Abstract
Organ fibrosis, a common consequence of chronic tissue injury, presents a significant health challenge. Recent research has revealed the regulatory role of N6-methyladenosine (m6A) RNA modification in fibrosis of various organs, including the lung, liver, kidney, and heart. In this comprehensive review, we summarize the latest findings on the mechanisms and functions of m6A modification in organ fibrosis. By highlighting the potential of m6A modification as a therapeutic target, our goal is to encourage further research in this emerging field and support advancements in the clinical treatment of organ fibrosis.
Keywords: m6A modification, organ fibrosis, RNA epitranscriptomics, fibrotic diseases, therapeutic targets
Introduction
In response to organ injuries, a cascade of sequential yet interrelated stages occurs, encompassing inflammation, proliferation, and remodeling [1]. Unfortunately, even in tissues with significant regenerative potential, such as the liver, wound repair cannot solely rely on the regeneration of parenchymal cells while disregarding the role of connective tissues [2]. Under ideal conditions, the excessive accumulation of extracellular matrix (ECM) resulting from acute organ injury is broken down and eliminated with the assistance of matrix metalloproteinases, leading to the restoration of both structure and function [3]. Nevertheless, repeated insults can disrupt the finely balanced ECM environment, disturbing the normal wound-healing process and resulting in progressive scarring [4]. Significantly, the formation of pathological scars during the tissue repair process, known as fibrosis, is characterized by the excessive synthesis, production, and deposition of ECM proteins such as collagen and fibronectin. This process plays a crucial role in organ failure and disease progression [5, 6]. Fibrosis has been implicated in the pathogenesis of numerous organ pathologies, including those affecting the lungs, liver, heart, and kidneys [7]. It contributes to 45% of all deaths in developed countries [2].
Fibrosis is a condition that encompasses a multitude of cell populations, including those responsible for the pathological deposition of ECM and non-fibroblast cell types that regulate fibrosis. In the context of pulmonary fibrosis, mesenchymal cells are traditionally considered the primary source of ECM production. However, Zepp et al. [8] have reported that mesenchymal cells in distinct spatial locations exhibit unique regulatory functions. Similarly, hepatic stellate cells (HSCs) have heretofore been regarded as the principal collagen-producing cells in the liver [9]. However, recent studies by Dobie et al. [10] have provided a more detailed understanding of HSCs’ functions, indicating that only central vein-associated HSCs are the primary pathogenic collagen-producing cells. Concurrently, Ramachandran et al. [11] have identified a distinct subpopulation of macrophages characterized by TREM2+CD9+, which plays a pro-fibrogenic role in liver cirrhosis.
Moreover, other non-fibroblast cell types, including epithelial [12] and endothelial [13, 14] cells, have also demonstrated the ability to undergo phenotypic transitions. This process entails the loss of their distinctive morphology and markers, accompanied by the acquisition of myofibroblast-like characteristics. This phenomenon is recognized as epithelial-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition. These transitions have been identified as contributory factors to the development of organ fibrosis. It is important to note that the various cell populations implicated in fibrosis do not operate in isolation; rather, their intricate intercellular interactions play a pivotal role in the pathogenesis of fibrosis [15].
In addition to the diverse cell populations, numerous signaling pathways are crucial in the fibrotic process. Interestingly, despite differences in the triggers that initiate disruption of the ECM in different organs, the process of fibrosis and the associated signaling pathways exhibit a high degree of conservation [5]. Among these pathways, the transforming growth factor-β (TGF-β) signaling pathway stands out as a critical contributor to the pathogenesis of fibrogenic responses in various organs [16]. However, it does not operate in isolation. Instead, it engages in extensive crosstalk with a network of other signaling pathways, including mitogen-activated protein kinase, Wnt/β-catenin, epidermal growth factor receptor, and more. Together, these pathways collectively govern the progression of fibrosis [17].
In the context of disease progression, the activation of specific signaling pathways and expression of cellular functions are intricately intertwined with the precise regulation of particular genes. Gene expression is regulated across regulation across multiple dimensions, encompassing epigenetic, transcriptional, post-transcriptional, translational, and post-translational modifications. The realm of RNA modifications, known as “epitranscriptomics,” plays a pivotal role in this intricate regulatory network. While DNA is known for its stability, RNA exhibits a wide range of structural diversity. This includes messenger RNA (mRNA) responsible for protein coding, as well as non-coding RNAs such as microRNA (miRNA), transfer RNA, short interfering RNAs, ribosomal RNA, and long non-coding RNAs (lncRNAs). Various RNA species have been found to contain modifications, with over 170 distinct RNA modification methods documented to date [18]. Of these modifications, N6-methyladenosine (m6A) is the most prevalent. m6A modifications can influence the structure, stability, degradation, and cellular interactions of target RNA molecules [19]. Therefore, m6A modifications play a crucial role in regulating various diseases, including cancer, neurological disorders, and metabolic conditions [20–22].
The development of RNA sequencing (RNA-Seq) has provided a powerful and high-throughput method for identifying methylated sites, offering unprecedented insights into the mechanisms underlying diseases associated with m6A modifications. In recent years, research has emerged uncovering the involvement of m6A modifications in organ fibrosis. This review aims to summarize and present the latest research findings on m6A modifications in organ fibrosis, with a focus on pulmonary, hepatic, renal, and cardiac fibrosis (Table 1). The purpose of this synthesis is to encourage further investigation into the mechanisms and treatments for organ fibrosis.
Table 1.
The involvement of m6A regulators in organ fibrosis.
Organ | Type | m6A regulator | Ref. | |
---|---|---|---|---|
Lung | Writer | METTL3; METTL14; WTAP; ZC3H13; RBM15/15B; HAKAI | [23–31] | Deng, et al., 2022; Zhou, et al., 2022; Zhang, et al., 2022; Huang, et al., 2022; Zhang, et al., 2022; Wang, et al., 2022; Ning, et al., 2022; Han, et al., 2020; Ji, et al., 2023 |
Reader | YTHDC1–2; YTHDF1–3; IGF2BP1–3; FMR1; SND1 | [23–27, 29, 32, 33] | Deng, et al., 2022; Zhou, et al., 2022; Zhang, et al., 2022; Huang, et al., 2022; Zhang, et al., 2022; Ji, et al., 2023; Zhang, et al., 2021; Zhang, et al., 2024 | |
Eraser | ALKBH5; FTO | [23, 26, 27, 34] | Deng, et al., 2022; Huang, et al., 2022; Zhang, et al., 2022; Sun, et al., 2022 | |
Liver | Writer | METTL3; METTL14; METTL16; WTAP; ZC3H13 | [35–44] | Gao, et al., 2022; Feng, et al., 2021; Li, et al., 2022; Qu, et al., 2021; Shu, et al., 2022; Fan, et al., 2022; Shu, et al., 2021; Zhu, et al., 2020; Feng, et al., 2022; Chen, et al., 2023 |
Reader | YTHDF1; YTHDF3; YTHDC1 | [41, 45–47] | Chen, et al., 2023; Sun, et al., 2022; Shen, et al., 2022; Feng et al., 2023 | |
Eraser | ALKBH5; FTO | [41, 47–50] | Chen, et al., 2023; Shen, et al., 2022; Yang, et al., 2022; Chen, et al., 2023 | |
Kidney | Writer | METTL3 | [51–55] | Jung, et al., 2024; Ni, et al., 2023; Tang, et al., 2022; Liu, et al., 2020; Liu, et al., 2021; |
Reader | YTHDF1; IGF2BP2 | [53, 56, 57] | Tang, et al., 2022; Xing, et al., 2022; Chen, et al., 2023 | |
Eraser | ALKBH5; FTO | [57–61] | Chen, et al., 2023; Ning, et al., 2020; Li, et al., 2022; Yang, et al., 2022; Zang, et al., 2022 | |
Heart | Writer | METTL3 | [62–66] | Ding, et al., 2023; Zhou, et al., 2022; Li, et al., 2021; Zhuang, et al., 2023; Cheng, et al., 2023 |
Reader | IGFBP2 | [67] | Peng, et al., 2022 | |
Eraser | FTO; ALKBH5 |
[68–71] | Li, et al., 2022; Liu, et al., 2022; Mathiyalagan, et al., 2019; Zhuang, et al., 2024. |
Abbreviations: METTL: Methyltransferase-like protein; WTAP: Wilms tumor 1-associated protein; ZC3H13: Zinc finger CCCH domain-containing protein 13; RBM: The RNA-binding motif protein; YTHDC: YTH domain-containing protein; YTHDF: YTH domain-containing family protein; IGF2BP: Insulin-like growth factor binding protein; FMR1: Fragile X messenger ribonucleoprotein 1; SND1: Staphylococcal nuclease and tudor domain containing 1; ALKBH5: Alpha-ketoglutarate-dependent dioxygenase AlkB homolog 5; FTO: FTO alpha-ketoglutarate dependent dioxygenase.
Brief view of m6A modification
The identification of m6A represents a significant milestone in RNA modifications. Its initial report dates back to 1974 by Desrosiers [72]. This modification is the most abundant among mRNA modifications and has been identified in various species, including mammals, yeast, plants, flies, and bacteria [19]. Importantly, m6A decoration is a reversible process regulated by three distinct categories of proteins known as “writers,” “erasers,” and “readers” (Fig. 1).
Figure 1.
The mechanism and key regulators of m6A modification; (a). writers: methyltransferase complex composed of METTL3, METTL14, WTAP, VIRMA, ZC3H13, HAKAI, and RBM15/15B; METTL16. (b). readers: YTH, HNRNPs, IGF2BPs, FMR1, and Prrc2a. (c). erasers: FTO, ALKBH5.
Within m6A modification, “writers” and “erasers” refer to methyltransferases and demethylases, respectively, which add and remove methylation from mRNA. Meanwhile, “readers” recognize m6A modifications [73]. The primary writer complex comprises multiple subunits, with methyltransferase-like 3 (METTL3) and methyltransferase-like 14 (METTL14) serving as the core components [74]. METTL3 is the catalytic subunit responsible for methyl group transfer, while METTL14 acts as a necessary RNA-binding platform [75]. Other subunits, such as Wilms tumor 1-associating protein (WTAP), Vir-like m6A methyltransferase-associated (VIRMA), zinc finger CCCH-type containing 13 (ZC3H13), HAKAI, and RNA-binding motif protein 15/15B (RBM15/15B), are essential for maintaining the stability, functionality, localization, and specificity of the writer complex [76–80]. In addition to the canonical m6A methyltransferase complex, METTL16 has gained attention as an m6A methyltransferase that prefers cytosolic localization, facilitating the translation of mRNA transcripts [81].
The group of “readers” encompasses widely recognized YT521-B homology (YTH) family proteins, heterogeneous nuclear ribonucleoproteins (HNRNPs), insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs), fragile X mental retardation 1 (FMR1), and the recently disclosed proline-rich coiled-coil 2A (Prrc2a). These proteins affect the fate of RNA molecules, either enhancing the stability or facilitating the degradation of target transcripts [82–86]. Conversely, “erasers,” represented by m6A demethylases like fat mass and obesity-associated protein (FTO) and AlkB homolog 5 (ALKBH5), excise the methyl group from target RNA, conferring the m6A modification its reversibility and precise regulation [87, 88].
m6A in organ fibrosis
Pulmonary fibrosis
The utilization of gene sequencing technology has yielded unforeseen revelations in the exploration of disease mechanisms. Bioinformatics, which utilizes mathematical and statistical methodologies, is extensively used to predict disease occurrence and prognosis based on gene expression data. Several bioinformatics investigations employing gene expression data related to m6A regulatory proteins and methylated RNA immunoprecipitation sequencing (MeRIP-Seq) have delved into the intricate interplay between m6A modification and pulmonary fibrosis.
Regarding m6A writers, the methyltransferases METTL3, METTL14, and ZC3H13 have been identified as potential biomarkers for early diagnosis of pulmonary fibrosis and as prognostic indicators for patients with pulmonary fibrosis [23–25]. Furthermore, m6A readers, such as YTH domain-containing protein one and IGF2BP2–3, have also been identified as biomarkers for pulmonary fibrosis [24]. Additionally, the m6A eraser FTO has been associated with differences in immune cell infiltration and is crucial for the onset, progression, and prognosis of idiopathic pulmonary fibrosis (IPF) [26].
In addition to the m6A regulatory proteins, there has been significant focus on analyzing the downstream target genes influenced by m6A modification. Zhang et al. [27] investigated silica-induced pulmonary fibrosis and investigated the up-regulation of METTL3 and the downregulation of ALKBH5, FTO, YTHDF1, and YTHDF3. They utilized MeRIP-Seq and RNA-Seq to screen 18 genes with significant mRNA and m6A level changes. Functional and KEGG pathway analyses revealed that these screened genes predominantly enriched in immune response, phagocytosis, antigen processing and presentation, phagosome, lysosome, apoptosis, and other pathways. Similarly, Wang et al. [89] identified four m6A-related genes—RBM11, RBM47, RIC3, TRAF5, and ZNF14—using a multivariate Cox model, demonstrating their strong and stable predictive efficiency for IPF.
Furthermore, several downstream target RNAs of m6A have been identified and associated with different processes in the context of pulmonary fibrosis. One of these targets is CDH1, which encodes the epithelial marker E-cadherin and undergoes a reduction during the EMT process. EMT is a crucial source of fibroblasts and contributes to tissue fibrosis [90]. Ning et al. [28] demonstrated that in a model of ambient airborne fine particulate matter-induced pulmonary fibrosis model that PM2.5 increases METTL3-mediated m6A modification of CDH1. This modified CDH1 is then recognized by YTHDF2, which is negatively regulated by miR-494-3p. This recognition ultimately leads to the inhibition of E-cadherin expression, thereby promoting the progression of EMT. Whereas METTL3 has been reported to enhance the translation of the transcription factor Nrf, which can alleviate PM2.5-induced pulmonary fibrosis. Nrf is recognized by YTHDF1 and IGF2BP1 [29] (Fig. 2(a)).
Figure 2.
Mechanisms of m6A RNA Modification Involved in Pulmonary Fibrosis. (a) Stimulation by PM2.5 increases the METTL3-mediated CDH1 m6A modification recognized by YTHDF2 and inhibits E-cadherin expression and promotes the development of EMT. Increased METTL3-mediated m6A modification also can enhance the translation of the transcription factor Nrf, which attenuates PM2.5-induced pulmonary fibrosis. (b) In bleomycin (BML)-induced lung fibrosis, KCNH6 expression was enhanced by METTL3-mediated m6A modification in a YTHDF1-dependent manner. (c) Stimulated by inhalation of black carbon, the METTL3-mediated m6A modification of pri-miRNA-126 decreases and binding of DGCR8 is reduced, hindering miRNA-126 maturation, leading to activation of the PI3K-Akt–mTOR/PFKFB3 pathway and promotion of lung fibrosis. Induced by inhaled silica, the m6A eraser ALKBH5 demethylates pri-miR-320a-3p and prevents the binding of DGCR8, inhibiting miR-320a-3p maturation and leading to fibroblast activation.
In a process resembling EMT, myofibroblasts, originating from fibroblasts, represent a predominant source of collagen production. This transition is termed fibroblast-to-myofibroblast transition (FMT). Potassium Voltage-Gated Channel Subfamily H Member 6 (KCNH6), previously known for its role in glucose homeostasis and insulin secretion, has been found to regulate FMT. The expression of KCNH6 is enhanced by METTL3-mediated m6A modification in a YTHDF1-dependent manner [32] (Fig. 2(b)).
In addition to mRNA, m6A modifications in non-coding RNAs (ncRNAs) assume a substantial role in the regulation of pulmonary fibrosis. The PI3K-Akt–mTOR/PFKFB3 pathway regulates pulmonary fibrosis by modulating fibroblast aerobic glycolysis and collagen synthesis [91]. In the context of pulmonary fibrosis, m6A modification of pri-miRNA-126 decreases. This, coupled with a reduction in its binding with DGCR8, a key component involved in mRNA maturation [92], hinders the maturation of miRNA-126. Consequently, this leads to the activation of the PI3K-Akt–mTOR/PFKFB3 pathway [30] (Fig. 2(c)).
Similarly, Sun et al. [34] demonstrated that miR-320a-3p regulates fibrosis by targeting the 3′-untranslated region (UTR) of FOXM1 mRNA. The m6A eraser ALKBH5 regulates the maturation of miR-320a-3p (Fig. 2(c)). Specifically, ALKBH5 demethylates pri-miR-320a-3p and prevents its binding with DGCR8, thereby inhibiting the maturation of miR-320a-3p (Fig. 2(c)). Additionally, Wang et al. [31] identified two circular RNAs, namely hsa_circ_0000672 and hsa_circ_0005654, as participants in pulmonary fibrosis. This revelation was facilitated through the utilization of m6A-circRNA epitranscriptomic chips. The study suggested that METTL3 promotes SiO(2)-induced pulmonary fibrosis through m6A modifications in these two circRNA.
In addition to their canonical role in m6A regulation, RNA-binding proteins involved in m6A modification pathways have also been demonstrated to contribute pulmonary fibrosis independently of m6A modification. For example, YTHDC1 has been reported to mitigate cellular senescence-induced pulmonary fibrosis by facilitating the interaction between DNA topoisomerase 2-binding protein 1 (TopBP1) and Meiotic Recombination 11 (MRE11), thereby enhancing DNA damage repair [33]. These findings further expand the functional scope of m6A regulators in organ fibrosis.
Hepatic fibrosis
Given the high prevalence of liver cirrhosis, extensive research has focused on m6A modifications in hepatic fibrosis. Using m6A-seq and RNA-seq methodologies, numerous studies have conducted screenings to identify distinct m6A methylation patterns associated with hepatic fibrosis. These patterns have demonstrated enrichment in various processes intricately linked to hepatic fibrosis [93, 94].
m6A modification assumes a dual role in the advancement and regression of hepatic fibrosis. Hepatic fibrosis progression is associated with oxidative stress and cytochrome metabolism, while fibrosis regression is intricately linked to immune response and apoptosis [95]. Chronic hepatitis B is among the most common causes of hepatic fibrosis. Gao et al. [35] demonstrated that METTL16 regulates the expression of the chronic hepatitis B-related gene HLA-DPB1 and contributes to the progression of hepatic fibrosis. Significantly, TGF-β1, a pivotal constituent of the canonical fibrotic signaling pathway, emanates from activated Kupffer cells (KC) and propagates hepatic fibrosis. Activation of METTL3 and METTL14 amplifies TGF-β1 mRNA levels in an m6A-dependent manner, further emphasizing the involvement of m6A modifications in hepatic fibrosis [36].
Since HSCs constitute the principal source of ECM and assume a pivotal role in hepatic fibrosis, considerable attention has been directed toward comprehending the regulatory role of m6A modification in HSC activation, thereby elucidating the mechanism underlying hepatic fibrosis.
Fan et al. [37] conducted a comprehensive analysis by examining 90 genes that displayed noteworthy alterations in both m6A and mRNA expression levels by applying m6A-seq and RNA-seq techniques. Their investigation substantiated distinctive expression patterns of m6A regulators, wherein the decreased expression of WTAP was determined to foster HSC activation, consequently instigating hepatic fibrosis. Conversely, the m6A methyltransferase METTL3 deficiency was shown to inhibit HSC activation in an m6A-dependent manner by targeting Lats2, a key modulator of the Hippo/YAP signaling pathway [38].
HSCs can also be activated by several types of immune cells, among which hepatic macrophages and their cross-talk with HSCs play a pivotal role in hepatic fibrosis. For example, exosomes derived from lipopolysaccharide (LPS)-induced macrophages have been observed to transfer miRNAs to promote HSC proliferation and activation during the progression of liver fibrosis [96]. It is noteworthy that METTL3 enhances the M1 polarization and inhibits M2 polarization of macrophages by directly methylating signal transducer and activator of transcription 1 (STAT1) mRNA at its coding sequence and 3′-untranslated regions, which encodes the master transcription factor facilitating M1 macrophage polarization [97]. In a study conducted by Qu et al. [39], the online prediction tool Starbase was used to report the potential binding of METTL3 and the lncRNA NEAT1, which has been demonstrated to accelerate the progression of liver fibrosis [98]. Additionally, METTL3 has been shown to promote NEAT1 expression in Kupffer cells and LPS-treated Kupffer cell exosomes, which can further enhance the activation of HSCs [40].
Several studies have also investigated the impact of m6A readers on HSC activation. For instance, Feng et al. [45] underscored the pivotal role of m6A modification in HSC activation and the excessive ECM production, which is mediated by the m6A reader YTHDF1. Furthermore, Chen et al. [41] reported the involvement of the circadian rhythm in the progression of liver fibrosis. They demonstrated that the downregulation of the liver clock gene NR1D1 promoted persistent activation of HSCs and liver fibrosis induced by CCl4. Importantly, they found that the expression of NR1D1 is regulated by m6A modification in a YTHDC1-dependent manner.
Furthermore, the m6A demethylase ALKBH5 has been implicated in the activation of HSCs through various downstream factors. Yang et al. [48] demonstrated, in a mouse model of CCl4-induced liver fibrosis, that ALKBH5 instigated Patched 1 (PTCH1) activation, a negative regulator of the hedgehog signaling pathway, suppressing HSC activation. Similarly, Chen et al. [49] reported that radiation could induce ALKBH5 expression, leading to TIRAP demethylation and subsequently activating downstream signaling pathways and HSCs.
Given the central role of mitochondrial metabolism in the onset and progression of chronic liver diseases [99], researchers have elucidated the involvement of m6A modification in the governance of mitochondrial function and its repercussions on hepatic fibrosis. Peroxiredoxins (PRDXs), a peroxidase superfamily crucial for inhibiting oxidative stress [100], have been investigated. Sun et al. [46] demonstrated that PRDX3 plays a role in restraining HSCs activation by regulating mitochondrial ROS production, thereby attenuating hepatic fibrosis. Notably, the expression of PRDX3 was found to be controlled by YTHDF3 in an m6A-dependent manner. Furthermore, Wang et al. [50] reported that the loss of ALKBH5 enhances the m6A modification of Dynamin-related protein 1 (Drp1), leading to increased expression. Drp1, in turn, mediates mitochondrial fission, thereby promoting HSC proliferation and migration in hepatic fibrosis.
In recent years, emerging forms of programmed cell death have undergone extensive study, revealing that ferroptosis and pyroptosis are implicated in hepatic fibrosis and can be modulated by m6A modification [42, 47]. Shen et al. [47] demonstrated that increased m6A modification of the autophagy-related gene, BECN1, plays a crucial role in the mechanism of dihydroartemisinin for treating hepatic fibrosis. This mechanism involves preventing the activation of HSCs through the ferroptosis pathway. They further established that the reduction of FTO mediates dihydroartemisin-induced up-regulation of m6A levels, and YTHDF1 is the key reader responsible for maintaining the stability of BECN1 mRNA. Shu et al. [42] conducted a study revealing that the METTL3/MALAT1/PTBP1/USP8/TAK1 axis promotes pyroptosis and inflammation in macrophages, leading to the activation of HSCs and their involvement in liver fibrosis.
Another mechanism involving m6A regulation in liver fibrosis was reported by Feng et al. [43], who investigated the impact of chronic corticosterone exposure on inflammation and fibrosis in the chicken liver. Their results suggested that m6A modification on heat shock proteins (HSPs) transcripts reduces HSPs, consequently suppressing the cytoprotective effect of HSPs in this model. Additionally, as mentioned in the pulmonary fibrosis section, the maturation of miRNA regulated by METTL3-mediated m6A modification was also found to be involved in hepatic fibrosis [44].
Renal fibrosis
m6A RNA methylation was found to be significantly elevated in renal fibrosis, accompanied by increased expression of key m6A regulators, including METTL3, METTL14, ALKBH5, YTHDF1, and YTHDF3 [51]. It is noteworthy that the authors demonstrated that genetic knockdown and pharmacological inhibition of METTL3 by STM2457, a selective inhibitor of METTL3 catalytic activity, suppressed the TGF-β-induced fibrosis. Similar results were observed in a mouse model of UUO-induced kidney fibrosis. Furthermore, the researchers observed that TGF-β enhances m6A methylation of the NET1 transcript in the vicinity of its transcription termination site, thereby promoting mRNA stability and contributing to the progression of renal fibrosis. These findings highlight the pivotal role of m6A modification in renal fibrosis.
Additionally, Ni et al. [52] reported the hyperactivation of METTL3 and m6A modifications in TGF-β-induced HK-2 cells and the UUO-induced mouse model. The researchers also identified another target of METTL3-regulated m6A modification, Ena/VASP-like (EVL) mRNA, which was stabilized by METTL3-induced m6A modification. In this study, the authors identified a TCM monomer, isoforsythiaside that can inhibit METTL3 activity using molecular docking and virtual screening. They found that pharmacological inhibition of METTL3 activity suppressed EVL m6A and subsequently reduced its interaction with Smad7.
The differential expression of m6A regulators has led to bioinformatic investigations to predict pivotal genes that could serve as diagnostic biomarkers for renal fibrosis and identify potential drug targets through machine-learning models [52]. For instance, Xing et al. [101] identified increased expression of YTHDF1 in human fibrotic kidneys. They established a correlation with Yes-associated protein (YAP), a pivotal regulator of ECM production triggered by myofibroblast transformation. Their experiments in mice and cultured cells supported the role of YTHDF1 in promoting renal fibrosis by regulating YAP. The co-location and specific binding of YTHDF1 to YAP were further confirmed through immunofluorescence and RNA-binding protein immunoprecipitation. Nonetheless, YTHDF1 has also been reported to play a role in mitigating renal interstitial fibrosis induced by diabetic nephropathy by enhancing the stability of nuclear receptor-binding SET domain protein 2 (NSD2) through METTL3-mediated m6A modification [56].
The significance of the m6A eraser in regulating renal fibrosis is noteworthy. However, the involvement of various m6A regulators and their specific downstream target genes can lead to divergent regulatory outcomes. Ning et al. [53] demonstrated that genistein promoted the restoration of ALKBH5 decline and reduced renal fibrosis, possibly by regulating EMT-related factors in an m6A-dependent manner. However, in an ischemia–reperfusion injury (IRI) model, Chen et al. [58] reported that ALKBH5 knockout alleviated IRI-induced renal fibrosis. Another m6A eraser, FTO, also contributes to renal fibrosis by regulating different mRNAs [57, 59]. Zhang et al. [57] showed that FTO reduced the m6A modification of KCNK5, leading to the up-regulation of TWIK-related acid-sensitive K(+) channel-2 (TASK-2, encoded by KCNK5) and promoting renal fibrosis. Meanwhile, Yang et al. [59] demonstrated that the decrease in FTO expression was associated with the anti-fibrotic effect of canagliflozin, achieved by stabilizing SQSTM1 mRNA.
Furthermore, m6A modification of non-coding RNA has also been implicated in the development and progression of renal fibrosis. In animal and cell culture models of renal fibrosis, m6A demethylase FTO has been shown to reduce the m6A decoration of lncRNA GAS5, promoting the EMT process [60]. Liu et al. [61] unveiled that lncRNA MALAT1 serves as a miRNA sponge, exerting inhibitory effects on miR-145, which, in turn, attenuates FAK expression, ultimately culminating in the promotion of EMT and renal fibrosis. Furthermore, they discovered that METTL3 plays a role in the upregulation of MALAT1. Similarly, METTL3 was found to fulfill a pivotal catalytic role in m6A modification in renal fibrosis, instigating obstructive renal fibrosis by promoting the maturation of miR-21-5p, consequently activating the SPRY1/ERK/NF-kB signaling pathway [54].
Cardiac fibrosis
Cardiac fibroblasts are the critical cells involved in myocardial fibrosis. The relationship between m6A modification and cardiac fibrosis has been mainly focused on the regulation of cardiac fibroblast activation. Numerous studies have documented that the m6A methyltransferase METTL3 can induce cardiac fibroblast activation in various mechanisms, ultimately promoting myocardial fibrosis [55, 62, 63]. For instance, Li et al. [63] discovered that METTL3-mediated m6A modification promotes cardiac fibroblast activation and collagen accumulation, and silencing METTL3 attenuates cardiac fibrosis, possibly through the regulation of m6A modification of collagen-related genes. Zhou et al. [62] revealed that enhanced glycolysis was crucial for fibroblast proliferation, and METTL3 could boost glycolysis by repressing the androgen receptor, which interacts with the glycolysis marker HIF-1α, inhibiting its activation. Ding et al. [55] identified a differentially expressed gene, IGFBP3, in cardiac fibrosis and demonstrated that its knockdown suppresses the migration and proliferation of cardiac fibroblasts. In their study, the researchers observed abnormal m6A modification patterns in models of cardiac fibrosis. They also demonstrated that silencing METTL3 could downregulate the expression of IGFBP3. Moreover, Tenascin-C, an ECM modulator implicated in cardiac fibrosis, was reported to be stabilized by METTL3-mediated m6A modification [64]. In their study, Zhuang et al. [65] investigated the upstream regulatory factor of METTL3 in cardiac fibrosis. They showed that the lncRNA MetBil protects METTL3 from degradation through the ubiquitin-proteasome pathway, thereby promoting cardiac fibrosis.
In line with the association between m6A levels and myocardial fibrosis, multiple studies have also reported the involvement of the m6A demethylase FTO in the attenuation of myocardial fibrosis. Li et al. [66] demonstrated that circCELF reduces the m6A modification of DKK2 by up-regulating FTO expression, thereby suppressing the binding of miR-636 to DKK2 and increasing the expression of DKK2, which plays an anti-fibrotic role in cardiac fibroblast activation by inhibiting the Wnt/β-catenin signaling pathway [68].
Mathiyalagan et al. [102] discovered that decreased FTO expression increased m6A modification and was associated with cardiac fibrosis and enhanced angiogenesis after myocardial infarction. On the other hand, in heart failure with preserved ejection fraction, Liu et al. [69] demonstrated that exercise increased total m6A levels and decreased FTO expression. Overexpression of FTO reversed the benefits of exercise, leading to enhanced myocyte apoptosis, myocyte hypertrophy, and myocardial fibrosis.
Additionally, the m6A demethylase ALKBH5 has been linked to the development of cardiac fibrosis by influencing the macrophage-to-myofibroblast transition (MMT) [70]. In this study, increased expression of ALKBH5 was observed during angiotensin II-induced MMT, while macrophage-specific knockout of ALKBH5 suppressed this transition and subsequently inhibited cardiac fibrosis. Furthermore, interleukin-11 (IL-11) was identified as a target of ALKBH5-mediated m6A demethylation through RNA immunoprecipitation sequencing, where ALKBH5 enhanced IL-11 mRNA stability and elevated its protein levels.
Aside from the m6A writer and eraser, Peng et al. [71] reported that the lncRNA Airn inhibited cardiac fibroblast activation, alleviating diabetic cardiac fibrosis. Mechanistically, Airn stabilized p53 mRNA in an m6A-dependent manner through the recognition of the m6A reader IGF2BP2, resulting in cardiac fibroblast cell cycle arrest and the suppression of cardiac fibrosis.
Collectively, the existing studies suggest that m6A modification plays a role in promoting cardiac fibrosis. This highlights the potential of key regulators of m6A modification as targets for treating cardiac fibrosis. Nevertheless, it is crucial to consider the widespread occurrence of m6A modification and its multifaceted functions, as a broad spectrum often signifies potential adverse impacts on the functionality of other tissues and organs. Further research is required to understand the various roles of m6A in the heart comprehensively.
Others
Numerous studies have investigated the role of m6A modification in fibrosis affecting various organs and tissues. Liu et al. [67] conducted a bioinformatics analysis to identify differentially expressed mRNA transcripts with unique m6A modification patterns in hyperplastic scars compared to normal skin. Notably, these modified m6A genes were found to be significantly enriched in pathways related to fibrosis.
In the study of vitreoretinal fibrosis, a condition characterized by abnormal growth of fibrous tissue within the vitreous of the eye, Ma et al. [103] observed a decrease in levels of METTL3 and a reduction in total m6A abundance. Importantly, increasing the expression of METTL3 reduced the EMT induced by TGF-β1, thus ameliorating proliferative vitreoretinopathy. Conversely, in the context of subretinal fibrosis, which is characterized by fibrotic changes in the subretinal space, METTL3 was observed to facilitate fibrosis progression. This effect was achieved through the induction of m6A modification in high mobility group AT-hook 2 (HMGA2), ultimately leading to the activation of SNAIL, a transcription factor known for inducing EMT [104].
Additionally, Li et al. [105] discovered a correlation between m6A modification and arecoline-induced oral submucosal fibrosis. They found that arecoline, a compound frequently linked to this condition, enhanced m6A levels by modulating the TGF-β signaling pathway. Consequently, they identified MYC as a downstream target of m6A modification, a process regulated by METTL14. Importantly, most of these published studies have primarily focused on assessing global changes in m6A levels and alterations in key m6A regulators, while the specific downstream targets remain an area ripe for exploration.
Conclusion and perspective
Research has shown that fibrosis progression often leads to organ dysfunction, with the lung, liver, kidney, and heart being extensively studied examples. Recent research has unveiled the pivotal regulatory role of m6A modification in fibrosis processes across various organs. Importantly, the principal m6A regulators, encompassing writers, readers, and erasers, are all implicated in the regulation of organ fibrosis. This widespread involvement can be attributed to the ubiquitous and abundant nature of m6A modification. However, it is crucial to acknowledge that the mechanisms of fibrosis, mediated by m6A regulators in different organs, are intricate and subject to variation due to distinct tissue and cellular environments [83]. As highlighted in the “m6A in organ fibrosis” section, even within the same organ, similar types of m6A regulators may produce diverse regulatory outcomes in fibrosis by interacting with specific downstream target RNAs.
Expression patterns of m6A regulators are primarily context-dependent and influenced by a myriad of factors. The distribution and functionality of m6A regulators differ among various cell lines [106]. For instance, METTL3 is mainly localized in the nucleus, where it forms a complex with other writers [107]. However, a fraction of METTL3 can localize to the cytoplasm and interact with downstream RNA independently of its catalytic activity, effectively functioning as an m6A reader rather than a writer [108, 109]. Additionally, the activation of m6A regulators depends on the specific cellular environment. Some m6A readers are constitutively active, while others are induced by distinct stimuli, such as heat shock-induced up-regulation and translocation of YTHDF2 or infection-induced overexpression of YTHDF1–2 [110, 111]. Furthermore, the functional impact of m6A modification on organ fibrosis occurs through interactions with specific downstream RNAs, each playing unique roles. Therefore, it is not surprising to observe divergent regulatory effects on fibrosis by the same m6A regulators.
Despite the encouraging potential of m6A modulation in the context of fibrosis, several significant challenges must be addressed prior to its clinical translation. These include the potential for off-target effects and the difficulty of efficiently delivering m6A modulators to individual transcripts. To address these challenges, novel strategies are being developed. For example, CRISPR-Cas13b-based delivery systems are being investigated for their capacity to precisely target and modify m6A modifications on specific transcripts, thereby improving specificity and reducing off-target effects [112, 113]. Furthermore, developments in nanoparticle-based delivery systems are intended to augment the efficacy of m6A modulator delivery to intended cells [114]. It is imperative that these challenges be addressed through such advanced strategies if m6A modulation is to be successfully applied in a clinical context.
The investigation of m6A in organ fibrosis presents a promising avenue for translational research, offering new possibilities for therapeutic intervention. However, the realization of this potential will necessitate not only further preclinical and clinical studies but also robust interdisciplinary collaboration. Engaging experts in molecular biology, pharmacology, clinical medicine, and engineering materials will be crucial for overcoming current challenges and advancing m6A-targeted therapies from bench to bedside.
Key Points
Fibrosis is a common consequence of chronic tissue injury affecting multiple organs, including the lung, liver, kidney, and heart, posing a major challenge to human health.
The m6A modifications involve three classes of proteins: writers, erasers, and readers, which are involved in adding, removing, and recognizing m6A marks, respectively.
N6-methyladenosine (m6A) RNA modifications play a key regulatory role in fibrosis in different organs, and the modifications can affect RNA structure, stability, degradation, and intracellular interactions.
The m6A modification has great potential as a therapeutic target for fibrosis in chronic diseases.
Acknowledgments
This work was supported by the National Natural Science Foundation of China (82360385 to S.C., 82060023 and 82160133 to Y.L.); Jiangxi Provincial Natural Science Foundation (20202ACBL206015 to S.C., 20224BAB206007, and 20212ACB216005 to Y.L.); and the Graduate Innovative Special Fund of Jiangxi Province [Yc2023-B072 to X.H.].
Contributor Information
Xiangfei Huang, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Zilu Yu, Queen Mary School, Medical College, Nanchang University, 1299 Xuefu Road, Honggutan District, Nanchang 330031, China.
Juan Tian, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Tao Chen, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Aiping Wei, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Chao Mei, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Shibiao Chen, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Yong Li, Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China.
Author contributions
YL and SC conceived and conceptualized ideas, compiled information, and edited the manuscript. XH and ZY conceptualized information, prepared the illustration, analyzed data, and wrote the original draft. JT participated in the writing and editing of the manuscript. AW, TC, and CM reviewed and edited the manuscript. All authors approved the submission to this journal.
Xiangfei Huang (Conceptualization [equal], Resources [equal], Writing—original draft [equal]), Zilu Yu (Resources [equal], Writing—original draft [equal], Writing—review & editing [equal]), Juan Tian (Resources [supporting], Writing—review & editing [supporting]), Tao Chen (Writing—review & editing [supporting]), Aiping Wei (Writing—review & editing [supporting]), Chao Mei (Writing—review & editing [supporting]), Shibiao Chen (Funding acquisition [supporting], Project administration [supporting], Writing—review & editing [supporting]), and Yong Li (Conceptualization [equal], Funding acquisition [lead], Investigation [lead], Project administration [lead], Supervision [lead], Writing—review & editing [lead]).
Funding
None declared.
Data availability
The data supporting this review are from previously reported studies and datasets, which have been cited. The processed data are available from the corresponding author upon request.
References
- 1. Eming SA, Wynn TA, Martin P. Inflammation and metabolism in tissue repair and regeneration. Science 2017;356:1026–30. [DOI] [PubMed] [Google Scholar]
- 2. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 2008;214:199–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Friedman SL, Sheppard D, Duffield JS. et al. Therapy for fibrotic diseases: nearing the starting line. Sci Transl Med 2013;5:167sr161. [DOI] [PubMed] [Google Scholar]
- 4. Schuppan D, Ruehl M, Somasundaram R. et al. Matrix as a modulator of hepatic fibrogenesis. Semin Liver Dis 2001;21:351–72. [DOI] [PubMed] [Google Scholar]
- 5. Mutsaers HA, Olinga P. Editorial: organ fibrosis: triggers, pathways, and cellular plasticity. Front Med (Lausanne) 2016;3:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012;18:1028–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Ghafouri-Fard S, Abak A, Talebi SF. et al. Role of miRNA and lncRNAs in organ fibrosis and aging. Biomed Pharmacother 2021;143:112132. [DOI] [PubMed] [Google Scholar]
- 8. Zepp JA, Zacharias WJ, Frank DB. et al. Distinct mesenchymal lineages and niches promote epithelial self-renewal and Myofibrogenesis in the lung. Cell 2017;170:1134–1148.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Friedman SL, Roll FJ, Boyles J. et al. Hepatic lipocytes: the principal collagen-producing cells of normal rat liver. Proc Natl Acad Sci U S A 1985;82:8681–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Dobie R, Wilson-Kanamori JR, Henderson BEP. et al. Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis. Cell Rep 2019;29:1832–1847.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ramachandran P, Dobie R, Wilson-Kanamori JR. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 2019;575:512–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2014;15:178–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Manetti M, Romano E, Rosa I. et al. Endothelial-to-mesenchymal transition contributes to endothelial dysfunction and dermal fibrosis in systemic sclerosis. Ann Rheum Dis 2017;76:924–34. [DOI] [PubMed] [Google Scholar]
- 14. Zeisberg EM, Tarnavski O, Zeisberg M. et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med 2007;13:952–61. [DOI] [PubMed] [Google Scholar]
- 15. Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020;587:555–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020;217:e20190103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol 2016;12:325–38. [DOI] [PubMed] [Google Scholar]
- 18. Boccaletto P, Stefaniak F, Ray A. et al. MODOMICS: a database of RNA modification pathways. 2021 update. Nucleic Acids Res 2022;50:D231–d235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Oerum S, Meynier V, Catala M. et al. A comprehensive review of m6A/m6Am RNA methyltransferase structures. Nucleic Acids Res 2021;49:7239–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Barbieri I, Kouzarides T. Role of RNA modifications in cancer. Nat Rev Cancer 2020;20:303–22. [DOI] [PubMed] [Google Scholar]
- 21. Haruehanroengra P, Zheng YY, Zhou Y. et al. RNA modifications and cancer. RNA Biol 2020;17:1560–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Destefanis E, Avşar G, Groza P. et al. A mark of disease: how mRNA modifications shape genetic and acquired pathologies. RNA 2021;27:367–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Deng MS, Chen KJ, Zhang DD. et al. m6A RNA methylation regulators contribute to predict and as a therapy target of pulmonary fibrosis. Evid Based Complement Alternat Med 2022;2022:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 24. Zhou Y, Fang C, Sun Q. et al. Relevance of RNA N6-Methyladenosine regulators for pulmonary fibrosis: implications for chronic hypersensitivity pneumonitis and idiopathic pulmonary fibrosis. Front Genet 2022;13:939175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Zhang J, Zhang Y, Wang Z. et al. Genes related to N6-methyladenosine in the diagnosis and prognosis of idiopathic pulmonary fibrosis. Front Genet 2022;13:1102422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Huang G, Huang S, Cui H. Effect of M6A regulators on diagnosis, subtype classification, prognosis and novel therapeutic target development of idiopathic pulmonary fibrosis. Front Pharmacol 2022;13:993567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Zhang Y, Gu P, Xie Y. et al. Insights into the mechanism underlying crystalline silica-induced pulmonary fibrosis via transcriptome-wide m(6)a methylation profile. Ecotoxicol Environ Saf 2022;247:114215. [DOI] [PubMed] [Google Scholar]
- 28. Ning J, Du H, Zhang Y. et al. N6-Methyladenosine modification of CDH1 mRNA promotes PM2.5-induced pulmonary fibrosis via mediating epithelial mesenchymal transition. Toxicol Sci 2022;185:143–57. [DOI] [PubMed] [Google Scholar]
- 29. Ji D, Hu C, Ning J. et al. N(6)-methyladenosine mediates Nrf2 protein expression involved in PM2.5-induced pulmonary fibrosis. Ecotoxicol Environ Saf 2023;254:114755. [DOI] [PubMed] [Google Scholar]
- 30. Han B, Chu C, Su X. et al. N(6)-methyladenosine-dependent primary microRNA-126 processing activated PI3K-AKT-mTOR pathway drove the development of pulmonary fibrosis induced by nanoscale carbon black particles in rats. Nanotoxicology 2020;14:1–20. [DOI] [PubMed] [Google Scholar]
- 31. Wang S, Luo W, Huang J. et al. The combined effects of circular RNA methylation promote pulmonary fibrosis. Am J Respir Cell Mol Biol 2022;66:510–23. [DOI] [PubMed] [Google Scholar]
- 32. Zhang JX, Huang PJ, Wang DP. et al. M(6)a modification regulates lung fibroblast-to-myofibroblast transition through modulating KCNH6 mRNA translation. Mol Ther 2021;29:3436–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Zhang C, Chen L, Xie C. et al. YTHDC1 delays cellular senescence and pulmonary fibrosis by activating ATR in an m6A-independent manner. EMBO J 2024;43:61–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Sun W, Li Y, Ma D. et al. ALKBH5 promotes lung fibroblast activation and silica-induced pulmonary fibrosis through miR-320a-3p and FOXM1. Cell Mol Biol Lett 2022;27:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Gao H, Wang X, Ma H. et al. METTL16 regulates m(6)a methylation on chronic hepatitis B associated gene HLA-DPB1 involved in liver fibrosis. Front Genet 2022;13:996245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Feng Y, Dong H, Sun B. et al. METTL3/METTL14 transactivation and m(6)A-dependent TGF-β1 translation in activated Kupffer cells. Cell Mol Gastroenterol Hepatol 2021;12:839–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Fan C, Ma Y, Chen S. et al. Comprehensive analysis of the transcriptome-wide m6A methylation modification difference in liver fibrosis mice by high-throughput m6A sequencing. Front Cell Dev Biol 2021;9:767051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Li Y, Kang X, Zhou Z. et al. The m(6)a methyltransferase Mettl3 deficiency attenuates hepatic stellate cell activation and liver fibrosis. Mol Ther 2022;30:3714–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Qu T, Mou Y, Dai J. et al. Changes and relationship of N6-methyladenosine modification and long non-coding RNAs in oxidative damage induced by cadmium in pancreatic β-cells. Toxicol Lett 2021;343:56–66. [DOI] [PubMed] [Google Scholar]
- 40. Shu B, Zhang R-Z, Zhou Y-X. et al. METTL3-mediated macrophage exosomal NEAT1 contributes to hepatic fibrosis progression through Sp1/TGF-β1/Smad signaling pathway. Cell Death Discovery 2022;8:266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Chen L, Xia S, Wang F. et al. M(6)a methylation-induced NR1D1 ablation disrupts the HSC circadian clock and promotes hepatic fibrosis. Pharmacol Res 2023;189:106704. [DOI] [PubMed] [Google Scholar]
- 42. Shu B, Zhou YX, Li H. et al. The METTL3/MALAT1/PTBP1/USP8/TAK1 axis promotes pyroptosis and M1 polarization of macrophages and contributes to liver fibrosis. Cell Death Discov 2021;7:368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Feng Y, Hu Y, Hou Z. et al. Chronic corticosterone exposure induces liver inflammation and fibrosis in association with m(6)A-linked post-transcriptional suppression of heat shock proteins in chicken. Cell Stress Chaperones 2020;25:47–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Zhu Y, Pan X, Du N. et al. ASIC1a regulates miR-350/SPRY2 by N(6) -methyladenosine to promote liver fibrosis. FASEB J 2020;34:14371–88. [DOI] [PubMed] [Google Scholar]
- 45. Feng Y, Guo S, Zhao Y. et al. DNA 5mC and RNA m(6)a modification successively facilitates the initiation and perpetuation stages of HSC activation in liver fibrosis progression. Cell Death Differ 2023;30:1211–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Sun R, Tian X, Li Y. et al. The m6A reader YTHDF3-mediated PRDX3 translation alleviates liver fibrosis. Redox Biol 2022;54:102378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Shen M, Guo M, Li Y. et al. M(6)a methylation is required for dihydroartemisinin to alleviate liver fibrosis by inducing ferroptosis in hepatic stellate cells. Free Radic Biol Med 2022;182:246–59. [DOI] [PubMed] [Google Scholar]
- 48. Yang JJ, Wang J, Yang Y. et al. ALKBH5 ameliorated liver fibrosis and suppressed HSCs activation via triggering PTCH1 activation in an m(6)a dependent manner. Eur J Pharmacol 2022;922:174900. [DOI] [PubMed] [Google Scholar]
- 49. Chen Y, Zhou P, Deng Y. et al. ALKBH5-mediated m(6) a demethylation of TIRAP mRNA promotes radiation-induced liver fibrosis and decreases radiosensitivity of hepatocellular carcinoma. Clin Transl Med 2023;13:e1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Wang J, Yang Y, Sun F. et al. ALKBH5 attenuates mitochondrial fission and ameliorates liver fibrosis by reducing Drp1 methylation. Pharmacol Res 2023;187:106608. [DOI] [PubMed] [Google Scholar]
- 51. Jung HR, Lee J, Hong S-P. et al. Targeting the m6A RNA methyltransferase METTL3 attenuates the development of kidney fibrosis. Exp Mol Med 2024;56:355–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Feng C, Wang Z, Liu C. et al. Integrated bioinformatical analysis, machine learning and in vitro experiment-identified m6A subtype, and predictive drug target signatures for diagnosing renal fibrosis. Front Pharmacol 2022;13:909784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ning Y, Chen J, Shi Y. et al. Genistein ameliorates renal fibrosis through regulation Snail via m6A RNA demethylase ALKBH5. Front Pharmacol 2020;11:579265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Liu E, Lv L, Zhan Y. et al. METTL3/N6-methyladenosine/ miR-21-5p promotes obstructive renal fibrosis by regulating inflammation through SPRY1/ERK/NF-κB pathway activation. J Cell Mol Med 2021;25:7660–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Ding JF, Sun H, Song K. et al. IGFBP3 epigenetic promotion induced by METTL3 boosts cardiac fibroblast activation and fibrosis. Eur J Pharmacol 2023;942:175494. [DOI] [PubMed] [Google Scholar]
- 56. Tang W, Zhao Y, Zhang H. et al. METTL3 enhances NSD2 mRNA stability to reduce renal impairment and interstitial fibrosis in mice with diabetic nephropathy. BMC Nephrol 2022;23:124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Zhang J, Chen J, Lu Y. et al. TWIK-related acid-sensitive K(+) channel 2 promotes renal fibrosis by inducing cell-cycle arrest. iScience 2022;25:105620. 10.1016/j.isci.2022.105620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Chen J, Xu C, Yang K. et al. Inhibition of ALKBH5 attenuates I/R-induced renal injury in male mice by promoting Ccl28 m6A modification and increasing Treg recruitment. Nat Commun 2023;14:1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Yang Y, Li Q, Ling Y. et al. m6A eraser FTO modulates autophagy by targeting SQSTM1/P62 in the prevention of canagliflozin against renal fibrosis. Front Immunol 2022;13:1094556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Li X, Li Y, Wang Y. et al. The m(6)a demethylase FTO promotes renal epithelial-mesenchymal transition by reducing the m(6)a modification of lncRNA GAS5. Cytokine 2022;159:156000. [DOI] [PubMed] [Google Scholar]
- 61. Liu P, Zhang B, Chen Z. et al. M(6)A-induced lncRNA MALAT1 aggravates renal fibrogenesis in obstructive nephropathy through the miR-145/FAK pathway. Aging (Albany NY) 2020;12:5280–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Zhou Y, Song K, Tu B. et al. METTL3 boosts glycolysis and cardiac fibroblast proliferation by increasing AR methylation. Int J Biol Macromol 2022;223:899–915. [DOI] [PubMed] [Google Scholar]
- 63. Li T, Zhuang Y, Yang W. et al. Silencing of METTL3 attenuates cardiac fibrosis induced by myocardial infarction via inhibiting the activation of cardiac fibroblasts. FASEB J 2021;35:e21162. [DOI] [PubMed] [Google Scholar]
- 64. Cheng H, Li L, Xue J. et al. TNC accelerates hypoxia-induced cardiac injury in a METTL3-dependent manner. Genes (Basel) 2023;14:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Zhuang Y, Li T, Hu X. et al. MetBil as a novel molecular regulator in ischemia-induced cardiac fibrosis via METTL3-mediated m6A modification. FASEB J 2023;37:e22797. [DOI] [PubMed] [Google Scholar]
- 66. Li XX, Mu B, Li X. et al. circCELF1 inhibits myocardial fibrosis by regulating the expression of DKK2 through FTO/m(6)a and miR-636. J Cardiovasc Transl Res 2022;15:998–1009. [DOI] [PubMed] [Google Scholar]
- 67. Liu SY, Wu JJ, Chen ZH. et al. The m(6)a RNA modification modulates gene expression and fibrosis-related pathways in hypertrophic scar. Front Cell Dev Biol 2021;9:748703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Sun LY, Bie ZD, Zhang CH. et al. MiR-154 directly suppresses DKK2 to activate Wnt signaling pathway and enhance activation of cardiac fibroblasts. Cell Biol Int 2016;40:1271–9. [DOI] [PubMed] [Google Scholar]
- 69. Liu K, Ju W, Ouyang S. et al. Exercise training ameliorates myocardial phenotypes in heart failure with preserved ejection fraction by changing N6-methyladenosine modification in mice model. Front Cell Dev Biol 2022;10:954769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Zhuang T, Chen M-H, Wu R-X. et al. ALKBH5-mediated m6A modification of IL-11 drives macrophage-to-myofibroblast transition and pathological cardiac fibrosis in mice. Nat Commun 2024;15:1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Peng T, Liu M, Hu L. et al. LncRNA Airn alleviates diabetic cardiac fibrosis by inhibiting activation of cardiac fibroblasts via a m6A-IMP2-p53 axis. Biol Direct 2022;17:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci U S A 1974;71:3971–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Shi H, Wei J, He C. Where, when, and how: context-dependent functions of RNA methylation writers, readers, and erasers. Mol Cell 2019;74:640–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Liu J, Yue Y, Han D. et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 2014;10:93–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Wang X, Feng J, Xue Y. et al. Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature 2016;534:575–8. [DOI] [PubMed] [Google Scholar]
- 76. Bawankar P, Lence T, Paolantoni C. et al. Hakai is required for stabilization of core components of the m(6)a mRNA methylation machinery. Nat Commun 2021;12:3778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Ping XL, Sun BF, Wang L. et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res 2014;24:177–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Yue Y, Liu J, Cui X. et al. VIRMA mediates preferential m(6)a mRNA methylation in 3'UTR and near stop codon and associates with alternative polyadenylation. Cell Discov 2018;4:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Knuckles P, Lence T, Haussmann IU. et al. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m(6)a machinery component Wtap/Fl(2)d. Genes Dev 2018;32:415–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Patil DP, Chen CK, Pickering BF. et al. M(6)a RNA methylation promotes XIST-mediated transcriptional repression. Nature 2016;537:369–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Su R, Dong L, Li Y. et al. METTL16 exerts an m(6)A-independent function to facilitate translation and tumorigenesis. Nat Cell Biol 2022;24:205–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Xu C, Wang X, Liu K. et al. Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol 2014;10:927–9. [DOI] [PubMed] [Google Scholar]
- 83. Alarcón CR, Goodarzi H, Lee H. et al. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell 2015;162:1299–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Li A, Chen YS, Ping XL. et al. Cytoplasmic m(6)a reader YTHDF3 promotes mRNA translation. Cell Res 2017;27:444–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Huang H, Weng H, Sun W. et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol 2018;20:285–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Wu R, Li A, Sun B. et al. A novel m(6)a reader Prrc2a controls oligodendroglial specification and myelination. Cell Res 2019;29:23–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Jia G, Fu Y, Zhao X. et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 2011;7:885–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Zheng G, Dahl JA, Niu Y. et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 2013;49:18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Wang Z, Shen L, Wang J. et al. Prognostic analysis of m6A-related genes as potential biomarkers in idiopathic pulmonary fibrosis. Front Genet 2022;13:1059325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003;112:1776–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Hu X, Xu Q, Wan H. et al. PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis. Lab Invest 2020;100:801–11. [DOI] [PubMed] [Google Scholar]
- 92. Han J, Lee Y, Yeom KH. et al. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev 2004;18:3016–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Petri BJ, Piell KM, Wahlang B. et al. Polychlorinated biphenyls alter hepatic m6A mRNA methylation in a mouse model of environmental liver disease. Environ Res 2023;216:114686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Zhao T, Qi J, Liu T. et al. N6-Methyladenosine modification participates in the progression of hepatitis B virus-related liver fibrosis by regulating immune cell infiltration. Front Med (Lausanne) 2022;9:821710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Cui Z, Huang N, Liu L. et al. Dynamic analysis of m6A methylation spectroscopy during progression and reversal of hepatic fibrosis. Epigenomics 2020;12:1707–23. [DOI] [PubMed] [Google Scholar]
- 96. Chen L, Yao X, Yao H. et al. Exosomal miR-103-3p from LPS-activated THP-1 macrophage contributes to the activation of hepatic stellate cells. FASEB J 2020;34:5178–92. [DOI] [PubMed] [Google Scholar]
- 97. Liu Y, Liu Z, Tang H. et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 facilitates M1 macrophage polarization through the methylation of STAT1 mRNA. Am J Physiol Cell Physiol 2019;317:C762–75. [DOI] [PubMed] [Google Scholar]
- 98. Yu F, Jiang Z, Chen B. et al. NEAT1 accelerates the progression of liver fibrosis via regulation of microRNA-122 and Kruppel-like factor 6. J Mol Med (Berl) 2017;95:1191–202. [DOI] [PubMed] [Google Scholar]
- 99. Mansouri A, Gattolliat CH, Asselah T. Mitochondrial dysfunction and Signaling in chronic liver diseases. Gastroenterology 2018;155:629–47. [DOI] [PubMed] [Google Scholar]
- 100. Wonsey DR, Zeller KI, Dang CV. The c-Myc target gene PRDX3 is required for mitochondrial homeostasis and neoplastic transformation. Proc Natl Acad Sci U S A 2002;99:6649–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Xing J, He YC, Wang KY. et al. Involvement of YTHDF1 in renal fibrosis progression via up-regulating YAP. FASEB J 2022;36:e22144. [DOI] [PubMed] [Google Scholar]
- 102. Mathiyalagan P, Adamiak M, Mayourian J. et al. FTO-dependent N(6)-Methyladenosine regulates cardiac function during Remodeling and repair. Circulation 2019;139:518–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Ma X, Long C, Wang F. et al. METTL3 attenuates proliferative vitreoretinopathy and epithelial-mesenchymal transition of retinal pigment epithelial cells via wnt/β-catenin pathway. J Cell Mol Med 2021;25:4220–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Wang Y, Chen Y, Liang J. et al. METTL3-mediated m6A modification of HMGA2 mRNA promotes subretinal fibrosis and epithelial-mesenchymal transition. J Mol Cell Biol 2023;15:mjad005. 10.1093/jmcb/mjad005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Li X, Gao Y, Chen W. et al. N6-methyladenosine modification contributes to arecoline-mediated oral submucosal fibrosis. J Oral Pathol Med 2022;51:474–82. [DOI] [PubMed] [Google Scholar]
- 106. Knuckles P, Carl SH, Musheev M. et al. RNA fate determination through cotranscriptional adenosine methylation and microprocessor binding. Nat Struct Mol Biol 2017;24:561–9. [DOI] [PubMed] [Google Scholar]
- 107. Schöller E, Weichmann F, Treiber T. et al. Interactions, localization, and phosphorylation of the m(6)a generating METTL3-METTL14-WTAP complex. RNA 2018;24:499–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Choe J, Lin S, Zhang W. et al. mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature 2018;561:556–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Lin S, Choe J, Du P. et al. The m(6)a methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell 2016;62:335–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Hao H, Hao S, Chen H. et al. N6-methyladenosine modification and METTL3 modulate enterovirus 71 replication. Nucleic Acids Res 2019;47:362–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Zhou J, Wan J, Gao X. et al. Dynamic m(6)a mRNA methylation directs translational control of heat shock response. Nature 2015;526:591–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Liu X-M, Zhou J, Mao Y. et al. Programmable RNA N6-methyladenosine editing by CRISPR-Cas9 conjugates. Nat Chem Biol 2019;15:865–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Li J, Chen Z, Chen F. et al. Targeted mRNA demethylation using an engineered dCas13b-ALKBH5 fusion protein. Nucleic Acids Res 2020;48:5684–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Chen S, He Y, Huang X. et al. Photosensitive and dual-targeted chromium nanoparticle delivering small interfering RNA YTHDF1 for molecular-targeted immunotherapy in liver cancer. J Nanobiotechnol 2024;22:348. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data supporting this review are from previously reported studies and datasets, which have been cited. The processed data are available from the corresponding author upon request.