Open Access

A gene interaction network‑based method to measure the common and heterogeneous mechanisms of gynecological cancer

  • Authors:
    • Mingyuan Wang
    • Liping Li
    • Jinglan Liu
    • Jinjin Wang
  • View Affiliations

  • Published online on: May 3, 2018     https://doi.org/10.3892/mmr.2018.8961
  • Pages: 230-242
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Gynecological malignancies are a leading cause of mortality in the female population. The present study intended to identify the association between three severe types of gynecological cancer, specifically ovarian cancer, cervical cancer and endometrial cancer, and to identify the connective driver genes, microRNAs (miRNAs) and biological processes associated with these types of gynecological cancer. In the present study, individual driver genes for each type of cancer were identified using integrated analysis of multiple microarray data. Gene Ontology (GO) has been used widely in functional annotation and enrichment analysis. In the present study, GO enrichment analysis revealed a number of common biological processes involved in gynecological cancer, including ‘cell cycle’ and ‘regulation of macromolecule metabolism’. Kyoto Encyclopedia of Genes and Genomes pathway analysis is a resource for understanding the high‑level functions and utilities of a biological system from molecular‑level information. In the present study, the most common pathway was ‘cell cycle’. A protein‑protein interaction network was constructed to identify a hub of connective genes, including minichromosome maintenance complex component 2 (MCM2), matrix metalloproteinase 2 (MMP2), collagen type I α1 chain (COL1A1) and Jun proto‑oncogene AP‑1 transcription factor subunit (JUN). Survival analysis revealed that the expression of MCM2, MMP2, COL1A1 and JUN was associated with the prognosis of the aforementioned gynecological cancer types. By constructing an miRNA‑driver gene network, let‑7 targeted the majority of the driver genes. In conclusion, the present study demonstrated a connection model across three types of gynecological cancer, which was useful in identifying potential diagnostic markers and novel therapeutic targets, in addition to in aiding the prediction of the development of cancer as it progresses.

Introduction

Gynecological malignancies, particularly ovarian cancer, cervical cancer and endometrial cancer, are serious medical conditions in women and have been leading causes of cancer mortality in recent years. However, the use of cancer markers for early and progressive detection remain lacking (1). In addition, research has demonstrated that there are close associations across the three aforementioned types of cancer. It has been demonstrated that the progress and the development of the three aforementioned types of cancer are similar, which may be useful when diagnosing any one of these three cancer types. In the case of endometrial cancer, prior to the development of endometrial carcinoma, the endometrium undergoes progressive neoplastic alterations in a parallel fashion to the premalignant alterations observed in the cervix prior to the development of cervical carcinoma (2). The rationale of oophorectomy in surgical management is that endometrial cancer may metastasize to the ovary, in which women with endometrial cancer are at risk for synchronous and metachronous ovarian cancer, and the source of estrogen may be eliminated by oophorectomy (3,4). In cancer cells, oncogenic transformation is associated with major alterations in gene expression (5). With the advent of large-scale screening of cancer genomes, hundreds of genes with alterations in different types of tumors from patients with cancer have been identified (610), which revealed that cancer is a complex disease caused by genetic alterations in multiple genes (11,12). In order to elucidate the cancer marker genes and biological processes associated with each type of gynecological tumor, and the potential underlying mechanism of associations among gynecological tumors, the contribution of identified differentially expressed genes (DEGs) to the pathogenesis of gynecological tumors must be understood.

To analyze different DEGs, high-throughput experimental methods, including microarray analysis, have been widely used in a number of studies (13,14). A vast quantity of microarray data has been produced and deposited in publicly-available data repositories, including the Gene Expression Omnibus (GEO) (15). With the methods of integrated bioinformatics analysis, researchers have been able to advance the identification of genetic signatures. This may provide insights into the underlying biological mechanisms of the development of gynecological tumors.

Chung et al (16) revealed that microRNA (miRNA)-200b/a is a direct transcriptional target of grainyhead like transcription factor 2, which is associated with development and overall survival in epithelial ovarian cancer. Halabi et al (17) demonstrated that 41 genes, including matrix metalloproteinase (MMP)7 and tumor protein 53, were involved in the potential underlying mechanisms of ovarian cancer. Espinosa et al (18) revealed that six genes encoding cyclin B2, cell-division cycle protein 20, protein regulator of cytokinesis 1, synaptonemal complex protein 2, nucleolar and spindle associated protein 1 and cyclin-dependent kinase inhibitor 2 belonging to the mitosis pathway, were potential markers for screening or therapeutic targets of cervical cancer. However, biomarkers which were identified in this way have had poor translation into actual clinical practices. Results have been non-concordant among studies due to small sample sizes. In addition, the studies into the associations of biomarker genes (driver genes) remain lacking among the different types of gynecological tumors.

A robust driver gene biomarker signature may be beneficial for the diagnosis and targeted treatment of gynecological tumors. In the present study, in order to identify a driver gene biomarker signature for the three types of gynecological tumors, data from the Metabolic Gene Rapid Visualizer database (MERAV, which is derived from GEO) was used (19). In MERAV, microarrays were normalized together to eliminate systematic errors caused by different batch experiments.

The present study devised a target network for ovarian cancer, cervical cancer and endometrial cancer using the selected driver genes, and further investigated the identified DEGs via functional enrichment analysis, pathway enrichment analysis and protein-protein interaction (PPI) networks. In addition, the present study extracted clinical information of ovarian cancer, cervical cancer and endometrial cancer from The Cancer Genome Atlas (TCGA) data portal. Subsequently, driver genes in each type of cancer were analyzed. It was important to investigate the underlying mechanism of each gynecological tumor and whether the identified driver genes contributed to these diseases. Subsequently, a network was generated between the miRNAs and the identified driver genes, using the method of mining the Mir2 disease and Tarbase databases which provide information on miRNAs, diseases and the interactions between miRNAs and genes. Finally, the present study determined hub-genes and hub-miRNAs across the gynecological tumors to study the potential underlying mechanisms of the developments of gynecological tumors, which may shed light on different strategies for the design of biological targets for cancer therapies.

Materials and methods

Identification of gene expression datasets

In the present study, DEGs were identified between normal tissues and tumors extracted from the MERAV database from the National Center for Biotechnology Information GEO database (MERAV, http://merav.wi.mit.edu). The experimental samples for the present study are presented in Tables I and II. The following information was extracted from each identified study: GEO accession number, sample type, number of cases and controls, and gene expression data. Studies in which the microarray data were uncertain were excluded. The experimental protocol for the present study is presented in Fig. 1.

Table I.

Datasets from the Metabolic Gene Rapid Visualizer database (cervix).

Table I.

Datasets from the Metabolic Gene Rapid Visualizer database (cervix).

Tissue typeDatasets
Normal, n=4GSM176135, GSM175833, GSM176130, GSM176140
Tumor
  Squamous cell carcinoma, n=5GSM152635, GSM277702, GSM46919, GSM102527, GSM152587
  Squamous cell carcinoma non-keratinizing, n=5GSM179907, GSM46942, GSM76614, GSM152580, GSM203742
  Squamous cell carcinoma keratinizing, n=3GSM117576, GSM152723, GSM152751
  Adenoma, n=6GSM179956, GSM152667, GSM152719, GSM179853, GSM325835, GSM203622

Table II.

Datasets from the Metabolic Gene Rapid Visualizer database (ovary and endometrium).

Table II.

Datasets from the Metabolic Gene Rapid Visualizer database (ovary and endometrium).

Datasets

Tissue typeOvary, n=4Endometrium, n=22
Normal tissuesGSM175789GSM175777, GSM175778,
GSM176131GSM175779, GSM175780,
GSM176136GSM175781, GSM175783,
GSM176318GSM175784, GSM175785,
GSM176039, GSM176040,
GSM176041, GSM176043,
GSM176093, GSM176099,
GSM176127, GSM176137,
GSM176141, GSM176142,
GSM176144, GSM176146,
GSM176143, GSM176145,

Tissue typeOvary serous adenocarcinoma, n=11Endometrioid carcinoma, n=12

TumorsGSM8897, GSM203626, GSM15267,GSM102425, GSM117582,
GSM102445, GSM46831, GSM152577,GSM117586, GSM117590,
GSM88973, GSM152581, GSM27769,GSM88952, GSM88966,
GSM277737, GSM301703GSM102469, GSM102492,
GSM53058, GSM88978,
GSM46923, GSM46937
Integrated analysis of DEGs identified in the extracted databases

Information was extracted from the microarray datasets in MERAV which are presented in Tables I and II, respectively. Following the intersection of the microarray datasets, the DEGs were established between the normal and cancer tissues. In the present study, the degree of differential gene expression was measured by fold-change based on the Student's t-test. A fold-change value >2 or <0.5 and t-test P<0.01 for a gene was considered to be significant. The differential expression analysis was conducted using the Linear Models for Microarray Data package in R (20).

Protein interaction network

The DEGs were subsequently applied to the Human Protein Reference Database (21) (HPRD, www.hprd.org), to identify the more complex functional interactive driver genes of separate cancer types. Genes with interactions with each other were extracted from the DEGs as mentioned above (presented in Tables IIIX). The PPI network is a useful research tool for investigating the cellular networks of protein interactions, and was downloaded from the HPRD. Cancer-associated gene-gene interaction networks were constructed by mapping the DEGs into the HPRD PPI network for each cancer (cervix tumor, ovarian tumor and endometrium tumor). To make it easier to identify the driver genes, the present study calculated the lines attached to each node, which was defined as the degree of the node. The nodes that exhibited degrees ≥4 were defined as driver genes. The nodes whose degree was ≥4 were considered to serve more complex roles in the development of the diseases of interest. These nodes were then extracted for the PPI network (Fig. 2). The present study constructed a connected network which contained the driver genes across the three cancer types. Through this method, it was determined whether the driver genes of the separate cancer types had any interaction with each other. The networks were constructed using Cytoscape version 3.3.0 (www.cytoscape.org).

Table III.

Driver genes identified by integrated analysis of the microarray datasets (cervical squamous cell carcinoma).

Table III.

Driver genes identified by integrated analysis of the microarray datasets (cervical squamous cell carcinoma).

Gene
RB1HTRA1MTORCLDN5NARFPURA
MCM7KPNA2PLSCR4CYBANCAPD2RBM8A
MCM2LMNB1PRKD1DCUN1D1NCF4RECK
PLK1MEIS1PSMA5DDAH2NME4REV3L
ARNCOA1PSMB10DMPKNPLOC4RFC3
PPP1CAPBX1PSMB9EPS8NR2F1RNF126
ABL1PIAS3PSMD2EXOSC5NR2F2RPA3
LMNAPOLA2RACGAP1GABBR1NRASRRM1
PTNPPP1R14ARTN3GAS6NTF3RRM2
TRIP13AXLSNRPBGCH1NTRK2SAT2
CAV1BUB1BTOR1AIP1GCHFRNUB1SDC2
CDC20CCL14TUBA4AGLRX3NUP210SEC24A
CDC6CCR5UBTFGMFBNUP50SELENBP1
FLNACOL4A5USP6NLGOLGA2PAFAH1B3SERBP1
FXR2CSNK1DUTP3HOXD13PAK2SH3BP5
ZHX1DBF4ACTN4ILKPAMSMC4
CCNA2DVL3ADAM10KANK1PCGF2SNRPD1
DGKZEFEMP2ANTXR2LAPTM5PHACTR4SNTB2
MCM10EIF4EBP1ARHGAP17LDB2PLK2SNX27
MCM6EZH2ASPMLDOC1PNO1SPIN1
PCNAFAM46ABIDLMO4PNPSSSCA1
RBPMSHOXD10BMP4LRP1PPIASTXBP2
RPS6KA1HSPA4BNIP2LRP6PPIHSUB1
SAT1ITGB3BPC1QALRRC41PRPF18TALDO1
BUB1KLF6CBX4LZTS2PSMA6TGFBR3
CSNK1EMAD2L2CCNE1MAGEH1PSMB7TNFRSF1A
DCNMAP2K4CCR1MELKPSMD4UFD1L
FGFR1MAPK10CDC42BPAMPDZPSME3WSB2
FXYD1MCM5CENPEMTA1PSMF1XPNPEP1
GMNNMITFCHFRMYCBPPSTPIP1YLPM1
HOXA10MMP9CIB1MYL9PTTG1ZMIZ1

Table X.

