguel MP, Pernía O, Rodriguez C, de Castro Carpeño J, Nistal M, Conde E, López-Ríos F, Belda-Iniesta C, Perona R. IGFBP-3 methylation-derived deficiency mediates the resistance to cisplatin through the activation of the IGFIR/Akt pathway in non-small cell lung cancer. Oncogene. 2013;32:1274-1283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 53]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
29.  Fu X, Feng J, Zeng D, Ding Y, Yu C, Yang B. PAK4 confers cisplatin resistance in gastric cancer cells via PI3K/Akt- and MEK/Erk-dependent pathways. Biosci Rep. 2014; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 70]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
30.  Zhang LL, Zhang J, Shen L, Xu XM, Yu HG. Overexpression of AKT decreases the chemosensitivity of gastric cancer cells to cisplatin in vitro and in vivo. Mol Med Rep. 2013;7:1387-1390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
31.  Zhi X, Tao J, Xiang G, Cao H, Liu Z, Yang K, Lv C, Ni S. APRIL induces cisplatin resistance in gastric cancer cells via activation of the NF-κB pathway. Cell Physiol Biochem. 2015;35:571-585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
32.  Lin J, Zhang Q, Lu Y, Xue W, Xu Y, Zhu Y, Hu X. Downregulation of HIPK2 increases resistance of bladder cancer cell to cisplatin by regulating Wip1. PLoS One. 2014;9:e98418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
33.  Woods DC, White YA, Dau C, Johnson AL. TLR4 activates NF-κB in human ovarian granulosa tumor cells. Biochem Biophys Res Commun. 2011;409:675-680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
34.  Tang XH, Li M, Deng S, Lu MS. Cross-reacting material 197, a heparin-binding EGF-like growth factor inhibitor, reverses the chemoresistance in human cisplatin-resistant ovarian cancer. Anticancer Drugs. 2014;25:1201-1210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]