Open Access

Profiles of m6A RNA methylation regulators for the prognosis of hepatocellular carcinoma

  • Authors:
    • Wang Li
    • Qi‑Feng Chen
    • Tao Huang
    • Lujun Shen
    • Zi‑Lin Huang
    • Peihong Wu
  • View Affiliations

  • Published online on: March 3, 2020     https://doi.org/10.3892/ol.2020.11435
  • Pages: 3296-3306
  • Copyright: © Li 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

N6‑methyladenosine (m6A) RNA methylation, which is related to cancer initiation and progression, is dynamically regulated by the m6A RNA methylation regulators (including ‘writers’, ‘erasers’ and ‘readers’). However, the prognostic value of m6A RNA methylation regulators involved in hepatocellular carcinoma (HCC) carcinogenesis and progression remains to be elucidated. The aim of the present study was to determine the prognostic score in predicting the prognosis of HCC patients based on these regulators. In The Cancer Genome Atlas, most of the 13 major m6A RNA methylation regulators were found to be differentially expressed between HCC and normal samples (P<0.001). In addition, two subgroups (clusters 1/2) had also been identified by applying consensus clustering in the m6A RNA methylation regulators. As compared with the cluster 1 subgroup, the cluster 2 subgroup was correlated with a poorer prognosis, as shown by the Kaplan‑Meier method (P=6.197e‑4). A risk signature was constructed based on these findings using six m6A RNA methylation regulators, which could not only predict the clinicopathological features of HCCs, but also serve as an independent prognostic marker, as shown by Cox regression analysis (hazard ratio=1.219, 95% confidence interval: 1.143‑1.299; P<0.001). Data from the International Cancer Genome Consortium were used for external validation. In addition, gene set enrichment analysis identified several pathways that m6A RNA methylation regulators were closely associated with. In conclusion, the m6A RNA methylation regulators are the crucial participants in the malignant progression of HCCs, which are potentially useful for prognosis stratification and therapeutic strategy development for HCC.

Introduction

Like histone and DNA, the epigenetic modification of RNA species has been extensively reported over the past few decades (1). Since the 1950s, over 100 chemical modification types have been described in RNA, particularly in rRNA and tRNA (2). Of note, any micro-event during base modification can result in potent influence on the metabolic pathways, as well as the resulting organism phenotype alterations. As a result, the abnormal alteration can result in the occurrence of abnormalities and disease initiation like tumors (3,4).

The m6A modification has attracted wide attention in the field of epitranscriptomics, which is associated with the highest prevalence among transcripts (5,6). Thanks to the developments of recent technology, N6-methyladenosine (m6A) modifications in mRNA have been identified (7,8). m6A modification reveals an extensive, while rare, epitranscriptomic landscape, which participates in various physiological processes, including cancer (9).

There are 3 protein classes that can regulate m6A modification, the ‘reader’ (m6A-binding protein), ‘eraser’ (m6A demethylating enzyme) and ‘writer’ (adenosine methyltransferase) (3,10,11). Specifically, m6A modification can be subjected to reversible installment and removal by writers and erasers, separately. This process is dynamic and reversible. However, the deregulated m6A modification, which is associated with abnormal expression levels or functions of the m6A readers, erasers and writers, may result in cancer genesis and progression (12).

Hepatocellular carcinoma (HCC), one of the most frequently observed liver cancer types, is a severe worldwide health problem (13,14). Nonetheless, no existing study has comprehensively analyzed the expression levels of m6A RNA methylation regulators among HCCs that have various clinical and pathological features, or their role and prognosis significance in the malignant development of HCC. The present study carried out a systemic analysis on the expression levels of 13 extensively identified m6A RNA regulators in HCCs, according to the RNA sequencing information extracted from The Cancer Genome Atlas (TCGA) (n=377) database. In addition, the expression profiles for all 13 m6A modification regulators were provided based on various clinical and pathological characteristics. According to the present results, the expression levels of the m6A RNA methylation regulators played an important role during HCC malignant development. A signature was also constructed using 6 screened m6A RNA methylation regulators for HCC prognosis stratification. The constructed signature was further confirmed by the International Cancer Genome Consortium (ICGC) database.

Materials and methods

Data extraction

Data were downloaded from the TCGA database. Gene expression data and the clinical information of HCC patients (https://tcga-data.nci.nih.gov/) were downloaded using the Data Transfer Tool (provided by GDC Apps). A total of 374 tumor and 50 normal samples from 377 HCC patients were used in this study to analyze the differentially expressed m6A RNA methylation regulators. Typically, the list of the 13 m6A RNA methylation regulators was determined with reference to published literature (4). All data were publicly available and open-access; as a result, Ethics Committee approval was not required. Data were processed in accordance with the data access policies, as well as the TCGA Human Subject Protection system formulated by the National Institutes of Health (NIH; http://cancergenome.nih.gov/publications/publicationguidelines). The LIRI-JP project from the ICGC database was used as an independent validation cohort (n=237).

Bioinformatic analysis

First, the expression patterns of m6A RNA methylation regulators were compared between tumors and normal samples, and Spearman's rank correlation coefficient was used for correlation analysis among the regulators. In addition, the interactions between m6A RNA methylation regulators would be examined using the Search Tool for the Retrieval of Interacting Genes/Proteins database (http://www.string-db.org/). To investigate the function of m6A RNA methylation regulators in HCCs, HCCs were clustered in various groups using the ‘ConsensusClusterPlus’ (http://www.bioconductor.org/). In addition, gene expression profiles of the various HCC groups were investigated using principal component analysis (PCA) as well as R package. Moreover, the c2.cp.kegg.v6.2.symbols were examined based on gene set enrichment analysis (GSEA) at 1,000 random sample permutations using JAVA procedure (http://software.broadinstitute.org/gsea/index.jsp).

Construction of a signature based on m6A RNA methylation regulators

The association between each m6A RNA methylation regulator and patient overall survival (OS) was calculated using the univariate Cox model. Subsequently, the thirteen m6A RNA methylation regulators were screened and verified by least absolute shrinkage and selection operator (LASSO) regression using the ‘glmnet’ R software. Finally, the regulator-based prognostic risk score was constructed through linearly multiplying the expression level with the regression model (β) according to the following formula: Risk=β regulator1 × regulator1 expression + β regulator2 × regulator2 expression + · ···· + β regulatorn × regulatorn expression (15,16).

Confirmation of the signature based on m6A RNA methylation regulators

Patients, together with their survival information, were distributed according to risk score. Furthermore, patients were classified as high- or low-risk, according to their median risk score value. Next, survival curves were drawn according to the Kaplan-Meier method, which could predict the high or low risk of patients. Subsequently, the sensitivity and specificity of survival prediction were compared using risk score, and the time-dependent receiver operating characteristic (ROC) curves were employed to evaluate the accuracy of predicting the 5-year prognosis. In addition, one-way analysis of variance or t-test were carried out to compare risk scores among different cases stratified according to their clinical and molecular pathological features, in order to assess the signature risk score for HCC cases possessing various clinical and pathological features. Univariate and multivariate Cox proportional hazards regression analysis was then conducted to examine whether the risk was predicted independently from other clinical factors.

Statistical analysis

A two-sided P<0.05 was considered to indicate a statistically significant difference. Prism 7 (GraphPad Software Inc., La Jolla, CA, USA) and R software (version 3.4.1; R Foundation, Vienna, Austria), were employed for all analyses.

Results

Expression difference in the m6A RNA methylation regulators between HCCs and normal tissues

The clinicopathological information of all patients is summarized in Table I. Considering the important biological functions of each m6A RNA methylation regulator during tumorigenesis and development, the differences in all m6A RNA methylation regulators between HCCs and normal samples were comprehensively examined. The expression level of each m6A RNA methylation regulator is presented in heatmaps (Fig. 1A) and violin plots (Fig. 1B), which showed that the expression of most m6A RNA methylation regulators was markedly upregulated in HCCs, namely ZC3H13 [not significant (NS)], METTL14 (NS), FTO (P<0.001), YTHDC2 (P<0.001), YTHDC1 (P<0.001), ALKBH5 (P=0.001), KIAA1429 (P<0.001), METTL3 (P<0.001), HNRNPC (P<0.001), RBM15 (P<0.001), YTHDF2 (P<0.001), WTAP (P<0.001) and YTHDF1 (P<0.001).

Table I.

Baseline patient characteristics.

Table I.

Baseline patient characteristics.

CharacteristicsNumberPercentage
Total377100.0
Median follow-up, days (range)557 (0–3,675)
Age, years (mean ± SD)59.5±13.5
Sex
  Male25567.6
  Female12232.4
Ethnicity
  White23562.3
  Others14237.7
Grade
  I  5514.6
  II18047.7
  III12432.9
  IV  133.4
  Unknown  51.3
Stage
  I17546.4
  II  8723.1
  III  8622.8
  IV  51.3
  Unknown  246.4
T stage
  I18549.1
  II  9525.2
  III  8121.5
  IV  133.4
  Unknown  30.8
N
  No25768.2
  Yes  41.1
  Unknown11630.8
M
  No27272.1
  Yes  41.1
  Unknown10126.8
Regulator correlation and interaction

For a better understanding of interactions between these 13 m6A RNA methylation regulators, the correlation (Fig. 2A) and interaction (Fig. 2B) among them was also analyzed. Clearly, ZC3H13 and ALKBH5 were negatively correlated, while the other pairs were positively correlated. FTO, WTAP, YTHDC1, METTL3 and HNRNPC exhibited a significantly positive correlation with the other 12 regulators. Of note, the correlation between HNRNPC and METTL3 (0.72), YTHDC1 (0.67) and YTHDF1 (0.62), ranked top among all correlations. In Fig. 2B, the interactions between 2 regulators were supported by experimental determination (pink lines), the existing databases (blue lines), co-expression (black lines), or text mining (dark olive green lines). In addition, there was a pink line connected to neither two erasers (FTO and ALKBH5) nor two readers (YTHDF1 and YTHDF2), suggesting that more experiments should be carried out on these 4 regulators to examine their interactions with other regulators.

m6A RNA methylation regulator cluster analysis

Based on the expression similarity of m6A RNA methylation regulators, it appeared that k=2 was a sufficient value from the clustering stability range of k=2-10 in the TCGA datasets (Fig. 3A-L). Thereafter, patients were clustered into one of the two subgroups. Therefore, the clinical and pathological characteristics between the two subgroups classified based on k=2 (clusters 1/2) were compared (Fig. 4A). The cluster 1 subgroup was markedly correlated with late stage at diagnosis (P<0.05) and high frequency of grade III/IV (P<0.001). Furthermore, PCA was also employed for comparing transcriptional patterns between the two subgroups. Our findings indicated that these two subgroups were distinctly different (Fig. 4B). In addition, the cluster 1 subgroup had an evidently reduced OS, as compared with that in the cluster 2 subgroup (P=6.197e-4) (Fig. 4C).

Prognosis of m6A RNA methylation regulator, as well as construction and validation of the risk signature

The prognostic value of the m6A RNA methylation regulators in HCCs was also examined. Specifically, gene expression in TCGA datasets was analyzed using the univariate Cox regression model. According to the findings, 9/13 genes examined in this study exhibited a marked correlation with OS (P<0.05; Fig. 5). Among these 9 genes, YTHDF2, YTHDF1, METTL3, KIAA1429, HNRNPC, WTAP, YTHDC1 and RBM15 were the risk genes with a hazard ratio (HR) of >1, while ZC3H13 was the protective gene with a HR of <1.

For a more precise prediction of HCC prognosis using the m6A RNA methylation regulators, the Cox regression algorithm LASSO was utilized (Fig. 6A and B). Six genes, including METTL3, KIAA1429, ZC3H13, YTHDF1, YTHDF2 and ALKBH5, were selected for the construction of a risk signature, according to the minimal standards. In addition, the associated coefficients were acquired based on the LASSO algorithm. Risk was formulated as follows: Risk=0.105*METTL3 expression + 0.041*KIAA1429 expression - 0.094*ZC3H13 expression + 0.025*YTHDF1 expression + 0.067*YTHDF2 expression - 0.005*ALKBH5 expression.

HCC cases obtained from TCGA datasets were classified as low- or high-risk, according to the median risk score value of 3.266, and the distinct heterogeneities with regard to OS were observed between these two subgroups, in order to examine the value of the as-constructed signature in predicting prognosis (P=1.062e-5; Fig. 6C). Furthermore, the ROC curves verified that, prognosis prediction using the risk signature could attain an area under the ROC curve (AUC) value of 0.774 (1 year), 0.732 (3 years) and 0.690 (5 years; Fig. 6D).

The risk signature showed a strong association between clinicopathological features and OS. The expression levels of 6 screened m6A RNA methylation regulators in patients from the high- and low-risk groups within the TCGA dataset are presented in the heatmap (Fig. 7A). Clearly, differences in T stage (P<0.05), grade (P<0.001), status (P<0.05) and stage (P<0.01) were statistically significant between the two groups. Moreover, the association between risk score and every clinicopathological characteristic was examined, and it was found that differences in the risk scores among patients were associated with T stage, stage, grade, and status subgroups, but not age, gender, N stage and M stage (Fig. 7B-I).

Meanwhile, the risk signature HR was 1.238 upon univariate Cox proportional hazards regression [95% confidence interval (CI): 1.168–1.313; P<0.001; Fig. 8A)]. In addition, the same results could be obtained by multivariate Cox proportional hazards regression analysis with adjusted clinical covariate (HR=1.219, 95% CI: 1.143–1.299; P<0.001; Fig. 8B).

The above findings suggested that risk scores determined based on the as-constructed signature were able to precisely estimate the prognosis and clinicopathological characteristics of HCC patients.

External validation of the prognostic signature in the ICGC cohort

To confirm the external validity, the prognostic signature was applied in the ICGC data. The expression levels of the 6 regulators were compared between the high- and low-risk groups, and the heatmap is presented in Fig. 9A. The high-risk group had a significantly shorter survival than the low-risk group in the ICGC cohorts (P=2.588e-3; Fig. 9B). ROC curve analysis showed that risk signature prognosis prediction could attain an AUC value of 0.693 (1 year), 0.723 (3 years) and 0.713 of (5 years; Fig. 9C). Using univariate (P=0.004) and multivariate (P=0.020) Cox regression analysis, the signature was further confirmed as an independent prognostic factor (Fig. 9D and E).

Functional analysis

mRNAs associated with the m6A RNA methylation regulators were applied into the GSEA for enrichment analysis, in order to examine the potential biological functions. As indicated in Fig. 10, the top enrichments included ATM_PATHWAY, CCR5_PATHWAY, CXCR4_PATHWAY, IL6_PATHWAY, MCM_PATHWAY, NGF_PATHWAY, P53HYPOXIA_PATHWAY and TCR_PATHWAY.

Discussion

The present findings showed that the expression of m6A RNA methylation regulators was closely associated with malignant grade and prognosis for HCCs. In addition, two HCC subgroups, namely cluster 1 and 2, were classified using consensus clustering on the basis of m6A RNA methylation regulator expression levels. Specifically, the cluster 1/2 subgroups affected patient prognosis and exhibited a close correlation with clinicopathological features. Furthermore, a risk signature for prognosis was also constructed based on the 6 screened m6A RNA methylation regulators, which could stratify patient OS into high- or low-risk subgroups.

The present study displayed obvious advantages. First, clustering analysis of m6A modification regulators was carried out. Specifically, clusters were formed so that patients in the same cluster were similar, while patients in different clusters were distinct. Second, with regard to methodology, the application of the LASSO-penalized regression could boost the accuracy of the bioinformatics analysis. Different from the conventional stepwise regression used in prior research, the LASSO algorithm could analyze all independent factors simultaneously, identifying the most significant variables (17). Consequently, this formulation approach displayed a higher accuracy than stepwise regression using the multivariate Cox model, particularly in huge datasets, such as genomics (18). Thirdly, the results were validated in the ICGC dataset to check the general applicability. Next, we comprehensively analyzed 13 regulators simultaneously, while previous published studies usually focused on one regulator. Cheng et al (19) reported that KIAA1429 could regulate HCC invasion and migration by changing the m6A modification in ID2 mRNA. In addition, Chen et al (20) reported that METTL3 expression was usually increased in human HCC, which contributed to the progression of HCC, while the SOCS2 level in HCC was repressed by a mechanism that depended on m6A-YTHDF2. Zhao et al (21) discovered that YTHDF1 played a vital role in the regulation of HCC metabolism, as well as cell cycle development. Ma et al (22) reported that METTL14 could suppress the metastatic capacity of HCC cells by regulating the primary miRNA processing of m6A-dependent tumor suppressors. In addition, it was found that YTHDF2 could modulate the m6A level in HCC (23). However, the aforementioned studies only focused on one m6A RNA methylation regulator. Recently, Zhou et al (24) reported the m6A-related genes in HCC, and confirmed the independent predictive value of both METTL3 and YTHDF1 in OS through multivariate Cox regression analysis; therefore, patients were further divided into three groups, based on METTL3 and YTHDF1 expression. Notably, no differential expression of ZC3H13 was observed between tumor and non-tumor samples (exact data not shown). However, ZC3H13 was a protective gene in univariate Cox regression analysis, and further investigations are needed.

The present study revealed that the m6A RNA methylation regulators are correlated with biological processes during the malignant development of HCC. The RNA m6A methylation function within the tumor was recently confirmed, and certain biological processes were found to be affected by it, including tumor stem cell growth, tumorigenesis and self-renewal (25,26), as well as DNA damage response secondary to radiotherapy or chemotherapy (27,28). Herein, the expression levels of m6A RNA methylation regulators in HCC were found to be correlated with HCC-related biological processes, such as ATM_PATHWAY (29), CXCR4_PATHWAY (30) and IL6_PATHWAY (31).

The present results showed that the expression levels of m6A RNA methylation regulators could serve as prognostic markers. The overexpression of YTHDF1 was associated with poor prognosis, which was consistent with the results of Zhao et al (21). In this study YTHDF2 overexpression was correlated with poor prognosis however YTHDF2 suppressed cell proliferation and growth in the study by Zhong et al (32). More importantly, the as-constructed risk signature for the prognosis of HCC based on the 6 selected m6A RNA methylation regulators was proven valuable, and its significance in predicting the T stage, stage, grade and survival status was determined. However, no significant difference in risk score was identified between the N and M stages, which might be partially due to the small number of patients at these stages (Table I). Moreover, risk significance was finally verified by multivariate Cox analysis.

The study, however, had the following limitations: First, more data are necessary to confirm these findings. Second, these 13 regulators, as well as others, require further investigation. Third, consensus clustering analysis was conducted based on the m6A RNA methylation regulator expression levels rather than writers, readers or erasers.

In conclusion, the present study comprehensively illustrated the expression patterns, possible role and prognostic significance of m6A RNA methylation regulators in HCC. Typically, the expression levels of m6A RNA methylation regulators exhibited a strong association with malignant clinical and pathological characteristics in HCCs, as well as with upregulated gene expression involved in biological processes to accelerate the malignant development of HCC. The present study provided critical support for future research into RNA m6A methylation function in HCCs.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81801804).

Availability of data and materials

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

Authors' contributions

WL, QFC, PW and ZLH conceived and designed the study. WL, QFC, TH, PW and LS analyzed the data. WL, TH, PW and LS wrote the paper. WL, QFC, PW and ZLH reviewed and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

m6A

N6-methyladenosine

HCC

hepatocellular carcinoma

TCGA

The Cancer Genome Atlas

ICGC

International Cancer Genome Consortium

NS

not significant

PCA

principal component analysis

OS

overall survival

HR

hazard ratio

LASSO

least absolute shrinkage and selection operator

ROC

receiver operating characteristic

AUC

area under the ROC curve

CI

confidence interval

KM

Kaplan-Meier

GSEA

gene set enrichment analysis

References

1 

He C: Grand challenge commentary: RNA epigenetics? Nat Chem Biol. 6:863–865. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Boccaletto P, Machnicka MA, Purta E, Piatkowski P, Baginski B, Wirecki TK, de Crécy-Lagard V, Ross R, Limbach PA, Kotter A, et al: MODOMICS: A database of RNA modification pathways. 2017 update. Nucleic Acids Res. 46:D303–D307. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Ji P, Wang X, Xie N and Li Y: N6-methyladenosine in RNA and DNA: An epitranscriptomic and epigenetic player implicated in determination of stem cell fate. Stem Cells Int. 2018:32565242018. View Article : Google Scholar : PubMed/NCBI

4 

Chai RC, Wu F, Wang QX, Zhang S, Zhang KN, Liu YQ, Zhao Z, Jiang T, Wang YZ and Kang CS: m(6)A RNA methylation regulators contribute to malignant progression and have clinical prognostic impact in gliomas. Aging (Albany NY). 11:1204–1225. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Meyer KD and Jaffrey SR: Rethinking m(6)A readers, writers, and erasers. Annu Rev Cell Dev Biol. 33:319–342. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Roundtree IA and He C: RNA epigenetics-chemical messages for posttranscriptional gene regulation. Curr Opin Chem Biol. 30:46–51. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, et al: Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 485:201–206. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Li X, Xiong X and Yi C: Epitranscriptome sequencing technologies: Decoding RNA modifications. Nat Methods. 14:23–31. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Wang S, Sun C, Li J, Zhang E, Ma Z, Xu W, Li H, Qiu M, Xu Y, Xia W, et al: Roles of RNA methylation by means of N(6)-methyladenosine (m(6)A) in human cancers. Cancer Lett. 408:112–120. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Huang J and Yin P: Structural insights into N(6)-methyladenosine (m(6)A) modification in the transcriptome. Genomics Proteomics Bioinformatics. 16:85–98. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Luo J, Liu H, Luan S, He C and Li Z: Aberrant regulation of mRNA m(6)A modification in cancer development. Int J Mol Sci. 19:E25152018. View Article : Google Scholar : PubMed/NCBI

12 

Deng X, Su R, Feng X, Wei M and Chen J: Role of N(6)-methyladenosine modification in cancer. Curr Opin Genet Dev. 48:1–7. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Chen QF, Jia ZY, Yang ZQ, Fan WL and Shi HB: Transarterial chemoembolization monotherapy versus combined transarterial chemoembolization-microwave ablation therapy for hepatocellular carcinoma tumors </=5 cm: A propensity analysis at a single center. Cardiovasc Intervent Radiol. 40:1748–1755. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Chen QF, Huang T, Shen L and Li W: Predictive value of a nomogram for hepatocellular carcinoma with brain metastasis at initial diagnosis: A population-based study. PLoS One. 14:e02092932019. View Article : Google Scholar : PubMed/NCBI

15 

Chen QF, Li W, Wu P, Shen L and Huang ZL: Alternative splicing events are prognostic in hepatocellular carcinoma. Aging (Albany NY). 11:4720–4735. 2019.PubMed/NCBI

16 

Chen QF, Li W, Wu PH, Shen LJ and Huang ZL: Significance of tumor-infiltrating immunocytes for predicting prognosis of hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol. 25:5266–5282. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Friedman J, Hastie T and Tibshirani R: Regularization paths for generalized linear models via coordinate descent. J Stat Softw. 33:1–22. 2010. View Article : Google Scholar : PubMed/NCBI

18 

McNeish DM: Using lasso for predictor selection and to assuage overfitting: A method long overlooked in behavioral sciences. Multivariate Behav Res. 50:471–484. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Cheng X, Li M, Rao X, Zhang W, Li X, Wang L and Huang G: KIAA1429 regulates the migration and invasion of hepatocellular carcinoma by altering m6A modification of ID2 mRNA. OncoTargets Ther. 12:3421–3428. 2019. View Article : Google Scholar

20 

Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, Tsang LH, Ho DW, Chiu DK, Lee JM, et al: RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 67:2254–2270. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Zhao X, Chen Y, Mao Q, Jiang X, Jiang W, Chen J, Xu W, Zhong L and Sun X: Overexpression of YTHDF1 is associated with poor prognosis in patients with hepatocellular carcinoma. Cancer Biomark. 21:859–868. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, Wang TT, Xu QG, Zhou WP and Sun SH: METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6 -methyladenosine-dependent primary MicroRNA processing. Hepatology. 65:529–543. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Yang Z, Li J, Feng G, Gao S, Wang Y, Zhang S, Liu Y, Ye L, Li Y and Zhang X: MicroRNA-145 modulates N6-methyladenosine levels by targeting the 3′-untranslated mRNA Region of the N6-methyladenosine binding YTH domain family 2 protein. J Biol Chem. 292:3614–3623. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Zhou Y, Yin Z, Hou B, Yu M, Chen R, Jin H and Jian Z: Expression profiles and prognostic significance of RNA N6-methyladenosine-related genes in patients with hepatocellular carcinoma: Evidence from independent datasets. Cancer Manag Res. 11:3921–3931. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Pan Y, Ma P, Liu Y, Li W and Shu Y: Multiple functions of m6A RNA methylation in cancer. J Hematol Oncol. 11:482018. View Article : Google Scholar : PubMed/NCBI

26 

Cui Q, Shi H, Ye P, Li L, Qu Q, Sun G, Sun G, Lu Z, Huang Y, Yang CG, et al: m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 18:2622–2634. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Dai D, Wang H, Zhu L, Jin H and Wang X: N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 9:1242018. View Article : Google Scholar : PubMed/NCBI

28 

Xiang Y, Laurent B, Hsu CH, Nachtergaele S, Lu Z, Sheng W, Xu C, Chen H, Ouyang J, Wang S, et al: RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature. 543:573–576. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Gao SB, Li KL, Qiu H, Zhu LY, Pan CB, Zhao Y, Wei SH, Shi S, Jin GH and Xue LX: Enhancing chemotherapy sensitivity by targeting PcG via the ATM/p53 pathway. Am J Cancer Res. 7:1874–1883. 2017.PubMed/NCBI

30 

Meng YM, Liang J, Wu C, Xu J, Zeng DN, Yu XJ, Ning H, Xu L and Zheng L: Monocytes/Macrophages promote vascular CXCR4 expression via the ERK pathway in hepatocellular carcinoma. Oncoimmunology. 7:e14087452017. View Article : Google Scholar : PubMed/NCBI

31 

Cheng Y, Li H, Deng Y, Tai Y, Zeng K, Zhang Y, Liu W, Zhang Q and Yang Y: Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 9:4222018. View Article : Google Scholar : PubMed/NCBI

32 

Zhong L, Liao D, Zhang M, Zeng C, Li X, Zhang R, Ma H and Kang T: YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett. 442:252–261. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2020
Volume 19 Issue 4

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
Li W, Chen QF, Huang T, Shen L, Huang ZL and Wu P: Profiles of m6A RNA methylation regulators for the prognosis of hepatocellular carcinoma. Oncol Lett 19: 3296-3306, 2020
APA
Li, W., Chen, Q., Huang, T., Shen, L., Huang, Z., & Wu, P. (2020). Profiles of m6A RNA methylation regulators for the prognosis of hepatocellular carcinoma. Oncology Letters, 19, 3296-3306. https://doi.org/10.3892/ol.2020.11435
MLA
Li, W., Chen, Q., Huang, T., Shen, L., Huang, Z., Wu, P."Profiles of m6A RNA methylation regulators for the prognosis of hepatocellular carcinoma". Oncology Letters 19.4 (2020): 3296-3306.
Chicago
Li, W., Chen, Q., Huang, T., Shen, L., Huang, Z., Wu, P."Profiles of m6A RNA methylation regulators for the prognosis of hepatocellular carcinoma". Oncology Letters 19, no. 4 (2020): 3296-3306. https://doi.org/10.3892/ol.2020.11435