Driver genes identified by the integrated analysis of the microarray datasets (endometrial carcinoma).

Table X.

Driver genes identified by the integrated analysis of the microarray datasets (endometrial carcinoma).

Gene
EP300CDKN2AF2RAMFREPN3MMP11
JUNCOL3A1FZD5AXLEPR1MMP26
CAV1EGR1HLA-DMBBCL11AFOSBMYO5B
CTNNB1ERBB4HOXA10BCL2A1GALNT10NRG2
ABL1FBLN1ID1BIKGAS6NRXN2
ARFBN1ID4BLNKGATA2PCOLCE
TCF4FLNAIDEC1RGCH1PDGFRB
THBS1FOXO1INADLC1SGCHFRPKD2
TUBA4AHLA-DRAJUNDC3AR1GPIPNP
ATXN1ID3LMO4CCND2GPRASP1PPP1R14A
COL1A1IGFBP5LNX1CDH11HLA-DQB1PRDM1
DCNLAMB3NCALDCDKN1AHLA-DRB1PSTPIP2
LRP1MITFNCF2CDKN2CHLFPTGDS
C3MYCNR2F2CFBHOXA9PTGS2
COL7A1PLATPDGFRACGNID2R3HDM2
FBLN2RUNX1T1PLEKHF2CLEC3BIGFBP4RAB25
FOSS100A8PTPN13CLK1IGFBP6RAB3IP
GNAI2SERPINA1RAB8BCXADRIL33RAPGEF6
IGF1SYKRABAC1CXCL10IRS1S100A9
LAMC2TGFB1I1ROR2DNM1KLF5SCRIB
MUC1CD14SFNDPYSL2LAPTM5SEC24D
NID1COL5A1SFRP1ECM1LDB2SNTB2
PRKD1CRMP1TFAP2AEDNRALUC7L3SOX9
PTPN12DBPTJP2EFEMP2MAFBSPINT1
VCANDDR2TRPC1EFSMAL2SPP1
CD74F10WNT5AENO2MAPK10ST14
SYTL1TJP3TLR3TROWASF2WNT4
TBL1XTLR2TPD52USP54WNT2ZEB1
miRNAs regulating gene network construction

The present study analyzed the association between miRNAs and the identified driver genes (Fig. 3). This process was performed by extracting a list of miRNAs which were associated with the type of cancer (cervical tumor, ovarian tumor or endometrial tumor) from the Mir2 Disease database (www.mir2disease.org) (22). Following this step, a network was created regarding the regulatory associations between the miRNA and the specific driver gene of each type of cancer in order to identify the hub-miRNAs of the gynecological tumors. The associations of the regulation were extracted from Tarbase (diana.cslab.ece.ntua.gr/tarbase) (23).

Functional and pathway enrichment analysis

In order to assess the functional relevance of the aforementioned DEGs, a pathway analysis was created based on the Database for Annotation, Visualization and Integrated Discovery (DAVID) (24). DAVID provides a useful tool to analyze large gene lists, including gene ontology (GO) and pathway analysis. DEGs in different diseases were applied to this database in order to detect potentially represented functions. GO-categories were organized based on the GO database (25) (www.geneontology.org). In addition, pathway analysis was based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (26) (genome.jp/kegg). Significant categories were identified by expression analysis systematic explorer scores, a modified Fisher's exact P-value. The threshold for significance for a category was considered to be P<0.01, with >4 genes for the corresponding term.

Survival analysis

The present study used TCGA database to extract clinical information and gene expression profile information. At the start of the analysis, the expression values of each driver gene were listed, which were identified via the PPI network. To find the median level of gene expression, the samples were divided into two groups by median of expression (high expression group and low expression group). Additionally, the corresponding clinical information of each sample was extracted. Survival data representing time between initial diagnosis and mortality were downloaded directly from TCGA data portal (tcga-data.nci.nih.gov/tcga/tcgaHome2.jsp) (27). With this information, the present study was able to estimate the association between the identified driver genes of the three types of cancer mentioned above and the survival rates of patients. All analyses were conducted using custom-written code in R (www.r-project.org).

Results

Integrated analysis of multiple studies to establish the driver genes in cancer

There are multiple genes that contribute to the cause of the aforementioned cancer types and, therefore, no single gene is a determining factor in diagnosis. It was identified that each type of cancer was driven by different variations of genes that serve key roles during the development of pathology. However, no single gene may explain the heterogeneity of each type of cancer. In the case of cervical cancer, 186 genes in squamous cell carcinoma of the cervix (Table III), 107 genes in keratinized squamous cell carcinoma of the cervix (Table IV), 96 genes in cervical adenocarcinoma Grade 3 (Table V), 133 genes in non-keratinized squamous cell carcinoma of the cervix (Table VI) and 203 genes in cervical adenocarcinoma Grade 2 (Table VII) were identified to be important. In addition, 120 genes and 76 genes were established, respectively, in adenocarcinoma of the ovary Grade 2 and Grade 3 (Tables VIII and IX). A total of 168 genes were established in endometrial carcinoma (Table X).

Table IV.

Driver genes identified by integrated analysis of the microarray datasets (cervical keratinized squamous cell carcinoma).

Table IV.

Driver genes identified by integrated analysis of the microarray datasets (cervical keratinized squamous cell carcinoma).

