Open Access

cDNA microarray analysis of the effect of cantharidin on DNA damage, cell cycle and apoptosis-associated gene expression in NCI-H460 human lung cancer cells in vitro

  • Authors:
    • Te‑Chun Hsia
    • Chien‑Chih Yu
    • Shu‑Chun Hsu
    • Nou‑Ying Tang
    • Hsu‑Feng Lu
    • Chun‑Shu Yu
    • Shin‑Hwar Wu
    • Jaung‑Geng Lin
    • Jing‑Gung Chung
  • View Affiliations

  • Published online on: March 24, 2015     https://doi.org/10.3892/mmr.2015.3538
  • Pages: 1030-1042
  • Copyright: © Hsia et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

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


Abstract

Cantharidin (CTD) induces cytotoxic effects in different types of human cancer cell; however, to date, there have been no studies on the effects of CTD on gene expression in human lung cancer cells and the potential associated signaling pathways. Therefore, the present study aimed to investigate how CTD affects the expression of key genes and functional pathways of human H460 lung cancer cells using complementary DNA microarray analysis. Human H460 lung cancer cells were cultured for 24 h in the presence or absence of 10 µM CTD; gene expression was then examined using microarray analysis. The results indicated that 8 genes were upregulated > 4‑fold, 29 genes were upregulated >3‑4‑fold and 156 genes were upregulated >2‑3‑fold. In addition, 1 gene was downregulated >4 fold, 14 genes were downregulated >3‑4‑fold and 150 genes were downregulated >2‑3 fold in H460 cells following exposure to CTD. It was found that CTD affected DNA damage genes, including DNIT3 and GADD45A, which were upregulated 2.26‑ and 2.60‑fold, respectively, as well as DdiT4, which was downregulated 3.14‑fold. In addition, the expression of genes associated with the cell cycle progression were altered, including CCND2, CDKL3 and RASA4, which were upregulated 2.72‑, 2.19‑ and 2.72‑fold, respectively; however, CDC42EP3 was downregulated 2.16‑fold. Furthermore, apoptosis‑associated genes were differentially expressed, including CARD6, which was upregulated 3.54‑fold. In conclusion, the present study demonstrated that CTD affected the expression of genes associated with DNA damage, cell cycle progression and apoptotic cell death in human lung cancer H460 cells.

Introduction

Lung cancer accounts for ~28% of cancer-associated mortali-ties (1), the occurrence of which is increasing worldwide. There are ~1.2 million novel cases of lung cancer and ~1 million mortalities from lung cancer each year (2). Lung cancer may be subdivided into small cell lung carcinoma and non-small cell lung carcinoma (NSCLC). The majority of lung cancer diagnoses are NSCLC (3,4), which has a five-year survival rate of ~33% (5). At present, the standard treatment for patients with resectable stage I to IIIA NSCLC is surgical excision; however, the prognosis remains poor (6). In addition, chemotherapy with or without surgery is not effective in the majority of cases; therefore, it is essential to identify novel compounds, including natural products, which may be employed for the treatment of lung cancer.

Cantharidin (CTD) is a component of mylabris (blister beetle), which has previously been used as a Traditional Chinese Medicine (7). Previous studies have reported that CTD induced cytotoxic effects in leukemia stem cells (8) as well as U937 (9), pancreatic cancer (10), hepatocellular carcinoma (11,12), colon cancer (13) and human lung cancer A549 (14) cells. In addition, CTD was found to inhibit the activity of protein phosphatase 2A (PP2A) (9) and heat shock factor 1 (HSF1) (15). Furthermore, it was shown that CTD induced cell death in human colorectal cancer cells, which was suggested to proceed through inhibiting the binding of heat shock protein 70 (HSP70), B cell lymphoma 2-associated athanogene domain 3 (BAG3) and HSF1 to promoters (15).

Genetic mutations in oncogenes and tumor suppressor genes are present in cancer cells (16,17). The development of cancer cells is well-known to be dependent on oncogenes for tumor initiation and progression; this concept has therefore been named oncogene addiction (18). Oncogenes are commonly used as targets for drug-screening programs (19); however, other signaling pathways have also been examined, such as the molecular chaperone pathway (20). The present study aimed to investigate the effect of CTD on the expression of key genes and functional pathways of human H460 lung cancer cells using complementary DNA microarray analysis. The results of the present study showed that CTD affected DNA damage, the cell cycle and the expression of apoptosis-associated genes in vitro. Differentially expressed genes were then used to generate interaction maps of signaling pathways. The epidermal growth factor and vascular endothelial growth factor receptor pathways, provided by the present study may be useful for the development of novel molecular targeted therapies against lung cancer (21).

Materials and methods

Chemicals and reagents

Cantharidin (CTD), propidium iodide and dimethyl sulfoxide (DMSO) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Minimum essential medium (MEM), fetal bovine serum (FBS), L-glutamine and penicillin-streptomycin were purchased from Gibco-BRL (Carlsbad, CA, USA). CTD was dissolved in DMSO and stored at 20°C.

Lung cancer cell culture

The NCI-H460 human lung cancer cell line was purchased from the Food Industry Research and Development Institute (Hsinchu, Taiwan). Cells were grown in MEM containing 10% (v/v) FBS as well as 100 U/ml penicillin and 100 μg/ml streptomycin in a 37°C humidified incubator with 5% CO2. Cells were then subcultured once they reached 80–90% confluence, as previously described (22).

Complementary (c)DNA microarray assay

H460 cells were placed on 12-well plates at a density of 5×105 cells/well in 2 ml MEM with 10% (v/v) FBS and 2 mM L-glutamine, as well as 100 U/ml penicillin and 100 μg/ml streptomycin for 24 h. Subsequently, cells were treated with or without 10 μM CTD for a further 24 h. Cells (3×106) were then harvested and washed twice with phosphate-buffered saline (Gibco-BRL). Cells were lysed in TRIzol® (Invitrogen Life Technologies, Carlsbad, CA, USA) and total RNA was extracted using a Qiagen RNeasy Mini kit (Qiagen, Valencia, CA, USA). RNA concentrations were determined using a Qubit™ Fluorocytometer (Invitrogen Life Technologies).

