Open Access

Comprehensive gene and pathway analysis of cervical cancer progression

  • Authors:
    • Yuexiong Yi
    • Yan Fang
    • Kejia Wu
    • Yanyan Liu
    • Wei Zhang
  • View Affiliations

  • Published online on: March 3, 2020     https://doi.org/10.3892/ol.2020.11439
  • Pages: 3316-3332
  • Copyright: © Yi 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

Cervical Cancer is one of the leading causes of cancer‑associated mortality in women. The present study aimed to identify key genes and pathways involved in cervical cancer (CC) progression, via a comprehensive bioinformatics analysis. The GSE63514 dataset from the Gene Expression Omnibus database was analyzed for hub genes and cancer progression was divided into four phases (phases I‑IV). Pathway enrichment, protein‑protein interaction (PPI) and pathway crosstalk analyses were performed, to identify key genes and pathways using a criterion nodal degree ≥5. Gene pathway analysis was determined by mapping the key genes into the key pathways. Co‑expression between key genes and their effect on overall survival (OS) time was assessed using The Cancer Genome Atlas database. A total of 3,446 differentially expressed genes with 107 hub genes were identified within the four phases. A total of 14 key genes with 11 key pathways were obtained, following extraction of ≥5 degree nodes from the PPI and pathway crosstalk networks. Gene pathway analysis revealed that CDK1 and CCNB1 regulated the cell cycle and were activated in phase I. Notably, the following terms, ‘pathways in cancer’, ‘focal adhesion’ and the ‘PI3K‑Akt signaling pathway’ ranked the highest in phases II‑IV. Furthermore, FN1, ITGB1 and MMP9 may be associated with metastasis of tumor cells. STAT1 was indicated to predominantly function at the phase IV via cancer‑associated signaling pathways, including ‘pathways in cancer’ and ‘Toll‑like receptor signaling pathway’. Survival analysis revealed that high ITGB1 and FN1 expression levels resulted in significantly worse OS. CDK1 and CCNB1 were revealed to regulate proliferation and differentiation through the cell cycle and viral tumorigenesis, while FN1 and ITGB1, which may be developed as novel prognostic factors, were co‑expressed to induce metastasis via cancer‑associated signaling pathways, including PI3K‑Art signaling pathway, and focal adhesion in CC; however, the underlying molecular mechanisms require further research.

Introduction

Cervical Cancer (CC) is a highly aggressive tumor and is one of the leading causes of cancer-associated mortality in women, with an estimated 570,000 new cases and 311,000 deaths in 2018 worldwide (1). Women with CC are considered to have a lower quality of life (2). The progression of CC, from normal cervical mucosal epithelium to cervical intraepithelial neoplasia (CIN) grade 1, 2, and 3, to CC (3) is associated with persistent high-risk human papillomavirus (HPV) infection (4). Furthermore, a number of risk factors, including early sexual activity (5), multiple sexual partners (6), long-term use of oral contraceptives (7), genetic factors [active oncogenes, including PIK3CA (8), ATAD2 (9) and CRNDE (10); tumor suppressor genes, including p53 (11), Ras association domain family 1 isoform A (12) and NOL7 (13)], tobacco use [current smoker, started smoking age ≤15 years, smoking duration ≥30 years, ≥20 cigarettes/day (14)] and other viral infections (such as HIV, herpes simplex virus (HSV) type II and bacterial infections caused by Chlamydia trachomatis) (15) have been associated with CC progression.

HPV infection plays a leading role in CC (16). The DNA of HPV integrates into the host cell genome [HPV16: q21-q31 of chromosome no. 13; HPV18: q24 of chromosome no. 8 (17)], disrupts the open reading frame and causes overexpression of E6 and E7 genes (18). It has been verified that E6 and E7 exert carcinogenic effects by binding to the cell cycle regulators, p53 and retinoblastoma (Rb) (19). While E6 and E7 proteins are upregulated, E6 can interact with its associated protein [E6-associated protein, E6AP (20)] to form a complex and bind to p53. This binding hydrolyzes p53 and results in the loss of p53-induced negative regulation of cell proliferation, thereby leading to unchecked cellular proliferation and malignant transformation (21). E7 has a high affinity for Rb, which controls the cell cycle. Binding of E7 to Rb can dissociate the Rb-E2F complex, thus releasing E2F to exert its role as a transcription factor, which leads to an uncontrolled cell cycle and cellular immortalization (22,23). Furthermore, centrosomes are central regulators of mitosis that are often increased in numbers in cancer cells (24). A previous study indicated that an abnormally increased number of centrosomes is associated with structural chromosomal abnormality in cervical lesions with high risk of HPV infection (25). Duensing and Münger (26) reported that abnormal number of centrosomes and associated spindle mitotic abnormality can be found in cells infected by the high-risk HPV16 E6 and E7 proteins, but not in cells infected by the low-risk HPV6. Although numerous experimental studies on genes [HPV16 L1 protein (27), sonic hedgehog (28) and FGFR4 (29)], and signaling pathways [Wnt/β-catenin signaling pathway (30), adenosinergic pathway (31) and ERK signal transduction (32)], as well as bioinformatics analyses have focused on microRNAs (33) and genes (34) associated with CC, and have provided an understanding of the pathophysiological mechanisms of the disease over the last decade, the underlying molecular mechanisms remain unclear.

The development of CC occurs over a number of years and its complexity presents clinical challenges in patients screening and treatment. Currently, The Bethesda System (35), which is a tool that is used to report Pap smear results for cervical cytologic diagnoses, provides useful data that allows research into the epidemiology, biology and pathology of cervical lesions; however, its diagnostic value remains poor (36). Instead, direct biopsy remains the gold standard for diagnosis. Nevertheless, invasive examinations may cause adverse psychological effects, including anxiety, depression or distress (37). Surgery, chemotherapy and radiotherapy (38) are the three major therapeutic strategies in the treatment of CC; however, their uses may be limited for various reasons. Surgery may be limited by the status and stage of patients, including late stage or tolerance to anesthesia (39), whereas chemotherapy is limited due to the lack of sensitivity and the development of drug resistance (40). In addition, radiotherapy can be limited by the maximum tolerated dose to adjacent normal tissues (41). Thus, it is essential to understand the underlying molecular mechanisms in the initiation and development of CC, in order to develop methods for its accurate diagnosis and effective treatment. A number of studies have reported that multiple genes [CXCL12 (42), FGFR4 (29) and SHH (43)], proteins [cyclin D1 (44), FOXO1 (45) and BASP1 (46)] and pathways [Toll-like signaling pathway (47), VEGF signaling pathway (48) and Wnt signaling pathway (30)] are involved in the natural progression of CC; however, few studies have investigated the fundamental pathological molecular mechanisms in the progression of CC (from normal, to CIN1, CIN2, CIN3, to cancer). Thus, the specific pathological processes remain unclear.

The present study provided a systematic investigation of the development of CC and further understanding of the associations between the four phases of CC progression, and thus revealed additional targets for the detection and treatment of CC. A flow diagram of the present study is presented in Fig. 1.

Materials and methods

Identification of differentially expressed genes (DEGs)

The CC gene expression profile in the GSE63514 dataset, acquired using the GPL570 platform (Affymetrix Human Genome U133 Plus 2.0 Array) provided by den Boon in 2015 (49), was downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/geo/). The profile contained 128 cervical specimens, including: Normal (n=24), CIN1 (n=14), CIN2 (n=22), CIN3 (n=40) and cancer (n=28) samples. All samples were divided into four phases as follows: Phase I, normal to CIN1; phase II, CIN1 to CIN2; phase III, CIN2 to CIN3 and phase IV, CIN3 to cancer, and GEO2R tools (https://www.ncbi.nlm.nih.gov/geo/geo2r/) (50) within the limma package version 3.26.8 (51) were used to screen the DEGs at the four phases. The criteria fold change (FC) of expression >2 and P<0.05 were used to identify DEGs.

Identification of hub genes

The Search Tool for the Retrieval of Interacting Genes (STRING) database (52) and Cytoscape software (version 3.5.1) (53) were used to identify the hub genes in the four phases. The PPI network was constructed by searching for gene symbols and the minimum required interaction score was set at 0.7, to ensure high confidence in the results. The nodes that not connect to the major network were removed to decrease the error detection rate. CytoHubba (54), a plug-in for Cytoscape software, was used to investigate notable nodes in the interactome network using 12 topological algorithms, including Degree, Edge Percolated Component, Maximum Neighborhood Component, Density of Maximum Neighborhood Component and Maximal Clique Centrality, and centralities based on shortest paths, such as Bottleneck, EcCentricity, Closeness, Radiality, Betweenness, Clustering Coefficient and Stress. The genes that ranked in the top 10 for each topological algorithm were extracted and the duplication of each gene was calculated. Genes duplicated <2 times were excluded, in order to guarantee that the genes were associated with CC. The remaining genes were considered as hub genes in the four phases.

