Next Article in Journal
Genome-Wide Characterization of the Sulfate Transporter Gene Family in Oilseed Crops: Camelina sativa and Brassica napus
Next Article in Special Issue
ASR1 and ASR2, Two Closely Related ABA-Induced Serine-Rich Transcription Repressors, Function Redundantly to Regulate ABA Responses in Arabidopsis
Previous Article in Journal
Identification and Characterization of Resistance to Rust in Lentil and Its Wild Relatives
Previous Article in Special Issue
Small Heat Shock Protein (sHSP) Gene Family from Sweet Pepper (Capsicum annuum L.) Fruits: Involvement in Ripening and Modulation by Nitric Oxide (NO)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Insights into m6A RNA Methylation and Its Emerging Role in Plant Circadian Clock

by
Nguyen Nguyen Chuong
1,†,
Phan Phuong Thao Doan
1,†,
Lanshuo Wang
1,
Jin Hee Kim
2,* and
Jeongsik Kim
1,2,3,*
1
Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 690756, Republic of Korea
2
Subtropical Horticulture Research Institute, Jeju National University, Jeju 690756, Republic of Korea
3
Faculty of Science Education, Jeju National University, Jeju 690756, Republic of Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Plants 2023, 12(3), 624; https://doi.org/10.3390/plants12030624
Submission received: 4 January 2023 / Revised: 24 January 2023 / Accepted: 27 January 2023 / Published: 31 January 2023
(This article belongs to the Special Issue Trends and Prospects of Genetic and Molecular Research in Plant)

Abstract

:
N6-adenosine methylation (m6A) is a prevalent form of RNA modification found in the expressed transcripts of many eukaryotic organisms. Moreover, m6A methylation is a dynamic and reversible process that requires the functioning of various proteins and their complexes that are evolutionarily conserved between species and include methylases, demethylases, and m6A-binding proteins. Over the past decade, the m6A methylation process in plants has been extensively studied and the understanding thereof has drastically increased, although the regulatory function of some components relies on information derived from animal systems. Notably, m6A has been found to be involved in a variety of factors in RNA processing, such as RNA stability, alternative polyadenylation, and miRNA regulation. The circadian clock in plants is a molecular timekeeping system that regulates the daily and rhythmic activity of many cellular and physiological processes in response to environmental changes such as the day-night cycle. The circadian clock regulates the rhythmic expression of genes through post-transcriptional regulation of mRNA. Recently, m6A methylation has emerged as an additional layer of post-transcriptional regulation that is necessary for the proper functioning of the plant circadian clock. In this review, we have compiled and summarized recent insights into the molecular mechanisms behind m6A modification and its various roles in the regulation of RNA. We discuss the potential role of m6A modification in regulating the plant circadian clock and outline potential future directions for the study of mRNA methylation in plants. A deeper understanding of the mechanism of m6A RNA regulation and its role in plant circadian clocks will contribute to a greater understanding of the plant circadian clock.

1. Introduction

Plants, as multicellular organisms, rely on a range of complex molecular mechanisms to control gene expression for normal development and stimuli response. Gene expression is controlled on two levels, which involve the amount of mRNA that is produced and the regulation of mRNA translation into proteins [1]. Recent studies have highlighted the importance of RNA modification as a crucial mechanism that dynamically modulates the cellular transcriptomic profile [2]. Currently, more than 200 types of RNA modifications have been identified [3], which are involved in various plant cellular and biological processes, including embryo development, shoot stem cell fate, floral transition, trichome morphogenesis, leaf initiation, and root development [4,5,6,7,8,9,10,11,12].
In eukaryotic mRNA, m6A is the most prevalent type of modification [13,14], which occurs through the addition of a methyl group (CH3) to the N6 position of adenosines of the mRNA [15]. The presence of m6A in plants was first identified in maize and later in many other plant species such as Arabidopsis, wheat, and oats [16]. The occurrence of m6A in Arabidopsis mRNA was determined to vary between 0.45–0.65% of the total adenosine bases, with an estimated 0.5–0.7 m6A peaks per 1000 nucleotides [17]. A recent study reported that m6A sites can be detected in 32–35% of all expressed transcripts [13]. The frequency of m6A is unevenly distributed within the mRNA and is predominantly clustered near the stop codon and 3′ untranslated regions (UTRs) [18,19,20]. The m6A of plants is also found to be enriched near the start codon [17,21]. Many chloroplast-associated and photosynthesis-related genes showed an abundance of m6A sites located around the start codon, which suggests a unique function of m6A in plant photosynthesis [17,22].
Though these modifications have been detected since the 1970s, studies on m6A were initially limited due to the lack of methods for identifying m6A sites. However, with the discovery of the first m6A demethylase (fat mass and obesity-associated protein [FTO]) in 2011 [23], m6A has been determined to be a dynamic and reversible process that may have regulatory functions. Since then, several approaches have been developed to facilitate the functional study of m6A methylation. Based on the strategies to identify or recognize m6A sites, these approaches can be broadly divided into two categories: antibody-dependent methods such as methylated RNA immunoprecipitation (MeRIP)-seq [18], UV cross-linking and immunoprecipitation (miCLIP)-seq [24], or super-low-input m6A (SLIM)-seq [25] and antibody-independent methods such as deamination adjacent to RNA modification targets (DART)-seq [26], m6A-selective chemical labeling (m6A-SEAL)-seq [27], m6A-selective allyl chemical labeling (m6A-SAC)-seq [28], or nanopore-based direct RNA (DR)-seq [29]. Each of these methods has its own advantages and limitations, which have been discussed in detail in previous reviews [30,31].
As more efficient mapping methods are developed and more m6A-related genes are identified, the important roles that m6A plays in various biological processes are being gradually discovered. Although most m6A functional studies were conducted in an animal system, several reports have shown that m6A modification plays a crucial role in the regulation of plant development [4,32,33,34,35] and stress resistance [7,32,36,37]. In this review, we provide the current progress on the understanding of m6A biogenesis and function and its involvement in the regulation of plant circadian clock.

2. m6A in Plants

2.1. General Mechanisms

Generally, the m6A modification process involves numerous components, which include methylases, demethylases, and m6A-recognizing proteins (Figure 1). These are commonly termed m6A writers, erasers, and readers, respectively [15,38]. In plants and other eukaryotes, N6-adenosine methylation occurs through the binding of m6A writers to a highly conserved consensus sequence, RRACH (R = G/A; H: U/A/C) [10], which leads to the transfer of a methyl group from an S-Adenosyl Methionine (SAM) molecule onto the sixth nitrogen of the adenosine base [39]. However, the m6A abundance is significantly lower than that of the RRACH motifs, indicating that not all of these consensus motifs are associated with m6A modification and that the mechanism by which these motifs are selected for m6A modification remains elusive [40]. The reverse process of oxidatively removing the methyl group from the adenosine base is modulated by m6A erasers [39]. Finally, m6A-modified RNA is recognized by the m6A readers which subsequently utilize different complexes to regulate the RNA fate [41]. The Arabidopsis genome contains 29 m6A regulatory genes (6 writers, 6 erasers, and 17 readers) which encode for 55 proteins (8 writers, 14 erasers, and 33 readers) [42]. Next, we discuss in detail the structural features and the mechanistic insight of each m6A component.

2.2. m6A Writers

Generally, the methylase-containing protein complexes that are responsible for the N6-adenosine methylation of mRNA are commonly called m6A writer complexes [43]. Components of these complexes are evolutionarily conserved between the main eukaryotic lineages [4]. Studies on mammalian and Drosophila systems indicate that the m6A methylase complex consists of two sub-complexes, which are the m6A-METTL complex (MAC) and the m6A-METTL associated complex (MACOM) [44]. METTL3 and METTL14 together form the stable heterodimer that is MAC, while the MACOM consists of WTAP, RBM15, VIRMA, ZC3H13, and HAKAI [45]. METTL3, the first discovered methylase, harbors a catalytic subunit that serves as the main catalytic core for the methylation reaction [46,47]. Though METTL14 shares a high homology with METTL3 it is unable to bind with SAM but rather functions as a facilitator for the interaction between METTL3 and the RNA target [48,49]. MACOM interacts weakly with MAC to support the MAC functions, with each of its components performing a distinct function. WTAP binds directly to METTL3 to improve the activity of methyl transferase by ensuring the proper localization of the METTL3-METTL14 heterodimer [50]. RBM15 functions by recruiting the complex to the specific sites on RNA to promote methylation [51]. VIRMA mediates the preferential m6A mRNA methylation in the 3′ UTR and near-stop codons [52]. ZC3H13 is required for the nuclear localization of the complex [53], while HAKAI appeared to stabilize the WTAP and VIRMA protein [54].
In Arabidopsis, several orthologues of animal m6A writer components have been identified with similar functions and shown to interact with each other [8,38]. These orthologues include the mRNA adenosine methylase A (MTA; an orthologue of the methyl transferase 3 [METTL3]) [55] and B (MTB; an orthologue of METTL14) [8], the splicing factor FKBP12 interacting protein 37 kDa (FIP37; an orthologue of the Wilms tumor-1 associated protein [WTAP]) [8], the protein virilizer (VIR; an orthologue of the m6A methyltransferase-associated virus [VIRMA]/KIAA1429) [8], the putative ubiquitin E3 ligase HAKAI [8], the RNA-binding protein FPA (an orthologue of the RNA-binding protein 15 [RBM15]) [56], and the HAKAI-interacting zinc finger protein HIZ2 (a possible orthologue of the CCCH type 13 zinc finger protein [ZC3H13]) [57] (Figure 1). Protein sequence analysis has identified the specific conserved domains that are present in each m6A writer, which are MT-A70, WTAP, VIR_N, HAKAI, and CCCH domains in MTA/MTB, FIP37, VIR, HAKAI, and HIZ2, respectively [42,57]. The MT-A70 domain is required for the binding of substrate SAM, which is essential for the formation of m6A in mRNA [46]. Similar to their animal orthologues, plant m6A writers are mainly localized in the nucleoplasm [8]. Interestingly, though the knockdown of RBM15 reduced global levels of m6A [51], the knockdown of FPA, the RBM15 orthologue in Arabidopsis did not [56]. This may indicate the distinct roles of RBM15/FPA in m6A methylation in animal and plant systems.
Apart from m6A methyl transferases in the MTA-MTB complex which are responsible for the majority of m6A sites in Arabidopsis [8,10,32,55], several methyl transferases have also been identified, including METTL16 [58], METTL5 [59], or the zinc finger CCHC type containing 4 (ZCCHC4) [60]. While there is currently no information regarding the orthologues of METTL5 and ZCCHC4 in plants, FIO1 has been identified as the orthologue of METTL16 in Arabidopsis [58,61]. However, FIO1 is distinct from METTL16 since FIO1 does not methylate Arabidopsis SAM synthetases or affect their transcript expression levels. Instead, FIO1 installs m6A into U6 small nuclear (sn)RNA, a small subset of poly(A)+ RNA, and several phenotype-related mRNAs, thereby regulating the mRNA stability and many developmental processes [62]. Another example of independent methyl transferase that is found in Arabidopsis is MTC (an orthologue of METTL4) [63]. The structural comparison revealed that Arabidopsis MTC homodimer shared similar features with the METTL3-METTL14 heterodimer, suggesting that MTC functions as the m6A writer [63]. MTC displayed N6−2′-O-dimethyladenosine (m6Am) in vivo and N6-methylation of 2′-O-methyladenosine (Am) within single-stranded RNA in vitro [63]. However, whether these methyl transferases act independently or cooperatively with other proteins for m6A methylation requires further investigation.
Though the m6A consensus sequences (RRACH) frequently appear in mRNA (once every ~57 nucleotides), only a few are methylated. Additionally, despite the abundance of RRACH sequences, m6A only occurs in specific transcripts. As previously mentioned, the regulatory mechanisms of this transcript- and site-specific selectivity remain unclear. Two models have been proposed for the recruitment of m6A writer complex to the specific transcript, which is mediated by (i) transcription factors or (ii) histone modifications. Similarly, there are also two proposed models for site-specific targeting of m6A, which are (i) the RNA-binding protein (RBP)-mediated and (ii) RNA polymerase II (Pol II)-mediated recruitment models. These models have been extensively discussed in a previous review [64]. However, it is noteworthy that these models only account for a small fraction of m6A-containing transcripts in the cell, suggesting that there are unknown mechanisms responsible for most of the mRNA N6-adenosine methylation.
The m6A modifications occur in response to various internal and external stimuli such as heat shock, DNA damage, or stress [65,66,67,68,69]. The m6A writer complex is suitable for regulation at different levels, such as changes in the abundance of individual components or post-translational modifications (PTMs). The catalytically active subunit METTL3, which is targeted by miRNAs [70] and SUMOylation [71], is an obvious candidate for this regulation. Expression and stability of other m6A writer complex components appear to be METTL3-dependent [72] and can be affected by PTMs [73]. Overall, much knowledge regarding the regulatory role of this methyl transferase activity and m6A landscape remains to be elucidated.

