Open Access

Bioinformatics analysis of gene expression alterations in microRNA‑122 knockout mice with hepatocellular carcinoma

  • Authors:
    • Bosheng He
    • Ying He
    • Weixiang Shi
    • Shenchu Gong
    • Xiaohong Chen
    • Jing Xiao
    • Jinhua Gu
    • Wenbin Ding
    • Yilang Wang
  • View Affiliations

  • Published online on: April 7, 2017     https://doi.org/10.3892/mmr.2017.6445
  • Pages: 3681-3689
  • Copyright: © He 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

Reduced microRNA (miR)‑122 expression levels are frequently observed in hepatocellular carcinoma (HCC). The present study was conducted to investigate potential targets of miR‑122 and determine the underlying regulatory mechanisms of miR‑122 in HCC development. The public dataset GSE31731 was utilized, consisting of 8 miR‑122 knockout (KO) mice (miR‑122 KO) and 8 age‑matched wild‑type mice (WT group). Following data preprocessing, the differentially expressed genes (DEGs) were selected, followed by enrichment analysis. A protein‑protein interaction (PPI) network was established, and a module network was further extracted. Combining the DEGs with microRNA targeting databases permitted the screening of the overlapping targets of miR‑122. Furthermore, previously reported genes were screened out by literature mining. Transcription factors (TFs) of the targets were subsequently investigated. DEGs between miR‑122 KO and WT groups were selected, including 713 upregulated and 395 downregulated genes. Of these, upregulated genes were enriched in cell cycle‑associated processes [including nucleolar and spindle associated protein 1 (NUSAP1)], the cytokine‑cytokine receptor interaction pathway [including C‑X‑C motif chemokine receptor 4 (CXCR4) and C‑C motif chemokine receptor 2 (CCR2)], and the extracellular matrix‑receptor interaction pathway [including integrin subunit alpha V (ITGAV)]. In addition, multiple overlapping targets were highlighted in the PPI network, including NUSAP1, CXCR4, CCR2 and ITGAV. Notably, CXCR4 and CCR2 were linked in module C, enriched in the cytokine‑cytokine receptor interaction pathway. Furthermore, upregulated sex determining region Y‑box 4 (SOX4) was identified as a TF. The results of the present study may provide a theoretical basis for further studies on the mechanisms of miR‑122 in the development of HCC.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the most frequent liver cancer globally (1). The majority of HCC cases occur in cirrhotic liver, and the primary risk factors are chronic hepatitis B virus or chronic hepatitis C virus (HCV) infection, which account for almost all HCC cases (2). The incidence of HCC varies between different geographical areas; however, it is increasing globally, particularly in Asia, with 6–11 per 100,000 people with the disease (3,4). A study of HCC epidemiology in Germany indicated that, despite the availability of various advanced chemotherapies and radiotherapies, including chemoembolization with drug-eluting beads, sorafenib and selective internal radiotherapy, the overall survival rate has not improved (5). Therefore, the development of more effective therapeutic methods, including molecular targeting therapy, is necessary.

Multiple studies have been conducted to investigate the molecular mechanisms underlying HCC pathogenesis and numerous gene markers have been identified in HCC, including alpha-fetoprotein, glypican-3 (a serum and histochemical marker) and transforming growth factor-β (68). As small, non-coding RNAs, microRNAs (miRNAs) are important regulators of cellular function and physiology (9). Controlling miRNA expression is essential for the maintenance of the steady state of cellular machinery (10). Various microRNAs (miRNAs) have been proposed as novel biomarkers of HCC prognosis, including the chromosome 19 miRNA cluster, which is overexpressed in HCC (11). HCV-induced alteration of miRNA expression regulates inflammation, leading to liver fibrosis. In addition, miRNA (miR)-449a has been reported to serve as an inhibitor in HCV patients, acting via the downregulation of chitinase-3-like protein 1 expression, which is an inflammatory marker for chronic liver diseases with fibrosis (12). Dysregulation of other miRNAs has been detected in HCC, including upregulated miR-23a, −146a and −181a, and downregulated miR-17, −338-3p and −378 (13). Notably, miR-122 is involved in HCC pathogenesis. It is commonly downregulated in HCC and the loss of miR-122 contributes to hepatocarcinogenesis in mice (14). miR-122 has additionally been reported to induce apoptosis in human HCC cell lines via targeting the anti-apoptosis gene B-cell lymphoma-2-like 2 (15). Furthermore, miR-122 inhibits cell proliferation in HCC by targeting the Wnt/β-catenin signaling pathway (16). To further understand the modulation of miR-122 in HCC development, previous studies have investigated the consequences of miR-122 deletion. Hsu et al (17) revealed that deletion of mouse mir-122 resulted in hepatocarcinogenesis and HCC-like tumor development. Although increased expression of multiple targets of miR-122 has been detected in miR-122 knockout (KO) mice, including aldolase, fructose bisphosphate A (ALDOA), solute carrier family 7 member 1, citrate synthase and cyclin G1, the functions and pathways of the targets, and the potential associations between them at the protein level, remain to be elucidated.

