Skip to main content
Molecular Medicine Reports logoLink to Molecular Medicine Reports
. 2015 Mar 24;12(1):1030–1042. doi: 10.3892/mmr.2015.3538

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

TE-CHUN HSIA 1,2, CHIEN-CHIH YU 3, SHU-CHUN HSU 4, NOU-YING TANG 1, HSU-FENG LU 5, CHUN-SHU YU 3, SHIN-HWAR WU 6, JAUNG-GENG LIN 1,, JING-GUNG CHUNG 4,7,
PMCID: PMC4438957  PMID: 25815777

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.

Keywords: cantharidin, H460 cells, DNA damage, cell cycle, apoptosis, in vitro

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.

Probe set ID Fold change Gene symbol Gene description mRNA accession no.
8108370 16.50 EGR1 Early growth response 1 NM_001964
8012949 13.05 CDRT1 CMT1A duplicated region transcript 1 NM_006382
8012951 10.85 CDRT1 CMT1A duplicated region transcript 1 NM_006382
7916609 8.05 JUN Jun oncogene NM_002228
7977075 6.45 SNORA28 Small nucleolar RNA, H/ACA box 28 NR_002964
8041168 5.54 SNORD53 Small nucleolar RNA, C/D box 53 NR_002741
7982084 5.06 SNORD115-11 Small nucleolar RNA, C/D box 115-11 NR_003303
8097991 4.27 TDO2 Tryptophan 2,3-dioxygenase NM_005651
8114468 3.99 SNORD63 Small nucleolar RNA, C/D box 63 NR_002913
8158862 3.87 SNORD62A Small nucleolar RNA, C/D box 62A NR_002914
8158864 3.87 SNORD62A Small nucleolar RNA, C/D box 62A NR_002914
8007420 3.85 AOC3 Amine oxidase, copper-containing 3 (vascular adhesion protein 1) NM_003734
7975779 3.81 FOS FBJ murine osteosarcoma viral oncogene homolog NM_005252
8001746 3.74 SNORA46 Small nucleolar RNA, H/ACA box 46 NR_002978
7914322 3.70 SNORD103A Small nucleolar RNA, C/D box 103A NR_004054
7914324 3.70 SNORD103A Small nucleolar RNA, C/D box 103A NR_004054
793342 3.69 PTPN20A Protein tyrosine phosphatase, non-receptor type 2 NR_001042389
7918467 3.65 Clorf103 Chromosome 1 open reading frame 103 NM_018372
8053797 3.60 LOC400986 Protein immuno-reactive with anti-parathyroid hormone polyclona ENST00000456556
8156848 3.59 NR4A3 Nuclear receptor subfamily 4, group A, member 3 NM_006981
8005483 3.56 FBXW10 F-box and WD repeat domain-containing 10 NM_031456
8105077 3.54 CARD6 Caspase recruitment domain family, member 6 NM_032587
8139840 3.48 ERV3 Endogenous retroviral sequence 3 (includes zinc) NM_001007253
8030831 3.45 ZNF175 Zinc finger protein 175 NM_007147
7958200 3.45 EID3 EP300-interacting inhibitor of differentiation 3 NM_001008394
7936637 3.44 SNORA19 Small nucleolar RNA, H/ACA box 19 NR_002917
8107353 3.43 ZRSR1 Zinc finger (CCCH type), RNA-binding motif and serine/arginine rich 1 BC104811
8173600 3.26 NAP1L2 Nucleosome assembly protein 1-like 2 NM_021963
8126853 3.25 C6orf138 Chromosome 6 open reading frame 138 NM_001013732
8025301 3.20 CD209 CD209 molecule NM_021155
7923119 3.17 ZBTB41 Zinc finger and BTB domain-containing 41 NM_194314
7985555 3.11 EFTUD1 Elongation factor Tu guanine triphosphate binding domain-containing NM_024580
7952986 3.09 HSN2 Hereditary sensory neuropathy, type II NM_213655
8114572 3.08 HBEGF Heparin-binding epidermal growth factor-like growth factor NM_001945
8049540 3.05 LRRFIP1 Leucine-rich repeat (in FLII) interacting protein 1 NM_001137550
8047926 3.03 MAP2 Microtubule-associated protein 2 NM_002374
7954382 3.02 PYROXD1 Pyridine nucleotide-disulphide oxidoreductase domain 1 NM_024854
7957260 2.99 GLIPR1 GLI pathogenesis-related 1 NM_006851
8054054 2.97 ANKRD36B Ankryin repeat domain 36B NM_025190
7907572 2.96 PAPPA2 Pappalysin 2 NM_020318
8090688 2.93 SNORA58 Small nucleolar RNA, H/ACA box 58 NR_002985
8139935 2.89 TYW1B tRNA-yW synthesizing protein 1 homolog B NM_001145440
7982028 2.87 SNORD115-11 Small nucleolar RNA, C/D box 115-11 NR_003303
7982050 2.87 SNORD115-11 Small nucleolar RNA, C/D box 115-11 NR_003303
7982064 2.87 SNORD115-11 Small nucleolar RNA, C/D box 115-11 NR_003303
7982078 2.87 SNORD115-11 Small nucleolar RNA, C/D box 115-11 NR_003303
7982092 2.87 SNORD115-11 Small nucleolar RNA, C/D box 115-11 NR_003303
7905339 2.86 GABPB2 GA binding protein transcription factor, β subunit NM_144618
8140782 2.84 ABCB1 ATP-binding cassette, sub-family B (multidrug resistance protein/transporter associated with antigen processing) NM_000927
8139482 2.83 SNORA5A Small nucleolar RNA, H/ACA box 5A NR_002919
8124756 2.83 PPP1R10 Protein phosphatase 1, regulatory (inhibitor) subunit NM_002714
8124756 2.83 PPP1R10 Protein phosphatase 1, regulatory (inhibitor) subunit NM_002714
8178358 2.83 PPP1R10 Protein phosphatase 1, regulatory (inhibitor) subunit NM_002714
8179664 2.83 PPP1R10 Protein phosphatase 1, regulatory (inhibitor) subunit NM_002714
8112731 2.77 F2RL2 Coagulation factor II (thrombin) receptor-like 2 NM_004101
8043687 2.74 ANKRD36 Ankryin repeat domain 36 NM_001164315
7977273 2.74 ADSSL1 Adenylosuccinate synthase like 1 NM_152328
7953200 2.72 CCND2 Cyclin D2 NM_001759
8057954 2.72 C2prf66 Chromosome 2 open reading frame 66 AY358249
8049532 2.72 LRRFIP1 Leucine-rich repeat (in FLII) interacting protein 1 NM_001137550
8141843 2.72 RASA4 RAS p21 protein activator 4 NM_006989
8098752 2.72 ABCA11P Adenosine triphosphate-binding cassette, sub-family A, member 11, pseudogene NR_002451
7982046 2.70 SNORD115-20 Small nucleolar RNA, C/D box 115-20 NR_003312
7982016 2.70 SNORD115-12 Small nucleolar RNA, C/D box 115-12 NR_003304
7982024 2.70 SNORD115-12 Small nucleolar RNA, C/D box 115-12 NR_003304
7982030 2.70 SNORD115-12 Small nucleolar RNA, C/D box 115-12 NR_003304
8054064 2.69 ANKRD36B Ankyrin repeat domain 36B NM_025190
7910047 2.68 DNAH14 Dynein, axonemal, heavy chain 14 NM_001373
8016239 2.68 PLEKHM1 Pleckstrin homology domain-containing, family M (with RUN domain) member 1 NR_027774
8060949 2.67 ANKRD5 Ankyrin repeat domain 5 NM_022096
8064375 2.62 SRXN1 Sulfiredoxin 1 homolog (S. Cerevisiae) NM_080725
8077612 2.61 TTLL3 Tubulin tyrosine ligase-like family, member 3 NM_001025930
8151559 2.60 SLC10A5 Solute carrier family 10 (sodium/bile acid cotransporter family), member 5 NM_001010893
7902227 2.60 GADD45A Growth arrest and DNA-damage-inducible, α NM_001924
7982058 2.59 SNORD115-26 Small nucleolar RNA, C/D box 115-26 NR_003343
8118023 2.55 GTF2H4 General transcription factor II human, polypeptide 4 NM_001517
8006336 2.54 LRRC37B Leucine-rich repeat-containing 37B NM_052888
8108006 2.51 LEAP2 Liver-expressed antimicrobial peptide 2 NM_052971
7971388 2.50 SLC25A30 Solute carrier family 25, member 30 NM_001010875
7987163 2.48 FMN1 Formin 1 ENST00000414268
7938293 2.47 SNORA45 Small nucleolar RNA, H/ACA box 45 NR_002977
8113651 2.45 ATG12 ATG12 autophagy-related 12 homolog (S. Cerevisiae) NR_033362
8049542 2.45 LRRFIP1 Leucine-rich repeat (in FLII) interacting protein 1 NM_001137550
8097435 2.45 C4orf33 Chromosome 4 open reading frame 33 NM_173487
8168345 2.45 ACRC Acidic repeat-containing NM_052957
7980828 2.42 CCDC88C Coiled-coil domain-containing 88C NM_001080414
8045423 2.42 SNORA40 Small nucleolar RNA, H/ACA box 40 NR_002973