2.3. m6A Erasers

m6A modification is a reversible process, by which the methyl group is removed catalytically by the m6A erasers in a dynamic, rapid, signal-dependent manner [74]. The m6A demethylases belong to the ALBHK protein family (a homolog of the α-ketoglutarate dependent dioxygenase AlkB family) [23,75]. To date, FTO and ALBHK5 are the only two identified m6A erasers in animals, although several members of the AlkB family have been documented to function by reversing m6A methylation [76]. Unlike FTO, which has no identified homolog in the plant system, the Arabidopsis genome contains 13 ALBHK family members with different subcellular localizations [77]. Five of these proteins are identified to be homologs of ALBHK5 including ALKBH9A, ALKBH9B, ALKBH9C, ALKBH10A, and ALKBH10B [6]. Among these, ALKBH9B is suggested to have demethylase activities in vitro and in vivo [7]. However, alkbh9b and alkbh9c mutants did not display any changes in the m6A/A ratio [6]. Another homologous protein, ALKBH10B, was shown to demethylate m6A in vivo and in vitro and the alkbh10b mutant consistently has increased m6A level [6]. Although m6A erasers have previously been suggested to play a crucial role in m6A functioning, they now appear to be limited to specific tissues and conditions [64].

2.4. m6A Readers

Eukaryotes can perceive m6A marks via the m6A readers. These m6A-binding proteins play a major role in the mechanism by which m6A regulates RNA metabolism. The characterization of m6A readers revealed that these proteins can be categorized into three classes based on their mechanism of recognizing m6A-containing RNA, which are (i) direct binding to the m6A base using a YT521-B homology (YTH) domain [78,79]; (ii) binding of the exposed single-stranded RNA motifs generated by m6A-induced structural changes [80,81]; or (iii) utilizing a common RNA-binding domain (RBD) and its flanking regions to recognize the m6A-modified transcripts [82]. Class I m6A readers are proteins that contain the YTH domain, which recognizes and binds to the N6-methyl group of adenosine via a hydrophobic binding pocket that contains conserved aromatic side chains [83]. The binding of class II readers to m6A-containing transcripts occurs via an m6A switch mechanism, in which the methylation destabilizes the Watson-Crick base-pairing and increases the accessibility of a single-stranded RNA-binding motif that is recognized by this class [80,81]. Class III readers lack of YTH domain and bind to m6A-containing transcripts in an RNA-structure-independent manner [82]. Though these are shown to utilize a common RBD and its flanking region for the recognition of m6A modifications, the exact mechanism behind these selective binding remains unclear. The different m6A-binding mechanisms indicate a broad and complex network of m6A-dependent pathways, and each class of m6A reader can be employed for specific targets or conditions.
Currently, there is no information regarding the homologs of class II and III readers in the plant system. In contrast, 13 proteins containing the YTH RNA-binding domain have been identified in the Arabidopsis genome [84]. These proteins can be categorized into two subgroups: (i) the evolutionarily conserved C-terminal region 1–11 proteins (ECT1–11) that belong to the YTHDF family [4] and (ii) the other two members that belong to the YTHDC family (ECT12 and the cleavage and polyadenylation specificity factor 30 [CPSF30]) [9,85]. Structural analysis revealed that the YTH domain of the first group is located near the C-terminus, in contrast to the other group whose YTH domain is located in the internal regions [86]. The conserved consensus sequence RRm6ACH is recognized by the YTH domain of ECTs. Concurrently, the intrinsic disorder region within the ECTs forms a stable interaction with the U-rich sequence in adjacent regions, thereby controlling the occupancy of the binding adjacent to m6A with different RNA-binding factors. These ECT-binding targets are mainly involved in the translation and metabolic processes [4]. Arabidopsis CPSF30 forms two splice isoforms via alternative polyadenylation: (i) a ~28 kDa isoform (CPSF30-S) that harbors three zinc finger domains and is homologous to mammalian CPSF30 and (ii) a ~70 kDa isoform (CPSF30-L) that has an additional YTH domain that is unique to plants [87,88]. The CPSF30 binding site is predominantly located in the mRNA 3′ UTR region and its m6A binding ability enhances the formation of CPSF30 liquid-like nuclear bodies to regulate polyadenylation [85].

2.5. m6A Regulates RNA Activity

m6A employs diverse mechanisms to regulate RNA metabolism, which involves different aspects such as stability, structure, translatability, localization, or splicing (Figure 1). However, most m6A studies regarding its molecular functions were conducted in animals. The impact of m6A on plant RNA activity is limited. Current evidence suggests that m6A in plants, like in the animal model system, also mediates the stability of RNA under various conditions. For example, the knockdown of m6A components such as the writer MTA [6,32] or reader ECT2 [11] and CPSF30 [85] in Arabidopsis was shown to accelerate the degradation of target transcripts. Similarly, the writer FIP37 destabilized mRNA of the key shoot meristem genes [10] while demethylation by the ALKBH10B eraser promoted the stability of its targets [6].
Several underlying mechanisms have been proposed to explain the m6A roles in RNA stability. The m6A binding by the mammalian reader YTHDF2 may directly or indirectly regulate transcript stability by either recruiting the CCR4-NOT deadenylase complex to destabilize the m6A-containing mRNA [89] or promoting the translocation of mRNA from the translation machinery to processing bodies for degradation [90]. In another study, m6A was found to regulate the interaction between mRNA and the mRNA stabilizer human antigen R (HuR) in a distance-dependent manner [91]. In close proximity, m6A promotes HuR binding and conversely decreases HuR binding when far apart [91]. The regulatory roles of m6A on mRNA stability may also lead to structural changes in mRNA, under certain conditions. For example, m6A increases the stability of mRNA and subsequently leads to a decrease in structural complexity in response to a change in salinity [92]. m6A may impact RNA structure by promoting the transition from paired to unpaired RNA, as suggested by the less structured mRNAs with the m6A consensus motif GGACU in wild-type mouse embryonic stem cells (mESCs) compared with that of METTL3-knockout mESCs [93].
Another potential mechanism that m6A may utilize to regulate RNA metabolism is alternative polyadenylation (APA), which has been suggested in several studies. Reducing expression of VIR, an m6A writer in Arabidopsis negatively affected mRNA 3′s end formation and subsequently resulted in the preferential proximal poly(A) site selection [13]. The reader CPSF30 was also found to regulate APA and poly(A) site selection [85,94]. Additionally, m6A is necessary to maintain transcriptome integrity by guiding site-specific mRNA polyadenylation in target genes with intrinsic transcription termination and polyadenylation defects [95]. This APA pathway termed the m6A-assisted polyadenylation requires the writer FIP37 and reader CPSF30-L for proper functioning [95]. Studies on other plant species support the involvement of m6A in the APA pathway, as m6A modification can affect poly(A) site selection in maize [96].
There are lines of evidence that suggest the interaction between m6A and other regulatory mechanisms of mRNA. m6A modifications were found to affect the primary-microRNA (pri-miRNA) secondary structure and recruitment of microprocessor complexes to the pri-miRNA in Arabidopsis [97]. Furthermore, the m6A writer MTA also interacted with proteins involved in miRNA biogenesis such as RNA Polymerase II and TOUGH [97]. This observation is consistent with the animal system, as N6-adenosine methylation of pri-miRNA mediated by METTL3 facilitated the DGCR8 microprocessor complex pri-miRNA processing [98]. Another m6A component involved in this process was the nuclear m6A reader hnRNPA2B1 [99]. In the opposite direction, miRNA machinery plays a role in regulating m6A formation, possibly by modulating METTL3-mRNA binding [100]. Crosstalk between histone modification and m6A has also been proposed in a previous study. H3K36me2 and m6A were found to co-occur near the 3 UTR region and affected the global level of each other in Arabidopsis [101]. Furthermore, the direct interaction between an H3K36me2 writer SET DOMAIN GROUP 8 (SDG8), and an m6A writer protein FIP37 was observed. Still, further investigation is required to fully understand this crosstalk. In conclusion, though has been indicated in many previous studies, the underlying mechanisms for the regulatory roles of m6A in RNA metabolism require further investigation, especially in plant systems.

3. m6A in the Plant Circadian Rhythm

3.1. Plant Circadian Clock and m6A Methylation

Plants, as sessile organisms, evolve to sense and predict environmental changes. The most dramatic changes in the environment are day-night cycles due to the earth’s rotation and many processes in plants are synchronized with their circadian cycles. The circadian clock is the endogenous timekeeper that governs and coordinates these rhythmic processes to increase plant fitness. This is based on the observation that altered period length or out-of-phase conditions in plants interfere with their optimal growth or defense against abiotic and biotic stress [102,103]. The fundamental and molecular aspects of the circadian clock rely on the transcriptional feedback among the central oscillator components with morning components, CIRCADIAN CLOCK-ASSOCIATED1 (CCA1)/LATE ELONGATED HYPOCOTYL (LHY), and an evening component, TIMING OF CAB EXPRESSION1 (TOC1) (Figure 2) [104]. Additional transcriptional feedback loops involve morning and evening complexes that link the central clock components. The morning components, such as the PSEUDORESPONSE REGULATOR9 (PRR9) and PRR7, act to reset the clock in the morning by inhibiting the transcription of LHY and CCA1 [105]. The evening complex with the LUX ARRHYTHMO (LUX), EARLY FLOWERING3 (ELF3), and ELF4 act to maintain the clock in the evening by repressing the transcription of PRR9 and PRR7 [106]. Cycling activators such as NIGHT LIGHT-INDUCIBLE AND CLOCK-REGULATED GENE1 (LNK1)/LNK2 and REVEILLE8 (REV8) are involved in the activation of evening complex components including LUX, ELF3, and ELF4 [107,108]. Post-translational feedback regulation has a role in the maintenance of the circadian clock. For example, ZEITLUPE (ZTL) promotes the degradation of TOC1, while the GIGANTEA (GI) and PSEUDORESPONSE REGULATOR3 (PRR3) function to stabilize and protect TOC1 [109,110]. Overall, this complex network of transcriptional and post-translational feedback loops allows the clock to maintain a consistent and robust circadian rhythm in Arabidopsis.
In plants, the circadian clock controls the rhythmic expression of thousands of genes through transcriptional activation or suppression by multiple transcription factors [112]. Recent studies using RNA-seq analysis have highlighted the importance of post-transcriptional regulation of mRNA, including alternative splicing, polyadenylation, mRNA nuclear export, mRNA degradation, and mRNA methylation, in the rhythmic expression of these genes [113,114,115]. In particular, m6A methylation has been proposed as a new and emerging layer of dynamic mRNA regulation in the circadian clock. In mammalian cells, many well-known clock genes, including Clock, Periods (Pers), Albumin D-site-binding protein, Nuclear receptor subfamily 1 group D member 1 (Nr1d1), and Casein kinase 1 delta (CK1δ), contain multiple m6A methylation sites on their transcripts [116,117]. Silencing of the m6A methylase METTL3 expression lengthened the circadian period through a delay in the nuclear export of mRNAs of the clock genes Per2 and Aryl hydrocarbon receptor nuclear translocator-like (Arntl) [116]. Cryptochrome 1 (CRY1) and CRY2 are core components of the mammalian circadian clock and interact with an m6A eraser FTO, which is required to maintain global m6A methylation levels [118]. In addition, cry1cry2 knockdown mice with a complete loss of circadian rhythm have significantly lower levels of m6A and a loss of the circadian rhythm of m6A [118]. In plants, daily oscillation of mRNA transcripts of the writers and erasers and accumulation of global m6A levels at midnight have been observed in seagrasses Cymodocea nodosa and Zostera marina [119].
Recent advancements in molecular technologies for direct detection of RNA methylation, such as nanopore mRNA-seq, miCLIP-seq, and m6A-seq, have enabled high-throughput quantitative analysis of the methylation levels and accurate identification of deposition sites on mRNA in various developmental conditions [13,120]. m6A methylation is more frequently found in mRNAs of clock-controlled genes (CCGs) and many mRNAs of clock components are m6A methylated in Arabidopsis [17,120]. m6A is the most prominent and dynamic type of RNA methylation and determines the fate of CCG mRNA, including the mRNA stability, splice-site choice, and RNA 3′ end formation [13,120]. The importance of m6A in the regulation of the circadian clock in plants has been emphasized by several recent studies on the genome-wide analysis of the function of (i) CRY1/2-mediated MTA-MTB-FIP37 methylase complexes [112,120]; (ii) VIR, another conserved m6A writer complex component [13]; and (iii) FIO1, a homolog of the m6A writer METTL16, in the regulation of the circadian clock and related physiology (Figure 2) [62,121,122,123,124].