In the present study, the expression profile dataset generated by Hsu et al (17), GSE31731, was reanalyzed and differentially expressed genes (DEGs) were identified between miR-122 KO mice and age-matched wild-type (WT) mice. Enrichment analysis of the identified DEGs was subsequently conducted, followed by protein-protein interaction (PPI) and module analysis. Furthermore, targets of miR-122 of these DEGs were selected by combining the results with relevant databases and literature mining, followed by transcription factor (TF) analysis. By means of these comprehensive bioinformatics analyses, the present study aimed to further elucidate the involvement of miR-122 in HCC, and identify potential regulators among its targets.

Materials and methods

Data resources

Gene expression of GSE31731 of HCCs, which was deposited by Hsu et al (17) in the public Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo) database, was utilized in the present study. The miR-122 KO mice (liver tumor samples) and age-matched WT mice (healthy liver samples) were contained in this dataset. There were 8 biological replicates. Two-channel microarray experiments were conducted for generation of the dataset, based on the platform GPL13912 (accession no. Agilent-028005 SurePrint G3 Mouse GE 8×60 K Microarray; Agilent Technologies, Inc., Santa Clara, CA, USA).

Data pretreatment and differential expression analysis

Raw data were preprocessed using R package in Bioconductor (version 3.4; www.bioconductor.org/packages/3.0/bioc/). Following background correction and normalization, the expression value was converted from the probe level to the gene level. Subsequently, DEGs between liver tumor samples and healthy liver samples were screened out, based on the Student's t-test in Linear Models for Microarray Analysis package (version 3.30.3; www.bioconductor.org/packages/release/bioc/html/limma.html) (18). The Benjamini-Hochberg method (19) was used to adjust the P-value. The selection criteria for significant DEGs were a false discovery rate <0.01 and a log2 fold change >1.5.

Enrichment analysis for DEGs

The Database for Annotation, Visualization and Integrated Discovery (DAVID; david.ncifcrf.gov/home.jsp) is a common tool for gene function and pathway annotation (2022). To examine biological functions and pathways of the identified DEGs, Gene Ontology (GO; www.geneontology.org/) (23) and Kyoto Encyclopedia of Genes and Genomes (KEGG; www.genome.jp/kegg/pathway.html) (24) pathway enrichment analyses were performed using DAVID (version 6.8). Cut-off values for significant GO and KEGG pathway terms were P<0.05 and enriched gene number ≥2.

PPI network construction

To predict potential interactions between DEGs at the protein level, the DEGs were entered into the Search Tool for the Retrieval of Interacting Genes (string-db.org/) database (25). The parameter of interplayed PPIs was set as 0.7, and a prerequisite for the network construction was that all the PPI nodes were DEGs. Finally, the PPI network was visualized by Cytoscape (version 3.4.0; cytoscape.org/) software (26).

Furthermore, module analysis was performed for the PPI network using ClusterONE (version 1.0; www.paccanarolab.org/clusterone/) (27), followed by KEGG pathway enrichment analysis. The threshold for significant module selection was P<1.0×10−6.

Analysis of miR-122 targets

Initially, targets of miR-122 in mouse were downloaded from three databases: miRecords (c1.accurascience.com/miRecords/) (28), TargetScan (www.targetscan.org/) (29) and microrna.org (www.microrna.org) (30), and only genes that appeared in at least two databases were deemed to be targets of miR-122. These predicted targets were compared with the DEGs, and the overlapping genes were screened out. Following this, TFs of the overlapped targets were predicted by the iRegulon plugin of Cytoscape (iregulon.aertslab.org), which integrates a set of TF databases including Transfac, Jaspar, Encode, Swissregulon and Homer to detect enriched TF motifs and their optimal sets of direct targets (31). Normalized Enrichment Score (NES) was the measurement index for TFs of the targets and the threshold used was NES >3. Furthermore, the Agilent Literature Search plugin (Agilent Technologies, Inc.) (32), which is complementary for protein interaction data, was used to analyze the literature mining association network. In the present study, the search terms were set as ‘targets of miR-122’, context ‘liver cancer’ and species ‘Mus’, to select the reported HCC-associated literature involving miR-122.

Results

DEGs between miR-122 KO and WT groups

Using predefined criteria, DEGs between miR-122 KO and WT groups were screened out, including 713 upregulated and 395 downregulated genes.