8112331 2.42 ISCA1 Iron-sulfur cluster assembly 1 homolog NM_030940
8043697 2.41 ANKRD36B Ankyrin repeat domain 36B NM_025190
8033667 2.40 ZNF558 Zinc finger protein 558 NM_144693
8142232 2.39 LAMB4 Laminin, β4 NM_007356
7960052 2.39 SNORA49 Small nuclear RNA, H/ACA box 49 NR_002979
8031837 2.39 ZNF587 Zinc finger protein 587 AF294842
8174715 2.38 SNORA69 Small nuclear RNA, H/ACA box 69 NR_002584
8001748 2.38 SNORA50 Small nuclear RNA, H/ACA box 50 NR_002980
8042503 2.38 MXD1 MAX dimerization protein 1 NM_002357
8072678 2.36 HMOX1 Heme oxygenase (decycling) 1 NM_002133
7997904 2.35 ZNF778 Zinc finger protein 778 AK295122
8053648 2.35 KRCC1 Lysine-rich coiled-coil 1 NM_016618
8035793 2.35 ZNF737 Zinc finger protein 737 NM_001159293
7977732 2.34 SNORD8 Small nuclear RNA, C/D box 8 NR_002916
8153457 2.31 EEF1D Eukaryotic translation elongation factor 1δ AY358690
8069574 2.31 C21orf91 Chromosome 21 open reading frame 91 NM_001100420
8112841 2.30 HOMER1 Homer homolog 1 (Drosophila) NM_004272
8038919 2.29 ZNF350 Zinc finger protein 50 NM_021632
9175288 2.29 MOSPD1 Motile sperm domain-containing 1 NM_019556
8160912 2.28 C9orf131 Chromosome 9 open reading frame 131 NM_203299
7934334 2.28 TTC18 Tetratricopeptide repeat domain 18 NM_145170
8056572 2.27 SPC25 SPC25, NDC80 kinetochore complex component NM_020675
8161919 2.27 TLE1 Transducin-like enhancer of split 1 (Drosophila) NM_005077
7964460 2.26 DDIT3 DNA-damage-inducible transcript 3 NM_004083
8019857 2.26 NDC80 NDC80 Homolog, kinetochore complex component (S. cerevisiae) NM_006101
8045587 2.24 ACVR2A Activin A receptor, type IIA NM_001616
8002660 2.24 TXNL4B Thioredoxin-like 4B NM_017853
7911329 2.24 14-Sep Septin 14 NM_207366
8080980 2.24 FLJ10213 Endogenous Borna-like N element-1 NM_018029
8014115 2.22 MYOID Myosin ID NM_015194
7949916 2.20 CHKA Choline kinase α NM_001277
7938295 2.20 RPL27A Ribosomal protein L27a NM_000990
8168146 2.20 KIF4A Kinesin family member 4A NM_012310
8114171 2.19 CDKL3 Cycline-dependent kinase-like 3 NM_001113575
8008700 2.19 FLJ11710 Hypothetical protein FLJ11710 AK021772
8108321 2.18 FAM53C Family with sequence similarity 53, member C NM_001135647
8047161 2.18 OBFC2A Oligonucleotide/oligosaccharide-binding fold-containing 2a NM_001031716
8053576 2.17 RNF103 Ring finger protein 103 NM_005667
8006237 2.17 LOC400590 Hypothetical LOC400590 ENST00000433145
8136341 2.17 BPGM 2,3-bisphosphoglycerate mutase NM_199186
8146225 2.16 C8orf40 Chromosome 8 open reading frame 40 NM_001135674
8147057 2.16 CHMP4C Chromatin modifying protein 4C NM_152284
7921228 2.15 ETV3 E26 transforming-specific variant 3 NM_001145312
8065607 2.15 PLAGL2 Pleiomorphic adenoma gene-like 2 NM_002657
8096511 2.14 BMPR1B Bone morphogenetic protein receptor, type 1B NM_001203
7927389 2.14 MAPK8 Mitogen-activated protein kinase 8 NM_002750
8098958 2.14 POLN Polymerase (DNA directed) nu NM_181808
8038989 2.14 ZNF600 Zinc finger protein 600 NM_198457
7951654 2.14 FDXACB1 Ferredoxin-fold anticodon binding domain-containing 1 NM_138378
8036341 2.13 ZNF461 Zinc finger protein 461 NM_153257
7981998 2.13 SNORD116-25 Small nucleolar RNA, C/D box 116-25 NM_003339
8041179 2.13 CLIP4 Cytoskeleton-associated protein-glycine-rich domain-containing linker protein family member 4 NM_024692
7969096 2.13 CDADC1 Cytidine and deoxycytidine monophosphate deaminase domain-containing 1 NM_030911
7969243 2.13 CKAP2 Cytoskeleton-associated protein 2 NM_018204
7986350 2.12 ARRDC4 Arrestin domain-containing 4 NM_183376
8063382 2.12 SNAI1 Snail homolog 1 (Drosophila) NM_005985
8053801 2.12 ANKRD36 Ankyrin repeat domain 36 NM_001164315
7999588 2.12 PLA2G10 Phospholipase A2, group X NM_003561
8008795 2.11 C17orf71 Chromosome 17 open reading frame 71 NM_018149
8029340 2.11 ZNF155 Zinc finger protein 155 NM_003445
8166104 2.11 OFD1 Oral-facial-digital syndrome 1 NM_003611
8123825 2.11 SLC35B3 Solute carrier family 35 member B3 NM_015948
7901052 2.11 SNORD38B Small nucleolar RNA, C/D box 38B NM_001457
8084880 2.10 HES1 Hairy and enhancer of split 1 NM_005524
7925672 2.10 ZNF670 Zinc finger protein 670 NM_033213
7982294 2.10 OTUD7A OTU domain-containing 7A NM_130901
7962112 2.09 CAPRIN2 Caprin family member 2 NM_001002259
7973948 2.09 BRMSIL Breast cancer metastasis-suppressor 1-like NM_032352
8117685 2.09 ZKSCAN3 Zinc finger with KRAB and SCAN domains 3 NM_024493
8010082 2.08 SNORD1A Small nucleolar RNA, C/D box 1A NR_004395
8041982 2.08 ACYP2 Acylphosphatase 2, muscle type NM_138448
8137693 2.08 COX19 Cytochrome c oxidase assembly homolog 19 NM_001031617
7917779 2.08 GCLM Glutamate-cysteine ligase, modifier subunit NM_002061
7938364 2.08 WEE1 WEE1 homolog 1 (S. Pombe) BX641032
8007414 2.08 AOC2 Amine oxidase, copper-containing 2 (retina-specific) NM_009590
8139656 2.08 GRB10 Growth factor receptor-bound protein 10 NM_001001555
8059852 2.08 MSL3L2/MSL3-like 2 Male-specific lethal 3-like 2 (Drosophila) NM_001166217
8109484 2.07 KIF4B Kinesin family member 4B NM_001099293
8022559 2.07 ANKRD29 Ankyrin repeat domain 29 NM_173505
7910030 2.07 DNAH14 Dynein, axonemal, heavy chain 14 NM_00145154
8052143 2.07 GPR75 G protein-coupled receptor 75 NM_006794
7931643 2.07 CYP2E1 Cytochrome P450, family 2, subfamily E, polypeptide 1 NM_000773
8102789 2.06 TERF1 Telomeric repeat binding factor (NIMA-interacting) 1 NM_003218
7953603 2.06 C1S Complement component 1, s subcomponent NM_201442
8104930 2.05 SLC1A3 Solute carrier family 1 (glial high affinity glutamate transporter), member 3 NM_004172
7953211 2.05 C12orf5 Chromosome 12 open reading frame 5 NM_020375
8114326 2.04 FAM13B Family with sequence similarity 13, member B NM_0166603
7936826 2.04 IKZF5 IKAROS family zinc finger (Pegasus) NM_022466
8013567 2.04 C17orf108 Chromosome 17 open reading frame 108 NM_001076680
7975066 2.04 AKAP5 A-kinase anchor protein 5 NM_004857
8142524 2.04 TSPAN12 Tetraspanin 12 NM_012338
7952673 2.04 FLJ45950 FLJ45950 protein AK127847
8081128 2.04 NSUN3 Nucleolar protein 2 homolog/Sun domain family, member 3 NM_022072
7922846 2.04 FAM129A Family with sequence similarity 129, member A NM_052966
8013305 2.04 ZNF286B Zinc finger protein 286B NM_001145045
8153935 2.03 ZNF252 Zinc finger protein 252 NM_023392
8162490 2.03 HIATL1 Hippocampus abundant transcript-like 1 NM_032558
8128698 2.02 SESN1 Sestrin 1 NM_014454
8010778 2.02 CSNK1D Casein kinase 1, δ NM_001893
8141311 2.02 FAM200A Family with sequence similarity 200, member A NM_145111
7944867 2.02 SIAE Sialic acid acetylesterase NM_170601
7961829 2.02 BCAT1 Branched-chain amino-acid transaminase 1, cytosolic NM_005504
7994161 2.02 RBBP6 Etinoblastoma binding protein 6 NM_006910
7981273 2.02 CCDC85C Coiled-coil domain-containing 85C NM_001144995
8110649 2.02 TRIM41 Tripartite motif-containing 41 NM_033549
8101839 2.01 EIF4E Eukaryotic translation initiation factor 4E NM_001968
8103226 2.01 TMEM154 Transmembrane protein 154 NM_152680

Table II.

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

Probe set ID Fold change Gene symbol Gene description mRNA accession no.
8175016 −3.08 APLN Apelin NM_017413
8124413 −3.06 HIST1H4D Histone cluster 1, H4d NM_003539
8105302 −3.05 FST Follistatin NM_006350
7953665 −3.04 DPPA3 Developmental pluripotency-associated 3 NM_199286
8117426 −2.99 HIST1H2BH Histone cluster 1, H2bh NM_003524
8117898 −2.97 HIST1H4J Histone cluster 1, H4j NM_021968
8117337 −2.95 HIST1H1E Histone cluster 1, H1e NM_005321
7911241 −2.93 OR2L8 Olfactory receptor, family 2, subfamily L, member 8 NM_001001963
8048749 −2.88 KCNE4 Potassium voltage-gated channel, IsK-related family, member 4 NM_080671
8124437 −2.87 HIST1H3F Histone cluster 1, H3f NM_021018
8117395 −2.83 HIST1H2BF Histone cluster 1, H2bf NM_003522
8015798 −2.79 LOC100130581 Hypothetical LOC100130581 NR_027413
7919642 −2.78 HIST2H2AB Histone cluster 2, H2ab NM_175065
8059470 −2.76 IRS1 Insulin receptor substrate 1 NM_005544
8077270 −2.75 CHL1 Cell adhesion molecule with homology to L1 cell adhesion molecule NM_006614
7915592 −2.74 RNU5D RNA, U5D small nuclear NR_002755
7906767 −2.74 FCGR2C Fc fragment of immunoglobulin G, low affinity IIc, receptor for (CD32) (gene/pseudogene) NM_201563
8117594 −2.74 HIST1H2BM Histone cluster 1, H2bm NM_003521
8117589 −2.72 HIST1H3H Histone cluster 1, H3h NM_003536
7981728 −2.71 LOC100293211 Similar to hCG2042717 ENST00000390601
8124397 −2.71 HIST1H1C Histone cluster 1, H1c NM_005319
8135734 −2.71 C7orf58 Chromosome 7 open reading frame 58 NM_024913
8138988 −2.70 DPY19L2P1 Dpy-19-like 2 pseudogene 1 (C. elegans) NR_002833
8117583 −2.65 HIST1H2AI Histone cluster 1, H2ai NM_003509
8153258 −2.65 SLC45A4 Solute carrier family 45, member 4 BC033223
7960865 −2.61 SLC2A3 Solute carrier family 2 (facilitated glucose transporter), member 3 NM_006931
8116921 −2.59 EDN1 Endothelin 1 NM_001955
8101757 −2.58 GPRIN3 G protein-regulated inducer of neurite outgrowth family member 3 NM_198281
7971723 −2.58 FLJ37307 Hypothetical LOC283521 NR_027047
8117580 −2.56 HIST1H2AI Histone cluster 1, H2ai NM_003509
8167573 −2.56 GAGE1 G antigen 1 NM_001468
8165295 −2.56 LCN8 Lipocalin 8 ENST00000371686
7927876 −2.53 TET1 Ten-eleven translocation oncogene 1 NM_030625
7963054 −2.52 TUBA1A Tubulin, α1a NM_006009
7957386 −2.51 ACSS3 Acyl-CoA synthetase short-chain family member 3 NM_024560
7915919 −2.49 TAL1 T cell acute lymphocytic leukemia 1 NM_003189
8174985 −2.48 SMARCA1 Switch/sucrose non-fermentable-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 1 NM_003069
8105495 −2.47 PART1 Prostate androgen-regulated transcript 1 (non-protein coding) NR_028508
8146967 −2.46 CRISPLD1 Cysteine-rich secretory protein LCCL domain-containing 1 NM_031461
8144786 −2.46 SLC7A2 Solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 NM_003046
8029280 −2.45 CD177 CD177 molecule NM_020406
7984524 −2.45 PAQR5 Progestin and adipoQ receptor family member V NM_001104554
7973182 −2.44 LOC554207 Hypothetical LOC554207 ENST00000320322
8145795 −2.44 LOC100293539 Similar to ribosomal protein 10 XM_002346094
8095697 −2.44 CXCL1 Chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, α) NM_001511
8124406 −2.42 HIST1H2BC Histone cluster 1, H2bc NM_003526
7978260 −2.42 DHRS1 Dehydrogenase/reductase family member 1 NM_001136050
7920271 −2.42 S100A4 S100 calcium binding protein A4 NM_019544
8166469 −2.42 SAT1 Spermidine/spermine N1-acetyltransferase 1 NR_027783
8124527 −2.40 HIST1H1B Histone cluster 1, H1b NM_005322
8008321 −2.40 ACSF2 Acyl-CoA synthetase family member 2 NM_025149
8124385 −2.39 HIST1H4B Histone cluster 1, H4b NM_003544
800731 −2.38 TUBG2 Tubulin, γ2 NM_016437
8122365 −2.36 GPR126 G protein-coupled receptor 126 NM_020455
8015273 −2.34 KRT31 Keratin 31 NM_002277
7904465 −2.34 HIST2H2BA Histone cluster 2, H2ba NR_027337
8124416 −2.33 HIST1H3D Histone cluster 1, H3d NM_003530
8074458 −2.33 C22orf39 Chromosome 22 open reading frame 39 NM_173793
8046048 −2.33 CSRNP3 Cysteine-serine-rich nuclear protein 3 NM_001172173
8007493 −2.32 ARL4D Adenosine diphosphate-ribosylation factor-like 4D NM_001661
8157804 −2.31 OLFML2A Olfactomedin-like 2A NM_182487
7948836 −2.31 TMEM223 Transmembrane protein 223 NM_001080501
8033458 −2.31 LYPLA2 Lysophospholipase II NM_007260
7919619 −2.30 HIST2H2AA3 histone cluster 2, H2aa3 NM_003516
7905079 −2.30 HIST2H2AA3 Histone cluster 2, H2aa3 NM_003516
7927631 −2.29 DKK1 Dickkopf homolog 1 (Xenopus laevis) NM_012242
7975598 −2.28 ACOT1 Acyl-CoA thioesterase 1 NM_001037161
8071801 −2.27 GSTTP1 Glutathione S-transferase θ pseudogene 1 NR_003081
7928429 −2.27 PLAU Plasminogen activator, urokinase NM_002658
8068898 −2.27 HIST1H2BK Histone cluster 1, H2bk NM_080593
8041467 −2.26 VIT Vitrin NM_053276
8077160 −2.26 ARSA Arylsulfatase A NM_000487
7991754 −2.25 HBZ Hemoglobin, ζ NM_005332
8049534 −2.24 LRRFIP1 Leucine-rich repeat (in FLII) interacting protein 1 NM_001137550
7909789 −2.23 TGFB2 Transforming growth factor β2 NM_001135599
7919612 −2.23 HIST2H3D Histone cluster 2, H3d NM_001123375
8100578 −2.22 EPHA5 Ephrin receptor A5 NM_004439
8169541 −2.22 DOCK11 Dedicator of cytokinesis 11 NM_144658
8124430 −2.21 HIST1H1D Histone cluster 1, H1d NM_005320
8124524 −2.21 HIST1H2AK Histone cluster 1, H2ak NM_003510
8124524 −2.21 HIST1H2AK Histone cluster 1, H2ak NM_003510
7906775 −2.20 HSPA7 Heat shock 70kDa protein 7 (HSP70B) NR_024151
7953291 −2.20 CD9 CD9 molecule NM_001769
8033319 −2.19 SH2D3A Src Homology 2 domain-containing 3A NM_005490
7905088 −2.19 HIST2H2AC Histone cluster 2, H2ac NM_003517
7975602 −2.19 ACOT2 Acyl-CoA thioesterase 2 NM_006821
7982854 −2.19 DLL4 δ-like 4 (Drosophila) NM_019074
8019778 −2.19 PCYT2 Phosphate cytidylyltransferase 2, ethanolamine NM_002861
8046048 −2.19 HIST1H4C Histone cluster 1, H4c NM_003542
8007493 −2.18 VWA5A Von Willebrand factor A domain-containing 5A NM_001130142
8157804 −2.18 PLG Plasminogen NM_000301
7948836 −2.18 CD24 CD24 molecule NM_013230
8033458 −2.17 FSTL4 Follistatin-like 4 NM_015082
7919619 −2.16 CA2 Carbonic anhydrase II NM_000067
7905079 −2.16 CDH19 Cadherin 19, type 2 NM_021153
7927631 −2.16 CDC42EP3 Cell division cycle 42 effector protein (Rho guanosine triphosphatase binding) 3 NM_006449
7975598 −2.16 ACCN2 Amiloride-sensitive cation channel 2, neuronal NM_020039
8071801 −2.15 HIST2H3D Histone cluster 2, H3d NM_001123375
7928429 −2.15 RFX2 Regulatory factor X, 2 (influences human leukocyte antigen class II expression) NM_000635
8068898 −2.15 NES Nestin NM_006617
8041467 −2.15 LOC25845 Hypothetical LOC25845 NR_024158
8077160 −2.15 THSD7A Thrombospondin, type I, domain-containing 7A NM_015204
7991754 −2.14 LOC147727 Hypothetical LOC147727 NR_024333
8049534 −2.14 CALML6 Calmodulin-like 6 NM_138705
7909789 −2.14 DEFB109P1B Defensin, β 109, pseudogene 1B NR_003668
7919612 −2.13 EPOR Erythropoietin receptor NM_000121
8100578 −2.13 EEF2K Eukaryotic elongation factor-2 kinase NM_013302
8169541 −2.13 EMP3 Epithelial membrane protein 3 NM_001425
8124430 −2.13 TMEM84 Transmembrane protein 84 NR_026949
8124524 −2.13 CXXC5 CXXC finger 5 NM_016463
7906775 −2.12 PCYT2 Phosphate cytidylyltransferase 2, ethanolamine NM_002861
7953291 −2.12 LYPD1 Ly6/plasminogen activator, urokinase 1 receptor domain-containing NM_144586
8033319 −2.12 PHLDB2 Pleckstrin homology-like domain, family B, member 2 NM_001134439
7905088 −2.11 LRFN2 Leucine-rich repeat and fibronectin type III domain-containing 2 NM_020737
7975602 −2.11 C9orf23 Chromosome 9 open reading frame 23 NM_148179
7982854 −2.11 FLJ13744 Hypothetical FLJ13744 BC070061
8018445 −2.11 UNK Unkempt homolog (Drosophila) NM_001080419
8038407 −2.10 RRAS Related RAS viral (r-ras) oncogene homolog NM_006270
7987230 −2.10 LPCAT4 Lysophosphatidylcholine acyltransferase 4 NM_153613
8031514 −2.10 LOC100133142 Hypothetical LOC100133142 XM_001718400
8130374 −2.10 FBXO5 F-box protein 5 NM_012177
7908409 −2.09 RGS2 Regulator of G-protein signaling 2 NM_002923
8111255 −2.09 CDH10 Cadherin 10, type 2 (T2-cadherin) NM_006727
7965335 −2.09 DUSP6 Dual specificity phosphatase 6 NM_001946
8065537 −2.09 LOC100134868 Hypothetical LOC100134868 NR_004846
8138466 −2.08 MACC1 Metastasis associated in colon cancer 1 NM_182762
7902687 −2.08 CYR61 Cysteine-rich, angiogenic inducer, 61 NM_001554
8036136 −2.08 TMEM149 Transmembrane protein 149 NM_024660
8098916 −2.08 TMEM129 Transmembrane protein 129 NM_001127266
7955663 −2.07 TENC1 Tensin-like C1 domain-containing phosphatase (tensin 2) NM_170754
7939897 −2.07 FOLH1 Folate hydrolase (prostate-specific membrane antigen) 1 NM_004476
7920191 −2.07 LCE3A Late cornified envelope 3A NM_178431
7951437 −2.06 GUCY1A2 Guanylate cyclase 1, soluble, α2 NM_000855
8022653 −2.06 LOC728606 Hypothetical LOC728606 NR_024259
7929816 −2.06 SCD Stearoyl-CoA desaturase (δ-9-desaturase) NM_005063
7940565 −2.06 FADS2 Fatty acid desaturase 2 NM_004265
7951157 −2.06 CCDC82 Coiled-coil domain-containing 82 AK313893
7936100 −2.06 CALHM2 Calcium homeostasis modulator 2 NM_015916
7954090 −2.06 EMP1 Epithelial membrane protein 1 NM_001423
8005951 −2.05 SNORD42B Small nucleolar RNA, C/D box 42B NR_000013
8148917 −2.05 MFSD3 Major facilitator superfamily domain-containing 3 NM_138431
7937990 −2.04 HBG1 Hemoglobin, γA NM_000559
7937993 −2.04 HBG2 Hemoglobin, γG NM_000184
8033233 −2.04 TUBB4 Tubulin, β4 NM_006087
8048350 −2.04 PLCD4 Phospholipase C, δ4 NM_032726
8037408 −2.04 KCNN4 Potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4 NM_002250
7964119 −2.04 STAT2 Signal transducer and activator of transcription 2 NM_005419
8016841 −2.03 TMEM100 Transmembrane protein 100 NM_001099640
7958948 −2.03 DDX54 DEAD box polypeptide 54 NM_0011111322
8151512 −2.02 PAG1 Phosphoprotein associated with glycosphingolipid microdomains 1 NM_018440
8005549 −2.02 GRAPL Growth factor receptor-bound protein 2-related adaptor protein-like NM_001129778
8033159 −2.02 PSPN Persephin NM_004158
7986639 −2.02 VSIG6 V-set and immunoglobulin domain-containing 6 ENST00000338567
7938741 −2.01 MRGPRX3 MAS-related G protein coupled receptor, member X3 NM_054031
8047174 −2.01 SLC39A10 Solute carrier family 39 (zinc transporter), member 10 NM_001127257

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. 13, 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.

Figure 1.

Figure 1

Top scored (by the number of pathways) AN network, as determined using GeneGo_cat_FC2 analysis. Thick cyan lines indicate the fragments of canonical pathways. Upregulated genes are marked with red circles and downregulated genes are indicated with blue circles. FC, Fold Change 2.0; AN, Analyze Networks algorithm.

Figure 2.

Figure 2

Second scored (by the number of pathways) AN network, as determined using GeneGo_cat_FC2 analysis. Thick cyan lines indicate the fragments of canonical pathways. Upregulated genes are marked with red circles and downregulated genes are indicated with blue circles. FC, Fold Change 2.0; AN, Analyze Networks algorithm.

Figure 3.

Figure 3

Third scored (by the number of pathways) AN network, as determined using GeneGo_cat_FC2 analysis. Thick cyan lines indicate the fragments of canonical pathways. Upregulated genes are marked with red circles and downregulated genes are indicated with blue circles. FC, Fold Change 2.0; AN, Analyze Networks algorithm.

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, Matthay RA. Epidemiology, etiology, and prevention of lung cancer. Clin Chest Med. 2002;23:1–25. doi: 10.1016/S0272-5231(03)00057-1. [DOI] [PubMed] [Google Scholar]
  • 2.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 3.Zhang P, Gao WY, Turner S, Ducatman BS. Gleevec (STI-571) inhibits lung cancer cell growth (A549) and potentiates the cisplatin effect in vitro. Mol Cancer. 2003;2:1. doi: 10.1186/1476-4598-2-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Midthun DE, Jett JR. Chemotherapy for advanced lung cancer. When to expect a response. Postgrad Med. 1997;101:187–194. doi: 10.3810/pgm.1997.03.180. [DOI] [PubMed] [Google Scholar]
  • 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. 2010;75:173–234. doi: 10.1016/j.critrevonc.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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. 2009;4:792–801. doi: 10.1097/JTO.0b013e3181a7716e. [DOI] [PubMed] [Google Scholar]
  • 7.Zhang W, Ma YZ, Song L, Wang CH, Qi TG, Shao GR. Effect of cantharidins in chemotherapy for hepatoma: A retrospective cohort study. Am J Chin Med. 2014;42:561–567. doi: 10.1142/S0192415X14500360. [DOI] [PubMed] [Google Scholar]
  • 8.Dorn DC, Kou CA, Png KJ, Moore MA. The effect of cantharidins on leukemic stem cells. Int J Cancer. 2009;124:2186–2199. doi: 10.1002/ijc.24157. [DOI] [PubMed] [Google Scholar]
  • 9.Huh JE, Kang KS, Chae C, Kim HM, Ahn KS, Kim SH. Roles of p38 and JNK mitogen-activated protein kinase pathways during cantharidin-induced apoptosis in U937 cells. Biochem Pharmacol. 2004;67:1811–1818. doi: 10.1016/j.bcp.2003.12.025. [DOI] [PubMed] [Google Scholar]
  • 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. 2010;101:1226–1233. doi: 10.1111/j.1349-7006.2010.01523.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yeh CB, Su CJ, Hwang JM, Chou MC. Therapeutic effects of cantharidin analogues without bridging ether oxygen on human hepatocellular carcinoma cells. Eur J Med Chem. 2010;45:3981–3985. doi: 10.1016/j.ejmech.2010.05.053. [DOI] [PubMed] [Google Scholar]
  • 12.Chang C, Zhu YQ, Mei JJ, Liu SQ, Luo J. Involvement of mitochondrial pathway in NCTD-induced cytotoxicity in human hepG2 cells. J Exp Clin Cancer Res. 2010;29:145. doi: 10.1186/1756-9966-29-145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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. 2011;38:1067–1073. doi: 10.3892/ijo.2011.922. [DOI] [PubMed] [Google Scholar]
  • 14.Zhang WD, Zhao HR, Yan Y, Wang XH, Zong ZH, Liu Y. Apoptosis induced by cantharidin in human pulmonary carcinoma cells A549 and its molecular mechanisms. Zhonghua Zhong Liu Za Zhi. 2005;27:330–334. In Chinese. [PubMed] [Google Scholar]
  • 15.Kim JA, Kim Y, Kwon BM, 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. 2013;288:28713–28726. doi: 10.1074/jbc.M113.488346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Osborne C, Wilson P, Tripathy D. Oncogenes and tumor suppressor genes in breast cancer: potential diagnostic and therapeutic applications. Oncologist. 2004;9:361–377. doi: 10.1634/theoncologist.9-4-361. [DOI] [PubMed] [Google Scholar]
  • 17.Heneghan HM, Miller N, Kerin MJ. MiRNAs as biomarkers and therapeutic targets in cancer. Curr Opin Pharmacol. 2010;10:543–550. doi: 10.1016/j.coph.2010.05.010. [DOI] [PubMed] [Google Scholar]
  • 18.Weinstein IB. Cancer. Addiction to oncogenes – the Achilles heal of cancer. Science. 2002;297:63–64. doi: 10.1126/science.1073096. [DOI] [PubMed] [Google Scholar]
  • 19.Luo J, Solimini NL, Elledge SJ. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell. 2009;136:823–837. doi: 10.1016/j.cell.2009.02.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Solimini NL, Luo J, Elledge SJ. Non-oncogene addiction and the stress phenotype of cancer cells. Cell. 2007;130:986–988. doi: 10.1016/j.cell.2007.09.007. [DOI] [PubMed] [Google Scholar]
  • 21.Méndez M, Custodio A, Provencio M. New molecular targeted therapies for advanced non-small cell lung cancer. J Thorac Dis. 2011;3:30–56. doi: 10.3978/j.issn.2072-1439.2010.12.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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. 2013;3:148–152. doi: 10.1016/j.biomed.2013.05.001. [DOI] [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. 2014;45:245–254. doi: 10.3892/ijo.2014.2428. [DOI] [PubMed] [Google Scholar]
  • 24.Ayala F, Dewar R, Kieran M, Kalluri R. Contribution of bone microenvironment to leukemogenesis and leukemia progression. Leukemia. 2009;23:2233–2241. doi: 10.1038/leu.2009.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Straussman R, Morikawa T, Shee K, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487:500–504. doi: 10.1038/nature11183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wilson TR, Fridlyand J, Yan Y, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487:505–509. doi: 10.1038/nature11249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Moniri MR, Dai LJ, Warnock GL. The challenge of pancreatic cancer therapy and novel treatment strategy using engineered mesenchynmal stem cells. Cancer Gene Ther. 2014;21:12–23. doi: 10.1038/cgt.2013.83. [DOI] [PubMed] [Google Scholar]
  • 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. 2008;26:3552–3559. doi: 10.1200/JCO.2007.13.9030. [DOI] [PubMed] [Google Scholar]
  • 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. 2007;25:1553–1561. doi: 10.1200/JCO.2006.09.5570. [DOI] [PubMed] [Google Scholar]
  • 30.Glynne-Jones R, Hoskin P. Neoadjuvant cisplatin chemotherapy before chemoradiation: A flawed paradigm? J Clin Oncol. 2007;25:5281–5286. doi: 10.1200/JCO.2007.12.3133. [DOI] [PubMed] [Google Scholar]
  • 31.Ricci MS, Zong WX. Chemotherapeutic approaches for targeting cell death pathways. Oncologist. 2006;11:342–357. doi: 10.1634/theoncologist.11-4-342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tan TT, White E. Therapeutic targeting of death pathways in cancer: Mechanisms for activating cell death in cancer cells. Adv Exp Med Biol. 2008;615:81–104. doi: 10.1007/978-1-4020-6554-5_5. [DOI] [PubMed] [Google Scholar]

Articles from Molecular Medicine Reports are provided here courtesy of Spandidos Publications

RESOURCES