Functional enrichment analyses of GO and pathways

The functional features of the genes associated with the four phases were examined using WebGestalt (55) and ToppGene (56). In WebGestalt, over-representation analysis was selected as the enrichment method, Biological Process in GO as the functional database, gene symbol as the gene ID type and genome as the reference set for enrichment analysis. In ToppGene, two frequently used databases, Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.kegg.jp/) and BioCarta (https://www.biocarta.com/), were utilized to perform pathway enrichment analysis, to improve the reliability of the results. Pathways with a false discovery rate of P<0.05 were considered to indicate significantly enriched pathways.

Pathway crosstalk analysis

Pathway crosstalk analysis was performed (57), to investigate the interactions among the significantly enriched pathways. The pathways with either a false discovery rate of P>0.05 or <3 genes were removed as selection criteria. The number of shared genes between pairwise pathways was calculated and pairwise pathways with <2 overlapping genes were removed. The Jaccard Coefficient (JC) and the Overlap Coefficient (OC) parameters were calculated, to measure the overlap between the pathways. Specifically, JC=|A∩BA∪B|, while OC=|A∩Bmin(|A|,|B|)|, where A and B represent the gene numbers of the tested pathways. The interrelationships between pathways were visualized using Cytoscape software.

To determine the overall progression and further detect an association between two pathways, the KEGG and BioCarta databases were used to identify the upstream or downstream associations between pathways. Furthermore, the nodal degree was calculated using Centiscape (58) to identify key nodes. According to Han et al (59), key nodes are considered as those with a nodal degree ≥5.

Integration of the PPI network

The PPI network (60) was used to identify key proteins for the four phases of CC. As Protein Interaction Network Analysis (PINA) (https://omics.bjcancer.org/pina/) (61) is an integrated platform for protein interaction network construction, analysis and visualization, it can identify the associations between the queried genes based on integration of data from six public PPI databases: IntAct (62), MINT (63), BioGRID (64), DIP (65), HPRD (66) and MIPS MPact (67). Thus, the PINA4MS plug-in for Cytoscape software was used to construct the PPI network, to identify CC progression-associated genes. As PINA4MS requires UniProt accession numbers, the UniProt Retrieve/ID mapping tool (https://www.uniprot.org/uploadlists/) was used to input gene symbols. The key nodes for the PPI network were also extracted using a criterion of nodal degree ≥5.

Comprehensive gene-pathway analysis

To determine the molecular mechanisms and associations between the key genes and pathways, the gene-pathway network was constructed by examining the key pathways, in order to determine which pathway contained at least one of the key genes.

Co-expression and survival analysis for key genes

To identify the co-expression of key genes and their impact on OS time, the LinkedOmics database (68) was used, which was based on TCGA (69). The co-expression analysis was performed using Pearson correlation and OS analysis was assessed with Cox regression method. For survival analysis, samples were divided by the median value of the investigated gene. P<0.05 was considered to indicate a statistically significant difference for both the co-expression correlation and OS time.

Results

Identification of DEGs

Analysis of the GSE63514 dataset using GEO2R, with a criteria of ≥2 FC and P<0.05, identified a total of 3,446 DEGs for the four phases as follows: 446 DEGs in phase I, of which 76 were upregulated and 370 were downregulaged; 382 DEGs in phase II, of which 146 were upregulated and 236 were downregulated; 756 DEGs in phase III, of which 435 were upregulated and 321 were downregulated; 1,862 DEGs in phase IV, of which 816 were upregulated and 1046 were downregulated.

Identification of hub genes

Following removal of 2,256 irrelevant genes (Phase I, 265; Phase II, 197; Phase III, 603; Phase IV, 1191), 12 topological algorithms were used and the top 10 genes for each method were extracted. A total of 107 genes that appeared at least twice were conserved as hub genes, as presented in Table I. A total of 29 genes were identified in phase I, among which five genes were members of the kinesin family (KIF11, KIF15, KIF23, KIF4A and KIF14), and five genes were associated with meiosis and the maturation of oocytes [BUB1B (70), BUB1 (71), CCNA2 (72), CCNB1 (72) and CDK1 (73)], as well as other genes associated with inflammation and innate immune responses [STAT1 (74), GBP1 (75) and RHOA (76)]. A total of 25 hub genes were verified in phase II, among which the involvement of seven interferon-induced genes was identified (IFI44L, IFIT3, IFIF1, IFIF5, IFI44, IFIT2 and IFI6), and several pattern recognition receptor-associated genes [IRF7 (77), STAT1 (78) and CXCL10 (79)], as well as some genes involved in invasion and metastasis of cancer cells [HGF (80), IGF1 (81), KIT (82), FN1 (83) and CXCL12 (84)]. A number of common cancer-associated signaling pathway genes were identified in phase III [CCND1 (85), STAT1 (86) and VEGFA (87)]. A total of three C-X-C motif chemokine ligands (CXCL8, CXCL11 and CXCL4), two integrin subunits (ITGB1 and ITGA1), and one mitogen-activated protein (MAPK12) were identified in phase IV. Furthermore, PIK3CA (88) and FOS (89) participated in cancer-associated pathways in phase IV. The diversity of genes within the four phases demonstrated that CC progression is a complex process and its molecular mechanisms are not constant.

Table I.

Hub genes in phases I–IV.

Table I.

Hub genes in phases I–IV.

PhaseGeneRegulationCountsLogFCP-value
Phase ICDK1+71.24<0.01
KIF11+71.270.01
BUB1B+61.130.02
BUB1+51.170.01
CCNA2+51.01<0.01
HLA-DPA1+51.490.03
CENPE+51.360.02
RHOA+41.47<0.01
KIF15+41.11<0.01
CCNB1+41.230.01
NDC80+41.470.01
TTK+41.14<0.01
STAT14−1.470.03
CXCL10+42.47<0.01
KIF23+41.480.01
KIF4A+31.000.05
PSMB9+31.120.03
GNG2+31.260.04
SPAG5+21.150.01
TRIP13+21.090.02
ANLN+21.190.02
CDKN3+21.83<0.01
KIF14+21.150.01
MKI67+21.450.01
NUSAP1+21.220.02
NEK2+21.300.01
NCAPG+21.18<0.01
DLGAP5+21.54<0.01
GBP1+21.45<0.01
Phase IISTAT19−1.030.02
CXCL106−2.160.02
CXCL126−1.480.04
DCN6−1.370.01
CCL25−1.540.01
KIT5−1.260.05
IGF15−1.86<0.01
OAS24−1.370.01
IRF74−1.080.01
ISG154−1.830.02
FN14−1.340.04
HGF4−1.220.02
HERC63−1.71<0.01
MX23−1.860.01
IFIT33−1.79<0.01
IFIT13−2.96<0.01
GBP13−1.200.01
CDC6+31.400.01
IFIT52−1.11<0.01
IFI62−1.550.01
SP1102−1.43<0.01
IFI442−1.87<0.01
DDX602−1.18<0.01
IFIT22−1.67<0.01
RSAD22−2.66<0.01
Phase IIIBIRC5+91.13<0.01
TOP2A+81.36<0.01
KIF2C+61.04<0.01
MCM10+61.160.01
VEGFA+61.27<0.01
MAD2L1+51.03<0.01
KIF15+51.45<0.01
ASPM+51.70<0.01
FOXM1+51.180.01
MX2+41.340.01
STAT1+41.20<0.01
PLXNA44−1.150.01
AR4−1.69<0.01
CCND131.20<0.01
OAS2+31.05<0.01
ACLY+31.550.01
GNG2+3−1.21<0.01
RSAD2+21.85<0.01
ISG15+21.82<0.01
IFI35+21.350.01
IRF5+21.46<0.01
SAMHD1+21.05<0.01
MKI67+21.20<0.01
PLK4+21.030.01
AHCTF1+21.03<0.01
NUDC+21.07<0.01
EXO1+21.29<0.01
PLXNA3+21.010.01
MMP9+22.55<0.01
PLAUR+21.01<0.01
Phase IVPIK3CA+91.42<0.01
CXCL8+81.280.05
ITGB1+82.20<0.01
PTK2+81.33<0.01
GNG2+61.180.05
ITGA1+61.33<0.01
GNG126−1.14<0.01
FOS5−1.130.05
EDN1+51.07<0.01
NMU4−2.23<0.01
LPAR54−1.36<0.01
STAT1+31.75<0.01
FN1+33.61<0.01
GSTM13−1.090.02
PLA2G4A3−1.410.02
CXCR4+21.58<0.01
HCAR32−1.60<0.01
S1PR52−1.56<0.01
CXCL52−2.100.01
NQO12−1.380.01
CXCL11+21.690.02
COMP+21.620.01
MAPK12+21.38<0.01

[i] +, upregulated; -, downregulated.

GO enrichment analysis of hub genes

To further identify the biological functions and locations of hub genes, GO enrichment analysis (90) was performed (Fig. 2). Hub genes were notably enriched in ‘biological regulation’, ‘metabolic process’ and ‘cellular component organization’ in phase I and II, while ‘responses to stimulus’ and ‘biological regulation’ were predominantly enriched at phases III–IV in biological process. For the cellular components, ‘nucleus’, ‘membrane-enclosed lumen’ and ‘macromolecular complex’ was enriched at phases I–III, while ‘chromosome’ and ‘membrane’ was identified in phases II and IV, respectively. ‘Protein binding’ was enriched at all four phases for Molecular Function. Furthermore, ‘nucleic acid binding’ and ‘hydrolase activity’ were enriched at phase II and III, while ‘ion binding’ was enriched at phase III and IV.

Pathway enrichment analysis of hub genes

As presented in Table II, a total of 10 notably enriched pathways were identified at phase I, of which five pathways were associated with virus infections including, ‘influenza A’, ‘tuberculosis’, ‘herpes simplex infection’, ‘viral carcinogenesis’ and ‘Epstein-Barr virus infection’, and additional pathways involved in the ‘cell cycle’, ‘oocyte meiosis’ and ‘progesterone-mediated oocyte maturation’. Furthermore, the chemokine signaling pathway was also identified in phase I. The RIG-I-like receptor and Toll-like receptor signaling pathways were identified in phase II, and are associated with pattern-recognition receptors (91). In addition, several pathways, including ‘focal adhesion’, ‘Rap1 signaling pathway’, ‘Ras signaling pathway’, ‘PI3K-Akt signaling pathway’ and ‘Proteoglycans in cancer’ were associated with invasion and metastasis (9296). The two common cancer-associated signaling pathways ‘Pathways in cancer’ and ‘Proteoglycans in cancer’, were enriched in phase III, while 70 pathways were significant enriched at phase IV (P<0.05). Apart from the common cancer-associated signaling pathways and virus infection pathways at phase IV, the ‘IL-17 signaling pathway’, ‘VEGF signaling pathway’ and ‘endocrine resistance’ also were also demonstrated to be associated with CC progression. Furthermore, the ‘AGE-RAGE signaling pathway in diabetic complications’ was also identified at phases II–IV.

Table II.

Pathway enrichment analysis for phases I–IV.

Table II.

Pathway enrichment analysis for phases I–IV.

PhasePathwayFDRInvolved genes
Phase ICell cycle<0.001CDK1, TTK, CCNA2, BUB1, CCNB1, BUB1B
Progesterone-mediated oocyte maturation0.002CDK1, CCNA2, BUB1, CCNB1
Chemokine signaling pathway0.006RHOA, CXCL10, STAT1, GNG2
Oocyte meiosis0.018CDK1, BUB1, CCNB1
NOD-like receptor signaling pathway0.036RHOA, GBP1, STAT1
Influenza A0.036HLA-DPA1, CXCL10, STAT1
Tuberculosis0.037RHOA, HLA-DPA1, STAT1
Herpes simplex infection0.038CDK1, HLA-DPA1, STAT1
Viral carcinogenesis0.044RHOA, CDK1, CCNA2
Epstein-Barr virus infection0.044CDK1, HLA-DPA1, CCNA2
Phase IIInfluenza A<0.001CCL2, OAS2, IRF7, RSAD2, CXCL10, STAT1
NOD-like receptor signaling pathway<0.001GBP1, CCL2, OAS2, IRF7, STAT1
Herpes simplex infection<0.001CCL2, OAS2, IRF7, IFIT1, STAT1
Pathways in cancer0.001HGF, IGF1, FN1, KIT, CXCL12, STAT1
Hepatitis C0.001OAS2, IRF7, IFIT1, STAT1
Cytokine-cytokine receptor interaction0.001HGF, CCL2, KIT, CXCL10, CXCL12
RIG-I-like receptor signaling pathway0.003IRF7, ISG15, CXCL10
Chemokine signaling pathway0.003CCL2, CXCL10, CXCL12, STAT1
Genes encoding secreted soluble factors0.003HGF, CCL2, IGF1, CXCL10, CXCL12
Proteoglycans in cancer0.004HGF, IGF1, FN1, DCN
AGE-RAGE signaling pathway in diabetic complications0.005CCL2, FN1, STAT1
Toll-like receptor signaling pathway0.006IRF7, CXCL10, STAT1
Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins0.008HGF, CCL2, IGF1, FN1, DCN, CXCL10, CXCL12
Measles0.009OAS2, IRF7, STAT1
PI3K-Akt signaling pathway0.015HGF, IGF1, FN1, KIT
Focal adhesion0.024HGF, IGF1, FN1
Rap1 signaling pathway0.026HGF, IGF1, KIT
Ras signaling pathway0.029HGF, IGF1, KIT
Ensemble of genes encoding ECM-associated proteins including ECM-affilaited proteins, ECM regulators and secreted factors0.032HGF, CCL2, IGF1, CXCL10, CXCL12
Phase IIIPathways in cancer0.002BIRC5, CCND1, MMP9, AR, STAT1, GNG2, VEGFA
Bladder cancer0.006CCND1, MMP9, VEGFA
Hepatitis B0.011BIRC5, CCND1, MMP9, STAT1
Pancreatic cancer0.012CCND1, STAT1, VEGFA
Proteoglycans in cancer0.025PLAUR, CCND1, MMP9, VEGFA
AGE-RAGE signaling pathway in diabetic complications0.027CCND1, STAT1, VEGFA
Phase IVChemokine signaling pathway<0.001GNG12, CXCL11, CXCL5, PIK3CA, CXCR4, PTK2, STAT1, CXCL8, GNG2
Pathways in cancer<0.001FN1, LPAR5, GNG12, ITGB1, PIK3CA, CXCR4, FOS, PTK2, STAT1, CXCL8, GNG2
Fluid shear stress and atherosclerosis<0.001GSTM1, NQO1, MAPK12, PIK3CA, FOS, EDN1, PTK2
Signaling of Hepatocyte Growth Factor Receptor<0.001ITGA1, ITGB1, PIK3CA, FOS, PTK2
PI3K-Akt signaling pathway<0.001ITGA1, FN1, COMP, LPAR5, GNG12, ITGB1, PIK3CA, PTK2, GNG2
B Cell Survival Pathway<0.001ITGA1, ITGB1, PIK3CA, FOS
AGE-RAGE signaling pathway in diabetic complications<0.001MAPK12, FN1, PIK3CA, EDN1, STAT1, CXCL8
Toll-like receptor signaling pathway<0.001MAPK12, CXCL11, PIK3CA, FOS, STAT1, CXCL8
Aspirin Blocks Signaling Pathway Involved in Platelet Activation<0.001PLA2G4A, ITGA1, ITGB1, PTK2
Erk and PI-3 Kinase Are Necessary for Collagen Binding in Corneal Epithelia<0.001ITGA1, ITGB1, PIK3CA, PTK2
Pertussis<0.001MAPK12, CXCL5, ITGB1, FOS, CXCL8
Focal adhesion<0.001ITGA1, FN1, COMP, ITGB1, PIK3CA, PTK2
TNF signaling pathway<0.001MAPK12, CXCL5, PIK3CA, FOS, EDN1
Leukocyte transendothelial migration<0.001MAPK12, ITGB1, PIK3CA, CXCR4, PTK2
Regulation of actin cytoskeleton<0.001ITGA1, FN1, GNG12, TGB1, PIK3CA, PTK2
VEGF signaling pathway<0.001PLA2G4A, MAPK12, PIK3CA, PTK2
PTEN dependent cell cycle arrest and apoptosis<0.001ITGB1, PIK3CA, PTK2
uCalpain and friends in Cell spread<0.001ITGA1, ITGB1, PTK2
Trefoil Factors Initiate Mucosal Healing<0.001ITGB1, PIK3CA, PTK2
Prolactin signaling pathway<0.001MAPK12, PIK3CA, FOS, STAT1
Leishmaniasis<0.001MAPK12, ITGB1, FOS, STAT1
Inhibition of Cellular Proliferation by Gleevec<0.001PIK3CA, FOS, STAT1
CXCR4 Signaling Pathway<0.001PIK3CA, CXCR4, PTK2
TPO Signaling Pathway<0.001PIK3CA, FOS, STAT1
Bacterial invasion of epithelial cells<0.001FN1, ITGB1, PIK3CA, PTK2
mCalpain and friends in Cell motility<0.001ITGA1, ITGB1, PTK2
ECM-receptor interaction<0.001ITGA1, FN1, COMP, ITGB1
Small cell lung cancer<0.001FN1, ITGB1, PIK3CA, PTK2
EGF Signaling Pathway<0.001PIK3CA, FOS, STAT1
IL-17 signaling pathway<0.001MAPK12, CXCL5, FOS, CXCL8
PDGF Signaling Pathway<0.001PIK3CA, FOS, STAT1
Amoebiasis<0.001FN1, PIK3CA, PTK2, CXCL8
Endocrine resistance<0.001MAPK12, PIK3CA, FOS, PTK2
Proteoglycans in cancer<0.001MAPK12, FN1, ITGB1, PIK3CA, PTK2
Chagas disease (American trypanosomiasis)<0.001MAPK12, PIK3CA, FOS, CXCL8
Agrin in Postsynaptic Differentiation<0.001ITGA1, ITGB1, PTK2
Integrin Signaling Pathway<0.001ITGA1, ITGB1, PTK2
Fc Epsilon Receptor I Signaling in Mast Cells<0.001PLA2G4A, PIK3CA, FOS
Cholinergic synapse<0.001GNG12, PIK3CA, FOS, GNG2
Platelet activation0.001PLA2G4A, MAPK12, ITGB1, PIK3CA
Osteoclast differentiation0.001MAPK12, PIK3CA, FOS, STAT1
Dopaminergic synapse0.001MAPK12, GNG12, FOS, GNG2
Hepatitis C0.001MAPK12, PIK3CA, STAT1, CXCL8
Cytokine-cytokine receptor interaction0.001CXCL11, CXCL5, CXCR4, ACKR3, CXCL8
Hepatitis B0.001PIK3CA, FOS, STAT1, CXCL8
Phospholipase D signaling pathway0.001PLA2G4A, LPAR5, PIK3CA, CXCL8
Shigellosis0.001MAPK12, ITGB1, CXCL8
Fc epsilon RI signaling pathway0.002PLA2G4A, MAPK12, PIK3CA
Influenza A0.002MAPK12, PIK3CA, STAT1, CXCL8
Axon guidance0.002ITGB1, PIK3CA, CXCR4, PTK2
Integrin Signaling Pathway0.002ITGA1, PIK3CA, PTK2
Salmonella infection0.003MAPK12, FOS, CXCL8
MAPKinase Signaling Pathway0.003MAPK12, FOS, STAT1
Rap1 signaling pathway0.003MAPK12, LPAR5, ITGB1, PIK3CA
Rheumatoid arthritis0.003CXCL5, FOS, CXCL8
Th1 and Th2 cell differentiation0.003MAPK12, FOS, STAT1
Circadian entrainment0.003GNG12, FOS, GNG2
Inflammatory mediator regulation of TRP channels0.003PLA2G4A, MAPK12, PIK3CA
Ras signaling pathway0.003PLA2G4A, GNG12, PIK3CA, GNG2
Choline metabolism in cancer0.003PLA2G4A, PIK3CA, FOS
Retrograde endocannabinoid signaling0.003 MAPK12,GNG12,GNG2
T cell receptor signaling pathway0.004MAPK12, PIK3CA, FOS
Th17 cell differentiation0.004MAPK12, FOS, STAT1
Serotonergic synapse0.004PLA2G4A, GNG12, GNG2
Toxoplasmosis0.004MAPK12, ITGB1, STAT1
Glutamatergic synapse0.004PLA2G4A, GNG12, GNG2
MAPK signaling pathway0.004PLA2G4A, MAPK12, GNG12, FOS
Sphingolipid signaling pathway0.005S1PR5, MAPK12, PIK3CA
NOD-like receptor signaling pathway0.012MAPK12, STAT1, CXCL8
cAMP signaling pathway0.018HCAR3, PIK3CA, FOS

[i] FDR, false discovery rate.

Pathway crosstalk analysis

The pathway crosstalk analysis results are presented in Fig. S1. A total of two major modules were identified in phase I, one of which was predominantly associated with inflammatory responses to viral infections, such as the NOD-like receptor signaling pathway and the chemokine signaling pathway, while the other module was associated with cycle regulation of oocytes, including cell cycle, progesterone-mediated oocyte maturation and oocyte meiosis (Fig. S1A). Similarly, the pathways were grouped into two modules in phase II. One module consisted of immune responses (RIG-I-like receptor and Toll-like receptor signaling pathways, and NOD-like receptor signaling pathway), which may trigger rapid activation of innate immunity by inducing the production of proinflammatory cytokines (9799). The other module was predominantly involved in the regulation of cell proliferation and invasion (Ras signaling pathway, PI3K-Akt signaling pathway, Rap1 signaling pathway, focal adhesion and other cancer-associated signaling pathways) (Fig. S1B). Notably, the two modules in phases I and II were not independent as they were demonstrated to connect with each other via several signaling pathways. All pathways formed a cluster and common cancer-associated signaling pathways were indicated to play a critical role in phase III (Fig. S1C), while the connection between the pathways became highly complex in phase IV (Fig. S1D). The possible molecular mechanisms, such as inflammation caused by virus infections, pathways associated with cell invasion, IL-17 and VEGF signaling pathways, and endocrine resistance, are associated with one another. These results further verified the complexity of CC. A comprehensive combination of pathway crosstalk analysis containing 47 nodes and 105 edges among the four phases is presented in Fig. 3A. By analyzing the nodes with degrees ≥5, a subnetwork containing 11 key pathways was extracted (Fig. 3B). It indicates that the MAPK signaling pathway (degree=33), PI3K-Akt signaling pathway (degree=21) and focal adhesion (degree=15), which are ranked as the top three nodes and most interactive, may play critical roles in the progression of CC.

PPI network analysis

A PPI network containing 51 nodes and 78 edges was constructed (Fig. 4A) by downloading the hub genes into the PINA database. Based on the description of a previous study (59), which defined the main nodes as nodes with degree >5, 14 key genes were identified from the PPI network (Fig. 4B). CDK1, FN1 and ITGB1 rank first, second and third, respectively, as the top three-degree levels (16, 12 and 8, respectively). STAT1 was the only gene demonstrated to be involved at all four phases. Furthermore, MMP9 presented self-regulating functions and was demonstrated to co-express with FN1 and ITGB1.

Comprehensive gene-pathway analysis

After mapping the key genes onto the key pathways using the KEGG and BioCarta databases, a potential gene-pathway flowchart, including eight key pathways and six key genes was constructed (Fig. 5). The results demonstrated the following: For phase I, CDK1 and CCNB1 participated in the regulation of the cell cycle, while CDK1 was also involved in viral carcinogenesis; for phases II–IV, ‘pathways in cancer,’ ‘focal adhesion’ and ‘PI3K-Akt signaling pathway’ were ranked the top three pathways according to the number of genes involved; FN1, ITGB1 and MMP9 may be associated with metastasis of tumor cells, and STAT1 participated in ‘pathways in cancer’ and ‘Toll-like receptor signaling pathway’, which functioned at a phase IV.

Co-expression network and survival analysis for key genes

By mining the data from LinkedOmics, the results of co-expression demonstrated that CDK1 had a significantly positive correlation with CCNB1 (P<0.0001), but negative correlation with FN1 (P=0.003) and MMP9 (P=0.001), respectively (Fig. 6). CCNB1 demonstrated a significantly negative correlation with ITGB1 (P=0.047); however, FN1, ITGB1 and MMP9 indicated a significantly positive correlation between each other (FN1 and ITGB1, P<0.001; FN1 and MMP9, P<0.0001; ITGB1 and MMP9, P=0.023). STAT1 was significantly positively correlated with MMP9 (P<0.0001).

Survival analysis indicated that patients with higher FN1 and ITGB1 expression levels had a significantly worse OS time (FN1, P=0.00080; ITGB1, P=0.00005; Fig. 7). However, CDK1, CCNB1, MMP9 and STAT1 were not demonstrated to have a significant effect on OS.

Discussion

To date, the occurrence and development of CC is hypothesized to be linked with persistent HPV infection (100); however, the specific molecular mechanisms require further investigation. In addition, although a number of studies are examining the molecular mechanisms of CC (101104), the detailed pathological process remains unclear.

The results of the present study indicated that CDK1 had the highest degree, and participated in the cell cycle with CCNB1 and viral carcinogenesis in phase I. CDK1 is one of the major cell cycle regulatory proteins and operates at the center of the cell cycle regulatory network (105). It regulates the G-S phase transition and initiates DNA replication (106). Furthermore, as a core molecule at the M phase checkpoint, CDK1 plays a role in the regulation of G2 phase, at M phase in the cell cycle (107). The HPV infection pathway is regulated within the viral carcinogenesis pathway (19). E6 proteins inactivate p53 by binding (108), while p53 negatively regulates CDK1 transcription under normal physiological conditions (109). In addition, the activation of cyclin B1-CDK1 is the key event that initiates the start of mitosis (110). Centrosome separation can be regulated by CDK1 (111), and cyclin B1-CDK1 remains activated following centrosome separation (110). Hence, overexpression of CDK1 can cause dysfunction in cell cycle progression, failure of normal proliferation and differentiation, and thereby lead to malignant proliferation of cancer cells and the formation of CC. CCNB1 is a notable member of the cyclin family, a key initiator and a stringent quality control step of mitosis (112). It also plays a key role in the regulation of CDK1, and its phosphorylated substrates can promote the transition of the cell cycle from G2 to mitosis (113,114). Amplification of the HPV genome depends on prolongation of the G2 phase in the cell cycle (115). CCNB1 is a downstream target of STAT3, which is a key gene that regulates the proliferation and differentiation of CC cells (116). In cells with inactivated STAT3, CCNB1 expression is downregulated and amplification of the HPV genome is also decreased, resulting in decreased activity of CC cells (116). As the results of the present study demonstrated that CDK1 and CCNB1 occurred in phase I and functioned as regulators of proliferation and differentiation, they may be potential promoters of CIN and CC.

In the process of tumor invasion and metastasis, cancer cells can bind to ligands of the extracellular matrix (ECM) via integrins and degrade the basement membrane (BM) by secreting proteases via the pathways of focal adhesion and the PI3K-Akt signaling pathway (117). This degradation is also the prerequisite for stromal infiltration and cancer cell migration (118). ITGB1 belongs to the integrin family and FN1 is the ligand. The binding of ITGB1 and FN1 induces the phosphorylation of tyrosine and directly affects cytoskeleton reconstruction and signal transduction activities of the Ras-MAPK signaling pathway via the RAP1 signaling pathway, which initiates the expression of MMP genes (119). MMPs are a family of calcium and zinc-dependent proteases that degrade a variety of components of the ECM (120). Collagen type IV is the main scaffold in the BM of the ECM and also the main substrate of MMP9 (121). MMP9 can decompose the nestin in the BM to destroy the cells integrity and promote the invasion and metastasis of cancer cells (122). MMP9 expression in HPV-positive patients with CC is higher than in HPV-negative patients (123). Cardeal et al (124) reported that MMP9 is upregulated in human keratinocytes expressing the HPV16 E7 protein. This may be due to TIMP2, an inhibitor of MMP9, which could be downregulated by HPV16 E7. It was also demonstrated that HPV can directly regulate the activity of MMP9 in lung cancer cells (125). There may be an association between HPV infection and the MMP family, which may be beneficial in the diagnosis of cervical precancerous lesions and CC as MMP9 may be considered as a novel biomarker. However, the specific molecular mechanisms require further investigation. As FN1 and ITGB1 were targets of miR-9-3p (126) and FN1 promoted migration and invasion by upregulating MMP9 in cancer (127), it is not surprising that these three genes are co-expressed as a reaction triplet. Furthermore, since higher levels of FN1 and ITGB1 are significantly associated with lower OS rate, these two genes may be developed as novel prognostic factors for CC.

STAT1, the only gene that participates in all four phases, in the present study, is involved in the cancer pathway at phases II, III and IV. It has been reported that STAT1 is upregulated in both CIN1 and CC (128), and the results of the present study that STAT1 is upregulated in phase I, III and IV confirmed this finding. A previous study demonstrated that activated STAT1 plays a tumor suppressive role in breast cancer cells (129). Nevertheless, STAT1 also exerts tumor promoter effects under specific conditions (130). In some malignant diseases, including breast and lung cancers, STAT1 can act as an oncoprotein or a tumor suppressor of the same cell type based on the specific genetic background (130). In CC, STAT1 may have a protective effect in the early stages of HPV infection but may act as a proto-oncogene during the invasive phase of the disease (128). STAT1 can promote cancer cell death by activating p53 expression, and it plays a role in immunosurveillance, and the inhibition of angiogenesis and metastasis in cancer cells (130); however, STAT1 can also promote tumor invasion and metastasis in chronic inflammation (131). The effect of STAT1 in CC still remains unclear; therefore, further verification is required as it may be a key target for the treatment of CC.

The current study presented several limitations. First, as this was an in-silico study, the identification of DEGs may change with additional data, thus the results of subsequent analyses may change accordingly. Secondly, some genes were excluded to decrease the false-positive rate; however, these genes may also have a vital effect on CC. Thirdly, although several genes associated with HPV infection (TP53TG1, RAC1, PAK2 and LTBP2) were identified in DEGs, HPV infection was not observed in the pathway analyses. This may be due to the fact that an insufficient amount of hub genes were identified, or the genes had a low or moderate effect on the HPV infection pathway.

In conclusion, the present study revealed that CDK1, CCNB1, ITGB1, FN1, MMP9 and STAT1 played different roles in the progression of CC through different signaling pathways. CDK1 and CCNB1 served as regulators of proliferation and differentiation via regulation of the cell cycle and viral tumorigenesis, and initiated CIN and CC, whereas FN1, ITGB1 and MMP9 were co-expressed as a reaction triplet to trigger metastasis via cancer pathways, PI3K-Akt signaling pathway and focal adhesion. FN1 and ITGB1 may be novel prognostic factors for CC. STAT1 may have a protective effect in the early stage of HPV infection, but may also act as a proto-oncogene during the invasive stage; however, the specific molecular mechanisms require further investigation.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated and/or analyzed during this study are included in this published article. The datasets generated and/or analyzed during the current study are available in the GEO (https://www.ncbi.nlm.nih.gov/geo/) and LinkOmics (http://www.linkedomics.org/) repository.

Authors' contributions

YY, YF and WZ designed the present study and drafted the initial manuscript. KW and YL performed the literature review, acquired the data and performed the statistical analyses. All authors have read and approved the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CC

cervical cancer

CIN

cervical intraepithelial neoplasia

GO

Gene Ontology

PPI

protein-protein interaction

TCGA

The Cancer Genome Atlas

DEG

differentially expressed gene

OS

overall survival

STRING

Search Tool for the Retrieval of Interacting Genes

KEGG

Kyoto Encyclopedia of Genes and Genomes

JC

jaccard coefficient

OC

overlap coefficient

PINA

protein interaction network analysis

References

1 

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

2 

Muliira RS, Salas AS and O'Brien B: Quality of life among female cancer survivors in Africa: An integrative literature review. Asia Pac J Oncol Nurs. 4:6–17. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Gravitt PE and Winer RL: Natural history of HPV infection across the lifespan: Role of viral latency. Viruses. 9:E2672017. View Article : Google Scholar : PubMed/NCBI

4 

Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, Snijders PJ, Peto J, Meijer CJ and Muñoz N: Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol. 189:12–19. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Louie KS, de Sanjose S, Diaz M, Castellsagué X, Herrero R, Meijer CJ, Shah K, Franceschi S, Muñoz N and Bosch FX; International Agency for Research on Cancer Multicenter Cervical Cancer Study Group, : Early age at first sexual intercourse and early pregnancy are risk factors for cervical cancer in developing countries. Br J Cancer. 100:1191–1197. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Liu ZC, Liu WD, Liu YH, Ye XH and Chen SD: Multiple sexual partners as a potential independent risk factor for cervical cancer: A meta-analysis of epidemiological studies. Asian Pac J Cancer Prev. 16:3893–3900. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Smith JS, Green J, Berrington de Gonzalez A, Appleby P, Peto J, Plummer M, Franceschi S and Beral V: Cervical cancer and use of hormonal contraceptives: A systematic review. Lancet. 361:1159–1167. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Ma YY, Wei SJ, Lin YC, Lung JC, Chang TC, Whang-Peng J, Liu JM, Yang DM, Yang WK and Shen CY: PIK3CA as an oncogene in cervical cancer. Oncogene. 19:2739–2744. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Zheng L, Li T, Zhang Y, Guo Y, Yao J, Dou L and Guo K: Oncogene ATAD2 promotes cell proliferation, invasion and migration in cervical cancer. Oncol Rep. 33:2337–2344. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Bai X, Wang W, Zhao P, Wen J, Guo X, Shen T, Shen J and Yang X: LncRNA CRNDE acts as an oncogene in cervical cancer through sponging miR-183 to regulate CCNB1 expression. Carcinogenesis. Oct 12–2019.(Epub ahead of print).

11 

Bremer GL, Tieboschb AT, van der Putten HW, de Haan J and Arends JW: p53 tumor suppressor gene protein expression in cervical cancer: Relationship to prognosis. Eur J Obstet Gynecol Reprod Biol. 63:55–59. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Cohen Y, Singer G, Lavie O, Dong SM, Beller U and Sidransky D: The RASSF1A tumor suppressor gene is commonly inactivated in adenocarcinoma of the uterine cervix. Clin Cancer Res. 9:2981–2984. 2003.PubMed/NCBI

13 

Hasina R, Pontier AL, Fekete MJ, Martin LE, Qi XM, Brigaudeau C, Pramanik R, Cline EI, Coignet LJ and Lingen MW: NOL7 is a nucleolar candidate tumor suppressor gene in cervical cancer that modulates the angiogenic phenotype. Oncogene. 25:588–598. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Roura E, Castellsagué X, Pawlita M, Travier N, Waterboer T, Margall N, Bosch FX, de Sanjosé S, Dillner J, Gram IT, et al: Smoking as a major risk factor for cervical cancer and pre-cancer: Results from the EPIC cohort. Int J cancer. 135:453–466. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Husain RS and Ramakrishnan V: Global variation of human papillomavirus genotypes and selected genes involved in cervical malignancies. Ann Glob Health. 81:675–683. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Soto D, Song C and McLaughlin-Drubin ME: Epigenetic alterations in human papillomavirus-associated cancers. Viruses. 9:E2482017. View Article : Google Scholar : PubMed/NCBI

17 

Mincheva A, Gissmann L and zur Hausen H: Chromosomal integration sites of human papillomavirus DNA in three cervical cancer cell lines mapped by in situ hybridization. Med Microbiol Immunol. 176:245–256. 1987. View Article : Google Scholar : PubMed/NCBI

18 

Senapati R, Senapati NN and Dwibedi B: Molecular mechanisms of HPV mediated neoplastic progression. Infect Agent Cancer. 11:592016. View Article : Google Scholar : PubMed/NCBI

19 

Münger K, Baldwin A, Edwards KM, Hayakawa H, Nguyen CL, Owens M, Grace M and Huh K: Mechanisms of human papillomavirus-induced oncogenesis. J Virol. 78:11451–11460. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Martinez-Zapien D, Ruiz FX, Poirson J, Mitschler A, Ramirez J, Forster A, Cousido-Siah A, Masson M, Vande Pol S, Podjarny A, et al: Structure of the E6/E6AP/p53 complex required for HPV-mediated degradation of p53. Nature. 529:541–545. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Scheffner M, Huibregtse JM, Vierstra RD and Howley PM: The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell. 75:495–505. 1993. View Article : Google Scholar : PubMed/NCBI

22 

Sherr CJ: The Pezcoller lecture: Cancer cell cycles revisited. Cancer Res. 60:3689–3695. 2000.PubMed/NCBI

23 

Martin LG, Demers GW and Galloway DA: Disruption of the G1/S transition in human papillomavirus type 16 E7-expressing human cells is associated with altered regulation of cyclin E. J Virol. 72:975–985. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Löffler H, Fechter A, Matuszewska M, Saffrich R, Mistrik M, Marhold J, Hornung C, Westermann F, Bartek J and Krämer A: Cep63 recruits Cdk1 to the centrosome: Implications for regulation of mitotic entry, centrosome amplification, and genome maintenance. Cancer Res. 71:2129–2139. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Skyldberg B, Fujioka K, Hellström AC, Sylvén L, Moberger B and Auer G: Human papillomavirus infection, centrosome aberration, and genetic stability in cervical lesions. Mod Pathol. 14:279–284. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Duensing S and Münger K: The human papillomavirus type 16 E6 and E7 oncoproteins independently induce numerical and structural chromosome instability. Cancer Res. 62:7075–7082. 2002.PubMed/NCBI

27 

Chaiwongkot A, Niruthisard S, Kitkumthorn N and Bhattarakosol P: Quantitative methylation analysis of human papillomavirus 16 L1 gene reveals potential biomarker for cervical cancer progression. Diagn Microbiol Infect Dis. 89:265–270. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Sharma A, De R, Javed S, Srinivasan R, Pal A and Bhattacharyya S: Sonic hedgehog pathway activation regulates cervical cancer stem cell characteristics during epithelial to mesenchymal transition. J Cell Physiol. Feb 4–2019.(Epub ahead of print). View Article : Google Scholar

29 

Li YP, Zhang L, Zou YL and Yu Y: Association between FGFR4 gene polymorphism and high-risk HPV infection cervical cancer. Asian Pac J Trop Med. 10:680–684. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Zhang J and Gao Y: CCAT-1 promotes proliferation and inhibits apoptosis of cervical cancer cells via the Wnt signaling pathway. Oncotarget. 8:68059–68070. 2017.PubMed/NCBI

31 

Mora-García ML, Ávila-Ibarra LR, García-Rocha R, Weiss-Steider B, Hernández-Montes J, Don-López CA, Gutiérrez-Serrano V, Titla-Vilchis IJ, Fuentes-Castañeda MC, Monroy-Mora A, et al: Cervical cancer cells suppress effector functions of cytotoxic T cells through the adenosinergic pathway. Cell Immunol. 320:46–55. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Song ZC, Ding L, Ren ZY, Sun XS, Yang Q, Wang L, Feng MJ, Liu CL and Wang JT: Effects of Src on cervical cancer cells proliferation and apoptosis through ERK signal transduction pathway. Zhonghua Liu Xing Bing Xue Za Zhi. 38:1246–1251. 2017.(In Chinese; Abstract available in Chinese from the publisher). PubMed/NCBI

33 

Xu Z, Zhou Y, Shi F, Cao Y, Dinh TLA, Wan J and Zhao M: Investigation of differentially-expressed microRNAs and genes in cervical cancer using an integrated bioinformatics analysis. Oncol Lett. 13:2784–2790. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Chen SZ, Ma WL and Zheng WL: Screening for cervical cancer-related genes and their bioinformatics analysis. Nan Fang Yi Ke Da Xue Xue Bao. 28:585–588. 2008.(In Chinese). PubMed/NCBI

35 

Nayar R and Wilbur DC: The bethesda system for reporting cervical cytology: A historical perspective. Acta Cytol. 61:359–372. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Shoji T, Takatori E, Takeuchi S, Yoshizaki A, Uesugi N, Sugai T and Sugiyama T: Clinical significance of atypical glandular cells in the bethesda system 2001: A comparison with the histopathological diagnosis of surgically resected specimens. Cancer Invest. 32:105–109. 2014. View Article : Google Scholar : PubMed/NCBI

37 

O'Connor M, Gallagher P, Waller J, Martin CM, O'Leary JJ and Sharp L; Irish Cervical Screening Research Consortium (CERVIVA), : Adverse psychological outcomes following colposcopy and related procedures: A systematic review. BJOG. 123:24–38. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Haque N, Uddin AFMK, Dey BR, Islam F and Goodman A: Challenges to cervical cancer treatment in Bangladesh: The development of a women's cancer ward at Dhaka Medical College Hospital. Gynecol Oncol Rep. 21:67–72. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Roque DR, Wysham WZ and Soper JT: The surgical management of cervical cancer: An overview and literature review. Obstet Gynecol Surv. 69:426–441. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Marin JJ, Romero MR, Blazquez AG, Herraez E, Keck E and Briz O: Importance and limitations of chemotherapy among the available treatments for gastrointestinal tumours. Anticancer Agents Med Chem. 9:162–184. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Chen HHW and Kuo MT: Improving radiotherapy in cancer treatment: Promises and challenges. Oncotarget. 8:62742–62758. 2017.PubMed/NCBI

42 

Lecavalier-Barsoum M, Chaudary N, Han K, Pintilie M, Hill RP and Milosevic M: Targeting CXCL12/CXCR4 and myeloid cells to improve the therapeutic ratio in patient-derived cervical cancer models treated with radio-chemotherapy. Br J Cancer. 121:249–256. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Zhang F, Ren CC, Liu L, Chen YN, Yang L, Zhang XA, Wang XM and Yu FJ: SHH gene silencing suppresses epithelial-mesenchymal transition, proliferation, invasion, and migration of cervical cancer cells by repressing the hedgehog signaling pathway. J Cell Biochem. 119:3829–3842. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Gu J, Zhang X, Yang Z and Wang N: Expression of cyclin D1 protein isoforms and its prognostic significance in cervical cancer. Cancer Manag Res. 11:9073–9083. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Chay DB, Han GH, Nam S, Cho H, Chung JY and Hewitt SM: Forkhead box protein O1 (FOXO1) and paired box gene 3 (PAX3) overexpression is associated with poor prognosis in patients with cervical cancer. Int J Clin Oncol. 24:1429–1439. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Tang H, Wang Y, Zhang B, Xiong S, Liu L, Chen W, Tan G and Li H: High brain acid soluble protein 1(BASP1) is a poor prognostic factor for cervical cancer and promotes tumor growth. Cancer Cell Int. 17:972017. View Article : Google Scholar : PubMed/NCBI

47 

Yang X, Cheng Y and Li C: The role of TLRs in cervical cancer with HPV infection: A review. Signal Transduct Target Ther. 2:170552017. View Article : Google Scholar : PubMed/NCBI

48 

Frumovitz M and Sood AK: Vascular endothelial growth factor (VEGF) pathway as a therapeutic target in gynecologic malignancies. Gynecol Oncol. 104:768–778. 2007. View Article : Google Scholar : PubMed/NCBI

49 

den Boon JA, Pyeon D, Wang SS, Horswill M, Schiffman M, Sherman M, Zuna RE, Wang Z, Hewitt SM, Pearson R, et al: Molecular transitions from papillomavirus infection to cervical precancer and cancer: Role of stromal estrogen receptor signaling. Proc Natl Acad Sci USA. 112:E3255–E3264. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res. 41:D991–D995. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

52 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43:D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Chin CH, Chen SH, Wu HH, Ho CW, Ko MT and Lin CY: cytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 8 (Suppl 4):S112014. View Article : Google Scholar : PubMed/NCBI

55 

Wang J, Vasaikar S, Shi Z, Greer M and Zhang B: WebGestalt 2017: A more comprehensive, powerful, flexible and interactive gene set enrichment analysis toolkit. Nucleic Acids Res. 45:W130–W137. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Chen J, Bardes EE, Aronow BJ and Jegga AG: ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 37:W305–W311. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Hu Y, Pan Z, Hu Y, Zhang L and Wang J: Network and pathway-based analyses of genes associated with Parkinson's disease. Mol Neurobiol. 54:4452–4465. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Scardoni G, Petterlini M and Laudanna C: Analyzing biological network parameters with CentiScaPe. Bioinformatics. 25:2857–2859. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Han JD, Berlin N, Hao T, Goldberg DS, Berriz GF, Zhang LV, Dupuy D, Walhout AJ, Cusick ME, Roth FP and Vidal M: Evidence for dynamically organized modularity in the yeast protein-protein interaction network. Nature. 430:88–93. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Kanwal A and Fazal S: Construction and analysis of protein-protein interaction network correlated with ankylosing spondylitis. Gene. 638:41–51. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Wu J, Vallenius T, Ovaska K, Westermarck J, Mäkelä TP and Hautaniemi S: Integrated network analysis platform for protein-protein interactions. Nat Methods. 6:75–77. 2009. View Article : Google Scholar : PubMed/NCBI

62 

Kerrien S, Alam-Faruque Y, Aranda B, Bancarz I, Bridge A, Derow C, Dimmer E, Feuermann M, Friedrichsen A, Huntley R, et al: IntAct-source resource for molecular interaction data. Nucleic Acids Res. 35:D561–D565. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Chatr-aryamontri A, Ceol A, Palazzi LM, Nardelli G, Schneider MV, Castagnoli L and Cesareni G: MINT: The Molecular INTeraction database. Nucleic Acids Res. 35:D572–D574. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Breitkreutz BJ, Stark C, Reguly T, Boucher L, Breitkreutz A, Livstone M, Oughtred R, Lackner DH, Bähler J, Wood V, et al: The BioGRID interaction database: 2008 update. Nucleic Acids Res. 36:D637–D640. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Salwinski L, Miller CS, Smith AJ, Pettit FK, Bowie JU and Eisenberg D: The database of interacting proteins: 2004 update. Nucleic Acids Res. 32:D449–D451. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Peri S, Navarro JD, Amanchy R, Kristiansen TZ, Jonnalagadda CK, Surendranath V, Niranjan V, Muthusamy B, Gandhi TK, Gronborg M, et al: Development of human protein reference database as an initial platform for approaching systems biology in humans. Genome Res. 13:2363–2371. 2003. View Article : Google Scholar : PubMed/NCBI

67 

Güldener U, Münsterkötter M, Oesterheld M, Pagel P, Ruepp A, Mewes HW and Stümpflen V: MPact: The MIPS protein interaction resource on yeast. Nucleic Acids Res. 34:D436–D441. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Vasaikar SV, Straub P, Wang J and Zhang B: LinkedOmics: Analyzing multi-omics data within and across 32 cancer types. Nucleic Acids Res. 46:D956–D963. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Cancer Genome Atlas Research Network, ; Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The cancer genome atlas pan-cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Gasca S, Pellestor F, Assou S, Loup V, Anahory T, Dechaud H, De Vos J and Hamamah S: Identifying new human oocyte marker genes: A microarray approach. Reprod Biomed Online. 14:175–183. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Schwab MS, Roberts BT, Gross SD, Tunquist BJ, Taieb FE, Lewellyn AL and Maller JL: Bub1 is activated by the protein kinase p90(Rsk) during Xenopus oocyte maturation. Curr Biol. 11:141–150. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Ouandaogo ZG, Frydman N, Hesters L, Assou S, Haouzi D, Dechaud H, Frydman R and Hamamah S: Differences in transcriptomic profiles of human cumulus cells isolated from oocytes at GV, MI and MII stages after in vivo and in vitro oocyte maturation. Hum Reprod. 27:2438–2447. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Li J, Qian WP and Sun QY: Cyclins regulating oocyte meiotic cell cycle progression†. Biol Reprod. 101:878–881. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Kaplan MH: STAT signaling in inflammation. JAKSTAT. 2:e241982013.PubMed/NCBI

75 

Qiu X, Guo H, Yang J, Ji Y, Wu CS and Chen X: Down-regulation of guanylate binding protein 1 causes mitochondrial dysfunction and cellular senescence in macrophages. Sci Rep. 8:16792018. View Article : Google Scholar : PubMed/NCBI

76 

Bros M, Haas K, Moll L and Grabbe S: RhoA as a key regulator of innate and adaptive immunity. Cells. 8:E7332019. View Article : Google Scholar : PubMed/NCBI

77 

Erdely A, Antonini JM, Salmen-Muniz R, Liston A, Hulderman T, Simeonova PP, Kashon ML, Li S, Gu JK, Stone S, et al: Type I interferon and pattern recognition receptor signaling following particulate matter inhalation. Part Fibre Toxicol. 9:252012. View Article : Google Scholar : PubMed/NCBI

78 

Luu K, Greenhill CJ, Majoros A, Decker T, Jenkins BJ and Mansell A: STAT1 plays a role in TLR signal transduction and inflammatory responses. Immunol Cell Biol. 92:761–769. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Smolen KK, Ruck CE, Fortuno ES III, Ho K, Dimitriu P, Mohn WW, Speert DP, Cooper PJ, Esser M, Goetghebuer T, et al: Pattern recognition receptor-mediated cytokine response in infants across 4 continents. J Allergy Clin Immunol. 133:818–826.e4. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Xiang C, Chen J and Fu P: HGF/Met signaling in cancer invasion: The impact on cytoskeleton remodeling. Cancers (Basel). 9:E442017. View Article : Google Scholar : PubMed/NCBI

81 

Lei T and Ling X: IGF-1 promotes the growth and metastasis of hepatocellular carcinoma via the inhibition of proteasome-mediated cathepsin B degradation. World J Gastroenterol. 21:10137–10149. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Cho S, Kitadai Y, Yoshida S, Tanaka S, Yoshihara M, Yoshida K and Chayama K: Deletion of the KIT gene is associated with liver metastasis and poor prognosis in patients with gastrointestinal stromal tumor in the stomach. Int J Oncol. 28:1361–1367. 2006.PubMed/NCBI

83 

Li B, Shen W, Peng H, Li Y, Chen F, Zheng L, Xu J and Jia L: Fibronectin 1 promotes melanoma proliferation and metastasis by inhibiting apoptosis and regulating EMT. Onco Targets Ther. 12:3207–3221. 2019. View Article : Google Scholar : PubMed/NCBI

84 

Liu Y, Ren CC, Yang L, Xu YM and Chen YN: Role of CXCL12-CXCR4 axis in ovarian cancer metastasis and CXCL12-CXCR4 blockade with AMD3100 suppresses tumor cell migration and invasion in vitro. J Cell Physiol. 234:3897–3909. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Chen Y, Jiang J, Zhao M, Luo X, Liang Z, Zhen Y, Fu Q, Deng X, Lin X, Li L, et al: microRNA-374a suppresses colon cancer progression by directly reducing CCND1 to inactivate the PI3K/AKT pathway. Oncotarget. 7:41306–41319. 2016.PubMed/NCBI

86 

Thomas SJ, Snowden JA, Zeidler MP and Danson SJ: The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br J Cancer. 113:365–371. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Stacker SA and Achen MG: The VEGF signaling pathway in cancer: The road ahead. Chin J Cancer. 32:297–302. 2013.PubMed/NCBI

88 

Shi X, Wang J, Lei Y, Cong C, Tan D and Zhou X: Research progress on the PI3K/AKT signaling pathway in gynecological cancer (Review). Mol Med Rep. 19:4529–4535. 2019.PubMed/NCBI

89 

Li X, Jiang S and Tapping RI: Toll-like receptor signaling in cell proliferation and survival. Cytokine. 49:1–9. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Gene Ontology Consortium: Gene ontology consortium: Going forward. Nucleic Acids Res. 43:D1049–D1056. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Kawai T and Akira S: Toll-like receptor and RIG-1-like receptor signaling. Ann N Y Acad Sci. 1143:1–20. 2008. View Article : Google Scholar : PubMed/NCBI

92 

Gkretsi V and Stylianopoulos T: Cell adhesion and matrix stiffness: Coordinating cancer cell invasion and metastasis. Front Oncol. 8:1452018. View Article : Google Scholar : PubMed/NCBI

93 

Zhang YL, Wang RC, Cheng K, Ring BZ and Su L: Roles of Rap1 signaling in tumor cell migration and invasion. Cancer Biol Med. 14:90–99. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Campbell PM and Der CJ: Oncogenic Ras and its role in tumor cell invasion and metastasis. Semin Cancer Biol. 14:105–114. 2004. View Article : Google Scholar : PubMed/NCBI

95 

Sun F, Wang J, Sun Q, Li F, Gao H, Xu L, Zhang J, Sun X, Tian Y, Zhao Q, et al: Interleukin-8 promotes integrin β3 upregulation and cell invasion through PI3K/Akt pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 38:4492019. View Article : Google Scholar : PubMed/NCBI

96 

Sanderson RD: Heparan sulfate proteoglycans in invasion and metastasis. Semin Cell Dev Biol. 12:89–98. 2001. View Article : Google Scholar : PubMed/NCBI

97 

Loo YM and Gale M Jr: Immune signaling by RIG-I-like receptors. Immunity. 34:680–692. 2011. View Article : Google Scholar : PubMed/NCBI

98 

Nyati KK and Prasad KN: Role of cytokines and Toll-like receptors in the immunopathogenesis of Guillain-Barré syndrome. Mediators Inflamm. 2014:7586392014. View Article : Google Scholar : PubMed/NCBI

99 

Saxena M and Yeretssian G: NOD-Like receptors: Master regulators of inflammation and cancer. Front Immunol. 5:3272014. View Article : Google Scholar : PubMed/NCBI

100 

Crosbie EJ, Einstein MH, Franceschi S and Kitchener HC: Human papillomavirus and cervical cancer. Lancet. 382:889–899. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Balasubramaniam SD, Balakrishnan V, Oon CE and Kaur G: Key molecular events in cervical cancer development. Medicina (Kaunas). 55:E3842019. View Article : Google Scholar : PubMed/NCBI

102 

Lin M, Ye M, Zhou J, Wang ZP and Zhu X: Recent advances on the molecular mechanism of cervical carcinogenesis based on systems biology technologies. Comput Struct Biotechnol J. 17:241–250. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Ye J, Yin L, Xie P, Wu J, Huang J, Zhou G, Xu H, Lu E and He X: Antiproliferative effects and molecular mechanisms of troglitazone in human cervical cancer in vitro. Onco Targets Ther. 8:1211–1218. 2015.PubMed/NCBI

104 

Yugawa T and Kiyono T: Molecular mechanisms of cervical carcinogenesis by high-risk human papillomaviruses: Novel functions of E6 and E7 oncoproteins. Rev Med Virol. 19:97–113. 2009. View Article : Google Scholar : PubMed/NCBI

105 

Jones MC, Askari JA, Humphries JD and Humphries MJ: Cell adhesion is regulated by CDK1 during the cell cycle. J Cell Biol. 217:3203–3218. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Hochegger H, Dejsuphong D, Sonoda E, Saberi A, Rajendra E, Kirk J, Hunt T and Takeda S: An essential role for Cdk1 in S phase control is revealed via chemical genetics in vertebrate cells. J Cell Biol. 178:257–268. 2007. View Article : Google Scholar : PubMed/NCBI

107 

Vassilev LT: Cell cycle synchronization at the G2/M phase border by reversible inhibition of CDK1. Cell Cycle. 5:2555–2556. 2006. View Article : Google Scholar : PubMed/NCBI

108 

Shaikh F, Sanehi P and Rawal R: Molecular screening of compounds to the predicted protein-protein interaction site of Rb1-E7 with p53-E6 in HPV. Bioinformation. 8:607–612. 2012. View Article : Google Scholar : PubMed/NCBI

109 

Yun J, Chae HD, Choy HE, Chung J, Yoo HS, Han MH and Shin DY: p53 negatively regulates cdc2 transcription via the CCAAT-binding NF-Y transcription factor. J Biol Chem. 274:29677–29682. 1999. View Article : Google Scholar : PubMed/NCBI

110 

Lindqvist A, van Zon W, Karlsson Rosenthal C and Wolthuis RM: Cyclin B1-Cdk1 activation continues after centrosome separation to control mitotic progression. PLoS Biol. 5:e1232007. View Article : Google Scholar : PubMed/NCBI

111 

Crasta K, Huang P, Morgan G, Winey M and Surana U: Cdk1 regulates centrosome separation by restraining proteolysis of microtubule-associated proteins. EMBO J. 25:2551–2563. 2006. View Article : Google Scholar : PubMed/NCBI

112 

Fang Y, Yu H, Liang X, Xu J and Cai X: Chk1-induced CCNB1 overexpression promotes cell proliferation and tumor growth in human colorectal cancer. Cancer Biol Ther. 15:1268–1279. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Krek W and Nigg EA: Differential phosphorylation of vertebrate p34cdc2 kinase at the G1/S and G2/M transitions of the cell cycle: Identification of major phosphorylation sites. EMBO J. 10:305–316. 1991. View Article : Google Scholar : PubMed/NCBI

114 

Morgan DO: Principles of CDK regulation. Nature. 374:131–134. 1995. View Article : Google Scholar : PubMed/NCBI

115 

Banerjee NS, Wang HK, Broker TR and Chow LT: Human papillomavirus (HPV) E7 induces prolonged G2 following S phase reentry in differentiated human keratinocytes. J Biol Chem. 286:15473–15482. 2011. View Article : Google Scholar : PubMed/NCBI

116 

Morgan EL, Wasson CW, Hanson L, Kealy D, Pentland I, McGuire V, Scarpini C, Coleman N, Arthur JSC, Parish JL, et al: STAT3 activation by E6 is essential for the differentiation-dependent HPV18 life cycle. PLoS Pathog. 14:e10069752018. View Article : Google Scholar : PubMed/NCBI

117 

Stewart DA, Cooper CR and Sikes RA: Changes in extracellular matrix (ECM) and ECM-associated proteins in the metastatic progression of prostate cancer. Reprod Biol Endocrinol. 2:22004. View Article : Google Scholar : PubMed/NCBI

118 

Chiu CF, Ho MY, Peng JM, Hung SW, Lee WH, Liang CM and Liang SM: Raf activation by Ras and promotion of cellular metastasis require phosphorylation of prohibitin in the raft domain of the plasma membrane. Oncogene. 32:777–787. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Matter ML and Ruoslahti E: A signaling pathway from the alpha5beta1 and alpha(v)beta3 integrins that elevates bcl-2 transcription. J Biol Chem. 276:27757–27763. 2001. View Article : Google Scholar : PubMed/NCBI

120 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002. View Article : Google Scholar : PubMed/NCBI

121 

Sand JM, Larsen L, Hogaboam C, Martinez F, Han M, Røssel Larsen M, Nawrocki A, Zheng Q, Karsdal MA and Leeming DJ: MMP mediated degradation of type IV collagen alpha 1 and alpha 3 chains reflects basement membrane remodeling in experimental and clinical fibrosis-validation of two novel biomarker assays. PLoS One. 8:e849342013. View Article : Google Scholar : PubMed/NCBI

122 

Bokhari AA, Baker TM, Dorjbal B, Waheed S, Zahn CM, Hamilton CA, Maxwell GL and Syed V: Nestin suppression attenuates invasive potential of endometrial cancer cells by downregulating TGF-β signaling pathway. Oncotarget. 7:69733–69748. 2016. View Article : Google Scholar : PubMed/NCBI

123 

da Silva Cardeal LB, Brohem CA, Corrêa TC, Winnischofer SM, Nakano F, Boccardo E, Villa LL, Sogayar MC and Maria-Engler SS: Higher expression and activity of metalloproteinases in human cervical carcinoma cell lines is associated with HPV presence. Biochem Cell Biol. 84:713–719. 2006. View Article : Google Scholar : PubMed/NCBI

124 

Cardeal LB, Boccardo E, Termini L, Rabachini T, Andreoli MA, di Loreto C, Longatto Filho A, Villa LL and Maria-Engler SS: HPV16 oncoproteins induce MMPs/RECK-TIMP-2 imbalance in primary keratinocytes: Possible implications in cervical carcinogenesis. PLoS One. 7:e335852012. View Article : Google Scholar : PubMed/NCBI

125 

Shiau MY, Fan LC, Yang SC, Tsao CH, Lee H, Cheng YW, Lai LC and Chang YH: Human papillomavirus up-regulates MMP-2 and MMP-9 expression and activity by inducing interleukin-8 in lung adenocarcinomas. PLoS One. 8:e544232013. View Article : Google Scholar : PubMed/NCBI

126 

Ding Y, Pan Y, Liu S, Jiang F and Jiao J: Elevation of MiR-9-3p suppresses the epithelial-mesenchymal transition of nasopharyngeal carcinoma cells via down-regulating FN1, ITGB1 and ITGAV. Cancer Biol Ther. 18:414–424. 2017. View Article : Google Scholar : PubMed/NCBI

127 

Wang J, Deng L, Huang J, Cai R, Zhu X, Liu F, Wang Q, Zhang J and Zheng Y: High expression of Fibronectin 1 suppresses apoptosis through the NF-κB pathway and is associated with migration in nasopharyngeal carcinoma. Am J Transl Res. 9:4502–4511. 2017.PubMed/NCBI

128 

Rajkumar T, Sabitha K, Vijayalakshmi N, Shirley S, Bose MV, Gopal G and Selvaluxmy G: Identification and validation of genes involved in cervical tumourigenesis. BMC Cancer. 11:802011. View Article : Google Scholar : PubMed/NCBI

129 

Koromilas AE and Sexl V: The tumor suppressor function of STAT1 in breast cancer. JAKSTAT. 2:e233532013.PubMed/NCBI

130 

Zhang Y and Liu Z: STAT1 in cancer: Friend or foe? Discov Med. 24:19–29. 2017.PubMed/NCBI

131 

Akram M, Kim KA, Kim ES, Shin YJ, Noh D, Kim E, Kim JH, Majid A, Chang SY, Kim JK and Bae ON: Selective inhibition of JAK2/STAT1 signaling and iNOS expression mediates the anti-inflammatory effects of coniferyl aldehyde. Chem Biol Interact. 256:102–110. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2020
Volume 19 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yi Y, Fang Y, Wu K, Liu Y and Zhang W: Comprehensive gene and pathway analysis of cervical cancer progression. Oncol Lett 19: 3316-3332, 2020
APA
Yi, Y., Fang, Y., Wu, K., Liu, Y., & Zhang, W. (2020). Comprehensive gene and pathway analysis of cervical cancer progression. Oncology Letters, 19, 3316-3332. https://doi.org/10.3892/ol.2020.11439
MLA
Yi, Y., Fang, Y., Wu, K., Liu, Y., Zhang, W."Comprehensive gene and pathway analysis of cervical cancer progression". Oncology Letters 19.4 (2020): 3316-3332.
Chicago
Yi, Y., Fang, Y., Wu, K., Liu, Y., Zhang, W."Comprehensive gene and pathway analysis of cervical cancer progression". Oncology Letters 19, no. 4 (2020): 3316-3332. https://doi.org/10.3892/ol.2020.11439