Total RNA of CTD-treated and untreated H460 cells was used for cDNA synthesis. Samples were hybridized using an Affymetrix GeneChip Human Gene 1.0 ST array (Affymetrix, Santa Clara, CA, USA) according to the manufacturer’s instructions. Sample fluorescence was quantified by Asia BioInnovations Corp. (Taipei, Taiwan), while data were analyzed using the Transcriptome Analysis Console™ 2.0 Version 2.0.0.9. (Affymetrix) with default robust multichip analysis parameters. A 2-fold change in gene expression was used as the threshold to indicate an effect on expression (710). An Oligo(dT) Maxime RT PreMix kit (iNtRON Biotechnology, Gyeonggi-do, South Korea) was used to reverse transcribe RNA into cDNA. The Affymetrix GeneChip® Whole Transcript Sense Target (ST) Labeling (cat. no. 900673; 30 Rxn; Affymetrix, Santa Clara, CA, USA) assay is designed to generate amplified and biotinylated sense-strand DNA targets from the entire expressed genome without bias. This assay and associated reagents have been optimized specifically for use with the GeneChip® ST arrays, and the probes on the arrays have been selected to be distributed throughout the entire length of each transcript. The gene list complete with Affymetrix transcript identifiers, was uploaded from a spreadsheet onto Metacore 5.0 software (GeneGo pathways analysis; http://www.genego.com). GeneGo recognizes the Affymetrix identifiers and maps the gene to the MetaCore™ data analysis suite, generating maps to describe common pathways or molecular connections between genes in the list. Graphical representations of the molecular associations between the genes were generated using the GeneGo pathway analysis, based upon processes exhibiting a significant association (P<0.05).

Gene ontology analysis

For detection of significantly over-represented GO biological processes, the DAVID functional annotation clustering tool (http://david.abcc.ncifcrf.gov) was used (DAVID Bioinformatics Resources 6.7). Enrichment was determined at the DAVID calculated Benjamini value <0.05. The significance of the overexpression of individual genes was determined using Student’s t-test.

Statistical analysis

Values are representative of three independent experiments. Differences between control and CTD-experimental groups are presented which >2-fold, where positive numbers represent upregulation and negative numbers represent downregulation.

Results

Upregulated and downregulated gene expression in H460 cells exposed to CTD

H460 cells were incubated in the presence or absence of 10 μM CTD in a 12-well plate for 24 h. Cells were then harvested and following the extraction of total RNA, RNA concentrations were determined and cDNA microarray analysis was performed in order to determine the expression of genes. The calculated upregulation and downregulation of gene expression, as determined by the microarray, are shown in Tables I and II, respectively. As shown in Table I, the results indicated that in CTD-treated H460 cells, 8 genes were upregulated >4-fold, 29 genes were upregulated >3–4-fold and 156 genes were upregulated >2–3-fold compared with expression levels in the untreated control cells. In addition, Table II indicated that one gene was downregulated >4 fold, 14 genes were downregulated >3–4 fold and 150 genes were downregulated >2–3 fold in H460 cells following exposure to CTD compared with those in the untreated control cells. The results presented in Table I demonstrated that genes associated with DNA damage, including DN1T3 and GADD45A were upregulated by 2.26-and 2.60-fold, respectively; in addition, the expression of genes associated with the cell cycle progression (check point proteins) were upregulated, including CCND2, CDKL3 and RASA4, which were upregulated 2.72-, 2.19- and 2.72-fold, respectively. Furthermore, the expression of apoptosis-associated genes was upregulated, such as CARD6, which was upregulated 3.54-fold (Table I). By contrast, the results presented in Table II demonstrated that genes associated with DNA damage, cell cycle progression and apoptosis were also downregulated, including DdiT4, CDC42EP3 and STAT2, respectively. These genes were found to be downregulated 3.14-, 2.16 and 2.04-fold, respectively (Table II). Overall, cDNA microarray analysis of H460 cells following treatment with CTD demonstrated that CTD induced the differential expression of numerous genes associated with DNA damage, cell cycle progression and apoptosis.

Table I

Upregulation of gene expression in catharidine-treated NCI-H460 cells.

Table I

Upregulation of gene expression in catharidine-treated NCI-H460 cells.

