scholarly journals Carcinogenesis effects of E2F transcription factor 8 (E2F8) in hepatocellular carcinoma outcomes: an integrated bioinformatic report

2020 ◽  
Vol 40 (2) ◽  
Author(s):  
Ying Lü ◽  
Jing Zhang ◽  
Lei Li ◽  
Shun Li ◽  
Zongguo Yang

Abstract This report aimed to investigate the carcinogenesis effects of E2F transcription factor 8 (E2F8) in hepatocellular carcinoma (HCC). E2F8 expression level was compared in Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA) and Oncomine. Survival analysis of E2F8 for HCC were conducted in Kaplan–Meier plotter. Correlations of E2F8 and clinico-pathological features were performed in TCGA. Enrichment of interacted and similar genes with E2F8 was evaluated in Gene Set Enrichment Analysis (GSEA) and Metascape. We found that E2F8 was significantly up-regulated in tumor tissues compared with nontumor tissues (all P < 0.01). Moreover, E2F8 was significantly overexpressed in peripheral blood mononuclear cell (PBMC) in HCC patients than that in healthy individuals (P < 0.001). Meta-analysis in Oncomine database confirmed that E2F8 was significantly higher in HCC tumors (P = 4.28E-08). Additionally, E2F8 elevation significantly correlated with overall survival (OS), recurrence-free survival (RFS), disease-specific survival (DSS) and progression-free survival (PFS) in HCC patients (all P < 0.01). E2F8 level was significantly higher in HCC patients with advanced neoplasm histologic grade, American Joint Committee on Cancer (AJCC) stage and α-fetoprotein (AFP) elevation (all P < 0.05). Cox regression model demonstrated that high E2F8 was an independent risk factor for OS and DFS in HCC patients (HR = 2.16, P = 0.003 and HR = 1.64, P = 0.002, respectively). Enrichment analysis revealed that genes interacted/similar with E2F8 were mainly enriched in cell cycle pathways/biological process. Conclusively, up-regulated in tumors, E2F8 might accelerate tumor progression and result in unfavorable outcomes in HCC patients.

2021 ◽  
Vol 2021 ◽  
pp. 1-8
Author(s):  
Honglan Guo ◽  
Qinqiao Fan

Background. We aimed to investigate the expression of the hyaluronan-mediated motility receptor (HMMR) gene in hepatocellular carcinoma (HCC) and nonneoplastic tissues and to investigate the diagnostic and prognostic value of HMMR. Method. With the reuse of the publicly available The Cancer Genome Atlas (TCGA) data, 374 HCC patients and 50 nonneoplastic tissues were used to investigate the diagnostic and prognostic values of HMMR genes by receiver operating characteristic (ROC) curve analysis and survival analysis. All patients were divided into low- and high-expression groups based on the median value of HMMR expression level. Univariate and multivariate Cox regression analysis were used to identify prognostic factors. Gene set enrichment analysis (GSEA) was performed to explore the potential mechanism of the HMMR genes involved in HCC. The diagnostic and prognostic values were further validated in an external cohort from the International Cancer Genome Consortium (ICGC). Results. HMMR mRNA expression was significantly elevated in HCC tissues compared with that in normal tissues from both TCGA and the ICGC cohorts (all P values <0.001). Increased HMMR expression was significantly associated with histologic grade, pathological stage, and survival status (all P values <0.05). The area under the ROC curve for HMMR expression in HCC and normal tissues was 0.969 (95% CI: 0.948–0.983) in the TCGA cohort and 0.956 (95% CI: 0.932–0.973) in the ICGC cohort. Patients with high HMMR expression had a poor prognosis than patients with low expression group in both cohorts (all P < 0.001 ). Univariate and multivariate analysis also showed that HMMR is an independent predictor factor associated with overall survival in both cohorts (all P values <0.001). GSEA showed that genes upregulated in the high-HMMR HCC subgroup were mainly significantly enriched in the cell cycle pathway, pathways in cancer, and P53 signaling pathway. Conclusion. HMMR is expressed at high levels in HCC. HMMR overexpression may be an unfavorable prognostic factor for HCC.


2021 ◽  
Vol 11 ◽  
Author(s):  
Yuan Nie ◽  
Mei-chun Jiang ◽  
Cong Liu ◽  
Qi Liu ◽  
Xuan Zhu

BackgroundsTumor microenvironment (TME) plays a crucial role in the initiation and progression of Hepatocellular Carcinoma (HCC), especially immune infiltrates. However, there is still a challenge in understanding the modulation of the immune and stromal components in TME, especially TME related genes.MethodsThe proportion of tumor-infiltrating immune cells (TICs) and the immune and stromal scores in 374 HCC patients from The Cancer Genome Atlas (TCGA) database were determined using CIBERSORT and ESTIMATE computational methods. The final screened genes were confirmed by the PPI network and univariate Cox regression of the differentially expressed genes based on different immune or stromal scores. The correlation between the expression levels of the final gene interactions and the clinical characteristics was based on TCGA database and local hospital data. Gene set enrichment analysis (GSEA) and the effect of CXCL5 expression on TICs were conducted.ResultsThere were correlations between the expression of CXCL5 and survival of HCC patients and TMN classification both in TCGA database and local hospital data. The immune-related activities were enriched in the high-expression group; however, the metabolic pathways were enriched in the low-expression group. The result of CIBERSORT analyzing had indicated that CXCL5 expression were correlated with the proportion of NK cells activated, macrophages M0, Mast cells resting, Neutrophils.ConclusionsCXCL5 was a potential prognostic marker for HCC and provides clues regarding immune infiltrates, which offers extra insight for therapeutics of HCC, however, more independent cohorts and functional experiments of CXCL5 are warranted.


2020 ◽  
Vol 2020 ◽  
pp. 1-8
Author(s):  
Yuan Zhang ◽  
Lei Yang ◽  
Jia Shi ◽  
Yunfei Lu ◽  
Xiaorong Chen ◽  
...  

Objective. This study is aimed at investigating the predictive value of CENPA in hepatocellular carcinoma (HCC) development. Methods. Using integrated bioinformatic analysis, we evaluated the CENPA mRNA expression in tumor and adjacent tissues and correlated it with HCC survival and clinicopathological features. A Cox regression hazard model was also performed. Results. CENPA mRNA was significantly upregulated in tumor tissues compared with that in adjacent tissues, which were validated in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) series (all P<0.01). In the Kaplan-Meier plotter platform, the high level of CENPA mRNA was significantly correlated with overall survival (OS), disease-free survival (DFS), recurrence-free survival (RFS), and progression-free survival (PFS) in HCC patients (all log rank P<0.01). For validation in GSE14520 and pan-TCGA dataset, HCC patients with CNEPA mRNA overexpression had poor OS compared with those with low CENPA mRNA (log rank P=0.025 and P<0.0001, respectively), and those with high CENPA had poor DFS in TCGA (log rank P=0.0001). Additionally, CENPA mRNA were upregulated in HCC patients with alpha-fetoprotein (AFP) elevation, advanced TNM stage, larger tumor size, advanced AJCC stage, advanced pathology grade, and vascular invasion (all P<0.05). A Cox regression model including CENPA, OIP5, and AURKB could predict OS in HCC patients effectively (AUC=0.683). Conclusion. Overexpressed in tumors, CENPA might be an oncogenic factor in the development of HCC patients.


2020 ◽  
Author(s):  
Bo Hu ◽  
Xiao-Bo Yang ◽  
Xinting Sang

Abstract Background: Abnormal Nei endonuclease VIII-like 3 (NEIL3)expression is associated with carcinogenesis. Methods: We used sequencing data from the Cancer Genome Atlas database, analyzed NEIL3 expression, gene regulation networks and the correlation with immune infiltrates in hepatocellular carcinoma (HCC). Clinicopathologic characteristics associated with overall survival in TCGA patients using Cox regression and the Kaplan-Meier method. Gene Set Enrichment Analysis was performed using TCGA data set. LinkedOmics was used to identify differential gene expression with NEIL3 and to analyze Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathways. Gene enrichment analysis examined target networks of kinases and transcription factors.Correlations between NEIL3 expression and cancer immune infiltrates and immune gene markers were analyzed by TIMER and GEPIA. Results: We found that overexpressed NEIL3 predicted poor prognosis. Functional network analysis suggested that NEIL3 regulates the DNA replication and cell cycle signaling via pathways involving several cancer-related kinases and E2F Transcription Factor 1.NEIL3 was also found to be associated with the infiltration of several immune cells. Conclusions: Our results demonstrate that data mining efficiently reveals information about NEIL3 expression, potential regulatory networks and the relationship with immune infiltration in HCC, laying a foundation for further study of the role of NEIL3 in carcinogenesis.


2020 ◽  
Author(s):  
Baohui Zhang ◽  
Bufu Tang ◽  
Jianyao Gao ◽  
Jiatong Li ◽  
Lingming Kong ◽  
...  

Abstract Background Hypoxia plays an indispensable role in the development of hepatocellular carcinoma (HCC). However, there are few studies on the application of hypoxia molecules in the prognosis predicting of HCC. We aimed to identify the hypoxia-related genes in HCC and construct reliable models for diagnosis, prognosis and recurrence of HCC patients as well as exploring the potential mechanism.Methods Differentially expressed genes (DEGs) analysis was performed using The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database and four clusters were determined by a consistent clustering analysis. Three DEGs closely related to overall survival(OS)were identified using Cox regression and LASSO analysis and the hypoxia-related signature was developed and validated in TCGA and International Cancer Genome Consortium (ICGC) database. Then the Gene Set Enrichment Analysis (GSEA) was performed to explore signaling pathways regulated by the signature and the CIBERSORT was used for estimating the fractions of immune cell types.Results A total of 397 hypoxia-related DEGs were detected and three genes (PDSS1, CDCA8 and SLC7A11) were selected to construct a prognosis, recurrence and diagnosis model. Then patients were divided into high- and low-risk groups. Our hypoxia-related signature was significantly associated with worse prognosis and higher recurrence rate. The diagnostic model also accurately distinguished HCC from normal samples and nodules. Furthermore, the hypoxia-related signature could positively regulate immune response and the high-risk group had higher fractions of macrophages, B memory cells and follicle-helper T cells, and exhibited higher expression of immunocheckpoints such as PD1and PDL1.Conclusions Altogether, our study showed that hypoxia-related signature is a potential biomarker for diagnosis, prognosis and recurrence of HCC, and it provided an immunological perspective for developing personalized therapies.


2020 ◽  
Author(s):  
Ze-bing Song ◽  
Guo-pei Zhang ◽  
shaoqiang li

Abstract Background: Hepatocellular carcinoma (HCC) is one of the most common malignant tumor in the world which prognosis is poor. Therefore, a precise biomarker is needed to guide treatment and improve prognosis. More and more studies have shown that lncRNAs and immune response are closely related to the prognosis of hepatocellular carcinoma. The aim of this study was to establish a prognostic signature based on immune related lncRNAs for HCC.Methods: Univariate cox regression analysis was performed to identify immune related lncRNAs, which had negative correlation with overall survival (OS) of 370 HCC patients from The Cancer Genome Atlas (TCGA). A prognostic signature based on OS related lncRNAs was identified by using multivariate cox regression analysis. Gene set enrichment analysis (GSEA) and a competing endogenous RNA (ceRNA) network were performed to clarify the potential mechanism of lncRNAs included in prognostic signature. Results: A prognostic signature based on OS related lncRNAs (AC145207.5, AL365203.2, AC009779.2, ZFPM2-AS1, PCAT6, LINC00942) showed moderately in prognosis prediction, and related with pathologic stage (Stage I&II VS Stage III&IV), distant metastasis status (M0 VS M1) and tumor stage (T1-2 VS T3-4). CeRNA network constructed 15 aixs among differentially expressed immune related genes, lncRNAs included in prognostic signature and differentially expressed miRNA. GSEA indicated that these lncRNAs were involved in cancer-related pathways. Conclusion: We constructed a prognostic signature based on immune related lncRNAs which can predict prognosis and guide therapies for HCC.


2020 ◽  
Author(s):  
Andi Ma ◽  
Yukai Sun ◽  
Racheal O. Ogbodu ◽  
Ling Xiao ◽  
Haibing Deng ◽  
...  

Abstract Background: It is well known that long non-coding RNAs (lncRNAs) play a vital role in cancer. We aimed to explore the prognostic value of potential immune-related lncRNAs in hepatocellular carcinoma (HCC). Methods: Validated the established lncRNA signature of 343 patients with HCC from The Cancer Genome Atlas (TCGA) and 81 samples from Gene Expression Omnibus (GEO). Immune-related lncRNAs for HCC prognosis were evaluated using Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO) analyses. LASSO analysis was performed to calculate a risk score formula to explore the difference in overall survival between high- and low-risk groups in TCGA, which was verified using GEO, Gene Ontology (GO), and pathway-enrichment analysis. These analyses were used to identify the function of screened genes and construct a co-expression network of these genes. Results: Using computational difference algorithms and lasso Cox regression analysis, the differentially expressed and survival-related immune-related genes (IRGs) among patients with HCC were established as five novel immune-related lncRNA signatures (AC099850.3, AL031985.3, PRRT3-AS1, AC023157.3, MSC-AS1). Patients in the low‐risk group showed significantly better survival than patients in the high‐risk group ( P = 3.033e−05). The signature identified can be an effective prognostic factor to predict patient survival. The nomogram showed some clinical net benefits predicted by overall survival. In order to explore its underlying mechanism, several methods of enrichment were elucidated using Gene Set Enrichment Analysis. Conclusion: Identifying five immune-related lncRNA signatures has important clinical implications for predicting patient outcome and guiding tailored therapy for patients with HCC with further prospective validation.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Qingmiao Shi ◽  
Chen Xue ◽  
Xin Yuan ◽  
Yuting He ◽  
Zujiang Yu

Abstract Hepatocellular carcinoma (HCC) ranks fourth in cancer-related mortality worldwide. N1-methyladenosine (m1A), a methylation modification on RNA, is gaining attention for its role across diverse biological processes. However, m1A-related regulatory genes expression, its relationship with clinical prognosis, and its role in HCC remain unclear. In this study, we utilized The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) database to investigate alterations within 10 m1A-related regulatory genes and observed a high mutation frequency (23/363). Cox regression analysis and least absolute shrinkage and selection operator were used to explore the association between m1A-related regulatory genes expression and HCC patient survival and identified four regulators that were remarkably associated with HCC patient prognosis. Additionally, an independent cohort from International Cancer Genome Consortium was studied to validate our discoveries and found to be consistent with those in the TCGA dataset. In terms of mechanism, gene set enrichment analysis linked these four genes with various physiological roles in cell division, the MYC pathway, protein metabolism, and mitosis. Kyoto Encyclopedia of Genes and Genomes analysis revealed that PI3K/Akt signaling pathway had potential relevance to m1A-related regulatory genes in HCC. These findings indicate that m1A-related regulatory genes may play crucial roles in regulating HCC progression and be exploited for diagnostic and prognostic purposes.


2021 ◽  
Author(s):  
Lili Li ◽  
Rongrong Xie ◽  
Qichun Wei

Abstract Background: Hepatocellular carcinoma (HCC) is one of the leading causes of mortality worldwide. N6-methyladenosine (m6A) methyltransferase, has been proved to act as an oncogene in several human cancers. However, little is known about its relationship with the long non-coding RNAs (lncRNAs) that remains elusive in HCC.Methods: We comprehensively integrated gene expression data acquired from 371 HCC and 50 normal tissues in The Cancer Genome Atlas (TCGA) database. Differentially expressed protein-coding genes (DE-PCGs)/lncRNAs (DE-lncRs) analysis and univariate regression & Kaplan-Meier (K-M) analysis was performed to identify m6A methyltransferase‑related lncRNAs that were related to overall survival (OS). m6A methyltransferase‑related lncRNA signature was constructed using the Least absolute shrinkage and selection operator (LASSO) Cox regression analyses. Furthermore, Cox regression analysis was applied to identify independent prognostic factors in HCC. The signature was validated in an internal validation set. Finally, the correlation analysis between gene signature and immune cells infiltration was also investigated via single-sample Gene Set Enrichment Analysis (ssGSEA) and immunotherapy response was calculated through Tumor Immune Dysfunction and Exclusion (TIDE) algorithm.Results: A total of 21 m6A methyltransferase-related lncRNAs were screened out according to Spearman correlation analysis with the immune score (|R| > 0.3, P < 0.05). We selected 3 prognostic lncRNAs to construct m6A methyltransferase-related lncRNA signature through univariate and LASSO Cox regression analyses. The univariate and multivariate Cox regression analyses demonstrated that the lncRNAs signature was a robust independent prognostic factor in OS prediction with high accuracy. The GSEA also suggested that the m6A methyltransferase-related lncRNAs were involved in the immune-related biological processes and pathways which were very well-known in the context of HCC tumorigenesis. Besides, we found that the lncRNAs signature was strikingly correlated with the tumor microenvironment (TME) immune cells infiltration and expression of critical immune checkpoints. Finally, results from the TIDE analysis revealed that the m6A methyltransferase-related lncRNAs could efficiently predict the clinical response of immunotherapy in HCC.Conclusion: Together, our study screened potential prognostic m6A methyltransferase related lncRNAs and established a novel m6A methyltransferase-based prognostic model of HCC, which not only provides new potential prognostic biomarkers and therapeutic targets but also deepens our understanding of tumor immune microenvironment status and lays a theoretical foundation for immunotherapy.


2021 ◽  
Author(s):  
Lili Li ◽  
Rongrong Xie ◽  
Guangrong Lu

N6-methyladenosine (m6A) methyltransferase has been shown to be an oncogene in a variety of cancers. Nevertheless, the relationship between the long non-coding RNAs (lncRNAs) and hepatocellular carcinoma (HCC) remains elusive. We integrated the gene expression data of 371 HCC and 50 normal tissues from The Cancer Genome Atlas (TCGA) database. Differentially expressed protein-coding genes (DE-PCGs)/lncRNAs (DE-lncRs) analysis and univariate regression & Kaplan-Meier (K-M) analysis were performed to identify m6A methyltransferase‑related lncRNAs. Three prognostic lncRNAs were selected by univariate and LASSO Cox regression analyses to construct the m6A methyltransferase-related lncRNA signature. Multivariate Cox regression analyses illustrated that this signature was an independent prognostic factor for overall survival (OS) prediction. The Gene Set Enrichment Analysis (GSEA) suggested that the m6A methyltransferase-related lncRNAs were involved in the immune-related biological processes and pathways. Besides, we discovered that the lncRNAs signature was correlated with the tumor microenvironment (TME) and the expression of critical immune checkpoints. Tumor Immune Dysfunction and Exclusion (TIDE) analysis revealed that the lncRNAs could predict the clinical response to immunotherapy. Our study had originated a prognostic signature for HCC based on the potential prognostic m6A methyltransferase-related lncRNAs. This study had deepened the understanding of the TME status of HCC patients and laid a theoretical foundation for the choice of immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document