Gene
FYNADAM10ARHGAP17HSPB2PHF1TMOD1
ZHX1ADAM17ARMCX2ID4PIK3C2BTMSB10
ABL1ANXA6BIN1LDB2PIP5K1CTPD52
BCL2L1AXLCBX3LDOC1PNPUBTF
FXR2BCL11ACLDN5LMO4PSME3ZHX2
TBPCSNK1ECNN3LRP1PSMF1ZMIZ1
ARDMPKCNNM3LRP6PTOV1ZNF76
BARD1ITGB3BPCNTNAP1LSM5PTPN12
BIDKPNA6CRYABMAGI2RAE1
DDX24MAD2L2CSE1LMAPK10REV3L
NCOA1MCL1CSTF1MIS12RUNX1T1
PDGFRBNR2F1EFEMP2MPDZSDC2
PRKD1NTRK2EXOSC5MTA1SFRP1
PSEN1PPP1R14AFGFR1MYCBPSH3BP5
RBPMSPTNFXYD1NPDC1TAF9
SPTAN1RTN3FZD6NR2F2TCF7L2
TCF4SYKGAS6NTF3TERF1
TGFAVIMGDI1NUDT21TFDP1
A2MANTXR2GTF3C3PBX1TGFBR3
ACP1AQP1HOXA10PDGFDTLN2

Table V.

Driver genes identified by integrated analysis of the microarray datasets (cervical adenocarcinoma G3).

Table V.

Driver genes identified by integrated analysis of the microarray datasets (cervical adenocarcinoma G3).

Gene
ARBADPLD2CIB1MAPK10SERPINA1
CAV1BAHD1PPA1CLDN5MED14SF1
FLNAC1QBPPRKD1CUL4BMPDZSMO
PPP1CACPSF6SAT1DMPKMYL9SPINT2
NCK2CSNK1DSMAD1EFNB1NR2F1SSBP3
PLSCR1DOCK1SNAP23F3NTF3SSR1
SUMO4DVL2TAF1DGDI1PCGF2STAM
LMNAFXR2TAF9HOXA10PDPK1SYNE1
LRP1FXYD1TCF4HOXD10PHACTR4TCF7L2
PSEN1ILKWIPI1HOXD13PHYHIPTGFBR3
PTNLDB1ACVR2AHSPA1BPLSCR4TMF1
CSNK1ELMO4ANTXR2HSPBAP1PNPLA2UBTF
DVL3MAP2K4ATG12KANK1PPP1R10VAMP8
MMP14NCOA1CD82KPNA6PTCH2WASF1
PPP1R14ANTRK2CDC42BPALDB2RNF138WASF2
ALDOAPBX1CDC42EP1LRP6RUNX1T1ZHX1

Table VI.

Driver genes identified by integrated analysis of the microarray datasets (cervical non-keratinized squamous cell carcinoma).

Table VI.

Driver genes identified by integrated analysis of the microarray datasets (cervical non-keratinized squamous cell carcinoma).

Gene
ARFXR2FOXO1TLR2FBN2NTF3
ABL1ILKGMNNTXNDC9FGRNTRK2
CAV1LMNAHOXD10XRCC4FXYD1NUBP1
CHD3MEIS1ICAM3YAP1GDI1PALLD
HIF1ANCOA1ITGB2ADCY6HCLS1PDPK1
PTPN6PAG1LCP2ADI1HLA-DMBPGK1
SAT1PBX1LRP1AGTPBP1HLA-DRAPGLS
FLNAPIAS1MAFGANTXR2HOXD13PIK3R3
HOXA10PSEN1MPDZANXA6HSPB2PLTP
PLSCR1PTNNDNARHGDIBLCP1PNP
RAF1WASF2NR2F2CDC37LDOC1PRRX1
DCNZHX1PAICSCITED2LILRB2RAB11FIP2
EZRACTR3PLSCR4CLDN5LRP6RAB18
MMP14BIN1PPP1R14ACNN3MAPK10RFXANK
PDGFRBC1QBPPP2R1ACOL4A5MED14RUNX1T1
ABCA1C1QCPRDX2DOCK1MTA1SAT2
C1QACSNK1DSNTB2DVL2MYO5BSEPHS1
CSNK1EDGKZSSSCA1ENO1NARFSF1
DMPKDVL3TCF4FAM46ANISCH
ELNEFEMP2TLR1FBLN1TICAM1
SNX2SYNE1TCF7L2VTA1TRAP1
TMEM8BTMOD1TMSB10TPD52SH3BP5
WASF3ZNF76TEAD3TIMP2NR2F1

Table VII.

Driver genes identified by integrated analysis of the microarray datasets (cervical adenocarcinoma G2).

Table VII.

Driver genes identified by integrated analysis of the microarray datasets (cervical adenocarcinoma G2).

Gene
ABL1HSPA5ASAP1PSMF1ASS1EHD2
ARHTRA1AXLQKIATRXENAH
CAV1LMNABCRRAB4AAURKAENO1
PPP1CAMEIS1BGNRNF138AURKBERBB3
FLNANTRK2BMP4SDC2BIN1FBLN1
FYNPRNPBRCA2SMARCE1BIRC5GAS6
MMP2PTPN12CDKN2ASNAP29CAPZBGLRX3
SMAD1SMAD5CSNK1ETAF7CAV2GOLGA2
NCK2TAF9DMPKTCF4CBX4GTF2I
RB1TTF2DOCK1TGFBR3CD81HAT1
PTNDVL2DR1THBS2CDT1HOXD10
PTPN6EFEMP2FGFR1TIFACEP76HSPA1B
SMAD7FXR2FXYD1TIMP2CLDN5HSPB2
SUMO4HOXA10GDF5TNFRSF1ACLUIDE
A2MHOXD13GNA12ZHX1CNN3IFI35
AP1M1LRP1KIDINS220ADI1CNTNAP1IFNAR1
CDC5LNCOA1LDOC1AHNAKCOL4A5ILK
EZRNOTCH2LRP6ALDOACOL6A3IQGAP1
MMP14PBX1MAFGANTXR1COX5AJAG1
PIAS1PDGFRBMAP2K4ANTXR2CUL4BKANK1
CD2APPRKD1MAPK10ANXA6CXCL12KDM2A
CDH1SAT1MEF2CAQP1DCLRE1AKPNA6
DCNWASF2POLE3ARHGAP17DDX24LCAT
DRAP1YAP1PPP1R14AARHGEF6EFNB1MAD2L1BP
ELNACVR2APRRX1ASH1LEFSMAP3K3
MCM4NR2F2PLSCR4RUNX1T1SYNE1WNK1
MED14NTF3PPA1SALL2TEAD3YLPM1
MPDZNUDT21PPP1R10SAT2TERF1ZMIZ1
MSNPALB2PPP2R1ASETD7THBS3
MYCBP2PALLDPSMB10SH3BP5TMEM8B
MYO5BPBX3PURASH3KBP1TSPAN4
NFE2L1PDGFDRAB11FIP1SKAP1TWIST2
NMIPHACTR4RAB11FIP2SPARCL1UBTF
NPHS2PIP4K2BRBPJSTX3VGLL4
NR2F1PKD2REPS2STX7WFDC2

Table VIII.

Driver genes identified by integrated analysis of the microarray datasets (adenocarcinoma of the ovary Grade 2).

Table VIII.

Driver genes identified by integrated analysis of the microarray datasets (adenocarcinoma of the ovary Grade 2).

Gene
JUNMEF2CHSPA1ACNNM3GNEPHF1
FXR2NCOA2HTRA1COX5AGNG4PKD2
RAF1NIF3L1IKZF4CRY2GPRASP1PLA2G16
RBPMSPCBD1LIFRCTF1HMGA1PLK1
ZBTB16PDGFRAMAPK10CTSDHSPA2PTPN13
PRKACAPRTFDC1MYO15ADCNICAM3RBBP8
CAV1STAT5ANFE2L1DSTIGFBP4RBP1
MAP3K3APBB1NR2F6ELF3IRS1SDC2
MAP3K5C1RPER1ELK1KIAA1217SGK1
NCOA1C1SPTPN6ENAHMAFGSH3BP5
PDGFRBCALCOCO2SERPING1ENGMRASSMC3
SIN3ACD2APSIN3BEPS8NBL1SNCA
ABLIM1DCTN1TGFBR3ETV6NFATC4SNRNP70
DDX17DMPKTSC22D3EYA2NINLSPOP
FEZ1DVL2UBQLN1FLAD1NR2F2SPTBN1
GATA4FHL2ACTA2FOXO1OLFML3SPTBN2
GOLGA2FLNABEGAINFOXO3PAICSST13
LRP1FXYD1CCT5FTH1PDGFDSTRBP
TCF4THRATPM2TXNUSP13ZC3H10
TEAD1TOP2ATRIM21TXNDC9WTIPZFPM2

Table IX.

Driver genes identified by integrated analysis of the microarray datasets (adenocarcinoma of the ovary Grade 3).

Table IX.

Driver genes identified by integrated analysis of the microarray datasets (adenocarcinoma of the ovary Grade 3).

Gene
CDK1HLA-DRACD14FCGR2BPDGFDNR2F2
AURKBICAM3CDC20FOSSLPI
CAV1KRT7CDH1GCASMC4
PTPN6MAD2L1CDKN2AGNESOX9
ZBTB16MAL2CEBPGGPRASP1SPINT1
BCL2L1MAP3K5CENPAHLA-DMBST14
HSPA1APDGFRACKS2HLA-DRB1STRBP
IRS1PDGFRBCLDN1LAPTM5TACC1
ITGB2PMAIP1CLDN3LCP1TOP2A
MCM2RACGAP1CRIP1LRP1TRIP13
NDC80RBPMSCTSSMSLNTYROBP
SYKTPD52CXCR4MUC1ZWINT
TPD52L1ALOX5DBF4MUC16ECT2
BCL11AALOX5APDSC2NCAPD2CCNB1
CCNB2BIKDSG2NR2F1ERBB3
Integrated PPI (protein-protein interactions) network construction

Based on the HPRD, the interaction network of the identified driver genes was constructed, which consisted of 101 nodes (genes that form associations) and 185 edges (biological association) (Fig. 2). Genes with a higher degree of association (degree ≥4) were observed to be larger in size, and included the genes CDK1, CAV1, ZBTB16, Jun proto-oncogene AP-1 transcription factor subunit (JUN), RAF1, RB1, minichromosome maintenance complex component 2 (MCM2), AR, ABL1, LMNA, FLNA, DCN, FYN, SMAD1, LRP1, PSEN1, EP300, CTNNB1, collagen type I α1 chain (COL1A1) and FOS. Through this method, it was identified that driver genes in each gynecological cancer have contact interactions.

Comprehensive analysis of miRNA regulation and the selected driver genes

Fig. 3 illustrates that certain miRNAs serve important roles in regulating the driver genes. In the present study, it was demonstrated that a number of miRNAs regulate separate networks [for example the let7 family, miRNA (miR)-23b, miR-21, miR-214 and miR-218]. miRNAs that were confirmed to be significant in cervical cancer, including let7c and let7b, are also found to be associated with the other two cancers in this study. This information may be important in establishing the connections between the three gynecological cancer types, which may be used in the development of targets for further research and diagnosis.

Functional and pathway enrichment analysis

GO analysis revealed that the identified genes of cervical tumors, ovarian tumors and endometrial tumors were predominantly involved in the illustrated biological processes (Fig. 4). The top three significant biological processes of cervical cancer were ‘mitotic cell cycle’, ‘cell cycle’ and ‘cell cycle process’, while for ovarian cancer, the biological processes consisted of ‘cell cycle process’, ‘cell cycle phase’ and ‘macromolecule metabolic process’. For the progression of endometrial cancer, the top three biological processes observed to be at fault for cancer progression were ‘response to organic substance’, ‘regulation of cell proliferation’ and ‘skeletal system development’.

Using the method of pathway analysis, it was revealed that genes in cervical cancer were significantly enriched in ‘cell cycle’, ‘pathways in cancer’ and ‘DNA replication’. Ovarian cancer was observed to be significantly enriched in ‘MAPK signaling pathway’, ‘cell cycle’ and ‘oocyte maturation’. Endometrial cancer was observed to be significantly enriched in ‘pathways in cancer’, ‘focal adhesion’ and ‘complement and coagulation cascades’ (Fig. 5).

Survival analysis of patients with gynecological tumor

Fig. 6 illustrates the association between survival time and survival rate in the high and low expression groups. The genes MCM2, MMP2, COL1A1 and JUN are presented in the figure, and it was observed that the driver genes of the expression groups were able to divide each of the target cancer types into two groups, one of which contained the high expression group with the other containing the low expression group. Therefore, in order to determine whether the driver genes had a key role in the development of gynecological tumors and the connective function of separate cancer types, the present study aimed to identify the association between the target cancer driver genes and other types of gynecological cancer.

Discussion

The principal challenge of high-throughput cancer genomics is to identify specific driver genes and the underlying mechanisms of carcinogenesis, apart from the vast quantity of heterogeneous genomic alteration data. Numerous studies have focused on identifying individual functional modules or pathways involved in cancer (2830). Based on this methodology, the analysis of the present study focused specifically on DEGs in order to reveal the transcriptional responses of gynecological tumors. The results of this analysis suggested that the common biological processes of cancer of the cervix, ovary and endometrium were those involved in the cell cycle and the regulation of macromolecule metabolism.

The cell cycle is the progression of biochemical and morphological phases and events that occur in a cell during successive cell replication or nuclear replication. Research has shown that interference with cell cycle components may lead to tumor formation (31). Certain cell cycle inhibitors, including retinoblastoma protein and tumor protein 53 may mutate during replication, causing the cell to proliferate uncontrollably, ultimately resulting in a tumor. Furthermore, the proportion of active cell division in tumors is much higher compared with the rate in normal tissue.

To clarify the hub genes in ovarian cancer, cervical cancer and endometrial cancer, DEGs were predicted to be biomarkers for each cancer using PPI networks. It is considered that hub nodes are genes that are highly connected with other genes and have been predicted to serve key roles in numerous networks. In addition, highly connected hub genes were proposed to have a considerable role in biological development. Hub nodes have more complex interactions compared with those of other nodes, which indicates that they have pivotal roles in the underlying mechanisms of disease. In addition, certain identified biomarkers of each type of cancer were extracted from each network and these driver genes were placed into one PPI network with the duplication hub genes eliminated. Therefore, the particular hub genes of each gynecological cancer and the connection nodes across the three types of cancers may be identified. Accordingly, the identification of hub genes and hub connected genes involved in each gynecological cancer may lead to the discovery of the association across ovarian cancer, cervical cancer and endometrial cancer, and may lead to the development of effective diagnostic and therapeutic approaches.

In order to ascertain a causal association across the three types of gynecological cancer, the present study extracted clinical information and gene expression profile information from TCGA database, and used the hub connected genes identified in the PPI network to perform survival analysis. In the present study, four noteworthy genes were identified, including MCM2, MMP2, COL1A1 and JUN.

The present study demonstrated that MCM2 may serve a key role in cervical cancer. A poor prognosis was associated with lower expression. Furthermore, MCM2 was highly connected with ovarian cancer and endometrial cancer. The results suggested that MCM2 is a component of the DNA replication licensing complex, with a rich binding surface that directs multiple regulatory interactions of cancer significance, marking DNA replication origins during the G1 phase of the cell cycle for use in the subsequent S-phase. A deficiency of MCM2 results in death or morbidity in the absence of an overt tumor (32). These processes of DNA replication have been studied and used as therapeutic targets. Simon and Schwacha (33) suggested that MCM2 was a promising target for blocking the proliferation of cancerous and precancerous cells.

In the present study, MMP2 was identified to be essential in causing cervical cancer. MMPs are zinc-containing endopeptidases with an extensive range of substrate specificities. These enzymes are able to degrade various components of extracellular matrix (ECM) proteins. In photocarcinogenesis, degradation of the ECM is the initial step towards tumor cell invasion, to intrude in the basement membrane and the surrounding stroma that primarily comprises fibrillary collagens. Additionally, MMP2 is involved in angiogenesis, which promotes cancer cell growth and migration (34).

COL1A1 and COL1A2 encode the α1 and α2 chains of type I collagen, respectively (35). The primary constituents of the ECM are collagens, adhesive glycoproteins and proteoglycans (36). Specific interactions between cells and ECM-mediated cell-surface-associated components and transmembrane molecules result in the control of cellular activities, including adhesion and migration (37). Collagen is the primary component of the ECM, which serves pivotal roles in maintaining skin and vessel elasticity, and increasing cartilage lubricity (38). Upregulation of type II collagen expression may contribute to ovarian cancer metastasis and biological processes, including cell proliferation, invasion and migration (39). The oncogene JUN is the putative transforming gene of avian sarcoma virus 17, which is the most extensively studied protein of the activator protein-1 complex and is involved in numerous cell activities, including proliferation, apoptosis, survival, tumorigenesis and tissue morphogenesis. The present study identified that COL1A1 was important in ovarian cancer, which was highly connected with cervical and endometrial cancer. Therefore, COL1A1 and JUN may be potentially important associated genes of the three types of gynecological malignancies.

miRNAs are small noncoding regulatory RNAs that downregulate transcription by targeting specific mRNAs. Furthermore, the present study identified that certain miRNAs were highly associated with hub connected genes, including let7, which is one of the founding members of the miRNA family. This miRNA was first identified in Caenorhabditis elegans. Lee and Dutta (40) identified six functional let7 target sites in the 3′-untranslated region of high mobility group AT-hook 2 (HMGA2), which reduced HMGA2 expression and cell proliferation in a lung cancer cell line. Using genome-wide mRNA expression analysis, Mi et al (41) identified that miRNA let7B was downregulated in acute lymphoblastic leukemia (ALL) compared with acute myeloid leukemia (AML). Quantitative polymerase chain reaction analysis confirmed the downregulation of let7B in ALL samples compared with AML samples and normal controls.

The present study identified that let7a, let7b and let7c had strong connections with the hub genes and that these miRNAs may serve an important part of the potential mechanism, which may explain the connections across the hub genes.

Overall, the present study identified a number of DEGs associated with gynecological cancer, in addition to the functions and signaling pathways in which these genes were involved. Comprehensive network analyses of the dysregulated gene expression in gynecological cancers identified a series of hub genes and the connection genes across ovarian cancer, cervical cancer and endometrial cancer in a PPI network. Subsequently, this study confirmed the driver genes by survival analysis using the TCGA database. Comprehensive network analyses of miRNAs and connection driver genes identified certain miRNAs which may be potential therapeutic and prevention targets of gynecological cancer. In addition, the present study demonstrated the associations across the different gynecological cancers, which may be useful for identifying potential useful diagnostic markers and novel therapeutic targets. The results of this study may provide an insight into the underlying mechanism of the aforementioned gynecological cancers and may lead to further improvement in diagnosis and treatment of them.

Acknowledgements

The authors would like to thank Professor Yunyan Gu (College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.) for technical support and critically reviewing the manuscript.

Funding

The present study was supported by grant no. RC2013QN004112 from Harbin Science and Technology Innovation Talents, China.

Availability of data and materials

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

Authors' contributions

MY and JW conceived and designed the study; MY, LL and JL performed the experiments and analyzed the data. MY wrote the paper, and JW revised the manuscript and gave final approval of the version to be published.

Ethics approval and consent to participate

The present study was approved by the Clinical Research Ethics Committee of the Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Zhuzhou, China), and written informed consent was obtained from all participants.

Consent for publication

Written informed consent was obtained from all volunteers for the publication of any associated data.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

miRNAs

microRNAs

GO

gene ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

PPI

Protein-protein Interaction

DEGs

differentially expressed genes

GEO

Gene Expression Omnibus

MERAV

Metabolic Gene Rapid Visualizer Database

HPRD

Human Protein Reference Database

MMPs

matrix metalloproteinases

ECM

extracellular matrix

References

1 

Li XY and Wang X: The role of human cervical cancer oncogene in cancer progression. Int J Clin Exp Med. 8:8363–8368. 2015.PubMed/NCBI

2 

Nisker JA: Screening for endometrial cancer. Can Fam Physician. 29:961–965. 1983.PubMed/NCBI

3 

Setiawan VW, Yang HP, Pike MC, McCann SE, Yu H, Xiang YB, Wolk A, Wentzensen N, Weiss NS, Webb PM, et al: Type I and II endometrial cancers: Have they different risk factors? J Clin Oncol. 31:2607–2618. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Wright JD: Take 'em or leave 'em: Management of the ovaries in young women with endometrial cancer. Gynecol Oncol. 131:287–288. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Lukk M, Kapushesky M, Nikkilä J, Parkinson H, Goncalves A, Hube W, Ukkonen E and Brazma A: A global map of human gene expression. Nat Biotechnol. 28:322–324. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Cancer Genome Atlas Research Network, . McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, Mastrogianakis GM, Olson JJ, Mikkelsen T, Lehman N, et al: Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 455:1061–1068. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Cancer Genome Atlas Research Network: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Guichard C, Amaddeo G, Imbeaud S, Ladeiro Y, Pelletier L, Maad IB, Calderaro J, Bioulac-Sage P, Letexier M, Degos F, et al: Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat Genet. 44:694–698. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Liu P, Morrison C, Wang L, Xiong D, Vedell P, Cui P, Hua X, Ding F, Lu Y, James M, et al: Identification of somatic mutations in non-small cell lung carcinomas using whole-exome sequencing. Carcinogenesis. 33:1270–1276. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Nik-Zainal S, Alexandrov LB, Wedge DC, Van Loo P, Greenman CD, Raine K, Jones D, Hinton J, Marshall J, Stebbings LA, et al: Mutational processes molding the genomes of 21 breast cancers. Cell. 149:979–993. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Baudot A, Real FX, Izarzugaza JM and Valencia A: From cancer genomes to cancer models: Bridging the gaps. EMBO Rep. 10:359–366. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Kreeger PK and Lauffenburger DA: Cancer systems biology: A network modeling perspective. Carcinogenesis. 31:2–8. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Lee S, Stewart S, Nagtegaal I, Luo J, Wu Y, Colditz G, Medina D and Allred DC: Differentially expressed genes regulating the progression of ductal carcinoma in situ to invasive breast cancer. Cancer Res. 72:4574–4586. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Wray CJ, Ko TC and Tan FK: Secondary use of existing public microarray data to predict outcome for hepatocellular carcinoma. J Surg Res. 188:137–142. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Barrett T, Troup DB, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, et al: NCBI GEO: Archive for functional genomics data sets-10 years on. Nucleic Acids Res. 39:(Database Issue). D1005–D1010. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Chung VY, Tan TZ, Tan M, Wong MK, Kuay KT, Yang Z, Ye J, Muller J, Koh CM, Guccione E, et al: GRHL2-miR-200-ZEB1 maintains the epithelial status of ovarian cancer through transcriptional regulation and histone modification. Sci Rep. 6:199432016. View Article : Google Scholar : PubMed/NCBI

17 

Halabi NM, Martinez A, Al-Farsi H, Mery E, Puydenus L, Pujol P, Khalak HG, McLurcan C, Ferron G, Querleu D, et al: Preferential allele expression analysis identifies shared germline and somatic driver genes in advanced ovarian cancer. PLoS Genet. 12:e10058922016. View Article : Google Scholar : PubMed/NCBI

18 

Espinosa AM, Alfaro A, Roman-Basaure E, Guardado-Estrada M, Palma Í, Serralde C, Medina I, Juárez E, Bermúdez M, Márquez E, et al: Mitosis is a source of potential markers for screening and survival and therapeutic targets in cervical cancer. PLoS One. 8:e559752013. View Article : Google Scholar : PubMed/NCBI

19 

Shaul YD, Yuan B, Thiru P, Nutter-Upham A, McCallum S, Lanzkron C, Bell GW and Sabatini DM: MERAV: A tool for comparing gene expression across human tissues and cell types. Nucleic Acids Res. 44:D560–D566. 2016. View Article : Google Scholar : PubMed/NCBI

20 

R Development Core Team, . R: A language and environment for statistical computingR Foundation for Statistical Computing. Vienna, Austria: 2017

21 

Prasad Keshava TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A, et al: Human protein reference database-2009 update. Nucleic Acids Res. 37:(Database Issue). D767–D772. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Jiang Q, Wang Y, Hao Y, Juan L, Teng M, Zhang X, Li M, Wang G and Liu Y: miR2Disease: A manually curated database for microRNA deregulation in human disease. Nucleic Acids Res. 37:(Database Issue). D98–D104. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Vlachos IS, Paraskevopoulou MD, Karagkouni D, Georgakilas G, Vergoulis T, Kanellos I, Anastasopoulos IL, Maniou S, Karathanou K, Kalfakakou D, et al: DIANA-TarBase v7.0: Indexing more than half a million experimentally supported miRNA:mRNA interactions. Nucleic Acids Res. 43:(Database Issue). D153–D159. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

25 

Kaimal V, Bardes EE, Tabar SC, Jegga AG and Aronow BJ: ToppCluster: A multiple gene list feature analyzer for comparative enrichment clustering and network-based dissection of biological systems. Nucleic Acids Res. 38:W96–W102. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Altermann E and Klaenhammer TR: PathwayVoyager: Pathway mapping using the Kyoto encyclopedia of genes and genomes (KEGG) database. BMC Genomics. 6:602005. View Article : Google Scholar : PubMed/NCBI

27 

Stamoulis C and Betensky RA: A novel signal processing approach for the detection of copy number variations in the human genome. Bioinformatics. 27:2338–2345. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Miller CA, Settle SH, Sulman EP, Aldape KD and Milosavljevic A: Discovering functional modules by identifying recurrent and mutually exclusive mutational patterns in tumors. BMC Med Genomics. 4:342011. View Article : Google Scholar : PubMed/NCBI

29 

Ciriello G, Cerami E, Sander C and Schultz N: Mutual exclusivity analysis identifies oncogenic network modules. Genome Res. 22:398–406. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Vandin F, Upfal E and Raphael BJ: De novo discovery of mutated driver pathways in cancer. Genome Res. 22:375–385. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Tsaniras Champeris S, Kanellakis N, Symeonidou IE, Nikolopoulou P, Lygerou Z and Taraviras S: Licensing of DNA replication, cancer, pluripotency and differentiation: An interlinked world? Semin Cell Dev Biol. 30:174–180. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Pruitt SC, Bailey KJ and Freeland A: Reduced Mcm2 expression results in severe stem/progenitor cell deficiency and cancer. Stem Cells. 25:3121–3132. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Simon NE and Schwacha A: The Mcm2-7 replicative helicase: A promising chemotherapeutic target. Biomed Res Int. 2014:5497192014. View Article : Google Scholar : PubMed/NCBI

34 

Pittayapruek P, Meephansan J, Prapapan O, Komine M and Ohtsuki M: Role of matrix metalloproteinases in photoaging and photocarcinogenesis. Int J Mol Sci. 17:pii: E8682016. View Article : Google Scholar

35 

Chan TF, Poon A, Basu A, Addleman NR, Chen J, Phong A, Byers PH, Klein TE and Kwok PY: Natural variation in four human collagen genes across an ethnically diverse population. Genomics. 91:307–314. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Bosman FT and Stamenkovic I: Functional structure and composition of the extracellular matrix. J Pathol. 200:423–428. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Uitto VJ and Larjava H: Extracellular matrix molecules and their receptors: An overview with special emphasis on periodontal tissues. Crit Rev Oral Biol Med. 2:323–354. 1991. View Article : Google Scholar : PubMed/NCBI

38 

Deyl Z, Miksik I and Eckhardt A: Preparative procedures and purity assessment of collagen proteins. J Chromatogr B Analyt Technol Biomed Life Sci. 790:245–275. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Dai J, Wang T, Wang W, Zhang S, Liao Y and Chen J: Role of MAPK7 in cell proliferation and metastasis in ovarian cancer. Int J Clin Exp Pathol. 8:10444–10451. 2015.PubMed/NCBI

40 

Lee YS and Dutta A: The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev. 21:1025–1030. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Mi S, Lu J, Sun M, Li Z, Zhang H, Neilly MB, Wang Y, Qian Z, Jin J, Zhang Y, et al: MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proc Natl Acad Sci USA. 104:19971–19976. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2018
Volume 18 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
Wang M, Li L, Liu J and Wang J: A gene interaction network‑based method to measure the common and heterogeneous mechanisms of gynecological cancer. Mol Med Rep 18: 230-242, 2018
APA
Wang, M., Li, L., Liu, J., & Wang, J. (2018). A gene interaction network‑based method to measure the common and heterogeneous mechanisms of gynecological cancer. Molecular Medicine Reports, 18, 230-242. https://doi.org/10.3892/mmr.2018.8961
MLA
Wang, M., Li, L., Liu, J., Wang, J."A gene interaction network‑based method to measure the common and heterogeneous mechanisms of gynecological cancer". Molecular Medicine Reports 18.1 (2018): 230-242.
Chicago
Wang, M., Li, L., Liu, J., Wang, J."A gene interaction network‑based method to measure the common and heterogeneous mechanisms of gynecological cancer". Molecular Medicine Reports 18, no. 1 (2018): 230-242. https://doi.org/10.3892/mmr.2018.8961