Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Oct 1.
Published in final edited form as: Sci China Life Sci. 2024 May 27;67(10):2105–2119. doi: 10.1007/s11427-023-2494-x

RNA modifications in the progression of liver diseases: from fatty liver to cancer

Simiao Li 1, Wajahat Z Mehal 1, Xinshou Ouyang 1,*
PMCID: PMC11545962  NIHMSID: NIHMS2018600  PMID: 38809498

Abstract

Non-alcoholic fatty liver disease (NAFLD) has emerged as a prominent global health concern associated with high risk of metabolic syndrome, and has impacted a substantial segment of the population. The disease spectrum ranges from simple fatty liver to non-alcoholic steatohepatitis (NASH), which can progress to cirrhosis and hepatocellular carcinoma (HCC) and is increasingly becoming a prevalent indication for liver transplantation. The existing therapeutic options for NAFLD, NASH, and HCC are limited, underscoring the urgent need for innovative treatment strategies. Insights into gene expression, particularly RNA modifications such as N6 methyladenosine (m6A), hold promising avenues for interventions. These modifications play integral roles in RNA metabolism and cellular functions, encompassing the entire NAFLD-NASH-HCC progression. This review will encompass recent insights on diverse RNA modifications, including m6A, pseudouridine (Ψ), N1-methyladenosine (m1A), and 5-methylcytidine (m5C) across various RNA species. It will uncover their significance in crucial aspects such as steatosis, inflammation, fibrosis, and tumorigenesis. Furthermore, prospective research directions and therapeutic implications will be explored, advancing our comprehensive understanding of the intricate interconnected nature of these pathological conditions.

Keywords: m6A RNA modification, NASH, NAFLD, HCC, fatty liver

Introduction

Non-alcoholic fatty liver disease (NAFLD) has emerged as a substantial global public health concern, impacting approximately 25% of the worldwide population, and has increasingly become a public health concern in recent years alongside obesity and metabolic syndrome (Huang et al., 2021). NAFLD comprises a spectrum of liver conditions characterized by the excessive accumulation of fat within the hepatic tissue, spanning from simple steatosis to the more severe non-alcoholic steatohepatitis (NASH). Marked by inflammation and fibrosis, NASH can potentially culminate in cirrhosis and hepatocellular carcinoma (HCC), and has become an increasingly prevalent indication for liver transplantation (Ipsen et al., 2018). Notably, NAFLD has taken the lead as the fastest-growing cause of HCC (Huang et al., 2021), with a model projecting a staggering 130% increase in NAFLD-related HCC cases in the US, from 10,820 cases in 2016 to an estimated 24,860 cases by 2030 (Estes et al., 2018).

Dietary and lifestyle modifications can ameliorate NAFLD, but there remains a dearth of pharmaceutical treatments for both NAFLD and NASH (Powell et al., 2021). First-line therapies for HCC include Sorafenib and Lenvatinib, but provide a response in a minority of patients (Yang et al., 2023a). Ultimately, understanding the molecular mechanisms underlying the progression from NAFLD to HCC is pivotal to advancing treatment options for these interconnected disorders.

A comprehensive grasp of gene expression at both transcriptional and translational levels is essential for advancing treatment strategies. Epigenetics encompasses histone modifications, DNA methylation, and chromatin rearrangement. Meanwhile, the burgeoning field of RNA modifications, known as “epitranscriptomics,” scrutinizes post-transcriptional gene regulation. RNA modification is observed across various RNA species, including mRNA, lncRNA, miRNA, circRNA, tRNA, and rRNA. Diverse modifications such as N6 methyladenosine (m6A), pseudouridine (Ψ), N1-methyladenosine (m1A), 5-methylcytidine (m5C), N6,2′-O-dimethyladenosine (m6Am), inosine, N4-acetylcytidine (ac4C), 2′-O-methylated nucleotide (Nm), and internal N7-methylguanosine (m7G) play multifaceted roles in RNA metabolism, influencing splicing, translation, stability, and export. Given the pivotal regulatory roles of RNA modifications in cellular activity, they hold potential as exciting targets for various diseases, including NAFLD, NASH, and HCC (Roundtree et al., 2017a).

In this review, we delve into the intricate roles of RNA modifications throughout the NAFLD-NASH-HCC progression. The exploration begins with an overview of the fundamental mechanisms of m6A methylations, subsequently revealing their impact on crucial aspects including steatosis, inflammation, fibrosis, and tumorigenesis. Finally, we contemplate the broader spectrum of RNA modifications and anticipate the future research pathways that will further illuminate this dynamic field.

m6A RNA modification

m6A is one of the most common and well-studied RNA modifications in eukaryotes. It is a dynamic and reversible modification typically installed on the motif RRACH (R=A or G; H=A, C, or U). The m6A modifications can be installed by methyltransferases (writers), removed by demethylases (erasers) and recognized by RNA-binding proteins (readers).

Writers, erasers, and readers

m6A modifications are primarily installed by the methyltransferase complex, which is localized in nuclear specks. The core of this complex is composed of a heterodimer between methyltransferase-like3 (METTL3) and methyltransferase-like14 (METTL14), with METTL3 being the catalytic subunit (Liu et al., 2014; Wang et al., 2016). Wilm’s tumor 1-associating protein (WTAP) is a regulatory subunit of the methyltransferase complex (Ping et al., 2014). Other m6A writers form the regulatory component of the methyltransferase as well, including vir like m6A methyltransferase associated (VIRMA), zinc finger CCCH domain-containing protein 13 (ZC3H13), E3 ubiquitin-protein ligase Hakai (HA-KAI), and RNA binding motif protein 15/15B (RBM15/15B) (Patil et al., 2016; Růžička et al., 2017; Wen et al., 2018; Yue et al., 2018). Additionally, METTL16 can install m6A modifications to specific motifs independently of the classic methyltransferase complex (Mendel et al., 2021).

Fat mass and obesity-associated protein (FTO) and alkB homolog 5 (ALKBH5) are the two m6A erasers characterized so far, and they can remove m6A markers from mRNAs independently of each other (Jia et al., 2011; Zheng et al., 2013).

A multitude of m6A readers are engaged in diverse roles in mediating RNA stability, decay, and translation. These readers can be categorized into three main groups: proteins featuring the YT521-B homology (YTH) domain, insulin-like growth factor 2 binding proteins (IGF2BPs), and heterogeneous nuclear ribonucleoproteins (hnRNPs). YTHDF2 promotes the degradation of its target transcripts, whereas YTH N6-methyladenosine RNA binding protein F1 (YTHDF1) and YTH N6-methyladenosine RNA binding protein F3 (YTHDF3) promote translation (Du et al., 2016; Shi et al., 2017). YTH N6-methyladenosine RNA binding protein C1 (YTHDC1) mediates different RNA fates including nuclear export, alternative splicing, RNA stabilization, and RNA decay, and YTH N6-methyladenosine RNA binding protein C2 (YTHDC2) regulates translation and stability (Roundtree et al., 2017b; Zhou et al., 2021). insulin-like growth factor 2 binding protein 1/2/3 (IGF2BP1, 2, and 3) all appear to stabilize their target transcripts (Bell et al., 2013; Huang et al., 2018). Finally, heterogeneous nuclear ribonucleoprotein C (HNRNPC), heterogeneous nuclear ribonucleoprotein G (HNRNPG), and heterogeneous nuclear ribonucleoprotein A2/B1 (HNRNPA2B1) regulate RNA splicing and processing (Alarcón et al., 2015). (Figure 1)

Figure 1.

Figure 1.

Fundamental mechanisms of m6A modifications. The writers such as METTL3, METTL14, and WTAP add m6A modifications to RNA, and the erasers, FTO and ALKBH5, remove it. Various readers, YTHDF1/2/3, YTHDC1/2, IGFBP1/2/3, and hnRNPs, recognize RNA with m6A modifications and promote its translation, decay, stability, or splicing.

m6A modifications have been shown to be implicated in diverse developmental and pathological processes. It is essential to hemopoiesis, immunity, the central nervous system, the immune system, the reproductive system, and adipose tissue development and homeostasis (Ben-Haim et al., 2015; Lence et al., 2016; Vu et al., 2017; Wu et al., 2023; Zhang et al., 2017). The dysregulation of m6A, notably the upregulation of METTL3, has been associated with a range of cancers (Liu et al., 2023b; Vu et al., 2017; Xiang et al., 2017; Yang et al., 2019). Additionally, m6A modifications are implicated in metabolic diseases including diabetes mellitus, cardiovascular diseases, obesity, and notably NAFLD (De Jesus et al., 2019; Jiang et al., 2021a; Li et al., 2023; Yang et al., 2020). Taken together, in view of the increased focus on exploring the intricate interplay between m6A and liver diseases, a comprehensive review dedicated to this subject is not only timely but also imperative for the advancing of this field.

The function of m6A in fatty liver progression

Dysregulation of m6A writers, readers, and erasers drives the progression of NAFLD through a variety of pathways. Cheng et al. (2022) observed elevated expression levels of METTL3, METTL14, FTO, and eukaryotic translation initiation factor 3 subunit H (EIF3H), along with decreased expression levels of WTAP, RBM15, YTHDC1, YTHDC2, IGF2BP1, HNRNPC, and HNRNPA2B1 in liver biopsy tissues from NAFLD patients. Another study found significant changes in m6A modification levels of 226 differentially expressed genes in high fructose diet induced NAFLD mice. Of these 226 genes, 193 were more methylated, suggesting that greater m6A modifications correlate with NAFLD (Luo et al., 2021). However, the amount and effect of m6A modifications in the NAFLD-NASH-HCC progression change based on cell types and mRNA targets.

Lipid metabolism regulation by m6A

An imbalance in lipid metabolism is a significant aspect of NAFLD. Liver steatosis occurs when fatty acid uptake and de novo lipogenesis (DNL) exceed fatty acid oxidation and lipid export. Each of these lipid metabolism processes is affected by m6A modifications (Figure 2).

Figure 2.

Figure 2.

m6A modifications in steatosis. Writers and erasers affect the m6A levels on the mRNA of various genes related to fatty acid uptake, lipogenesis, lipid export, and fatty acid oxidations. Dysregulation of m6A-related proteins leads to dysregulation of lipid metabolism-related gene expression, which causes steatosis.

Lipogenesis and fatty acid uptake

Lipogenesis is the process in which acetyl-CoA from excess carbohydrates is converted into new fatty acids, which are then stored as triglycerides. It is largely regulated by two transcription factors: sterol regulatory binding protein 1c (SREBP-1c) and ChREBP. SREBP1c activates lipogenic genes such as fatty acid synthase (FASN), stearoyl-coenzyme A desaturase 1 (SCD1), adenosine triphosphate citrate lyase (ACLY), and acetyl-CoA carboxylase 1 (ACC1) (Ipsen et al., 2018). The expression of SREBP-1c, ChREBP, FASN, SCD1, and ACLY are all heavily affected by m6A modifications.

FTO, by decreasing levels of m6A methylation of SREBF1 and ChREBP mRNA, stabilizes their mRNA transcripts and promotes the expression of SREBF1 and ChREBP as well as downstream lipogenic genes FASN, SCD1, ACLY, and ACC1 (Tang et al., 2023). This is supported by Zhou et al. (2021), and they showed that YTHDC2, as a reader, bound to the m6A locations on SREBP1c, FASN, ACC1, and SCD1 mRNA and destabilized them. Glucocorticoids play a role in increasing lipid accumulation via this pathway, as the glucocorticoid receptor binds to the promoter of FTO, which leads to increased FTO levels and subsequently upregulates lipogenic genes including Srebf1, Fasn, Acaca, and Scd1 (Hu et al., 2020). Additionally, the increased FTO activity leads to the upregulation of SREBP1c, thereby promoting the activation of Cell death-inducing DFFA (DNA fragmentation factor-α)-like effector c (CIDEC), a regulator of lipid droplet size and triglyceride accumulation (Chen et al., 2018a). Conversely, a conflicting study revealed that in a NAFLD mouse model and HCC patients, SREBP1 was not upregulated. Instead, METTL3 and METTL14 directly methylated ACLY and SCD1 mRNA, resulting in enhanced expressions of ACLY and SCD1, and promoting lipogenesis (Yang et al., 2022b).

In addition to SREBP1c, m6A modifications also govern pathways associated with fatty acid synthesis, triglyceride synthesis, and fatty acid uptake. For instance, METTL3 stabilizes the lipogenesis-related lncRNA, LINC00958, thereby leading to the elevation of cellular cholesterol and triglyceride levels in HCC (Zuo et al., 2020). Furthermore, another study revealed that METTL3 stabilized the mRNA of diverse genes involved in glucose and lipid metabolism within the context of liver metabolic disorders and hematogenous diabetes, with Lpin1 being a notable example (Li et al., 2020). The m6A reader IGF2BP2, activated by Hilnc, stabilizes Pparγ mRNA, facilitating an increase in liver PPARγ level (Jiang et al., 2021b). While typically not abundantly expressed in the liver, its expression is associated with metabolic syndrome and triggers the upregulation of essential components such as fatty acid binding protein 4, Cidec, CD36, monoacylglycerol O-acyltransferase 1, and perilipin 2, which collectively contribute to the processes involved in triglyceride synthesis, free fatty acid uptake, and lipid droplet formation (Wang et al., 2020). Cd36, a key regulator of free fatty acid uptake, has also been directly governed by METTL3 and WTAP, but through an m6A independent mechanism. Li et al. revealed that METTL3 and WTAP inhibited Cd36 expression via histone modification (Li et al., 2021b). Additionally, the surplus of these free fatty acids might arise from increased lipoatrophy in white adipose tissues, which is regulated by WTAP suppression of IGFBP1 (Li et al., 2022c).