Probe set IDFold changeGene symbolGene descriptionmRNA accession no.
810837016.50EGR1Early growth response 1NM_001964
801294913.05CDRT1CMT1A duplicated region transcript 1NM_006382
801295110.85CDRT1CMT1A duplicated region transcript 1NM_006382
79166098.05JUNJun oncogeneNM_002228
79770756.45SNORA28Small nucleolar RNA, H/ACA box 28NR_002964
80411685.54SNORD53Small nucleolar RNA, C/D box 53NR_002741
79820845.06SNORD115-11Small nucleolar RNA, C/D box 115-11NR_003303
80979914.27TDO2Tryptophan 2,3-dioxygenaseNM_005651
81144683.99SNORD63Small nucleolar RNA, C/D box 63NR_002913
81588623.87SNORD62ASmall nucleolar RNA, C/D box 62ANR_002914
81588643.87SNORD62ASmall nucleolar RNA, C/D box 62ANR_002914
80074203.85AOC3Amine oxidase, copper-containing 3 (vascular adhesion protein 1)NM_003734
79757793.81FOSFBJ murine osteosarcoma viral oncogene homologNM_005252
80017463.74SNORA46Small nucleolar RNA, H/ACA box 46NR_002978
79143223.70SNORD103ASmall nucleolar RNA, C/D box 103ANR_004054
79143243.70SNORD103ASmall nucleolar RNA, C/D box 103ANR_004054
7933423.69PTPN20AProtein tyrosine phosphatase, non-receptor type 2NR_001042389
79184673.65Clorf103Chromosome 1 open reading frame 103NM_018372
80537973.60LOC400986Protein immuno-reactive with anti-parathyroid hormone polyclona ENST00000456556
81568483.59NR4A3Nuclear receptor subfamily 4, group A, member 3NM_006981
80054833.56FBXW10F-box and WD repeat domain-containing 10NM_031456
81050773.54CARD6Caspase recruitment domain family, member 6NM_032587
81398403.48ERV3Endogenous retroviral sequence 3 (includes zinc)NM_001007253
80308313.45ZNF175Zinc finger protein 175NM_007147
79582003.45EID3EP300-interacting inhibitor of differentiation 3NM_001008394
79366373.44SNORA19Small nucleolar RNA, H/ACA box 19NR_002917
81073533.43ZRSR1Zinc finger (CCCH type), RNA-binding motif and serine/arginine rich 1BC104811
81736003.26NAP1L2Nucleosome assembly protein 1-like 2NM_021963
81268533.25C6orf138Chromosome 6 open reading frame 138NM_001013732
80253013.20CD209CD209 moleculeNM_021155
79231193.17ZBTB41Zinc finger and BTB domain-containing 41NM_194314
79855553.11EFTUD1Elongation factor Tu guanine triphosphate binding domain-containingNM_024580
79529863.09HSN2Hereditary sensory neuropathy, type IINM_213655
81145723.08HBEGFHeparin-binding epidermal growth factor-like growth factorNM_001945
80495403.05LRRFIP1Leucine-rich repeat (in FLII) interacting protein 1NM_001137550
80479263.03MAP2 Microtubule-associated protein 2NM_002374
79543823.02PYROXD1Pyridine nucleotide-disulphide oxidoreductase domain 1NM_024854
79572602.99GLIPR1GLI pathogenesis-related 1NM_006851
80540542.97ANKRD36BAnkryin repeat domain 36BNM_025190
79075722.96PAPPA2Pappalysin 2NM_020318
80906882.93SNORA58Small nucleolar RNA, H/ACA box 58NR_002985
81399352.89TYW1BtRNA-yW synthesizing protein 1 homolog BNM_001145440
79820282.87SNORD115-11Small nucleolar RNA, C/D box 115-11NR_003303
79820502.87SNORD115-11Small nucleolar RNA, C/D box 115-11NR_003303
79820642.87SNORD115-11Small nucleolar RNA, C/D box 115-11NR_003303
79820782.87SNORD115-11Small nucleolar RNA, C/D box 115-11NR_003303
79820922.87SNORD115-11Small nucleolar RNA, C/D box 115-11NR_003303
79053392.86GABPB2GA binding protein transcription factor, β subunitNM_144618
81407822.84ABCB1ATP-binding cassette, sub-family B (multidrug resistance protein/transporter associated with antigen processing)NM_000927
81394822.83SNORA5ASmall nucleolar RNA, H/ACA box 5ANR_002919
81247562.83PPP1R10Protein phosphatase 1, regulatory (inhibitor) subunitNM_002714
81247562.83PPP1R10Protein phosphatase 1, regulatory (inhibitor) subunitNM_002714
81783582.83PPP1R10Protein phosphatase 1, regulatory (inhibitor) subunitNM_002714
81796642.83PPP1R10Protein phosphatase 1, regulatory (inhibitor) subunitNM_002714
81127312.77F2RL2Coagulation factor II (thrombin) receptor-like 2NM_004101
80436872.74ANKRD36Ankryin repeat domain 36NM_001164315
79772732.74ADSSL1Adenylosuccinate synthase like 1NM_152328
79532002.72CCND2Cyclin D2NM_001759
80579542.72C2prf66Chromosome 2 open reading frame 66AY358249
80495322.72LRRFIP1Leucine-rich repeat (in FLII) interacting protein 1NM_001137550
81418432.72RASA4RAS p21 protein activator 4NM_006989
80987522.72ABCA11PAdenosine triphosphate-binding cassette, sub-family A, member 11, pseudogeneNR_002451
79820462.70SNORD115-20Small nucleolar RNA, C/D box 115-20NR_003312
79820162.70SNORD115-12Small nucleolar RNA, C/D box 115-12NR_003304
79820242.70SNORD115-12Small nucleolar RNA, C/D box 115-12NR_003304
79820302.70SNORD115-12Small nucleolar RNA, C/D box 115-12NR_003304
80540642.69ANKRD36BAnkyrin repeat domain 36BNM_025190
79100472.68DNAH14Dynein, axonemal, heavy chain 14NM_001373
80162392.68PLEKHM1Pleckstrin homology domain-containing, family M (with RUN domain) member 1NR_027774
80609492.67ANKRD5Ankyrin repeat domain 5NM_022096
80643752.62SRXN1Sulfiredoxin 1 homolog (S. Cerevisiae)NM_080725
80776122.61TTLL3Tubulin tyrosine ligase-like family, member 3NM_001025930
81515592.60SLC10A5Solute carrier family 10 (sodium/bile acid cotransporter family), member 5NM_001010893
79022272.60GADD45AGrowth arrest and DNA-damage-inducible, αNM_001924
79820582.59SNORD115-26Small nucleolar RNA, C/D box 115-26NR_003343
81180232.55GTF2H4General transcription factor II human, polypeptide 4NM_001517
80063362.54LRRC37BLeucine-rich repeat-containing 37BNM_052888
81080062.51LEAP2Liver-expressed antimicrobial peptide 2NM_052971
79713882.50SLC25A30Solute carrier family 25, member 30NM_001010875
79871632.48FMN1Formin 1 ENST00000414268
79382932.47SNORA45Small nucleolar RNA, H/ACA box 45NR_002977
81136512.45ATG12ATG12 autophagy-related 12 homolog (S. Cerevisiae)NR_033362
80495422.45LRRFIP1Leucine-rich repeat (in FLII) interacting protein 1NM_001137550
80974352.45C4orf33Chromosome 4 open reading frame 33NM_173487
81683452.45ACRCAcidic repeat-containingNM_052957
79808282.42CCDC88CCoiled-coil domain-containing 88CNM_001080414
80454232.42SNORA40Small nucleolar RNA, H/ACA box 40NR_002973
81123312.42ISCA1Iron-sulfur cluster assembly 1 homologNM_030940
80436972.41ANKRD36BAnkyrin repeat domain 36BNM_025190
80336672.40ZNF558Zinc finger protein 558NM_144693
81422322.39LAMB4Laminin, β4NM_007356
79600522.39SNORA49Small nuclear RNA, H/ACA box 49NR_002979
