Open Access

Identification of the association between HMMR expression and progression of hepatocellular carcinoma via construction of a co‑expression network

  • Authors:
    • Donglan Lu
    • Xue Bai
    • Qiyuan Zou
    • Zuhuan Gan
    • Yufeng Lv
  • View Affiliations

  • Published online on: July 9, 2020     https://doi.org/10.3892/ol.2020.11844
  • Pages: 2645-2654
  • Copyright: © Lu 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

The aim of the present study was to identify key genes involved in the progression of hepatocellular carcinoma (HCC). According to the theory of the multistep process of hepatocarcinogenesis and weighted gene co‑expression network analysis, hub genes associated with the progression of HCC were identified using the gene expression profiles of patients with normal to chronic hepatitis/cirrhosis and dysplastic nodules to HCC. An independent dataset was used to verify the association between hub gene and clinical phenotype. The diagnostic and prognostic value of hub genes regarding HCC were evaluated. Gene set enrichment analysis (GSEA) was performed to explore the function of hub genes. A co‑expression gene module positively associated with HCC progression was identified. Combined with a protein‑protein interaction (PPI) network, a total of 10 common hub genes common to both the module of interest and the PPI network were selected as hub genes. Hyaluronan mediated motility receptor (HMMR) was selected as the candidate gene and was significantly upregulated in HCC at the mRNA and protein expression levels. HMMR is a promising diagnostic biomarker for HCC, and is also associated with its progression. The expression of HMMR was positively correlated with HCC tumor grade, pathological stage, tumor stage and Ishak score. The expression of HMMR was an independent prognostic factor compared with clinicopathological features. Patients with high expression levels of HMMR exhibited a less favorable prognosis. GSEA identified 6 representative gene sets that were associated with cancer. Overall, HMMR may serve an important role in HCC and may have potential as a biomarker of HCC diagnosis and progression.

Introduction

Liver cancer, of which 75–85% of cases consist of hepatocellular carcinoma (HCC), was the sixth most commonly diagnosed cancer and the fourth leading cause of cancer-associated death worldwide in 2018 (1). HCC is one of the few types of cancer which has had a continued increase in incidence over the last decade (2). The risk factors for HCC include chronic infection with hepatitis C or hepatitis B virus, high alcohol intake, aflatoxin B1, obesity, smoking and type 2 diabetes (35). Hepatocarcinogenesis is considered to be a multistep process evolving from normal to chronic hepatitis/cirrhosis and dysplastic nodules to HCC (6,7). Curative treatment options are limited to surgical resection of the tumor or liver transplantation; however, >70% of patients with HCC will encounter recurrence within 5 years after surgery (810). The specific mechanisms underlying the progression from healthy liver to chronic hepatitis/cirrhosis and dysplastic nodules to HCC are still elusive. The investigation of these mechanisms may help identify potential therapeutic targets to prevent the development and recurrence of HCC and biomarkers of these processes may help clinicians monitor this disease progression.

Previously, with the development of high-throughput and microarray technology, gene expression profiles have been used to identify genes associated with the progression of HCC (1113). However, the majority of these studies focused on the screening of differentially expressed genes without considering the correlations between genes, despite the fact that genes with similar expression patterns may be functionally related (14). Weighted gene co-expression network analysis (WGCNA) can be used to analyze the associations between gene sets and indicators of tumor progression, including tumor stages and grades (15,16). In the present study, the various stages of progression from healthy liver to chronic hepatitis/cirrhosis and dysplastic nodules to HCC were treated as phenotypes. It was hypothesized that the expression patterns of certain genes would be closely associated with these phenotypes.

According to the theory of the multistep process of hepatocarcinogenesis (6,7) and WGCNA, the present study aimed to identify network-centric genes associated with HCC progression by constructing a co-expression network using gene expression profiles from normal to chronic hepatitis/cirrhosis and dysplastic nodules to HCC. Hyaluronan mediated motility receptor (HMMR) was identified to exhibit a strong correlation with the progression of HCC and may represent a promising marker for the prevention and treatment of HCC.

Materials and methods

Data collection

The microarray dataset (GSE89377) of HCC was retrieved from the Gene Expression Omnibus (GEO) database (ncbi.nlm.nih.gov/geo/) using the R (http://www.R-project.org/) package GEOquery (17) in RStudio (http://www.rstudio.com/). The GSE89377 dataset was collected using Illumina HumanHT-12 version 3.0 expression beadchip (Illumina, Inc.) and was used to construct co-expression networks and identify hub genes in the present study. There were 13 healthy liver tissue samples and 94 tissues covering 9 stages of HCC progression in the GSE89377 dataset, including low grade chronic hepatitis (n=8), high grade chronic hepatitis (n=12), cirrhosis (n=12), low grade dysplastic nodules (n=11), high grade dysplastic nodules (n=11), early HCC (n=5), HCC with grade 1 (n=9), HCC with grade 2 (n=12) and HCC with grade 3 (n=14). The gene expression profiles had been normalized using quantile normalization with GenPlex version 3.0 by Jung Woo Eun from The Catholic University of Korea. An independent dataset including RNA-sequencing data and clinical information was obtained from The Cancer Genome Atlas (TCGA) database (cancer.gov/tcga) to further verify the association of hub genes and clinical phenotypes. The gene expression profiles of GSE87630 (18) based on the GPL6947 dataset included 64 HCC and 30 non-tumor profiles used to validate the aberrant expression of the hub genes. The gene expression profiles of GSE87630 were processed using the lumi package (19) in R. As these data are publicly available and open-access, ethical approval was not necessary for the present study.

