Skip to main content
  • Research article
  • Open access
  • Published:

Prognostic implications of alcohol dehydrogenases in hepatocellular carcinoma

Abstract

Background

Hepatocellular carcinoma (HCC) is a malignancy with high incidence and mortality rates worldwide. Alcohol dehydrogenases (ADHs) are huge family of dehydrogenase enzymes and associated with the prognosis of various cancers. However, comprehensive analysis of prognostic implications related to ADHs in HCC is still lacking and largely unknown.

Methods

The expression profiles and corresponding clinical information of HCC were obtained from The Cancer Genome Atlas (TCGA). Wilcoxon signed-rank test was employed to evaluate the expression of ADHs. Cox regression and Kaplan-Meier analyses were used to investigate the association between clinicopathological characteristics and survival. GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analyses were performed and visualized using R/BiocManager package.

Results

We found that the expression of ADH1A, ADH1B, ADH1C, ADH4, and ADH6 was significantly downregulated in HCC samples compared to normal liver samples. Our univariate and multivariate Cox regression analyses results showed that high expression of ADH1A, ADH1B, ADH1C, ADH4, and ADH6 was considered as an independent factor with an improved prognosis for the survival of HCC patients. Moreover, our Kaplan-Meier analysis results also revealed that high expression of AHD1A, ADH1B, ADH1C, ADH4, and ADH6 was significantly associated with good survival rate in HCC patients. In addition, GO, KEGG, and GSEA analyses unveiled several oncogenic signaling pathways were negatively associated high expression of ADHs in HCC.

Conclusion

In the present study, our results provide the potential prognostic biomarkers or molecular targets for the patients with HCC.

Peer Review reports

Background

Liver cancer is the sixth most common diagnosed cancer and the fourth most common cause of cancer-related deaths, which leads to about 841,080 (4.7%) new cases diagnosed and 781,631 (8.2%) deaths according to global cancer statistics 2018 [1]. Hepatocellular carcinoma (HCC) is the primary liver cancers, and accounts for approximately 75–85% of liver cancers worldwide [1]. Over the past few decades, considerable molecules have been developed to diagnose and treat HCC; where these molecules have been applied and shown their efficacy in detection and treatment of the disease [2, 3]. However, most of HCC patients are diagnosed at advanced stages resulting in the increasing mortality and poor prognosis in many parts of the world [4]. Therefore, identification of novel diagnostic biomarkers for early diagnosis of HCC is urgently needed.

Increasing evidence revealed that hepatitis B and C virus infection drove most of the global burden of HCC and accounted for 80% of HCC cases [5]. Moreover, it has been well documented the association between liver cirrhosis and the development of HCC [6]. In Europe and USA, alcoholic cirrhosis is considered as the second most important risk factor for HCC [7]. Therefore, alcohol is recognized as a primary cause or cofactor for the development of HCC in the patients who are heavy alcoholic [8, 9]. In liver, alcohol is primarily metabolized into acetaldehyde, which is also carcinogenic, through alcohol dehydrogenases [10]. Several studies have demonstrated that ADHs are the major rate-limiting factors in the process of alcohol metabolism [11].

Alcohol dehydrogenases (ADHs) are super family of dehydrogenase enzymes located on chromosome 4q22-q24, including class I (ADH1A, ADH1B, and ADH1C), class II (ADH4), class III (ADH5), class IV (ADH6), and class V (ADH7) [12]. ADH family members are widely expressed in several human tissues; interestingly, ADH7 is the only one which is not expressed in human liver [12, 13]. Previous studies have reported that members of ADH gene family are associated with various cancers [14, 15], and the genetic variation of ADHs also affects the risk of cancer for alcohol dependent individuals [16,17,18]. Recent investigations have shown the prognostic values of ADH family members for non-small cell lung cancer and gastric cancer [19, 20]. However, to the best of our knowledge, the prognostic value of ADH family members in HCC is still unclear. Therefore, the aim of our present study was to explore the potential prognostic value of ADH genes for HCC patients.

Methods

Data acquisition

The transcriptomic profiles and clinicopathological information of Liver Hepatocellular Carcinoma in TCGA (The Cancer Genome Atlas) were obtained from GDC (Genomic Data Commons Data Portal) website (https://portal.gdc.cancer.gov/). For transcriptomic data, FPKM (Fragments Per Kilobase of transcript per Million mapped reads) was used as a unit representing the gene expression levels. Tumor samples, which have full clinicopathological characteristics including age, gender, histologic grade, pathologic T stage, vital status, OS (overall survival), alcohol consumption status, recurrence status (new tumor event after initial treatment) and days to new tumor event after initial treatment, were selected for further analyses, resulting in 269 samples. RFS (recurrence free survival) was calculated depending on recurrence status and days to new tumor event after initial treatment. GSEA (Gene Set Enrichment Analysis) was performed with the JAVA program GSEA 4.0 and annotated gene set database including KEGG “c2.cp.kegg.v7.0.symbols.gmt” and PID “c2.cp.pid.v7.0.symbols.gmt” were chosen as the reference gene sets (minimal set size = 15, maximal set size = 500) [21]. The results were shown in the form of multiple-GSEA using R packages including plyr, ggplot2, grid, and gridExtra.

Statistical analysis

Wilcoxon signed-rank test was employed to compare the statistical significance of ADHs’ expression in different samples. Cases were categorized into high and low expression subgroups based on Youden index calculation according to ADHs’ expression levels [22]. Survival analysis of cases was performed on Kaplan-Meier method with a log-rank test. The association between clinicopathological characteristics and survival was carried out by univariate Cox regression and multivariate Cox regression analyses with HR (Hazard Ratio) and 95% CI (Confidence Intervals). GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analyses were performed and visualized using R/BiocManager package [23, 24]. Analyses were performed with R version 3.6.2 [25] and Graphpad Prism 5.0 (GraphPad Software, Inc., USA). All statistical analyses with a p-value < 0.05 were considered to be significant, and GSEA gene sets with p-value < 0.05 and a false discovery rate (FDR) < 0.25 were considered as a significantly enrichment.

Results

Gene expression signatures of ADHs across different HCC samples

In order to distinguish the expression levels of ADHs between normal and tumor liver tissues, the transcriptome of 50 normal liver and 269 HCC samples was identified. As shown in Fig. 1, the expression level of ADH1A, ADH1B, ADH1C, ADH4, and ADH6 was significantly downregulated in HCC samples compared to normal liver samples. Interestingly, the expression level of ADH5 was slightly but significantly upregulated in HCC samples (Fig. 1e). Furthermore, all the ADH genes showed a positive correlation with each other (Fig. 2).

Fig. 1
figure 1

Gene expression level of ADHs in normal liver and HCC tissues. The gene expression level of ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f) in normal liver (n = 50) and HCC (n = 269) tissue samples. The relative expression level of ADHs in each sample was shown as log2FPKM, all data were collected from TCGA. Data shown represent box and whisker plots (whiskers = 10–90 percentile). The significant difference was calculated with Wilcoxon signed-rank test for gene expression in normal liver and HCC tissue samples, p < 0.05 was recognized as significant difference, and the significant differences were denoted by **** for p < 0.0001

Fig. 2
figure 2

Co-expression of ADHs in HCC patients. Pearson correlation analysis was performed to characterize the co-expression of ADHs in HCC patients (n = 269). The relative expression level of ADHs in each sample was shown as log2FPKM, all data were collected from TCGA. p < 0.05 was recognized as significant difference

Alcohol consumption increases the risk for liver cancer, and is also considered as a primary cause of HCC through the development of cirrhosis [26]. In the present study, our data showed that the expression level of ADH family members including ADH1A-ADH6 was significantly increased in alcohol consumption HCC patients compared to non-alcohol consumption ones (Fig. 3). Obviously, the expression of ADH1B and ADH4 was strongly upregulated (Fig. 3b and d).

Fig. 3
figure 3

Gene expression level of ADHs in HCC patients with alcohol consumption and non-alcohol consumption. The gene expression level of ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f) in non-alcohol consumption HCC patients (No, n = 175) and alcohol consumption HCC patients (Yes, n = 94). The relative expression level of ADHs in each sample was shown as log2FPKM, all data were collected from TCGA. Data shown represent box and whisker plots (whiskers = 10–90 percentile). The significant difference was calculated with Wilcoxon signed-rank test for gene expression in non- and alcohol consumption HCC patients, p < 0.05 was recognized as significant difference, and the significant differences were denoted by **** for p < 0.0001

In order to characterize the correlation between ADHs and tumor stages, the expression level of ADHs was identified in HCC samples with different pathologic T stages. Our results showed that the expression level of ADHs including ADH1A, ADH1B, ADH1C, and ADH6 was obviously decreased with the progression of tumor malignancy (Fig. 4a-c and f). And the expression level of ADH4 was higher in HCC patients with T2 pathologic stage than T1 and T3 pathologic stage, but it was lowest in HCC samples with T3 pathologic stage (Fig. 4d). While the expression level of ADH5 was remarkably increased with the progression of tumor malignancy (Fig. 4e).

Fig. 4
figure 4

Gene expression level of ADHs in HCC patients with different pathologic T stage. The gene expression level of ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f) in HCC patients with pathologic T1 (n = 133), T2 (n = 62) and T3 (n = 63) stage. The relative expression level of ADHs in each sample was shown as log2FPKM, all data were collected from TCGA. Data shown represent box and whisker plots (whiskers = 10–90 percentile). The significant difference was calculated with Wilcoxon signed-rank test for gene expression in different pathologic T stage HCC tissue samples, p < 0.05 was recognized as significant difference, and the significant differences were denoted by **** for p < 0.0001

Association between ADHs expression and survival time of HCC patients

In order to explore the prognostic value of ADHs expression level in the patients with HCC, all the patients were categorized into high and low expression groups based on the expression level of ADHs as described above. Primarily, the association of ADHs expression and clinicopathological characteristics was determined with two-sided Fisher’s exact tests. Our results showed that the expression of ADH1B, ADH4, and ADH5 was significantly associated with age. Moreover, the expression of ADH1A, ADH1B, and ADH4 was significantly associated with histologic grade. In addition, ADH1A, ADH1B and ADH4 expressions were significantly associated with pathologic T stage (Table 1).

Table 1 Association analysis between ADHs expression and clinicopathological variables in 269 HCC patients

Furthermore, univariate and multivariate Cox regression analyses were also performed to assess the prognostic value of ADHs expression and clinicopathological characteristics. As shown in Table 2, univariate Cox regression analysis results presented that high pathologic T stage, alcohol consumption, tumor recurrence were significantly associated with poor OS for HCC patients. Moreover, higher pathologic T stage and tumor recurrence were also significantly associated with poor RFS. Interestingly, high expression of ADH1A, ADH1C, ADH4, and ADH6 was considered as an independent factor with an improved prognosis for OS and RFS. While high expression of ADH1B was only considered as an independent factor with an improved prognosis for RFS (Table 2). In addition, our multivariate Cox regression analysis results showed that higher pathologic T stage and tumor recurrence were also significantly associated with poor OS and RFS (Figs. 5 and 6). Interestingly, high expression of ADH1A, ADH1C, ADH4, and ADH6 was also considered as an independent factor with an improved prognosis for OS (Fig. 5). And high expression of ADHs without ADH5 was considered as an independent factor with an improved prognosis for RFS (Fig. 6).

Table 2 Univariate analysis of the expression of ADHs expression with OS and RFS in 269 patients with HCC
Fig. 5
figure 5

Multivariate Cox regression analysis for overall survival in HCC patients. Forest-plot showing the hazard ratio (HR) with 95% confidence index (CI) and p value for overall survival (OS) in patients with HCC (n = 269) based on high versus low expression of ADH family members, including ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f). p < 0.05 was considered as significant difference

Fig. 6
figure 6

Multivariate Cox regression analysis for recurrence free survival in HCC patients. Forest-plot showing the HR with 95%CI and p value for recurrence free survival (RFS) in patients with HCC (n = 269) based on high versus low expression of ADH family members, including ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f). p < 0.05 was considered as significant difference

Next, the correlation between ADHs expression and OS/RFS was evaluated with Kaplan-Meier analysis and log-rank test. Our results showed that high expression of AHD1A, ADH1C, ADH4, and ADH6 was significantly associated with good OS and RFS in HCC patients (Figs. 7 and 8). Interestingly, the RFS rate of HCC patients with high ADH1B expression was significantly better than that of patients with low ADH1B expression (Fig. 8b).

Fig. 7
figure 7

Overall survival analysis in HCC patients. Kaplan-Meier survival plots representing the probability of OS for HCC patients (n = 269) according to the expression level of ADH family members, including ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f). p < 0.05 was considered as significant difference

Fig. 8
figure 8

Recurrence free survival analysis in HCC patients. Kaplan-Meier survival plots representing the probability of RFS for HCC patients (n = 269) according to the expression level of ADH family members, including ADH1A (a), AHD1B (b), ADH1C (c), ADH4 (d), ADH5 (e), and ADH6 (f). p < 0.05 was considered as significant difference

Identification of involved pathways related to ADHs expression in HCC

In order to identify the potential biological functions of ADHs, GO terms and KEGG pathways enrichment analysis was performed with R project as above description. GO terms enrichment analysis revealed that plenty of pathways were well enriched, and 9 of them including ethanol oxidation, ethanol metabolic process, primary alcohol metabolic process, retinoid metabolic process, diterpenoid metabolic process, terpenoid metabolic process, isoprenoid metabolic process, antibiotic metabolic process, and alcohol metabolic process were enriched for all the members of ADHs family (Fig. 9a and Table S1). Moreover, KEGG pathways enrichment analysis results showed that 7 pathways were significantly enriched, including tyrosine metabolism, fatty acid degradation, retinol metabolism, glycolysis/gluconeogenesis, drug metabolism-cytochrome P450, metabolism of xenobiotics by cytochrome P450, chemical carcinogenesis (Fig. 9b and Table S2).

Fig. 9
figure 9

Functional enrichment analysis of ADH family members. a Dot-plot showing the significant enrichment terms (p < 0.05) in the Gene Ontology of ADH family members. b Dot-plot showing the significant enrichment pathways (p < 0.05) in the KEGG pathway of ADH family members

In order to investigate the role of ADHs in the pathogenesis of HCC, GSEA was performed between datasets with ADHs high expression and low expression. Our results unveiled that ADHs were enriched in serval signaling pathways (Table S3 and S4). Based on the enrichment with KEGG database, the high expression group of ADHs was positively associated with retinol metabolism, and fatty acid metabolism with the significant difference (Fig. 10a and b). And the low expression group of ADHs family members not including ADH5 was significantly but negatively associated with pathways in cancer (Fig. 10c). Furthermore, we utilized GSEA to enrich in cancer related pathways based on pathway interaction database (PID). As can be seen from Fig. 10d-i, low expression group of ADHs family members without ADH5 was significantly enriched in several cancer related pathways, including ATR, FOXM1, FOXO, MTOR, NOTCH, and P53 downstream pathway.

Fig. 10
figure 10

Gene set enrichment analysis of ADHs in HCC patients. Representative multi-GSEA plots showing ADHs high and low expression group in KEGG retinol metabolism (a), KEGG fatty acid metabolism (b), KEGG pathways in cancer (c), PID ATR pathway (d), PID FOXM1 pathway (e), PID FOXO pathway (f), PID MTOR 4pathway (g), PID NOTCH pathway (h), and PID P53 downstream pathway (i). GSEA gene sets with p < 0.05 and FDR < 0.25 were considered as a significant difference

Discussion

HCC is a malignant tumor with high incidence and mortality rates worldwide [27]. The occurrence and progression of HCC are complex process, which is modulated through various numbers of oncogenes and anti-tumor genes. ADHs are huge family covering 7 members that are mainly involved in the conversion between alcohol and acetaldehyde, and also correlated to several hepatic diseases [14, 28]. However, the prognostic value of ADHs in the patients with HCC is still unclear. In the present study, we performed comprehensive analysis to investigate ADH genes association with the progression and prognosis of the patients with HCC, and to explore a series of diagnostic biomarkers of HCC.

The ADH family members are widely expressed in human liver without ADH7 [12, 13]. Therefore, ADH1A-ADH6 were selected to evaluate the prognostic value of ADHs in the patients with HCC. Primarily, our results revealed that the expression levels of ADH1A-ADH4, and ADH6 were significantly decreased in HCC tissues compared to normal liver tissues (Fig. 1), which were similar to their expression in non-small cell lung cancer (NSCLC) and pancreatic adenocarcinoma [19, 29]. Therefore, we speculate that ADH1A-ADH4, and ADH6 may serve as tumor suppressors in HCC. Whereas, the expression of ADH5 were significantly upregulated in HCC tissues compared to normal liver tissues (Fig. 1e), consistent with what is already known in NSCLC, gastric cancer, and pancreatic adenocarcinoma [19, 20, 29]. In addition, previous studies have reported the positive correlation of ADHs with each other in numerous cancers [19, 20]; here, our results also confirmed that in HCC (Fig. 2).

Smoking is an important risk factor for lung cancer, a recent study reported that the expression levels of ADHs without ADH1A were significantly associated with smoking status of the NSCLC patients [19]. Interestingly, alcohol consumption status is considered as a primary cause for HCC; here, our results revealed that the expression levels of ADH1A-ADH6 were obviously increased in alcohol consumption HCC patients (Fig. 3). Moreover, accumulating evidences have demonstrated that ADH family members were correlated with clinical stages, pathological grades, and TNM classifications in several cancers [19, 29, 30]. In our present study, it showed that the expression levels of ADH1A-ADH1C, and ADH6 were remarkably downregulated according to the pathologic T stage progression of HCC (Fig. 4). In addition, the expression of ADH1A, ADH1B and ADH4 was significantly associated with pathologic T stage (Table 1). Altogether, ADH family members play an important role in the development process of HCC. It is meaningful to evaluate the prognostic value for HCC patients.

In recent years, large numbers of investigations have reported that gene polymorphism of ADHs was correlated with cancer risk [15, 17, 18, 31, 32] . Moreover, the activity of ADH isoenzymes was significantly higher in liver cancer tissues than in healthy tissues [33], suggested the diagnostic value of ADH for the patients with liver cancer. However, rare studies have been performed to evaluate the prognostic value of ADHs mRNA expression in HCC. In the present study, our univariate and multivariate Cox regression analyses results indicated that high ADH1A, ADH1C, ADH4, and ADH6 levels independently predicted improved OS and RFS in HCC patients; whereas, high ADH1B levels independently predicted improved RFS in HCC patients (Table 2, Figs. 5 and 6). Furthermore, our Kaplan-Meier analysis data also revealed that high ADH1A, ADH1C, ADH4, and ADH6 levels predicted good OS and RFS in HCC patients; while, high ADH1B only predicted good RFS in HCC patients (Figs. 7 and 8). Recently, the expression of ADH1A was measured by using MS/MS and TMA in CHCC-HBV patients, which indicated the robust prognostic value of ADH1A for potential clinical application [34]. Moreover, Chen Q, et al also reported that high expression of ADH1C was associated with a good prognosis for HCC patients by using the TCGA internal and three GEO (GSE76427, GSE15654, and GSE14520) external validation cohorts [35]. In addition, the prognostic value of ADH4 was also confirmed by immunohistochemical analysis with 91 paraffin-embedded HCC specimens [36]. Although, a recent study reported that decreased ADH5 expression in HBV-related HCC tumor tissue predicted earlier recurrence [30]. Surprisingly, ADH5 expression could not play a significant role in prediction of OS and RFS in HCC patients depending on our present data. Therefore, our data supposed that ADH family members without ADH5 might serve as the potential biomarkers for the patients with HCC.

ADHs play a pivotal role in the metabolic process of ethanol [26]. Our GO enrichment analysis showed that ADHs were contributed to ethanol metabolism, such as ethanol oxidation, ethanol metabolic process, and primary alcohol metabolic process. Moreover, KEGG enrichment analysis indicated that ADHs were involved in fatty acid degradation, retinol metabolism, and so on (Fig. 9), constitute with what is investigated using GSEA (Fig. 10a and b). In addition, our GSEA results also showed that high expression group of ADHs was significantly and negatively associated with pathways in cancer without ADH5 (Fig. 10c), which suggested that high expression of ADHs could inhibit cancer related pathways and ADHs presented the tumor suppressor role. Increasing evidences indicated that various numbers of signaling pathways participated in the progression of HCC [37]. In order to determine the signaling pathways related to ADHs in HCC patients, GSEA was performed based on PID dataset [38]. Our results showed that low expression group of ADHs without ADH5 was positively related to various pathways (Fig. 10, Table S3 and S4). Interestingly, most of the signaling pathways contributed to promote tumorigenesis, such as ATR pathway [39], FOXM1 and FOXO pathways [40, 41], MTOR pathway [42], NOTCH pathway [43], and P53 downstream pathway [44]. Accordingly, high expression of ADHs could inhibit the malignant process of HCC, which promote the OS and RFS probability of HCC patients. Although low expression group of ADH5 was positively related to oncogenic signaling pathways, such as ATR, FOXM1, MTOR, NOTCH, and P53 downstream pathways, the statistic difference was not significant. Interestingly, low expression group of ADH5 was not positively related to FOXO signaling pathway, which may be associated with the high expression of ADH5 in HCC. By summarizing all the points, ADHs without ADH5 might act as the tumor suppressor via inhibiting oncogenic signaling pathway in HCC.

There were some limitations in our present study, which should be known. Primarily, the clinicopathological information of HCC patients from TCGA website, such as tumor size, hepatitis virus infection, α-fetoprotein, non-alcoholic fatty liver and cirrhosis, was not comprehensive or lost. Secondly, our present study investigates the prognostic value of ADHs for HCC patients at mRNA levels, which is not consummate. Therefore, their prognostic value for HCC patients at protein levels should be further evaluated in our future studies. Thirdly, ADH1A, ADH1B, ADH1C, ADH4, and ADH6 were all associated with the prognosis of HCC patients. In the future, the in vitro and in vivo experiments should be performed to investigate their roles in the progress of HCC, then the best one of them could be selected as diagnostic and prognostic biomarkers for HCC patients. In addition, further effort is also needed to elucidate the mechanisms, which ADHs contribute to the progress of HCC in the future.

Conclusions

In the present study, our results were to explore the expression levels of ADHs in different grouping HCC patients, and also evaluated the correlation between prognosis of HCC and the expression patterns of ADHs. We found that the expression of ADH1A, ADH1B, ADH1C, ADH4, and ADH6 was significantly downregulated in HCC samples compared to normal liver samples; but upregulated in alcohol consumption HCC patients. Moreover, high expression of ADH1A, ADH1B, ADH1C, ADH4, and ADH6 was associated with good prognosis for HCC patients. Therefore, our results provide the potential prognostic biomarkers or molecular targets for the patients with HCC.

Availability of data and materials

The datasets analyzed in the current study are available in the TCGA repository (https://portal.gdc.cancer.gov/repository).

Abbreviations

ADHs:

Alcohol dehydrogenases

ATR:

Ataxia telangiectasia and rad3-related

CHCC-HBV:

Chinese HCC patients with HBV infection

CI:

Confidence interval

FOX:

Forkhead box

GO:

Gene ontology

GSEA:

Gene set enrichment analysis

HCC:

Hepatocellular carcinoma

HR:

Hazard ratio

KEGG:

Kyoto encyclopedia of genes and genomes

MS:

Mass spectrometry

MTOR:

Mammalian target of rapamycin

NSCLC:

Non-small cell lung cancer

OS:

Overall survival

PID:

Pathway interaction database

RFS:

Recurrence-free survival

TCGA:

The Cancer Genome Atlas

TMA:

Tissue microarray

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.

    Google Scholar 

  2. Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol. 2019;16(10):589–604.

    Article  Google Scholar 

  3. Singh AK, Kumar R, Pandey AK. Hepatocellular carcinoma: causes, mechanism of progression and biomarkers. Curr Chem Genom Transl Med. 2018;12:9–26.

    Article  CAS  Google Scholar 

  4. Kulik L, El-Serag HB. Epidemiology and management of hepatocellular carcinoma. Gastroenterology. 2019;156(2):477–91 e1.

  5. Yang JD, Roberts LR. Hepatocellular carcinoma: a global view. Nat Rev Gastroenterol Hepatol. 2010;7(8):448–58.

    Article  Google Scholar 

  6. Kanda T, Goto T, Hirotsu Y, Moriyama M, Omata M. Molecular mechanisms driving progression of liver cirrhosis towards hepatocellular carcinoma in chronic hepatitis B and C infections: a review. Int J Mol Sci. 2019;20(6):1358.

    Article  CAS  Google Scholar 

  7. Park JW, Chen M, Colombo M, Roberts LR, Schwartz M, Chen PJ, et al. Global patterns of hepatocellular carcinoma management from diagnosis to death: the BRIDGE study. Liver Int. 2015;35(9):2155–66.

    Article  Google Scholar 

  8. Morgan TR, Mandayam S, Jamal MM. Alcohol and hepatocellular carcinoma. Gastroenterology. 2004;127(5 Suppl 1):S87–96.

    Article  CAS  Google Scholar 

  9. Testino G, Leone S, Borro P. Alcohol and hepatocellular carcinoma: a review and a point of view. World J Gastroenterol. 2014;20(43):15943–54.

    Article  CAS  Google Scholar 

  10. Seitz HK, Stickel F. Acetaldehyde as an underestimated risk factor for cancer development: role of genetics in ethanol metabolism. Genes Nutr. 2010;5(2):121–8.

    Article  CAS  Google Scholar 

  11. Cederbaum AI. Alcohol metabolism. Clin Liver Dis. 2012;16(4):667–85.

    Article  Google Scholar 

  12. Edenberg HJ, McClintick JN. Alcohol dehydrogenases, aldehyde dehydrogenases, and alcohol use disorders: a critical review. Alcohol Clin Exp Res. 2018;42(12):2281–97.

    Article  CAS  Google Scholar 

  13. Jairam S, Edenberg HJ. Single-nucleotide polymorphisms interact to affect ADH7 transcription. Alcohol Clin Exp Res. 2014;38(4):921–9.

    Article  CAS  Google Scholar 

  14. Jelski W, Zalewski B, Szmitkowski M. Alcohol dehydrogenase (ADH) isoenzymes and aldehyde dehydrogenase (ALDH) activity in the sera of patients with liver cancer. J Clin Lab Anal. 2008;22(3):204–9.

    Article  CAS  Google Scholar 

  15. Hashibe M, Mckay JD, Curado MP, Oliveira JC, Koifman S, Koifman R, et al. Multiple ADH genes are associated with upper aerodigestive cancers. Nat Genet. 2008;40(6):707–9.

    Article  CAS  Google Scholar 

  16. Ding J, Li S, Wu J, Gao C, Zhou J, Cao H, et al. Alcohol dehydrogenase-2 and aldehyde dehydrogenase-2 genotypes, alcohol drinking and the risk of primary hepatocellular carcinoma in a Chinese population. Asian Pac J Cancer Prev. 2008;9(1):31–5.

    CAS  PubMed  Google Scholar 

  17. Abe H, Aida Y, Seki N, Sugita T, Tomita Y, Nagano T, et al. Aldehyde dehydrogenase 2 polymorphism for development to hepatocellular carcinoma in east Asian alcoholic liver cirrhosis. J Gastroenterol Hepatol. 2015;30(9):1376–83.

    Article  CAS  Google Scholar 

  18. Homann N, Stickel F, Konig IR, Jacobs A, Junghanns K, Benesova M, et al. Alcohol dehydrogenase 1C*1 allele is a genetic marker for alcohol-associated cancer in heavy drinkers. Int J Cancer. 2006;118(8):1998–2002.

    Article  CAS  Google Scholar 

  19. Wang P, Zhang L, Huang C, Huang P, Zhang J. Distinct prognostic values of alcohol dehydrogenase family members for non-small cell lung cancer. Med Sci Monit. 2018;24:3578–90.

    Article  CAS  Google Scholar 

  20. Guo E, Wei H, Liao X, Xu Y, Li S, Zeng X. Prognostic value of alcohol dehydrogenase mRNA expression in gastric cancer. Oncol Lett. 2018;15(4):5505–16.

    PubMed  PubMed Central  Google Scholar 

  21. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50.

    Article  CAS  Google Scholar 

  22. Cao H, Chu X, Wang Z, Guo C, Shao S, Xiao J, et al. High FOXK1 expression correlates with poor outcomes in hepatocellular carcinoma and regulates stemness of hepatocellular carcinoma cells. Life Sci. 2019;228:128–34.

    Article  CAS  Google Scholar 

  23. Carlson M. GO.db: A set of annotation maps describing the entire Gene Ontology. R package version 3.8.2. 2019.

  24. Carlson M. KEGG. db: a set of annotation maps for KEGG. R package version 3.1.2. 2016.

  25. Team RC. R: a language and environment for statistical computing; 2013.

    Google Scholar 

  26. Seitz HK, Becker P. Alcohol metabolism and cancer risk. Alcohol Res Health. 2007;30(1):38–41 44-37.

    PubMed  PubMed Central  Google Scholar 

  27. Jemal A, Center MM, DeSantis C, Ward EM. Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev. 2010;19(8):1893–907.

    Article  Google Scholar 

  28. Gao N, Li J, Li MR, Qi B, Wang Z, Wang GJ, et al. Higher activity of alcohol dehydrogenase is correlated with hepatic fibrogenesis. J Pharmacol Exp Ther. 2018;367(3):473–82.

    Article  CAS  Google Scholar 

  29. Liao X, Huang R, Liu X, Han C, Yu L, Wang S, et al. Distinct prognostic values of alcohol dehydrogenase mRNA expression in pancreatic adenocarcinoma. Onco Targets Ther. 2017;10:3719–32.

    Article  Google Scholar 

  30. Shang L, Zhu G, Su H, Chen B, Ye X, Chen X, et al. Identification of alcohol dehydrogenase as a potential prognostic marker in HBV-related hepatocellular carcinoma. Int J Clin Exp Med. 2017;10(3):4457–72.

    CAS  Google Scholar 

  31. Mohelnikova-Duchonova B, Vrana D, Holcatova I, Ryska M, Smerhovsky Z, Soucek P. CYP2A13, ADH1B, and ADH1C gene polymorphisms and pancreatic cancer risk. Pancreas. 2010;39(2):144–8.

    Article  CAS  Google Scholar 

  32. Ghosh S, Bankura B, Ghosh S, Saha ML, Pattanayak AK, Ghatak S, et al. Polymorphisms in ADH1B and ALDH2 genes associated with the increased risk of gastric cancer in West Bengal, India. BMC Cancer. 2017;17(1):782.

    Article  Google Scholar 

  33. Jelski W, Zalewski B, Szmitkowski M. The activity of class I, II, III, and IV alcohol dehydrogenase (ADH) isoenzymes and aldehyde dehydrogenase (ALDH) in liver cancer. Dig Dis Sci. 2008;53(9):2550–5.

    Article  CAS  Google Scholar 

  34. Gao Q, Zhu H, Dong L, Shi W, Chen R, Song Z, et al. Integrated proteogenomic characterization of HBV-related hepatocellular carcinoma. Cell. 2019;179(2):561–77 e22.

  35. Chen Q, Li F, Gao Y, Xu G, Liang L, Xu J. Identification of energy metabolism genes for the prediction of survival in hepatocellular carcinoma. Front Oncol. 2020;10:1210.

    Article  Google Scholar 

  36. Wei RR, Zhang MY, Rao HL, Pu HY, Zhang HZ, Wang HY. Identification of ADH4 as a novel and potential prognostic marker in hepatocellular carcinoma. Med Oncol. 2012;29(4):2737–43.

    Article  CAS  Google Scholar 

  37. Blagotinsek K, Rozman D. Targeting signalling pathways in hepatocellular carcinoma. Curr Pharm Des. 2017;23(1):170–5.

    CAS  PubMed  Google Scholar 

  38. Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, et al. PID: the pathway interaction database. Nucleic Acids Res. 2009;37(Database issue):D674–9.

    Article  CAS  Google Scholar 

  39. Karnitz LM, Zou L. Molecular pathways: targeting ATR in cancer therapy. Clin Cancer Res. 2015;21(21):4780–5.

    Article  CAS  Google Scholar 

  40. Raychaudhuri P, Park HJ. FoxM1: a master regulator of tumor metastasis. Cancer Res. 2011;71(13):4329–33.

    Article  CAS  Google Scholar 

  41. Lu M, Hartmann D, Braren R, Gupta A, Wang B, Wang Y, et al. Oncogenic Akt-FOXO3 loop favors tumor-promoting modes and enhances oxidative damage-associated hepatocellular carcinogenesis. BMC Cancer. 2019;19(1):887.

    Article  Google Scholar 

  42. Zahid KR, Yao S, Khan ARR, Raza U, Gou D. mTOR/HDAC1 crosstalk mediated suppression of ADH1A and ALDH2 links alcohol metabolism to hepatocellular carcinoma onset and progression in silico. Front Oncol. 2019;9:1000.

    Article  Google Scholar 

  43. Huang Q, Li J, Zheng J, Wei A. The carcinogenic role of the Notch signaling pathway in the development of hepatocellular carcinoma. J Cancer. 2019;10(6):1570–9.

    Article  Google Scholar 

  44. Tang Q, Su Z, Gu W, Rustgi AK. Mutant p53 on the path to metastasis. Trends Cancer. 2020;6(1):62–73.

    Article  Google Scholar 

Download references

Acknowledgments

The authors thank the contributions of the members of TCGA who provided samples and data used in the present study.

Funding

This work was supported by the National Natural Science Foundation of China (Grant No. 81802389), Qinglan Project in Jiangsu Province, and A Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). The funders have played no role in the research design, data collection and analysis, or writing of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

XYL, TTL, DLK, and RXT conceived and designed the experiments. XYL, DLK, and TTL analyzed the data. XYL, TTL, DLK, HJY, FYK and RXT contributed materials and analysis tools. XYL, TTL, DLK, HJY, FYK, and RXT wrote and reviewed the paper. The authors read and approved the final version of the manuscript.

Corresponding authors

Correspondence to Xiangye Liu or Renxian Tang.

Ethics declarations

Ethics approval and consent to participate

No ethics approval was required for the present study. The datasets analyzed in the present study are publicly available.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

Enrichment analysis of GO terms for ADHs.

Additional file 2: Table S2.

Enrichment analysis of KEGG pathways for ADHs.

Additional file 3: Table S3.

Signaling pathways enriched in HCC samples corresponding ADHs expression by GSEA based on KEGG.

Additional file 4: Table S4.

Signaling pathways enriched in HCC samples corresponding ADHs expression by GSEA based on PID.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X., Li, T., Kong, D. et al. Prognostic implications of alcohol dehydrogenases in hepatocellular carcinoma. BMC Cancer 20, 1204 (2020). https://doi.org/10.1186/s12885-020-07689-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-020-07689-1

Keywords