80318372.39ZNF587Zinc finger protein 587AF294842
81747152.38SNORA69Small nuclear RNA, H/ACA box 69NR_002584
80017482.38SNORA50Small nuclear RNA, H/ACA box 50NR_002980
80425032.38MXD1MAX dimerization protein 1NM_002357
80726782.36HMOX1Heme oxygenase (decycling) 1NM_002133
79979042.35ZNF778Zinc finger protein 778AK295122
80536482.35KRCC1Lysine-rich coiled-coil 1NM_016618
80357932.35ZNF737Zinc finger protein 737NM_001159293
79777322.34SNORD8Small nuclear RNA, C/D box 8NR_002916
81534572.31EEF1DEukaryotic translation elongation factor 1δAY358690
80695742.31C21orf91Chromosome 21 open reading frame 91NM_001100420
81128412.30HOMER1Homer homolog 1 (Drosophila)NM_004272
80389192.29ZNF350Zinc finger protein 50NM_021632
91752882.29MOSPD1Motile sperm domain-containing 1NM_019556
81609122.28C9orf131Chromosome 9 open reading frame 131NM_203299
79343342.28TTC18Tetratricopeptide repeat domain 18NM_145170
80565722.27SPC25SPC25, NDC80 kinetochore complex componentNM_020675
81619192.27TLE1Transducin-like enhancer of split 1 (Drosophila)NM_005077
79644602.26DDIT3 DNA-damage-inducible transcript 3NM_004083
80198572.26NDC80NDC80 Homolog, kinetochore complex component (S. cerevisiae)NM_006101
80455872.24ACVR2AActivin A receptor, type IIANM_001616
80026602.24TXNL4BThioredoxin-like 4BNM_017853
79113292.2414-SepSeptin 14NM_207366
80809802.24FLJ10213Endogenous Borna-like N element-1NM_018029
80141152.22MYOIDMyosin IDNM_015194
79499162.20CHKACholine kinase αNM_001277
79382952.20RPL27ARibosomal protein L27aNM_000990
81681462.20KIF4AKinesin family member 4ANM_012310
81141712.19CDKL3Cycline-dependent kinase-like 3NM_001113575
80087002.19FLJ11710Hypothetical protein FLJ11710AK021772
81083212.18FAM53CFamily with sequence similarity 53, member CNM_001135647
80471612.18OBFC2A Oligonucleotide/oligosaccharide-binding fold-containing 2aNM_001031716
80535762.17RNF103Ring finger protein 103NM_005667
80062372.17LOC400590Hypothetical LOC400590 ENST00000433145
81363412.17BPGM 2,3-bisphosphoglycerate mutaseNM_199186
81462252.16C8orf40Chromosome 8 open reading frame 40NM_001135674
81470572.16CHMP4CChromatin modifying protein 4CNM_152284
79212282.15ETV3E26 transforming-specific variant 3NM_001145312
80656072.15PLAGL2Pleiomorphic adenoma gene-like 2NM_002657
80965112.14BMPR1BBone morphogenetic protein receptor, type 1BNM_001203
79273892.14MAPK8Mitogen-activated protein kinase 8NM_002750
80989582.14POLNPolymerase (DNA directed) nuNM_181808
80389892.14ZNF600Zinc finger protein 600NM_198457
79516542.14FDXACB1Ferredoxin-fold anticodon binding domain-containing 1NM_138378
80363412.13ZNF461Zinc finger protein 461NM_153257
79819982.13SNORD116-25Small nucleolar RNA, C/D box 116-25NM_003339
80411792.13CLIP4 Cytoskeleton-associated protein-glycine-rich domain-containing linker protein family member 4NM_024692
79690962.13CDADC1Cytidine and deoxycytidine monophosphate deaminase domain-containing 1NM_030911
79692432.13CKAP2 Cytoskeleton-associated protein 2NM_018204
79863502.12ARRDC4Arrestin domain-containing 4NM_183376
80633822.12SNAI1Snail homolog 1 (Drosophila)NM_005985
80538012.12ANKRD36Ankyrin repeat domain 36NM_001164315
79995882.12PLA2G10Phospholipase A2, group XNM_003561
80087952.11C17orf71Chromosome 17 open reading frame 71NM_018149
80293402.11ZNF155Zinc finger protein 155NM_003445
81661042.11OFD1Oral-facial-digital syndrome 1NM_003611
81238252.11SLC35B3Solute carrier family 35 member B3NM_015948
79010522.11SNORD38BSmall nucleolar RNA, C/D box 38BNM_001457
80848802.10HES1Hairy and enhancer of split 1NM_005524
79256722.10ZNF670Zinc finger protein 670NM_033213
79822942.10OTUD7AOTU domain-containing 7ANM_130901
79621122.09CAPRIN2Caprin family member 2NM_001002259
79739482.09BRMSILBreast cancer metastasis-suppressor 1-likeNM_032352
81176852.09ZKSCAN3Zinc finger with KRAB and SCAN domains 3NM_024493
80100822.08SNORD1ASmall nucleolar RNA, C/D box 1ANR_004395
80419822.08ACYP2Acylphosphatase 2, muscle typeNM_138448
81376932.08COX19Cytochrome c oxidase assembly homolog 19NM_001031617
79177792.08GCLMGlutamate-cysteine ligase, modifier subunitNM_002061
79383642.08WEE1WEE1 homolog 1 (S. Pombe)BX641032
80074142.08AOC2Amine oxidase, copper-containing 2 (retina-specific)NM_009590
81396562.08GRB10Growth factor receptor-bound protein 10NM_001001555
80598522.08MSL3L2/MSL3-like 2Male-specific lethal 3-like 2 (Drosophila)NM_001166217
81094842.07KIF4BKinesin family member 4BNM_001099293
80225592.07ANKRD29Ankyrin repeat domain 29NM_173505
79100302.07DNAH14Dynein, axonemal, heavy chain 14NM_00145154
80521432.07GPR75G protein-coupled receptor 75NM_006794
79316432.07CYP2E1Cytochrome P450, family 2, subfamily E, polypeptide 1NM_000773
81027892.06TERF1Telomeric repeat binding factor (NIMA-interacting) 1NM_003218
79536032.06C1SComplement component 1, s subcomponentNM_201442
81049302.05SLC1A3Solute carrier family 1 (glial high affinity glutamate transporter), member 3NM_004172
79532112.05C12orf5Chromosome 12 open reading frame 5NM_020375
81143262.04FAM13BFamily with sequence similarity 13, member BNM_0166603
79368262.04IKZF5IKAROS family zinc finger (Pegasus)NM_022466
80135672.04C17orf108Chromosome 17 open reading frame 108NM_001076680
79750662.04AKAP5A-kinase anchor protein 5NM_004857
81425242.04TSPAN12Tetraspanin 12NM_012338
79526732.04FLJ45950FLJ45950 proteinAK127847
80811282.04NSUN3Nucleolar protein 2 homolog/Sun domain family, member 3NM_022072
79228462.04FAM129AFamily with sequence similarity 129, member ANM_052966
80133052.04ZNF286BZinc finger protein 286BNM_001145045
81539352.03ZNF252Zinc finger protein 252NM_023392
81624902.03HIATL1Hippocampus abundant transcript-like 1NM_032558
81286982.02SESN1Sestrin 1NM_014454
80107782.02CSNK1DCasein kinase 1, δNM_001893
81413112.02FAM200AFamily with sequence similarity 200, member ANM_145111
79448672.02SIAESialic acid acetylesteraseNM_170601
79618292.02BCAT1Branched-chain amino-acid transaminase 1, cytosolicNM_005504
79941612.02RBBP6Etinoblastoma binding protein 6NM_006910
79812732.02CCDC85CCoiled-coil domain-containing 85CNM_001144995
81106492.02TRIM41Tripartite motif-containing 41NM_033549
81018392.01EIF4EEukaryotic translation initiation factor 4ENM_001968
81032262.01TMEM154Transmembrane protein 154NM_152680

Table II

Downregulation of gene expressions in catharidine-treated NCI-H460 cells.

Table II

Downregulation of gene expressions in catharidine-treated NCI-H460 cells.

Probe set IDFold changeGene symbolGene descriptionmRNA accession no.
8175016−3.08APLNApelinNM_017413
8124413−3.06HIST1H4DHistone cluster 1, H4dNM_003539
8105302−3.05FSTFollistatinNM_006350
7953665−3.04DPPA3Developmental pluripotency-associated 3NM_199286
8117426−2.99HIST1H2BHHistone cluster 1, H2bhNM_003524
8117898−2.97HIST1H4JHistone cluster 1, H4jNM_021968
8117337−2.95HIST1H1EHistone cluster 1, H1eNM_005321
7911241−2.93OR2L8Olfactory receptor, family 2, subfamily L, member 8NM_001001963
8048749−2.88KCNE4Potassium voltage-gated channel, IsK-related family, member 4NM_080671
8124437−2.87HIST1H3FHistone cluster 1, H3fNM_021018
8117395−2.83HIST1H2BFHistone cluster 1, H2bfNM_003522
8015798−2.79LOC100130581Hypothetical LOC100130581NR_027413
7919642−2.78HIST2H2ABHistone cluster 2, H2abNM_175065
8059470−2.76IRS1Insulin receptor substrate 1NM_005544
8077270−2.75CHL1Cell adhesion molecule with homology to L1 cell adhesion moleculeNM_006614
7915592−2.74RNU5DRNA, U5D small nuclearNR_002755
7906767−2.74FCGR2CFc fragment of immunoglobulin G, low affinity IIc, receptor for (CD32) (gene/pseudogene)NM_201563
8117594−2.74HIST1H2BMHistone cluster 1, H2bmNM_003521
8117589−2.72HIST1H3HHistone cluster 1, H3hNM_003536
7981728−2.71LOC100293211Similar to hCG2042717 ENST00000390601
8124397−2.71HIST1H1CHistone cluster 1, H1cNM_005319
8135734−2.71C7orf58Chromosome 7 open reading frame 58NM_024913
8138988−2.70DPY19L2P1Dpy-19-like 2 pseudogene 1 (C. elegans)NR_002833
8117583−2.65HIST1H2AIHistone cluster 1, H2aiNM_003509
8153258−2.65SLC45A4Solute carrier family 45, member 4BC033223
7960865−2.61SLC2A3Solute carrier family 2 (facilitated glucose transporter), member 3NM_006931
8116921−2.59EDN1Endothelin 1NM_001955
8101757−2.58GPRIN3G protein-regulated inducer of neurite outgrowth family member 3NM_198281
7971723−2.58FLJ37307Hypothetical LOC283521NR_027047
8117580−2.56HIST1H2AIHistone cluster 1, H2aiNM_003509
8167573−2.56GAGE1G antigen 1NM_001468
8165295−2.56LCN8Lipocalin 8 ENST00000371686
7927876−2.53TET1Ten-eleven translocation oncogene 1NM_030625
7963054−2.52TUBA1ATubulin, α1aNM_006009
7957386−2.51ACSS3Acyl-CoA synthetase short-chain family member 3NM_024560
7915919−2.49TAL1T cell acute lymphocytic leukemia 1NM_003189
8174985−2.48SMARCA1Switch/sucrose non-fermentable-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 1NM_003069
8105495−2.47PART1Prostate androgen-regulated transcript 1 (non-protein coding)NR_028508
8146967−2.46CRISPLD1Cysteine-rich secretory protein LCCL domain-containing 1NM_031461
8144786−2.46SLC7A2Solute carrier family 7 (cationic amino acid transporter, y+ system), member 2NM_003046
8029280−2.45CD177CD177 moleculeNM_020406
7984524−2.45PAQR5Progestin and adipoQ receptor family member VNM_001104554
7973182−2.44LOC554207Hypothetical LOC554207 ENST00000320322
8145795−2.44LOC100293539Similar to ribosomal protein 10XM_002346094
8095697−2.44CXCL1Chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, α)NM_001511
8124406−2.42HIST1H2BCHistone cluster 1, H2bcNM_003526
7978260−2.42DHRS1 Dehydrogenase/reductase family member 1NM_001136050
7920271−2.42S100A4S100 calcium binding protein A4NM_019544
8166469−2.42SAT1Spermidine/spermine N1-acetyltransferase 1NR_027783
8124527−2.40HIST1H1BHistone cluster 1, H1bNM_005322
8008321−2.40ACSF2Acyl-CoA synthetase family member 2NM_025149
8124385−2.39HIST1H4BHistone cluster 1, H4bNM_003544
800731−2.38TUBG2Tubulin, γ2NM_016437
8122365−2.36GPR126G protein-coupled receptor 126NM_020455
8015273−2.34KRT31Keratin 31NM_002277
7904465−2.34HIST2H2BAHistone cluster 2, H2baNR_027337
8124416−2.33HIST1H3DHistone cluster 1, H3dNM_003530
8074458−2.33C22orf39Chromosome 22 open reading frame 39NM_173793
8046048−2.33CSRNP3 Cysteine-serine-rich nuclear protein 3NM_001172173
8007493−2.32ARL4DAdenosine diphosphate-ribosylation factor-like 4DNM_001661
8157804−2.31OLFML2AOlfactomedin-like 2ANM_182487
7948836−2.31TMEM223Transmembrane protein 223NM_001080501
8033458−2.31LYPLA2Lysophospholipase IINM_007260
7919619−2.30HIST2H2AA3histone cluster 2, H2aa3NM_003516
7905079−2.30HIST2H2AA3Histone cluster 2, H2aa3NM_003516
7927631−2.29DKK1Dickkopf homolog 1 (Xenopus laevis)NM_012242
7975598−2.28ACOT1Acyl-CoA thioesterase 1NM_001037161
8071801−2.27GSTTP1Glutathione S-transferase θ pseudogene 1NR_003081
7928429−2.27PLAUPlasminogen activator, urokinaseNM_002658
8068898−2.27HIST1H2BKHistone cluster 1, H2bkNM_080593
8041467−2.26VITVitrinNM_053276
8077160−2.26ARSAArylsulfatase ANM_000487
7991754−2.25HBZHemoglobin, ζNM_005332
8049534−2.24LRRFIP1Leucine-rich repeat (in FLII) interacting protein 1NM_001137550
7909789−2.23TGFB2Transforming growth factor β2NM_001135599
7919612−2.23HIST2H3DHistone cluster 2, H3dNM_001123375
8100578−2.22EPHA5Ephrin receptor A5NM_004439
8169541−2.22DOCK11Dedicator of cytokinesis 11NM_144658
8124430−2.21HIST1H1DHistone cluster 1, H1dNM_005320
8124524−2.21HIST1H2AKHistone cluster 1, H2akNM_003510
8124524−2.21HIST1H2AKHistone cluster 1, H2akNM_003510
7906775−2.20HSPA7Heat shock 70kDa protein 7 (HSP70B)NR_024151
7953291−2.20CD9CD9 moleculeNM_001769
8033319−2.19SH2D3ASrc Homology 2 domain-containing 3ANM_005490
7905088−2.19HIST2H2ACHistone cluster 2, H2acNM_003517
7975602−2.19ACOT2Acyl-CoA thioesterase 2NM_006821
7982854−2.19DLL4δ-like 4 (Drosophila)NM_019074
8019778−2.19PCYT2Phosphate cytidylyltransferase 2, ethanolamineNM_002861
8046048−2.19HIST1H4CHistone cluster 1, H4cNM_003542
8007493−2.18VWA5AVon Willebrand factor A domain-containing 5ANM_001130142
8157804−2.18PLGPlasminogenNM_000301
7948836−2.18CD24CD24 moleculeNM_013230
8033458−2.17FSTL4Follistatin-like 4NM_015082
7919619−2.16CA2Carbonic anhydrase IINM_000067
7905079−2.16CDH19Cadherin 19, type 2NM_021153
7927631−2.16CDC42EP3Cell division cycle 42 effector protein (Rho guanosine triphosphatase binding) 3NM_006449
7975598−2.16ACCN2Amiloride-sensitive cation channel 2, neuronalNM_020039
8071801−2.15HIST2H3DHistone cluster 2, H3dNM_001123375
7928429−2.15RFX2Regulatory factor X, 2 (influences human leukocyte antigen class II expression)NM_000635
8068898−2.15NESNestinNM_006617
8041467−2.15LOC25845Hypothetical LOC25845NR_024158
8077160−2.15THSD7AThrombospondin, type I, domain-containing 7ANM_015204
7991754−2.14LOC147727Hypothetical LOC147727NR_024333
8049534−2.14CALML6Calmodulin-like 6NM_138705
7909789−2.14DEFB109P1BDefensin, β 109, pseudogene 1BNR_003668
7919612−2.13EPORErythropoietin receptorNM_000121
8100578−2.13EEF2KEukaryotic elongation factor-2 kinaseNM_013302
8169541−2.13EMP3Epithelial membrane protein 3NM_001425
8124430−2.13TMEM84Transmembrane protein 84NR_026949
8124524−2.13CXXC5CXXC finger 5NM_016463
7906775−2.12PCYT2Phosphate cytidylyltransferase 2, ethanolamineNM_002861
7953291−2.12LYPD1Ly6/plasminogen activator, urokinase 1 receptor domain-containingNM_144586
8033319−2.12PHLDB2Pleckstrin homology-like domain, family B, member 2NM_001134439
7905088−2.11LRFN2Leucine-rich repeat and fibronectin type III domain-containing 2NM_020737
7975602−2.11C9orf23Chromosome 9 open reading frame 23NM_148179
7982854−2.11FLJ13744Hypothetical FLJ13744BC070061
8018445−2.11UNKUnkempt homolog (Drosophila)NM_001080419
8038407−2.10RRASRelated RAS viral (r-ras) oncogene homologNM_006270
7987230−2.10LPCAT4 Lysophosphatidylcholine acyltransferase 4NM_153613
8031514−2.10LOC100133142Hypothetical LOC100133142XM_001718400
8130374−2.10FBXO5F-box protein 5NM_012177
7908409−2.09RGS2Regulator of G-protein signaling 2NM_002923
8111255−2.09CDH10Cadherin 10, type 2 (T2-cadherin)NM_006727
7965335−2.09DUSP6Dual specificity phosphatase 6NM_001946
8065537−2.09LOC100134868Hypothetical LOC100134868NR_004846
8138466−2.08MACC1Metastasis associated in colon cancer 1NM_182762
7902687−2.08CYR61Cysteine-rich, angiogenic inducer, 61NM_001554
8036136−2.08TMEM149Transmembrane protein 149NM_024660
8098916−2.08TMEM129Transmembrane protein 129NM_001127266
7955663−2.07TENC1Tensin-like C1 domain-containing phosphatase (tensin 2)NM_170754
7939897−2.07FOLH1Folate hydrolase (prostate-specific membrane antigen) 1NM_004476
7920191−2.07LCE3ALate cornified envelope 3ANM_178431
7951437−2.06GUCY1A2Guanylate cyclase 1, soluble, α2NM_000855
8022653−2.06LOC728606Hypothetical LOC728606NR_024259
7929816−2.06SCDStearoyl-CoA desaturase (δ-9-desaturase)NM_005063
7940565−2.06FADS2Fatty acid desaturase 2NM_004265
7951157−2.06CCDC82Coiled-coil domain-containing 82AK313893
7936100−2.06CALHM2Calcium homeostasis modulator 2NM_015916
7954090−2.06EMP1Epithelial membrane protein 1NM_001423
8005951−2.05SNORD42BSmall nucleolar RNA, C/D box 42BNR_000013
8148917−2.05MFSD3Major facilitator superfamily domain-containing 3NM_138431
7937990−2.04HBG1Hemoglobin, γANM_000559
7937993−2.04HBG2Hemoglobin, γGNM_000184
8033233−2.04TUBB4Tubulin, β4NM_006087
8048350−2.04PLCD4Phospholipase C, δ4NM_032726
8037408−2.04KCNN4Potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4NM_002250
7964119−2.04STAT2Signal transducer and activator of transcription 2NM_005419
8016841−2.03TMEM100Transmembrane protein 100NM_001099640
7958948−2.03DDX54DEAD box polypeptide 54NM_0011111322
8151512−2.02PAG1Phosphoprotein associated with glycosphingolipid microdomains 1NM_018440
8005549−2.02GRAPLGrowth factor receptor-bound protein 2-related adaptor protein-likeNM_001129778
8033159−2.02PSPNPersephinNM_004158
7986639−2.02VSIG6V-set and immunoglobulin domain-containing 6 ENST00000338567
7938741−2.01MRGPRX3MAS-related G protein coupled receptor, member X3NM_054031
8047174−2.01SLC39A10Solute carrier family 39 (zinc transporter), member 10NM_001127257

[i] Acyl-CoA, acyl coenzyme A; DEAD, (Asp-Glu-Ala-Asp).

GeneGo analysis

A GeneGo analysis program was used to analyze the CTD-treated NCI-H460 cells in order to determine the top scoring genes which were differentially expressed, as determined by the number of pathway networks involved. The results of the GeneGo analyses are shown in Figs. 1Figure 23, which reveal the top, second and third scored genes by the number of pathways, respectively. Experimental data were used to generate maps of the pathway interactions and genes which were upregulated (indicated by red circles) and down-regulated (indicated by blue circles) in H460 cells following treatment with CTD. It was indicated that these genes may also be involved in DNA damage, cell cycle arrest and apop-tosis-associated responses in CTD-treated H460 cells.

Discussion

CTD has been reported to have cytotoxic effects in numerous different types of cancer cell (815). The results of previous studies have also demonstrated that CTD-induced cell death occurred due to the induction of apoptosis in human lung cancer cells (data not shown) (23). However, the effects of CTD on gene expression in cancer cells have remained to be elucdiated. To the best of our knowledge, the present study was the first to report on the effects of CTD on gene expression in H460 cells. Therefore, the present study not only advanced the understanding of the differential gene expression following treatment with CTD in lung cancer cells, but may additionally provide several potential biomarkers for use as future therapeutic clinical targets for the treatment of lung cancer.

It has been well documented that the tumor microenvironment, which contains matrix proteins, stromal cells and associated secreted molecules, including cytokines and associated genes, which may be used as targets of cancer therapeutic drugs (2426). Therefore, an increasing number of studies focus on elucidating the tumor microenvironment and associated gene expression in order to determine potential novel therapeutic agents for treating cancer patients (27). Over the past decade, there have been numerous clinical trials of treatments for lung cancer patients, including adjuvant chemotherapy trials and neo-adjuvant chemotherapy trials (2830); however, the results of these trials have not yet provided a successful, effective treatment for lung cancer. Numerous studies have demonstrated that chemotherapeutics may result in cell death through DNA damage, cell cycle arrest and the induction of apoptosis (31,32). In the present study, H460 cells were treated with CTD and incubated in 12-well plates, and their RNA was then isolated in order to determine which genes exhibited altered expression following treatment with CTD. The results revealed that CTD effected the upregulation and downregulation, respectively, of the expression of certain genes which are known to be associated with DNA damage, cell cycle progression and apoptosis in H460 cells.

In order to further elucidate the molecular signaling pathways associated with altered gene expression in H460 cells following exposure to CTD, GeneGo Process Networks were used in the present study in order to analyze the altered gene expression results of the microarray, in order to determine the possible signaling pathways involved. Based on GeneGo pathway and canonical pathway maps, which represent a set of ~650 signaling and metabolic maps covering human biology (signaling and metabolism) in a comprehensive way. A preset network of protein interaction characteristics for the process was used for each process, and the experimental data were mapped regarding the specific process. The obtained hypothetical molecular signaling pathways indicated that CTD affects numerous associated signaling pathways, indicated by the involvement of the differentially expressed genes in the network of the respective the signaling pathways. The gene content of the uploaded files was used as the input list for the generation of biological networks using the Analyze Networks algorithm with default settings. This is a variant of the shortest paths algorithm, with main parameters of relative enrichment with the uploaded data, and relative saturation of the networks with canonical pathways. The network provides data listing interacting proteins. In this workflow the network is prioritized based on the number of fragments of canonical pathways on the network.

In conclusion, the results of the present study revealed that treatment with CTD induced the upregulation and downregulation of numerous genes in H460 cells. In addition, these differentially expressed genes were associated with DNA damage, cell cycle progression and apoptotic cell death in human lung cancer H460 cells. The present study also revealed possible signaling pathways, which may provide more information on the possible mechanism of CTD in H460 cells; however, further studies are required.

Acknowledgments

The present study was supported by a grant from China Medical University [grant no. MU 101-AWARD-03(1/2); Taichung, Taiwan].

References

1 

Bilello KS, Murin S and Matthay RA: Epidemiology, etiology, and prevention of lung cancer. Clin Chest Med. 23:1–25. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Zhang P, Gao WY, Turner S and Ducatman BS: Gleevec (STI-571) inhibits lung cancer cell growth (A549) and potentiates the cisplatin effect in vitro. Mol Cancer. 2:12003. View Article : Google Scholar : PubMed/NCBI

4 

Midthun DE and Jett JR: Chemotherapy for advanced lung cancer. When to expect a response. Postgrad Med. 101:187–194. 1997. View Article : Google Scholar : PubMed/NCBI

5 

Stewart DJ: Tumor and host factors that may limit efficacy of chemotherapy in non-small cell and small cell lung cancer. Crit Rev Oncol Hematol. 75:173–234. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Chansky K, Sculier JP, Crowley JJ, et al: International Staging Committee and Participating Institutions: The International Association for the Study of Lung Cancer Staging Project: prognostic factors and pathologic TNM stage in surgically managed non-small cell lung cancer. J Thorac Oncol. 4:792–801. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Zhang W, Ma YZ, Song L, Wang CH, Qi TG and Shao GR: Effect of cantharidins in chemotherapy for hepatoma: A retrospective cohort study. Am J Chin Med. 42:561–567. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Dorn DC, Kou CA, Png KJ and Moore MA: The effect of cantharidins on leukemic stem cells. Int J Cancer. 124:2186–2199. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Huh JE, Kang KS, Chae C, Kim HM, Ahn KS and Kim SH: Roles of p38 and JNK mitogen-activated protein kinase pathways during cantharidin-induced apoptosis in U937 cells. Biochem Pharmacol. 67:1811–1818. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Li W, Xie L, Chen Z, et al: Cantharidin, a potent and selective PP2A inhibitor, induces an oxidative stress-independent growth inhibition of pancreatic cancer cells through G2/M cell-cycle arrest and apoptosis. Cancer Sci. 101:1226–1233. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Yeh CB, Su CJ, Hwang JM and Chou MC: Therapeutic effects of cantharidin analogues without bridging ether oxygen on human hepatocellular carcinoma cells. Eur J Med Chem. 45:3981–3985. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Chang C, Zhu YQ, Mei JJ, Liu SQ and Luo J: Involvement of mitochondrial pathway in NCTD-induced cytotoxicity in human hepG2 cells. J Exp Clin Cancer Res. 29:1452010. View Article : Google Scholar : PubMed/NCBI

13 

Huang WW, Ko SW, Tsai HY, et al: Cantharidin induces G2/M phase arrest and apoptosis in human colorectal cancer colo 205 cells through inhibition of CDK1 activity and caspase-dependent signaling pathways. Int J Oncol. 38:1067–1073. 2011.PubMed/NCBI

14 

Zhang WD, Zhao HR, Yan Y, Wang XH, Zong ZH and Liu Y: Apoptosis induced by cantharidin in human pulmonary carcinoma cells A549 and its molecular mechanisms. Zhonghua Zhong Liu Za Zhi. 27:330–334. 2005.In Chinese. PubMed/NCBI

15 

Kim JA, Kim Y, Kwon BM and Han DC: The natural compound cantharidin induces cancer cell death through inhibition of heat shock protein 70 (HSP70) and Bcl-2-associated athanogene domain 3 (BAG3) expression by blocking heat shock factor 1 (HSF1) binding to promoters. J Biol Chem. 288:28713–28726. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Osborne C, Wilson P and Tripathy D: Oncogenes and tumor suppressor genes in breast cancer: potential diagnostic and therapeutic applications. Oncologist. 9:361–377. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Heneghan HM, Miller N and Kerin MJ: MiRNAs as biomarkers and therapeutic targets in cancer. Curr Opin Pharmacol. 10:543–550. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Weinstein IB: Cancer. Addiction to oncogenes – the Achilles heal of cancer. Science. 297:63–64. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Luo J, Solimini NL and Elledge SJ: Principles of cancer therapy: oncogene and non-oncogene addiction. Cell. 136:823–837. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Solimini NL, Luo J and Elledge SJ: Non-oncogene addiction and the stress phenotype of cancer cells. Cell. 130:986–988. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Méndez M, Custodio A and Provencio M: New molecular targeted therapies for advanced non-small cell lung cancer. J Thorac Dis. 3:30–56. 2011.

22 

Chang YM, Velmurugan BK, Kuo WW, et al: Inhibitory effect of alpinate Oxyphyllae fructus extracts on Ang II-induced cardiac pathological remodeling-related pathways in H9c2 cardio-myoblast cells. BioMedicine. 3:148–152. 2013. View Article : Google Scholar

23 

Hsia TC, Yu CC, Hsu SC, et al: Cantharidin induces apoptosis of H460 human lung cancer cells through mitochondria-dependent pathways. Int J Oncol. 45:245–254. 2014.PubMed/NCBI

24 

Ayala F, Dewar R, Kieran M and Kalluri R: Contribution of bone microenvironment to leukemogenesis and leukemia progression. Leukemia. 23:2233–2241. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Straussman R, Morikawa T, Shee K, et al: Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 487:500–504. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Wilson TR, Fridlyand J, Yan Y, et al: Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 487:505–509. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Moniri MR, Dai LJ and Warnock GL: The challenge of pancreatic cancer therapy and novel treatment strategy using engineered mesenchynmal stem cells. Cancer Gene Ther. 21:12–23. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Pignon JP, Tribodet H, Sagliotti GV, et al: LACE Collaborative Group: Lung adjuvant cisplatin evaluation: A pooled analysis by the LACE Collaborative Group. J Clin Oncol. 26:3552–3559. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Pepe C, Hasan B, Winton TL, et al National Cancer Institute of Canada and Intergroup Study FBR.10: Adjuvant vinorelbine and cisplatin in elderly patients: National Cancer Institute of Canada and Intergroup Study JBR.10. J Clin Oncol. 25:1553–1561. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Glynne-Jones R and Hoskin P: Neoadjuvant cisplatin chemotherapy before chemoradiation: A flawed paradigm? J Clin Oncol. 25:5281–5286. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Ricci MS and Zong WX: Chemotherapeutic approaches for targeting cell death pathways. Oncologist. 11:342–357. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Tan TT and White E: Therapeutic targeting of death pathways in cancer: Mechanisms for activating cell death in cancer cells. Adv Exp Med Biol. 615:81–104. 2008.PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 12 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hsia TC, Yu CC, Hsu SC, Tang NY, Lu HF, Yu CS, Wu SH, Lin JG and Chung JG: cDNA microarray analysis of the effect of cantharidin on DNA damage, cell cycle and apoptosis-associated gene expression in NCI-H460 human lung cancer cells in vitro. Mol Med Rep 12: 1030-1042, 2015
APA
Hsia, T., Yu, C., Hsu, S., Tang, N., Lu, H., Yu, C. ... Chung, J. (2015). cDNA microarray analysis of the effect of cantharidin on DNA damage, cell cycle and apoptosis-associated gene expression in NCI-H460 human lung cancer cells in vitro. Molecular Medicine Reports, 12, 1030-1042. https://doi.org/10.3892/mmr.2015.3538
MLA
Hsia, T., Yu, C., Hsu, S., Tang, N., Lu, H., Yu, C., Wu, S., Lin, J., Chung, J."cDNA microarray analysis of the effect of cantharidin on DNA damage, cell cycle and apoptosis-associated gene expression in NCI-H460 human lung cancer cells in vitro". Molecular Medicine Reports 12.1 (2015): 1030-1042.
Chicago
Hsia, T., Yu, C., Hsu, S., Tang, N., Lu, H., Yu, C., Wu, S., Lin, J., Chung, J."cDNA microarray analysis of the effect of cantharidin on DNA damage, cell cycle and apoptosis-associated gene expression in NCI-H460 human lung cancer cells in vitro". Molecular Medicine Reports 12, no. 1 (2015): 1030-1042. https://doi.org/10.3892/mmr.2015.3538