scholarly journals Exploring the Prognostic Value and Biological Function of CPNE1 Gene in Hepatocellular Carcinoma

Author(s):  
Jinfang Su ◽  
Yongbiao Huang ◽  
Yali Wang ◽  
Rui Li ◽  
Wanjun Deng ◽  
...  

Abstract BackgroundLiver hepatocellular carcinoma (LIHC), the major histology subtype of primary liver cancer, accounts for 70-80% proportion of total liver cancer cases. Copine1 (CPNE1), the first discovered CPNE1 family member, participates in the process of carcinogenesis and development of diverse tumors. Our study aimed to investigate the expression and prognostic value of CPNE1 gene in hepatocellular carcinoma (HCC), to explore its functional network in HCC and its effects on biological behaviors such as proliferation, migration and invasion of HCC cells, and to explore its related signaling pathways.METHODSHCCDB, CCLE and HPA online databases were used to explore the expression of CPNE1 gene in HCC tissues; LinkedOmics online database was used to analyze the co-expression network of CPNE1 in hepatocellular carcinoma, and gene set enrichment analysis (GSEA) was used for GO functional annotation, KEGG pathway enrichment analysis, kinase target enrichment, miRNA target enrichment and transcription factor target enrichment analysis. The expression levels of CPNE1 in normal hepatocytes and several hepatocellular carcinoma cell lines were detected by RT-qPCR, and finally HepG2 and MHCC-97H cells were selected to construct CPNE1 knockdown cell lines by transfection with siRNA, and the knockdown efficiency was detected by Western Blot and RT-qPCR. The effect of CPNE1 knockdown on the proliferation of hepatocellular carcinoma cells was examined by CCK8 assay and clone formation assay; the effect of CPNE1 knockdown on the migration ability of hepatocellular carcinoma cells was assessed by cell scratch assay and Transwell cell migration assay; finally, the expression of related signaling pathway proteins was examined by Western Blot. The correlation of CPNE1 expression with immune infiltration and immune checkpoint molecules in HCC tissues was analyzed using TIMER online database.RESULTSAnalysis in several databases showed that CPNE1 was highly expressed in HCC tissues and significantly correlated with sex, age, cancer stage and tumor grade. Overall survival (OS) was significantly lower in patients with high CPNE1 expression than in patients with low CPNE1 expression, and CPNE1 could be used as an independent prognostic indicator for HCC. GSEA analysis showed that co-expressed genes of CPNE1 were mainly involved in biological processes such as establishment of protein localization to membrane, ribonucleoprotein complex biogenesis and lipid localization. Q-PCR showed that CPNE1 expression was upregulated in HCC cells compared with normal hepatocytes, and knockdown of CPNE1 gene inhibited the AKT/P53 pathway, resulting in decreased proliferation, migration and invasion of HCC cells. The level of CPNE1 expression in HCC was significantly and positively correlated with the level of infiltration of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells (p<0.001), and with the expression of immune checkpoint molecules PDCD1, CD274, CTLA4, LAG3, HAVCR2, and TIGIT.CONCLUSIONThe expression of CPNE1 was significantly higher in HCC tissues than in normal liver tissues, and high CPNE1 expression was associated with poor prognosis. Knockdown of CPNE1 inhibited AKT/P53 pathway activation and suppressed HCC cell proliferation and migration. There was a significant correlation between CPNE1 expression and tumor immune infiltration in HCC.

2020 ◽  
Vol 7 ◽  
Author(s):  
Saiyan Bian ◽  
Wenkai Ni ◽  
Mengqi Zhu ◽  
Qianqian Song ◽  
Jianping Zhang ◽  
...  

Purpose: N6-methyladenosine (m6A) RNA methylation has been implicated in various malignancies. This study aimed to identify the m6A methylation regulator-based prognostic signature for hepatocellular carcinoma (HCC) as well as provide candidate targets for HCC treatment.Methods: The least absolute shrinkage and selection operator (LASSO) analyses were performed to identify a risk signature in The Cancer Genome Atlas (TCGA) datasets. The risk signature was further validated in International Cancer Genome Consortium (ICGC) and Pan-Cancer Analysis of Whole Genomes (PCAWG) datasets. Following transfection of short hairpin RNA (shRNA) targeting YTHDF1, the biological activities of HCC cells were evaluated by Cell Counting Kit-8 (CCK-8), wound-healing, Transwell, flow cytometry, and xenograft tumor assays, respectively. The potential mechanisms mediated by YTHDF1 were predicted by overrepresentation enrichment analysis (ORA)/gene set enrichment analysis (GSEA) and validated by Western blotting.Results: Overexpression of m6A RNA methylation regulators was correlated with malignant clinicopathological characteristics of HCC patients. The Cox regression and LASSO analyses identified a risk signature with five m6A methylation regulators (KIAA1429, ZC3H13, YTHDF1, YTHDF2, and METTL3). In accordance with HCC cases in TCGA, the prognostic value of risk signature was also determined in ICGC and PCAWG datasets. Following analyzing the expression and clinical implications in TCGA and Gene Expression Omnibus (GEO), YTHDF1 was chosen for further experimental validation. Knockdown of YTHDF1 significantly inhibited the proliferation, migration, and invasion of HCC cells, as well as enhanced the apoptosis in vitro. Moreover, silencing YTHDF1 repressed the growth of xenograft tumors in vivo. Mechanism investigation indicated that YTHDF1 might promote the aggressive phenotypes by facilitating epithelial–mesenchymal transition (EMT) and activating AKT/glycogen synthase kinase (GSK)-3β/β-catenin signaling.Conclusion: The current study identified a robust risk signature consisting of m6A RNA methylation regulators for HCC prognosis. In addition, YTHDF1 was a potential molecular target for HCC treatment.


RSC Advances ◽  
2015 ◽  
Vol 5 (3) ◽  
pp. 1841-1845 ◽  
Author(s):  
Baiqi Wang ◽  
Hetao Chen ◽  
Rui Yang ◽  
Fang Wang ◽  
Ping Zhou ◽  
...  

The red signals from the cytoplasm of HCC cells reveal that the QD probes can specifically label liver cancer cells.


2021 ◽  
Author(s):  
Chun Duan ◽  
Bin Quan ◽  
Ni Wang ◽  
Jianghua Yang ◽  
Yan-Lin Yu

Abstract Background: Hepatocellular carcinoma (HCC) is a common malignancy with high morbidity. The current study aimed to explore the molecular mechannism of lncRNA SLC16A1-AS1 in the tumorigenesis of HCC.Material and Methods: The expression of SLC16A1-AS1 and miR-411 were examined in clinical HCC tissues. HCC cell lines Hep3B and Huh-7 were employed and transfected with si-SLC16A1-AS1. The correlation between SLC16A1-AS1 and miR-411 was verified by luciferase reporter assay. Cell viability was detected by CCK-8 assay. Cell migration and invasion capacity were examined by transwell assay. The protein level of MITD1 was analyzed by western blotting.Results: The expression of SLC16A1-AS1 markedly increased in HCC tissues and cell lines. Subsequent studies identified SLC16A1-AS1 as a downstream target of miR-411. In addition, SLC16A1-AS1 knockdown and miR-411 overexpression significantly stagnated progression of HCC cells. SLC16A1-AS1 knockdown also downregulated MITD1 levels. Conclusion: Our findings showed that SLC16A1-AS1 was overexpressed in HCC cells and tissues. SLC16A1-AS1 promoted the malignant characteristics of HCC cells and acted as an oncogene. Its regulatory effect may be associated with miR-411/MITD1 axis. Therefore, SLC16A1-AS1 has a potential be used as a biomarker or therapeutic target for the treatment of HCC.


2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Sijia Lei ◽  
Bin Zhang ◽  
Luyuan Huang ◽  
Ziyou Zheng ◽  
Shaohan Xie ◽  
...  

AbstractSteroid receptor RNA activator 1 (SRA1) has been described as a novel transcriptional co-activator that affects the migration of cancer cells. Through RT-PCR, we identified that skipping exon 3 of SRA1 produces two isoforms, including the truncated short isoform, SRA1-S, and the long isoform, SRA1-L. However, the effect of these two isomers on the migration of HCC cells, as well as the specific mechanism of exon 3 skipping remain unclear. In this study, we found up regulated expression of SRSF1 and SRA1-L in highly metastatic HCCLM3, as well as in HCCs with SRSF1 demonstrating the strongest correlation with SRA1-L. In contrast, we observed a constitutively low expression of SRA1-S and SRSF1 in lowly metastatic HepG2 cells. Overexpression of SRSF1 or SRA1-L promoted migration and invasion by increasing the expression of CD44, while SRA1-S reversed the effect of SRSF1 and SRA1-L in vitro. In addition, lung metastasis in mice revealed that, knockdown of SRSF1 or SRA1-L inhibited the migration of HCC cells, while SRA1-L overexpression abolished the effect of SRSF1 knockout and instead promoted HCC cells migration in vivo. More importantly, RNA immunoprecipitation and Cross-link immunoprecipitation analyses showed that SRSF1 interacts with exon 3 of SRA1 to up regulate the expression of SRA1-L in HCC cells. RNA pull-down results indicated that SRSF1 could also bind to exon 3 of SRA1 in vitro. Finally, minigene -MS2 mutation experiments showed that mutation of the SRA1 exon 3 binding site for SRSF1 prevented the binding of SRA1 pre-mRNA. In summary, our results provide experimental evidence that SRA1 exon 3 inclusion is up regulated by SRSF1 to promote tumor invasion and metastasis in hepatocellular carcinoma.


2021 ◽  
Vol 11 (4) ◽  
pp. 718-724
Author(s):  
Jiuwu Zhuo ◽  
Yishan Zheng ◽  
Wanying Hu ◽  
Guoping Yin

Sufentanil is a powerful analgesic that acts on μ-receptors, but there are few studies on sufentanil in cancer. The biological function and underlying mechanisms of sufentanil on the hepatocellular carcinoma (HCC) cells were explored in the present study. HCC cells were first treated with different concentrations of sufentanil and the most optimum concentration of sufentanil was determined. The expression of miR-204 in HCC cells was changed by transfected with miR-204 inhibitor and the transfection efficiency was assessed by qRT-PCR. CCK-8, wound-healing and Transwell assays were performed to evaluate the proliferation, migration and invasion of HCC cells, respectively. The level of AKT and PI3K phosphorylation (p-AKT and p-PI3K) were assessed by western blot analysis. Our results demonstrated that sufentanil effectively inhibited cell proliferation,migration and invasion in both Huh7 and Hep3B cells, and significantly decreased the expression of p-AKT and p-PI3K. In addition, miR-204 was upregulated in Huh7 and Hep3B cells treated with sufentanil, and low expression of miR-204 attenuated the damage of sufentanil on the viability of Huh7 and Hep3B cells. Taken together, sufentanil suppressed the proliferation, migration and invasion of HCC cells via inhibiting AKT/PI3K signaling pathway by targeting miR-204.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Wenpeng Zhao ◽  
Jiang Guo ◽  
Honglu Li ◽  
Liang Cai ◽  
Youjia Duan ◽  
...  

Abstract Background Abundant evidence has manifested that long noncoding RNAs (lncRNAs) are closely implicated in human cancers, including hepatocellular carcinoma (HCC). Remarkably, lncRNA FAM83H antisense RNA 1 (FAM83H-AS1) has been reported to be a tumor-propeller in multiple cancers. However, its effect on HCC progression remains unknown. Methods FAM83H-AS1 expression was analyzed by RT-qPCR. Colony formation, EdU, and flow cytometry as well as transwell assays were implemented to analyze the biological functions of FAM83H-AS1 on HCC progression. Luciferase reporter, RIP and RNA pull-down assays were implemented to detect the interaction among FAM83H-AS1, microRNA-485-5p (miR-485-5p), and myocyte enhancer factor 2D (MEF2D) in HCC cells. Results FAM83H-AS1 expression in HCC cells was markedly elevated. FAM83H-AS1 accelerated cell proliferation, migration and invasion whereas inhibiting cell apoptosis in HCC. Besides, we confirmed that FAM83H-AS1 acts as a miR-485-5p sponge in HCC cells. Additionally, MEF2D was verified to be a direct target of miR-485-5p. FAM83H-AS1 could upregulate MEF2D expression via sponging miR-485-5p. Further, rescue experiments testified that MEF2D upregulation or miR-485-5p downregulation offset the repressive effect of FAM83H-AS1 depletion on HCC cell progression. Conclusions FAM83H-AS1 facilitates HCC malignant progression via targeting miR-485-5p/MEF2D axis, suggesting that FAM83H-AS1 may be a promising biomarker for HCC treatment in the future.


2021 ◽  
Author(s):  
Sha She ◽  
Min Yang ◽  
Shi Ying Li ◽  
Huai Dong Hu ◽  
YiXuan Yang ◽  
...  

Abstract Background C-reactive protein (CRP), an acute phase protein, has been increasingly implicated in various tumors, and the role of CRP is positively correlated with invasion and metastasis in hepatocellular carcinoma cells. However, the mechanism of CRP affecting HCC progression remains poorly investigated. The present study investigated the role of CRP in HCC and the underlying mechanisms. Methods In the current study, CRP overexpression and suppression expression experiments were used to evaluate the effect of CRP on malignant biological behavior of liver cancer cells in vitro. Then iTRAQ-mass spectrometry analysis was used to identify CRP co-immunoprecipitation complexes. Detecting the interaction between CRP and Eph receptor B3 (EphB3) by co-precipitation. Moreover, immunofluorescence colocalization and co-precipitation, and Western Blot, in vivo model were applied to study the molecular mechanism of CRP affecting the development of Hepatocellular Carcinoma. Results We first found that CRP was significantly upregulated in HCC tissues and HCC cells, the expression level correlated with the metastatic ability of HCC cells. Knockdown of CRP significantly suppresses migration and invasion capacity in HCC cells. Through a proteomic analysis of CRP co-immunoprecipitation complexes, the EphB3 was identified as a new CRP interactor. Then we found that the expression and functions of EphB3 were consistent with CRP in HCC. In addition, co-immunoprecipitation and immunofluorescence assays suggested that EphB3 was able to interact with MAPK/ERK to activate MAPK/ERK signaling pathways. Furthermore, we showed that CRP can induce the phosphorylation of MAPK/ERK by binding EphB3. CRP also significantly stimulated MMP-9 expression, mainly by activating HIF-1α via the MAPK/ERK pathways. Conclusions Our findings showed that CRP increased HCC cells migration and invasion by binding EphB3 to activate MAPK/ERK signaling pathways. It suggested that CRP may become a prognostic factor and a potential therapeutic target for liver cancer.


2020 ◽  
Author(s):  
jiahui Wan ◽  
Xiuli Wang ◽  
Fusheng Zhao ◽  
Ying Jiang ◽  
Wei Ma ◽  
...  

Abstract Background: Increasing evidences show that long non-coding RNA (lncRNA) plays the role of competitive endogenous RNAs (ceRNAs) in the development and progression of cancers. The purpose of our study was to identify potential lncRNA biomarkers that serve as a therapeutic target and prognostic biomarker in HCC.Methods: The differential expression of RNAs was examined using the edgeR package. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to predict potential functions. Survival analysis and ROC curve analysis were performed to predict ceRNA network had significant prognostic value. The biological functions of MYLK-AS1 on HCC cells were studied by RNA interference approaches in vitro. Cell proliferation, migration and invasion were detected by Cell Counting Kit-8(CCK-8) assay, wound healing and transwell assay relatively. The mechanism of competitive endogenous RNAs (ceRNAs) were predicted and verified by bioinformatic analysis, western blot analysis and luciferase assays.Results: The newly constructed ceRNA network comprised 76 HCC specific lncRNAs, 15 miRNAs, and 35 mRNAs from the database (Targetscan, miRTarBase, and miRDB). 10 differentially expressed lncRNAs and 10 differentially expressed mRNAs were significantly associated with overall survival in HCC (P value < 0.05). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes(KEGG) pathways enrichment analysis results showed the differentially expressed mRNAs were involved primarily in the most significant cancer-related signaling pathways. ROC curve analysis demonstrated that MYLK-AS1—has-mir-424—CCNE1 ceRNA network had significant prognostic value. The expression of MYLK-AS1 was up-regulated in HCC cells and tissues. Biological function analyses indicated that down-regulation of MYLK-AS1 suppressed cell proliferation, migration and invasion . MYLK-AS1 and miR-424-5p bound directly and reversibly to each other. MYLK-AS1 could positively regulate CCNE1 expression by competitively binding to miR-424-5p. Conclusion: The current study provides novel insights into the lncRNA-related ceRNA network in HCC and the MYLK-AS1 may be a candidate biomarker for molecular diagnosis and prognosis monitoring of HCC.


2020 ◽  
Author(s):  
Weiwei Liu ◽  
Qian Feng ◽  
Wenjun Liao ◽  
Jun Gao ◽  
Jiyuan Ai ◽  
...  

Abstract Background: Numerous studies have shown that TUG1 has an important relationship with tumorigenesis. TUG1 is highly expressed in most tumors and can promote tumor development. However, the role of TUG1 in hepatocellular carcinoma (HCC) remains to be studied. miR-29a plays a tumor suppressor role in a variety of tumors, and there is a relationship between TUG1 and miR-29a, but the specific relationship and mechanism of action are still unclear. miR-29a can inhibit the expression of IFITM3. However, the regulatory relationship between these three components requires elucidation. This study aimed to investigate the regulatory relationship between TUG1, miR-29a, and IFITM3 in human hepatocarcinogenesis.Methods: The expression levels of TUG1 and miR-29a in tumor tissues and adjacent non-tumor tissues of 41 HCC patients were detected by real-time quantitative polymerase chain reaction. The migration and invasion of liver cancer cells were studied by a wound healing assay and the Transwell method. The apoptosis rate of hepatocarcinoma cells was detected by flow cytometry, and the proliferation rate of hepatoma cells was detected by the EdU method. Immunofluorescence was selected to detect the expression of TUG1 and IFITM3 in HCC-LM3 and HL-7702. The relationship between TUG1 and miR-29a was detected using a double luciferase report and FISH. Tumors were established in vivo by subcutaneous injection of hepatocellular carcinoma cells into nude mice and injection of these cells into the tail vein. Western blotting was used to quantify the biomarkers. Results: TUG1 expression increased significantly in both tumor tissues and HCC cells. The expression of miR-29a in liver cancer tissues was also significantly lower than that in normal human liver tissues. The expression of TUG1 in HCC tissue samples was negatively correlated with that of miR-29a. Moreover, the expression of TUG1 was positively correlated with the expression of IFITM3. TUG1 can regulate the migration, invasion, apoptosis, and proliferation of HCC lines in vitro and regulate the development of tumors in vivo. Knocking down TUG1 will increase miR-29a expression, and thus, weaken the invasion, migration, and proliferation of HCC cells and enhance their apoptosis. miR-29a can affect the occurrence and progression of liver cancer through IFITM3. It was found that TUG1 regulates IFITM3 in HCC cells via miR-29a, and its expression affects cell invasion, migration, proliferation, and apoptosis.Conclusion: As a CeRNA, TUG1 competitively binds mir-29a to regulate IFITM3 and promote the development of liver cancer. Downregulation of TUG1 can significantly inhibit the migration, invasion, and proliferation of liver cancer cells, and TUG1 is expected to serve as a key gene to improve the prognosis of patients.


Sign in / Sign up

Export Citation Format

Share Document