Fatty acid oxidation and lipid export

Upon uptake by hepatocytes or synthesis through DNL, fatty acids face two pathways: oxidation for energy or conversion into triglycerides. These triglycerides can then be either released into bloodstream as very low-density lipoproteins (VLDL) or stored as lipid droplets within hepatocytes.

Given that steatosis indicates the accumulation of lipid droplets, diminished fatty acid oxidation could contribute to the onset of NAFLD. Wei et al. (2022) demonstrated the involvement of FTO in the demethylation of peroxisome proliferator-activated receptor α (PPARα), a master regulator of fatty acid oxidation. The reduction in methylation of PPARα mRNA results in diminished PPARα expression, reduced fatty acid oxidation, and increased lipid accumulation. Building on this, another study confirms that PPARα undergoes methylation by METTL3 and destabilization by YTHDF2. The m6A methylation of PPARα constitutes an element of the Bmal1-regulated rhythmic epigenomic programming driven by the circadian clock as Bmal1 governs METTL3 and YTHDF2 levels (Zhong et al., 2018).

Beyond its impact on fatty acid oxidation, alternations in m6A levels also exert an influence on lipid clearance. Written by METTL3 and read by YTHDF1, the m6A methylation of Rubicon mRNA promotes its stability. The run domain Beclin-1-interacting and cysteine-rich domain-containing protein (RUBICON), localized to lysosomes, binds to PI2KC3 and inhibits autophagosome-lysosome fusion, thus decreasing the rate of lipid droplet clearance (Peng et al., 2022c).

The function of m6A in NAFLD to NASH progression: inflammation and fibrosis

Inflammation

The transition from NAFLD to NASH is marked by the inclusion of inflammation and hepatocyte damage. In the context of NAFLD, the buildup of lipid droplets in hepatocytes triggers endoplasmic reticulum (ER) stress, leading to apoptosis and the subsequent release of chemokines. Notably, m6A methylation is intricately involved in regulating multiple aspects of these inflammatory processes.

METTL14 appears to exert a protective function against ER stress and apoptosis in hepatocytes. Wei et al. (2021) demonstrated that the unfolded protein response (UPR) promoted an augmentation in METTL14 expression. Operating an m6A-dependent mechanism, METTL14 facilitates the degradation of C/EBP-homologous protein (CHOP) mRNA, which in turn suppresses the expression of downstream proapoptotic target genes. This intricate process functions as a protective role against ER proteotoxic stress and the consequential liver injury. Moreover, the depletion of METTL14 results in reduced stability and translation of heat shock protein 90 beta family member 1 (Hsp90b1), endoplasmic reticulum protein 29 (Erp29), STT3 oligosaccharyltransferase complex catalytic subunit A (Stt3a), prolyl 4-hydroxylase subunit beta (P4hb), and lectin, mannose binding 1 (Lman1) mRNA transcripts. Given that these genes encode polypeptide-processing proteins, their diminished expressions can lead to an accumulation of unfolded or misfolded proteins, triggering the UPR and inducing ER stress (Cao et al., 2021).

Numerous proinflammatory pathways undergo regulations through m6A modifications. For example, TAK1-binding protein 2 (Tab2), a target of IGF2BP2, can promote the activation of JNK and NF-κB pathways, consequently leading to the expression of a range of chemokines and cytokines (Zhou et al., 2022a). Gan et al. (2022) showed that FTO, through demethylation of IL-17RA, enhanced its expression, thereby promoting liver inflammation. In the context of NASH development induced by arsenic exposure and hepatic insulin resistance, Qiu et al. (2023) found that METTL14 and IGF2BP2 contributed to the stabilization of NLRP3 mRNA, subsequently promoting NLRP3 inflammasome activation and resulting in the release of IL-1β and IL-18 (Figure 3).

Figure 3.

Figure 3.

The m6A modifications in liver inflammation and fibrosis. Hepatocytes, hepatic stellate cells, Kupffer cells, and recruited immune cells all play a role in NASH progression. The multifaceted role of m6A modifications in NASH relies on its decorated mRNAs among distinct immune cell types.

The inflammatory cytokine CCL2 has been associated with the progression of steatosis to NASH (Pan et al., 2020). Within hepatocytes, METTL3 and WTAP act to inhibit CCL2 expression, whereas IGF2BP2 contributes to the elevation of CCL2 levels. Interestingly, the roles of METTL3, WTAP, and IGF2BP2 in this context may not rely solely on m6A modifications; instead, they might involve DNA and histone modifications as a mechanism for regulating CCL2 (Li et al., 2021b; Simon et al., 2014)

Immune cell activation

Stimulated by the inflammatory signals originating from distressed hepatocytes, an immune response is triggered, initiating with the liver’s resident macrophages, Kupffer cells. Feng et al. (2021) unveiled that NF-κB engaged with the promotors of METTL3 and METTL14, leading to increased m6A methylation across Kupffer cells. Moreover, the methylation of transforming growth factor β1 (TGF-β1) mRNA facilitates its translation.

Hepatic macrophages are classified as pro-inflammatory and cytokine-producing M1 macrophages, and the anti-inflammatory M2 macrophages. Shu et al. (2021) uncovered that METTL3 methylated and stabilized the lncRNA MALAT1, which holds significance for the polarization of M1 macrophages. Furthermore, as a consequence of the MALAT1 pathway, the engagement of TAK1 induces pyroptosis, a type of programmed cell death distinguished by the creation of cell membrane pores that release inflammatory cytokines. This aligns with another investigation where alcohol consumption-induced pyroptosis in Kupffer cells was mitigated by silencing METTL3 (Pan et al., 2023a). Additionally, METTL3 was found to potentially enhance M2 polarization in Kupffer cells through the stabilization of RBM14 mRNA facilitated by YTHDF1 (Hu et al., 2022).

The progression towards NASH is marked by the infiltration of neutrophils into the liver. Elevation in the expression of m6A reader IGF2BP2 within mouse hepatocytes triggers the activation of NF-κB signaling, resulting in the increased expression of Cxcl1 and Cxcl2. These changes contribute to the recruitment of neutrophils to the liver (Zhou et al., 2022a). The activation of additional myeloid cells also plays a role in the progression of NASH. Qin et al. (2021) demonstrated that METTL3 methylated and destabilized DNA damage inducible transcript 4 (DDIT4) mRNA. DDIT4 typically inhibits mTORC1 and NF-κB signaling pathways. Therefore, increased m6A methylation of DDIT4 caused by excessive nutrient load and metabolic ER stress results in the upregulation of mTORC1 and NF-κB signaling. This, in turn, leads to an enhancement of macrophage effector function. Taken together, the distress of hepatocytes initiates the activation of Kupffer cells via NF-κB-mediated m6A methylation, subsequently influencing macrophage polarization. Within the context of m6A methylation processes, where METTL3, METTL14 and IGFBP2 play pivotal roles, these functions encompass the safeguarding against ER stress, the regulation of proinflammatory pathways, and shaping of immune responses throughout the progression of NASH development.

Hepatic stellate cell activation and fibrogenesis

In the healthy liver, hepatic stellate cells (HSCs) are in a quiescent state, undergo activation by signaling from hepatocytes and macrophages, and turn into ECM-producing myofibroblasts. An investigation on the variations in m6A modifications across the transcriptome of mice with liver fibrosis (Fan et al., 2021) showed that among the 3,315 genes with noteworthy differences in m6A levels, 2,498 displayed hypermethylation, while 817 showed hypomethylation. Notably, a significant number of these genes were associated with key elements of HSC activation, including ER stress response, PPAR, and TGF-β signaling pathways. Indeed, Sun et al. (2022) revealed that peroxiredoxin 3 (PRDX3), whose translation was upregulated by m6A reader YTHDF3, operated as a suppressor of HSC activation and liver fibrosis through the (ROS)/TGF-β1/Smad2/3 pathway. The primary function of PRDX3 is to eliminate a significant proportion of mitochondrial hydrogen peroxide (H2O2). Therefore, a decrease in PRDX3 translation leads to an accumulation of reactive oxygen species in mitochondria, consequently triggering the activation on the TGF-β signaling pathway. It is found (Yang et al., 2022a) that TGF-β stimulation diminishes the levels of ALKBH5 in HSCs. Through its role in demethylating patched 1 (Ptch1) mRNA, ALKBH5 increases PTCH1 levels, thus impeding hedgehog signaling and subsequently reducing HSC activation. Additionally, ALKBH5 functions by eliminating the m6A modification on Dynamin-related protein 1 (Drp1), which in turn prevents YTHDF1-facilitated Drp1 translation. DRP1 is instrumental in driving mitochondrial fission, a pivotal process contributing to TGF-β induced HSC proliferation (Wang et al., 2023a). The m6A modification could indirectly influence DRP1 levels, as increased methylation of nuclear receptor subfamily 1 group d member 1 (NR1D1), a clock gene, leads to its degradation mediated by YTHDC1, thereby inhibiting the phosphorylation of DRP1 (Chen et al., 2023). Within HSCs, the reduction in DRP1 levels, arising from m6A methylation of either Nr1d1 or Drp1, leads to a reduced mitochondrial fission. This consequently triggers an increase of mtDNA release, activating the cGAS pathway and promoting liver fibrosis. A promising avenue emerges with the potential role of dihydroartemisinin (DHA) in alleviating liver fibrosis. DHA achieves this by orchestrating the proteasomal degradation of YTHDC1, thus contributing to the moderation of NR1D1 degradation (Chen et al., 2023) (Figure 3).

Upon activation, m6A modifications are essential for the perpetuation of HSC activation. The transcripts of major collagen genes undergo hypermethylation, with YTHDF1 contributing to their stability (Feng et al., 2023). Simultaneously, METTL3 engages in the modification of large tumor suppressor kinase 2 (Lats2) mRNA, an integral participant in the Hippo/Yap signaling pathway, thereby enhancing its stability. This increased LATS2 expression prompts increased phosphorylation of the downstream transcription factor YAP, resulting in the attenuation of pro-fibrotic gene expression (Li et al., 2022d). Hence, the excessive production of ECM in HSCs is due to the interplay of distinct methylation patterns, encompassing both hyper- and hypo-methylation events across various transcripts.

A prospective approach to treat liver fibrosis involves the elimination of HSCs through ferroptosis, a non-apoptotic form of programmed cell death. In this regard, m6A modifications play crucial roles in DHA, erastin, and sorafenib-induced ferroptosis. These compounds, known to induce ferroptosis, elicit an elevation in METTL14 levels alongside a concurrent reduction in FTO levels, and this orchestrated modulation results in an overall increase in the levels of m6A modifications throughout the transcriptome (Shen et al., 2021). Of notable significance is the role played by YTHDF1 in the stabilization of m6A decorated beclin 1 (BECN1) transcripts, a pivotal event that sets the stage for the amplification of BECN1 expression. This increased expression in turn fuels the autophagic degradation of ferritin, ultimately culminating in the execution of ferroptosis (Shen et al., 2022).

The involvement of m6A function in HCC tumorigenesis

NAFLD and NASH stand as significant risk factors for the development of HCC, particularly in cases involving NASH-associated cirrhosis. Notably, in contrast to other etiological factors contributing to HCC, a substantial portion—approximately 30%—of NAFLD-related HCC cases emerge without the presence of cirrhosis, complicating the early-stage detection (Huang et al., 2021). m6A modifications take center stage in HCC, playing an indispensable role in tumorigenesis (Table 1). Utilizing The Cancer Genome Atlas (TCGA) database, Liu et al. (2020a) conducted an in-depth investigation that unveiled the upregulation of eleven m6A writers, readers, and erasers in the context of HCC. The five components within this spectrum—writers METTL3, KIAA1429, and ZC3H13, as well as readers YTHDF1 and YTHDF2—displayed a stronger correlation with worse outcomes due to their marked overexpressions.

Table 1.

mRNA m6A modifications in HCC

Type Regulator Effect of m6A on mRNA Target gene Effect of m6A on HCC Ref.

Writer METTL3 Translation Stat3 HCC proliferation (Liu et al., 2023a)
METTL3 Translation Yap1 Vasculogenic mimicry (Qiao et al., 2021)
METTL3 Translation Survivin Inhibits apoptosis (Zhang et al., 2022b)
METTL3 Translation EGFR Lenvatinib resistance, HCC cell viability and metastasis (Wang et al., 2023c)
METTL3 Stability Snail EMT (Xu et al., 2020a)
METTL3 Stability FEN1 HCC growth (Pu et al., 2020)
METTL3 Stability FZD10 CSCs, lenvatinib resistance (Wang et al., 2023b)
METTL3 Upregulation HIF-1α Glycolysis and glutaminolysis (Yang et al., 2021)
METTL14 Stability USP48 Impedes metabolic reprogramming and glycolysis (Du et al., 2021)
METTL14 Stability HNF3γ CSC differentiation, sorafenib sensitivity (Zhou et al., 2020)

Eraser FTO Decay SOX2, KLF4, NANOG HCC stemness (Bian et al., 2021)
ALKBH5 Decay MAP3K8 HCC cell proliferation, metastasis, and macrophage recruitment (You et al., 2022)
ALKBH5 Stability LYPD1 Proliferation and invasiveness of HCC cells (Chen et al., 2020)
WTAP Destabilization ETS1 HCC tumor growth and proliferation (Chen et al., 2019)

Reader YTHDF1 Translation ATG2A and ATG14 HCC survival under hypoxia (Li et al., 2021a)
YTHDF1
YTHDF3
Translation EGFR Lenvatinib resistance, HCC cell viability and metastasis (Hu et al., 2023; Su et al., 2021; Wang et al., 2023c)
YTHDF2 Decay IL-11 Inflammation-mediated malignancy (Hou et al., 2019)
YTHDF2 Decay SERPINE2 Disruption of vascular normalization (Chen et al., 2018b)
YTHDF2 Decay SOCS Inhibits HCC cell migration, invasion, and EMT (Chen et al., 2018b)
YTHDF2 Decay MAP3K8 HCC cell proliferation, metastasis, and macrophage recruitment (You et al., 2022)
YTHDF2 Translation OCT4 CSC phenotype (Zhang et al., 2020a)
YTHDF1
YTHDF3
Translation EGFR Lenvatinib resistance, HCC cell viability and metastasis (Hu et al., 2023; Su et al., 2021; Wang et al., 2023c)
IGF2BP2 Stability FEN1 HCC growth (Pu et al., 2020)
IGF2BP2 Stability LYPD1 Proliferation and invasiveness of HCC cells (Chen et al., 2020)
YTHDF2 Stability FZD10 CSCs, lenvatinib resistance (Wang et al., 2023b)
IGF2BP2 Stability HNF3γ CSC differentiation, sorafenib sensitivity (Zhou et al., 2020)
YTHDF1
YTHDF3
Translation EGFR Lenvatinib resistance, HCC cell viability and metastasis (Hu et al., 2023; Su et al., 2021; Wang et al., 2023c)
RALYL Stability TGF-β2 HCC stemness (Wang et al., 2021a)
HuR Destabilization ETS1
GATA3
HCC tumor growth, proliferation, and metastasis (Chen et al., 2019) (Zhang et al., 2022c)

STAT3 signaling holds a central role in obesity-driven HCC (Grohmann et al., 2018). An intriguing positive feedback loop exists between METTL3 and STAT3 in HCC, i.e., METTL3 promotes STAT3 translation through m6A methylation, while STAT3 facilitates the nuclear localization of METTL3 by upregulating WTAP (Liu et al., 2023a). Upon exposure to lipopolysaccharide stimulation, m6A modifications and the expression of GNAS undergo a substantial increase (Ding et al., 2020). The increase in GNAS expression prevents the interaction between long non-coding RNA TPTEP1 and STAT3, ultimately resulting in STAT3 downregulation. This sequence of events leads to an intensified m6A modification on GNAS, thereby amplifying STAT3 Y705 phosphorylation and consequently promoting the proliferation and invasion of inflammation-related HCC.

The upregulation of METTL3 levels contributes to HCC progression through multiple additional pathways. Vasculogenic mimicry (VM), a process in which aggressive tumor cells stimulate vasculogenic networks, plays a crucial role in the malignancy of HCC. METTL3 methylates and increases the translation of YAP1, thereby facilitating the development of VM by activating the hippo pathway (Qiao et al., 2021). Moreover, METTL3 is implicated in several dimensions of cellular regulation. It methylates the anti-apoptotic protein Survivin (Zhang et al., 2022b), promotes glycolysis through activating mTOR signaling (Lin et al., 2020), and contributes to the induction of epithelial-to-mesenchymal transition (EMT) by modulating Snail (Xu et al., 2020a). Additionally, METTL3 plays a pivotal role in reprograming intracellular metabolism by methylating HIF-1α mRNA, thereby promoting glycolysis and glutaminolysis (Yang et al., 2021). This in turn triggers the increase of m6A readers including YTHDF1. As a result, YTHDF1 facilitates the translation of autophagy-related genes like ATG2A and ATG14 to help HCC cells survive under hypoxia (Li et al., 2021a).

The increased levels of METTL3 and YTHDF1 are likely contributors to the increased translation of epidermal growth factor receptor (EGFR). This statement gains support from the study conducted by Wang et al. (2023c), wherein they observed that the upregulation of METTL3 expression resulted in Lenvatinib resistance due to the methylation of EGFR. Su et al. (2021) found that YTHDF1 promoted HCC cell viability and metastasis by facilitating the translation of EGFR. YTHDF3 also plays a role in promoting EGFR translation (Hu et al., 2023). However, an alternative study yielded contrasting results, indicating that m6A methylation and subsequent posttranscriptional destabilization of EGFR by METTL14 inhibit HCC cell migration, invasion, and EMT process (Shi et al., 2020). This discrepancy could be due to these m6A modifications being recognized by a different reader, namely YTHDF2, as elucidated by Zhong et al. (2019), who identified its role in promoting the degradation of EGFR mRNA in HCC cells. Clearly, the regulation of EGFR by m6A is a multifaced process influenced by numerous m6A readers and writers.

While HIF-1α leads to an elevation of YTHDF1 levels, HIF-2α results in a reduction of YTHDF2 levels. YTHDF2 processes the decay of interleukin 11 (IL-11) and serpin family E member 2 (SERPINE2) mRNAs. A decrease in YTHDF2 levels facilitates inflammation-mediated malignancy and disrupts vascular normalization (Hou et al., 2019). YTHDF2 is responsible for the decay of suppressor of cytokine signaling 2 (Socs2) mRNA. Consequently, the elevated METTL3 levels in HCC lead to a decrease in SOCS2 expression and an increase in tumorgenicity (Chen et al., 2018b). The last of the YTH domain family proteins, YTHDF3, is also upregulated in HCC and forms a positive feedback loop with phosphofructokinase PFKL, thereby promoting glycolysis (Zhou et al., 2022b).

Numerous other m6A-regulated proteins also contribute to the dynamics of HCC. For example, IGF2BP2 stabilizes flap endonuclease-1 (FEN1) transcripts methylated by METTL3, promoting liver cancer growth (Pu et al., 2020). IGF2BP1 stabilizes and ALKBH5 demethylates LY6/PLAUR domain containing 1 (LYPD1), which increases the proliferation and invasiveness of HCC cells. Notably, the decreased expression of ALKBH5 in HCC leads to an increase in LYPD1 methylation and expression (Chen et al., 2020). However, an alternative study presents a different perspective, indicating an upregulation of ALKBH5 in HCC. This study found that ALKBH5 upregulated MAP3K8 by preventing YTHDF2-mediated decay of MAP3K8 mRNA, thus promoting HCC cell proliferation, metastasis, and PD-L1+ macrophage recruitment (You et al., 2022). METTL14 is downregulated in HCC, and m6A methylation of USP48 by METTL14 plays a protective role in HCC as USP48 stabilizes SIRT6, which impedes metabolic reprogramming and glycolysis (Du et al., 2021). Upregulated WTAP destabilizes ETS proto-oncogene 1 (ETS1) transcripts by preventing them from binding to HuR, an RNA stabilizer that targets non-m6A modified transcripts, thus driving HCC tumor growth and proliferation (Chen et al., 2019). Similarly, upregulated KIAA1429 downregulates the tumor-suppressing gene GATA3 by adding m6A modifications that prevent GATA3 pre-mRNA from binding to HuR (Zhang et al., 2022c).

Further dysregulation of m6A modifications in HCC could potentially result from the SUMOylation of m6A readers and erasers. For example, the SUMOylation of Mettl3 by small ubiquitin-like modifier SUMO1 is increased upon mitogen stimulation (Xu et al., 2020a), and the induction of FTO SUMOylation at a specific site, which is caused by the activation of RANBP2 by SIRT1, promotes FTO degradation (Liu et al., 2020b).

Cancer stem cells

Cancer stem cells (CSCs) represent a small subset of cells within a tumor with increased tumorigenic potential compared with their more differentiated counterparts. Consequently, an increased CSC population in HCC corresponds to an enhanced cancer progression and augmented resistance to treatments. Notably, the m6A reader YTHDF2 is associated with poor survival rates in HCC patients. YTHDF2 recognizes m6A modifications on OCT4 mRNA, leading to an increase in OCT translation (Zhang et al., 2020a). As a result, this mechanism contributes to the CSC phenotype. YTHDF2 plays a distinctive role in stabilizing Frizzled-10 (FZD10) mRNA following the addition of m6A modifications by METTL3. This unique function holds immense importance, as increased FZD10 expression promotes the WNT/β-catenin and Hippo pathways. Consequently, this initiation leads to the transcription of specific genes that drive self-renewal, tumorigenesis, and metastasis of liver CSCs. Downstream of WNT/β-catenin is c-Jun, which in turn promotes the expression of METTL3, creating a positive feedback loop interconnecting METTL3, FZD10, β-catenin, and c-Jun. Furthermore, c-Jun activates the MAPK pathway, leading to lenvatinib resistance (Wang et al., 2023b). m6A modifications in CSCs are also involved in sorafenib resistance. HNF3γ, methylated by METTL14 and stabilized by IGF2BP2, promotes differentiation in both HCC cells and liver CSCs. This results in a reduction of the proportion of CSCs within HCC cells and the suppression of HCC growth. Moreover, HNF3γ promotes the expressions of OATP1B1 and OATP1B3, two major membrane transporters responsible for sorafenib uptake, thus sensitizing HCC cells to the treatment with sorafenib (Zhou et al., 2020).

FTO is also associated with HCC stemness as it promotes the expression of SRY-box transcription factor 2 (SOX2), Kruppel like factor 4 (KLF4), and NANOG by m6A demethylation (Bian et al., 2021). Within the scope of m6A modifications, RALYL, a component of the hnRNP family of m6A readers, contributes to the stabilization of TGF-β2 mRNA. This stabilization event subsequently activates the PI3K/AKT and STAT3 pathways and promotes HCC stemness (Wang et al., 2021a).

Non-coding RNAs

The scope of m6A modifications extends beyond mRNA, encompassing a diverse range of molecules such as lncRNAs, circRNAs, miRNAs, and rRNAs. These types of RNAs collectively contribute to the intricate landscape of HCC progression (Table 2).

Table 2.

Non-coding RNA networks regulated by m6A modifications in HCC

Type Regulator lncRNA/clrcRNA miRNA or RNA binding protein mRNA; downstream effectors Effect in HCC Ref.

Writer METTL3 LINC00958 miR-3619-5p HDGF; lipogenic genes HCC progression (Zuo et al., 2020)
METTL3 DUXAP8 miR-584-5p MAPK1; MAPK/ERK pathway Chemoresistance (Liu et al., 2021b)
METTL3 NIFK-AS1 miR-637 AKT1; MMP-7/9 and OATP1B1/3 HCC progression, Sorenifib resistance (Chen et al., 2021c)
METTL3 MEG3 miR544b BTG2 HCC suppression (Wu et al., 2021)
METTL3 LINC00106 let7f periostin HCC stemness and metastasis (Liang et al., 2021)
METTL3 circHPS5 miR-270 HMGA2 EMT, CSCs (Rong et al., 2021)
METTL3/14 circRNA-SORE miR-103a-2-5p miR-660-3p Wnt/β-catenin pathway Sorenifib resistance (Xu et al., 2020b)
METTL14 MIR155HG miR-223 STAT1 and PD-L1 Immune escape (Peng et al., 2022b)
METTL14 DGCR8 microRNA 126 HCC suppression (Ma et al., 2017)
METTL5 18S rRNA 80S ribosome assembly fatty acid metabolism-related mRNAs HCC oncogenic transformation (Peng et al., 2022a)
KIAA1429 circDLC1 HuR MMP1 (Liu et al., 2021a)

Eraser FTO circGPR137B miR-4739 FTO Inhibition of HCC tumorigenesis and metastasis (Liu et al., 2022b)
FTO circRNA-SORE miR-103a-2-5p miR-660-3p Wnt/β-catenin pathway Sorenifib resistance (Xu et al., 2020b)
ALKBH5 circCPSF6 PCBP2 YAP1 HCC malignancy (Chen et al., 2022)

Reader IGF2BP1 circMDK miR-346 miR-874-3p ATG16L1 PI3K/AKT/mTOR pathway HCC proliferation, apoptosis inhibition (Du et al., 2022)
IGF2BP1 LINC00106 let7f periostin HCC stemness and metastasis (Liang et al., 2021)
YTHDC1 circHPS5 miR-270 HMGA2 EMT, CSCs (Rong et al., 2021)
YTHDF1/2 circRNA-SORE miR-103a-2-5p miR-660-3p Wnt/β-catenin pathway Sorenifib resistance (Xu et al., 2020b)
YTHDF2 circCPSF6 PCBP2 YAP1 HCC malignancy (Chen et al., 2022)
IGF2BP1 circMAP3K4 N/A AIF protects HCC cells against cisplatin-induced apoptosis (Duan et al., 2022)

N/A Has_circ_0008583 miR-1301-3p METTL3 HCC progression (Wang et al., 2022a)
circMAP2K4 miR-139-5p YTHDF1 HCC progression (Chi et al., 2021)

Long non-coding RNAs (lncRNAs) both regulate m6A modifications and are affected by m6A modifications in HCC. For example, the lncRNA CASC11 recruits ALKBH5 to ubiquitin conjugating enzyme E2 T (UBE2T) mRNA and inhibits the interaction between UBE2T and YTHDF2, thus preventing m6A-dependent decay of UBE2T mRNA and promoting HCC growth and metastasis (Chen et al., 2021a). Similarly, ILF3 AS1 recruits METTL3 to ILF3 mRNA and enhances its interaction with IGF2BP2, thus promoting ILF3 mRNA stability and HCC tumorigenesis (Bo et al., 2021). AC115619 is a lncRNA that encodes the micropeptide, AC11619-22a, which binds to WTAP, preventing the assembly of the m6A methyltransferase complex and reducing global m6A levels. AC115619 is downregulated in HCC, and thus m6A levels are generally increased (Zhang et al., 2023b).

Increased m6A modifications dysregulate various lncRNAs in HCC. METTL3 and IGF2BP2 stabilize lnc-CTHCC, which binds to hnRNP K and activates YAP1 transcription, thus promoting HCC growth and metastasis (Xia et al., 2022). METTL14 and IGF2BP2 stabilize ARHGAP5-AS1, which weakens the interactions between cold shock domain containing E1 (CSDE1) and tripartite motif containing 28 (TRIM28), preventing CSDE1 degradation and coordinating oncogenic RNA regulons that activate the ERK pathway (Liu et al., 2022a). METTL16 also promotes HCC progression through destabilizing lncRNA RAB11B-AS1 (Dai et al., 2022). YTHDF2 upregulates LncAY in HCC, and through promoting BMI1 expression, lncAY activates Wnt/β-catenin signaling (Chen et al., 2021b). Increased m6A modifications on the lncRNA FAM111A-DT further HCC progression by promoting FAM111A transcription through interactions with YTHDC1 (Pu et al., 2023). ALKBH5 is generally downregulated in HCC, and decreased demethylation of LINC01468 increased its expression, driving NAFLD-HCC progression through CUL4A-linked degradation of SHIP2 and subsequent activation of the PI3K/AKT/mTOR signaling pathway (Wang et al., 2022b). Likewise, LINC02551, downregulated by ALKBH5, promotes HCC growth and metastasis by blocking the interaction between DEAD-box helicase 24 (DDX24) and an E3 ligase TRIM27, thus inhibiting DDX24 degradation (Zhang et al., 2022a). Overall, it appears that m6A modifications on lncRNAs are increased in HCC, with most m6A readers and writers being upregulated and erasers downregulated. However, a study by Kong et al. (2022) found that LINC01273, miR-600, and METTL3 form a feedback loop that reduces METTL3 levels in sorafenib-resistant HCC. LINC01273 stabilizes miR-600, thus increasing the repressive effect of miR-600 on METTL3 mRNA and downregulation of METTL3. METTL3 and YTHDF2 promote the degradation of LINC01273, and higher levels of LINC01273 decrease METTL3 levels thus further upregulating LINCO1273.

Several other lncRNAs also interact with miRNAs in ceRNA networks in which lncRNAs, by sponging miRNAs, prevent them from interacting with their mRNA targets. For example, LINC00958, stabilized by METTL3 methylation in HCC, upregulates hepatoma-derived growth factor (HDGF) expression by sponging miR-3619-5p, and increased HDGF promotes the expression of lipogenic genes and furthers HCC progression (Zuo et al., 2020). Similarly, METTL3 upregulates double homeobox A pseudogene 8 (DUXAP8), which, by sponging miR-584-5p, promotes MAPK1 expression and activates the MAPK/ERK pathway, causing chemoresistance in HCC (Liu et al., 2021b). METTL3 also regulates the NIFK-AS1/miR-637/AKT1 axis in the same way. Downstream of AKT1 includes MMP-7 and MMP-9, as well as OATP1B1 and OATP1B3, therefore METTL3-regulated NIFK-AS1 promotes HCC progression and Sorenifib resistance (Chen et al., 2021c). m6A modifications on LINC00106 by METTL3 and the subsequent stabilization by IGF2BP1 upregulate LINC00106 levels. This lncRNA sponges the miRNA let7f to activate periostin, thus promoting the properties of stemness and metastasis in HCC cells (Liang et al., 2021). METTL3-mediated m6A-modified lncRNAs can also downregulate them, as in the case of MEG3. Decreased sponging of miR544b by MEG3 enhances the binding of miR544b to BTG2, lowering BTG2 expression and furthering HCC progression (Wu et al., 2021). Peng et al. (2022b) found that lipopolysaccharide promoted m6A methylation by METTL14 of lncRNA MIR155HG, which is stabilized through a HuR-dependant pathway. By sponging miR-223, MIR155HG upregulates STAT1 and PD-L1, facilitating the immune escape of HCC cells.

m6A-regulated circRNAs are also able to act as ceRNAs and sponge miRNAs in HCC. CircMDK stabilizes IGF2BP1, by sponging miR-346 and miR-874-3p, it promotes the expression of ATG16L1. This activates the PI3K/AKT/mTOR signaling pathway, causing a greater proliferation of HCC cells, and an inhibition of apoptosis (Du et al., 2022). In a similar fashion, circRNA-SORE is upregulated by m6A modifications and activates the Wnt/β-catenin pathway by sponging miR-103a-2-5p and miR-660-3p, thus inducing Sorenifib resistance (Xu et al., 2020b). METTL3 and YTHDC1 upregulate circHPS5, which by sponging miR-270, increases HMGA2 expression, promoting EMT and CSCs in HCC (Rong et al., 2021). FTO-mediated demethylation of circGPR137B promotes its expression, and by sponging miR-4739, circGPR137B upregulates FTO. This circGPR137B/miR-4739/FTO feedback loop inhibits HCC tumorigenesis and metastasis, and circGPR137B is downregulated in HCC tissues (Liu et al., 2022b). Other circRNAs regulate m6A-related proteins as well. Has_circ_0008583 sponges miR-1301-3p to promote METTL3 expression (Wang et al., 2022a), and circMAP2K4 sponges miR-139-5p to promote YTHDF1 expression (Chi et al., 2021), both of which further HCC progression.

CircRNAs bind to not only miRNA but also RNA-binding proteins to regulate mRNA. ALKBH5 and YTHDF2 upregulate circCPSF6, which sustains the stability of YAP1 and drives HCC malignancy by competitively binding to PCBP2 (Chen et al., 2022). m6A writer KIAA1429 downregulates circDLC1, decreasing its binding to HuR and thus promoting the binding of HuR to MMP1 in HCC (Liu et al., 2021a). Another mechanism in which m6A modifications interact with circRNAs is by promoting the circRNA’s translation into a short peptide. The m6A modifications on the coding circRNA, circMAP3K4, are recognized by IGF2BP1, which promotes its translation into circMAP3K4-455aa. This peptide interacts with AIF and protects HCC cells against cisplatin-induced apoptosis, promoting cancer progression (Duan et al., 2022).

Other than lncRNAs and circRNAs, various other non-coding RNAs can be dysregulated by m6A modifications in HCC. For example, METTL14 interacts with the microprocessor protein DGCR8 microprocessor complex subunit (DGCR8) to promote the processing of primary microRNA 126 in an m6A-dependent manner, suppressing HCC progression (Ma et al., 2017). METTL5 promotes HCC tumorigenesis both in vitro and in mouse models. Depletion of METTL5-mediated 18S rRNA m6A modification results in an impaired 80S ribosome assembly and decreases the translation of fatty acid metabolism-related mRNAs (Peng et al., 2022a).

Other RNA modifications in NAFLD-NASH-HCC progression

Although m6A is one of the most well-studied RNA modifications, it remains crucial to emphasize its concurrent existence alongside other modifications. In addition to m6A, genes related to m5C and m1A could also predict the prognosis of HCC (Li et al., 2022a). The overexpression of m1A writers TRMT10C, TRMT6, and ribosomal RNA processing protein 8 (RRP8) as well as m5A reader Y box binding protein 1 (YBX1) were correlated with poor prognosis. TRMT6 and TRMT61A-mediated m1A modifications on tRNA have been discovered to enhance PPARδ translation. This translation initiation subsequently stimulates cholesterol synthesis, triggering hedgehog signaling and thereby promoting the self-renewal of liver CSCs, ultimately liver tumorigenesis (Wang et al., 2021b). Additionally, Aly/REF export factor (ALYREF) has been identified as reader protein to have an oncogenic function in HCC development through directly binding to target genes by m5C modifications (Xue et al., 2023), while m5C writer, NSUN2, can enhance the stability of H19 lncRNA. This in turn leads to the recruitment of the G3BP1 oncoprotein, therefore promoting tumor development in HCC (Sun et al., 2020). The emerging functions of m5C, m1A, m3C, ψ and m7G have also surfaced in the complex landscape of HCC. This highlights the potential efficacy of diverse targeting of these modification types in cancer cells as a promising strategy for HCC treatment (Feng et al., 2022; Li et al., 2022a). The ongoing exploration of these RNA modifications, including the unfolding biological roles of m1A, holds substantial promise for refining our understanding of HCC progression. A crucial objective is to gain comprehensive insights into the pathophysiological functions and altered pathways governed by m6A, m5C, m1A, m3C, ψ and m7G in HCC, thereby unraveling their implications for therapeutic targeting.

Avenues for therapeutic intervention

The absence of currently effective drugs for NAFLD and NASH has prompted exploration into innovative therapeutic avenues. Approaches center around targeting RNA modifications and related processes to intervene in disease progression. Investigating m6A RNA modifications and their roles in inflammation, fibrosis, and metabolism offers potential intervention points. Additionally, strategies to modulate macrophage polarization through m6A modifications could mitigate inflammatory responses. These novel directions aim to address the urgent need for effective treatments for NAFLD and NASH.

Targeting RNA methylation with small molecules

Current therapeutic pursuits have predominantly concentrated on FTO and METTL3, the two most promising therapeutic enzymes. The development of inhibitors or activators for RNA methylation regulators holds significant potential in the realm of liver cancer therapy. Notable examples include STM2457, a specific METTL3 inhibitor, and the newly introduced second-generation METTL3 inhibitor, STM3006. They both show an increased potency compared with their predecessors. The rapid metabolism of STM3006 has facilitated the clinical advancement of STC15, a clinical compound maintaining similar potency and specificity (Fiorentino et al., 2023; Guirguis et al., 2023; Pan et al., 2023b; Xu et al., 2022; Yankova et al., 2021). Currently, STC15 is undergoing Phase 1 trials, which makes a significant stride in the exploration of advanced METTL3 inhibition strategies and their potential applications in cancer therapeutics (Guirguis et al., 2023).

Moreover, the potential therapeutic benefits of RNA methylation modulation through the targeting of FTO by small-molecule inhibitors are promising. Noteworthy inhibitors in this pursuit including rhein, MO-I-500, meclofenamic acid, R-2HG, fluorescein, FB23-2, and entacapone all exhibitefficacies in suppressing FTO demethylation activities (Chen et al., 2012; He et al., 2015; Huang et al., 2019; Huang et al., 2015; Peng et al., 2019; Toh et al., 2015; Wang et al., 2015). Additionally, CS1 and CS2 have been identified as portent FTO inhibitor to sensitize leukemia cells to T cell cytotoxicity (Su et al., 2020). Recent progression also highlights the specific FTO inhibitor 18097 (Xie et al., 2022), as well as small molecules targeting key players like IGF2BP2, ALKBH5, along with the ALKBH5-specific inhibitor 20m (Fang et al., 2022). Intriguingly, the impact of STM2457 on AML development and the suppressive effects of FTO inhibitor FB23-2 on AML cell proliferation highlight the complex interplay between methylation and demethylation processes (Huang et al., 2019; Yankova et al., 2021). Nevertheless, STM2457 synergizes with anti-programmed cell death protein 1 (PD-1) to reinvigorate cytotoxic CD8+ T cells and mediate tumor regression in NAFLD-related HCC model (Pan et al., 2023b). Targeting METTL3 using the specific inhibitor STM2457 improves the sensitivity to lenvatinib in vitro and in vivo, indicating that METTL3 may be a potential therapeutic target to overcome lenvatinib resistance in HCC (Wang et al., 2023c).

Extensive research has been dedicated to investigating numerous small molecules derived from natural products as potential regulators of RNA methylations (Bedi et al., 2020; He et al., 2015; Huang et al., 2019; Peng et al., 2019; Qiao et al., 2016; Wang et al., 2018). This exploration delves into the utilization of natural product-derived small-molecule inhibitors. Among these, Rhein stands out due to its ability to competitively bind to the active site of FTO, leading to the effective inhibition of intracellular m6A demethylation effectively (Chen et al., 2012). This highlights a shifting trend in modifying drugs epigenetically, which traditionally is confined to tumor-centric domains, this field is now extending its scope to leverage the broader potential of RNA biology for advancing therapeutic strategies. This evolution holds significant promise for potential applications in HCC therapy.

The growing prominence of immunotherapy underscores the pivotal role of RNA methylation within the HCC tumor microenvironment (Xu et al., 2021a; Yin et al., 2022). Notable research underscores its potential to amplify the impact of immunotherapies by influencing immune surveillance and countering immunosuppressive cell infiltration (Cai et al., 2021) (Xu et al., 2021b). Encouragingly, the targeting of m6A and m5C emerges as a potential strategy to overcome chemo- and radio-resistance in HCC, synergizing with established therapeutic approaches (Li et al., 2022b; Zhang et al., 2023a). In light of these insights, the integration of RNA methylation modulations into liver cancer therapy holds immense promise for advancing the field and ultimately enhancing patient outcomes in HCC.

Targeting RNA methylation in non-coding RNAs and mRNA translation

Indirect regulatory effects on translation by m6A-related enzymes also warrant attention. An illustrative example is METTL3’s direct activation of the PI3K-Akt-mTOR signaling pathway in retinoblastoma cancer cells (Zhang et al., 2020b). This emphasizes the complex interplay between m6A methylation and downstream pathways. Notably, ribosome migration, crucial for translation, can be influenced by RNA’s secondary structure and binding proteins, as exemplified by m6A binding protein YTHDC2 (Hsu et al., 2017; Mao et al., 2019). This intricate process potentially underlies m6A-facilitated translation. However, the necessity of m6A for YTHDC2’s roadblock-clearing function requires further elucidation (Zhang et al., 2023c). Furthermore, while current studies showcase translational regulations by m6A-binding proteins that impact target genes, with minimal influence on mRNA levels but significant effects on protein levels, the indirect mechanisms involved demand deeper scrutiny.

In the realm of cellular regulation, non-coding RNAs like transfer RNAs (tRNAs) and ribosomal RNAs (rRNAs) play a pivotal role in post-transcriptional control of mRNAs (Li and Mason, 2014). These non-coding RNAs are extensively modified, with the functions of these modifications still not fully elucidated. By influencing the dynamics of non-coding RNAs, these modifications could potentially impact mRNA translation, either directly or indirectly (Li and Mason, 2014). This opens intriguing possibilities for RNA-based therapeutic interventions, particularly in liver diseases like NAFLD, NASH, and HCC (Yang et al., 2023b; Zhao et al., 2020). The unique modifications found in tRNAs and rRNAs could be harnessed to modulate the translation of key mRNAs central to the progression of liver diseases (Huang et al., 2023; Zhu et al., 2023). Additionally, some m6A regulators oversee various RNA modifications, each with distinct regulatory functions in mRNA translation (Sun et al., 2023). The interplay between these modifications could create a composite effect, shaping the specificity of targeted mRNAs and influencing immune cell responses within the context of liver diseases (Sun et al., 2023). As we delve deeper into the intricacies of RNA modifications, there is an emerging potential to develop RNA-based therapies tailored to liver diseases. By manipulating non-coding RNAs, scientists might be able to directly influence the translation of disease-relevant mRNAs, offering a precise approach to addressing the molecular basis of liver disorders. This innovative strategy could revolutionize our treatment approaches, leading to improved outcomes for patients with NAFLD, NASH, and HCC.

Concluding remarks

In this review, we illuminated the multifaced roles of RNA modifications, notably m6A, within the intricate landscape of NAFLD, NASH, and HCC. The dynamics of m6A within the liver exert substantial influence over critical processes including lipid metabolism, inflammation, fibrosis, and tumorigenesis. However, it is important to recognize that the impact of m6A is complex and context-dependent, capable of both protective and detrimental effects.

Extensive research has yielded valuable insights into the role of m6A in driving the progression of HCC and its associated regulatory pathways. Elevated m6A methylation has been identified as a driving force behind HCC advancement, exerting its influence over a multitude of key mRNAs and signaling pathways. However, the landscape in NAFLD and NASH appears less defined due to limited investigations and conflicting outcomes. Unraveling these intricate dynamics demands dedicated exploration into the function of m6A within HSCs, Kupffer cells and infiltrating immune cells, as well as the impact of m6A on non-coding RNAs specific to NAFLD and NASH. This encompasses cell-autonomous RNA modifications that influence other types of liver cell populations. It involves interactions between immune cells and hepatocytes, as well as the interplay between immune cells and HSCs, alongside the interaction between hepatocytes and HSCs. Additionally, it is crucial to investigate RNA modifications beyond m6A, given the limited exploration of Ψ, m1A, m5C, m6Am, and other modifications in the NAFLD-to-HCC progression. State-of-the-art techniques such as single-cell m6A sequencing offer the potential to unravel the contradictory findings concerning the influence of m6A. These approaches hold the possibility to shed light on the contradictory information surrounding the role of m6A and determine whether certain mechanisms are truly m6A dependent.

The potential for therapeutic interventions in NAFLD and NASH not only encompasses m6A-related small molecules but also extends to other potential targets including non-coding RNAs and diverse RNA modifications. This comprehensive viewpoint not only enhances our understanding but also has the potential to significantly reshape treatment approaches, offering a more promising outlook for individuals affected by these medical conditions.

Consequently, it is essential to thoroughly investigate various m6A targeting molecules for their potential efficacies in addressing these liver disorders. However, given the essential roles that m6A modifications play in regular cellular functions, a broad range of m6A targeting molecules might inadvertently induce undesirable side effects. Hence, a comprehensive understanding of m6A regulation is indispensable before designating it as a therapeutic target. Moreover, recognizing the role of m6A in liver diseases could also provide a valuable diagnostic tool, in addition to advancements in drug development.

Acknowledgement

This work was supported by NIH R01 grant (R01DK134549 to X.O.), Yale Liver Center Award NIH P30 DK034989 Morphology Core and the Cellular/Molecular Core (to X.O.), and NIH U01 grant (5U01AA026962-02 to W.Z.M. and X.O.). The Yale Liver Center core facilities were funded by NIH grant (DK P30-034989).

Footnotes

Compliance and ethics

The author(s) declare that they have no conflict of interest.

References

  1. Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, and Tavazoie SF (2015). HNRNPA2B1 is a mediator of m6A-dependent nuclear RNA processing events. Cell 162, 1299–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Bedi RK, Huang D, Eberle SA, Wiedmer L, Śledź P, and Caflisch A (2020). Small-molecule inhibitors of METTL3, the major human epitranscriptomic writer. Chemmedchem 15, 744–748. [DOI] [PubMed] [Google Scholar]
  3. Bell JL, Wächter K, Mühleck B, Pazaitis N, Köhn M, Lederer M, and Hüttelmaier S (2013). Insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs): post-transcriptional drivers of cancer progression? Cell Mol Life Sci 70, 2657–2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ben-Haim MS, Moshitch-Moshkovitz S, and Rechavi G (2015). FTO: linking m6A demethylation to adipogenesis. Cell Res 25, 3–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bian X, Shi D, Xing K, Zhou H, Lu L, Yu D, and Wu W (2021). AMD1 upregulates hepatocellular carcinoma cells stemness by FTO mediated mRNA demethylation. Clin Transl Med 11, e352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bo C, Li N, He L, Zhang S, and An Y (2021). Long non-coding RNA ILF3-AS1 facilitates hepatocellular carcinoma progression by stabilizing ILF3 mRNA in an m6 A-dependent manner. Hum Cell 34, 1843–1854. [DOI] [PubMed] [Google Scholar]
  7. Cai J, Zhou M, and Xu J (2021). N6-methyladenosine (m6A) RNA methylation regulator SNRPC is a prognostic biomarker and is correlated with immunotherapy in hepatocellular carcinoma. World J Surg Onc 19, 241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Cao X, Shu Y, Chen Y, Xu Q, Guo G, Wu Z, Shao M, Zhou Y, Chen M, Gong Y, et al. (2021). Mettl14-mediated m6A modification facilitates liver regeneration by maintaining endoplasmic reticulum homeostasis. Cell Mol Gastroenterol Hepatol 12, 633–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Chen A, Chen X, Cheng S, Shu L, Yan M, Yao L, Wang B, Huang S, Zhou L, Yang Z, et al. (2018a). FTO promotes SREBP1c maturation and enhances CIDEC transcription during lipid accumulation in HepG2 cells. Biochim Biophys Acta 1863, 538–548. [DOI] [PubMed] [Google Scholar]
  10. Chen B, Ye F, Yu L, Jia G, Huang X, Zhang X, Peng S, Chen K, Wang M, Gong S, et al. (2012). Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J Am Chem Soc 134, 17963–17971. [DOI] [PubMed] [Google Scholar]
  11. Chen F, Li M, and Wang L (2021a). LncRNA CASC11 promotes hepatocellular carcinoma progression via upregulation of UBE2T in a m6A-dependent manner. Front Oncol 11, 772671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chen L, Xia S, Wang F, Zhou Y, Wang S, Yang T, Li Y, Xu M, Zhou Y, Kong D, et al. (2023). m6A methylation-induced NR1D1 ablation disrupts the HSC circadian clock and promotes hepatic fibrosis. Pharmacol Res 189, 106704. [DOI] [PubMed] [Google Scholar]
  13. Chen M, Wei L, Law C, Tsang FH, Shen J, Cheng CL, Tsang L, Ho DW, Chiu DK, Lee JM, et al. (2018b). RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology 67, 2254–2270. [DOI] [PubMed] [Google Scholar]
  14. Chen MH, Fu LS, Zhang F, Yang Y, and Wu XZ (2021b). LncAY controls BMI1 expression and activates BMI1/Wnt/β-catenin signaling axis in hepatocellular carcinoma. Life Sci 280, 119748. [DOI] [PubMed] [Google Scholar]
  15. Chen Y, Ling Z, Cai X, Xu Y, Lv Z, Man D, Ge J, Yu C, Zhang D, Zhang Y, et al. (2022). Activation of YAP1 by N6-methyladenosine-modified circCPSF6 drives malignancy in hepatocellular carcinoma. Cancer Res 82, 599–614. [DOI] [PubMed] [Google Scholar]
  16. Chen Y, Peng C, Chen J, Chen D, Yang B, He B, Hu W, Zhang Y, Liu H, Dai L, et al. (2019). WTAP facilitates progression of hepatocellular carcinoma via m6 A-HuR-dependent epigenetic silencing of ETS1. Mol Cancer 18, 127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chen Y, Zhao Y, Chen J, Peng C, Zhang Y, Tong R, Cheng Q, Yang B, Feng X, Lu Y, et al. (2020). ALKBH5 suppresses malignancy of hepatocellular carcinoma via m6A-guided epigenetic inhibition of LYPD1. Mol Cancer 19, 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chen Y, Xiang D, Zhao X, and Chu X (2021c). Upregulation of lncRNA NIFK-AS1 in hepatocellular carcinoma by m6A methylation promotes disease progression and sorafenib resistance. Hum Cell 34, 1800–1811. [DOI] [PubMed] [Google Scholar]
  19. Cheng W, Li M, Zhang L, Zhou C, Yu S, Peng X, Zhang W, and Zhang W (2022). New roles of N6-methyladenosine methylation system regulating the occurrence of non-alcoholic fatty liver disease with N6-methyladenosine-modified MYC. Front Pharmacol 13, 973116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chi F, Cao Y, and Chen Y (2021). Analysis and validation of circRNA-miRNA network in regulating m6A RNA methylation modulators reveals circMAP2K4/miR-139-5p/YTHDF1 axis involving the proliferation of hepatocellular carcinoma. Front Oncol 11, 560506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Dai Y, Liu Y, Li J, Chen M, Huang M, Wang F, Yang Q, Yuan J, and Sun S (2022). METTL16 promotes hepatocellular carcinoma progression through downregulating RAB11B-AS1 in an m6A-dependent manner. Cell Mol Biol Lett 27, 41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. De Jesus DF, Zhang Z, Kahraman S, Brown NK, Chen M, Hu J, Gupta MK, He C, and Kulkarni RN (2019). m6A mRNA methylation regulates human β-cell biology in physiological states and in type 2 diabetes. Nat Metab 1, 765–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Ding H, Zhang X, Su Y, Jia C, and Dai C (2020). GNAS promotes inflammation-related hepatocellular carcinoma progression by promoting STAT3 activation. Cell Mol Biol Lett 25, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Du A, Li S, Zhou Y, Disoma C, Liao Y, Zhang Y, Chen Z, Yang Q, Liu P, Liu S, et al. (2022). m6A-mediated upregulation of circMDK promotes tumorigenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. Mol Cancer 21, 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, Ma J, and Wu L (2016). YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun 7, 12626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Du L, Li Y, Kang M, Feng M, Ren Y, Dai H, Wang Y, Wang Y, and Tang B (2021). USP48 is upregulated by Mettl14 to attenuate hepatocellular carcinoma via regulating SIRT6 stabilization. Cancer Res 81, 3822–3834. [DOI] [PubMed] [Google Scholar]
  27. Duan JL, Chen W, Xie JJ, Zhang ML, Nie RC, Liang H, Mei J, Han K, Xiang ZC, Wang FW, et al. (2022). A novel peptide encoded by N6-methyladenosine modified circMAP3K4 prevents apoptosis in hepatocellular carcinoma. Mol Cancer 21, 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Estes C, Anstee QM, Arias-Loste MT, Bantel H, Bellentani S, Caballeria J, Colombo M, Craxi A, Crespo J, Day CP, et al. (2018). Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J Hepatol 69, 896–904. [DOI] [PubMed] [Google Scholar]
  29. Fan C, Ma Y, Chen S, Zhou Q, Jiang H, Zhang J, and Wu F (2021). Comprehensive analysis of the transcriptome-wide m6A methylation modification difference in liver fibrosis mice by high-throughput m6A sequencing. Front Cell Dev Biol 9, 767051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fang Z, Mu B, Liu Y, Guo N, Xiong L, Guo Y, Xia A, Zhang R, Zhang H, Yao R, et al. (2022). Discovery of a potent, selective and cell active inhibitor of m6 A demethylase ALKBH5. Eur J Med Chem 238, 114446. [DOI] [PubMed] [Google Scholar]
  31. Feng Q, Wang D, Xue T, Lin C, Gao Y, Sun L, Jin Y, and Liu D (2022). The role of RNA modification in hepatocellular carcinoma. Front Pharmacol 13, 984453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Feng Y, Dong H, Sun B, Hu Y, Yang Y, Jia Y, Jia L, Zhong X, and Zhao R (2021). METTL3/METTL14 transactivation and m6A-dependent TGF-β1 translation in activated kupffer cells. Cell Mol Gastroenterol Hepatol 12, 839–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Feng Y, Guo S, Zhao Y, Dong H, Qian J, Hu Y, Wu L, Jia Y, and Zhao R (2023). DNA 5mC and RNA m6A modification successively facilitates the initiation and perpetuation stages of HSC activation in liver fibrosis progression. Cell Death Differ 30, 1211–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Fiorentino F, Menna M, Rotili D, Valente S, and Mai A (2023). METTL3 from target validation to the first small-molecule inhibitors: a medicinal chemistry journey. J Med Chem 66, 1654–1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Gan X, Dai Z, Ge C, Yin H, Wang Y, Tan J, Sun S, Zhou W, Yuan S, and Yang F (2022). FTO promotes liver inflammation by suppressing m6A mRNA methylation of IL-17RA. Front Oncol 12, 989353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Grohmann M, Wiede F, Dodd GT, Gurzov EN, Ooi GJ, Butt T, Rasmiena AA, Kaur S, Gulati T, Goh PK, et al. (2018). Obesity drives STAT-1-dependent NASH and STAT-3-dependent HCC. Cell 175, 1289–1306.e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Guirguis AA, Ofir-Rosenfeld Y, Knezevic K, Blackaby W, Hardick D, Chan YC, Motazedian A, Gillespie A, Vassiliadis D, Lam EYN, et al. (2023). Inhibition of METTL3 results in a cell-intrinsic interferon response that enhances antitumor immunity. Cancer Discov 13, 2228–2247. [DOI] [PubMed] [Google Scholar]
  38. He W, Zhou B, Liu W, Zhang M, Shen Z, Han Z, Jiang Q, Yang Q, Song C, Wang R, et al. (2015). Identification of a novel small-molecule binding site of the fat mass and obesity associated protein (FTO). J Med Chem 58, 7341–7348. [DOI] [PubMed] [Google Scholar]
  39. Hou J, Zhang H, Liu J, Zhao Z, Wang J, Lu Z, Hu B, Zhou J, Zhao Z, Feng M, et al. (2019). YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol Cancer 18, 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Hsu PJ, Zhu Y, Ma H, Guo Y, Shi X, Liu Y, Qi M, Lu Z, Shi H, Wang J, et al. (2017). Ythdc2 is an N6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res 27, 1115–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Hu B, Gao J, Shi J, Wen P, Guo W, and Zhang S (2023). m6A reader YTHDF3 triggers the progression of hepatocellular carcinoma through the YTHDF3/m6A-EGFR/STAT3 axis and EMT. Mol Carcinog 62, 1599–1614. [DOI] [PubMed] [Google Scholar]
  42. Hu J, Yang L, Peng X, Mao M, Liu X, Song J, Li H, and Chen F (2022). METTL3 promotes m6A hypermethylation of RBM14 via YTHDF1 leading to the progression of hepatocellular carcinoma. Hum Cell 35, 1838–1855. [DOI] [PubMed] [Google Scholar]
  43. Hu Y, Feng Y, Zhang L, Jia Y, Cai D, Qian SB, Du M, and Zhao R (2020). GR-mediated FTO transactivation induces lipid accumulation in hepatocytes via demethylation of m6A on lipogenic mRNAs. RNA Biol 17, 930–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Huang DQ, El-Serag HB, and Loomba R (2021). Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 18, 223–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, et al. (2018). Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol 20, 285–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Huang M, Long J, Yao Z, Zhao Y, Zhao Y, Liao J, Lei K, Xiao H, Dai Z, Peng S, et al. (2023). METTL1-mediated m7G tRNA modification promotes lenvatinib resistance in hepatocellular carcinoma. Cancer Res 83, 89–102. [DOI] [PubMed] [Google Scholar]
  47. Huang Y, Su R, Sheng Y, Dong L, Dong Z, Xu H, Ni T, Zhang ZS, Zhang T, Li C, et al. (2019). Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Huang Y, Yan J, Li Q, Li J, Gong S, Zhou H, Gan J, Jiang H, Jia GF, Luo C, et al. (2015). Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res 43, 373–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ipsen DH, Lykkesfeldt J, and Tveden-Nyborg P (2018). Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell Mol Life Sci 75, 3313–3327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang YG, et al. (2011). N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 7, 885–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, Yang C, and Chen Y (2021a). The role of m6A modification in the biological functions and diseases. Sig Transduct Target Ther 6, 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Jiang Y, Peng J, Song J, He J, Jiang M, Wang J, Ma L, Wang Y, Lin M, Wu H, et al. (2021b). Loss of Hilnc prevents diet-induced hepatic steatosis through binding of IGF2BP2. Nat Metab 3, 1569–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Kong H, Sun J, Zhang W, Zhang H, and Li H (2022). Long intergenic non-protein coding RNA 1273 confers sorafenib resistance in hepatocellular carcinoma via regulation of methyltransferase 3. Bioengineered 13, 3108–3121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Lence T, Akhtar J, Bayer M, Schmid K, Spindler L, Ho CH, Kreim N, Andrade-Navarro MA, Poeck B, Helm M, et al. (2016). m6A modulates neuronal functions and sex determination in Drosophila. Nature 540, 242–247. [DOI] [PubMed] [Google Scholar]
  55. Li D, Li K, Zhang W, Yang KW, Mu DA, Jiang GJ, Shi RS, and Ke D (2022a). The m6A/m5C/m1A regulated gene signature predicts the prognosis and correlates with the immune status of hepatocellular carcinoma. Front Immunol 13, 918140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Li J, Zhang H, and Wang H (2022b). N1-methyladenosine modification in cancer biology: current status and future perspectives. Comput Struct Biotechnol J 20, 6578–6585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Li Q, Ni Y, Zhang L, Jiang R, Xu J, Yang H, Hu Y, Qiu J, Pu L, Tang J, et al. (2021a). HIF-1α-induced expression of m6A reader YTHDF1 drives hypoxia-induced autophagy and malignancy of hepatocellular carcinoma by promoting ATG2A and ATG14 translation. Sig Transduct Target Ther 6, 76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Li S, and Mason CE (2014). The pivotal regulatory landscape of RNA modifications. Annu Rev Genom Hum Genet 15, 127–150. [DOI] [PubMed] [Google Scholar]
  59. Li S, Shen S, Xu H, Cai S, Yuan X, Wang C, Zhang X, Chen S, Chen J, Shi DL, et al. (2023). IGF2BP3 promotes adult myocardial regeneration by stabilizing MMP3 mRNA through interaction with m6A modification. Cell Death Discov 9, 164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Li X, Ding K, Li X, Yuan B, Wang Y, Yao Z, Wang S, Huang H, Xu B, Xie L, et al. (2022c). Deficiency of WTAP in hepatocytes induces lipoatrophy and nonalcoholic steatohepatitis (NASH). Nat Commun 13, 4549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Li X, Yuan B, Lu M, Wang Y, Ding N, Liu C, Gao M, Yao Z, Zhang S, Zhao Y, et al. (2021b). The methyltransferase METTL3 negatively regulates nonalcoholic steatohepatitis (NASH) progression. Nat Commun 12, 7213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Li Y, Kang X, Zhou Z, Pan L, Chen H, Liang X, Chu J, Dong S, Liu C, Yu S, et al. (2022d). The m6A methyltransferase Mettl3 deficiency attenuates hepatic stellate cell activation and liver fibrosis. Mol Ther 30, 3714–3728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Li Y, Zhang Q, Cui G, Zhao F, Tian X, Sun BF, Yang Y, and Li W (2020). m6 A regulates liver metabolic disorders and hepatogenous diabetes. Genomics Proteomics BioInf 18, 371–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Liang W, Wang Y, Zhang Q, Gao M, Zhou H, and Wang Z (2021). m6A-mediated upregulation of LINC00106 promotes stemness and metastasis properties of hepatocellular carcinoma via sponging Let7f. Front Cell Dev Biol 9, 781867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lin Y, Wei X, Jian Z, and Zhang X (2020). METTL3 expression is associated with glycolysis metabolism and sensitivity to glycolytic stress in hepatocellular carcinoma. Cancer Med 9, 2859–2867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Liu B, Cao J, Wu B, Hao K, Wang X, Chen X, and Shen Z (2023a). METTL3 and STAT3 form a positive feedback loop to promote cell metastasis in hepatocellular carcinoma. Cell Commun Signal 21, 121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Liu H, Lan T, Li H, Xu L, Chen X, Liao H, Chen X, Du J, Cai Y, Wang J, et al. (2021a). Circular RNA circDLC1 inhibits MMP1-mediated liver cancer progression via interaction with HuR. Theranostics 11, 1396–1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Liu J, Sun G, Pan S, Qin M, Ouyang R, Li Z, and Huang J (2020a). The Cancer Genome Atlas (TCGA) based m6A methylation-related genes predict prognosis in hepatocellular carcinoma. Bioengineered 11, 759–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al. (2014). A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 10, 93–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Liu J, Zhang N, Zeng J, Wang T, Shen Y, Ma C, and Yang M (2022a). N6-methyladenosine-modified lncRNA ARHGAP5-AS1 stabilises CSDE1 and coordinates oncogenic RNA regulons in hepatocellular carcinoma. Clin Transl Med 12, e1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Liu L, Gu M, Ma J, Wang Y, Li M, Wang H, Yin X, and Li X (2022b). CircGPR137B/miR-4739/FTO feedback loop suppresses tumorigenesis and metastasis of hepatocellular carcinoma. Mol Cancer 21, 149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Liu X, Feng M, Hao X, Gao Z, Wu Z, Wang Y, Du L, and Wang C (2023b). m6A methylation regulates hypoxia-induced pancreatic cancer glycolytic metabolism through ALKBH5-HDAC4-HIF1α positive feedback loop. Oncogene 42, 2047–2060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Liu X, Liu J, Xiao W, Zeng Q, Bo H, Zhu Y, Gong L, He D, Xing X, Li R, et al. (2020b). SIRT1 regulates N6-methyladenosine RNA modification in hepatocarcinogenesis by inducing RANBP2-dependent FTO SUMOylation. Hepatology 72, 2029–2050. [DOI] [PubMed] [Google Scholar]
  74. Liu Z, Lu J, Fang H, Sheng J, Cui M, Yang Y, Tang B, and Zhang X (2021b). m6A modification-mediated DUXAP8 regulation of malignant phenotype and chemotherapy resistance of hepatocellular carcinoma through miR-584-5p/MAPK1/ERK pathway axis. Front Cell Dev Biol 9, 783385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Luo Y, Zhang Z, Xiang L, Zhou B, Wang X, Lin Y, Ding X, Liu F, Lu Y, and Peng Y (2021). Analysis of N6-methyladenosine methylation modification in fructose-induced non-alcoholic fatty liver disease. Front Endocrinol 12, 780617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Ma J, Yang F, Zhou C, Liu F, Yuan J, Wang F, Wang T, Xu Q, Zhou W, and Sun S (2017). METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methyladenosine-dependent primary MicroRNA processing. Hepatology 65, 529–543. [DOI] [PubMed] [Google Scholar]
  77. Mao Y, Dong L, Liu XM, Guo J, Ma H, Shen B, and Qian SB (2019). m6A in mRNA coding regions promotes translation via the RNA helicase-containing YTHDC2. Nat Commun 10, 5332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Mendel M, Delaney K, Pandey RR, Chen KM, Wenda JM, Vågbø CB, Steiner FA, Homolka D, and Pillai RS (2021). Splice site m6A methylation prevents binding of U2AF35 to inhibit RNA splicing. Cell 184, 3125–3142.e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Pan X, Chiwanda Kaminga A, Liu A, Wen SW, Chen J, and Luo J (2020). Chemokines in non-alcoholic fatty liver disease: a systematic review and network meta-analysis. Front Immunol 11, 1802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Pan X, Li B, Wang L, Li N, Lin H, Zhang J, Du N, Zhu Y, Wu X, Hu C, et al. (2023a). Kupffer cell pyroptosis mediated by METTL3 contributes to the progression of alcoholic steatohepatitis. FASEB J 37, e22965. [DOI] [PubMed] [Google Scholar]
  81. Pan Y, Chen H, Zhang X, Liu W, Ding Y, Huang D, Zhai J, Wei W, Wen J, Chen D, et al. (2023b). METTL3 drives NAFLD-related hepatocellular carcinoma and is a therapeutic target for boosting immunotherapy. Cell Rep Med 4, 101144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Patil DP, Chen CK, Pickering BF, Chow A, Jackson C, Guttman M, and Jaffrey SR (2016). m6A RNA methylation promotes XIST-mediated transcriptional repression. Nature 537, 369–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Peng H, Chen B, Wei W, Guo S, Han H, Yang C, Ma J, Wang L, Peng S, Kuang M, et al. (2022a). N6-methyladenosine (m6A) in 18S rRNA promotes fatty acid metabolism and oncogenic transformation. Nat Metab 4, 1041–1054. [DOI] [PubMed] [Google Scholar]
  84. Peng L, Pan B, Zhang X, Wang Z, Qiu J, Wang X, and Tang N (2022b). Lipopolysaccharide facilitates immune escape of hepatocellular carcinoma cells via m6A modification of lncRNA MIR155HG to upregulate PD-L1 expression. Cell Biol Toxicol 38, 1159–1173. [DOI] [PubMed] [Google Scholar]
  85. Peng S, Xiao W, Ju D, Sun B, Hou N, Liu Q, Wang Y, Zhao H, Gao C, Zhang S, et al. (2019). Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1. Sci Transl Med 11, eaau7116. [DOI] [PubMed] [Google Scholar]
  86. Peng Z, Gong Y, Wang X, He W, Wu L, Zhang L, Xiong L, Huang Y, Su L, Shi P, et al. (2022c). METTL3-m6A-Rubicon axis inhibits autophagy in nonalcoholic fatty liver disease. Mol Ther 30, 932–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, et al. (2014). Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res 24, 177–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Powell EE, Wong VWS, and Rinella M (2021). Non-alcoholic fatty liver disease. Lancet 397, 2212–2224. [DOI] [PubMed] [Google Scholar]
  89. Pu J, Wang J, Qin Z, Wang A, Zhang Y, Wu X, Wu Y, Li W, Xu Z, Lu Y, et al. (2020). IGF2BP2 promotes liver cancer growth through an m6A-FEN1-dependent mechanism. Front Oncol 10, 578816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Pu J, Xu Z, Huang Y, Nian J, Yang M, Fang Q, Wei Q, Huang Z, Liu G, Wang J, et al. (2023). N6-methyladenosine-modified FAM111A-DT promotes hepatocellular carcinoma growth via epigenetically activating FAM111A. Cancer Sci 114, 3649–3665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Qiao K, Liu Y, Xu Z, Zhang H, Zhang H, Zhang C, Chang Z, Lu X, Li Z, Luo C, et al. (2021). RNA m6A methylation promotes the formation of vasculogenic mimicry in hepatocellular carcinoma via Hippo pathway. Angiogenesis 24, 83–96. [DOI] [PubMed] [Google Scholar]
  92. Qiao Y, Zhou B, Zhang M, Liu W, Han Z, Song C, Yu W, Yang Q, Wang R, Wang S, et al. (2016). A novel inhibitor of the obesity-related protein FTO. Biochemistry 55, 1516–1522. [DOI] [PubMed] [Google Scholar]
  93. Qin Y, Li B, Arumugam S, Lu Q, Mankash SM, Li J, Sun B, Li J, Flavell RA, Li HB, et al. (2021). m6A mRNA methylation-directed myeloid cell activation controls progression of NAFLD and obesity. Cell Rep 37, 109968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Qiu T, Wu C, Yao X, Han Q, Wang N, Yuan W, Zhang J, Shi Y, Jiang L, Liu X, et al. (2023). AS3MT facilitates NLRP3 inflammasome activation by m6A modification during arsenic-induced hepatic insulin resistance. Cell Biol Toxicol 39, 2165–2181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Rong D, Wu F, Lu C, Sun G, Shi X, Chen X, Dai Y, Zhong W, Hao X, Zhou J, et al. (2021). m6A modification of circHPS5 and hepatocellular carcinoma progression through HMGA2 expression. Mol Ther Nucleic Acids 26, 637–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Roundtree IA, Evans ME, Pan T, and He C (2017a). Dynamic RNA modifications in gene expression regulation. Cell 169, 1187–1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Roundtree IA, Luo GZ, Zhang Z, Wang X, Zhou T, Cui Y, Sha J, Huang X, Guerrero L, Xie P, et al. (2017b). YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. eLife 6, e31311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Růžička K, Zhang M, Campilho A, Bodi Z, Kashif M, Saleh M, Eeckhout D, El-Showk S, Li H, Zhong S, et al. (2017). Identification of factors required for m6A mRNA methylation in Arabidopsis reveals a role for the conserved E3 ubiquitin ligase HAKAI. New Phytol 215, 157–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Shen M, Guo M, Li Y, Wang Y, Qiu Y, Shao J, Zhang F, Xu X, Yin G, Wang S, et al. (2022). m6A methylation is required for dihydroartemisinin to alleviate liver fibrosis by inducing ferroptosis in hepatic stellate cells. Free Radic Biol Med 182, 246–259. [DOI] [PubMed] [Google Scholar]
  100. Shen M, Li Y, Wang Y, Shao J, Zhang F, Yin G, Chen A, Zhang Z, and Zheng S (2021). N6-methyladenosine modification regulates ferroptosis through autophagy signaling pathway in hepatic stellate cells. Redox Biol 47, 102151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Shi H, Wang X, Lu Z, Zhao BS, Ma H, Hsu PJ, Liu C, and He C (2017). YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res 27, 315–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Shi Y, Zhuang Y, Zhang J, Chen M, and Wu S (2020). METTL14 inhibits hepatocellular carcinoma metastasis through regulating EGFR/PI3K/AKT signaling pathway in an m6A-dependent manner. CMAR Volume 12, 13173–13184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Shu B, Zhou YX, Li H, Zhang RZ, He C, and Yang X (2021). The METTL3/MALAT1/PTBP1/USP8/TAK1 axis promotes pyroptosis and M1 polarization of macrophages and contributes to liver fibrosis. Cell Death Discov 7, 368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Simon Y, Kessler SM, Bohle RM, Haybaeck J, and Kiemer AK (2014). The insulin-like growth factor 2 (IGF2) mRNA-binding protein p62/IGF2BP2-2 as a promoter of NAFLD and HCC? Gut 63, 861–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Su R, Dong L, Li Y, Gao M, Han L, Wunderlich M, Deng X, Li H, Huang Y, Gao L, et al. (2020). Targeting FTO suppresses cancer stem cell maintenance and immune evasion. Cancer Cell 38, 79–96.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Su T, Huang M, Liao J, Lin S, Yu P, Yang J, Cai Y, Zhu S, Xu L, Peng Z, et al. (2021). Insufficient radiofrequency ablation promotes hepatocellular carcinoma metastasis through N6-methyladenosine mRNA methylation-dependent mechanism. Hepatology 74, 1339–1356. [DOI] [PubMed] [Google Scholar]
  107. Sun H, Li K, Liu C, and Yi C (2023). Regulation and functions of non-m6A mRNA modifications. Nat Rev Mol Cell Biol 24, 714–731. [DOI] [PubMed] [Google Scholar]
  108. Sun R, Tian X, Li Y, Zhao Y, Wang Z, Hu Y, Zhang L, Wang Y, Gao D, Zheng S, et al. (2022). The m6A reader YTHDF3-mediated PRDX3 translation alleviates liver fibrosis. Redox Biol 54, 102378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Sun Z, Xue S, Zhang M, Xu H, Hu X, Chen S, Liu Y, Guo M, and Cui H (2020). Aberrant NSUN2-mediated m5C modification of H19 lncRNA is associated with poor differentiation of hepatocellular carcinoma. Oncogene 39, 6906–6919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Tang Z, Sun C, Yan Y, Niu Z, Li Y, Xu X, Zhang J, Wu Y, Li Y, Wang L, et al. (2023). Aberrant elevation of FTO levels promotes liver steatosis by decreasing the m6A methylation and increasing the stability of SREBF1 and ChREBP mRNAs. J Mol Cell Biol 14, mjac061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Toh JDW, Sun L, Lau LZM, Tan J, Low JJA, Tang CWQ, Cheong EJY, Tan MJH, Chen Y, Hong W, et al. (2015). A strategy based on nucleotide specificity leads to a subfamily-selective and cell-active inhibitor of N6-methyladenosine demethylase FTO. Chem Sci 6, 112–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Vu LP, Pickering BF, Cheng Y, Zaccara S, Nguyen D, Minuesa G, Chou T, Chow A, Saletore Y, MacKay M, et al. (2017). The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 23, 1369–1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Wang F, Xie Z, Zhang N, Ding H, Xiong K, Guo L, Huang H, and Wen Z (2022a). RETRACTED ARTICLE: Has_circ_0008583 modulates hepatocellular carcinoma progression through the miR-1301-3p/METTL3 pathway. Bioengineered 13, 1185–1197. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  114. Wang H, Wang Y, Lai S, Zhao L, Liu W, Liu S, Chen H, Wang J, Du G, and Tang B (2022b). LINC01468 drives NAFLD-HCC progression through CUL4A-linked degradation of SHIP2. Cell Death Discov 8, 449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Wang J, Yang Y, Sun F, Luo Y, Yang Y, Li J, Hu W, Tao H, Lu C, and Yang JJ (2023a). ALKBH5 attenuates mitochondrial fission and ameliorates liver fibrosis by reducing Drp1 methylation. Pharmacol Res 187, 106608. [DOI] [PubMed] [Google Scholar]
  116. Wang J, Yu H, Dong W, Zhang C, Hu M, Ma W, Jiang X, Li H, Yang P, and Xiang D (2023b). N6-methyladenosine-mediated up-regulation of FZD10 regulates liver cancer stem cells’ properties and lenvatinib resistance through WNT/β-catenin and Hippo signaling pathways. Gastroenterology 164, 990–1005. [DOI] [PubMed] [Google Scholar]
  117. Wang L, Yang Q, Zhou Q, Fang F, Lei K, Liu Z, Zheng G, Zhu L, Huo J, Li X, et al. (2023c). METTL3-m6A-EGFR-axis drives lenvatinib resistance in hepatocellular carcinoma. Cancer Lett 559, 216122. [DOI] [PubMed] [Google Scholar]
  118. Wang R, Han Z, Liu B, Zhou B, Wang N, Jiang Q, Qiao Y, Song C, Chai J, and Chang J (2018). Identification of natural compound radicicol as a potent FTO inhibitor. Mol Pharm 15, 4092–4098. [DOI] [PubMed] [Google Scholar]
  119. Wang T, Hong T, Huang Y, Su H, Wu F, Chen Y, Wei L, Huang W, Hua X, Xia Y, et al. (2015). Fluorescein derivatives as bifunctional molecules for the simultaneous inhibiting and labeling of FTO protein. J Am Chem Soc 137, 13736–13739. [DOI] [PubMed] [Google Scholar]
  120. Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, Gong Z, Wang Q, Huang J, Tang C, et al. (2016). Structural basis of N6-adenosine methylation by the METTL3-METTL14 complex. Nature 534, 575–578. [DOI] [PubMed] [Google Scholar]
  121. Wang X, Wang J, Tsui YM, Shi C, Wang Y, Zhang X, Yan Q, Chen M, Jiang C, Yuan YF, et al. (2021a). RALYL increases hepatocellular carcinoma stemness by sustaining the mRNA stability of TGF-β2. Nat Commun 12, 1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Wang Y, Nakajima T, Gonzalez FJ, and Tanaka N (2020). PPArs as metabolic regulators in the liver: lessons from liver-specific PPAR-null mice. Int J Mol Sci 21, 2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Wang Y, Wang J, Li X, Xiong X, Wang J, Zhou Z, Zhu X, Gu Y, Dominissini D, He L, et al. (2021b). N1-methyladenosine methylation in tRNA drives liver tumourigenesis by regulating cholesterol metabolism. Nat Commun 12, 6314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Wei J, Harada BT, Lu D, Ma R, Gao B, Xu Y, Montauti E, Mani N, Chaudhuri SM, Gregory S, et al. (2021). HRD1-mediated METTL14 degradation regulates m6A mRNA modification to suppress ER proteotoxic liver disease. Mol Cell 81, 5052–5065.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Wei X, Zhang J, Tang M, Wang X, Fan N, and Peng Y (2022). Fat mass and obesity-associated protein promotes liver steatosis by targeting PPARα. Lipids Health Dis 21, 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Wen J, Lv R, Ma H, Shen H, He C, Wang J, Jiao F, Liu H, Yang P, Tan L, et al. (2018). Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol Cell 69, 1028–1038.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Wu J, Pang R, Li M, Chen B, Huang J, and Zhu Y (2021). m6A-induced lncRNA MEG3 suppresses the proliferation, migration and invasion of hepatocellular carcinoma cell through miR-544b/BTG2 signaling. Onco Targets Ther Volume 14, 3745–3755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Wu Z, Lu M, Liu D, Shi Y, Ren J, Wang S, Jing Y, Zhang S, Zhao Q, Li H, et al. (2023). m6A epitranscriptomic regulation of tissue homeostasis during primate aging. Nat Aging 3, 705–721. [DOI] [PubMed] [Google Scholar]
  129. Xia A, Yuan W, Wang Q, Xu J, Gu Y, Zhang L, Chen C, Wang Z, Wu D, He Q, et al. (2022). The cancer-testis lncRNA lnc-CTHCC promotes hepatocellular carcinogenesis by binding hnRNP K and activating YAP1 transcription. Nat Cancer 3, 203–218. [DOI] [PubMed] [Google Scholar]
  130. Xiang Y, Laurent B, Hsu CH, Nachtergaele S, Lu Z, Sheng W, Xu C, Chen H, Ouyang J, Wang S, et al. (2017). RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature 543, 573–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Xie G, Wu XN, Ling Y, Rui Y, Wu D, Zhou J, Li J, Lin S, Peng Q, Li Z, et al. (2022). A novel inhibitor of N6-methyladenosine demethylase FTO induces mRNA methylation and shows anti-cancer activities. Acta Pharm Sin B 12, 853–866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Xu H, Wang H, Zhao W, Fu S, Li Y, Ni W, Xin Y, Li W, Yang C, Bai Y, et al. (2020a). SUMO1 modification of methyltransferase-like 3 promotes tumor progression via regulating Snail mRNA homeostasis in hepatocellular carcinoma. Theranostics 10, 5671–5686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Xu J, Wan Z, Tang M, Lin Z, Jiang S, Ji L, Gorshkov K, Mao Q, Xia S, Cen D, et al. (2020b). N6-methyladenosine-modified CircRNA-SORE sustains sorafenib resistance in hepatocellular carcinoma by regulating β-catenin signaling. Mol Cancer 19, 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Xu Q, Xu H, Deng R, Li N, Mu R, Qi Z, Shen Y, Wang Z, Wen J, Zhao J, et al. (2021a). Landscape of prognostic m6A RNA methylation regulators in hepatocellular carcinoma to aid immunotherapy. Front Cell Dev Biol 9, 669145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Xu QC, Tien YC, Shi YH, Chen S, Zhu YQ, Huang XT, Huang CS, Zhao W, and Yin XY (2022). METTL3 promotes intrahepatic cholangiocarcinoma progression by regulating IFIT2 expression in an m6A-YTHDF2-dependent manner. Oncogene 41, 1622–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Xu Y, He X, Deng J, Xiong L, Li Y, Zhang X, Chen W, Liu X, and Xu X (2021b). Comprehensive analysis of the immune infiltrates and PD-L1 of m6A RNA methylation regulators in hepatocellular carcinoma. Front Cell Dev Biol 9, 681745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Xue C, Gu X, Zheng Q, Shi Q, Yuan X, Su Y, Jia J, Jiang J, Lu J, and Li L (2023). ALYREF mediates RNA m5C modification to promote hepatocellular carcinoma progression. Sig Transduct Target Ther 8, 130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Yang C, Hu Y, Zhou B, Bao Y, Li Z, Gong C, Yang H, Wang S, and Xiao Y (2020). The role of m6A modification in physiology and disease. Cell Death Dis 11, 960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Yang C, Zhang H, Zhang L, Zhu AX, Bernards R, Qin W, and Wang C (2023a). Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 20, 203–222. [DOI] [PubMed] [Google Scholar]
  140. Yang JJ, Wang J, Yang Y, Yang Y, Li J, Lu D, and Lu C (2022a). ALKBH5 ameliorated liver fibrosis and suppressed HSCs activation via triggering PTCH1 activation in an m6A dependent manner. Eur J Pharmacol 922, 174900. [DOI] [PubMed] [Google Scholar]
  141. Yang L, Tian S, Zheng X, Zhang M, Zhou X, Shang Y, and Han Y (2023b). N6-methyladenosine RNA methylation in liver diseases: from mechanism to treatment. J Gastroenterol 58, 718–733. [DOI] [PubMed] [Google Scholar]
  142. Yang N, Wang T, Li Q, Han F, Wang Z, Zhu R, and Zhou J (2021). HBXIP drives metabolic reprogramming in hepatocellular carcinoma cells via METTL3-mediated m6A modification of HIF-1α. J Cell Physiol 236, 3863–3880. [DOI] [PubMed] [Google Scholar]
  143. Yang S, Wei J, Cui YH, Park G, Shah P, Deng Y, Aplin AE, Lu Z, Hwang S, He C, et al. (2019). m6A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat Commun 10, 2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Yang Y, Cai J, Yang X, Wang K, Sun K, Yang Z, Zhang L, Yang L, Gu C, Huang X, et al. (2022b). Dysregulated m6A modification promotes lipogenesis and development of non-alcoholic fatty liver disease and hepatocellular carcinoma. Mol Ther 30, 2342–2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Yankova E, Blackaby W, Albertella M, Rak J, De Braekeleer E, Tsagkogeorga G, Pilka ES, Aspris D, Leggate D, Hendrick AG, et al. (2021). Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature 593, 597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Yin T, Zhao L, and Yao S (2022). Comprehensive characterization of m6A methylation and its impact on prognosis, genome instability, and tumor microenvironment in hepatocellular carcinoma. BMC Med Genomics 15, 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. You Y, Wen D, Zeng L, Lu J, Xiao X, Chen Y, Song H, and Liu Z (2022). ALKBH5/MAP3K8 axis regulates PD-L1+ macrophage infiltration and promotes hepatocellular carcinoma progression. Int J Biol Sci 18, 5001–5018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Yue Y, Liu J, Cui X, Cao J, Luo G, Zhang Z, Cheng T, Gao M, Shu X, Ma H, et al. (2018). VIRMA mediates preferential m6A mRNA methylation in 3′UTR and near stop codon and associates with alternative polyadenylation. Cell Discov 4, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Zhang C, Chen Y, Sun B, Wang L, Yang Y, Ma D, Lv J, Heng J, Ding Y, Xue Y, et al. (2017). m6A modulates haematopoietic stem and progenitor cell specification. Nature 549, 273–276. [DOI] [PubMed] [Google Scholar]
  150. Zhang C, Huang S, Zhuang H, Ruan S, Zhou Z, Huang K, Ji F, Ma Z, Hou B, and He X (2020a). YTHDF2 promotes the liver cancer stem cell phenotype and cancer metastasis by regulating OCT4 expression via m6A RNA methylation. Oncogene 39, 4507–4518. [DOI] [PubMed] [Google Scholar]
  151. Zhang H, Liu Y, Wang W, Liu F, Wang W, Su C, Zhu H, Liao Z, Zhang B, and Chen X (2022a). ALKBH5-mediated m6A modification of lincRNA LINC02551 enhances the stability of DDX24 to promote hepatocellular carcinoma growth and metastasis. Cell Death Dis 13, 926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Zhang H, Zhang P, Long C, Ma X, Huang H, Kuang X, Du H, Tang H, Ling X, Ning J, et al. (2020b). m6A methyltransferase METTL3 promotes retinoblastoma progression via PI3K/AKT/mTOR pathway. J Cell Mol Medi 24, 12368–12378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Zhang J, Gao J, Hu M, Xu S, Cheng C, Zheng W, and Zhang J (2023a). Integrated investigation of the clinical implications and targeted landscape for RNA methylation modifications in hepatocellular carcinoma. Eur J Med Res 28, 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Zhang L, Ke W, Zhao X, and Lu Z (2022b). Resina Draconis extract exerts anti-HCC effects through METTL3-m6A-Survivin axis. Phytother Res 36, 2542–2557. [DOI] [PubMed] [Google Scholar]
  155. Zhang Q, Wei T, Yan L, Zhu S, Jin W, Bai Y, Zeng Y, Zhang XF, Yin Z, Yang J, et al. (2023b). Hypoxia-responsive lncRNA AC115619 encodes a micropeptide that suppresses m6A modifications and hepatocellular carcinoma progression. Cancer Res 83, 2496–2512. [DOI] [PubMed] [Google Scholar]
  156. Zhang X, Li M, Xia L, and Zhang H (2022c). The biological function of m6A methyltransferase KIAA1429 and its role in human disease. PeerJ 10, e14334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Zhang Y, Hu W, and Li HB (2023c). RNA modification-mediated translational control in immune cells. RNA Biol 20, 603–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Zhao Z, Meng J, Su R, Zhang J, Chen J, Ma X, and Xia Q (2020). Epitranscriptomics in liver disease: basic concepts and therapeutic potential. J Hepatol 73, 664–679. [DOI] [PubMed] [Google Scholar]
  159. Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang CM, Li CJ, Vågbø CB, Shi Y, Wang WL, Song SH, et al. (2013). ALKBH5 is a mammalian RNA Demethylase that Impacts RNA Metabolism and Mouse Fertility. Mol Cell 49, 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Zhong L, Liao D, Zhang M, Zeng C, Li X, Zhang R, Ma H, and Kang T (2019). YTHDF2 suppresses cell proliferation and growth via destabilizing the EGFR mRNA in hepatocellular carcinoma. Cancer Lett 442, 252–261. [DOI] [PubMed] [Google Scholar]
  161. Zhong X, Yu J, Frazier K, Weng X, Li Y, Cham CM, Dolan K, Zhu X, Hubert N, Tao Y, et al. (2018). Circadian clock regulation of hepatic lipid metabolism by modulation of m6A mRNA methylation. Cell Rep 25, 1816–1828.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Zhou B, Liu C, Xu L, Yuan Y, Zhao J, Zhao W, Chen Y, Qiu J, Meng M, Zheng Y, et al. (2021). N6-methyladenosine reader protein YT521-B homology domain-containing 2 suppresses liver steatosis by regulation of mRNA stability of lipogenic genes. Hepatology 73, 91–103. [DOI] [PubMed] [Google Scholar]
  163. Zhou B, Luo Y, Ji N, Mao F, Xiang L, Bian H, Zheng MH, Hu C, Li Y, and Lu Y (2022a). Promotion of nonalcoholic steatohepatitis by RNA N6-methyladenosine reader IGF2BP2 in mice. Life Metab 1, 161–174. [Google Scholar]
  164. Zhou R, Ni W, Qin C, Zhou Y, Li Y, Huo J, Bian L, Zhou A, and Li J (2022b). A functional loop between YTH domain family protein YTHDF3 mediated m6A modification and phosphofructokinase PFKL in glycolysis of hepatocellular carcinoma. J Exp Clin Cancer Res 41, 334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Zhou T, Li S, Xiang D, Liu J, Sun W, Cui X, Ning B, Li X, Cheng Z, Jiang W, et al. (2020). m6A RNA methylation-mediated HNF3γ reduction renders hepatocellular carcinoma dedifferentiation and sorafenib resistance. Sig Transduct Target Ther 5, 296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Zhu S, Wu Y, Zhang X, Peng S, Xiao H, Chen S, Xu L, Su T, and Kuang M (2023). Targeting N7-methylguanosine tRNA modification blocks hepatocellular carcinoma metastasis after insufficient radiofrequency ablation. Mol Ther 31, 1596–1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Zuo X, Chen Z, Gao W, Zhang Y, Wang J, Wang J, Cao M, Cai J, Wu J, and Wang X (2020). m6A-mediated upregulation of LINC00958 increases lipogenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. J Hematol Oncol 13, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES