Skip to main content

ORIGINAL RESEARCH article

Front. Genet., 01 September 2021
Sec. Systems Biology Archive

Identification of Key Genes Associated With the Process of Hepatitis B Inflammation and Cancer Transformation by Integrated Bioinformatics Analysis

\r\nJingyuan ZhangJingyuan Zhang1Xinkui LiuXinkui Liu1Wei ZhouWei Zhou1Shan LuShan Lu1Chao WuChao Wu1Zhishan WuZhishan Wu1Runping LiuRunping Liu1Xiaojiaoyang LiXiaojiaoyang Li2Jiarui Wu*Jiarui Wu1*Yingying LiuYingying Liu1Siyu GuoSiyu Guo1Shanshan JiaShanshan Jia1Xiaomeng ZhangXiaomeng Zhang1Miaomiao WangMiaomiao Wang1
  • 1School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
  • 2School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China

Background: Hepatocellular carcinoma (HCC) has become the main cause of cancer death worldwide. More than half of hepatocellular carcinoma developed from hepatitis B virus infection (HBV). The purpose of this study is to find the key genes in the transformation process of liver inflammation and cancer and to inhibit the development of chronic inflammation and the transformation from disease to cancer.

Methods: Two groups of GEO data (including normal/HBV and HBV/HBV-HCC) were selected for differential expression analysis. The differential expression genes of HBV-HCC in TCGA were verified to coincide with the above genes to obtain overlapping genes. Then, functional enrichment analysis, modular analysis, and survival analysis were carried out on the key genes.

Results: We identified nine central genes (CDK1, MAD2L1, CCNA2, PTTG1, NEK2) that may be closely related to the transformation of hepatitis B. The survival and prognosis gene markers composed of PTTG1, MAD2L1, RRM2, TPX2, CDK1, NEK2, DEPDC1, and ZWINT were constructed, which performed well in predicting the overall survival rate.

Conclusion: The findings of this study have certain guiding significance for further research on the transformation of hepatitis B inflammatory cancer, inhibition of chronic inflammation, and molecular targeted therapy of cancer.

Introduction

Epidemiological studies have shown that chronic low-level inflammation can significantly increase the risk of cancer. On the one hand, during chronic inflammation caused by viral infections, a long-term abnormal expression of related proteins may induce physiological diseases and form a potential carcinogenic microenvironment. On the other hand, the occurrence and development of tumors also affect the inflammatory response process (Colotta et al., 2009; Huang et al., 2019). The global burden of hepatitis B virus (HBV) is enormous, with 257 million people chronically infected, causing more than 880,000 deaths worldwide each year (Iannacone and Guidotti, 2021). HBV has all the characteristics of ancient human pathogens, has chronic infections, including a prolonged asymptomatic period, and then gradually develops into clinical diseases. Persistent antiviral inflammation during chronic infection, immune clearance of virally infected cells, and hepatocyte regeneration all increase the risk of viral infectious liver disease developing into liver cancer (Xu et al., 2017; Revill et al., 2020). At present, vaccines and nucleoside or nucleotide drugs have been developed, with high coverage and efficacy. However, related studies have shown that vaccination and antiviral therapy can reduce infections but not completely eliminate risks, and reduce the rate of new infections and the development of liver disease (Chan et al., 2016; Fanning et al., 2019; Musa et al., 2019). Overall, up to 40% of men and women infected with HBV during the perinatal period will die from cirrhosis or hepatocellular carcinoma (Trépo et al., 2014; Schweitzer et al., 2015). There are many studies on the basis of clinical epidemiological studies on the mechanism of the relationship between hepatitis B and hepatitis B-related hepatocellular carcinoma, and significant progress has been made (Huang et al., 2019; Sun et al., 2019; Xie et al., 2020). However, few molecular targeted studies can comprehensively summarize the diagnosis, treatment, and prognosis of patients with progressive hepatitis B.

The rise of high-throughput gene chips and transcriptome sequencing and other transcriptome research methods has completely changed the previous systematic analysis methods for disease research (Kulasingam and Diamandis, 2008; Mair et al., 2019; Bustoros et al., 2020). High-throughput microarrays and RNA sequencing can detect changes in disease gene expression and transcriptome levels. These methods help to find reliable biological markers, classify diseases, and reveal the molecular mechanisms of disease development (Chen S. et al., 2020; Jin et al., 2020; Li X. et al., 2020). The purpose of this study is to find the key genes in the process of liver inflammation and cancer transformation and to provide reference for further study of the transformation of hepatitis B inflammatory cancer, inhibition of chronic inflammation, and molecular targeted therapy of cancer. In this study, we conducted a comprehensive analysis, selecting microarray data of normal tissues and HBV samples and microarray data of chronic hepatitis B-induced HCC and adjacent normal tissues, and separately analyzing the differentially expressed genes (DEGs) of the two groups of gene chips. Combining the TCGA DEG data of human hepatitis B-related hepatocellular carcinoma and normal liver tissue with the abovementioned chip data to obtain the key DEGs that directly affect the diagnosis and treatment of hepatitis B and later. Afterward, further functional enrichment analysis was conducted to analyze the main biological functions regulated by DEGs. Finally, through the use of protein–protein interaction (PPI) networks and survival analysis, key genes affecting the diagnosis, treatment, and prognosis of patients with progressive hepatitis B are identified. The detailed workflow of the study is shown in Figure 1.

FIGURE 1
www.frontiersin.org

Figure 1. The workflow for identifying key genes associated with HBV in inflammation and cancer transformation.

Materials and Methods

Gene Expression Profile Data

Gene expression profiles were extracted from the GSE83148 and GSE121248 data set, which was downloaded from the publicly available Gene Expression Omnibus database (GEO)1 (Clough and Barrett, 2016). GSE83148 (Zhou et al., 2017; Li et al., 2018; Chen Z. et al., 2019) and GSE121248 (Wang et al., 2007) are both based on the GPL570 ([HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array). The title of the GSE83148 data set is “Expression data of HBV infected liver tissue.” All hepatitis samples were HBV infected, which was validated by positive HBsAg or serum HBV-DNA. The samples with HCV infection or metabolic liver injury (e.g., fatty liver, chronic alcoholic hepatitis) were excluded. GSE83148 contains six human normal liver tissue samples and 122 HBV-infected hepatitis samples. The title of the GSE121248 data set is “Gene expression profiling of chronic hepatitis B induced HCC and adjacent-normal tissues.” Tissues from chronic hepatitis B-induced HCC and their adjacent normal tissues were isolated, and total RNA was extracted for Affymetrix gene microarray analysis. GSE121248 contains 37 chronic hepatitis B-induced HCC adjacent normal tissues and 70 human chronic hepatitis B-induced HCC liver tissues.

Screening of DEGs and Functional Enrichment Analysis

The limma software package in the R 3.6.3 software2 was used to normalize the matrix data of each GEO data set and the logarithm conversion based on 2. The DEG between the two groups of controls was screened through the limma software package. Corrected p < 0.05 and | log FC| > 1 were used as the cutoff criteria (Ritchie et al., 2015; Zhou et al., 2020). After that, functional enrichment analyses were performed on the obtained differential genes, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) (Rhee et al., 2008; Kanehisa et al., 2017). The DEGs of GO and KEGG pathway analysis were performed using Bioconductor clusterProfiler, org.Hs.eg.db, and DOSE, which are three R packages used for the enrichment analysis of gene clusters (Yu et al., 2012; Zou et al., 2019). p < 0.05 and q < 0.05 were defined as the cutoff criteria. In the GO analysis, p < 0.01 and q < 0.05 were defined as the cutoff criteria. Furthermore, in the KEGG analysis, p < 0.05 and q < 0.05 were defined as the cutoff criteria.

DEG Validation by TCGA

Using the RNA sequencing data in the TCGA HBV-related HCC data set, the results of the comprehensive analysis of the differential genes in the two GEO data sets were verified (Liu et al., 2018). The TCGA-Liver Hepatocellular Carcinoma (HCC) cohort with publicly available data3 was used for this study. From this cohort, 78 HCC cases with gene expression data set, epigenetic data, and copy number alteration data were selected. It contained 60 cases of HBV-related HCC, and 18 cases were HBV-related adjacent tissues. The above data were analyzed by the edgeR software package in the Sanger box.4 Genes with | log FC| > 1 and FDR < 0.05 are considered significant (FDR: false discovery rate). The common up-and-down overlapping genes between TCGA and the two GEO data sets were integrated for the next study. These genes are considered to be overlapping genes related to the occurrence and development of hepatitis B-related inflammation and cancer transformation. The obtained overlapping DEGs were visualized by TBtools for heat map analysis (Chen S. et al., 2020).

GO and KEGG Pathway Enrichment Analysis of Key Genes

To elucidate potential biological processes, molecular functions, cellular components, and signaling pathways associated with the overlapping DEGs, we performed GO enrichment analysis and KEGG enrichment analysis utilizing the Database for Annotation, Visualization and Integrated Discovery5 (DAVID 6.8) (Dennis et al., 2003; Huang da et al., 2009). FDR < 0.05 was defined as the cutoff criterion. The results of the GO functional enrichment analysis were visualized via GOplot software package in the R 3.6.3 software (Walter et al., 2015). The results of KEGG functional enrichment analysis are drawn by Sanger box.6

PPI Network and Module Analysis

The String 11.0 database7 is a database that searches for interactions between known proteins and predicted proteins (Szklarczyk et al., 2017). The database is used to study PPI networks, which helps to mine core regulatory genes. In this study, we selected protein interaction results with confidence greater than 0.7 for the next analysis. Data of protein interaction were imported into Cytoscape 3.7.18 for visual analysis (Franz et al., 2016). In addition, in order to detect the hub cluster module in the PPI network, we used the Molecular Complex Detection (MCODE) application with default parameters in Cytoscape 3.7.1 for module analysis (Zhang et al., 2020).

Expression Level Analysis and Correlation Analysis of the Key Genes

The violin diagram tool in Sanger Box9 was used to show the difference in the expression of key genes in HBV-related HCC tissues and normal tissues. Gene Expression Profiling Interactive Analysis10 (GEPIA) is the dynamic analysis of gene expression profiling data. It is a newly developed public database for cancer and normal gene expression profiling. GEPIA analyzed the RNA sequencing expression data of 9,736 tumors and 8,587 normal samples from TCGA and GTEx projects (Tang et al., 2017). Perform pairwise gene correlation analysis on any given TCGA and/or GTEx expression dataset and check the relative ratio between the two genes.

Survival Analysis

Clinical information for patients with hepatocellular carcinoma can also be downloaded from TCGA (see text footnote 3). After screening HBV-related HCC, after deleting patients without overall survival (OS) data and overlapping DEG gene expression profiles, 60 patients with HBV-related HCC were used for survival analysis. Univariate Cox proportional hazards regression analysis was used to identify candidate genes that were highly correlated with survival. Cox proportional hazards regression analysis screened prognostic gene signatures from DEGs, p < 0.05. A Cox proportional hazards regression model was constructed with key prognostic genes as dependent variables, with the purpose of evaluating the relative contribution of key prognostic genes to patient survival prediction. We have constructed a prediction formula for gene characteristics. The following formula of the model is as follows: risk score = gene 1 × β1 gene 1 expression + gene 2 × β2 gene 2 expression + … gene n × βn expression gene. The formula is a linear combination in which the gene expression value of each gene and the regression coefficient (β) were obtained from the multiple Cox proportional hazards regression model (George et al., 2014; Zhou et al., 2016; Huang et al., 2017, 2018; Liu et al., 2019). The survminer package and ggrisk package in the R language were used to draw a riskplot and K-M survival curves (Cox, 1972; Zhou et al., 2016; Li X. et al., 2020). The LIRI data were downloaded in the ICGC database,11 and 260 primary solid tumor tissue samples were extracted. Samples with complete expression profile data and clinical information were selected, and RNA-seq data and clinical information of 231 tumor samples were obtained. These samples were mainly from Japanese people with hepatocellular carcinoma, and the FPKM values from genes were used. The data in ICGC were taken as the test set. Nine prognostic genes in the current TCGA were selected as the test set, the training set and the test set were modeled, and the model was verified (He et al., 2020; Liang et al., 2020). In order to analyze the accuracy of survival prediction performance through a risk scoring model, a time-dependent receiver operating characteristic (ROC) curve was constructed. The ability of prognostic gene signatures to predict clinical outcome depends on the area of the AUC curve. When AUC > 0.5, the closer AUC is to 1, the better the prognosis (Heagerty and Zheng, 2005).

Results

Identification of DEGs

The GSE83148 data set includes six human normal liver tissue samples and 122 HBV-infected hepatitis samples. Supplementary Table 1 and Figure 2A show the results of the differential analysis of the GSE83148 data set, including 263 DEGs, 83 down-regulated genes, and 180 up-regulated genes. GSE121248 contains 37 chronic hepatitis B-induced HCC adjacent normal tissues and 70 human chronic hepatitis B-induced HCC liver tissues. Supplementary Table 2 and Figure 2B show the results of the differential analysis of the GSE121248 data set, including 798 DEGs, 559 down-regulated genes, and 239 up-regulated genes.

FIGURE 2
www.frontiersin.org

Figure 2. (A) Volcano map of differential genes in the GSE83148 data set. (B) Volcano map of differential genes in the GSE121248 data set. Blue indicates down-regulated genes, red indicates up-regulated genes.

Enrichment Analysis of Two Groups of DEGs

The two groups of DEGs in GSE83148 and GSE121248 were analyzed by GO and KEGG enrichment, respectively (Figures 3A–D). The DEGs in the GSE83148 data set are enriched in different functional entries. In GO analysis, DEGs are mainly enriched in the entry of leukocyte migration in the biological process (BP), mainly in the side of membrane in terms of cell component (CC), and mainly in glycosaminoglycan binding in terms of molecular function (MF). According to KEGG pathway enrichment analysis, the DEGs are mainly enriched in the cytokine–cytokine receptor interaction pathway, cell cycle pathway, hepatitis B pathway, oocyte meiosis pathway, viral carcinogenesis pathway, etc. Then, in the DEG enrichment analysis of GSE121248, DEGs were mainly enriched in the organic acid catabolic process, extracellular matrix, and cofactor binding in BP, CC, and MF. In addition, in the KEGG pathway, DEGs are mainly enriched in chemical carcinogenesis pathway, cell cycle pathway, etc. The results of KEGG pathway enrichment suggested that there were two identical pathways in the two data sets, including cell cycle pathway and P53 signaling pathway.

FIGURE 3
www.frontiersin.org

Figure 3. Functional enrichment analysis of the two groups of DEGs. (A) GO analysis of DEGs in the GSE83148 data set. (B) KEGG analysis of DEGs in the GSE83148 data set. (C) GO analysis of DEGs in the GSE121248 data set. (D) KEGG analysis of DEGs in the GSE121248 data set.

Overlapping DEGs

In the mRNA sequencing data in TCGA, 60 HBV-positive HCC and 18 HBV-related adjacent tissue RNA-seq reading count data were screened. The clinical characteristics of all patients are shown in Supplementary Table 3. The results of the DEG analysis of TCGA are listed in Supplementary Table 4. By sequencing TCGA HBV-related HCC, 1,641 DEGs were obtained, including 1,104 up-regulated genes and 537 down-regulated genes. The DEGs in the above two HBV-related liver disease gene chip data sets and the genes identified as differentially expressed in the TCGA HBV-related HCC sequencing data set are taken to intersect to screen for common overlapping DEGs. The common overlapping DEGs were screened. Figure 4 shows that a total of 22 overlapping DEGs were obtained, including 17 overlapping up-regulated DEGs (Figure 4A) and 5 overlapping down-regulated DEGs (Figure 4B). They may be the DEGs during the progression of HBV infection to HBV-related hepatocellular carcinoma. The visual analysis of 22 overlapping DEGs was performed by TBtools (Figure 4C). The green band in the figure represents the normal group samples, and the red band represents HBV-related HCC samples. The gradual change of color from blue to red represents the process of gene down-regulation to up-regulation.

FIGURE 4
www.frontiersin.org

Figure 4. Identification of genes common to chronic HBV infection and hepatocellular carcinoma. (A) The Venn diagram of the up-regulated DEGs between the two GEO data sets and the TCGA HBV-HCC data set (drawn by SangerBox, http://sangerbox.com/Signin). (B) The Venn diagram of the down-regulated DEGs between the two GEO data sets and the TCGA HBV-HCC data set (drawn by SangerBox, http://sangerbox.com/Signin). (C) The heat map of 5 down-regulated DEGs and 17 up-regulated DEGs in the integrated microarray analysis. The green band in the figure represents the normal group samples, and the red band represents HBV-related HCC samples. The gradual color ranging from blue to red represents the changing process from down-regulation to up-regulation.

Functional Annotation of Overlapping DEGs by GO and KEGG Pathway Analyses

Through the GO and KEGG pathway analysis, 22 overlapping DEGs were functionally annotated to clarify their potential biological functions. GO analysis of overlapping DEGs induced by HBV was enriched in items with significant differences (Figure 5A and Supplementary Table 5). These three entries are cell division, mitotic sister chromatid segregation, and nucleus. To further analyze the pathogenic mechanism of HBV, KEGG pathway analysis was performed on the identified overlapping DEGs (Figure 5B and Supplementary Table 6). The results showed that the overlapping DEGs were mainly enriched on the oocyte meiosis pathway and cell cycle pathway (Figure 5C).

FIGURE 5
www.frontiersin.org

Figure 5. Functional enrichment analysis of the overlapping DEGs (genes common to chronic HBV infection and hepatocellular carcinoma). (A) GO enrichment analysis of the overlapping DEGs. Red indicates the up-regulated gene, and blue indicates the down-regulated gene. The thicker the red circle in the middle of the graph, the more significant the difference, and the darker the red, the greater the proportion of up-regulated genes in the entry. (B) GO enrichment analysis of the overlapping DEGs. The right side of the outermost circle is the term, and the color corresponding to the gene on the left is the gene expression multiple. The inner circle on the left indicates the significance p of the gene corresponding pathway. (C) The key targets and key biological processes involved in hepatitis B-related inflammation and cancer transformation.

Key Gene Analysis

PPI Network and Module Analysis

A PPI network was constructed in the STRING 11.0 database, including 56 nodes and 869 interactions. As shown in Figure 6A, the size of the node is proportional to the degree value. Red nodes indicate up-regulated genes, blue nodes indicate down-regulated genes, and green nodes indicate secondary proteins obtained by protein interaction. The protein interaction results show that the nodes with interactions are mainly up-regulated genes. The top 5 genes with the highest degree are considered key genes, and they are all up-regulated genes. In addition, this study uses MCODE plug-in in Cytoscape to analyze the PPI network module and obtains important cluster modules. A total of three modules were obtained (Figure 6B–D). Module 1 has the highest score of 29.722. In addition, five key genes are concentrated in module 1, which also indicates that it may be the main functional module. The score of module 2 is 6.222, and the score of module 3 is 3. The five key genes include cyclin-dependent kinase 1 (CDK1), mitotic spindle assembly checkpoint protein MAD2A (MAD2L1), Cyclin-A2 (CCNA2), Securin (PTTG1), and serine/threonine-protein kinase Nek2 (NEK2). They are defined as the main hub nodes in the PPI network; the change in gene expression is shown in Figure 6E.

FIGURE 6
www.frontiersin.org

Figure 6. (A) The PPI network of overlapping DEGs (genes common to chronic HBV infection and hepatocellular carcinoma). (B) Module 1 (MCODE score = 29.722). (C) Module 2 (MCODE score = 6.222). (D) Module 3 (MCODE score = 3). Blue circles represent down-regulated genes, red circles represent up-regulated genes, and green circles indicate secondary proteins obtained by protein interaction. (E) Expression of the five key DEGs in HBV-related HCC and normal tissues (TCGA data set). (F) Correlation analysis of five key genes. From left to right, from top to bottom: CDK1-MAD2L1, CDK1-CCNA2, CDK1-PTTG1, CDK1-NEK2, PTTG1-NEK2, MAD2L2-CCNA2, MAD2L1-PTTG1, MAD2L1-NEK2, CCNA2-PTTG1, CCNA2-NEK2.

Correlation Analysis of Key Gene Expression Levels

We applied GEPIA to capture the correlation of expression levels between key genes. Correlation analysis was conducted on any two genes of CDK1, MAD2L1, CCNA2, PTTG1, and NEK2 and five key genes (Figure 6F). The results showed that the significance between any two genes was p < 0.01, indicating that the correlation coefficient was statistically different. The larger the correlation coefficient “R” is, the better the correlation between the two genes is. The four results (CCNA2-CDK1, CCNA2-MAD2L1, PTTG1-CCNA2, CCNA2-NEK2) are relatively weakly correlated (R < 0.5), but p is still extremely low. The above results indicate that the up-regulation of one of them will affect the high expression of other genes. This may indicate that they are all regulated by the same transcription factors and epigenetic modifications.

Survival Analysis

Cox Regression Analysis

The univariate Cox proportional hazards regression model was used to analyze 22 overlapping DEGs, and nine genes that were significantly related to survival time were identified (p < 0.05). After using the multivariate Cox proportional hazards regression model, a prognostic gene signature consisting of nine genes was developed, including Securin (PTTG1), mitotic spindle assembly checkpoint protein MAD2A (MAD2L1), PCNA-associated factor (PCLAF), ribonucleoside-diphosphate reductase subunit M2 (RRM2), targeting protein for Xklp2 (TPX2), cyclin-dependent kinase 1 (CDK1), serine/threonine-protein kinase Nek2 (NEK2), DEP domain-containing protein 1A (DEPDC1), and ZW10 interactor (ZWINT) (Supplementary Table 7). Figure 7A shows the forest plot of Cox regression. Using the survminer software package to multiply gene expression by the linear combination regression coefficient obtained through multiple Cox regression, the optimal cutoff threshold can be calculated, and more suitable high-risk groups and low-risk groups can be obtained. The risk scores of the patients are ranked, and then the survival status of the patients is displayed through a dot graph, and the expression of nine prognostic genes is displayed through a heat map. Figure 7B shows the patients’ risk scores in order from low to high: red indicates the high-risk group, and blue indicates the low-risk group. Different patients have different survival times: blue indicates survival during follow-up, and red indicates death during follow-up. The heat map of the nine prognostic genes shows that as the risk value increases, the survival time of patients tends to be shortened, the proportion of deaths tends to increase, and the nine prognostic genes tend to be highly expressed. The K-M curve in Figure 7C shows the relationship between patient survival time and survival probability (p < 0.0001, statistically significant). The red and blue solid lines represent the changes in survival rates of the high-risk group and the low-risk group, and the dotted lines represent the 95 and 5% confidence intervals. Figure 7D is the ROC curve of the patient in the training set. The results showed that the AUC of 1-, 2-, 3-, 4-, and 5-years OS were 0.86, 0.82, 0.83, 0.83, and 0.74, respectively, so the prognostic gene characteristics showed good performance in survival prediction.

FIGURE 7
www.frontiersin.org

Figure 7. Prognostic analysis of the nine-gene signature model in the TCGA cohort. (A) Training set forest diagram. (B) Risk score in the TCGA cohort. (C) Kaplan–Meier curve of OS for patients in the high-risk group and low-risk group. (D) The AUC of the time-dependent ROC curve in the TCGA cohort. Validation of the eight-gene signature in the ICGC cohort. (E) Test set forest diagram. (F) Risk score in the ICGC cohort. (G) Kaplan–Meier curve of OS for patients in high-risk group and low-risk group. (H) The AUC of the time-dependent ROC curve in the ICGC cohort.

Cox Model Verification

The 260 primary solid tissue tumor samples can be screened from the ICGC portal website12 (Supplementary Table 8). Samples with complete expression profile data and clinical information were selected, and RNA-seq data and clinical information of 231 tumor samples were obtained. These samples were mainly from Japanese people with hepatocellular carcinoma, and the FPKM values from genes were used. Sixty patients in TCGA were taken as the training set, and 231 patients in ICGC were taken as the test set for modeling. Because the PCLAF gene was not found in the test set, the remaining eight genes were thus used for fitting the model in the test set. The coefficients of the training set were extracted, and the expression of eight genes in the test set was multiplied to verify the model. The survminer package was used to recalculate the optimal cutoff threshold to obtain a more suitable high- and low-risk group. The risk score can be calculated by multiplying the gene expression by the linear combination regression coefficient obtained through multiple Cox regression. Figure 7E shows the forest plot of Cox regression. The survminer software package was used to recalculate the optimal cutoff threshold to obtain more suitable high-risk and low-risk populations. Figure 7F shows that the cutoff is 0.32, and the patients’ risk scores are sorted from low to high: red represents the high-risk group, and blue represents the low-risk group. Different patients have different survival times: blue indicates survival during follow-up, and red indicates death during follow-up. The heat map of the eight prognostic genes shows that as the risk value increases, the survival time of patients tends to be shortened, the proportion of deaths tends to increase, and the eight prognostic genes tend to be highly expressed. The K-M curve in Figure 7G shows the relationship between patient survival time and survival probability (p = 0.00042, statistically significant). The red and blue solid lines represent the changes in survival rates of the high-risk group and the low-risk group, and the dotted lines represent the 95 and 5% confidence intervals. Figure 7H shows the ROC curve of patients in the training set. The results showed that the AUC of the 2-, 3-, and 4-year OS were 0.73, 0.69, and 0.73, respectively, so the prognostic gene signatures showed good performance in survival prediction. The results show that the area under the ROC curve in the training set and the test set is more than 0.5, and the model is better.

Discussion

The risk factors of chronic hepatitis B disease progression can be divided into three categories: host factor element, virus factor, and liver factor (Wong et al., 2013). Host factors include age, male, family history of liver cancer, obesity, genetic susceptibility, smoking, alcoholism, diabetes, and immune status (Wong and Janssen, 2015). The viral factors include HBsAg positive, HBeAg positive, high level of HBV DNA, HBV genotype, and HBV mutant. Especially, high viral load is an independent and effective predictor (Wong and Sung, 2012). Liver factors include progressive fibrosis and cirrhosis, poor liver function, hepatitis activity, and other accompanying liver diseases, such as hepatitis C virus or coinfection of alcoholic and non-alcoholic fatty liver (Wong and Wong, 2013). Inflammatory reaction of the liver caused by virus replication in patients with chronic HBV infection is the main factor of liver disease progression. Chronic hepatitis B-cirrhosis-hepatocellular carcinoma is a common law of disease development and transformation in clinic, and its disease transformation process can also be regarded as a typical “inflammatory cancer transformation” process. Finding the key genes in the process of inflammatory cancer transformation is helpful to inhibit the progression of chronic hepatitis B and the transformation from chronic inflammation to cancer. The analysis of comprehensive bioinformatics mainly focuses on the screening of DEGs, the construction of related protein interaction networks, the screening and survival analysis of key genes, and the analysis of gene association. The above method has been widely used to identify potential biomarkers related to the diagnosis, treatment, and prognosis of HBV and HCC. Xie et al. (2020) studied the genetic characteristics of HBV positive (HBV +) HCC and revealed its potential carcinogenic mechanism by using the methods of differential gene screening, functional enrichment analysis, protein interaction network construction, survival analysis, immunohistochemistry, and statistical analysis. Sun et al. (2019) characterized the genome size of HBV and HCV-infected HCC by comparing the publicly available data of the Cancer Genome Atlas Project (TCGA), comparing their gene expression patterns, methylation profiles, and copy number variation. Huang et al. (2019) identified common gene disorders between HBV and HCC by screening DEGs. In addition, through modular methods such as PPI networks and hypergeometric tests, targeted drugs with regulatory effects on diseases are predicted.

In this study, we obtained two microarray data sets about hepatitis progression and integrated them. GSE83148 identified 263 DEGs between the normal group and HBV, including 180 up-regulated genes and 83 down-regulated genes; GSE121248 identified 798 DEGs of HBV and HBV-related HCC, including 239 up-regulated genes and 559 down-regulated genes. The above two microarray data sets were integrated with TCGA’s RNA sequencing data to identify 22 DEGs, including 17 down-regulated genes and 5 up-regulated genes. They are considered to be differential genes that jointly affect the occurrence and development of hepatitis B inflammatory cancer transformation. GO analysis of overlapping differential genes induced by HBV showed that the differential genes were enriched in cell division, mitotic sister chromatid separation, and nuclear entries. The results of KEGG pathway enrichment analysis showed that overlapping differential genes were mainly enriched in meiosis pathway and cell cycle pathway of oocytes.

In addition, we also identified five key genes in the PPI network and module analysis, namely, CDK1, MAD2L1, CCNA2, PTTG1, and NEK2. Coincidentally, they are all up-regulated genes in HBV disease changes. The CDK family is a Ser/Thr kinase system that corresponds to the cell cycle progression. Various CDKs are alternately activated along the cell cycle phase, phosphorylating the corresponding substrate. Through synergy with cyclin, cell cycle events proceed in an orderly manner. The activity of CDK1 is closely related to the content of CyclinB. CyclinB is generally synthesized in the late G1 phase. Through the S phase and G2 phase, the CyclinB content reached a certain level, entered the nucleus, and bound to CDK1. Then, CDK1 kinase activity began to appear (Yang et al., 2011; Hu et al., 2018). Activated CDK1 can phosphorylate target proteins to produce corresponding physiological effects, such as phosphorylation of nuclear laminin leading to the disintegration of nuclear fibrils, disappearance of nuclear membrane, and phosphorylation of histone H1, leading to the condensation of chromosomes. The final result of these effects is to keep the cell cycle running. Li Y. et al. (2019) also found that CDK1 is an important biomarker in the study of lncRNA-related comprehensive analysis of the ceRNA network to reveal potential biomarkers for the prognosis of HBV-related HCC. They verified the up-regulation of CDK1 in liver cancer in the microarray data set and TCGA database (Li Y. et al., 2019). In addition, some researchers believe that HBx is different from other viral proteins. HBx can continuously activate the cyclin B1-CDK1 kinase. However, the results of some researchers indicate that HBx induces G2/M arrest and apoptosis, which in turn inhibits the growth of HCC cells and vascular endothelial cells in vitro and in vivo. Another part of the researchers believed that HBx accelerates the appearance of cells entering the S phase from Go/Gland by promoting the rapid and strong activation of CDK kinase activity. HBx may promote viral carcinogenesis through molecular mechanisms (Benn and Schneider, 1995; Cheng et al., 2009; Kim et al., 2015). The contradiction between them may be due to differences of experimental environment and experimental materials, or the different expression levels of HBx. High HBx expression leads to cell cycle arrest and apoptosis, while low HBx expression indicates adverse effects (Bréchot et al., 2000). The results of the study reflect that the persistent chronic expression of HBX may be an important factor in the final progression of HBV to HCC. Mitotic arrest defect protein 2 (MAD2), also known as mitotic spindle assembly checkpoint protein, is encoded by the MAD2L1 gene (Chen Z. et al., 2019). Moreover, it has been reported that MAD2 and CDC20 form mitotic checkpoint complexes to monitor the attachment process of mitochondria spindle and inhibit the activity of late-stage promoting complexes (Luo et al., 2000). It regulates the mitotic process of cells and then affects the malignant progression of a variety of tumors (Fang et al., 1998; Guo et al., 2020). For example, the work of researchers using integrated bioinformatics analysis has shown that MAD2L1 may be a potential therapeutic target for HCC (Yang et al., 2019). The results of another study showed that MiR-200c-5p inhibited the proliferation, migration, and invasion of HCC cells by down-regulating MAD2L1 (Li et al., 2017). This indicates that the expression of MAD2L1 in HCC is significantly higher and is related to poor prognosis. Cyclin A2 (CCNA2) is a member of the cyclin family. Different cyclins can selectively activate specific substrates and cause different cell cycle events (Manni et al., 2001; Loog and Morgan, 2005; Chen et al., 2018). The researchers believe that the overexpression of CCNA2 is related to the carcinogenesis of the liver. There are many exons in CCNA2, and HBV integration occurs in introns. Because cyclin is important in controlling cell division, disrupting the cyclin A gene through viral insertion may help in tumorigenesis (Wang et al., 1990; Bayard et al., 2018). PTTG1 may work by blocking key proteins. Its gene product has in vitro transformation activity and in vivo tumorigenic activity and is highly expressed in various tumors (Zou et al., 1999). PTTG1 may play a role by blocking the key protein, and its gene product has in vitro transformation activity and in vivo tumorigenicity and is highly expressed in various tumors. Many studies have also proved that PTTG1 may be an important gene for HBV-related hepatitis to progress into hepatocellular carcinoma (Lin et al., 2019; Shen et al., 2019). The results of Li et al. (2013) suggested that the loss of miR-122 expression will lead to the up-regulation of its target PBF, thereby initiating the nuclear translocation of PTTG1 and promoting the transcriptional activity of PTTG1, thereby enhancing cell growth and invasion. With the development of chronic hepatitis B to cirrhosis and HCC, PTTG1 expression increased. In vitro experiments showed that HBx induced significant accumulation of PTTG1 protein without affecting the level of its mRNA. This may provide new insights for the pathogenesis of HBV-related inflammatory cancer transformation (Molina-Jiménez et al., 2010). NEK2 is involved in the control of centrosome separation and bipolar spindle formation in mitotic cells and chromatin condensation in meiotic cells (Hames and Fry, 2002). Researchers such as Xie identified important genes and pathways related to HBV-related HCC through bioinformatics analysis and found that NEK2 is a key gene in the protein interaction network. This is very similar to our results (Xie et al., 2019). Cheng et al. (2018) found in a cohort study that a high expression of NEK2 was an independent risk factor for decreased OS. The results of the study suggest that a high expression of NEK2 is a risk factor for poor survival of liver cancer patients (Cheng et al., 2018; Ren et al., 2018).

The current study identified nine key genes for prognosis of HBV-related liver disease changes and constructed a prognostic gene marker composed of these genes. It is worth noting that these nine genes are all identified as dangerous prognostic genes. Among them, PTTG1, MAD2L1, CDK1, and NEK2 are also the key genes obtained from the protein interaction network. They may be the key risk prognostic genes for hepatitis B inflammation and cancer transformation and play a key role in the progression of hepatitis B to hepatitis B-related HCC. In addition, DEPDC1, ZWINT, PCLAF, RRM2, and TPX2 have also been identified as dangerous prognostic genes. DEPDC1 overexpression promotes HCC cell proliferation, colony formation, and invasion (Guo et al., 2019). Studies have shown that high DEPDC1 expression is an independent predictor of cancer-related death and recurrence. The high expression of DEPDC1 in non-tumor liver is an independent risk factor for late relapse (Amisaki et al., 2019). ZWINT is part of the MIS12 complex, which is necessary for mitochondrial formation and spindle checkpoint activity (Musio et al., 2004). The dysregulation of ZWINT enhanced the chromosomal instability in tumorigenesis and contributed to poor prognosis in malignancies (Pérez de Castro et al., 2007). PCLAF acts as a PCNA-binding protein for DNA repair regulators during DNA replication (Kais et al., 2011). Studies have confirmed that overexpression of PCLAF in adrenal cortical tumors, nasopharyngeal carcinoma, and hepatocellular carcinoma may promote the growth and invasion of cancer cells (Jain et al., 2011; Abdelgawad et al., 2016; Ma et al., 2020). RRM2 is a key protein for DNA synthesis and repair, which can promote cell proliferation and inhibit apoptosis. In previous studies, it has been demonstrated that inhibition of RRM2 significantly inhibits the proliferation of liver cancer cells (Wang et al., 2018). TPX2 is a microtubule-associated protein that involves targeting the kinesin Xklp2 to microtubules. The expression of TPX2 in tumor tissues is higher than that in non-tumor tissues. Overexpression of TPX2 is positively correlated with poor prognosis (Liang et al., 2015).

KEGG pathway enrichment shows that key genes are mainly enriched in oocyte meiosis pathway and cell cycle pathway. Interestingly, in GSE83148, the result of pathway enrichment of differential gene KEGG also included oocyte meiosis pathway and cell cycle pathway; in GSE121248, the result of KEGG included the cell cycle pathway. The pathway enrichment results of 22 overlapping differential genes are also very similar to the pathway enrichment results of the two chips. An epidemiological and virological study of occult hepatitis B infection and hepatocellular carcinoma found that HBV DNA integration affects the liver cell cycle and tumor development, and the promotion of cancer-promoting proteins (such as HBx proteins and mutated surface proteins) produces and continues low-grade hepatic necrotizing inflammation. Inflammation can lead to liver fibrosis and cirrhosis, which is the pathogenic mechanism of occult hepatitis B infection-related hepatocellular carcinoma (Li Y. et al., 2019; Mak et al., 2020). In addition, studies have shown that the HBx gene can be expressed at the one-cell and two-cell stages of embryonic development. The data shows that sperm may be used as a carrier for the vertical transmission of HBV DNA to the next generation (Ali et al., 2005).

Conclusion

In general, through biological information research methods, we have identified five key genes and nine dangerous prognostic genes. Among them, PTTG1, MAD2L1, CDK1, and NEK2 may be the key prognostic genes of the hepatitis B inflammation and cancer transformation. However, since our research is based on data analysis, further experiments are needed to confirm. At the same time, we hope that our research results have a certain guiding significance for the prognosis and treatment of liver disease in hepatitis B progression.

Data Availability Statement

Publicly available datasets were analyzed in this study. This data can be found here: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE83148 and https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE121248.

Author Contributions

JZ and XKL contributed to the conception and design of the study. WZ and SL organized the database. CW performed the statistical analysis. JZ and ZW wrote the first draft of the manuscript. RL, XJL, and JW supervised the project and acquired the funding. YL, SG, and SJ wrote sections of the manuscript. XZ and MW critically reviewed the manuscript for important intellectual content. All the authors contributed to the manuscript revision and read and approved the submitted version.

Funding

This research was funded by the National Nature Science Foundation of China (grant no. 81673829) and the Young Scientists Training Program of Beijing University of Chinese Medicine.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fgene.2021.654517/full#supplementary-material

Supplementary Table 1 | The results of the differential analysis of the GSE83148 data set.

Supplementary Table 2 | The results of the differential analysis of the GSE121248 data set.

Supplementary Table 3 | The clinical characteristics of all patients.

Supplementary Table 4 | The results of DEGs analysis of TCGA.

Supplementary Table 5 | GO analysis of overlapping DEGs induced by HBV.

Supplementary Table 6 | KEGG analysis of overlapping DEGs induced by HBV.

Supplementary Table 7 | Prognostic value of the nine genes in the HBV-related HCC patients of the TCGA cohort.

Supplementary Table 8 | Sample data of primary solid tissue tumors in ICGC.

Abbreviations

BP, biological process; CC, cell component; CCNA2, Cyclin-A2; CDK1, Cyclin-dependent kinase 1; DEGs, differentially expressed genes; DEPDC1, DEP domain-containing protein 1A; FDR, false discovery rate; GEO, Gene Expression Omnibus database; GEPIA, Gene Expression Profiling Interactive Analysis; GO, Gene Ontology; HBV, hepatitis B virus; HCC, Hepatocellular Carcinoma; KEGG, Kyoto Encyclopedia of Genes and Genomes; MAD2, Mitotic arrest defect protein 2; MAD2L1, Mitotic spindle assembly checkpoint protein MAD2A; MCODE, Molecular Complex Detection; MF, molecular function; NEK2, Serine/threonine-protein kinase Nek2; NEK2, Serine/threonine-protein kinase Nek2; OS, overall survival; PCLAF, PCNA-associated factor; PPI, protein-protein interaction; PTTG1, Securin; RRM2, Ribonucleoside-diphosphate reductase subunit M2; TCGA, the Cancer Genome Atlas Projec; TPX2, Targeting protein for Xklp2; ZWINT, ZW10 interactor.

Footnotes

  1. ^ 1http://www.ncbi.nlm.nih.gov/geo/
  2. ^ 2https://cran.r-project.org/doc/FAQ/R-FAQ.html#Citing-R
  3. ^ 3http://sangerbox.com/TcgaDown
  4. ^ 4http://sangerbox.com/AllTools?tool_id=9699507
  5. ^ 5https://david.ncifcrf.gov/
  6. ^ 6http://sangerbox.com/AllTools?tool_id=9698327
  7. ^ 7https://string-db.org/
  8. ^ 8http://www.cytoscape.org/
  9. ^ 9http://sangerbox.com/AllTools?tool_id=9698305
  10. ^ 10http://gepia.cancer-pku.cn/index.html
  11. ^ 11https://icgc.org
  12. ^ 12https://dcc.icgc.org/projects/LIRI-JP

References

Abdelgawad, I. A., Radwan, N. H., and Hassanein, H. R. (2016). KIAA0101 mRNA expression in the peripheral blood of Hepatocellular carcinoma patients: association with some clinicopathological features. Clin. Biochem. 49, 787–791. doi: 10.1016/j.clinbiochem.2015.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Ali, B. A., Huang, T. H., and Xie, Q. D. (2005). Detection and expression of hepatitis B virus X gene in one and two-cell embryos from golden hamster oocytes in vitro fertilized with human spermatozoa carrying HBV DNA. Mol. Reprod. Dev. 70, 30–36. doi: 10.1002/mrd.20185

PubMed Abstract | CrossRef Full Text | Google Scholar

Amisaki, M., Yagyu, T., Uchinaka, E. I., Morimoto, M., Hanaki, T., Watanabe, J., et al. (2019). Prognostic value of DEPDC1 expression in tumor and non-tumor tissue of patients with Hepatocellular carcinoma. Anticancer Res. 39, 4423–4430. doi: 10.21873/anticanres.13614

PubMed Abstract | CrossRef Full Text | Google Scholar

Bayard, Q., Meunier, L., Peneau, C., Renault, V., Shinde, J., Nault, J. C., et al. (2018). Cyclin A2/E1 activation defines a Hepatocellular carcinoma subclass with a rearrangement signature of replication stress. Nat. Commun. 9:5235.

Google Scholar

Benn, J., and Schneider, R. J. (1995). Hepatitis B virus HBx protein deregulates cell cycle checkpoint controls. Proc. Natl. Acad. Sci. U. S. A. 92, 11215–11219. doi: 10.1073/pnas.92.24.11215

PubMed Abstract | CrossRef Full Text | Google Scholar

Bréchot, C., Gozuacik, D., Murakami, Y., and Paterlini-Bréchot, P. (2000). Molecular bases for the development of hepatitis B virus (HBV)-related Hepatocellular carcinoma (HCC). Semin. Cancer Biol. 10, 211–231. doi: 10.1006/scbi.2000.0321

PubMed Abstract | CrossRef Full Text | Google Scholar

Bustoros, M., Sklavenitis-Pistofidis, R., Park, J., Redd, R., Zhitomirsky, B., Dunford, A. J., et al. (2020). Genomic profiling of smoldering multiple myeloma identifies patients at a high risk of disease progression. J. Clin. Oncol. 38, 2380–2389.

Google Scholar

Chan, S. L., Wong, V. W., Qin, S., and Chan, H. L. (2016). Infection and cancer: the case of Hepatitis B. J. Clin. Oncol. 34, 83–90.

Google Scholar

Chen, C., Chen, H., Zhang, Y., Thomas, H. R., Frank, M. H., He, Y., et al. (2020). TBtools: an integrative toolkit developed for interactive analyses of big biological data. Mol. Plant 13, 1194–1202. doi: 10.1016/j.molp.2020.06.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Q. F., Xia, J. G., Li, W., Shen, L. J., Huang, T., and Wu, P. (2018). Examining the key genes and pathways in Hepatocellular carcinoma development from hepatitis B virus-positive cirrhosis. Mol. Med. Rep. 18, 4940–4950.

Google Scholar

Chen, S., Gao, C., Wu, Y., and Huang, Z. (2020). Identification of prognostic miRNA signature and lymph node metastasis-related key genes in cervical cancer. Front. Pharmacol. 11:544.

Google Scholar

Chen, Y. Y., Lin, Y., Han, P. Y., Jiang, S., Che, L., He, C. Y., et al. (2019). HBx combined with AFB1 triggers hepatic steatosis via COX-2-mediated necrosome formation and mitochondrial dynamics disorder. J. Cell Mol. Med. 23, 5920–5933. doi: 10.1111/jcmm.14388

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Chen, J., Huang, X., Wu, Y., Huang, K., Xu, W., et al. (2019). Identification of potential key genes for Hepatitis B virus-associated hepatocellular carcinoma by bioinformatics analysis. J. Comput. Biol. 26, 485–494. doi: 10.1089/cmb.2018.0244

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, P., Li, Y., Yang, L., Wen, Y., Shi, W., Mao, Y., et al. (2009). Hepatitis B virus X protein (HBx) induces G2/M arrest and apoptosis through sustained activation of cyclin B1-CDK1 kinase. Oncol. Rep. 22, 1101–1107.

Google Scholar

Cheng, Y., Chen, X., Ye, L., Zhang, Y., Liang, J., Liu, W., et al. (2018). The prognostic significance of NEK2 in Hepatocellular carcinoma: evidence from a meta-analysis and retrospective cohort study. Cell. Physiol. Biochem. 51, 2746–2759. doi: 10.1159/000495966

PubMed Abstract | CrossRef Full Text | Google Scholar

Clough, E., and Barrett, T. (2016). The gene expression omnibus database. Methods Mol. Biol. 1418, 93–110. doi: 10.1007/978-1-4939-3578-9_5

CrossRef Full Text | Google Scholar

Colotta, F., Allavena, P., Sica, A., Garlanda, C., and Mantovani, A. (2009). Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30, 1073–1081. doi: 10.1093/carcin/bgp127

PubMed Abstract | CrossRef Full Text | Google Scholar

Cox, D. R. (1972). Regression models and life-tables. J. R. Stat. Soc. Ser. B 34, 187–202.

Google Scholar

Dennis, G., Sherman, B. T., Hosack, D. A., Yang, J., Gao, W., Lane, H. C., et al. (2003). DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 4:3.

Google Scholar

Fang, G., Yu, H., and Kirschner, M. W. (1998). The checkpoint protein MAD2 and the mitotic regulator CDC20 form a ternary complex with the anaphase-promoting complex to control anaphase initiation. Genes Dev. 12, 1871–1883. doi: 10.1101/gad.12.12.1871

PubMed Abstract | CrossRef Full Text | Google Scholar

Fanning, G. C., Zoulim, F., Hou, J., and Bertoletti, A. (2019). Therapeutic strategies for hepatitis B virus infection: towards a cure. Nat. Rev. Drug Discov. 18, 827–844. doi: 10.1038/s41573-019-0037-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Franz, M., Lopes, C. T., Huck, G., Dong, Y., Sumer, O., and Bader, G. D. (2016). Cytoscape.js: a graph theory library for visualisation and analysis. Bioinformatics 32, 309–311.

Google Scholar

George, B., Seals, S., and Aban, I. (2014). Survival analysis and regression models. J. Nuclear Cardiol. 21, 686–694.

Google Scholar

Guo, W., Li, H., Liu, H., Ma, X., Yang, S., and Wang, Z. (2019). DEPDC1 drives hepatocellular carcinoma cell proliferation, invasion and angiogenesis by regulating the CCL20/CCR6 signaling pathway. Oncol. Rep. 42, 1075–1089.

Google Scholar

Guo, Y., Huang, P., Ning, W., Zhang, H., and Yu, C. (2020). Identification of core genes and pathways in medulloblastoma by integrated bioinformatics analysis. J. Mol. Neurosci. 70, 1702–1712. doi: 10.1007/s12031-020-01556-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Hames, R. S., and Fry, A. M. (2002). Alternative splice variants of the human centrosome kinase Nek2 exhibit distinct patterns of expression in mitosis. Biochem. J. 361, 77–85. doi: 10.1042/0264-6021:3610077

CrossRef Full Text | Google Scholar

He, L., Chen, J., Xu, F., Li, J., and Li, J. (2020). Prognostic implication of a metabolism-associated gene signature in lung adenocarcinoma. Mol. Ther. Oncolytics 19, 265–277. doi: 10.1016/j.omto.2020.09.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Heagerty, P. J., and Zheng, Y. (2005). Survival model predictive accuracy and ROC curves. Biometrics 61, 92–105. doi: 10.1111/j.0006-341x.2005.030814.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, J., Qiao, M., Chen, Y., Tang, H., Zhang, W., Tang, D., et al. (2018). Cyclin E2-CDK2 mediates SAMHD1 phosphorylation to abrogate its restriction of HBV replication in hepatoma cells. FEBS Lett. 592, 1893–1904. doi: 10.1002/1873-3468.13105

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, da, W., Sherman, B. T., and Lempicki, R. A. (2009). Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57. doi: 10.1038/nprot.2008.211

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, R., Liao, X., and Li, Q. (2017). Identification and validation of potential prognostic gene biomarkers for predicting survival in patients with acute myeloid leukemia. Onco Targets Ther. 10, 5243–5254. doi: 10.2147/ott.s147717

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, X. B., He, Y. G., Zheng, L., Feng, H., Li, Y. M., Li, H. Y., et al. (2019). Identification of hepatitis B virus and liver cancer bridge molecules based on functional module network. World J. Gastroenterol. 25, 4921–4932. doi: 10.3748/wjg.v25.i33.4921

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Z., Yang, Q., and Huang, Z. (2018). Identification of critical genes and five prognostic biomarkers associated with colorectal cancer. Med. Sci. Monit. 24, 4625–4633. doi: 10.12659/msm.907224

PubMed Abstract | CrossRef Full Text | Google Scholar

Iannacone, M., and Guidotti, L. G. (2021). Immunobiology and pathogenesis of hepatitis B virus infection. Nat. Rev. Immunol. doi: 10.1038/s41577-021-00549-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Jain, M., Zhang, L., Patterson, E. E., and Kebebew, E. (2011). KIAA0101 is overexpressed, and promotes growth and invasion in adrenal cancer. PLoS One 6:e26866. doi: 10.1371/journal.pone.0026866

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, L., Li, C., Liu, T., and Wang, L. (2020). A potential prognostic prediction model of colon adenocarcinoma with recurrence based on prognostic lncRNA signatures. Hum. Genom. 14:24.

Google Scholar

Kais, Z., Barsky, S. H., Mathsyaraja, H., Zha, A., Ransburgh, D. J., He, G., et al. (2011). KIAA0101 interacts with BRCA1 and regulates centrosome number. Mol. Cancer Res. 9, 1091–1099. doi: 10.1158/1541-7786.mcr-10-0503

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanehisa, M., Furumichi, M., Tanabe, M., Sato, Y., and Morishima, K. (2017). KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45, D353–D361.

Google Scholar

Kim, S., Lee, H. S., Ji, J. H., Cho, M. Y., Yoo, Y. S., Park, Y. Y., et al. (2015). Hepatitis B virus X protein activates the ATM-Chk2 pathway and delays cell cycle progression. J. Gen. Virol. 96, 2242–2251. doi: 10.1099/vir.0.000150

PubMed Abstract | CrossRef Full Text | Google Scholar

Kulasingam, V., and Diamandis, E. P. (2008). Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat. Clin. Pract. Oncol. 5, 588–599. doi: 10.1038/ncponc1187

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C. Y., Cai, J. H., Tsai, J. J. P., and Wang, C. C. N. (2020). Identification of hub genes associated with development of head and neck squamous cell carcinoma by integrated bioinformatics analysis. Front. Oncol. 10:681.

Google Scholar

Li, C., Wang, Y., Wang, S., Wu, B., Hao, J., Fan, H., et al. (2013). Hepatitis B virus mRNA-mediated miR-122 inhibition upregulates PTTG1-binding protein, which promotes hepatocellular carcinoma tumor growth and cell invasion. J. Virol. 87, 2193–2205. doi: 10.1128/jvi.02831-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., Zhao, X., Li, C., Sheng, C., and Bai, Z. (2019). Integrated analysis of lncRNA-associated ceRNA network reveals potential biomarkers for the prognosis of hepatitis B virus-related hepatocellular carcinoma. Cancer Manag. Res. 11, 877–897. doi: 10.2147/cmar.s186561

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Li, Z., Zhu, H., and Yu, X. (2020). Autophagy regulatory genes MET and RIPK2 play a prognostic role in pancreatic ductal adenocarcinoma: a bioinformatic analysis based on GEO and TCGA. BioMed. Res. Int. 2020:8537381.

Google Scholar

Li, Y., Bai, W., and Zhang, J. (2017). MiR-200c-5p suppresses proliferation and metastasis of human hepatocellular carcinoma (HCC) via suppressing MAD2L1. Biomed. Pharmacother. 92, 1038–1044. doi: 10.1016/j.biopha.2017.05.092

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Fu, Y., Hu, X., Sun, L., Tang, D., Li, N., et al. (2019). The HBx-CTTN interaction promotes cell proliferation and migration of hepatocellular carcinoma via CREB1. Cell Death Dis. 10:405.

Google Scholar

Li, Y., Liu, X., Ma, Y., Wang, Y., Zhou, W., Hao, M., et al. (2018). knnAUC: an open-source R package for detecting nonlinear dependence between one continuous variable and one binary variable. BMC Bioinformatics 19:448.

Google Scholar

Liang, B., Jia, C., Huang, Y., He, H., Li, J., Liao, H., et al. (2015). TPX2 level correlates with hepatocellular carcinoma cell proliferation, apoptosis, and EMT. Dig. Dis. Sci. 60, 2360–2372. doi: 10.1007/s10620-015-3730-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, J. Y., Wang, D. S., Lin, H. C., Chen, X. X., Yang, H., Zheng, Y., et al. (2020). A novel ferroptosis-related gene signature for overall survival prediction in patients with Hepatocellular carcinoma. Int. J. Biol. Sci. 16, 2430–2441. doi: 10.7150/ijbs.45050

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Y., Liang, R., Ye, J., Li, Q., Liu, Z., Gao, X., et al. (2019). A twenty gene-based gene set variation score reflects the pathological progression from cirrhosis to Hepatocellular carcinoma. Aging 11, 11157–11169. doi: 10.18632/aging.102518

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Lin, J., and He, H. (2019). Identification of potential crucial genes associated with the pathogenesis and prognosis of endometrial cancer. Front. Genet. 10:373.

Google Scholar

Liu, X., Wu, J., Zhang, D., Bing, Z., Tian, J., Ni, M., et al. (2018). Identification of potential key genes associated with the pathogenesis and prognosis of gastric cancer based on integrated bioinformatics analysis. Front Genet. 9:265.

Google Scholar

Loog, M., and Morgan, D. O. (2005). Cyclin specificity in the phosphorylation of cyclin-dependent kinase substrates. Nature 434, 104–108. doi: 10.1038/nature03329

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, X., Fang, G., Coldiron, M., Lin, Y., Yu, H., Kirschner, M. W., et al. (2000). Structure of the Mad2 spindle assembly checkpoint protein and its interaction with Cdc20. Nat. Struct. Biol. 7, 224–229.

Google Scholar

Ma, F., Zhi, C., Wang, M., Li, T., Khan, S. A., Ma, Z., et al. (2020). Dysregulated NF-κB signal promotes the hub gene PCLAF expression to facilitate nasopharyngeal carcinoma proliferation and metastasis. Biomed. Pharmacother. 125:109905. doi: 10.1016/j.biopha.2020.109905

PubMed Abstract | CrossRef Full Text | Google Scholar

Mair, B., Aldridge, P. M., Atwal, R. S., Philpott, D., Zhang, M., Masud, S. N., et al. (2019). High-throughput genome-wide phenotypic screening via immunomagnetic cell sorting. Nat. Biomed. Eng. 3, 796–805. doi: 10.1038/s41551-019-0454-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Mak, L. Y., Wong, D. K., Pollicino, T., Raimondo, G., Hollinger, F. B., and Yuen, M. F. (2020). Occult hepatitis B infection and hepatocellular carcinoma: epidemiology, virology, hepatocarcinogenesis and clinical significance. J. Hepatol. 73, 952–964. doi: 10.1016/j.jhep.2020.05.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Manni, I., Mazzaro, G., Gurtner, A., Mantovani, R., Haugwitz, U., Krause, K., et al. (2001). NF-Y mediates the transcriptional inhibition of the cyclin B1, cyclin B2, and cdc25C promoters upon induced G2 arrest. J. Biol. Chem. 276, 5570–5576. doi: 10.1074/jbc.m006052200

PubMed Abstract | CrossRef Full Text | Google Scholar

Molina-Jiménez, F., Benedicto, I., Murata, M., Martín-Vílchez, S., Seki, T., Antonio Pintor-Toro, J., et al. (2010). 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. doi: 10.1002/hep.23468

PubMed Abstract | CrossRef Full Text | Google Scholar

Musa, J., Li, J., and Grünewald, T. G. (2019). Hepatitis B virus large surface protein is priming for hepatocellular carcinoma development via induction of cytokinesis failure. J. Pathol. 247, 6–8. doi: 10.1002/path.5169

PubMed Abstract | CrossRef Full Text | Google Scholar

Musio, A., Mariani, T., Montagna, C., Zambroni, D., Ascoli, C., Ried, T., et al. (2004). Recapitulation of the roberts syndrome cellular phenotype by inhibition of INCENP, ZWINT-1 and ZW10 genes. Gene 331, 33–40. doi: 10.1016/j.gene.2004.01.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Pérez de Castro, I., de Cárcer, G., and Malumbres, M. (2007). A census of mitotic cancer genes: new insights into tumor cell biology and cancer therapy. Carcinogenesis 28, 899–912. doi: 10.1093/carcin/bgm019

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, Q., Li, B., Liu, M., Hu, Z., and Wang, Y. (2018). Prognostic value of NEK2 overexpression in digestive system cancers: a meta-analysis and systematic review. Onco Targets Ther. 11, 7169–7178. doi: 10.2147/ott.s169911

PubMed Abstract | CrossRef Full Text | Google Scholar

Revill, P. A., Tu, T., Netter, H. J., Yuen, L. K. W., Locarnini, S. A., and Littlejohn, M. (2020). The evolution and clinical impact of hepatitis B virus genome diversity. Nat. Rev. Gastroenterol. Hepatol. 17, 618–634. doi: 10.1038/s41575-020-0296-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Rhee, S. Y., Wood, V., Dolinski, K., and Draghici, S. (2008). Use and misuse of the gene ontology annotations. Nat. Rev. Genet. 9, 509–515. doi: 10.1038/nrg2363

PubMed Abstract | CrossRef Full Text | Google Scholar

Ritchie, M. E., Phipson, B., Wu, D., Hu, Y., Law, C. W., Shi, W., et al. (2015). limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e47. doi: 10.1093/nar/gkv007

PubMed Abstract | CrossRef Full Text | Google Scholar

Schweitzer, A., Horn, J., Mikolajczyk, R. T., Krause, G., and Ott, J. J. (2015). Estimations of worldwide prevalence of chronic hepatitis B virus infection: a systematic review of data published between 1965 and 2013. Lancet 386, 1546–1555. doi: 10.1016/s0140-6736(15)61412-x

CrossRef Full Text | Google Scholar

Shen, S., Kong, J., Qiu, Y., Yang, X., Wang, W., and Yan, L. (2019). Identification of core genes and outcomes in hepatocellular carcinoma by bioinformatics analysis. J. Cell. Biochem. 120, 10069–10081. doi: 10.1002/jcb.28290

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, S., Li, Y., Han, S., Jia, H., Li, X., and Li, X. (2019). A comprehensive genome-wide profiling comparison between HBV and HCV infected hepatocellular carcinoma. BMC Med. Genom. 12:147.

Google Scholar

Szklarczyk, D., Morris, J. H., Cook, H., Kuhn, M., Wyder, S., Simonovic, M., et al. (2017). The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45, D362–D368.

Google Scholar

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

Google Scholar

Trépo, C., Chan, H. L. Y., and Lok, A. (2014). Hepatitis B virus infection. Lancet 384, 2053–2063.

Google Scholar

Walter, W., Sánchez-Cabo, F., and Ricote, M. (2015). GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics 31, 2912–2914. doi: 10.1093/bioinformatics/btv300

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Chenivesse, X., Henglein, B., and Bréchot, C. (1990). Hepatitis B virus integration in a cyclin a gene in a hepatocellular carcinoma. Nature 343, 555–557. doi: 10.1038/343555a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S. M., Ooi, L. L., and Hui, K. M. (2007). Identification and validation of a novel gene signature associated with the recurrence of human Hepatocellular carcinoma. Clin. Cancer Res. 13, 6275–6283. doi: 10.1158/1078-0432.ccr-06-2236

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, X., Wang, X., Sun, J., and Fu, S. (2018). An enhanced RRM2 siRNA delivery to rheumatoid arthritis fibroblast-like synoviocytes through a liposome-protamine-DNA-siRNA complex with cell permeable peptides. Int. J. Mol. Med. 42, 2393–2402.

Google Scholar

Wong, G. L., and Wong, V. W. (2013). Risk prediction of hepatitis B virus- related hepatocellular carcinoma in the era of antiviral therapy. World J. Gastroenterol. 19, 6515–6522. doi: 10.3748/wjg.v19.i39.6515

PubMed Abstract | CrossRef Full Text | Google Scholar

Wong, G. L., Chan, H. L., Yiu, K. K., Lai, J. W., Chan, V. K., Cheung, K. K., et al. (2013). Meta-analysis: the association of hepatitis B virus genotypes and Hepatocellular carcinoma. Aliment. Pharmacol. Ther. 37, 517–526.

Google Scholar

Wong, V. W., and Janssen, H. L. (2015). Can we use HCC risk scores to individualize surveillance in chronic hepatitis B infection. J. Hepatol. 63, 722–732. doi: 10.1016/j.jhep.2015.05.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Wong, V. W., and Sung, J. J. (2012). Diagnosis and personalized management of hepatitis B including significance of genotypes. Curr. Opin. Infect. Dis. 25, 570–577. doi: 10.1097/qco.0b013e328357f2f8

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, S., Jiang, X., Zhang, J., Xie, S., Hua, Y., Wang, R., et al. (2019). Identification of significant gene and pathways involved in HBV-related Hepatocellular carcinoma by bioinformatics analysis. PeerJ. 7:e7408. doi: 10.7717/peerj.7408

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, W., Wang, B., Wang, X., Hou, D., Su, H., and Huang, H. (2020). Nine hub genes related to the prognosis of HBV-positive Hepatocellular carcinoma identified by protein interaction analysis. Ann. Transl. Med. 8:478. doi: 10.21037/atm.2020.03.94

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, W., Yu, J., and Wong, V. W. (2017). Mechanism and prediction of HCC development in HBV infection. Best Pract. Res. Clin. Gastroenterol. 31, 291–298. doi: 10.1016/j.bpg.2017.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, H., Gu, J., Zheng, Q., Li, M., Lian, X., Miao, J., et al. (2011). RPB5-mediating protein is required for the proliferation of hepatocellular carcinoma cells. J. Biol. Chem. 286, 11865–11874. doi: 10.1074/jbc.m110.136929

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, W. X., Pan, Y. Y., and You, C. G. (2019). CDK1, CCNB1, CDC20, BUB1, MAD2L1, MCM3, BUB1B, MCM2, and RFC4 may be potential therapeutic targets for Hepatocellular carcinoma using integrated bioinformatic analysis. BioMed Res. Int. 2019:1245072.

Google Scholar

Yu, G., Wang, L. G., Han, Y., and He, Q. Y. (2012). clusterProfiler: an R package for comparing biological themes among gene clusters. Omics J. Integr. Biol. 16, 284–287. doi: 10.1089/omi.2011.0118

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Liu, X., Wu, J., Zhou, W., Tian, J., Guo, S., et al. (2020). A bioinformatics investigation into the pharmacological mechanisms of the effect of the Yinchenhao decoction on hepatitis C based on network pharmacology. BMC Complement. Med. Ther. 20:50.

Google Scholar

Zhou, W., Ma, Y., Zhang, J., Hu, J., Zhang, M., Wang, Y., et al. (2017). Predictive model for inflammation grades of chronic hepatitis B: large-scale analysis of clinical parameters and gene expressions. Liver Int. 37, 1632–1641. doi: 10.1111/liv.13427

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, W., Wu, J., Liu, X., Ni, M., Meng, Z., Liu, S., et al. (2020). Identification of crucial genes correlated with esophageal cancer by integrated high-throughput data analysis. Medicine 99:e20340. doi: 10.1097/md.0000000000020340

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, X., Huang, Z., Xu, L., Zhu, M., Zhang, L., Zhang, H., et al. (2016). A panel of 13-miRNA signature as a potential biomarker for predicting survival in pancreatic cancer. Oncotarget 7, 69616–69624. doi: 10.18632/oncotarget.11903

PubMed Abstract | CrossRef Full Text | Google Scholar

Zou, H., McGarry, T. J., Bernal, T., and Kirschner, M. W. (1999). Identification of a vertebrate sister-chromatid separation inhibitor involved in transformation and tumorigenesis. Science 285, 418–422. doi: 10.1126/science.285.5426.418

PubMed Abstract | CrossRef Full Text | Google Scholar

Zou, J. B., Chai, H. B., Zhang, X. F., Guo, D. Y., Tai, J., and Wang, Y. (2019). Reconstruction of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveal functional lncRNAs in cerebral infarction. Sci. Rep. 9:12176.

Google Scholar

Keywords: hepatitis B, hepatocellular carcinoma, inflammation and cancer transformation, bioinformatics, differentially expressed genes, survival rate, biomarkers

Citation: Zhang J, Liu X, Zhou W, Lu S, Wu C, Wu Z, Liu R, Li X, Wu J, Liu Y, Guo S, Jia S, Zhang X and Wang M (2021) Identification of Key Genes Associated With the Process of Hepatitis B Inflammation and Cancer Transformation by Integrated Bioinformatics Analysis. Front. Genet. 12:654517. doi: 10.3389/fgene.2021.654517

Received: 16 January 2021; Accepted: 21 June 2021;
Published: 01 September 2021.

Edited by:

Josselin Noirel, Conservatoire National des Arts et Métiers (CNAM), France

Reviewed by:

Oksana Sorokina, University of Edinburgh, United Kingdom
Pratip Rana, Engineer Research and Development Center (ERDC), United States

Copyright © 2021 Zhang, Liu, Zhou, Lu, Wu, Wu, Liu, Li, Wu, Liu, Guo, Jia, Zhang and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jiarui Wu, exogamy@163.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.