3.2. Circadian Clock Regulation through m6A Methylation by a General m6A Writer Complex

Deposition of m6A on the majority of mRNAs is catalyzed by a multiprotein RNA methylases complex, which consists of two methylases (MAC) including MTA and MTB, as well as the additional proteins (MACOM) including FIP37, VIR, and HAKAI [8,74]. MTA, MTB, and FIP37 are the core components of the m6A methylase complex as counterpart subunits of METTL3, METTL14, and WTAP in the mammalian METTL3/14-type general m6A writer complex [38].
In Arabidopsis, the m6A deposition is more commonly detected in the mRNAs of CCGs, particularly those involved in chloroplast-related or photosynthesis processes, with 57% of CCG mRNAs relative to the 41% of expressed mRNAs [120]. m6A methylation is also more frequently found in mRNAs of clock regulatory genes, with a 1.4-fold enrichment. These include the morning components, CCA1 and LHY, and the evening components, GI and ELF3 [120]. To understand the role of the general m6A writer complex in the circadian clock, the m6A profiles and circadian rhythms should be examined in these mutants. However, these loss-of-function mutations result in embryonic lethality, which is a major obstacle in exploring the role of m6A in the circadian clock. Instead, partially rescued plants expressing the embryo-specific ABSCISIC ACID INSENSITIVE3 (ABI3) promoter-driven MTA in the mta mutant (ABI3::MTA/mta) were used to reveal the role of the MTA methylase in the circadian clock [120,125]. These plants demonstrated a 90% reduction in m6A methylation in leaves and a long-period rhythm in white light with reduced m6A methylation in the 3′ UTR of CCA1 mRNA. Interestingly, MTA and FIP37 physically interact with CRY1 and CRY2, blue light receptors, and MTB also interacts with CRY2 [55,112,120].
CRY1 and CRY2 mediate blue light input in the circadian clock depending on the fluence rate, with CRY1 for strong light and CRY2 for weak light [126,127]. However, the detailed mechanisms of how CRY1/2 blue light inputs are transduced to the circadian oscillators are not clearly understood and may include GI and PIFs [128,129]. Recently, it has been reported that CRY2 interacts with MTA to elicit m6A methylation of the CCGs to mediate the blue light input to the oscillator [120]. CRY-mediated blue light induces m6A modification in 10% mRNAs within a few hours in a fluence rate-dependent manner [112,120]. In blue light, CRY1 interacts with FIP37 and accelerates the m6A-mediated mRNA degradation of PIF3, PIF4, and PIF5 which are negative regulators of light and circadian clock-mediated hypocotyl growth [112]. On the other hand, CRY2 forms acute and dynamic blue light-mediated photobodies that concentrate molecules in the complex through liquid-liquid phase separation [120]. CRY2 recruits MTA, MTB, and FIP37 in the photobodies within a few seconds, which induces the mRNA m6A modification of ten core clock component genes including CCA1 and enhances their mRNA stability. The CRY2-mediated photobodies promote the enrichment of m6A writers and are regarded as another photo-regulatory layer in the regulation of the circadian clock [120,130].
Although CRY2 and the MTA-MTB-FIP37 methylase complex form photobodies and transfer the blue light input to the circadian clock, detailed analysis of their circadian clock phenotypes indicates that an additional and separate role of MTA in regulating the circadian clock exists [120]. The cry1cry2 double mutant showed reduced responses to period shortening effects by increased intensity of blue light, indicating the involvement of CRY1/2 in parametric entrainment of blue light input to the circadian clock through continuous modulation of the circadian clock by recognizing the intensity of blue light. However, mta showed a similar period shortening responsiveness as the wild type in blue light, and a longer period in white light than in blue light, indicating that this additional role may be mediating other light input or the modulation of the core clock oscillator. However, the effect of blue light-induced photobodies on the dynamic mRNA m6A modification of circadian regulatory genes remains unknown.
VIR, another conserved component of the general m6A writer complex, is also linked to circadian clock regulation [13]. The vir-1 mutant, a weak mutant allele of the VIR gene, exhibited a remarkable reduction in the m6A ratio to between 5–15 % in 3′ UTR and further exhibited multiple developmental defects as well as a lengthened circadian period. In vir-1, the mRNA abundance in the circadian core clock components including PRR7, LNK1/2, and GI, which had m6A modification, was increased. Unlike with the mta mutant, the CCA1 level was increased in vir-1, which is associated with a long-period rhythm in vir-1. VIR was also found in splicing speckles, where it was co-localized with the splicing factor SR34, but the alteration in splicing patterns of mRNA in the vir-1 mutant was minor [8,13]. In contrast, vir-1 mutation led to a global shift of the poly (A) tail length distribution of CCGs [13]. Additionally, vir-1 mutation abolished the 3′ end formation in mRNAs, which is similar to the function of human VIRMA, an orthologue of VIR, in the polyadenylation selection [52]. Human VIRMA associates with the polyadenylation cleavage factors, CPSF5 and CPSF6, which facilitates the selection of proximal polyadenylation sites within the 3′ UTR of mRNAs. Plant VIR might regulate the stability of mRNA encoding circadian clock genes through nonsense-mediated decay of mRNA with lengthened 3′ UTRs or aberrant poly(A) tail lengths.
Although MTA and VIR function in the same methylase complexes and mta and vir-1 mutations have similar lengthening effects on the circadian period, their functional role in regulating the circadian clock appears to be different [13,120]. MTA and VIR mediate the stabilization and 3′ end formation of their target mRNAs, respectively, despite their similar preference for m6A modification at the 3′ UTR. Interestingly, the levels of CCA1 mRNA, which are the result of these mutations, are opposite in their mutants relative to the wild type. This may be due to the cumulative effect of the altered activity of circadian clock genes, which are selectively guided by MTA and VIR. A more detailed investigation into the molecular effects of these mutations on clock-regulatory genes may reveal how they cooperate and act independently in regulating the circadian clock. It has been observed that m6A methylation of mRNAs can be regulated by the circadian clock, as global methylation profiles peak at midnight in marine plants [119]. Interestingly, the expression of several components in the general m6A writer complex, including MTA, MTB, and HAKAI, is diurnally regulated with a peak time at night and MTB expression is regulated by the circadian clock (Figure 3a,d) [131]. Further investigation into whether m6A methylation in CCGs is regulated by the circadian clock can suggest the dynamic role of m6A methylation by general m6A writer complexes in regulating the circadian clock.
Overall, it is evident that m6A methylation induced by the MTA-MTB methyl writer complex is essential for the photo-input or maintenance of the plant circadian clock, but the dynamic regulation and the role of m6A readers and erasers in regulating m6A methylation need to be established.

3.3. FIO1, a Core Clock Component as m6A Methylase

FIO1 was first identified by the screening of photoperiodic flowering responses and was further characterized as a core clock component for the maintenance of the proper circadian period [61]. FIO1 is predicted to be a tentative methylase with a SAM binding domain and an orthologous protein to mammalian METTL16. Mammalian METTL16, another type of m6A methylase, is known to function as a specific m6A writer on the mRNA MAT2A and the U6 snRNA without uncertainty, although a hundred candidate mRNAs could be predicted as its targets through a genome-wide m6A IP-seq [133]. MAT2A, the first identified target of METTL16, is the key enzyme in synthesizing SAM, which is required as a methyl donor for most cellular methylation events [134]. METTL16 establishes homeostasis of SAM through the stabilization of MAT2A through direct deposition of m6A on its mRNA when the availability of the SAM is limited in the cell. Another target of METTL16 is U6 snRNA which is an RNA component of U6 ribonucleoprotein in the spliceosome [135]. METTL16 binds and deposits a single m6A at the UACAGAGAA site within the stem-loop structure in the U6 snRNA, which is required for its incorporation into U4/U6 snRNA. METTL16 mediates the stability and splicing sites of the mRNAs by influencing the U6 spliceosome [136,137]. Recent research on FIO1 suggests that it possesses bona fide methylase activity with the catalytic motif with a stretch of amino acids NPPF for the binding of adenine substate RNAs in vitro and in vivo [62,123,124].
Recently, several reports have highlighted how FIO1 mediates m6A methylation by regulating various circadian clock-related developmental processes including photomorphogenesis and floral transition (Figure 2) [62,121,122,123,124]. In fio1 mutants, about 10%–15% of m6A in the total mRNA and 10–60% of the U6 snRNAs were reduced in seedlings, rosette leaves, and floral buds, which were relatively minor effects compared to the 80%–90% m6A loss in fip37 and mta. Global profiling of m6A methylation in fio1 mutants revealed more than 2-fold hypomethylated m6A peaks in around 1000–2500 mRNAs, which are mainly located around the 3′ UTR and near the stop codon. In Arabidopsis, FIO1 installs an m6A at the consensus sequence of U6 m6A motif ACAGA and plant-specific mRNA m6A motifs GGACC and UGUAU, with higher activity in the consensus sequence and different preferential activity for single-stranded over-stem-loop structures in U6 snRNA [62,123]. It is noted that multiple motif variants have been found in FIO1-mediated methylation sites with YHAGA (Y = C/U; H = C/A/U) in the CDS region [121]. The GAACU and UGUAA consensus sequences in the 3′ UTR [123], and DRACH (D = A/G/U; H = A/C/U) in the 3′ UTR [124]. Interestingly, there were no global pattern changes in the remaining m6A between the wild type and fio1 mutant, indicating that FIO1 has no preferential choice in m6A position and motif. FIO1 has a lesser effect on global m6A levels, but a higher m6A reduction in specific groups of mRNAs, indicating that FIO1 likely mediates certain biological and physiological processes [62,121,122,123,124]. Gene ontology analysis of the differentially expressed genes and genes with hypomethylated mRNA in fio1 mutant revealed characteristics of circadian rhythm and flower development [62]. These results support that FIO1 regulates the m6A deposition in mRNAs of specific target genes involved in certain biological processes including the circadian rhythm.
The fio1 mutant exhibits several developmental phenotypes, including a long circadian period, early flowering, and long hypocotyls, which may be due to an aberrant circadian rhythm or an independent function of FIO1 in developmental processes [61]. Based on the differential expression and hypomethylated mRNAs in the fio1 mutant, many important regulatory genes involved in the circadian clock, floral transition, and photomorphogenesis are estimated to be the direct targets of FIO1 (Figure 2). For example, FIO1 affects the m6A modification of transcripts of CCA1 and LHY in the circadian clock pathway, CO and CRY2 in the photoperiodic flowering pathway, FLC in the autonomous and vernalization flowering pathway, SOC1, SVP, SPL3, and SEP3 in the floral integration pathway, and PIF4 in photomorphogenesis [62,121,123]. However, the effectiveness of m6A modification by FIO1 on the target mRNAs is different among them, with the degradation of LHY, CO, CRY2, SOC1, SPL3, SEP3, and PIF4 mRNAs, and stabilization and altered splicing of SVP and FLC mRNAs [123]. Additionally, RNA-IP with FIO1 contributes to revealing a direct target of FIO1 as evidenced by the association of FIO1 with SOC1, SVP, CCA1, and LHY mRNAs [121], PIF4, CRY2, CO, and FLC mRNAs [62], and SPL3 and SEP3 mRNAs [123]. The effect of FIO1 on a wide range of targets supports the concept that FIO1 might be independently involved in various physiological processes through its m6A methylation of various target transcripts. However, several additional clock regulatory genes including ZTL, LKP2, LIP1, and LCL1 [62], ELF3, TIC, and PHYA [123], and WNK1, CKB3, and CRY1 [121] were predicted to be potential targets of FIO1, because their mRNAs were hypomethylated in fio1 mutants, although their expression was not dramatically or consistently affected. Additionally, gene ontology analysis suggests that the regulation of the circadian clock is enriched in hypomethylated mRNAs in fio1 mutants [62]. In this regard, it cannot be excluded that FIO1 plays a specific role in the regulation of the circadian clock through m6A methylation.
FIO1 is required for the maintenance of the period length in the core clock, but it is not clear how FIO1 contributes to the proper functioning of the circadian clock. As FIO1 functions as an m6A methylase, it may be involved in the depositing of m6A on the mRNAs of central oscillator genes, which affects their stability. CCA1 and LHY mRNAs are representative targets of FIO1-mediated m6A methylation, but their mRNA levels are not consistently affected in fio1 mutants in different studies, potentially due to different experimental conditions or alleles used. The cumulative effect of m6A on a subset of FIO1 target mRNAs induces changes in RNA stability, alternative 3′ ending, or splicing, which can modulate the activity of the circadian rhythm [113,114,115,138]. Alternatively, other effects of m6A methylation on target mRNAs such as changes in histone modification, nuclear retention, or translational activity, which have not been examined in these studies, may be involved [101,113,116,139].
Another important factor necessary to understand the role of FIO1 in regulating the circadian clock is whether the methylation activity of FIO1 is dynamically regulated by the circadian clock. FIO1 mRNA and protein levels are not controlled by the circadian clock [61]. It is possible that FIO1 generates a dynamic methylation profile on the mRNAs through its association with other m6A methylases, such as MTA-MTB m6A methylase complex and MTC m6A methylase, whose component and own expression, respectively, are regulated in a rhythmic manner (Figure 3a,d) [131]. Alternatively, FIO1 may have rhythmic collaborators that assist it to regulate the dynamic m6A methylation. Expression of genes encoding putative RNA methylases (AT5G51130 and AT5G10620) is diurnally rhythmic and their orthologous proteins can interact with METTL16 in humans and Drosophila [140,141,142]. The rhythmic features of m6A CCGs mRNA could be regulated in other layers of m6A methylation processing by m6A readers or erasers, which might act on different Zeitgeber times (Figure 3b,c,e,f). Global m6A methylation and expression profiles in fio1 mutants under diurnal or free-running conditions can provide a comprehensive and accurate evaluation of the dynamic role of FIO1 on targets in the circadian clock.
FIO1 shares structural similarities with the mammalian METTL16, but their functional effect on m6A methylation might differ. This may be due to the difference in their subcellular localization; FIO1 is localized exclusively in the nucleus, while METTL16 protein exists in both the nuclear and cytoplasmic regions and also in the nucleolus in the G1/S phase [61,143,144]. The cytosolic mammalian METTL16 has additional functions in enhancing the translation efficiency of transcripts through interaction with the eukaryotic translation machinery beyond m6A deposition [133,144]. However, FIO1 is likely to function as a nuclear methylase in the nucleoplasm, although the possibility of residual cytosolic FIO1 having additional functions cannot be excluded. In contrast to METTL16, FIO1 did not induce changes in the global alternative splicing patterns, with only 43 alternative splicing events, although FIO1 also methylated m6A in U6 snRNAs [120]. Additionally, the m6A in four mRNAs that encode for the synthesis of SAM was not affected by FIO1 [62,124]. Importantly, FIO1 installs m6A in plant-specific m6A motifs beyond the consensus sequence motif of U6 snRNAs.
A recent report by Simpson and colleagues (2022), however, highlighted the importance of FIO1-mediated m6A modification in U6 snRNA rather than in mRNAs [124]. The m6A deposition on U6 snRNA by FIO1 is essential for accurate splicing through the preferential selection of the canonical site from the two major 5′ splice sites. RNA-seq analysis of the fio1 mutant revealed widespread deregulation of pre-mRNA splicing by more than 2300 alternative 5′ splicing site selections, which is different from other reports [120,121]. Although m6A levels in more than 2800 sites are altered in the fio1 mutant, over 85% of them overlapped with the hypomethylated sites in the fip37 mutant, implying that FIO1-dependent methylation on mRNAs is relatively small. This might be due to the indirect effect of a 40% loss of MTB activity with premature termination due to altered splicing. Additionally, splicing changes in the mRNAs of several circadian clock genes, LHY and WNK1, and additional clock regulators, including HOS1 and SAR1, were found in fio1. Interestingly, hos1 and sar1 mutants showed early flowering phenotypes with high levels of CO and reduced levels of FLC expression [145,146] and lengthened circadian periods [147], which is similar to that of the fio1 mutant. In addition, global changes to the alternative splicing in loss-of-function mutants of the splicing of factor PRMT5, Type II protein arginine methyltransferase 5 [148,149], and SPLICEOSOMAL TIMEKEEPER LOCUS1 (STIPL1) [150] lengthened the circadian periods, which supports the indirect effect of the hypomethylation of U6 snRNA in fio1 being responsible for its aberrant circadian rhythm. However, FIO1 can directly interact with their targets and affect the m6A levels in their mRNAs [62,121,123], supporting that FIO1 installs direct m6A deposition on specific mRNAs for the regulation of developmental processes. Regardless, FIO1 is not likely a major writer of m6A modifications but rather a more selective methylase that acts on specific mRNAs and U6 snRNAs for the regulation of the circadian clock. Further investigations into the biological implications of FIO1-mediated m6A on U6 snRNA and its target mRNAs are required.

4. Conclusions and Future Directions

In recent years, m6A has emerged as an essential mechanism for RNA metabolism regulation. However, a substantial amount of m6A studies have been conducted in animal systems, but there is a lack of information regarding the mechanistic insights of this RNA modification in plants. Although it is highly conserved among species, plant m6A machinery also contains distinct features or components compared to that of the animal systems such as the plant-specific CPSF30-L isoform [87,88] or the different roles of RBM15 and FPA in animal and plant systems [51,56]. Studies on the RNA regulatory mechanisms of plant m6A in conjunction with animal m6A may provide other perspectives on the conserved and distinct functions of m6A in plants and animals. Furthermore, the number of identified m6A proteins in plants is relatively low compared to that of animals, which is also an obstacle to fully understanding the functions of m6A in plants. Further investigation into the mechanisms for the site- and transcript-specific selection of m6A modification is an interesting direction to be focused on in the future.
The development of novel sequencing technologies has facilitated the functional studies of m6A and provided considerable insights into this regulatory mechanism. However, there are still limitations in these methods that hinder the m6A studies such as the large amounts of inputs, lack of stoichiometric information, and low specificity and efficiency. Furthermore, many of these approaches have not been tested on plant systems. Therefore, the development of an improved, plant-specific method may accelerate the progress in studying m6A in plant systems.
The plant circadian clock is a complex and dynamic system that is coordinately regulated at multiple levels. m6A methylation, a newly emerging layer of epitranscriptomic regulation, has been observed to be prevalent in transcripts of photoreceptors, clock regulatory genes, and CCGs, indicating its role in various hierarchical structures of the clock, including inputs, the central oscillator, and outputs. Genetic evidence from mutant studies for methyl writers has demonstrated the importance of m6A methylation in the regulation of the plant circadian clock.
There is still much to understand regarding the effects and mechanisms of m6A modification on transcripts in clock regulatory genes and how it influences the regulation of the plant circadian clock. Further research should delve deeper into this topic and explore the kinetics of RNA methylation and demethylation in relation to the circadian rhythm and specific regulatory pathways involved in the circadian clock. The function of m6A erasers or readers, as well as other m6A writers, in regulating the circadian clock and their related processes also needs to be clarified. Further investigation into the impact of m6A modification on RNA metabolism and key circadian RNA molecules will improve our understanding of the molecular mechanisms underlying the plant circadian clock.

Author Contributions

Conceptualization, J.H.K. and J.K.; data curation, P.P.T.D., L.W. and J.K.; writing—original draft preparation, N.N.C., P.P.T.D., J.H.K. and J.K.; writing—review and editing, N.N.C., P.P.T.D., J.H.K. and J.K.; visualization, N.N.C. and P.P.T.D.; supervision, J.H.K. and J.K.; funding acquisition, J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the 2021 scientific promotion program funded by Jeju National University.

Acknowledgments

We apologize to all authors whose studies are not included in the present review due to space limitations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Phillips, T. Regulation of transcription and gene expression in eukaryotes. Nat. Educ. 2008, 1, 199. [Google Scholar]
  2. Frye, M.; Harada, B.T.; Behm, M.; He, C. RNA modifications modulate gene expression during development. Science 2018, 361, 1346–1349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Boccaletto, P.; Stefaniak, F.; Ray, A.; Cappannini, A.; Mukherjee, S.; Purta, E.; Kurkowska, M.; Shirvanizadeh, N.; Destefanis, E.; Groza, P.; et al. MODOMICS: A database of RNA modification pathways. 2021 update. Nucleic. Acids. Res. 2022, 50, D231–D235. [Google Scholar] [CrossRef] [PubMed]
  4. Arribas-Hernandez, L.; Bressendorff, S.; Hansen, M.H.; Poulsen, C.; Erdmann, S.; Brodersen, P. An m6A-YTH module controls developmental timing and morphogenesis in Arabidopsis. Plant Cell 2018, 30, 952–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Cui, X.A.; Liang, Z.; Shen, L.S.; Zhang, Q.; Bao, S.J.; Geng, Y.K.; Zhang, B.; Leo, V.; Vardy, L.A.; Lu, T.G.; et al. 5-methylcytosine RNA methylation in Arabidopsis thaliana. Mol. Plant 2017, 10, 1387–1399. [Google Scholar] [CrossRef] [Green Version]
  6. Duan, H.C.; Wei, L.H.; Zhang, C.; Wang, Y.; Chen, L.; Lu, Z.K.; Chen, P.R.; He, C.; Jia, G.F. ALKBH10B is an RNA N-6-methyladenosine demethylase affecting Arabidopsis floral transition. Plant Cell 2017, 29, 2995–3011. [Google Scholar] [CrossRef] [Green Version]
  7. Martinez-Perez, M.; Aparicio, F.; Lopez-Gresa, M.P.; Belles, J.M.; Sanchez-Navarro, J.A.; Pallas, V. Arabidopsis m6A demethylase activity modulates viral infection of a plant virus and the m6A abundance in its genomic RNAs. Proc. Natl. Acad. Sci. USA 2017, 114, 10755–10760. [Google Scholar] [CrossRef] [Green Version]
  8. Ruzicka, K.; Zhang, M.; Campilho, A.; Bodi, Z.; Kashif, M.; Saleh, M.; Eeckhout, D.; El-Showk, S.; Li, H.Y.; Zhong, S.L.; et al. Identification of factors required for m6A mRNA methylation in Arabidopsis reveals a role for the conserved E3 ubiquitin ligase HAKAI. New Phytol. 2017, 215, 157–172. [Google Scholar] [CrossRef] [Green Version]
  9. Scutenaire, J.; Deragon, J.M.; Jean, V.; Benhamed, M.; Raynaud, C.; Favory, J.J.; Merret, R.; Bousquet-Antonelli, C. The YTH domain protein ECT2 is an m6A reader required for normal trichome branching in Arabidopsis. Plant Cell 2018, 30, 986–1005. [Google Scholar] [CrossRef] [Green Version]
  10. Shen, L.S.; Liang, Z.; Gu, X.F.; Chen, Y.; Teo, Z.W.N.; Hou, X.L.; Cai, W.M.; Dedon, P.C.; Liu, L.; Yu, H. N-6-methyladenosine RNA modification regulates shoot stem cell fate in Arabidopsis. Dev. Cell 2016, 38, 186–200. [Google Scholar] [CrossRef] [Green Version]
  11. Wei, L.H.; Song, P.Z.; Wang, Y.; Lu, Z.K.; Tang, Q.; Yu, Q.; Xiao, Y.; Zhang, X.; Duan, H.C.; Jia, G.F. The m6A reader ECT2 controls trichome morphology by affecting mRNA stability in Arabidopsis. Plant Cell 2018, 30, 968–985. [Google Scholar] [CrossRef] [PubMed]
  12. Zuber, H.; Scheer, H.; Ferier, E.; Sement, F.M.; Mercier, P.; Stupfler, B.; Gagliardi, D. Uridylation and PABP cooperate to repair mRNA deadenylated ends in Arabidopsis. Cell Rep. 2016, 14, 2707–2717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Parker, M.T.; Knop, K.; Sherwood, A.V.; Schurch, N.J.; Mackinnon, K.; Gould, P.D.; Hall, A.J.W.; Barton, G.J.; Simpson, G.G. Nanopore direct RNA sequencing maps the complexity of Arabidopsis mRNA processing and m6A modification. Elife 2020, 9, 35. [Google Scholar] [CrossRef] [PubMed]
  14. Wan, Y.Z.; Tang, K.; Zhang, D.Y.; Xie, S.J.; Zhu, X.H.; Wang, Z.G.; Lang, Z.B. Transcriptome-wide high-throughput deep m6A-seq reveals unique differential m6A methylation patterns between three organs in Arabidopsis thaliana. Genome Biol. 2015, 16, 26. [Google Scholar] [CrossRef] [Green Version]
  15. Shi, H.L.; Wei, J.B.; He, C. Where, when, and how: Context-dependent functions of RNA methylation writers, readers, and erasers. Mol. Cell 2019, 74, 640–650. [Google Scholar] [CrossRef]
  16. Yue, H.; Nie, X.J.; Yan, Z.G.; Song, W.N. N6-methyladenosine regulatory machinery in plants: Composition, function and evolution. Plant Biotechnol. J. 2019, 17, 1194–1208. [Google Scholar] [CrossRef] [Green Version]
  17. Luo, G.Z.; MacQueen, A.; Zheng, G.Q.; Duan, H.C.; Dore, L.C.; Lu, Z.K.; Liu, J.; Chen, K.; Jia, G.F.; Bergelson, J.; et al. Unique features of the m6A methylome in Arabidopsis thaliana. Nat. Commun. 2014, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  18. Dominissini, D.; Moshitch-Moshkovitz, S.; Schwartz, S.; Salmon-Divon, M.; Ungar, L.; Osenberg, S.; Cesarkas, K.; Jacob-Hirsch, J.; Amariglio, N.; Kupiec, M.; et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 2012, 485, U201–U284. [Google Scholar] [CrossRef] [PubMed]
  19. Ke, S.D.; Alemu, E.A.; Mertens, C.; Gantman, E.C.; Fak, J.J.; Mele, A.; Haripal, B.; Zucker-Scharff, I.; Moore, M.J.; Park, C.Y.; et al. A majority of m6A residues are in the last exons, allowing the potential for 3′UTR regulation. Genes Dev. 2015, 29, 2037–2053. [Google Scholar] [CrossRef] [Green Version]
  20. Meyer, K.D.; Saletore, Y.; Zumbo, P.; Elemento, O.; Mason, C.E.; Jaffrey, S.R. Comprehensive analysis of mRNA methylation reveals enrichment in 3′UTRs and near stop codons. Cell 2012, 149, 1635–1646. [Google Scholar] [CrossRef] [Green Version]
  21. Slobodin, B.; Han, R.Q.; Calderone, V.; Vrielink, J.; Loayza-Puch, F.; Elkon, R.; Agami, R. Transcription impacts the efficiency of mRNA translation via co-transcriptional N6-adenosine methylation. Cell 2017, 169, 326–337. [Google Scholar] [CrossRef]
  22. Li, Y.L.; Wang, X.L.; Li, C.P.; Hu, S.N.; Yu, J.; Song, S.H. Transcriptome-wide N-6-methyladenosine profiling of rice callus and leaf reveals the presence of tissue-specific competitors involved in selective mRNA modification. RNA Biol. 2014, 11, 1180–1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jia, G.F.; Fu, Y.; Zhao, X.; Dai, Q.; Zheng, G.Q.; Yang, Y.; Yi, C.Q.; Lindahl, T.; Pan, T.; Yang, Y.G.; et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 2011, 7, 885–887. [Google Scholar] [CrossRef] [PubMed]
  24. Linder, B.; Grozhik, A.V.; Olarerin-George, A.O.; Meydan, C.; Mason, C.E.; Jaffrey, S.R. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat. Methods 2015, 12, 767–772. [Google Scholar] [CrossRef] [PubMed]
  25. Yin, R.; Chang, J.W.; Li, Y.S.; Gao, Z.Y.; Qiu, Q.; Wang, Q.F.; Han, G.Q.; Chai, J.H.; Feng, M.D.; Wang, P.P.; et al. Differential m6A RNA landscapes across hematopoiesis reveal a role for IGF2BP2 in preserving hematopoietic stem cell function. Cell Stem Cell 2022, 29, 149–159. [Google Scholar] [CrossRef]
  26. Meyer, K.D. DART-seq: An antibody-free method for global m6A detection. Nat. Methods 2019, 16, 1275–1280. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, Y.; Xiao, Y.; Dong, S.Q.; Yu, Q.; Jia, G.F. Antibody-free enzyme-assisted chemical approach for detection of N-6-methyladenosine. Nat. Chem. Biol. 2020, 16, 896–903. [Google Scholar] [CrossRef]
  28. Hu, L.L.; Liu, S.; Peng, Y.; Ge, R.Q.; Su, R.; Senevirathne, C.; Harada, B.T.; Dai, Q.; Wei, J.B.; Zhang, L.S.; et al. m6A RNA modifications are measured at single-base resolution across the mammalian transcriptome. Nat. Biotechnol. 2022, 40, 1210–1219. [Google Scholar] [CrossRef]
  29. Garalde, D.R.; Snell, E.A.; Jachimowicz, D.; Sipos, B.; Lloyd, J.H.; Bruce, M.; Pantic, N.; Admassu, T.; James, P.; Warland, A.; et al. Highly parallel direct RNA sequencing on an array of nanopores. Nat. Methods 2018, 15, 201–206. [Google Scholar] [CrossRef]
  30. Yin, R.; Li, Y.; Tian, W.; Zhou, F.; Zhang, H. RNA m6A modification: Mapping methods, roles, and mechanisms in acute myeloid leukemia. Blood Sci. 2022, 4, 116–124. [Google Scholar] [CrossRef]
  31. Zhao, X.; Zhang, Y.; Hang, D.; Meng, J.; Wei, Z. Detecting RNA modification using direct RNA sequencing: A systematic review. Comput. Struct. Biotechnol. J. 2022, 20, 5740–5749. [Google Scholar] [CrossRef] [PubMed]
  32. Anderson, S.J.; Kramer, M.C.; Gosai, S.J.; Yu, X.; Vandivier, L.E.; Nelson, A.D.L.; Anderson, Z.D.; Beilstein, M.A.; Fray, R.G.; Lyons, E.; et al. N-6-methyladenosine inhibits local ribonucleolytic cleavage to stabilize mRNAs in Arabidopsis. Cell Rep. 2018, 25, 1146–1157. [Google Scholar] [CrossRef] [Green Version]
  33. Chen, M.J.; Urs, M.J.; Sanchez-Gonzalez, I.; Olayioye, M.A.; Herde, M.; Witte, C.P. m6A RNA degradation products are catabolized by an evolutionarily conserved N-6-methyl-AMP deaminase in plant and mammalian Cells. Plant Cell 2018, 30, 1511–1522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, F.; Zhang, Y.C.; Liao, J.Y.; Yu, Y.; Zhou, Y.F.; Feng, Y.Z.; Yang, Y.W.; Lei, M.Q.; Bai, M.; Wu, H.; et al. The subunit of RNA N6-methyladenosine methyltransferase OsFIP regulates early degeneration of microspores in rice. PLoS Genet. 2019, 15, 19. [Google Scholar] [CrossRef] [Green Version]
  35. Zhou, L.L.; Tian, S.P.; Qin, G.Z. RNA methylomes reveal the m6A-mediated regulation of DNA demethylase gene SlDML2 in tomato fruit ripening. Genome Biol. 2019, 20, 23. [Google Scholar] [CrossRef] [Green Version]
  36. Li, Z.R.; Shi, J.; Yu, L.; Zhao, X.Z.; Ran, L.L.; Hu, D.Y.; Song, B.A. N-6-methyl-adenosine level in Nicotiana tabacum is associated with tobacco mosaic virus. Virol. J. 2018, 15, 10. [Google Scholar] [CrossRef] [Green Version]
  37. Miao, Z.Y.; Zhang, T.; Qi, Y.H.; Song, J.; Han, Z.X.; Ma, C. Evolution of the RNA N-6-methyladenosine methylome mediated by genomic duplication. Plant Physiol. 2020, 182, 345–360. [Google Scholar] [CrossRef] [Green Version]
  38. Reichel, M.; Koster, T.; Staiger, D. Marking RNA: m6A writers, readers, and functions in Arabidopsis. J. Mol. Cell. Biol. 2019, 11, 899–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Heck, A.M.; Wilusz, C.J. Small changes, big implications: The impact of m6A RNA methylation on gene expression in pluripotency and development. Biochim. Biophys. Acta-Gene Regul. Mech. 2019, 1862, 194402. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, N.; Pan, T. N-6-methyladenosine-encoded epitranscriptomics. Nat. Struct. Mol. Biol. 2016, 23, 98–102. [Google Scholar] [CrossRef]
  41. Liao, S.; Sun, H.; Xu, C. YTH domain: A family of N6-methyladenosine (m6A) readers. Genom. Proteom. Bioinf. 2018, 16, 99–107. [Google Scholar] [CrossRef] [PubMed]
  42. Nishat, Z.S.; Hasan, M.S.; Islam, M.S.; Hossain, T.; Ghosh, A. Identification of epitranscriptomic methylation marker genes in Arabidopsis and their expression profiling in response to developmental, anatomical, and environmental modulations. Curr. Plant Biol. 2022, 30, 100247. [Google Scholar] [CrossRef]
  43. Bujnicki, J.M.; Feder, M.; Radlinska, M.; Blumenthal, R.M. Structure prediction and phylogenetic analysis of a functionally diverse family of proteins homologous to the MT-A70 subunit of the human mRNA: m6 a methyltransferase. J. Mol. Evol. 2002, 55, 431–444. [Google Scholar] [CrossRef] [PubMed]
  44. Lence, T.; Paolantoni, C.; Worpenberg, L.; Roignant, J.Y. Mechanistic insights into m6A RNA enzymes. Biochim. Biophys. Acta-Gene Regul. Mech. 2019, 1862, 222–229. [Google Scholar] [CrossRef]
  45. Knuckles, P.; Lence, T.; Haussmann, I.U.; Jacob, D.; Kreim, N.; Carl, S.H.; Masiello, I.; Hares, T.; Villasenor, R.; Hess, D.; et al. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m6A machinery component Wtap/Fl(2)d. Genes Dev. 2018, 32, 415–429. [Google Scholar] [CrossRef] [Green Version]
  46. Bokar, J.A.; Shambaugh, M.E.; Polayes, D.; Matera, A.G.; Rottman, F.M. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N-6-adenosine)-methyltransferase. RNA 1997, 3, 1233–1247. [Google Scholar]
  47. Wang, P.; Doxtader, K.A.; Nam, Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol. Cell 2016, 63, 306–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Liu, J.Z.; Yue, Y.N.; Han, D.L.; Wang, X.; Fu, Y.; Zhang, L.; Jia, G.F.; Yu, M.; Lu, Z.K.; Deng, X.; et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N-6-adenosine methylation. Nat. Chem. Biol. 2014, 10, 93–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Wang, X.; Feng, J.; Xue, Y.; Guan, Z.Y.; Zhang, D.L.; Liu, Z.; Gong, Z.; Wang, Q.; Huang, J.B.; Tang, C.; et al. Structural basis of N-6-adenosine methylation by the METTL3-METTL14 complex. Nature 2016, 534, 575–578. [Google Scholar] [CrossRef] [PubMed]
  50. Ping, X.L.; Sun, B.F.; Wang, L.; Xiao, W.; Yang, X.; Wang, W.J.; Adhikari, S.; Shi, Y.; Lv, Y.; Chen, Y.S.; et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014, 24, 177–189. [Google Scholar] [CrossRef] [Green Version]
  51. Patil, D.P.; Chen, C.K.; Pickering, B.F.; Chow, A.; Jackson, C.; Guttman, M.; Jaffrey, S.R. m6A RNA methylation promotes XIST-mediated transcriptional repression. Nature 2016, 537, 369–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yue, Y.; Liu, J.; Cui, X.; Cao, J.; Luo, G.; Zhang, Z.; Cheng, T.; Gao, M.; Shu, X.; Ma, H.; et al. VIRMA mediates preferential m6A mRNA methylation in 3′UTR and near stop codon and associates with alternative polyadenylation. Cell Discov. 2018, 4, 10. [Google Scholar] [CrossRef]
  53. Wen, J.; Lv, R.T.; Ma, H.H.; Shen, H.J.; He, C.X.; Wang, J.H.; Jiao, F.F.; Liu, H.; Yang, P.Y.; Tan, L.; et al. Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol. Cell 2018, 69, 1028–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bawankar, P.; Lence, T.; Paolantoni, C.; Haussmann, I.U.; Kazlauskiene, M.; Jacob, D.; Heidelberger, J.B.; Richter, F.M.; Nallasivan, M.P.; Morin, V.; et al. Hakai is required for stabilization of core components of the m6A mRNA methylation machinery. Nat. Commun. 2021, 12, 3778. [Google Scholar] [CrossRef] [PubMed]
  55. Zhong, S.L.; Li, H.Y.; Bodi, Z.; Button, J.; Vespa, L.; Herzog, M.; Fray, R.G. MTA is an Arabidopsis messenger RNA adenosine methylase and interacts with a homolog of a sex-specific splicing factor. Plant Cell 2008, 20, 1278–1288. [Google Scholar] [CrossRef] [Green Version]
  56. Parker, M.T.; Knop, K.; Zacharaki, V.; Sherwood, A.V.; Tome, D.; Yu, X.H.; Martin, P.G.; Beynon, J.; Michaels, S.D.; Barton, G.J.; et al. Widespread premature transcription termination of Arabidopsis thaliana NLR genes by the spen protein FPA. Elife 2021, 10, e65537. [Google Scholar] [CrossRef]
  57. Zhang, M.; Bodi, Z.; Mackinnon, K.; Zhong, S.L.; Archer, N.; Mongan, N.P.; Simpson, G.G.; Fray, R.G. Two zinc finger proteins with functions in m6A writing interact with HAKAI. Nat. Commun. 2022, 13, 1227. [Google Scholar] [CrossRef]
  58. Pendleton, K.E.; Chen, B.B.; Liu, K.Q.; Hunter, O.V.; Xie, Y.; Tu, B.P.; Conrad, N.K. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell 2017, 169, 824–835. [Google Scholar] [CrossRef] [Green Version]
  59. Tran, N.V.; Ernst, F.G.M.; Hawley, B.R.; Zorbas, C.; Ulryck, N.; Hackert, P.; Bohnsack, K.E.; Bohnsack, M.T.; Jaffrey, S.R.; Graille, M.; et al. The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res. 2019, 47, 7719–7733. [Google Scholar] [CrossRef] [Green Version]
  60. Ma, H.H.; Wang, X.Y.; Cai, J.B.; Dai, Q.; Natchiar, S.K.; Lv, R.T.; Chen, K.; Lu, Z.K.; Chen, H.; Shi, Y.G.; et al. N-6-methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat. Chem. Biol. 2019, 15, 88–94. [Google Scholar] [CrossRef]
  61. Kim, J.; Kim, Y.; Yeom, M.; Kim, J.H.; Nam, H.G. FIONA1 is essential for regulating period length in the Arabidopsis circadian clock. Plant Cell 2008, 20, 307–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Wang, C.L.; Yang, J.B.; Song, P.Z.; Zhang, W.; Lu, Q.; Yu, Q.; Jia, G.F. FIONA1 is an RNA N-6-methyladenosine methyltransferase affecting Arabidopsis photomorphogenesis and flowering. Genome Biol. 2022, 23, 40. [Google Scholar] [CrossRef] [PubMed]
  63. Luo, Q.; Mo, J.Z.; Chen, H.; Hu, Z.T.; Wang, B.H.; Wu, J.B.; Liang, Z.Y.; Xie, W.H.; Du, K.X.; Peng, M.L.; et al. Structural insights into molecular mechanism for N-6-adenosine methylation by MT-A70 family methyltransferase METTL4. Nat. Commun. 2022, 13, 5636. [Google Scholar] [CrossRef]
  64. Zaccara, S.; Ries, R.J.; Jaffrey, S.R. Reading, writing and erasing mRNA methylation. Nat. Rev. Mol. Cell. Bio. 2019, 20, 608–624. [Google Scholar] [CrossRef] [PubMed]
  65. Batista, P.J.; Molinie, B.; Wang, J.K.; Qu, K.; Zhang, J.J.; Li, L.J.; Bouley, D.M.; Lujan, E.; Haddad, B.; Daneshvar, K.; et al. m6A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 2014, 15, 707–719. [Google Scholar] [CrossRef] [Green Version]
  66. Engel, M.; Eggert, C.; Kaplick, P.M.; Eder, M.; Roh, S.; Tietze, L.; Namendorf, C.; Arloth, J.; Weber, P.; Rex-Haffner, M.; et al. The role of m6A/m-RNA methylation in stress response regulation. Neuron 2018, 99, 389–403. [Google Scholar] [CrossRef] [PubMed]
  67. Xiang, Y.; Laurent, B.; Hsu, C.H.; Nachtergaele, S.; Lu, Z.K.; Sheng, W.Q.; Xu, C.Y.; Hen, H.C.; Jian, O.Y.; Wang, S.Q.; et al. RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature 2017, 543, 573–576. [Google Scholar] [CrossRef] [Green Version]
  68. Yu, J.Y.; Li, Y.; Wang, T.; Zhong, X. Modification of N-6-methyladenosine RNA methylation on heat shock protein expression. PLoS ONE 2018, 13, e0198604. [Google Scholar] [CrossRef]
  69. Zhou, J.; Wan, J.; Gao, X.W.; Zhang, X.Q.; Jaffrey, S.R.; Qian, S.B. Dynamic m6A mRNA methylation directs translational control of heat shock response. Nature 2015, 526, 591–594. [Google Scholar] [CrossRef] [Green Version]
  70. Du, M.J.; Zhang, Y.J.; Mao, Y.S.; Mou, J.W.; Zhao, J.; Xue, Q.; Wang, D.L.; Huang, J.F.; Gao, S.G.; Gao, Y.S. MiR-33a suppresses proliferation of NSCLC cells via targeting METTL3 mRNA. Biochem. Biophys. Res. Commun. 2017, 482, 582–589. [Google Scholar] [CrossRef]
  71. Du, Y.Z.; Hou, G.F.; Zhang, H.L.; Dou, J.Z.; He, J.F.; Guo, Y.M.; Li, L.; Chen, R.; Wang, Y.L.; Deng, R.; et al. SUMOylation of the m6A-RNA methyltransferase METTL3 modulates its function. Nucleic Acids Res. 2018, 46, 5195–5208. [Google Scholar] [CrossRef] [PubMed]
  72. Sorci, M.; Ianniello, Z.; Cruciani, S.; Larivera, S.; Ginistrelli, L.C.; Capuano, E.; Marchioni, M.; Fazi, F.; Fatica, A. METTL3 regulates WTAP protein homeostasis. Cell Death Dis. 2018, 9, 796. [Google Scholar] [CrossRef] [PubMed]
  73. Scholler, E.; Weichmann, F.; Treiber, T.; Ringle, S.; Treiber, N.; Flatley, A.; Feederle, R.; Bruckmann, A.; Meister, G. Interactions, localization, and phosphorylation of the m6A generating METTL3-METTL14-WTAP complex. RNA 2018, 24, 499–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhao, B.X.S.; Roundtree, I.A.; He, C. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 2017, 18, 31–42. [Google Scholar] [CrossRef] [Green Version]
  75. Zheng, G.Q.; Dahl, J.A.; Niu, Y.M.; Fedorcsak, P.; Huang, C.M.; Li, C.J.; Vagbo, C.B.; Shi, Y.; Wang, W.L.; Song, S.H.; et al. ALKBH5 Is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 2013, 49, 18–29. [Google Scholar] [CrossRef] [Green Version]
  76. Scarrow, M.; Chen, N.; Sun, G.L. Insights into the N-6-methyladenosine mechanism and its functionality: Progress and questions. Crit. Rev. Biotechnol. 2020, 40, 639–652. [Google Scholar] [CrossRef]
  77. Mielecki, D.; Zugaj, D.L.; Muszewska, A.; Piwowarski, J.; Chojnacka, A.; Mielecki, M.; Nieminuszczy, J.; Grynberg, M.; Grzesiuk, E. Novel AlkB dioxygenases-alternative models for in silico and in vivo studies. PLoS ONE 2012, 7, e30588. [Google Scholar] [CrossRef] [Green Version]
  78. Patil, D.P.; Pickering, B.F.; Jaffrey, S.R. Reading m6A in the transcriptome: m6A-binding proteins. Trends Cell Biol. 2018, 28, 113–127. [Google Scholar] [CrossRef]
  79. Wojtas, M.N.; Pandey, R.R.; Mendel, M.; Homolka, D.; Sachidanandam, R.; Pillai, R.S. Regulation of m6 A transcripts by the 3′ -> 5′ RNA helicase YTHDC2 is essential for a successful meiotic program in the mammalian germline. Mol. Cell 2017, 68, 374–387. [Google Scholar] [CrossRef] [Green Version]
  80. Liu, N.; Dai, Q.; Zheng, G.Q.; He, C.; Parisien, M.; Pan, T. N-6-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature 2015, 518, 560–564. [Google Scholar] [CrossRef] [Green Version]
  81. Liu, N.A.; Zhou, K.I.; Parisien, M.; Dai, Q.; Diatchenko, L.; Pan, T. N-6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 2017, 45, 6051–6063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Huang, H.L.; Weng, H.Y.; Sun, W.J.; Qin, X.; Shi, H.L.; Wu, H.Z.; Zhao, B.S.; Mesquita, A.; Liu, C.; Yuan, C.L.; et al. Recognition of RNA N-6- methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 2018, 20, 285–295. [Google Scholar] [CrossRef] [PubMed]
  83. Theler, D.; Dominguez, C.; Blatter, M.; Boudet, J.; Allain, F.H.T. Solution structure of the YTH domain in complex with N6-methyladenosine RNA: A reader of methylated RNA. Nucleic Acids Res. 2014, 42, 13911–13919. [Google Scholar] [CrossRef] [PubMed]
  84. Stoilov, P.; Rafalska, I.; Stamm, S. YTH: A new domain in nuclear proteins. Trends Biochem. Sci. 2002, 27, 495–497. [Google Scholar] [CrossRef] [PubMed]
  85. Song, P.Z.; Yang, J.B.; Wang, C.L.; Lu, Q.; Shi, L.Q.; Tayier, S.; Jia, G.F. Arabidopsis N-6-methyladenosine reader CPSF30-L recognizes FUE signals to control polyadenylation site choice in liquid-like nuclear bodies. Mol. Plant 2021, 14, 571–587. [Google Scholar] [CrossRef]
  86. Li, D.Y.; Zhang, H.J.; Hong, Y.B.; Huang, L.; Li, X.H.; Zhang, Y.F.; Ouyang, Z.G.; Song, F.M. Genome-wide identification, biochemical characterization, and expression analyses of the YTH domain-containing RNA-binding protein family in Arabidopsis and Rice. Plant Mol. Biol. Rep. 2014, 32, 1169–1186. [Google Scholar] [CrossRef]
  87. Delaney, K.J.; Xu, R.Q.; Zhang, J.X.; Li, Q.Q.; Yun, K.Y.; Falcone, D.L.; Hunt, A.G. Calmodulin interacts with and regulates the RNA-binding activity of an Arabidopsis polyadenylation factor subunit. Plant Physiol. 2006, 140, 1507–1521. [Google Scholar] [CrossRef] [Green Version]
  88. Hunt, A.G.; Xing, D.H.; Li, Q.S.Q. Plant polyadenylation factors: Conservation and variety in the polyadenylation complex in plants. BMC Genom. 2012, 13, 641. [Google Scholar] [CrossRef] [Green Version]
  89. Du, H.; Zhao, Y.; He, J.Q.; Zhang, Y.; Xi, H.R.; Liu, M.F.; Ma, J.B.; Wu, L.G. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat. Commun. 2016, 7, 11. [Google Scholar] [CrossRef]
  90. Wang, X.; Lu, Z.K.; Gomez, A.; Hon, G.C.; Yue, Y.N.; Han, D.L.; Fu, Y.; Parisien, M.; Dai, Q.; Jia, G.F.; et al. N-6-methyladenosine-dependent regulation of messenger RNA stability. Nature 2014, 505, 117–120. [Google Scholar] [CrossRef] [Green Version]
  91. Wang, Y.; Li, Y.; Toth, J.I.; Petroski, M.D.; Zhang, Z.L.; Zhao, J.C. N-6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat. Cell Biol. 2014, 16, 191–198. [Google Scholar] [CrossRef] [Green Version]
  92. Kramer, M.C.; Janssen, K.A.; Palos, K.; Nelson, A.D.L.; Vandivier, L.E.; Garcia, B.A.; Lyons, E.; Beilstein, M.A.; Gregory, B.D. N-6-methyladenosine and RNA secondary structure affect transcript stability and protein abundance during systemic salt stress in Arabidopsis. Plant Direct 2020, 4, e00239. [Google Scholar] [CrossRef] [PubMed]
  93. Spitale, R.C.; Flynn, R.A.; Zhang, Q.C.; Crisalli, P.; Lee, B.; Jung, J.W.; Kuchelmeister, H.Y.; Batista, P.J.; Torre, E.A.; Kool, E.T.; et al. Structural imprints in vivo decode RNA regulatory mechanisms. Nature 2015, 519, 486–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Hou, Y.F.; Sun, J.; Wu, B.X.; Gao, Y.Y.; Nie, H.B.; Nie, Z.T.; Quan, S.X.; Wang, Y.; Cao, X.F.; Li, S.S. CPSF30-L-mediated recognition of mRNA m6A modification controls alternative polyadenylation of nitrate signaling-related gene transcripts in Arabidopsis. Mol. Plant 2021, 14, 688–699. [Google Scholar] [CrossRef]
  95. Pontier, D.; Picart, C.; El Baidouri, M.; Roudier, F.; Xu, T.; Lahmy, S.; Llauro, C.; Azevedo, J.; Laudie, M.; Attina, A.; et al. The m6A pathway protects the transcriptome integrity by restricting RNA chimera formation in plants. Life Sci. Alliance 2019, 2, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Luo, J.H.; Wang, Y.; Wang, M.; Zhang, L.Y.; Peng, H.R.; Zhou, Y.Y.; Jia, G.F.; He, Y. Natural variation in RNA m6A methylation and its relationship with translational status. Plant Physiol. 2020, 182, 332–344. [Google Scholar] [CrossRef] [Green Version]
  97. Bhat, S.S.; Bielewicz, D.; Gulanicz, T.; Bodi, Z.; Yu, X.; Anderson, S.J.; Szewc, L.; Bajczyk, M.; Dolata, J.; Grzelak, N.; et al. mRNA adenosine methylase (MTA) deposits m6A on pri-miRNAs to modulate miRNA biogenesis in Arabidopsis thaliana. Proc. Natl. Acad. Sci. USA 2020, 117, 21785–21795. [Google Scholar] [CrossRef]
  98. Alarcon, C.R.; Lee, H.; Goodarzi, H.; Halberg, N.; Tavazoie, S.F. N-6-methyladenosine marks primary microRNAs for processing. Nature 2015, 519, 482–485. [Google Scholar] [CrossRef] [Green Version]
  99. Alarcon, C.R.; Goodarzi, H.; Lee, H.; Liu, X.H.; Tavazoie, S.; Tavazoie, S.F. HNRNPA2B1 is a mediator of m6A-dependent nuclear RNA processing events. Cell 2015, 162, 1299–1308. [Google Scholar] [CrossRef] [Green Version]
  100. Chen, T.; Hao, Y.J.; Zhang, Y.; Li, M.M.; Wang, M.; Han, W.F.; Wu, Y.S.; Lv, Y.; Hao, J.; Wang, L.B.; et al. m6A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. Cell Stem Cell 2015, 16, 289–301. [Google Scholar] [CrossRef] [Green Version]
  101. Shim, S.; Lee, H.G.; Lee, H.; Seo, P.J. H3K36me2 is highly correlated with m6 A modifications in plants. J. Integr. Plant. Biol. 2020, 62, 1455–1460. [Google Scholar] [CrossRef]
  102. Dodd, A.N.; Salathia, N.; Hall, A.; Kevei, E.; Toth, R.; Nagy, F.; Hibberd, J.M.; Millar, A.J.; Webb, A.A. Plant circadian clocks increase photosynthesis, growth, survival, and competitive advantage. Science 2005, 309, 630–633. [Google Scholar] [CrossRef] [PubMed]
  103. Goodspeed, D.; Chehab, E.W.; Min-Venditti, A.; Braam, J.; Covington, M.F. Arabidopsis synchronizes jasmonate-mediated defense with insect circadian behavior. Proc. Natl. Acad. Sci. USA 2012, 109, 4674–4677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Alabadi, D.; Oyama, T.; Yanovsky, M.J.; Harmon, F.G.; Mas, P.; Kay, S.A. Reciprocal regulation between TOC1 and LHY/CCA1 within the Arabidopsis circadian clock. Science 2001, 293, 880–883. [Google Scholar] [CrossRef] [PubMed]
  105. Farre, E.M.; Harmer, S.L.; Harmon, F.G.; Yanovsky, M.J.; Kay, S.A. Overlapping and distinct roles of PRR7 and PRR9 in the Arabidopsis circadian clock. Curr. Biol. 2005, 15, 47–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Herrero, E.; Kolmos, E.; Bujdoso, N.; Yuan, Y.; Wang, M.; Berns, M.C.; Uhlworm, H.; Coupland, G.; Saini, R.; Jaskolski, M.; et al. EARLY FLOWERING4 recruitment of EARLY FLOWERING3 in the nucleus sustains the Arabidopsis circadian clock. Plant Cell 2012, 24, 428–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Hsu, P.Y.; Devisetty, U.K.; Harmer, S.L. Accurate timekeeping is controlled by a cycling activator in Arabidopsis. Elife 2013, 2, e00473. [Google Scholar] [CrossRef]
  108. Rugnone, M.L.; Faigon Soverna, A.; Sanchez, S.E.; Schlaen, R.G.; Hernando, C.E.; Seymour, D.K.; Mancini, E.; Chernomoretz, A.; Weigel, D.; Mas, P.; et al. LNK genes integrate light and clock signaling networks at the core of the Arabidopsis oscillator. Proc. Natl. Acad. Sci. USA 2013, 110, 12120–12125. [Google Scholar] [CrossRef] [Green Version]
  109. Mas, P.; Alabadi, D.; Yanovsky, M.J.; Oyama, T.; Kay, S.A. Dual role of TOC1 in the control of circadian and photomorphogenic responses in Arabidopsis. Plant Cell 2003, 15, 223–236. [Google Scholar] [CrossRef] [Green Version]
  110. Fujiwara, S.; Wang, L.; Han, L.; Suh, S.S.; Salome, P.A.; McClung, C.R.; Somers, D.E. Post-translational regulation of the Arabidopsis circadian clock through selective proteolysis and phosphorylation of pseudo-response regulator proteins. J. Biol. Chem. 2008, 283, 23073–23083. [Google Scholar] [CrossRef] [Green Version]
  111. Yang, J.; Li, L.; Li, X.; Zhong, M.; Li, X.; Qu, L.; Zhang, H.; Tang, D.; Liu, X.; He, C.; et al. The blue light receptor CRY1 interacts with FIP37 to promote N(6)-methyladenosine RNA modification and photomorphogenesis in Arabidopsis. New Phytol. 2022, 237, 840–854. [Google Scholar] [CrossRef] [PubMed]
  112. Covington, M.F.; Maloof, J.N.; Straume, M.; Kay, S.A.; Harmer, S.L. Global transcriptome analysis reveals circadian regulation of key pathways in plant growth and development. Genome Biol. 2008, 9, R130. [Google Scholar] [CrossRef] [PubMed]
  113. Romanowski, A.; Yanovsky, M.J. Circadian rhythms and post-transcriptional regulation in higher plants. Front. Plant Sci. 2015, 6, 437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kojima, S.; Shingle, D.L.; Green, C.B. Post-transcriptional control of circadian rhythms. J. Cell. Sci. 2011, 124, 311–320. [Google Scholar] [CrossRef] [Green Version]
  115. Mateos, J.L.; de Leone, M.J.; Torchio, J.; Reichel, M.; Staiger, D. Beyond transcription: Fine-tuning of circadian timekeeping by post-transcriptional regulation. Genes 2018, 9, 616. [Google Scholar] [CrossRef] [Green Version]
  116. Fustin, J.M.; Doi, M.; Yamaguchi, Y.; Hida, H.; Nishimura, S.; Yoshida, M.; Isagawa, T.; Morioka, M.S.; Kakeya, H.; Manabe, I.; et al. RNA-methylation-dependent RNA processing controls the speed of the circadian clock. Cell 2013, 155, 793–806. [Google Scholar] [CrossRef] [Green Version]
  117. Fustin, J.M.; Kojima, R.; Itoh, K.; Chang, H.Y.; Ye, S.; Zhuang, B.; Oji, A.; Gibo, S.; Narasimamurthy, R.; Virshup, D.; et al. Two Ck1delta transcripts regulated by m6A methylation code for two antagonistic kinases in the control of the circadian clock. Proc. Natl. Acad. Sci. USA 2018, 115, 5980–5985. [Google Scholar] [CrossRef] [Green Version]
  118. Wang, C.Y.; Yeh, J.K.; Shie, S.S.; Hsieh, I.C.; Wen, M.S. Circadian rhythm of RNA N6-methyladenosine and the role of cryptochrome. Biochem. Biophys. Res. Commun. 2015, 465, 88–94. [Google Scholar] [CrossRef]
  119. Ruocco, M.; Ambrosino, L.; Jahnke, M.; Chiusano, M.L.; Barrote, I.; Procaccini, G.; Silva, J.; Dattolo, E. m6A RNA methylation in marine plants: First insights and relevance for Biological Rhythms. Int. J. Mol. Sci. 2020, 21, 7508. [Google Scholar] [CrossRef]
  120. Wang, X.; Jiang, B.; Gu, L.; Chen, Y.; Mora, M.; Zhu, M.; Noory, E.; Wang, Q.; Lin, C. A photoregulatory mechanism of the circadian clock in Arabidopsis. Nat. Plants 2021, 7, 1397–1408. [Google Scholar] [CrossRef]
  121. Xu, T.; Wu, X.; Wong, C.E.; Fan, S.; Zhang, Y.; Zhang, S.; Liang, Z.; Yu, H.; Shen, L. FIONA1-mediated m6A modification regulates the floral transition in Arabidopsis. Adv. Sci. 2022, 9, e2103628. [Google Scholar] [CrossRef] [PubMed]
  122. Sun, B.; Bhati, K.K.; Song, P.; Edwards, A.; Petri, L.; Kruusvee, V.; Blaakmeer, A.; Dolde, U.; Rodrigues, V.; Straub, D.; et al. FIONA1-mediated methylation of the 3′UTR of FLC affects FLC transcript levels and flowering in Arabidopsis. PLoS Genet. 2022, 18, e1010386. [Google Scholar] [CrossRef] [PubMed]
  123. Jing, C.; Hu, J.; Amara, U.; Park, S.J.; Li, Y.; Jeong, D.; Lee, I.; Xu, T.; Kang, H. FIONA1 is an N6-methyladenosine methyltransferase that plays a vital role in floral transition via affecting the splicing of FLC and the stability of SPL3 and SEP3 in Arabidopsis. J. Exp. Bot. 2022, erac461. [Google Scholar] [CrossRef]
  124. Parker, M.T.; Soanes, B.K.; Kusakina, J.; Larrieu, A.; Knop, K.; Joy, N.; Breidenbach, F.; Sherwood, A.V.; Barton, G.J.; Fica, S.M.; et al. m6A modification of U6 snRNA modulates usage of two major classes of pre-mRNA 5′ splice site. Elife 2022, 11, e78808. [Google Scholar] [CrossRef] [PubMed]
  125. Bodi, Z.; Zhong, S.; Mehra, S.; Song, J.; Graham, N.; Li, H.; May, S.; Fray, R.G. Adenosine methylation in Arabidopsis mRNA is associated with the 3′ end and reduced levels cause developmental defects. Front. Plant. Sci. 2012, 3, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Somers, D.E.; Devlin, P.F.; Kay, S.A. Phytochromes and cryptochromes in the entrainment of the Arabidopsis circadian clock. Science 1998, 282, 1488–1490. [Google Scholar] [CrossRef] [Green Version]
  127. Devlin, P.F.; Kay, S.A. Cryptochromes are required for phytochrome signaling to the circadian clock but not for rhythmicity. Plant Cell 2000, 12, 2499–2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Nohales, M.A.; Kay, S.A. GIGANTEA gates gibberellin signaling through stabilization of the DELLA proteins in Arabidopsis. Proc. Natl. Acad. Sci. USA 2019, 116, 21893–21899. [Google Scholar] [CrossRef] [Green Version]
  129. Pedmale, U.V.; Huang, S.C.; Zander, M.; Cole, B.J.; Hetzel, J.; Ljung, K.; Reis, P.A.B.; Sridevi, P.; Nito, K.; Nery, J.R.; et al. Cryptochromes interact directly with PIFs to control plant growth in limiting blue light. Cell 2016, 164, 233–245. [Google Scholar] [CrossRef] [Green Version]
  130. Quail, P.H. Photobodies reveal their secret. Nat. Plants 2021, 7, 1326–1327. [Google Scholar] [CrossRef]
  131. Mockler, T.C.; Michael, T.P.; Priest, H.D.; Shen, R.; Sullivan, C.M.; Givan, S.A.; McEntee, C.; Kay, S.A.; Chory, J. The DIURNAL project: DIURNAL and circadian expression profiling, model-based pattern matching, and promoter analysis. Cold Spring Harb. Symp. Quant. Biol. 2007, 72, 353–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Arribas-Hernandez, L.; Brodersen, P. Occurrence and functions of m6A and other covalent modifications in plant mRNA. Plant Physiol. 2020, 182, 79–96. [Google Scholar] [CrossRef] [PubMed]
  133. Su, R.; Dong, L.; Li, Y.; Gao, M.; He, P.C.; Liu, W.; Wei, J.; Zhao, Z.; Gao, L.; Han, L.; et al. METTL16 exerts an m6A-independent function to facilitate translation and tumorigenesis. Nat. Cell Biol. 2022, 24, 205–216. [Google Scholar] [CrossRef] [PubMed]
  134. Doxtader, K.A.; Wang, P.; Scarborough, A.M.; Seo, D.; Conrad, N.K.; Nam, Y. Structural basis for regulation of METTL16, an S-adenosylmethionine homeostasis factor. Mol. Cell 2018, 71, 1001–1011.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Madhani, H.D.; Bordonne, R.; Guthrie, C. Multiple roles for U6 snRNA in the splicing pathway. Genes Dev. 1990, 4, 2264–2277. [Google Scholar] [CrossRef] [PubMed]
  136. Ishigami, Y.; Ohira, T.; Isokawa, Y.; Suzuki, Y.; Suzuki, T. A single m6A modification in U6 snRNA diversifies exon sequence at the 5′ splice site. Nat. Commun. 2021, 12, 3244. [Google Scholar] [CrossRef] [PubMed]
  137. Mendel, M.; Chen, K.M.; Homolka, D.; Gos, P.; Pandey, R.R.; McCarthy, A.A.; Pillai, R.S. Methylation of structured RNA by the m6A writer METTL16 Is essential for mouse embryonic development. Mol. Cell 2018, 71, 986–1000.e11. [Google Scholar] [CrossRef] [Green Version]
  138. Malapeira, J.; Khaitova, L.C.; Mas, P. Ordered changes in histone modifications at the core of the Arabidopsis circadian clock. Proc. Natl. Acad. Sci. USA 2012, 109, 21540–21545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Lin, S.B.; Choe, J.; Du, P.; Triboulet, R.; Gregory, R.I. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell 2016, 62, 335–345. [Google Scholar] [CrossRef] [Green Version]
  140. Blomen, V.A.; Majek, P.; Jae, L.T.; Bigenzahn, J.W.; Nieuwenhuis, J.; Staring, J.; Sacco, R.; van Diemen, F.R.; Olk, N.; Stukalov, A.; et al. Gene essentiality and synthetic lethality in haploid human cells. Science 2015, 350, 1092–1096. [Google Scholar] [CrossRef]
  141. Guruharsha, K.G.; Rual, J.F.; Zhai, B.; Mintseris, J.; Vaidya, P.; Vaidya, N.; Beekman, C.; Wong, C.; Rhee, D.Y.; Cenaj, O.; et al. A protein complex network of Drosophila melanogaster. Cell 2011, 147, 690–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Szklarczyk, D.; Gable, A.L.; Lyon, D.; Junge, A.; Wyder, S.; Huerta-Cepas, J.; Simonovic, M.; Doncheva, N.T.; Morris, J.H.; Bork, P.; et al. STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019, 47, D607–D613. [Google Scholar] [CrossRef]
  143. Stixova, L.; Komurkova, D.; Svobodova Kovarikova, A.; Fagherazzi, P.; Bartova, E. Localization of METTL16 at the nuclear periphery and the nucleolus is cell cycle-specific and METTL16 interacts with several nucleolar proteins. Life 2021, 11, 669. [Google Scholar] [CrossRef] [PubMed]
  144. Nance, D.J.; Satterwhite, E.R.; Bhaskar, B.; Misra, S.; Carraway, K.R.; Mansfield, K.D. Characterization of METTL16 as a cytoplasmic RNA binding protein. PLoS ONE 2020, 15, e0227647. [Google Scholar] [CrossRef] [PubMed]
  145. Lazaro, A.; Valverde, F.; Pineiro, M.; Jarillo, J.A. The Arabidopsis E3 ubiquitin ligase HOS1 negatively regulates CONSTANS abundance in the photoperiodic control of flowering. Plant Cell 2012, 24, 982–999. [Google Scholar] [CrossRef] [Green Version]
  146. Cheng, Z.; Zhang, X.; Huang, P.; Huang, G.; Zhu, J.; Chen, F.; Miao, Y.; Liu, L.; Fu, Y.F.; Wang, X. Nup96 and HOS1 are mutually stabilized and gate CONSTANS protein level, conferring long-day photoperiodic flowering regulation in Arabidopsis. Plant Cell 2020, 32, 374–391. [Google Scholar] [CrossRef]
  147. MacGregor, D.R.; Gould, P.; Foreman, J.; Griffiths, J.; Bird, S.; Page, R.; Stewart, K.; Steel, G.; Young, J.; Paszkiewicz, K.; et al. High expression of osmotically responsive genes1 is required for circadian periodicity through the promotion of nucleo-cytoplasmic mRNA export in Arabidopsis. Plant Cell 2013, 25, 4391–4404. [Google Scholar] [CrossRef] [Green Version]
  148. Sanchez, S.E.; Petrillo, E.; Beckwith, E.J.; Zhang, X.; Rugnone, M.L.; Hernando, C.E.; Cuevas, J.C.; Godoy Herz, M.A.; Depetris-Chauvin, A.; Simpson, C.G.; et al. A methyl transferase links the circadian clock to the regulation of alternative splicing. Nature 2010, 468, 112–116. [Google Scholar] [CrossRef] [Green Version]
  149. Hong, S.; Song, H.R.; Lutz, K.; Kerstetter, R.A.; Michael, T.P.; McClung, C.R. Type II protein arginine methyltransferase 5 (PRMT5) is required for circadian period determination in Arabidopsis thaliana. Proc. Natl. Acad. Sci. USA 2010, 107, 21211–21216. [Google Scholar] [CrossRef] [Green Version]
  150. Jones, M.A.; Williams, B.A.; McNicol, J.; Simpson, C.G.; Brown, J.W.; Harmer, S.L. Mutation of Arabidopsis spliceosomal timekeeper locus1 causes circadian clock defects. Plant Cell 2012, 24, 4066–4082. [Google Scholar] [CrossRef]
Figure 1. Schematic model of m6A modifications and its regulatory roles in RNA metabolism. m6A writers, erasers, readers, and other proteins/protein complexes are illustrated in yellow, blue, red, and green, respectively. The colorful circles with dashed lines indicate unidentified/unconfirmed components/factors/regulations in the plant system. m6A writers install a methyl group at the N-6 position of an adenosine base after binding to the RRACH (R = G/A; H: U/A/C) consensus sequence. Various plant writers have been identified along with their homologs in the animal system. The presence of other plant writers and motifs, should they exist, remains to be identified. m6A modifications can be removed enzymatically by erasers. Plant erasers contain only members from the ALBHKs family. m6A readers are responsible for recognizing m6A-containing transcripts. Plants readers include members from the ECT family and CPSF30. Structural changes in RNA after m6A modification or the recognition of m6A modifications by m6A readers can facilitate or inhibit the interaction between RNA and various protein complexes, thus directly or indirectly regulating RNA metabolisms including RNA degradation, RNA stabilization, RNA splicing, miRNA processing, and histone modification.
Figure 1. Schematic model of m6A modifications and its regulatory roles in RNA metabolism. m6A writers, erasers, readers, and other proteins/protein complexes are illustrated in yellow, blue, red, and green, respectively. The colorful circles with dashed lines indicate unidentified/unconfirmed components/factors/regulations in the plant system. m6A writers install a methyl group at the N-6 position of an adenosine base after binding to the RRACH (R = G/A; H: U/A/C) consensus sequence. Various plant writers have been identified along with their homologs in the animal system. The presence of other plant writers and motifs, should they exist, remains to be identified. m6A modifications can be removed enzymatically by erasers. Plant erasers contain only members from the ALBHKs family. m6A readers are responsible for recognizing m6A-containing transcripts. Plants readers include members from the ECT family and CPSF30. Structural changes in RNA after m6A modification or the recognition of m6A modifications by m6A readers can facilitate or inhibit the interaction between RNA and various protein complexes, thus directly or indirectly regulating RNA metabolisms including RNA degradation, RNA stabilization, RNA splicing, miRNA processing, and histone modification.
Plants 12 00624 g001
Figure 2. A proposed model depicting the involvement of FIO1 and general m6A writer complexes in regulating circadian clock, flowering, and hypocotyl elongation in Arabidopsis. Diverse pathways respond to various external (photoperiod, vernalization, and ambient temperature) and internal (autonomous, gibberellins, and circadian oscillator) stimuli/signals to regulate these processes are shown in red and blue, respectively. FIO1 prevents premature flowering and maintains the circadian period by affecting the expression level, splicing, and/or stability of several key circadian clock transcripts (e.g., LHY and CCA1), flowering regulator transcripts (e.g., SOC1 and FLC), and photomorphogenesis-related genes (e.g., PIF4) through direct m6A methylation of the 3′ UTR and a subset on the CDS region, or indirectly through m6A methylation of U6 snRNA. The general m6A writer complex (including MTA, MTB, FIP37, VIR, and HAKAI) recognition motifs are mainly present in the 3′ UTR region. PIF37 can interact with cryptochromes (CRY1) and increases the m6A modification of PIF3, PIF4, and PIF5, which consequently reduces their RNA stability and promotes photomorphogenesis [111]. However, CRY2 recruits MTA, MTB, and PIF37 to methylate several core circadian clock genes and enhances their mRNA stability [62]. The involvement of other m6A writer components such as VIR and HAKAI in the regulation of these processes requires further investigation.
Figure 2. A proposed model depicting the involvement of FIO1 and general m6A writer complexes in regulating circadian clock, flowering, and hypocotyl elongation in Arabidopsis. Diverse pathways respond to various external (photoperiod, vernalization, and ambient temperature) and internal (autonomous, gibberellins, and circadian oscillator) stimuli/signals to regulate these processes are shown in red and blue, respectively. FIO1 prevents premature flowering and maintains the circadian period by affecting the expression level, splicing, and/or stability of several key circadian clock transcripts (e.g., LHY and CCA1), flowering regulator transcripts (e.g., SOC1 and FLC), and photomorphogenesis-related genes (e.g., PIF4) through direct m6A methylation of the 3′ UTR and a subset on the CDS region, or indirectly through m6A methylation of U6 snRNA. The general m6A writer complex (including MTA, MTB, FIP37, VIR, and HAKAI) recognition motifs are mainly present in the 3′ UTR region. PIF37 can interact with cryptochromes (CRY1) and increases the m6A modification of PIF3, PIF4, and PIF5, which consequently reduces their RNA stability and promotes photomorphogenesis [111]. However, CRY2 recruits MTA, MTB, and PIF37 to methylate several core circadian clock genes and enhances their mRNA stability [62]. The involvement of other m6A writer components such as VIR and HAKAI in the regulation of these processes requires further investigation.
Plants 12 00624 g002
Figure 3. Expression of genes encoding proteins involved in m6A modification in plants under diurnal and free-running conditions. Relative expression of rhythmic genes in the m6A writer (a,d), m6A eraser (b,e), and m6A reader (c,f) in diurnal (ac) and free-running conditions (df) was determined as a normalized value against the maximum value of expression at all-time points. Genes involved in m6A metabolism are selected according to [132]. Raw expression data were derived from the DIURNAL project (http://diurnal.mocklerlab.org, accessed on 2 January 2023) and rhythmic genes in diurnal and free-running conditions were determined based on the correction cutoff value = 0.8 and the condition = LDHC and LL23_LDHH (the DIURNAL project), respectively [131].
Figure 3. Expression of genes encoding proteins involved in m6A modification in plants under diurnal and free-running conditions. Relative expression of rhythmic genes in the m6A writer (a,d), m6A eraser (b,e), and m6A reader (c,f) in diurnal (ac) and free-running conditions (df) was determined as a normalized value against the maximum value of expression at all-time points. Genes involved in m6A metabolism are selected according to [132]. Raw expression data were derived from the DIURNAL project (http://diurnal.mocklerlab.org, accessed on 2 January 2023) and rhythmic genes in diurnal and free-running conditions were determined based on the correction cutoff value = 0.8 and the condition = LDHC and LL23_LDHH (the DIURNAL project), respectively [131].
Plants 12 00624 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Chuong, N.N.; Doan, P.P.T.; Wang, L.; Kim, J.H.; Kim, J. Current Insights into m6A RNA Methylation and Its Emerging Role in Plant Circadian Clock. Plants 2023, 12, 624. https://doi.org/10.3390/plants12030624

AMA Style

Chuong NN, Doan PPT, Wang L, Kim JH, Kim J. Current Insights into m6A RNA Methylation and Its Emerging Role in Plant Circadian Clock. Plants. 2023; 12(3):624. https://doi.org/10.3390/plants12030624

Chicago/Turabian Style

Chuong, Nguyen Nguyen, Phan Phuong Thao Doan, Lanshuo Wang, Jin Hee Kim, and Jeongsik Kim. 2023. "Current Insights into m6A RNA Methylation and Its Emerging Role in Plant Circadian Clock" Plants 12, no. 3: 624. https://doi.org/10.3390/plants12030624

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop