Original Article/Liver
Integrating transcriptomes and somatic mutations to identify RNA methylation regulators as a prognostic marker in hepatocellular carcinoma

https://doi.org/10.1016/j.hbpd.2020.05.002Get rights and content

Abstract

Background

RNA methylation modifying plays an important role in the occurrence and progression of a range of human cancers including hepatocellular carcinoma (HCC), which is characterized by a mass of genetic and epigenetic alterations. However, the treatment targeting these alterations is limited.

Methods

We used comprehensive bioinformatics analysis to analyze the correlation between cancer-associated RNA methylation regulators and HCC malignant features in network datasets.

Results

We identified two HCC subgroups (cluster 1 and 2), which had clearly distinct clinicopathological, biofunctional and prognostic characteristics, by consensus clustering. The cluster 2 subgroup correlated with malignancy of the primary tumor, higher tumor stage, higher histopathological grade and higher frequency of TP53 mutation, as well as with shorter survival when compared with cluster 1. Gene enrichment indicated that the cluster 2 correlated to the tumor malignancy signaling and biological processes. Based on these findings, an 11-gene risk signature was built, which not only was an independent prognostic marker but also had an excellent power to predict the tumor features.

Conclusions

Our study indicated that RNA methylation regulators are vital for HCC malignant progression and provide an important evidence for RNA methylation, methylation regulators are actionable targets for anticancer drug discovery.

Introduction

Hepatocellular carcinoma (HCC), with approximately 750,000 deaths each year, is one of the most common lethal cancers worldwide [1]. At present, HCC occurrence and progression are considered an intricate molecular pathological process involving multi-gene alterations and multi-step processes [2]. Persistent inflammatory microenvironment, such as viral infection, obesity and diabetes, causes hepatic parenchymal necrosis and regeneration. The long term inflammation results in the chromosomal instability and gene mutations in the hepatocytes [3], [4], [5]. However, the specific mechanics are still unclear and therefore, the potent drugs for molecular targeted therapy are sparse. Sorafenib is so far the only FDA-approved drug for advanced HCCs [6].

Emerging data indicated that RNA methylation regulates RNA splicing, nuclear export, translation and degradation. RNA methylation impacts almost all RNAs, like messenger RNAs (mRNAs) [7], tRNAs [8], microRNAs (miRNAs) [9], circular RNAs (cirRNAs) [10], ribosomal RNAs (rRNAs) [11], small nuclear RNAs (snRNAs) [12] and small nucleolar RNAs (snoRNAs) [13]. The majority of those RNAs are in the forms of N1-methyladenosine (m1A), 5-methylcytosine (m5C) and N6,2′-O-dimethyladenosine (m6A). However, these cancer-associated RNA methylations are on the RNA molecules, and the downstream biological functions are not consistent. A recent study found that m6A demethylase fat mass and obesity associated protein (FTO), acting as an oncogene, is up-regulated in acute myelocytic leukemia (AML), resulting in the decreased m6A and differentiation inhibition; FTO depletion enhances differentiation and decreases cell proliferation, cell colonies formation and promotes apoptosis [14]. Agent inhibiting demethylase FTO catalytic activity has anti-tumor effect via targeting FTO/m6A/MYC/CEBPA signaling pathway [15]. Methyltransferase like 3 (METTL3) has an inhibitory reaction to haematopoietic stem cell differentiation and is up-regulated in AMLs, while METTL3 depletion induces myeloid differentiation and cellular apoptosis [16,17]. This irreconcilable opposition is also seen in the gliomas. ALKBH5 as a demethylase is up-regulated in glioblastomas and correlated with poor prognosis, and its down-regulation blocks self-renewal of glioblastoma stem cells and prolongs mice survival [18]. Similarly, depletion of the METTL3-METTL14 complex, which is vital for initiation of m6A methylation process, decreases m6A burdens and facilitates glioblastoma stem cells growth leading to a poor survival [19]. But the recent research has suggested that reducing levels of m6A by silencing METTL3 could inhibit glioblastomas progression and improve the survival [20].

The data above indicate that different tumors or the same tumor at different phases have the differential sensitivity to methylations. HCC is characterized by a mass of genetic and epigenetic alterations, including aberrant activation of WNT signaling pathway and loss-of-function mutation in the TP53 suppressor gene [21,22]. This study aimed to elucidate the role of methylation regulators in HCC at different phase and different differentiation grade, the correlation between each key regulator and HCC clinicopathological feature. We tried to uncover the specific mechanisms of HCC evolution and to facilitate further anticancer drugs discovery.

Section snippets

Datasets acquisition and preprocessing

Transcriptome sequencing profiling data, corresponding clinical features and annotated somatic mutations of 424 HCC patients were acquired from the Cancer Genome Atlas (TCGA) [23]. A meta-data comprising 7 independent expression datasets of 401 HCC patients, including GSE75271, GSE45436, GSE62232, GSE29721, GSE41804, GSE40873 and GSE6222, were obtained from Gene Expression Omnibus (GEO) [24]. The 7 GEO datasets, from Affymetrix Human Genome U133 plus 2.0 Array platforms, have been normalized

HCC patients showed clear correlations with up-regulated expression of most cancer-associated RNA methylation regulators

As to these chosen RNA methylation regulators linked to specific human cancers, we first studied the relationships between the genes and HCC pathological features. Expression of each gene in different histological types, including 50 normal liver samples and 374 HCC samples from the TCGA dataset, and 137 normal liver samples and 264 HCC samples from the GEO meta-data, is presented as heat maps (Fig. 1A and B). It showed that the expressions of 45 (77.6%) cancer-associated RNA methylation

Discussion

HCC is one of malignant neoplasms with the highest incidence and mortality worldwide [33]. It is demonstrated that molecular signatures play important roles in early detection and prognosis of variety of cancers [34], [35], [36], which logically also play important roles in HCC patients. In this study, we comprehensively investigated the correlations between clinicopathological features and gene expression patterns, and the potential prognostic value within 58 selected cancer-associated RNA

Acknowledgments

We are very grateful to the contributions from the TCGA and GEO networks, the R project for statistical computing (including the Rtools) as well as all the selfless authors of all statistics packages in R.

CRediT authorship contribution statement

Yue Yan: Conceptualization, Formal analysis, Methodology, Software, Visualization, Writing - original draft. Yun-Hai Luo: Data curation, Resources, Validation, Writing - review & editing. Dao-Feng Zheng: Data curation, Validation, Writing - review & editing. Tong Mu: Data curation, Validation, Writing - review & editing. Zhong-Jun Wu: Conceptualization, Data curation, Resources, Supervision, Validation, Writing - review & editing.

Funding

None.

Ethical approval

The study was approved by the Committee for Medical Research Ethics of the First Affiliated Hospital of Chongqing Medical University.

Competing interest

No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.

References (55)

  • M.L. Tornesello et al.

    Mutations in TP53, CTNNB1 and PIK3CA genes in hepatocellular carcinoma associated with hepatitis B and hepatitis C virus infections

    Genomics

    (2013)
  • J.T. Nhieu et al.

    Nuclear accumulation of mutated beta-catenin in hepatocellular carcinoma is associated with increased cell proliferation

    Am J Pathol

    (1999)
  • M.D. Metodiev et al.

    Recessive Mutations in TRMT10C Cause Defects in Mitochondrial RNA Processing and Multiple Respiratory Chain Deficiencies

    Am J Hum Genet

    (2016)
  • J. Ferlay et al.

    Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012

    Int J Cancer

    (2015)
  • J. Zucman-Rossi et al.

    Genetic landscape and biomarkers of hepatocellular carcinoma

    Gastroenterology

    (2015)
  • R. Dhanasekaran et al.

    Molecular pathogenesis of hepatocellular carcinoma and impact of therapeutic advances

    F1000Res

    (2016)
  • J.D. Yang et al.

    Hepatocellular carcinoma: a global view

    Nat Rev Gastroenterol Hepatol

    (2010)
  • J.M. Llovet et al.

    SHARP investigators study group. Sorafenib in advanced hepatocellular carcinoma

    N Engl J Med

    (2008)
  • W.V. Gilbert et al.

    Messenger RNA modifications: form, distribution, and function

    Science

    (2016)
  • H. Singh et al.

    The alkali-stable dinucleotide sequences in 18s+28s ribonucleates from wheat germ

    Can J Biochem

    (1964)
  • A. Krol et al.

    Primary and secondary structures of chicken, rat and man nuclear U4 RNAs. Homologies with U1 and U5 RNAs

    Nucleic Acids Res

    (1981)
  • B. Linder et al.

    Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome

    Nat Methods

    (2015)
  • R. Su et al.

    R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling

    Cell

    (2018)
  • L.P. Vu et al.

    The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells

    Nat Med

    (2017)
  • I. Barbieri et al.

    Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control

    Nature

    (2017)
  • S. Zhang et al.

    m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program

    Cancer Cell

    (2017)
  • A. Visvanathan et al.

    Essential role of METTL3-mediated m6A modification in glioma stem-like cells maintenance and radioresistance

    Oncogene

    (2018)
  • Cited by (2)

    View full text