Altered functions and pathways of DEGs

Based on GO and KEGG enrichment analyses, upregulated DEGs were identified to be significantly enriched in cell cycle associated biological processes (BPs), including the cell cycle, M phase and mitotic cell cycle [for example nucleolar and spindle associated protein 1 (NUSAP1); Table I], the cytokine-cytokine receptor interaction pathway [for example C-X-C motif chemokine receptor 4 (CXCR4) and C-C motif chemokine receptor 2 (CCR2); Table II], and various cancer-associated pathways, including small cell lung cancer and pathways in cancer [for example integrin subunit alpha V (ITGAV); Table II]. The downregulated DEGs were associated with oxidation-reduction (Table I) and metabolism-associated pathways, including drug metabolism, linoleic acid metabolism and retinol metabolism (Table II).

Table I.

Functions altered by differentially expressed genes.

Table I.

Functions altered by differentially expressed genes.

CategoryTermCountP-value
Upregulated
  BPGO:0007049~cell cycle57 4.96×10−11
GO:0000279~M phase34 1.64×10−09
GO:0000278~mitotic cell cycle31 2.64×10−09
GO:0007067~mitosis27 3.21×10−09
GO:0000280~nuclear division27 3.21×10−09
  CC GO:0005576~extracellular region133 9.92×10−18
GO:0044421~extracellular region part74 2.00×10−13
GO:0005578~proteinaceous extracellular matrix35 7.33×10−09
GO:0031012~extracellular matrix35 2.01×10−08
GO:0044420~extracellular matrix part18 4.96×10−08
  MFGO:0005509~calcium ion binding60 1.62×10−07
GO:0008009~chemokine activity10 4.56×10−06
GO:0008201~heparin binding14 5.26×10−06
GO:0042379~chemokine receptor binding10 5.74×10−06
GO:0001871~pattern binding17 9.14×10−06
Downregulated
  BP GO:0055114~oxidation-reduction64 9.82×10−28
GO:0006631~fatty acid metabolic process24 4.36×10−13
GO:0008202~steroid metabolic process21 1.75×10−11
GO:0006694~steroid biosynthetic process15 3.73×10−11
GO:0006956~complement activation10 1.37×10−08
  CC GO:0005777~peroxisome22 4.28×10−15
GO:0042579~microbody22 4.28×10−15
GO:0005792~microsome26 5.15×10−14
GO:0042598~vesicular fraction26 1.13×10−13
GO:0005739~mitochondrion66 7.86×10−11
  MFGO:0009055~electron carrier activity34 4.36×10−21
GO:0020037~heme binding23 9.42×10−14
GO:0046906~tetrapyrrole binding23 2.57×10−13
GO:0005506~iron ion binding33 3.02×10−13

[i] The top 5 functions are presented for each category, ranked by the enrichment significance. Up, upregulated differentially expressed genes; down, downregulated differentially expressed genes; BP, biological process; CC, cellular component; MF, molecular function; GO, gene ontology; count, gene numbers enriched in a specific gene ontology term.

Table II.

Pathways altered by differentially expressed genes.

Table II.

Pathways altered by differentially expressed genes.

TermCountGenesP-value
Upregulated
  mmu04060:Cytokine-cytokine receptor interaction27CCL2, CXCL5, CXCR4, CXCL14, CCR2 4.82×10−06
  mmu04512:ECM-receptor interaction15COL3A1, LAMA2, ITGAV, COL1A2, LAMC1 4.87×10−06
  mmu04510:Focal adhesion22COL3A1, LAMA2, ITGAV, LAMC1 4.43×10−05
  mmu00480:Glutathione metabolism10GPX2, GSTA1, GSTA2, GSTM3, G6PDX 2.05×10−04
  mmu04110:Cell cycle14CCNB2, KMYT1, BUB1B, ESPL1, CDC200.001940583
  mmu05222:Small cell lung cancer11LAMA2, COL4A1, ITGAV, LAMC2, LAMC10.002161153
  mmu04810:Regulation of actin cytoskeleton19ITGAX, ITGAV, PDGFRB, PAK1, DIAP30.002891419
  mmu04062:Chemokine signaling pathway16CCL2, CXCR4, CXCL16, CCR2, CX3CR10.006803694
  mmu05200:Pathways in cancer23COL4A1, LAMA2, ITGAV, LAMC2, LAMC10.011257034
  mmu00590:Arachidonic acid metabolism9GPX2, CBR1, CYP4F18, GPX3, GGT1 et al0.018763259
Downregulated
  mmu00982:Drug metabolism18CYP2C37, CYP3A16, CYP2C54, CYP2C44, ADH4 1.83×10−12
  mmu00980:Metabolism of xenobiotics by cytochrome P45017CYP2C37, CYP3A16, CYP2C54, CYP2C44, CYP2C68 2.81×10−12
  mmu00591:Linoleic acid metabolism14CYP2J5, CYP2C37, CYP3A16, CYP2C54, CYP2C44 4.16×10−11
  mmu00830:Retinol metabolism16CYP2C37, CYP3A16, CYP2C54, CYP2C44, CYP2C68 6.09×10−11
  mmu03320:PPAR signaling pathway16ACOX1, ACSL1, CYP4A12A, HMGCS2, SCP2 5.86×10−10
  mmu00590:Arachidonic acid metabolism15CYP2J5, CYP2C37, CYP2C54, CYP2C44, CYP2J8 1.14×10−08
  mmu00120:Primary bile acid biosynthesis8CYP7B1, HSD3B7, CYP7A1, CYP8B1, SCP2 2.81×10−08
  mmu00071:Fatty acid metabolism9CYP4A12B, GCDH, ACOX1, ACSL1, ADH4 1.21×10−05
  mmu00140:Steroid hormone biosynthesis9CYP7B1, CYP3A16, HSD3B6, HSD17B2, CYP7A1 1.21×10−05
  mmu04610:Complement and coagulation cascades11MBL1, C8A, MBL2, C8B, CD55 1.40×10−05

[i] The top 10 pathways are presented, ranked by the enrichment significance. Up, upregulated differentially expressed genes; down, downregulated differentially expressed genes; count, gene numbers enriched in a specific pathway term.

PPI network and functional module analysis

A PPI network was established, involving 549 nodes and 2,243 interplayed protein interactions (Fig. 1). Three sub-networks [modules A (Fig. 2A), B (Fig. 2B) and C (Fig. 2C)] were extracted from the PPI network. Enrichment analysis revealed that the majority of the genes in module A were upregulated and enriched in DNA replication and cell cycle-associated pathways, whereas the majority of genes in module B were downregulated and enriched in metabolism of xenobiotics by cytochrome P450 pathway (Fig. 3). In module C, the majority of the genes were upregulated and involved with the chemokine signaling pathway and the cytokine-cytokine receptor interaction pathway (Fig. 3).

Targets of miR-122

Integrating the information from miRNA databases with identified DEGs, a total of 76 overlapping genes were selected as the targets of miR-122. Enrichment analysis indicated that these genes were significantly involved in pathways in cancer (for example ITGAV; Table II), regulation of actin cytoskeleton (for example ITGAV; Table II) and cytokine-cytokine receptor interaction (for example CXCR4 and CCR2; Table II).

Notably, 39 genes of the 76 overlapping targets were additionally the predominant nodes with high degree in the PPI network, including upregulated NUSAP1 (degree=30), CXCR4 (degree=21), CCR2 (degree=20), ITGAV (degree=17) and ALDOA (degree=14); and the downregulated acyl-CoA synthetase short-chain family member 2 (degree=10). NUSAP1 was also highlighted in module A, whereas CXCR4 and CCR2 were prominent in module C.

In total, 12 TFs targeting 62 overlapping genes were predicted, including sex determining region Y-box 4 (SOX4), heterogeneous nuclear ribonucleoprotein H3, NK2 homeobox 1, inhibitor of growth family member 4, early B-cell factor 1, sex determining region Y-box 15, nuclear receptor subfamily 3 group C member 1, zinc finger protein 263, IKAROS family zinc finger 2, paired like homeodomain 3, eukaryotic translation initiation factor 5A2 and chromobox 7. The TF-target regulatory network is presented in Fig. 4. Notably, of the TFs, SOX4 was additionally an upregulated DEG.

According to literature mining, a total of 47 genes of the 76 overlapping targets were reported to be associated with HCC, including ITGAV and CXCR4. Furthermore, significantly altered expression of these genes was detected following miR-122 KO, which suggested the involvement of miR-122 in HCC development.

Discussion

miR-122 deficiency results in chronic steatohepatitis and spontaneous HCC (14). By re-analyzing the dataset GSE31731, numerous DEGs were identified between miR-122 KO and WT groups. Of these, upregulated genes were significantly enriched in cell cycle-associated processes (for example NUSAP1), cytokine-cytokine receptor interaction pathways (for example CXCR4 and CCR2), and extracellular matrix (ECM) -receptor interactions (for example ITGAV). Various overlapping targets were highlighted in the PPI network, including NUSAP1, CXCR4, CCR2 and ITGAV. Notably, NUSAP1 was predominant in module A, which was associated with the cell cycle-associated pathway, whereas CXCR4 and CCR2 were linked in module C, enriched in the cytokine-cytokine receptor interaction pathway. Furthermore, upregulated SOX4 was identified as a TF.

Restoration of miR-122 suppressed HCC tumor cell growth, and the antitumor activity was closely associated with cell cycle arrest (33). The protein encoded by NUSAP1 is a nucleolar-spindle-associated protein that acts as a positive regulator of mitosis (34). It has been identified as a cell cycle progression gene in numerous cancer types, including prostate and lung cancer (35,36). In aggressive HCC, expression of NUSAP1 is affected by other cell cycle-associated genes, including L2DTL (37). In the present study, upregulated NUSAP1 in the miR-122 KO group was significantly enriched in cell cycle-associated BPs, and additionally served as a node in module A of the PPI network, as well as one of the overlapped targets of miR-122. However, NUSAP1 has not been previously reported to be directly involved in HCC based on the literature mining results, suggesting that this gene may be a novel target of miR-122 involved in cell cycle-associated processes in HCC progression.

Dysregulation of the cytokine-cytokine receptor interaction pathway has been detected in HCC development (38,39). Activation of various chemokines in this pathway is involved in the development of numerous cancers, including HCC (40). As a chemokine receptor, the function of CXCR4 has been extensively investigated. Increased expression of CXCR4 has previously been reported to have a close association with the progression of HCC (41). In addition, CXCR4 inhibition results in antitumor effects, including the inhibition of tumor growth and the improvement of survival in mice with HCC (42). Elevated expression of CXCR4 in the miR-122 deficiency group, combined with the target information in miRNA databases, indicated that CXCR4 may be a potential target of miR-122 in HCC. CCR2 is the chemokine receptor of C-C motif chemokine ligand 2 (CCL2). Increased expression of CCR2 has previously been observed in liver leukocytes from patients with HCC (43). CCL2 is also involved in the cytokine-cytokine receptor interaction pathway and its expression is associated with HCC progression (38). Notably, CCL2 has been identified as a target of miR-122, and restoration of miR-122 results in the suppression of CCL2 in HCC (44). The results of the present study indicated that the DEG CCR2 was upregulated in the miR-122 KO group and significantly enriched in the cytokine-cytokine receptor interaction pathway. Notably, it was additionally identified as an overlapping target based on the miRNA targeting database, and was linked to CXCR4 in module C of the PPI network. These data collectively suggested that CCR2 may be a target of miR-122, and co-regulate the cytokine-cytokine receptor interaction pathway with CXCR4 in HCC development.

ECM-receptor interaction is a common pathway that is disturbed by altered gene expressions in various cancers (45,46). The gene ITGAV encodes a protein that belongs to the integrin superfamily. It has been reported to be involved in the ECM-receptor interaction pathway. It is a part of the ECM system (47), suggesting its involvement in the ECM-receptor interaction pathway. In other cancer types, including gastric cancer, ITGAV is enriched in this pathway (48). Notably, overexpressed ITGAV is induced by the gene forkhead box Q1 (FOXQ1), a member of the forkhead TF family that influences HCC metastasis (49), suggesting the potential involvement of this gene in HCC. Upregulated ITGAV in the miR-122 KO group, combined with enrichment analysis and the overlapped prediction target, indicated that this gene may be a target of miR-122 that modulates the ECM-receptor interaction pathway in HCC progression.

The intron-lacking gene SOX4 contributes to hepatocarcinogenesis and its overexpression may be a useful prognostic marker for survival after surgical resection (50). It has been demonstrated in vitro that overexpressed SOX4 is involved in p53-mediated apoptosis in HCC (50). In addition, SOX4 overexpression has previously been reported to control the metastasis of HCC (51). Thus, SOX4 has been identified as a marker gene for HCC (52). Furthermore, SOX4 serves as a TF that regulates cell differentiation. In HCC, various targets have been experimentally validated using chromatin immunoprecipitation and small interfering RNA assays, including aldo-keto reductase family 1 member B10, coiled-coil domain containing 97, dickkopf Wnt signaling pathway inhibitor 1, FOXQ1 and microtubule associated protein 4 (51). miR-191 inhibition has previously been reported to result in the upregulation of SOX4, and increased SOX4 expression promotes cell apoptosis and suppresses tumorigenesis of HCC (53). The present study indicated that the TF SOX4 may additionally be a target of miR-122.

Despite these comprehensive bioinformatics analyses, the present study has limitations. The data was downloaded from the GEO database, and the sample size was relatively small. In addition, experimental validation of the associations between miR-122 and the predicted targets was lacking, and will be addressed in follow-up studies. Furthermore, the target expression levels following miR-122 restoration were not investigated, although this would have further confirmed the targeting associations. However, the present study has significant value as it provides novel insights into the consequences of miR-122 KO in HCC progression.

In conclusion, various crucial targets of miR-122 in HCC progression were identified, including NUSAP1, CXCR4, CCR2 and ITGAV. Cell cycle-associated processes, the cytokine-cytokine receptor interaction pathway, which may be co-regulated by CXCR4 and CCR2, and the ECM-receptor interaction pathway were altered by these targets. In addition, the target SOX4 may be a TF. The results of the present study may provide a theoretical basis for further studies on the mechanisms of miR-122 in the development of HCC.

Acknowledgements

The present study was supported by the Social Development Fund of Nantong (grant no. HS2014063), the 12th Talent Summit of Top Six Industries in Jiangsu Province (grant no. WSW080) and the Project of Enhancing Medicine with Science and Education under the 13th Five-Year Plan of Nantong [grant no. Nantong MED Sci & Edu (2016) 23].

References

1 

Yang J, Cai X, Lu W, Hu C, Xu X, Yu Q and Cao P: Evodiamine inhibits STAT3 signaling by inducing phosphatase shatterproof 1 in hepatocellular carcinoma cells. Cancer Lett. 328:243–251. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Borel F, Konstantinova P and Jansen PL: Diagnostic and therapeutic potential of miRNA signatures in patients with hepatocellular carcinoma. J Hepatol. 56:1371–1383. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Al-Mahtab M, Uddin H and Fazle Akbar SM: Epidemiology and Risk Factors of Hepatocellular Carcinoma in Asia. J Gastroenterology and Hepatology Res. 3:2014.

4 

Paranaguá-Vezozzo DC, Ono SK, Alvarado-Mora MV, Farias AQ, Cunha-Silva M, França JI, Alves VA, Sherman M and Carrilho FJ: Epidemiology of HCC in Brazil: Incidence and risk factors in a ten-year cohort. Ann Hepatol. 13:386–393. 2014.PubMed/NCBI

5 

Weinmann A, Koch S, Niederle IM, Schulze-Bergkamen H, König J, Hoppe-Lotichius M, Hansen T, Pitton MB, Düber C, Otto G, et al: Trends in epidemiology, treatment, and survival of hepatocellular carcinoma patients between 1998 and 2009: An analysis of 1066 cases of a German HCC registry. J Clin Gastroenterol. 48:279–289. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Soresi M, Magliarisi C, Campagna P, Leto G, Bonfissuto G, Riili A, Carroccio A, Sesti R, Tripi S and Montalto G: Usefulness of alpha-fetoprotein in the diagnosis of hepatocellular carcinoma. Anticancer Res. 23:1747–1753. 2003.PubMed/NCBI

7 

Capurro M, Wanless IR, Sherman M, Deboer G, Shi W, Miyoshi E and Filmus J: Glypican-3: A novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology. 125:89–97. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Murata M, Matsuzaki K, Yoshida K, Sekimoto G, Tahashi Y, Mori S, Uemura Y, Sakaida N, Fujisawa J, Seki T, et al: Hepatitis B virus X protein shifts human hepatic transforming growth factor (TGF)-beta signaling from tumor suppression to oncogenesis in early chronic hepatitis B. Hepatology. 49:1203–1217. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Beta M, Venkatesan N, Vasudevan M, Vetrivel U, Khetan V and Krishnakumar S: Identification and insilico analysis of retinoblastoma serum microRNA profile and gene targets towards prediction of novel serum biomarkers. Bioinform Biol Insights. 7:21–34. 2013.PubMed/NCBI

10 

Kosaka N, Iguchi H and Ochiya T: Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 101:2087–2092. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Augello C, Vaira V, Caruso L, Destro A, Maggioni M, Park YN, Montorsi M, Santambrogio R, Roncalli M and Bosari S: MicroRNA profiling of hepatocarcinogenesis identifies C19MC cluster as a novel prognostic biomarker in hepatocellular carcinoma. Liver Int. 32:772–782. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Sarma NJ, Tiriveedhi V, Subramanian V, Shenoy S, Crippin JS, Chapman WC and Mohanakumar T: Hepatitis C virus mediated changes in miRNA-449a modulates inflammatory biomarker YKL40 through components of the NOTCH signaling pathway. PLoS One. 7:e508262012. View Article : Google Scholar : PubMed/NCBI

13 

Zhang ZZ, Liu X, Wang DQ, Teng MK, Niu LW, Huang AL and Liang Z: Hepatitis B virus and hepatocellular carcinoma at the miRNA level. World J Gastroenterol. 17:3353–3358. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Hsu SH, Wang B, Kutay H, Bid H, Shreve J, Zhang X, Costinean S, Bratasz A, Houghton P and Ghoshal K: Hepatic loss of miR-122 predisposes mice to hepatobiliary cyst and hepatocellular carcinoma upon diethylnitrosamine exposure. Am J Pathol. 183:1719–1730. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Lin CJ, Gong HY, Tseng HC, Wang WL and Wu JL: miR-122 targets an anti-apoptotic gene, Bcl-w, in human hepatocellular carcinoma cell lines. Biochem Biophys Res Commun. 375:315–320. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Xu J, Zhu X, Wu L, Yang R, Yang Z, Wang Q and Wu F: MicroRNA-122 suppresses cell proliferation and induces cell apoptosis in hepatocellular carcinoma by directly targeting Wnt/β-catenin pathway. Liver Int. 32:752–760. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, Yu L, Bai S, La Perle K, Chivukula RR, et al: Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest. 122:2871–2883. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Smyth GK: Limma: Linear models for microarray dataBioinformatics and computational biology solutions using R and Bioconductor. Springer; pp. 397–420. 2005, View Article : Google Scholar

19 

Haynes W: Benjamini-Hochberg MethodEncyclopedia of Systems Biology. Springer; pp. 78. 2013, View Article : Google Scholar

20 

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

21 

da W Huang, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009.PubMed/NCBI

22 

da W Huang, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Gene Ontology Consortium, ; Blake JA, Dolan M, Drabkin H, Hill DP, Li N, Sitnikov D, Bridges S, Burgess S, Buza T, McCarthy F, et al: Gene Ontology annotations and resources. Nucleic Acids Res. 41:D530–D535. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Kanehisa M, Goto S, Sato Y, Furumichi M and Tanabe M: KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40:D109–D114. 2012. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: New features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Wan FC, Cui YP, Wu JT, Wang JM-Z, Liu Q and Gao ZL: The PPI network and cluster ONE analysis to explain the mechanism of bladder cancer. Eur Rev Med Pharmacol Sci. 17:618–623. 2013.PubMed/NCBI

28 

Xiao F, Zuo Z, Cai G, Kang S, Gao X and Li T: miRecords: An integrated resource for microRNA-target interactions. Nucleic Acids Res. 37:D105–D110. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP and Burge CB: Prediction of mammalian microRNA targets. Cell. 115:787–798. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Betel D, Wilson M, Gabow A, Marks DS and Sander C: The microRNA. org resource: Targets and expression. Nucleic Acids Res. 36:D149–D153. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Janky R, Verfaillie A, Imrichová H, Van de Sande B, Standaert L, Christiaens V, Hulselmans G, Herten K, Sanchez M Naval, Potier D, et al: iRegulon: from a gene list to a gene regulatory network using large motif and track collections. PLoS Comput Biol. 10:e10037312014. View Article : Google Scholar : PubMed/NCBI

32 

Cline MS, Smoot M, Cerami E, Kuchinsky A, Landys N, Workman C, Christmas R, Avila-Campilo I, Creech M, Gross B, et al: Integration of biological networks and gene expression data using Cytoscape. Nat Protoc. 2:2366–2382. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Ma L, Liu J, Shen J, Liu L, Wu J, Li W, Luo J, Chen Q and Qian C: Expression of miR-122 mediated by adenoviral vector induces apoptosis and cell cycle arrest of cancer cells. Cancer Biol Ther. 9:554–561. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Raemaekers T, Ribbeck K, Beaudouin J, Annaert W, Van Camp M, Stockmans I, Smets N, Bouillon R, Ellenberg J and Carmeliet G: NuSAP, a novel microtubule-associated protein involved in mitotic spindle organization. J Cell Biol. 162:1017–1029. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Cuzick J, Swanson GP, Fisher G, Brothman AR, Berney DM, Reid JE, Mesher D, Speights VO, Stankiewicz E, Foster CS, et al: Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: A retrospective study. Lancet Oncol. 12:245–255. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Zhou C, Chen H, Han L, Wang A and Chen LA: Identification of featured biomarkers in different types of lung cancer with DNA microarray. Mol Biol Rep. 41:6357–6363. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Pan HW, Chou HY, Liu SH, Peng SY, Liu CL and Hsu HC: Role of L2DTL, cell cycle-regulated nuclear and centrosome protein, in aggressive hepatocellular carcinoma. Cell Cycle. 5:2676–2687. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Li T, Wan B, Huang J and Zhang X: Comparison of gene expression in hepatocellular carcinoma, liver development, and liver regeneration. Mol Genet Genomics. 283:485–492. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Lin ZY, Chuang WL and Chuang YH: Amphotericin B up-regulates angiogenic genes in hepatocellular carcinoma cell lines. Eur J Clin Invest. 39:239–245. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Huang F and Geng XP: Chemokines and hepatocellular carcinoma. World J Gastroenterol. 16:1832–1836. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Schimanski CC, Bahre R, Gockel I, Müller A, Frerichs K, Hörner V, Teufel A, Simiantonaki N, Biesterfeld S, Wehler T, et al: Dissemination of hepatocellular carcinoma is mediated via chemokine receptor CXCR4. Br J Cancer. 95:210–217. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Chen Y, Ramjiawan RR, Reiberger T, Ng MR, Hato T, Huang Y, Ochiai H, Kitahara S, Unan EC, Reddy TP, et al: CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology. 61:1591–1602. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Chew V, Tow C, Teo M, Wong HL, Chan J, Gehring A, Loh M, Bolze A, Quek R, Lee VK, et al: Inflammatory tumour microenvironment is associated with superior survival in hepatocellular carcinoma patients. J Hepatol. 52:370–379. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Bandiera S, Pfeffer S, Baumert TF and Zeisel MB: MiR-122-a key factor and therapeutic target in liver disease. J Hepatol. 62:448–457. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Navab R, Strumpf D, Bandarchi B, Zhu CQ, Pintilie M, Ramnarine VR, Ibrahimov E, Radulovich N, Leung L, Barczyk M, et al: Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer. Proc Natl Acad Sci USA. 108:pp. 7160–7165. 2011; View Article : Google Scholar : PubMed/NCBI

46 

Lee HJ, Jang M, Kim H, Kwak W, Park W, Hwang JY, Lee CK, Jang GW, Park MN, Kim HC, et al: Comparative transcriptome analysis of adipose tissues reveals that ECM-receptor interaction is involved in the depot-specific adipogenesis in cattle. PLoS One. 8:e662672013. View Article : Google Scholar : PubMed/NCBI

47 

Fukui T, Shaykhiev R, Agosto-Perez F, Mezey JG, Downey RJ, Travis WD and Crystal RG: Lung adenocarcinoma subtypes based on expression of human airway basal cell genes. Eur Respir J. 42:1332–1344. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Hou Y: Molecular basis of the effect of midkine on tumor growth in human gastric cancer cell BGC823. Biomedicine and pharmacotherapy. 62:4222008. View Article : Google Scholar

49 

Xia L, Huang W, Tian D, Zhang L, Qi X, Chen Z, Shang X, Nie Y and Wu K: Forkhead box Q1 promotes hepatocellular carcinoma metastasis by transactivating ZEB2 and VersicanV1 expression. Hepatology. 59:958–973. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Hur W, Rhim H, Jung CK, Kim JD, Bae SH, Jang JW, Yang JM, Oh ST, Kim DG, Wang HJ, et al: SOX4 overexpression regulates the p53-mediated apoptosis in hepatocellular carcinoma: Clinical implication and functional analysis in vitro. Carcinogenesis. 31:1298–1307. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Liao YL, Sun YM, Chau GY, Chau YP, Lai TC, Wang JL, Horng JT, Hsiao M and Tsou AP: Identification of SOX4 target genes using phylogenetic footprinting-based prediction from expression microarrays suggests that overexpression of SOX4 potentiates metastasis in hepatocellular carcinoma. Oncogene. 27:5578–5589. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Chang Q, Chen J, Beezhold KJ, Castranova V, Shi X and Chen F: JNK1 activation predicts the prognostic outcome of the human hepatocellular carcinoma. Mol Cancer. 8:1–14. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Elyakim E, Sitbon E, Faerman A, Tabak S, Montia E, Belanis L, Dov A, Marcusson EG, Bennett CF, Chajut A, et al: hsa-miR-191 is a candidate oncogene target for hepatocellular carcinoma therapy. Cancer Res. 70:8077–8087. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 15 Issue 6

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
He B, He Y, Shi W, Gong S, Chen X, Xiao J, Gu J, Ding W and Wang Y: Bioinformatics analysis of gene expression alterations in microRNA‑122 knockout mice with hepatocellular carcinoma. Mol Med Rep 15: 3681-3689, 2017
APA
He, B., He, Y., Shi, W., Gong, S., Chen, X., Xiao, J. ... Wang, Y. (2017). Bioinformatics analysis of gene expression alterations in microRNA‑122 knockout mice with hepatocellular carcinoma. Molecular Medicine Reports, 15, 3681-3689. https://doi.org/10.3892/mmr.2017.6445
MLA
He, B., He, Y., Shi, W., Gong, S., Chen, X., Xiao, J., Gu, J., Ding, W., Wang, Y."Bioinformatics analysis of gene expression alterations in microRNA‑122 knockout mice with hepatocellular carcinoma". Molecular Medicine Reports 15.6 (2017): 3681-3689.
Chicago
He, B., He, Y., Shi, W., Gong, S., Chen, X., Xiao, J., Gu, J., Ding, W., Wang, Y."Bioinformatics analysis of gene expression alterations in microRNA‑122 knockout mice with hepatocellular carcinoma". Molecular Medicine Reports 15, no. 6 (2017): 3681-3689. https://doi.org/10.3892/mmr.2017.6445