Open Access

ACTG1 and TLR3 are biomarkers for alcohol‑associated hepatocellular carcinoma

  • Authors:
    • Bing Gao
    • Shicheng Li
    • Zhen Tan
    • Leina Ma
    • Jia Liu
  • View Affiliations

  • Published online on: November 26, 2018     https://doi.org/10.3892/ol.2018.9757
  • Pages: 1714-1722
  • Copyright: © Gao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Alcohol consumption is a risk factor for the development of hepatocellular carcinoma (HCC); however, the association between alcohol and HCC remains unknown. The present study aimed to identify key genes related to alcohol‑associated HCC to improve the current understanding of the pathology of this disease. Alcohol‑associated and non‑alcohol‑associated HCC samples in the GSE50579 dataset of the Gene Omnibus Database were analyzed to investigate altered gene expression. Integrated bioinformatics methods were employed to clarify the biological functions of the differentially expressed genes (DEGs), including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein‑protein interactions (PPIs). The present study reported that candidate biomarker micro (mi)RNAs via TargetScan Human 7.1. DEGs and their associated miRNAs (according to bioinformatics analysis) were validated using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Additionally, 284  EGs from the GSE50579 dataset were revealed. In GO term analysis, DEGs were closely associated with the ‘regulation of nucleic acid metabolism’. KEGG pathway analysis indicated that the DEGs were tightly engaged in the ‘VEGF and VEGF receptor signaling network’, ‘proteoglycan syndecan‑mediated signaling events’, ‘erbB receptor signaling’ and ‘β1 integrin cell surface interactions’. According to the results of PPI and heat map analysis, the main hub genes were centrin 3 (CETN3), Toll‑like receptor 3 (TLR3), receptor tyrosine‑protein kinase (ERBB4), heat shock protein family member 8, actin γ1 (ACTG1) and α‑smooth muscle actin. it was demonstrated that the ACTG1, TLR3, miR‑6819‑3p and miRΝΑ (miR)‑6877‑3P had undefined associations. Furthermore, RT‑qPCR analysis revealed that miR‑6819‑3p and miR‑6877‑3P may enhance the expression levels of ACTG1 and inhibit the expression levels of TLR3 in alcohol‑associated HCC tissues. TLR3 and ACTG1 were proposed as potential biomarkers of alcohol‑associated HCC. Investigation into the regulatory functions of miR‑6819‑3p and miR‑6877‑3P may provide novel insights into the treatment of alcohol‑associated HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common types of cancer to result in mortality, worldwide. It has been acknowledged that the primary carcinogenic factors of HCC include hepatitis B virus (HBV) and hepatitis C virus (HCV) infection, alcohol and non-alcoholic steatohepatitis (1). Of these principal etiologies, HBV and HCV predominate in Asian cohorts, whereas European cohorts are associated with heterogeneous etiologies, including alcohol consumption, and viral hepatitis within cirrhotic and non-cirrhotic livers (2). The analysis of patients with chronic HCV infection, in addition to alcohol abuse and dependence, indicated that elevated proton pump activity increased the risk of hepatic decompensation and hepatocellular carcinoma (3). According to the study of non-alcoholic fatty liver disease (NAFLD) in Indian patients, 11 patients with a history of considerable alcohol consumption had at least one risk factor for NAFLD, including alcohol-associated HCC (4). Asian countries have been reported to have a high incidence of HBV infection. A previous study of the Indian population demonstrated a lower prevalence of HBV infection of unknown origin (4); however, the direct role of alcohol in the carcinogenesis of HCC, or the mechanism underlying alcohol-induced hepatotoxicity associated with alcohol metabolites, oxidative stress and iron metabolism, are yet to be elucidated (2,5). The administration of alcohol/diethyl nitrosamine/carbon tetrachloride in mice successfully induced an α-fetoprotein-secreting HCC model in adult male BALB/c mice. This research may improve current knowledge of the stages of occurrence and development associated with HCC (5). The underlying mechanism of the alcohol-induced development of liver disease remains unknown. Recently, the epidemiological evidence regarding the association of alcohol and liver cancer was summarized; this publication demonstrated that it is crucial to identify all aspects associated with the effects of alcohol metabolism on alterations in hepatocyte metabolism, particularly neoplasia-associated alterations (6).

Microarray data analysis of the expression of different genes has provided powerful and feasible evidence for the clinical diagnosis and treatment of HCC. In the present study, the GSE50579 dataset was downloaded from the Gene Expression Omnibus database (GEO; http://www.ncbi.nlm.nih.gov/geo), which includes data regarding alcohol-associated HCC and HCC liver tissue samples; the differentially expressed genes (DEGs) were screened, and bioinformatics analysis was conducted for functional and pathway enrichment, and for the investigation of protein-protein interaction (PPI) networks. Statistical analysis and functional annotation revealed that actin γ1 (ACTG1; DFN20/26) and Toll-like receptor 3 (TLR3) may be biomarkers for alcohol-induced HCC. Notably, the present study reported that DEG-associated microRNAs (miRNAs) may be considered as potential targets in the treatment of alcohol-induced HCC. In addition, these biomarkers were assessed by reverse-transcription-quantitative polymerase chain reaction (RT-qPCR) in samples collected from patients with HCC.

Materials and methods

Ethics statement

The present study was conducted with the approval of the Ethics Committee of the Affiliated Hospital of Qingdao University (Qingdao, China). Patients provided informed consent for the use of their tissue specimens. The mean age of 30 patients, including 18 males and 12 females, was 52.6±16.8 years. A total of 30 clinical tissue samples, including 12 from patients with alcohol-associated HCC, 12 from patients with non-alcohol-associated HCC tissues, and six from healthy individuals were obtained from the Department of Hepatobiliary Surgery of the Affiliated Hospital of Qingdao University between March 2017 and July 2017.

Identification of differentially expressed genes

The microarray data set GSE50579 (7) was downloaded from the GEO, in which 10 alcohol-associated HCC and 16 clinical HCC liver samples were analyzed. The platform employed for analysis was the Agilent-028004 SurePrint G3 Human GE 8×60K Microarray (Agilent Technologies, Inc., Santa Clara, CA, USA). The annotation file was also acquired; the original CEL format was converted into an expression matrix using a function to study RNA with an Affy package (8). Probes were mapped to genes according to the annotation file in R version 3.3.3 (9). Average expression levels were calculated for the probes corresponding to the same gene. DEGs between alcohol-associated HCC tissue samples and non-alcohol associated individuals were removed via the goodness of fit test; genes that were not differentially expressed were subject to average distribution. The R limma package (9) was adopted for differential analysis. |logFC| (fold change) >1.0 and P<0.05 were set as the criteria to screen out DEGs between alcohol-associated HCC liver tissue samples and non-alcohol associated HCC samples.

Bioinformatics analysis of DEGs

Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses (https://www.genome.jp/kegg) were performed for the DEGs using the Cluego APP in Cytoscape (version 3.5.1; www.cytoscape.org). A previous study (10) demonstrated that certain genes sharing the same pathway or similar biological functions and gene expression patterns exhibit the same pathological effects. Therefore, the construction of a gene co-expression network may help identify gene sets associated with specific pathways and biological processes. In the present study, Pearson correlation analysis of co-expression was performed with a coefficient >0.85 and P<0.05 as the criteria for investigation. In addition, PPIs were constructed using Cytoscape software. Hub genes were identified within the PPIs of DEGs, and the edge length between nodes from these hub genes was determined.

miRNA analysis of the DEGs

The present study investigated numerous genes, including, centrin 3 (CETN3), TLR3, erbB2 receptor tyrosine kinase 4 (ERBB4), heat shock protein family member 8 (HSPA8), ACTG1 and α-smooth muscle actin (ACTA2) on the TargetScan Human 7.1 website (www.targetscan.org/vert_71) to predict the associated miRNAs of these genes. A diagram of the miRNAs and their predicted target genes was produced using Fun Rich software (http://funrich.org; version 3.0). Scores ≥90 points and overlapping miRNA quantity ≥3 were set to obtain the desired miRNAs and target DEGs.

RNA extraction

Tissues were stored at −80°C until use. Total RNA was extracted using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturers instructions. The RNA solutions were stored at −80°C. The RNA quality was determined by spectrophotometer.

RT-qPCR

RNA (1 µg) was reverse transcribed into cDNA using FastQuant RT kit (with gDNase) (Tiangen Biotech Co., Ltd., Beijing, China), according to the manufacturers protocol. cDNA was amplified using SYBR Green Real Master mix (Tiangen Biotech Co., Ltd.), according to the manufacturers protocol. The PCR reaction was conducted at 95°C for 15 min, followed by 40 cycles at 95°C for 10 sec, 58°C for 20 sec and 72°C for 30 sec. RT-qPCR was performed on the ABI StepOnePlus thermocycler (Thermo Fisher Scientific, Inc.). U6 single nuclear (sn)RNA was employed as the loading control for micro (mi)RNA expression, and β-actin for the expression of genes; stem-loop RT primers were used for miRNAs, and oligo (dinucleotide) primers for genes. Primers are displayed in Table I. All reactions were repeated three times and the data were calculated by the comparative 2−ΔΔCq method (11).

Table I.

Primers for differentially-expressed genes and their associated miRNAs.

Table I.

Primers for differentially-expressed genes and their associated miRNAs.

Gene/miRNA nameForward primer (53)Reverse primer (53)Stem-loop RT primer (53)
ACTG1 ATGGAAGGAAACACGGCTC CACTCTGTTCTTCCGCCG
TLR3 AGTGCACTTGGTGGTGGAG AGGAAAGGCTAGCAGTCATCC
β-actin TGACGGGGTCACCCACACTG AAGCTGTAGCCGCGCTCGGT
hsa-miR-6819 AAGCCTCTGTCCCCA CAGTGCGTGTCGTGGAGT GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGAC
hsa-miR-6877 CAGCCTCTGCCCTTG CAGTGCGTGTCGTGGAGT GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGAC
U6 CTCGCTTCGGCAGCACA AACGCTTCACGAATTTGCGT AACGCTTCACGAATTTGCGT

[i] miRNA/miR, microRNA; RT, reverse transcription.

Statistical analysis

All statistical analyses were performed using R software version 3.3 and GraphPad Prism 6 software (GraphPad Software, Inc. La Jolla, CA, USA). The data are presented as the mean ± standard deviation. Statistical significance was examined using Students t-test or one-way analysis of variance followed by Dunnetts test. P<0.05 was considered to indicate a statistically significance difference. To identify the prognostic value of the differentially expressed coding genes, Kaplan-Meier survival analysis was conducted.

Results

Identification of DEGs

A total of 284 DEGs from the GSE50579 dataset were obtained, which included 68 upregulated and 216 downregulated DEGs. The top DEGs were presented as a heat map (Fig. 1).

Bioinformatics analysis of the DEGs

To further understand the functions of the DEGs, GO and KEGG pathway analyses were conducted. The GO analysis results indicated that the DEGs were involved in ‘immune response’ and ‘regulation of nucleobase, nucleoside, nucleotide and nucleic acid metabolism’ (Fig. 2). In addition, KEGG pathway analysis revealed that the DEGs were closely associated with ‘VEGF and VEGFR signaling network’, ‘proteoglycan syndecan-mediated signaling events’, ‘erbB receptor signaling network’ and ‘β1 integrin cell surface interactions’ (Fig. 3).

According to the PPI network in Fig. 4, including the upregulated (Fig. 4A) and downregulated PPI network analyses (Fig. 4B), seven hub genes were identified, including CETN3, TLR3, ERBB4, HSPA8, ACTG1, ACTA2 and interleukin-8. For consistency with the heat map, CETN3, TLR3, ERBB4, HSPA8, ACTG1 and ACTA2 were selected for further investigation.

A diagram was generated according to the results of TargetScan analysis of DEGs and their associated miRNAs (Fig. 5). In accordance with the criteria previously set, miRNA (miR)-6819-3P and miR-6877-3P, and their common target genes ACTG1 and TLR3, were selected for further analysis via RT-qPCR.

Validation of DEGs and miRNAs via RT-qPCR

The relative mRNA expression levels of the DEGs and selected miRNAs are presented in Fig. 6. The significantly upregulated mRNA expression levels of ACTG1 (Fig. 6A) were consistent with the bioinformatics analysis. Similarly, the results for TLR3 (Fig. 6B) were in line with the bioinformatics analysis, in which the gene was downregulated in alcohol-associated HCC tissues. It was also indicated that miR-6819 (Fig. 6C) and miR-6877 (Fig. 6D) were upregulated in alcohol-associated HCC compared with non-alcohol-associated HCC. These results indicated that miR-6819 and miR-6877 may positively influence the ACTG1 gene, and exert a negative regulatory effect on TLR3.

Discussion

The role of chronic alcohol consumption in the induction and development of cancer is well known; heavy alcohol consumption has been reported to exhibit negative effects on human health, and there is increasing evidence to suggest that alcohol increases the risk of carcinogenesis (12).

The global HCC BRIDGE study reported that the most common risk factor in North America, Europe and Japan is HCV, and HBV in China, South Korea and Taiwan; together with estimates of alcohol consumption per capita (World Health Organization), alcohol has been reported as a strong risk factor for HCC. Additionally, the reported rate of alcohol abuse in China, Taiwan and Japan was estimated to be 24, 18 and 2%, respectively (1). Studies of the alcohol-induced liver injury model revealed that hypothalamic β-endorphin neuron transplants may reduce liver weight and triglyceride accumulation; fewer pathological alterations occurred, including infiltration of inflammatory cells and steatosis of hepatocytes (13). In addition, investigation into the association between alcohol consumption and the risk of gastric cancer suggested that aldehyde dehydrogenase [ALDH2; National Center for Biotechnology Information (NCBI) accession no. rs671], rather than alcohol dehydrogenase 1B (ADH1B; NCBI accession no. rs1229984) and 1C (ADH1C; NCBI accession no. rs698), which modify acetaldehyde, may serve an important role in gastric cancer (14). Xu et al (15) reported that alcohol may promote the metastasis of colorectal cancer cells by modulating the glycogen synthase kinase 3β/β-catenin/monocyte chemoattractant protein-1 signaling pathway. Furthermore, it has been demonstrated that an alcohol intake >30 g/day may be associated with an increased risk of colon cancer (12). Similarly, research into breast cancer suggested that the accumulation of alcohol at a sufficient level may induce cytochrome P450 2E1, which causes mutagenic DNA adducts (16). In the present study, DEGs were compared between alcohol-associated HCC (n=10) and HCC (n=16) tissues in the GSE50579 dataset; ACTG1 and TLR3 were selected among 284 DEGs as potential therapeutic biomarkers in alcohol-associated HCC, as determined by bioinformatics analysis.

The ACTG1 gene encodes actin γ1, which belongs to the actin family of six highly conserved proteins. These actin filaments are critical for the structure of the cytoskeleton and the shape of the cell, and as such have been reported to regulate cell motility, contraction and growth (17). In a family with DFN20/26-associated hearing loss, it was reported that the cause of the condition may be due to a missense mutation in the ACTG1 gene. The Thr278lle mutation in ACTG1 has been predicted to affect the structure of the protein and induce germline mutations in cytoplasmic actin isoforms (18). Notably, researchers observed that within this Norwegian family, ACTG1 mutations were not frequently associated with hereditary hearing impairment. In addition, the p.V370A mutation may impair actin function, which was demonstrated by a yeast growth assay (17). K118N and E241K in ACTG1 have been considered as two novel mutations that cause an aberrant multi-vacuolar pattern in yeast and mammalian cells (19). According to whole exome sequencing data and massive parallel sequencing, p.M305T and p.E316k have been selected as nonsyndromic hearing loss variants (20,21). Moreover, ACTG1 is positively implicated in the growth and survival of mammals, as ACTG−/− animals were observed to be smaller in size and exhibited a delay in the development of the cardiac outflow tract (22). Utilizing targeted next generation sequencing analysis, another study reported that in Japanese families, ACTG1, nonsyndromic hearing impairment 5, POU class 4 homeobox 3, solute carrier family 26 member 5, SIX homeobox 1, myosin VIIA, cadherin related 23, protocadherin related 15 and Usher syndrome 2A may be candidate genes for early-childhood hearing loss (23).

ACTG1 has also been associated with other types of cancer. Studies have demonstrated that miR-888 may reduce the mRNA expression levels of all four adherens junction pathway-associated genes, including E-cadherin, ACTG1, receptor-type tyrosine-protein phosphatase T and cell division cycle 42 in MCF-7 cells (24). Additionally, ACTG1 has been reported to exhibit high expression levels in skin cancer tissue, where it may regulate A431 cell proliferation and migration via the Rho-associated protein kinase signaling pathway (25). ACTG1 has also been predicted to be a target gene of miR-145-5P in non-small cell lung cancer (26). Furthermore, ACTG1 and matrix metalloproteinase 14 (MMP14) were validated as targets of miR-10a in colorectal cancer cells; ectopic expression of ACTG1 and MMP14 may delay decreases in cell adhesion and anoikis resistance activity (27). It has also been reported that in HBx-induced HCC, ATCG1 was upregulated in human liver tissues (28).

TLR3 belongs to the Toll-like receptor family and is a receptor for double-stranded (ds)RNA (29); as such, TLR3 is reported to exhibit protective immunity during viral infection (30). TLR3 has also been reported in a number of cancer types and the TLR3 signaling pathway may serve an important role in cell homeostasis. BM-06, a 25-nucleotide dsRNA, may activate TLR3 to inhibit the proliferation and promote the apoptosis of HepG2.2.15 cells (29). TLR3-TIR-domain-containing adapter-inducing interferon-β (TRIF) signaling may contribute to myocardial inflammation and infarction when ischemia-reperfusion-associated extracellular RNA is released (31). In addition, TLR3 is positively associated with HBV antigen (HBsAg) in HepG2.2.15 cells, and may promote interstitial immunoreactive cell infiltration and HCC cell apoptosis (32). According to RNA-sequencing analysis, TLR3 was proposed as an immune gene, particularly within BV-2 microglial cells (33). High expression levels of TLR3 are a distinctive feature of CD8+ dendritic cells, where activation of the receptor may induce an interferon (IFN)-dependent antiviral response in dendritic cell subtypes (34). It has been reported that in Chinese newborns with severe hepatitis, TLR3, TLR2, TLR4 and TLR9 may serve as potential therapeutic targets for treatment (35). Of note, TLR3 exhibits lower levels of expression in HCC when compared with adjacent tissues, and is positively associated with TRIF, nuclear factor-κB and IFN regulatory factor 3, which may inhibit HCC proliferation and promote HCC cell apoptosis (36). The expression levels of TLR3, TRIF and mitochondrial antiviral-signaling proteins were consistently decreased in chronic HCV-infected liver tissue, compared to that of than non-diseased liver tissue (37). Additionally, endogenous miR-155 can negatively regulate TLR3 expression and inhibit IFN-β production (38). Another study has reported that a TLR3 agonist may enhance the clinical efficacy of sorafenib in treating HCC (29,39). A TLR3 agonist may also inhibit angiogenesis and induce the apoptosis of human HCC cells (40). In addition, the cytoplasmic expression of TLR3 was positively associated with HBV infection (32,36). Compared with that of adjacent tissues, the expression of TLR3 is lower in HCC tissues and is correlated with longer durations of survival (36). The evidence that dsRNA induces the activation of TLR3 further strengthens the hypothesis that TLR3, as a cytotoxic agent, may contribute to developments in HCC therapy (41).

miRNAs engage in numerous cellular activities and affect important physiological functions. In the present study, ACTG1 and TLR3 were proposed to be targets of miR-6819-3P and miR-6877-3P. In addition, the expression levels of miR-6819-3P and miR-6877-3P were observed to be higher in alcohol-associated HCC compared with non alcohol-associated HCC tissues; the upregulation of miR-6877-3P appeared to be higher compared with the upregulation of miR-6819-3P. Additionally, ACTG1 and TLR3 may regulate the VEGF and VEGFR signaling, proteoglycan syndecan-mediated signaling, erbB receptor signaling, and β1 integrin cell surface interactions. ACTG1 and TLR3 may also function in the immune response and nucleic acid metabolism. The results of the present study indicated that miR-6819-3P and miR-6877-3P enhanced the expression of ACTG1 and inhibited that of TLR3. Further investigation is required to determine the association between ACTG1, TLR3, miR-6819-3P and miR-6877-3P in alcohol-associated HCC tissues.

In conclusion, comprehensive bioinformatics analysis was performed to screen candidate genes for the treatment of alcohol-associated HCC; the physiological functions and regulatory mechanisms of TLR3 and ACTG1 require further study. To further investigate the effects of alcohol on HCC, molecular biological analysis in established HCC animal models may be advantageous; however, the findings of the present study may provide novel insight into the alcohol-associated etiopathogenesis of HCC and subsequently improve the clinical diagnosis of the disease.

Acknowledgements

Not applicable.

Funding

The present study was supported by funds from the National Natural Sciences Foundation of China (grant no. 81502065), the China Postdoctoral Science Foundation Funded Project (grant nos. 2016T90613 and 2015M580574), the Natural Sciences Foundation of Shandong Province (grant nos. ZR2014HQ009 and ZR2014HM004), the Postdoctoral Innovation Project of Shandong Province (grant no. 201602037), the Qingdao Postdoctoral Research Project (grant no. 2015167) and the Qingdao Innovation Project (grant no. 16-5-1-56-JCH).

Availability of data and materials

The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.

Authors contributions

JL and LM designed the experiments. BG, SL and ZT conducted the experiments. SL and ZT collected the clinical data and were responsible for ethics approval and consent of patients to participate in the study. BG and SL performed the bioinformatics and statistical analysis; JL and LM were responsible for the overall design and funding of the project. JL, LM and BG prepared the figures and wrote the manuscript. All authors have and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the Affiliated Hospital of Qingdao University. Written informed consent was obtained from patients.

Patient consent for publication

Written informed consent was obtained from all participating patients.

Competing interests

The authors declare that they have no competing interests.

References

1 

Park JW, Chen M, Colombo M, Roberts LR, Schwartz M, Chen PJ, Kudo M, Johnson P, Wagner S, Orsini LS and Sherman M: Global patterns of hepatocellular carcinoma management from diagnosis to death: The BRIDGE Study. Liver Int. 35:2155–2166. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Nahon P and Nault JC: Constitutional and functional genetics of human alcohol-related hepatocellular carcinoma. Liver Int. 37:1591–1601. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Li DK, Yan P, Abou-Samra AB, Chung RT and Butt AA: Proton pump inhibitors are associated with accelerated development of cirrhosis, hepatic decompensation and hepatocellular carcinoma in noncirrhotic patients with chronic hepatitis C infection: Results from ERCHIVES. Aliment pharmacol Ther. 47:246–258. 2018. View Article : Google Scholar : PubMed/NCBI

4 

David D, Raghavendran A, Goel A Bharath, Kumar C, Kodiatte TA, Burad D, Abraham P, Ramakrishna B, Joseph P, Ramachandran J and Eapen CE: Risk factors for non-alcoholic fatty liver disease are common in patients with non-B non-C hepatocellular carcinoma in India. Indian J Gastroenterol. 36:373–379. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Xin B, Cui Y, Wang Y, Wang L, Yin J, Zhang L, Pang H, Zhang H and Wang RA: Combined use of alcohol in conventional chemical-induced mouse liver cancer model improves the simulation of clinical characteristics of human hepatocellular carcinoma. Oncol Lett. 14:4722–4728. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Ramadori P, Cubero FJ, Liedtke C, Trautwein C and Nevzorova YA: Alcohol and hepatocellular carcinoma: Adding fuel to the flame. Cancers (Basel). 9:E1302017. View Article : Google Scholar : PubMed/NCBI

7 

Neumann O, Kesselmeier M, Geffers R, Pellegrino R, Radlwimmer B, Hoffmann K, Ehemann V, Schemmer P, Schirmacher P, Lorenzo Bermejo J and Longerich T: Methylome analysis and integrative profiling of human HCCs identify novel protumorigenic factors. Hepatology. 56:1817–1827. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Harbig J, Sprinkle R and Enkemann SA: A sequence-based identification of the genes detected by probesets on the Affymetrix U133 plus 2.0 array. Nucleic Acids Res. 33:e312005. View Article : Google Scholar : PubMed/NCBI

9 

Wettenhall JM, Simpson KM, Satterley K and Smyth GK: affylmGUI: A graphical user interface for linear modeling of single channel microarray data. Bioinformatics. 22:897–899. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Han L, Suzek TO, Wang Y and Bryant SH: The text-mining based pubchem bioassay neighboring analysis. BMC Bioinformatics. 11:5492010. View Article : Google Scholar : PubMed/NCBI

11 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Cho E, Lee JE, Rimm EB, Fuchs CS and Giovannucci EL: Alcohol consumption and the risk of colon cancer by family history of colorectal cancer. Am J Clin Nut. 95:413–419. 2012. View Article : Google Scholar

13 

Murugan S, Boyadjieva N and Sarkar DK: Protective effects of hypothalamic beta-endorphin neurons against alcohol-induced liver injuries and liver cancers in rat animal models. Alcohol Clin Exp Res. 38:2988–2997. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Hidaka A, Sasazuki S, Matsuo K, Ito H, Sawada N, Shimazu T, Yamaji T, Iwasaki M, Inoue M and Tsugane S: JPHC Study Group: Genetic polymorphisms of ADH1B, ADH1C and ALDH2, alcohol consumption, and the risk of gastric cancer: The Japan Public Health Center-based prospective study. Carcinogenesis. 36:223–231. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Xu M, Wang S, Qi Y, Chen L, Frank JA, Yang XH, Zhang Z, Shi X and Luo J: Role of MCP-1 in alcohol-induced aggressiveness of colorectal cancer cells. Mol Carcinog. 55:1002–1011. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Brooks PJ and Zakhari S: Moderate alcohol consumption and breast cancer in women: From epidemiology to mechanisms and interventions. Alcohol Clin Exp Res. 37:23–30. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Rendtorff ND, Zhu M, Fagerheim T, Antal TL, Jones M, Teslovich TM, Gillanders EM, Barmada M, Teig E, Trent JM, et al: A novel missense mutation in ACTG1 causes dominant deafness in a Norwegian DFNA20/26 family, but ACTG1 mutations are not frequent among families with hereditary hearing impairment. Eur J Hum Genet. 14:1097–1105. 2006. View Article : Google Scholar : PubMed/NCBI

18 

van Wijk E, Krieger E, Kemperman MH, De Leenheer EM, Huygen PL, Cremers CW, Cremers FP and Kremer H: A mutation in the gamma actin 1 (ACTG1) gene causes autosomal dominant hearing loss (DFNA20/26). J Med Genet. 40:879–884. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Morín M, Bryan KE, Mayo-Merino F, Goodyear R, Mencía A, Modamio-Høybjør S, del Castillo I, Cabalka JM, Richardson G, Moreno F, et al: In vivo and in vitro effects of two novel gamma-actin (ACTG1) mutations that cause DFNA20/26 hearing impairment. Hum Mol Genet. 18:3075–3089. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Park G, Gim J, Kim AR, Han KH, Kim HS, Oh SH, Park T, Park WY and Choi BY: Multiphasic analysis of whole exome sequencing data identifies a novel mutation of ACTG1 in a nonsyndromic hearing loss family. BMC Genomics. 14:1912013. View Article : Google Scholar : PubMed/NCBI

21 

Wei Q, Zhu H, Qian X, Chen Z, Yao J, Lu Y, Cao X and Xing G: Targeted genomic capture and massively parallel sequencing to identify novel variants causing Chinese hereditary hearing loss. J Transl Med. 12:3112014. View Article : Google Scholar : PubMed/NCBI

22 

Bunnell TM and Ervasti JM: Delayed embryonic development and impaired cell growth and survival in Actg1 null mice. Cytoskeleton (Hoboken). 67:564–572. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Mutai H, Suzuki N, Shimizu A, Torii C, Namba K, Morimoto N, Kudoh J, Kaga K, Kosaki K and Matsunaga T: Diverse spectrum of rare deafness genes underlies early-childhood hearing loss in Japanese patients: A cross-sectional, multi-center next-generation sequencing study. Orphanet J Rare Dis. 8:1722013. View Article : Google Scholar : PubMed/NCBI

24 

Huang S, Cai M, Zheng Y, Zhou L, Wang Q and Chen L: miR-888 in MCF-7 side population sphere cells directly targets E-cadherin. J Genet Genomics. 41:35–42. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Dong X, Han Y, Sun Z and Xu J: Actin Gamma 1, a new skin cancer pathogenic gene, identified by the biological feature-based classification. J Cell Biochem. 119:1406–1419. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Gan TQ, Xie ZC, Tang RX, Zhang TT, Li DY, Li ZY and Chen G: Clinical value of miR-145-5p in NSCLC and potential molecular mechanism exploration: A retrospective study based on GEO, qRT-PCR, and TCGA data. Tumour Biol. 39:10104283176916832017. View Article : Google Scholar : PubMed/NCBI

27 

Liu Y, Zhang Y, Wu H, Li Y, Zhang Y, Liu M, Li X and Tang H: miR-10a suppresses colorectal cancer metastasis by modulating the epithelial-to-mesenchymal transition and anoikis. Cell Death Dis. 8:e27392017. View Article : Google Scholar : PubMed/NCBI

28 

Sun Q, Wang Y, Zhang Y, Liu F, Cheng X, Hou N, Zhao X and Yang X: Expression profiling reveals dysregulation of cellular cytoskeletal genes in HBx–induced hepatocarcinogenesis. Cancer Biol Ther. 6:668–674. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Xu YY, Chen L, Wang GL, Zhou JM, Zhang YX, Wei YZ, Zhu YY and Qin J: A synthetic dsRNA, as a TLR3 pathwaysynergist, combined with sorafenib suppresses HCC in vitro and in vivo. BMC Cancer. 13:5272013. View Article : Google Scholar : PubMed/NCBI

30 

Zhang SY, Herman M, Ciancanelli MJ, Pérez de Diego R, Sancho-Shimizu V, Abel L and Casanova JL: TLR3 immunity to infection in mice and humans. Curr Opin Immunol. 25:19–33. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Chen C, Feng Y, Zou L, Wang L, Chen HH, Cai JY, Xu JM, Sosnovik DE and Chao W: Role of extracellular RNA and TLR3-Trif signaling in myocardial ischemia-reperfusion injury. J Am Heart Assoc. 3:e0006832014. View Article : Google Scholar : PubMed/NCBI

32 

Chen XL, Xu YY, Chen L, Wang GL and Shen Y: TLR3 plays significant roles against HBV-associated HCC. Gastroenterol Res Prac. 2015:5721712015.

33 

Das A, Chai JC, Kim SH, Lee YS, Park KS, Jung KH and Chai YG: Transcriptome sequencing of microglial cells stimulated with TLR3 and TLR4 ligands. BMC Genomics. 16:5172015. View Article : Google Scholar : PubMed/NCBI

34 

Szeles L, Meissner F, Dunand-Sauthier I, Thelemann C, Hersch M, Singovski S, Haller S, Gobet F, Fuertes Marraco SA, Mann M, et al: TLR3-mediated CD8+ dendritic cell activation is coupled with establishment of a cell-intrinsic antiviral state. J Immunol. 195:1025–1033. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Qiu X, Dong Y, Cao Y and Luo Y: Correlation between TLR2, TLR3, TLR4, and TLR9 polymorphisms and susceptibility to and prognosis of severe hepatitis among the newborns. J Clin Lab Anal. 32:2018. View Article : Google Scholar

36 

Yuan MM, Xu YY, Chen L, Li XY, Qin J and Shen Y: TLR3 expression correlates with apoptosis, proliferation and angiogenesis in hepatocellular carcinoma and predicts prognosis. BMC Cancer. 15:2452015. View Article : Google Scholar : PubMed/NCBI

37 

Kar P, Kumar D, Gumma PK, Chowdhury SJ and Karra VK: Down regulation of TRIF TLR3, and MAVS in HCV infected liver correlates with the outcome of infection. J Med Virol. 89:2165–2172. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Hu X, Ye J, Qin A, Zou H, Shao H and Qian K: Both microRNA-155 and virus-encoded MiR-155 ortholog regulate TLR3 expression. PLoS One. 10:e01260122015. View Article : Google Scholar : PubMed/NCBI

39 

Ho V, Lim TS, Lee J, Steinberg J, Szmyd R, Tham M, Yaligar J, Kaldis P, Abastado JP and Chew V: TLR3 agonist and Sorafenib combinatorial therapy promotes immune activation and controls hepatocellular carcinoma progression. Oncotarget. 6:27252–27266. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Guo Z, Chen L, Zhu Y, Zhang Y, He S, Qin J, Tang X, Zhou J and Wei Y: Double-stranded RNA-induced TLR3 activation inhibits angiogenesis and triggers apoptosis of human hepatocellular carcinoma cells. Oncol Rep. 27:396–402. 2012.PubMed/NCBI

41 

Yoneda K, Sugimoto K, Shiraki K, Tanaka J, Beppu T, Fuke H, Yamamoto N, Masuya M, Horie R, Uchida K and Takei Y: Dual topology of functional Toll-like receptor 3 expression in human hepatocellular carcinoma: Differential signaling mechanisms of TLR3-induced NF-kappaB activation and apoptosis. Int J Oncol. 33:929–936. 2008.PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 17 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao B, Li S, Tan Z, Ma L and Liu J: ACTG1 and TLR3 are biomarkers for alcohol‑associated hepatocellular carcinoma. Oncol Lett 17: 1714-1722, 2019
APA
Gao, B., Li, S., Tan, Z., Ma, L., & Liu, J. (2019). ACTG1 and TLR3 are biomarkers for alcohol‑associated hepatocellular carcinoma. Oncology Letters, 17, 1714-1722. https://doi.org/10.3892/ol.2018.9757
MLA
Gao, B., Li, S., Tan, Z., Ma, L., Liu, J."ACTG1 and TLR3 are biomarkers for alcohol‑associated hepatocellular carcinoma". Oncology Letters 17.2 (2019): 1714-1722.
Chicago
Gao, B., Li, S., Tan, Z., Ma, L., Liu, J."ACTG1 and TLR3 are biomarkers for alcohol‑associated hepatocellular carcinoma". Oncology Letters 17, no. 2 (2019): 1714-1722. https://doi.org/10.3892/ol.2018.9757