Weighted gene co-expression network construction

Probes were filtered by variance as recommended (15), and the 4,881 probes with the highest variance were selected from 48,803 probes. The ‘WGCNA’ package (15) was used to construct a co-expression network for the 4,881 probes according to the protocols of WGCNA and R software. First, Pearson's correlation matrices were performed for all pair-wise genes. Subsequently, an adjacency matrix was constructed using a power adjacency function [αmn=Power (Smnβ)=|Smn|β; αmn, adjacency between two genes; Smn, Pearson's correlations between two genes]. β is a soft-thresholding parameter that emphasizes strong correlations between genes and penalizes weak correlations. In the present study, the power of β=9 (scale-free R2=0.85) was chosen in accordance with the scale-free topology criterion (Fig. 1A). Next, the adjacency was transformed into a topological overlap matrix that measured the network connectivity of a gene, defined as the sum of its adjacency with all other genes for network generation (20). Finally, the ‘cutreeStaticColor’ function was applied to classify similar expression genes into gene modules (minModuleSize=30; mergeCutHeight=0.25).

Identification of clinically significant modules and functional enrichment analysis

Gene significance (GS) was defined as the log10 transformation of the P-value in the linear regression between gene expression and HCC progress. Module significance (MS) was the mean GS for all the genes in a module. In general, the module with the absolute MS ranked first or second (ranked by MS) amongst all modules was considered as the module correlating with HCC progression. In the present study, the module exhibiting the strongest positive correlation with HCC progression was selected for further analysis and termed the primary module. Module eigengenes (MEs) were considered as the major components in the principal component analysis for each gene module and the expression patterns of all genes could be summarized into a single characteristic expression profile within a given module. The correlation between MEs and clinical traits was calculated to identify the relevant module. As there is more potential of oncogene as a marker or therapeutic target (21), the focus was on modules that are positively associated with HCC progression. In addition, functional Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using the ‘clusterProfiler’ R package (22) in order to uncover the biologic function of genes in the primary module. P-value [adjusted by false discovery rate (FDR)]<0.01 was set as the cutoff criteria.

Identification of hub genes

In the present study, hub genes in the primary module were defined by module connectivity, measured by the absolute value of the Pearson's correlation (cor.geneModuleMembership ≥0.8) and clinical trait relationship, measured by absolute value of the Pearson's correlation (cor.geneTraitSignificance ≥0.7) (23). Furthermore, a protein-protein interaction (PPI) network was constructed by uploading all genes in the primary module to the Search Tool for the Retrieval of Interacting Genes (STRING) database (24). Overall, 50 genes with the highest connectivity degree were defined as hub genes in the PPI network. The connectivity degree for each gene in the PPI network was calculated using the ‘cytoHubba’ (25) plugin in Cytoscape version 3.6.1 software (26). The hub genes common to both co-expression network and PPI networks were selected as ‘real’ hub genes and included for further analyses.

Hub gene validation and survival analysis

The Human Protein Atlas (27) was used to validate the expression of the hub genes at the protein level. The Human Protein Atlas is a publicly available database, all data and images are available for free download and non-commercial use. For validation of the correlation of hub genes and HCC progression, 371 HCC samples from TCGA database were analyzed to calculate the Pearson's correlation coefficient between hub gene expression and certain clinicopathological features. To evaluate the impact of the hub genes on the prognosis of patients with HCC, overall and disease-free survival rate were analyzed using Gene Expression Profiling Interactive Analysis tools (GEPIA) (28). The predictive value for prognosis between hub genes and routine clinicopathological factors were compared using univariate and multivariate Cox regression analyses in an HCC dataset from TCGA (TCGA-LIHC). These clinicopathological factors comprised alpha feto protein (AFP) (29), vascular invasion (30), Ishak score (32), and tumor pathological staging (33). P<0.05 were set as the cut-off criteria for significance.

Gene set enrichment analysis (GSEA)

371 HCC samples from RNA-sequencing data (displayed as read counts) were divided into two groups (high vs. low) according to the expression level of the candidate gene and the median expression value was selected as the cut-off point. The RNA-sequencing data was normalized using the limma package (34) in R. To determine the potential function of candidate gene, GSEA (35) was performed between the 2 groups. Hallmark gene sets summarize and represent specific well-defined biological states or processes and display coherent expression. These gene sets were generated by a computational methodology based on identifying overlaps between gene sets in other MSigDB collections (36) and retaining genes that display coordinate expression. Thus, the Hallmark gene sets (37) were selected as the reference gene sets. FDR <0.05 was set as the cut-off criteria.

Statistical analysis

The expression levels of the hub genes were analyzed using unpaired Student's t-tests in the comparison of two groups. ANOVA and Dunnett's post-hoc test were used for multiple comparisons using the multcomp package (CRAN.R-project.org/package=multcomp) in R. Univariate/multivariate Cox proportional hazards analyses and Kaplan-Meier survival analysis with log-rank method were used to compare survival between the two groups of patients. P<0.05 was considered to indicate a statistically significant difference.

Results

Weighted co-expression network construction and primary module identification

Overall, 13 healthy liver samples and 94 samples from different stages (low grade chronic hepatitis, high grade chronic hepatitis, cirrhosis, low grade dysplastic nodules, high grade dysplastic nodules, early HCC, HCC with grade 1, HCC with grade 2 and HCC with grade 3) of HCC progression were included in co-expression analysis (Fig. 1B). In the present study, the power of β=9 (scale free R2=0.85) was selected as the soft-threshold to ensure a scale-free network (Fig. 1A) and 11 modules were identified (Fig. 1C). The highest association between module and phenotype was revealed to be between the yellow module and clinical phenotype (r=−0.77; P=8×10−22; Fig. 1D); however, the brown module and clinical phenotype exhibited the highest positive correlation (r=0.73; P=9×10−19; Fig. 1D). This indicated that the brown module genes may be oncogenes and the yellow module genes may be tumor suppressor genes. As there is more potential of oncogene as a marker or therapeutic target (21), the brown module was selected and included in subsequent analyses.

To explore the biological relevance of the brown module, GO and KEGG enrichment analyses were performed for 671 genes using the ‘clusterProfiler’ package. GO enrichment analyses contained three parts: Cellular component (CC; Fig. 2A); biological process (BP; Fig. 2B); and molecular function (MF; Fig. 2C). The brown module genes were involved in mitotic-related CCs and BPs, such as ‘microtubule’, ‘nuclear division’ and ‘cell cycle G2/M phase transition’. While the brown module genes were involved in kinase-related MFs, such as ‘cyclin-dependent protein kinase activity’. The results of enrichment analyses indicated that the brown module genes were involved in various cancer-associated pathways (Fig. 2D), such as ‘cell cycle’ and ‘p53 signaling pathway’. The brown module genes associated with HCC are also involved in some viral infection-related pathways, such as human T-cell leukemia virus 1 infection.

Identification of hub genes

In the present study, 20 genes with high connectivity (cor.geneModuleMembership; ≥0.8) and high clinical trait relationship (cor.geneTraitSignificance; ≥0.7) in the brown module were selected as hub genes in WGCNA. A PPI network (Table SI; Fig. 3) was constructed using Cytoscape according to the STRING database and 50 genes with the highest connectivity degree were defined as hub genes in the PPI network. Then, a total of 10 genes (TOP2A, CDC20, CCNB2, PRC1, UBE2C, PTTG1, KIF20A, HMMR, NUSAP1 and RACGAP1) common to both the co-expression network and the PPI network were selected as ‘real’ hub genes (Table I). The aberrant expression data of the 10 hub genes were validated in an independent data set (Fig. 4). HMMR was selected as the candidate gene for further analysis due to the few existing reports about its role in HCC.

Table I.

Common hub genes in the brown module from weighted gene co-expression network analysis in GSE89377 and PPI network.

Table I.

Common hub genes in the brown module from weighted gene co-expression network analysis in GSE89377 and PPI network.

Weighted gene co-expression network analysisPPI network


ProbeGene cor.geneModuleMembership cor.geneTraitSignificanceConnectivity degree
ILMN_1686097TOP2A0.9560.748142
ILMN_1663390CDC200.9610.724121
ILMN_1801939CCNB20.9620.712111
ILMN_1728934PRC10.9620.725102
ILMN_1714730UBE2C0.9540.71097
ILMN_1753196PTTG10.9460.70692
ILMN_1695658KIF20A0.9380.71592
ILMN_1781942HMMR0.9160.70697
ILMN_1726720NUSAP10.9330.70986
ILMN_2077550RACGAP10.9590.71783

[i] PPI, Protein-Protein Interaction.

Hub gene validation and survival analysis

The expression of HMMR at the mRNA and protein levels were both significantly higher in HCC tissue compared with healthy liver tissue (Fig. 5A-C). In the GSE89377 dataset, HMMR exhibited diagnostic efficiency for HCC with an area under curve (AUC)=0.949, sensitivity=0.875 and specificity=0.910 (Fig. 5D). Based the results of WGCNA, the expression of HMMR was positively correlated with the progression of HCC (cor.geneTraitSignificance r=0.706; P=2.00×10−17). This correlation was validated in the HCC dataset from TCGA (r=0.290; P=3.57×10−6; Fig. 5E). The expression of HMMR was positively correlated with HCC pathological stage (r=0.062; P=0.008), tumor (T) stage (r=0.069; P=4.19×10−4) and Ishak score (Pearson correlation coefficient=0.178; P=0.004; Fig. 5E). Using GEPIA tools, it was revealed that patients with higher expression levels of HMMR exhibited significantly shorter overall survival (Fig. 5F, left) and disease-free survival rate (Fig. 5F, right). Furthermore, the expression of HMMR is an independent prognostic factor compared with routine clinicopathological features, not only in overall survival but also disease-free survival rate, in the HCC dataset from TCGA (Tables II and III) (multivariate Cox regression analysis P<0.05).

Figure 5.

Validation of aberrant expression of HMMR at transcription and protein levels and the prognostic value of HMMR in HCC. (A) Expression of HMMR at different pathological stages from normal to chronic hepatitis/cirrhosis and dysplastic nodules to HCC in the GSE89377 dataset. (B) Expression of HMMR between tumor tissues and non-tumor tissues in hepatocellular carcinoma dataset from TCGA. (C) HMMR protein was upregulated in hepatocellular carcinoma (images.proteinatlas.org/2433/6865_B_8_6.jpg) compared with healthy liver tissue (images.proteinatlas.org/2433/6892_A_8_4.jpg) (antibody CAB002433) using data from the Human Protein Atlas database. The healthy liver tissue was from a female (patient ID: 1846) and the hepatocellular carcinoma tissue was from a male (patient ID: 2325). (D) ROC curves of the expression of HMMR for hepatocellular carcinoma diagnosis in the GSE89377 dataset. (E) Pearson correlation between HMMR expression and routine clinicopathological features. This shows the correlation coefficient when P<0.01. (F) Kaplan-Meier curves obtained using the median value of HMMR expression to separate patients into high- and low-expression groups in Gene Expression Profiling Interactive Analysis. HCC, hepatocellular carcinoma; HMRR, Hyaluronan mediated motility receptor; LIHC, liver hepatocellular carcinoma; TCGA, The Cancer Genome Atlas; ROC, receiver operating characteristic; HR, hazard ratio; T, tumor; CHLG, chronic hepatitis with low grade; CHHG, chronic hepatitis with high grade; DNLG, dysplastic nodules with low grade; DNHG, dysplastic nodules with high grade; eHCC, early hepatocellular carcinoma; HCC-TG1, hepatocellular carcinoma with grade 1; HCC-TG2, hepatocellular carcinoma with grade 2; HCC-TG3, hepatocellular carcinoma with grade 3.

Table II.

Univariate and multivariate COX regression analyses for overall survival in hepatocellular carcinoma dataset of The Cancer Genome Atlas.

Table II.

Univariate and multivariate COX regression analyses for overall survival in hepatocellular carcinoma dataset of The Cancer Genome Atlas.

Univariate analysisMultivariate analysis


FactorsP-valueHRHR, 95% CIP-valueHRHR, 95% CI
Sex (male vs. female)0.2621.2250.859–1.745
Age, years (>65 vs. ≤65)0.1861.2650.893–1.791
AFP, ng/ml (>20 vs. ≤20) (29)0.026a1.6411.061–2.5400.5631.1500.716–1.846
Vascular invasion (positive vs. negative) (30)0.1551.3510.892–2.047
Child-pugh score (B/C vs. A) (31)0.1841.6140.796–3.270
Ishak score (5–6 vs. 0–4) (32)0.4970.8290.483–1.424
Tumor Grade (G3/4 vs. G1/2)0.5421.1190.780–1.604
Pathological T stage (T3/4 vs. T1/2) (33) <0.001a2.5371.783–3.6090.8891.1530.155–8.607
Pathological stage (III/IV vs. I/II) (33) <0.001a2.4461.687–3.5450.6341.6200.222–11.808
HMMR expression level (high vs. low) <0.001a2.1361.498–3.0440.007a1.9171.192–3.085

a P<0.05. T, tumor; HR, hazard ratio; CI, confidence interval; HMMR, hyaluronan mediated motility receptor; AFP, alpha-fetoprotein.

Table III.

Univariate and multivariate Cox regression analyses for disease-free survival in HCC dataset of TCGA.

Table III.

Univariate and multivariate Cox regression analyses for disease-free survival in HCC dataset of TCGA.

Univariate analysisMultivariate analysis


Clinicopathological factorsP-valueHRHR (95% CI)P-valueHRHR (95% CI)
Sex (male vs. female)0.9191.0190.714–1.454
Age, years (>65 vs. ≤65)0.0811.0430.739–1.472
AFP, ng/ml (>20 vs. ≤20) (29)0.4961.1490.771–1.712
Vascular invasion (positive vs. negative) (30)0.029a1.5401.045–2.2680.3971.1980.788–1.821
Child-pugh score (B/C vs. A) (31)0.2461.5420.742–3.204
Ishak score (5–6 vs. 0–4) (32)0.6231.1160.721–1.727
Tumor Grade (G3/4 vs. G1/2)0.8291.0390.733–1.474
Pathological T stage (T3/4 vs. T1/2) (33) <0.001b2.9402.071–4.1730.5180.5120.067–3.902
Pathological stage (III/IV vs. I/II) (34) <0.001b2.8852.009–4.1420.1264.7690.644–35.330
HMMR expression level (high vs. low)0.002b1.6971.213–2.3730.038a1.5271.024–2.277

a P<0.05

b P<0.01. HCC, hepatocellular carcinoma; TCGA, The Cancer Genome Atlas; HR, hazard ratio; CI, confidence interval; T, tumor; G, grade; AFP, alpha-fetoprotein; HMMR, Hyaluronan mediated motility receptor expression.

GSEA

To analyze the function of HMMR in HCC, GSEA was conducted to compare HMMR with hallmark gene sets. 371 HCC samples were divided into two groups (high vs. low) according to the HMMR median expression level (2.62). Under the cut-off criteria of FDR <0.05, a total of 8 functional gene sets were enriched. Overall, 6 representative gene sets were significantly associated with cancer, including ‘mitotic spindle’, ‘G2/M checkpoint’, ‘MYC targets v1’, ‘E2F targets’, ‘DNA repair’ and ‘mTORC1 signaling’ (Fig. 6).

Discussion

In previous years, the concept of multi-step human hepatocarcinogenesis has been well documented (6,7,38). Chronic liver inflammation can result in repeated cell injury, death and regeneration cycles, resulting in subsequent epigenetic and genetic alterations of hepatocytes (39). Phenotypically abnormal precursor hepatic lesions, including cirrhotic nodules, low-grade dysplastic nodules and high-grade dysplastic nodules dedifferentiate and gradually evolve to HCC (40). This process exists on a biologic continuum and may occur simultaneously at various rates throughout the liver; however, the specific molecular mechanisms underlying this process are yet to be elucidated. In the present study, several modules associated with this process were identified using WGCNA. In particular, genes in the brown module exhibited a strong positive correlation with this process, indicating that gene expression in the module gradually increase as the process progresses. Functional enrichment analysis revealed that genes in the brown module significantly influenced cell cycle-associated biological processes, for example ‘cell cycle G2/M phase transition’, ‘cell cycle G1/S phase transition’ and ‘mitotic nuclear division’, and cancer-related pathways, including ‘p53 signaling pathway’ and ‘cell cycle’.

Markers which accurately reflect the process from normal to chronic hepatitis/cirrhosis and dysplastic nodules to HCC are lacking in clinical practice and novel candidate molecules are needed. In the present study, according to the theory of the multistep process of hepatocarcinogenesis and WGCNA, a total of 10 hub genes common to the primary module and PPI network were selected as hub genes, including TOP2A, CDC20, CCNB2, PRC1, UBE2C, PTTG1, KIF20A, HMMR, NUSAP1 and RACGAP1. Previous studies had reported almost all ten genes to be associated with the progression of HCC (4149). HMMR was chosen as the candidate gene, since few studies have identified its role in HCC (50). HMMR (also known as CD168/IHABP/RHAMM) (51) is highly expressed in various solid tumors and it is described as a cancer-associated antigen, which is involved in both tumorigenesis and progression/metastasis (5256). HMMR was identified as a breast cancer susceptibility gene (57) and was once considered an ideal target antigen for immunotherapy of acute myeloid leukemia (58); however, the association between HMMR and HCC has been rarely reported.

In the present study, HMMR was significantly upregulated in HCC tissue compared with healthy liver tissue at both the mRNA and protein expression levels. HMMR is a promising diagnostic biomarker for HCC (AUC=0.949; sensitivity=0.875; specificity=0.910). In addition, the progression of HCC was associated with the upregulation of HMMR. Notably, the expression of HMMR was positively correlated with HCC tumor grade, pathological stage, T stage and Ishak score. Patients with HCC with higher expression levels of HMMR exhibited significantly shorter overall survival and disease-free survival times. Moreover, the expression of HMMR remained an independent prognostic factor compared with routine clinicopathological features. The current results indicated that HMMR may serve as a biomarker of HCC progression. Thus, patients with HCC and high expression levels of HMMR have a higher risk for recurrence and should be followed up more frequently than the routine schedule.

In order to reveal the function of HMMR in HCC, GSEA was performed. Overall, 6 representative gene sets, including ‘mitotic spindle’, ‘G2/M checkpoint’, ‘MYC targets v1’, ‘E2F targets’, ‘DNA repair’ and ‘mTORC1 signaling’, were significantly associated with cancer and enriched in samples with high expression levels of HMMR. This indicates that HMMR may interact with these genes or pathways to promote the progression of HCC. The present findings may improve our understanding of the role of HMMR in HCC and inform future research.

Notably, there were certain limitations to the present study. Firstly, the expression of HMMR was quantified and the values may vary on different platforms. The establishment of a standard is required before being applied to clinical practice. Secondly, as the present study only performed a bioinformatic analysis, including GSEA analysis to help identify the function of HMMR in HCC, it is not clear whether HMMR expression is causal or merely a biomarker of HCC progression. Whether HMMR can be used as a therapeutic target for HCC requires further molecular experimental verification.

In conclusion, the present study revealed that patients with HCC with high expression of HMMR exhibit a less favorable prognosis, suggesting that HMMR may serve an important role in HCC and has potential as a biomarker of HCC diagnosis and progression.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This research was supported by the Youth Science Foundation of Guangxi Medical University, Nanning, China (grant no. GXMUYSF 201716).

Availability of data and materials

The dataset of GSE89377 and GSE87630 are available from the Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo). The dataset of TCGA-LIHC are available from The Cancer Genome Atlas (cancer.gov/tcga).

Authors' contributions

YL designed the study and reviewed the manuscript. DL and XB analyzed the data and wrote the manuscript. ZG and QZ assisted with analyzing the data and writing the manuscript. All authors read and approved the final 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.

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 

Forner A, Reig M and Bruix J: Hepatocellular carcinoma. Lancet. 391:1301–1314. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Heidelbaugh JJ and Bruderly M: Cirrhosis and chronic liver failure: Part I. Diagnosis and evaluation. Am Fam Physician. 74:756–762. 2006.PubMed/NCBI

4 

Alter MJ: Epidemiology of hepatitis C virus infection. World J Gastroenterol. 13:2436–2441. 2007. View Article : Google Scholar : PubMed/NCBI

5 

El-Serag HB, Hampel H and Javadi F: The association between diabetes and hepatocellular carcinoma: A systematic review of epidemiologic evidence. Clin Gastroenterol Hepatol. 4:369–380. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Marquardt JU, Seo D, Andersen JB, Gillen MC, Kim MS, Conner EA, Galle PR, Factor VM, Park YN and Thorgeirsson SS: Sequential transcriptome analysis of human liver cancer indicates late stage acquisition of malignant traits. J Hepatol. 60:346–353. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Kudo M: Multistep human hepatocarcinogenesis: Correlation of imaging with pathology. J Gastroenterol. 44 (Suppl 19):S112–S118. 2009. View Article : Google Scholar

8 

Rahbari NN, Mehrabi A, Mollberg NM, Müller SA, Koch M, Büchler MW and Weitz J: Hepatocellular carcinoma: Current management and perspectives for the future. Ann Surg. 253:453–469. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Hasegawa K, Kokudo N, Makuuchi M, Izumi N, Ichida T, Kudo M, Ku Y, Sakamoto M, Nakashima O, Matsui O and Matsuyama Y: Comparison of resection and ablation for hepatocellular carcinoma: A cohort study based on a Japanese nationwide survey. J Hepatol. 58:724–729. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Poon RT, Fan ST, Lo CM, Liu CL and Wong J: Long-term survival and pattern of recurrence after resection of small hepatocellular carcinoma in patients with preserved liver function: Implications for a strategy of salvage transplantation. Ann Surg. 235:373–382. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Cheng J, Wei D, Ji Y, Chen L, Yang L, Li G, Wu L, Hou T, Xie L, Ding G, et al: Integrative analysis of DNA methylation and gene expression reveals hepatocellular carcinoma-specific diagnostic biomarkers. Genome Med. 10:422018. View Article : Google Scholar : PubMed/NCBI

12 

Kalinich M, Bhan I, Kwan TT, Miyamoto DT, Javaid S, LiCausi JA, Milner JD, Hong X, Goyal L, Sil S, et al: An RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinoma. Proc Natl Acad Sci USA. 114:1123–1128. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C, Castro de Moura M, Putra J, Camprecios G, Bassaganyas L, Akers N, et al: Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology. 153:812–826. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Tavazoie S, Hughes JD, Campbell MJ, Cho RJ and Church GM: Systematic determination of genetic network architecture. Nat Genet. 22:281–285. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Langfelder P and Horvath S: WGCNA: An R package for weighted correlation network analysis. BMC Bioinformatics. 9:5592008. View Article : Google Scholar : PubMed/NCBI

16 

Chen L, Yuan L, Qian K, Qian G, Zhu Y, Wu CL, Dan HC, Xiao Y and Wang X: Identification of biomarkers associated with pathological stage and prognosis of clear cell renal cell carcinoma by co-expression network analysis. Front Physiol. 9:3992018. View Article : Google Scholar : PubMed/NCBI

17 

Davis S and Meltzer PS: GEOquery: A bridge between the gene expression omnibus (GEO) and BioConductor. Bioinformatics. 23:1846–1847. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Woo HG, Choi JH, Yoon S, Jee BA, Cho EJ, Lee JH, Yu SJ, Yoon JH, Yi NJ, Lee KW, et al: Integrative analysis of genomic and epigenomic regulation of the transcriptome in liver cancer. Nat Commun. 8:8392017. View Article : Google Scholar : PubMed/NCBI

19 

Du P, Kibbe WA and Lin SM: Lumi: A pipeline for processing Illumina microarray. Bioinformatics. 24:1547–1548. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Yip AM and Horvath S: Gene network interconnectedness and the generalized topological overlap measure. BMC Bioinformatics. 8:222007. View Article : Google Scholar : PubMed/NCBI

21 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Horvath S and Dong J: Geometric interpretation of gene coexpression network analysis. PLoS Comput Biol. 4:e10001172008. View Article : Google Scholar : PubMed/NCBI

23 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1): D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

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

26 

Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al: Proteomics. Tissue-based map of the human proteome. Science. 347:12604192015. View Article : Google Scholar : PubMed/NCBI

27 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res 45 (W1). W98–W102. 2017. View Article : Google Scholar

28 

Nomura F, Ohnishi K and Tanabe Y: Clinical features and prognosis of hepatocellular carcinoma with reference to serum alpha-fetoprotein levels. Analysis of 606 patients. Cancer. 64:1700–1707. 1989. View Article : Google Scholar : PubMed/NCBI

29 

Llovet JM, Schwartz M and Mazzaferro V: Resection and liver transplantation for hepatocellular carcinoma. Semin Liver Dis. 25:181–200. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Cholongitas E, Papatheodoridis GV, Vangeli M, Terreni N, Patch D and Burroughs AK: Systematic review: The model for end-stage liver disease-should it replace Child-Pugh's classification for assessing prognosis in cirrhosis? Aliment Pharmacol Ther. 22:1079–1089. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Ishak K, Baptista A, Bianchi L, Callea F, De Groote J, Gudat F, Denk H, Desmet V, Korb G, MacSween RN, et al: Histological grading and staging of chronic hepatitis. J Hepatol. 22:696–699. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Sobin LH, Gospodarowicz MK and Wittekind C: TNM classification of malignant tumours. 7th edition. John Wiley & Sons; 2009

33 

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

34 

Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 34:267–273. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP and Tamayo P: The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 1:417–425. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Niu ZS, Niu XJ, Wang WH and Zhao J: Latest developments in precancerous lesions of hepatocellular carcinoma. World J Gastroenterol. 22:3305–3314. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Batts KP and Ludwig J: Chronic hepatitis. An update on terminology and reporting. Am J Surg Pathol. 19:1409–1417. 1995. View Article : Google Scholar : PubMed/NCBI

39 

Choi JY, Lee JM and Sirlin CB: CT and MR imaging diagnosis and staging of hepatocellular carcinoma: Part I. Development, growth, and spread: Key pathologic and imaging aspects. Radiology. 272:635–654. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Wong N, Yeo W, Wong WL, Wong NL, Chan KY, Mo FK, Koh J, Chan SL, Chan AT, Lai PB, et al: TOP2A overexpression in hepatocellular carcinoma correlates with early age onset, shorter patients survival and chemoresistance. Int J Cancer. 124:644–652. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Li J, Gao JZ, Du JL, Huang ZX and Wei LX: Increased CDC20 expression is associated with development and progression of hepatocellular carcinoma. Int J Oncol. 45:1547–1555. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Gao CL, Wang GW, Yang GQ, Yang H and Zhuang L: Karyopherin subunit-α 2 expression accelerates cell cycle progression by upregulating CCNB2 and CDK1 in hepatocellular carcinoma. Oncol Lett. 15:2815–2820. 2018.PubMed/NCBI

43 

Chen J, Rajasekaran M, Xia H, Zhang X, Kong SN, Sekar K, Seshachalam VP, Deivasigamani A, Goh BK, Ooi LL, et al: The microtubule-associated protein PRC1 promotes early recurrence of hepatocellular carcinoma in association with the Wnt/β-catenin signalling pathway. Gut. 65:1522–1534. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Ieta K, Ojima E, Tanaka F, Nakamura Y, Haraguchi N, Mimori K, Inoue H, Kuwano H and Mori M: Identification of overexpressed genes in hepatocellular carcinoma, with special reference to ubiquitin-conjugating enzyme E2C gene expression. Int J Cancer. 121:33–38. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Molina-Jimenez F, Benedicto I, Murata M, Martín-Vílchez S, Seki T, Antonio Pintor-Toro J, Tortolero M, Moreno-Otero R, Okazaki K, Koike K, et al: Expression of pituitary tumor-transforming gene 1 (PTTG1)/securin in hepatitis B virus (HBV)-associated liver diseases: Evidence for an HBV X protein-mediated inhibition of PTTG1 ubiquitination and degradation. Hepatology. 51:777–787. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Lu M, Huang X, Chen Y, Fu Y, Xu C, Xiang W, Li C, Zhang S and Yu C: Aberrant KIF20A expression might independently predict poor overall survival and recurrence-free survival of hepatocellular carcinoma. IUBMB Life. 70:328–335. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Zhang M, Yang D, Liu X, Liu Y, Liang J, He H, Zhong K, Lin L, Tao G, Zhang C and Zhou J: Expression of Nusap1 in the surgical margins of hepatocellular carcinoma and its association with early recurrence. Nan Fang Yi Ke Da Xue Xue Bao. 33:937–938, inside back cover, 2013 (In Chinese). PubMed/NCBI

48 

Yang XM, Cao XY, He P, Li J, Feng MX, Zhang YL, Zhang XL, Wang YH, Yang Q, Zhu L, et al: Overexpression of Rac GTPase activating protein 1 contributes to proliferation of cancer cells by reducing hippo signaling to promote cytokinesis. Gastroenterology. 155:1233–1249 e22. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Wurmbach E, Chen YB, Khitrov G, Zhang W, Roayaie S, Schwartz M, Fiel I, Thung S, Mazzaferro V, Bruix J, et al: Genome-wide molecular profiles of HCV-induced dysplasia and hepatocellular carcinoma. Hepatology. 45:938–947. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Willemen Y, Van den Bergh JM, Bonte SM, Anguille S, Heirman C, Stein BM, Goossens H, Kerre T, Thielemans K, Peeters M, et al: The tumor-associated antigen RHAMM (HMMR/CD168) is expressed by monocyte-derived dendritic cells and presented to T cells. Oncotarget. 7:73960–73970. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Rein DT, Roehrig K, Schondorf T, Lazar A, Fleisch M, Niederacher D, Bender HG and Dall P: Expression of the hyaluronan receptor RHAMM in endometrial carcinomas suggests a role in tumour progression and metastasis. J Cancer Res Clin Oncol. 129:161–164. 2003. View Article : Google Scholar : PubMed/NCBI

52 

Kalmyrzaev B, Pharoah PD, Easton DF, Ponder BA and Dunning AM; SEARCH Team, : Hyaluronan-mediated motility receptor gene single nucleotide polymorphisms and risk of breast cancer. Cancer Epidemiol Biomarkers Prev. 17:3618–3620. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Buttermore ST, Hoffman MS, Kumar A, Champeaux A, Nicosia SV and Kruk PA: Increased RHAMM expression relates to ovarian cancer progression. J Ovarian Res. 10:662017. View Article : Google Scholar : PubMed/NCBI

54 

Koelzer VH, Huber B, Mele V, Iezzi G, Trippel M, Karamitopoulou E, Zlobec I and Lugli A: Expression of the hyaluronan-mediated motility receptor RHAMM in tumor budding cells identifies aggressive colorectal cancers. Hum Pathol. 46:1573–1581. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Ishigami S, Ueno S, Nishizono Y, Matsumoto M, Kurahara H, Arigami T, Uchikado Y, Setoyama T, Arima H, Yoshiaki K, et al: Prognostic impact of CD168 expression in gastric cancer. BMC Cancer. 11:1062011. View Article : Google Scholar : PubMed/NCBI

56 

Pujana MA, Han JD, Starita LM, Stevens KN, Tewari M, Ahn JS, Rennert G, Moreno V, Kirchhoff T, Gold B, et al: Network modeling links breast cancer susceptibility and centrosome dysfunction. Nat Genet. 39:1338–1349. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Snauwaert S, Vanhee S, Goetgeluk G, Verstichel G, Van Caeneghem Y, Velghe I, Philippé J, Berneman ZN, Plum J, Taghon T, et al: RHAMM/HMMR (CD168) is not an ideal target antigen for immunotherapy of acute myeloid leukemia. Haematologica. 97:1539–1547. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 20 Issue 3

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
Lu D, Bai X, Zou Q, Gan Z and Lv Y: Identification of the association between HMMR expression and progression of hepatocellular carcinoma via construction of a co‑expression network. Oncol Lett 20: 2645-2654, 2020
APA
Lu, D., Bai, X., Zou, Q., Gan, Z., & Lv, Y. (2020). Identification of the association between HMMR expression and progression of hepatocellular carcinoma via construction of a co‑expression network. Oncology Letters, 20, 2645-2654. https://doi.org/10.3892/ol.2020.11844
MLA
Lu, D., Bai, X., Zou, Q., Gan, Z., Lv, Y."Identification of the association between HMMR expression and progression of hepatocellular carcinoma via construction of a co‑expression network". Oncology Letters 20.3 (2020): 2645-2654.
Chicago
Lu, D., Bai, X., Zou, Q., Gan, Z., Lv, Y."Identification of the association between HMMR expression and progression of hepatocellular carcinoma via construction of a co‑expression network". Oncology Letters 20, no. 3 (2020): 2645-2654. https://doi.org/10.3892/ol.2020.11844