Next Article in Journal
Wild Species from the Asteraceae Family, Traditionally Consumed in Some Mediterranean Countries
Previous Article in Journal
Genome-Wide Identification and Salt Stress-Responsive Expression Analysis of the GmPLATZ Gene Family in Soybean (Glycine max L.)
Previous Article in Special Issue
Understanding the Brassinosteroid-Dependent Environmental Adaption in Brassicaceae Plants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epitranscriptomic Control of Drought Tolerance in Rice: The Role of RNA Methylation

1
School of Chemistry and Life Science, Anshan Normal University, Anshan 114007, China
2
Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan 114007, China
3
Wuxi Branch of Jiangsu Academy of Agricultural, Wuxi 214000, China
4
State Key Laboratory of Green Pesticide and Guizhou University, Guiyang 550025, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Plants 2025, 14(13), 2002; https://doi.org/10.3390/plants14132002
Submission received: 30 May 2025 / Revised: 23 June 2025 / Accepted: 29 June 2025 / Published: 30 June 2025

Abstract

Drought stress is a predominant abiotic constraint adversely affecting global rice (Oryza sativa) production and threatening food security. While the transcriptional and post-transcriptional regulation of drought-responsive pathways has been widely investigated, the emerging field of epitranscriptomics, particularly RNA chemical modifications such as N6-methyladenosine (m6A), adds a new dimension to gene regulation under stress. The most prevalent internal modification in eukaryotic messenger RNA influences RNA metabolism by interacting dynamically with enzymes that add, remove, or recognize the modification. Recent studies in rice reveal that m6A deposition is not static but dynamically regulated in response to water-deficit conditions, influencing transcript stability, splicing, nuclear export, and translation efficiency of key drought-responsive genes. This review critically synthesizes current findings on the distribution and functional implications of m6A and other epitranscriptomic marks (e.g., 5-methylcytosine [m5C], pseudouridine [Ψ]) in modulating rice responses to drought. We discuss the regulatory circuitry involving m6A effectors such as OsMTA, OsFIP37, and YTH domain proteins and their integration with known drought-signaling pathways including ABA and reactive oxygen species (ROS) cascades. We also highlight emerging high-resolution technologies such as m6A-seq, direct RNA sequencing, and nanopore-based detection that facilitate epitranscriptomic profiling in rice. Finally, we propose future directions for translating epitranscriptomic knowledge into crop improvement, including CRISPR/Cas-based modulation of RNA modification machinery to enhance drought tolerance.

1. Introduction

Rice (Oryza sativa L.) is one of the most widely cultivated and consumed cereal crops worldwide, serving as a primary food source for over 50% of the global population [1]. However, rice is particularly vulnerable to drought stress due to its semi-aquatic origin and high water requirements during critical growth stages such as flowering and grain filling [2]. Prolonged drought leads to stunted growth, reduced biomass, spikelet sterility, and significant yield loss, posing a serious threat to global food security. Several rice cultivars, such as IR64, Swarna, and Pusa Basmati-1, are known to be highly sensitive to water-deficit conditions [3], while drought-tolerant varieties like N22, Sahbhagi Dhan, and Apo exhibit more robust physiological and molecular responses to stress.
Drought is one of the most damaging abiotic stresses impacting global crop productivity, significantly reducing both yield and quality. Rice, a staple for more than half of the world’s population, is particularly sensitive to drought due to its high water requirements during key developmental stages [4]. As climate change intensifies evapotranspiration and diminishes freshwater availability, drought-induced yield losses in rice have already surpassed those from other environmental stressors combined. Consequently, enhancing drought tolerance in rice is a priority for sustainable agriculture and food security [5]. While extensive research has uncovered key transcriptional, hormonal, and signaling mechanisms underlying drought responses, post-transcriptional regulation via epitranscriptomic modifications has only recently gained attention. Among these, N6-methyladenosine (m6A) methylation has emerged as a critical RNA modification involved in fine-tuning gene expression during plant stress responses. Historically, efforts to enhance drought tolerance in rice have primarily focused on genetic and transcriptional regulation involving transcription factors, drought-inducible promoters, and hormonal pathways, particularly abscisic acid (ABA) signaling. However, accumulating evidence now highlights the critical role of epitranscriptomic regulation, especially RNA methylation, as a key modulator of gene expression in response to environmental stresses [6].
Among the various RNA modifications, m6A is the most prevalent and dynamically regulated internal modification of eukaryotic mRNA. The addition of m6A is catalyzed by a conserved multiprotein methyltransferase complex, commonly referred to as the “writer” complex, which includes homologs of Methyltransferase-Like 3 (METTL3), e.g., (OsMTA), METTL14, and Wilms’ Tumor 1-Associating Protein (WTAP) in rice (OsFIP37); enzymes such as ALKBH10B remove methyl groups to enable reversibility, while mainly YTH-domain-containing proteins recognize methylated transcripts and regulate their splicing, stability, translation, and localization within the cell [7].
In rice, recent transcriptome-wide m6A profiling under drought stress conditions has revealed that m6A deposition is not random but dynamically modulated in response to stress stimuli [8,9]. For instance, drought stress induces selective methylation of transcripts encoding heat shock proteins and transcription factors (e.g., Dehydration-Responsive Element-Binding protein 2A (DREB2A); NAC-family proteins, a group of plant-specific transcription factors including No Apical Meristem (NAM), (Arabidopsis Transcription Activation Factor 1/2 ATAF1/2), and Cup-shaped Cotyledon 2 (CUC2); and reactive oxygen species (ROS) scavenging enzymes), thereby enhancing mRNA stability and translation efficiency [10]. Moreover, m6A methylation facilitates rapid post-transcriptional responses without requiring new transcription, allowing rice plants to fine-tune gene expression in a timely and energy-efficient manner under stress [11].
Importantly, the pattern and degree of m6A methylation vary significantly between drought-sensitive and drought-tolerant rice varieties. Studies have shown that drought-tolerant cultivars maintain more stable m6A methylomes and better preserve the expression of protective genes under stress, whereas drought-sensitive cultivars often exhibit m6A dysregulation, leading to impaired stress responses [12,13].
This review aims to systematically examine the role of RNA methylation, particularly m6A, in shaping drought stress responses in rice [14]. We describe the key components of the m6A machinery in rice, highlight the dynamic changes in RNA methylation patterns during drought, and discuss how this epitranscriptomic layer of regulation can be leveraged to breed drought-resilient rice through molecular breeding and genome editing strategies.

2. Epitranscriptomic RNA Methylation: A Critical Layer in Drought Stress Adaptation of Rice

Drought stress adversely affects key physiological functions in rice, including reducing stomatal conductance, limiting carbon dioxide (CO2) uptake, and impairing photosynthetic efficiency. Drought stress adversely affects key physiological functions in rice by triggering a cascade of water-conservation responses that ultimately hinder photosynthetic performance. One of the earliest responses is a reduction in stomatal conductance, as the stomata close to minimize water loss through transpiration [15]. However, this also limits the diffusion of CO2 into the leaf, thereby restricting CO2 assimilation in the chloroplasts. As a result, the Calvin cycle is compromised, leading to reduced carbohydrate synthesis and lower biomass accumulation. Moreover, drought-induced oxidative stress and damage to the photosynthetic apparatus, particularly photosystem II, further decrease photosynthetic efficiency. These combined effects not only impair energy production but also disrupt metabolic homeostasis, severely constraining plant growth and productivity under drought conditions [16].
Drought stress impairs chloroplast photochemistry and the efficient utilization of absorbed light energy, leading to a significant reduction in photosynthetic performance and ultimately hampering CO2 fixation. If drought or other stress impairs photochemistry, it means the plant’s ability to absorb light, excite electrons, or convert light energy into chemical energy is disrupted, leading to reduced carbon fixation and growth [17]. When drought or other environmental stresses disrupt photochemistry, the plant’s ability to capture light, excite electrons, and generate energy-rich molecules like Adenosine Triphosphate (ATP) and Nicotinamide Adenine Dinucleotide Phosphate (NADPH) is significantly compromised. This disruption limits the energy supply necessary for the Calvin cycle, where CO2 is fixed into carbohydrates. The reduction in carbon fixation leads to lower production of sugars and other essential metabolites, which are crucial for cellular growth, maintenance, and reproduction. Over time, this hampers overall plant biomass accumulation and yield [18]. Furthermore, impaired photochemistry can lead to the overaccumulation of excess light energy, causing the generation of ROS that can damage cellular components unless adequately scavenged by antioxidant systems. Thus, drought-induced impairment of photochemistry not only reduces energy conversion efficiency but also increases oxidative stress, further challenging plant survival and productivity [19]. It impairs chloroplast photochemistry and light energy utilization, ultimately hampering carbon fixation. Being sessile organisms, plants must rely on intricate regulatory networks to perceive and adapt to such stressors while maintaining growth and reproduction [20]. Among these, epigenetic regulation plays a crucial role in mediating gene expression under drought. Within the broader epigenetic framework including DNA methylation, histone modifications, and small RNAs, RNA methylation, particularly m6A, has emerged as a critical and reversible epitranscriptomic mark [21]. m6A is the most prevalent internal RNA modification in eukaryotes, including Oryza sativa, Arabidopsis thaliana, and Nicotiana benthamiana. It is enriched at RRACH (R = purine; H = A/C/U) and plant-specific UGUA motifs, especially near 3′ untranslated regions (3′ UTRs), stop codons, and transcription start sites [22].
In rice, as in A. thaliana, m6A methylation influences transcript stability, alternative splicing, nuclear export, and translation efficiency, playing a vital role in drought adaptation (Table 1) [7]. This modification is catalyzed by a conserved “writer” complex, comprising METTL3 (MTA homolog), METTL14 (MTB homolog), FIP37 (WTAP homolog), VIRILIZER (KIAA1429 homolog), and HAKAI components with putative homologs in rice that shape its drought-responsive m6A landscape [23]. Demethylation is mediated by “eraser” enzymes such as ALKBH9B and ALKBH10B (identified in A. thaliana), which allow dynamic reversibility. “Reader” proteins like ECT2, ECT3, and ECT4 interpret m6A marks, directing methylated transcripts toward degradation or enhanced translation, enabling a rapid and flexible stress response. Transcriptome-wide analyses using MeRIP-seq and m6A-seq in drought-stressed rice have revealed dynamic m6A modifications in transcripts encoding transcription factors, ABA signaling components, and other stress-responsive proteins [24].
Similar findings in A. thaliana and tobacco underscore m6A’s conserved role in stabilizing or destabilizing mRNAs involved in stress adaptation. Despite its importance, m6A detection remains technically challenging. Immunoprecipitation-based methods such as MeRIP-seq and m6A-seq offer transcriptome-scale insights but lack single-nucleotide resolution [25]. While the limitations of m6A detection methods such as MeRIP-seq and miCLIP are acknowledged, a more comprehensive discussion is needed on how these technical constraints impact the interpretation of methylation data and the identification of biologically functional m6A sites [26]. MeRIP-seq offers transcriptome-wide coverage but suffers from low resolution (typically ~100 nucleotides), making it difficult to precisely localize methylation sites and potentially leading to ambiguous associations with regulatory elements such as stop codons or 3′ UTRs. miCLIP, though capable of single-nucleotide resolution, is technically demanding and prone to crosslinking artifacts and biases related to antibody specificity [27]. These limitations can complicate downstream analyses, including motif enrichment studies, correlation with gene expression changes, and integration with other omics datasets. A more detailed explanation from the authors on how such methodological constraints may lead to false positives/negatives or misinterpretation of m6A’s functional roles would significantly strengthen the manuscript’s discussion of data reliability and biological relevance [28]. Advanced techniques like methylation-iCLIP (miCLIP) achieve base-resolution mapping by capturing crosslinking-induced mutations during reverse transcription [29]. These technologies have elucidated both conserved and species-specific m6A landscapes critical for drought stress regulation in rice (Figure 1) [30].
Table 1. Core components of the m6A machinery and their putative functions in rice under drought stress.
Table 1. Core components of the m6A machinery and their putative functions in rice under drought stress.
Component TypeGene Name (Rice)Arabidopsis HomologPutative FunctionKnown/Proposed Role in Drought StressReference
WriterOsMTAAtMTA (METTL3)Catalytic subunit of the m6A methyltransferase complexMay regulate drought-responsive transcripts via m6A deposition; expression modulated under abiotic stress[31,32]
WriterOsMTBAtMTB (METTL14)Forms heterodimer with OsMTA; provides structural supportFacilitates target specificity of OsMTA; role in drought not yet clarified[33]
WriterOsFIP37FIP37Adaptor protein linking MTA-MTB complex to RNA targetsEssential for embryogenesis in Arabidopsis; drought role in rice unknown[33]
EraserOsALKBH2ALKBH9B/10Bm6A demethylasePotentially removes m6A from transcripts to modulate gene expression under stress; deregulated under stress[13,34]
ReaderOsECT2ECT2YTH-domain-containing protein that binds m6A-modified RNAsMay regulate transcript stability and translation during drought[35]
ReaderOsYTHDF1-likeYTHDF1/2/3Cytoplasmic reader of m6A marksControls translation efficiency and mRNA decay of stress-responsive genes[35,36,37]
This table summarizes current knowledge and gaps regarding the rice m6A machinery, useful for highlighting future gene-targeting strategies.
The figure illustrates the role of epitranscriptomic RNA methylation, particularly m6A, as a critical regulatory layer in rice adaptation to drought stress. Under drought conditions, specific methyltransferase enzymes such as OsMTA7 and OsNUN2 catalyze the addition of methyl groups (m6A and m5C) on mRNA transcripts. These modifications occur at various stages, including in pre-mRNA during splicing and in mature mRNA. The methylation enhances mRNA stability and translation efficiency in the cytoplasm, facilitating the expression of drought-adaptive genes. Conversely, OsALKBH enzymes remove methyl groups, ensuring the dynamic and reversible nature of this epitranscriptomic mark. This balance between writers and erasers regulates the expression of genes involved in key drought response pathways, including ABA signaling, root development, osmoprotectant accumulation, and antioxidant enzyme production. Collectively, these molecular processes support enhanced drought tolerance in rice by fine-tuning gene expression at the RNA level. This dynamic regulation of mRNA through methylation and demethylation underscores the importance of epitranscriptomic modifications as a critical adaptive mechanism in plants under abiotic stress.
Several core components of the m6A methylation machinery have been identified in rice, with homologs in A. thaliana showing conserved roles in stress response (Table 1). For instance, OsMTA, homologous to AtMTA (METTL3), serves as the catalytic subunit of the methyltransferase complex and is known to be modulated under abiotic stress. OsMTB (METTL14-like) interacts with OsMTA to ensure target specificity, although its direct involvement in drought stress adaptation in rice remains to be clarified. Additionally, proteins such as OsFIP37 and OsALKBH2 may act as RNA-binding adaptors and demethylases, respectively, suggesting a dynamic regulation of m6A deposition and removal in stress-responsive mRNAs. Reader proteins like OsECT2 and OsYTHDF1-like further influence transcript fate post methylation, regulating their stability and translation efficiency (Table 1).

2.1. Overview of RNA Methylation in Plants: Types, Distribution, and Detection Technologies

RNA methylation is a crucial epitranscriptomic modification that plays a significant role in regulating RNA stability, processing, and overall function in plants [38,39]. Among the various types of RNA modifications identified, N6-methyladenosine (m6A) is the most prevalent internal modification found within messenger RNA (mRNA). Other important modifications include 5-methylcytosine (m5C) and pseudouridine (Ψ), which occur in a wide range of RNA species such as transfer RNA (tRNA), ribosomal RNA (rRNA), and long non-coding RNAs [40]. These modifications collectively influence numerous biological processes by modulating RNA metabolism and gene expression. In plants, m6A methylation occurs predominantly at conserved sequence motifs such as RRACH (where R is a purine and H is A, C, or U) and the plant-specific UGUA motif [22]. This modification is enriched near critical transcript regions including stop codons, 3′ untranslated regions (3′UTRs), and transcription start sites, where it regulates mRNA splicing, stability, export, and translation. Meanwhile, m5C is primarily detected in tRNAs and rRNAs but has recently been observed in mRNAs, where it affects RNA stability and translation efficiency. Pseudouridine, formed by isomerization of uridine, contributes to RNA structural stability and enhances translation fidelity [4].
The distribution and abundance of these methylations vary across plant species and tissues, reflecting their specific roles in development and environmental responses. For instance, studies in model plants like A. thaliana and key crops such as rice (O. sativa) and tobacco (N. benthamiana) reveal conserved yet species-specific methylation patterns that underpin their adaptive capabilities [22]. Recent technological advances have greatly enhanced our ability to detect and map RNA methylation, deepening our understanding of epitranscriptomic regulation in plants. Commonly used methods like methylated RNA immunoprecipitation sequencing (MeRIP-seq) and m6A-seq allow researchers to profile methylation across the transcriptome, though they lack precise single-nucleotide resolution [41]. To address this limitation, techniques such as methylation individual-nucleotide-resolution crosslinking and immunoprecipitation (miCLIP) have been developed, enabling base-resolution mapping of m6A sites by detecting antibody-induced mutations during reverse transcription. Emerging tools like nanopore direct RNA sequencing, which does not require reverse transcription, and chemical labeling approaches for m5C and pseudouridine are further expanding the scope and accuracy of RNA modification detection. Collectively, these technologies are revealing the dynamic and complex roles of RNA methylations in plant stress responses, including critical adaptations to drought conditions [42].

2.1.1. Translational Potential of Epitranscriptomics in Crop Improvement

Recent advances in understanding the role of m6A RNA methylation in plant stress responses have opened new avenues for crop improvement [43]. Functionally validated m6A-associated genes, particularly those involved in the regulation of drought-responsive transcription factors (e.g., DREB, NAC, bZIP families), ABA signaling components, and antioxidant enzymes, represent promising candidates for molecular breeding [44]. These genes and their epitranscriptomic modifications can serve as molecular markers for marker-assisted selection (MAS), enabling breeders to screen and select varieties with enhanced drought tolerance more efficiently [45].
Furthermore, the rapid progress in genome editing technologies, such as CRISPR/Cas systems, offers opportunities to precisely manipulate m6A-related genes or the m6A consensus motifs on target transcripts [29]. Editing m6A writer (e.g., METTL3 homologs), eraser (e.g., ALKBH homologs), or reader proteins can modulate the m6A landscape, thereby stabilizing or enhancing the translation of key stress-responsive mRNAs. Such targeted epitranscriptomic engineering can improve plant resilience without altering the underlying DNA sequence significantly, offering a novel layer of genetic regulation to exploit [46].
Integration of epitranscriptomic data into genomic selection (GS) models could further enhance breeding accuracy by capturing regulatory variation not detectable at the DNA sequence level alone. Multi-omics datasets incorporating m6A profiles alongside genomics, transcriptomics, and phenomics can provide a comprehensive framework for predicting drought tolerance phenotypes [47,48]. Proof-of-concept studies in model plants have demonstrated that overexpression of m6A writers or readers can enhance stress tolerance by improving mRNA stability and translation of drought-related genes. Translating these findings to crops holds considerable promise but requires careful functional validation and field evaluation [49]. Overall, leveraging the translational potential of epitranscriptomics offers a powerful complement to traditional and molecular breeding approaches, accelerating the development of drought-resilient crop varieties to meet the challenges of climate change and food security [50].

2.1.2. N6-Methyladenosine (m6A): A Key Epitranscriptomic Mark

N6-methyladenosine (m6A) is the most prevalent internal modification found in eukaryotic messenger RNA (mRNA), playing a crucial role in regulating RNA metabolism and gene expression (Figure 2). This dynamic and reversible epitranscriptomic mark affects multiple aspects of RNA function, including stability, translation, splicing, and localization, thus influencing vital biological processes such as plant responses to environmental stresses like drought. m6A modifications predominantly occur within conserved sequence motifs, most notably the RRACH consensus (where R represents purines A or G, and H is A, C, or U). In plants, a distinctive UGUA motif also serves as a methylation site, underscoring species-specific methylation patterns. These methylations are enriched near transcript features such as stop codons, 3′UTRs, and transcription start sites (TSSs), which is fundamental to their role in controlling mRNA, including its stability and translational efficiency. The addition of m6A marks is catalyzed by a multicomponent methyltransferase complex, and the key components of this complex include MTA (the METTL3 homolog responsible for catalytic activity), MTB (METTL14 homolog that enhances substrate specificity and complex stability), FIP37 (WTAP homolog guiding the complex to RNA targets), VIRILIZER (KIAA1429 homolog involved in complex assembly), and HAKAI, an E3-ubiquitin-ligase-like protein that supports complex integrity. These writer proteins are evolutionarily conserved across eukaryotes, including important plant models such as A. thaliana and major crops like Oryza sativa, highlighting their indispensable role in epitranscriptomic regulation. Importantly, m6A marks are reversible through the action of enzymes, such as ALKBH9B and ALKBH10B in plants, which belong to the AlkB family of dioxygenases that catalyze oxidative demethylation. This reversibility allows plants to dynamically modulate gene expression in response to developmental signals and environmental stresses, including drought. The biological outcomes of m6A modifications are mediated by proteins that specifically recognize methylated RNA and influence its fate. In plants, prominent readers include the YTH domain-containing proteins ECT2, ECT3, and ECT4, which bind m6A-modified transcripts to regulate mRNA stability, translation, and subcellular localization.
Figure 2 illustrates the core components involved in m6A RNA methylation in plants, including the methyltransferase complexes that catalyze the addition of m6A marks on RNA molecules. Demethylases remove these modifications, enabling dynamic regulation. Specific proteins recognize and bind m6A sites to mediate downstream effects on RNA stability, splicing, and translation. This machinery collectively modulates gene expression and plays critical roles in plant development and stress responses.
These readers enable plants to rapidly adjust gene expression post-transcriptionally under stress conditions by directing the degradation or enhanced translation of specific transcripts. To study m6A distribution and function, several high-throughput mapping technologies have been developed. Techniques such as MeRIP-seq and m6A-seq use antibody-based enrichment coupled with sequencing to identify m6A-enriched regions transcriptome-wide, but lack single-nucleotide precision. The miCLIP method addresses this limitation by employing UV crosslinking and antibody-induced mutations during reverse transcription to achieve single-nucleotide resolution mapping of m6A sites. Emerging tools like nanopore direct RNA sequencing enable detection of RNA modifications without the need for reverse transcription or amplification, while chemical labeling strategies enhance the mapping of other RNA methylation marks, collectively advancing our understanding of m6A’s dynamic role in plant stress responses, including drought tolerance.

2.2. Tools for Mapping m6A: MeRIP-Seq, m6A-Seq, miCLIP

Mapping m6A modifications across the transcriptome relies on specialized high-throughput sequencing techniques that combine antibody-based enrichment with next-generation sequencing. MeRIP-seq (methylated RNA immunoprecipitation sequencing) and m6A-seq are widely used methods that immunoprecipitate fragmented RNA using m6A-specific antibodies, followed by sequencing to identify methylated regions transcriptome-wide. While these approaches efficiently reveal m6A-enriched peaks, their resolution is limited to about 100–200 nucleotides, making it difficult to pinpoint the exact modified adenosine within the enriched region.
To overcome this, miCLIP (methylation individual-nucleotide-resolution crosslinking and immunoprecipitation) was developed. miCLIP utilizes UV-induced crosslinking between the antibody and RNA, causing specific mutations or truncations during reverse transcription that serve as precise markers form m6A sites. This enables single-nucleotide-resolution mapping, providing a more detailed and accurate landscape of m6A modifications. Emerging technologies, such as nanopore direct RNA sequencing, also show promise for detecting m6A modifications directly on native RNA molecules without the need for amplification or conversion, further enhancing epitranscriptomic analysis (Figure 2).

Functional Validation of m6A RNA Methylation

Functional validation of m6A RNA methylation is crucial for understanding its regulatory role in plant drought stress responses. While transcriptome-wide studies such as MeRIP-seq have identified numerous m6A-modified genes under drought, these findings remain largely correlative without experimental validation. In A. thaliana, genetic studies using loss- and gain-of-function mutants have demonstrated that core m6A writers like MTA, MTB, and FIP37 are essential for normal growth and stress adaptation [36,51]. For example, MTA-deficient plants show increased sensitivity to drought, indicating that m6A plays a positive role in stress tolerance. Additionally, functional analyses of m6A reader proteins, such as ECT2, have revealed their ability to bind methylated transcripts and regulate their stability and translation, further supporting the functional importance of m6A in stress-responsive gene expression [52]. More precise techniques, including miCLIP and CRISPR-mediated mutagenesis of specific m6A sites, have shown that m6A modifications can directly influence the fate of target transcripts like DREB2A and RD29A, which are critical for drought response. Despite these advances, functional validation in rice remains limited. Most rice m6A machinery components have been identified based on homology to Arabidopsis genes, but their physiological roles under drought stress have not been experimentally confirmed [53]. Future studies should focus on generating knockout or overexpression lines in rice, performing RNA stability assays, and integrating epitranscriptomic data with other regulatory layers such as histone modifications and DNA methylation to gain a comprehensive understanding of m6A-mediated drought adaptation [54].
m6A modification has been implicated in regulating various drought adaptation pathways, including transcription factor activity, ABA signaling, and oxidative stress responses (Table 2). ABA improves the efficiency of stress responses by coordinating physiological, molecular, and biochemical pathways enabling plants to survive and recover from adverse environmental conditions with minimal energy cost and maximum resilience [55]. For instance, in A. thaliana, m6A increases the stability of DREB2A and NAC transcripts, crucial for drought-induced transcriptional reprogramming. However, similar functional validations are lacking in rice. Moreover, components of the ABA signaling cascade such as PYR/PYL receptors and SnRK2 kinases are known m6A targets in maize and A. thaliana, but their m6A-mediated regulation in rice remains poorly characterized. Importantly, the interplay between m6A and chromatin-level epigenetic mechanisms, as well as the circadian clock and antioxidant defenses, underscores the epitranscriptomic control over multiple layers of stress adaptation (Table 2).

2.3. Role of m6A RNA Methylation in Drought Stress Adaptation

m6A RNA methylation has emerged as a crucial epitranscriptomic mechanism that modulates plant responses to drought stress by fine-tuning gene expression at multiple regulatory levels. One key role of m6A is its influence on the expression of transcription factors (TFs) that orchestrate drought-responsive pathways [63]. By affecting the stability and translation efficiency of mRNAs encoding TFs such as DREB, NAC, and MYB families, m6A ensures rapid and dynamic control of the transcriptional programs necessary for stress adaptation. This regulation allows plants to activate defense genes promptly without permanently altering their genome [64]. In addition to transcription factors, m6A methylation modulates components of the ABA signaling pathway, which is central to drought response. ABA mediates stomatal closure, osmotic adjustment, and expression of protective proteins under drought. Studies have shown that m6A marks regulate the transcript levels and translational output of ABA receptors, signaling intermediates, and downstream effectors, thereby fine-tuning ABA sensitivity and response intensity [65]. This epitranscriptomic control helps plants balance growth and stress tolerance efficiently. Beyond individual genes, m6A exerts broad regulation over complex stress-responsive gene networks. By selectively methylating transcripts involved in reactive oxygen species (ROS) detoxification, osmolyte biosynthesis, and cellular repair, m6A shapes the overall gene expression landscape that underpins drought resilience. This systemic regulation ensures coordinated activation of protective pathways while preventing unnecessary energy expenditure [66].
Mechanistically, m6A impacts transcript stability, alternative splicing, and translation during drought stress. Methylated RNAs may exhibit altered degradation rates, resulting in either stabilization or rapid turnover depending on the context and the associated reader proteins [67]. m6A can influence splicing patterns to generate stress-specific isoforms, and enhance or repress translation of key drought-responsive proteins. These multifaceted roles enable plants to dynamically reprogram their proteome in response to fluctuating water availability. Comparative studies in A. thaliana and other model plants have provided valuable insights into the conserved and species-specific aspects of m6A-mediated drought adaptation [68]. While the core machinery and many target transcripts are conserved, variations in methylation patterns and reader protein functions contribute to differential drought tolerance phenotypes. This comparative knowledge is critical for translating fundamental epitranscriptomic discoveries into crop improvement strategies for drought resilience [69].

3. Dynamic m6A Methylation Landscape Under Drought in Rice

Recent transcriptome-wide studies have begun to unravel the dynamic changes in m6A RNA methylation profiles in rice subjected to drought stress [46]. Using techniques like MeRIP-seq and miCLIP, researchers have mapped thousands of m6A sites across the rice transcriptome, revealing a global remodeling of methylation patterns under water-deficit conditions. These studies highlight that drought stress triggers both gain and loss of m6A marks on specific sets of mRNAs, suggesting that m6A modification acts as a rapid and reversible layer of gene regulation that complements transcriptional responses [46].
Temporal and spatial profiling of m6A under drought further reveals that methylation patterns vary significantly with the duration of stress and among different tissues [70]. For instance, early drought exposure may induce m6A modifications in transcripts involved in signaling and transcriptional regulation, while prolonged stress targets genes related to cellular protection and metabolic adjustments [71]. Tissue-specific differences also exist; roots often show distinct m6A remodeling compared to leaves, reflecting their unique roles in water sensing and conservation. This spatiotemporal plasticity underscores m6A’s role as a finely tuned regulator adapting to complex drought scenarios [72].
Functional enrichment analyses of drought-responsive m6A-modified transcripts consistently point to categories involved in stress signaling, hormone pathways (notably ABC), osmoprotection, and ROS scavenging. Genes associated with these processes are often methylated to modulate their expression post-transcriptionally, thereby enhancing the plant’s ability to mitigate drought-induced damage. This selective m6A methylation ensures prioritized regulation of critical pathways required for survival under limited water availability [73].
Importantly, changes in the m6A methylation landscape correlate strongly with physiological and metabolic responses observed in drought-stressed rice [46]. Enhanced m6A modification of transcripts involved in stomatal closure, root architecture remodeling, and osmolyte accumulation coincide with observed phenotypic traits such as reduced transpiration, deeper rooting, and improved cellular osmotic balance [74]. This correlation demonstrates that m6A-mediated post-transcriptional regulation is integral to linking environmental cues to adaptive physiological mechanisms, ultimately influencing drought tolerance at the whole-plant level [34].

3.1. Cross-Talk Between RNA Methylation and Other Regulatory Pathways

3.1.1. Interplay with Small RNAs (miRNAs, siRNAs)

RNA methylation, particularly m6A, interacts closely with small RNA pathways to orchestrate gene expression under drought stress (Figure 3) [4]. m6A modifications can influence the biogenesis and stability of microRNAs (miRNAs) and small interfering RNAs (siRNAs), which are crucial post-transcriptional regulators [75]. For example, m6A methylation of primary miRNA transcripts may affect their processing efficiency by Dicer-like enzymes, altering the pool of mature miRNAs that target stress-responsive genes [4]. Conversely, small RNAs can regulate the expression of RNA methylation machinery components, creating a regulatory feedback loop. This cross-talk ensures coordinated fine-tuning of gene silencing and activation during drought adaptation [76].
Figure 3 depicts the key molecular components responsible for adding, removing, and interpreting m6A modifications on RNA in plants. The methyltransferase complex catalyzes the methylation of adenosine residues, marking RNA molecules with m6A. Demethylase enzymes can reverse this modification, allowing dynamic regulation of RNA function. Specialized reader proteins recognize these m6A marks and influence RNA processes such as splicing, stability, export, and translation. Together, these components form a regulatory network that controls gene expression and impacts various physiological processes including growth, development, and responses to environmental stress in plants like A. thaliana [77].

3.1.2. Interaction with Chromatin Modifiers and Histone Marks

RNA methylation pathways do not operate in isolation but interact with chromatin-level regulators to achieve integrated control of gene expression [78]. Evidence suggests that m6A-modified transcripts can influence chromatin state indirectly by modulating the expression or activity of chromatin remodelers and histone-modifying enzymes [79]. This interplay facilitates coordinated regulation at both transcriptional and post-transcriptional levels, enhancing the plasticity of gene expression programs under drought stress [80].

3.1.3. Coordination with Transcriptional and Translational Controls

m6A RNA methylation acts as a bridge between transcriptional regulation and translational control, modulating the flow of genetic information in response to drought [4]. While transcription factors control the initial mRNA synthesis, m6A modifications influence mRNA splicing, nuclear export, stability, and translation efficiency [7]. This coordination allows plants to rapidly adjust protein synthesis independently of transcriptional changes, which is crucial during acute stress. By affecting mRNA fate, m6A ensures that transcripts encoding key drought-responsive proteins are timely translated or degraded, optimizing resource allocation and cellular homeostasis [81].

3.1.4. Potential Feedback Loops Between ABA Signaling and RNA Methylation

ABA is a central hormone in drought response, and emerging evidence highlights bidirectional feedback between ABA signaling and RNA methylation pathways [81]. ABA can modulate the expression and activity of m6A writers, erasers, and readers, altering the epitranscriptomic landscape to favor drought tolerance. In turn, m6A modifications regulate transcripts involved in ABA biosynthesis, signaling components such as receptors and transcription factors, thus fine-tuning hormone sensitivity and response amplitude [75]. These feedback loops create a dynamic regulatory network allowing plants to adaptively recalibrate ABA signaling according to stress severity and duration, ensuring robust yet flexible drought tolerance mechanisms [67].

4. Critical Issues and Research Gaps

Despite rapid advances in understanding RNA modifications, particularly m6A, several critical issues and research gaps remain in elucidating their role in drought tolerance in rice [61]. One major limitation is the insufficient functional characterization of the core m6A machinery including writers, erasers, and readers in rice. Although these components have been identified based on homology with A. thaliana, their specific roles under drought stress have not been thoroughly explored. There is a notable absence of mutant or overexpression lines in rice that could help define their contributions to drought adaptation [82].
Furthermore, a comprehensive understanding of their spatial expression patterns and regulatory dynamics in response to drought is lacking. Another key gap lies in the incomplete identification of drought-responsive m6A-modified transcripts in rice. While some studies have applied MeRIP-seq or m6A-seq in general stress contexts, few have examined the transcriptome-wide m6A landscape specifically under drought, particularly with high resolution or temporal depth [83]. This limits our understanding of how m6A marks shift over the course of drought stress, and there is minimal insight into tissue- or cell-type-specific methylation changes. Additionally, integration of m6A methylation data with transcriptomics, proteomics, and physiological traits remains underdeveloped, restricting the identification of functionally relevant targets [84].
The biological consequences of m6A modifications in rice drought response also remain ambiguous. Whether m6A predominantly stabilizes or destabilizes transcripts, or how it modulates splicing and translation of specific mRNAs under drought stress, is not well established [4]. These mechanisms are often inferred from model systems rather than directly demonstrated in rice, leaving a knowledge gap regarding their true regulatory outcomes. This gap is compounded by the limited investigation into how m6A interacts with other regulatory layers such as small RNAs, histone modifications, or hormone signaling pathways [85]. In particular, the cross-talk between m6A methylation and ABA biosynthesis and signaling central to drought response is poorly understood. There is also limited information on how m6A might influence miRNA activity or interact with the chromatin landscape in a stress-responsive manner [86].
From an applied perspective, epitranscriptomic insights have yet to be translated into breeding strategies or practical applications for drought-resilient rice. There are currently no established biomarkers based on RNA methylation for selecting drought-tolerant cultivars [87]. Tools for precise epigenome editing of m6A marks in rice are still in early stages, and natural genetic variation in m6A machinery across rice varieties remains largely unexplored. Finally, technical constraints continue to hinder progress, as commonly used m6A mapping methods like MeRIP-seq lack single-nucleotide resolution, which is crucial for identifying functional methylation sites [88]. Low abundance of modified transcripts under specific conditions may also obscure key targets. To overcome these limitations, multi-omics platforms integrating RNA-seq, m6A profiling, ribosome footprinting, and metabolic data are urgently needed to create a holistic view of m6A-mediated drought response in rice [89].

5. Future Research Directions

To fully realize the potential of RNA methylation as a target for improving drought tolerance in rice, several key research directions must be prioritized. First, functional genomics studies are urgently needed to elucidate the roles of RNA methylation regulatory proteins writers, erasers, and readers using CRISPR/Cas9-based gene editing, overexpression, or knockdown approaches. Particular attention should be given to tissue-specific and stress-inducible expression patterns to uncover spatial and temporal regulation under drought conditions.
Second, high-resolution mapping of RNA modifications using techniques such as miCLIP or nanopore-based direct RNA sequencing should be extended to drought-treated rice plants. These studies should include time-course analyses and organ-specific profiling to capture how the modification landscape dynamically changes during drought progression and recovery. Integrating these data with transcriptomic, proteomic, and metabolomic datasets will be essential for identifying functionally significant target genes and pathways.
Third, mechanistic studies are needed to dissect the functional effects of RNA methylation on RNA stability, alternative splicing, nuclear export, and translational efficiency under drought stress. Additionally, the cross-regulatory interactions between RNA methylation, small RNAs, histone modifications, and phytohormone signaling, particularly ABA, require detailed investigation using multi-omic and genetic approaches.
Fourth, future efforts should focus on translating epitranscriptomic insights into breeding programs by identifying natural allelic variations in RNA methylation regulators across diverse rice germplasms. The development of epigenome-editing tools to manipulate RNA modifications in a site-specific manner could open new avenues for precision crop improvement. Moreover, the identification of RNA modification-based biomarkers associated with drought resilience may facilitate marker-assisted selection in breeding pipelines.
Fifth, multi-omics approaches including epitranscriptomics (e.g., m6A-seq), transcriptomics (RNA-seq), proteomics, metabolomics, and chromatin accessibility assays (ATAC-seq or ChIP-seq) can be integrated to achieve a systems-level understanding of plant drought responses. By correlating m6A modifications with changes in transcript abundance, translation efficiency (assessed via ribosome profiling), and protein or metabolite levels, researchers can identify key regulatory nodes that coordinate stress adaptation. However, the integration of these diverse data types presents significant methodological and analytical challenges, such as ensuring high spatial and temporal resolution, effectively normalizing heterogeneous datasets, and applying advanced machine-learning-based network inference models to unravel complex regulatory interactions.

6. Conclusions

RNA methylation, especially m6A, adds a crucial regulatory layer to plant stress responses, with emerging evidence pointing to its role in modulating drought tolerance in rice. While the current understanding remains preliminary and largely correlative, advancing technologies such as high-resolution methylome mapping and functional validation of m6A regulators will be key to unlocking its full potential. As research progresses, epitranscriptomic regulation may offer innovative strategies for breeding drought-resilient rice varieties, supporting sustainable agriculture in the face of climate change.

Author Contributions

Conceptualization, M.N. and X.F.; methodology, M.N.; software, M.N.; validation, Y.Z., P.G., and X.F.; formal analysis, M.N.; investigation, X.F.; resources, M.N.; data curation, M.N.; writing—original draft preparation, M.N.; writing—review and editing, X.F., Y.Z., and P.G.; visualization, M.N.; supervision, M.N.; project administration, M.N.; funding acquisition, X.F. and Y.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Liaoning Provincial Department of Science and Technology Doctoral Research Initiation Fund Program (2023-BS-209 and 2024-BS-281); Wuxi’s “Taihu Light” Scientific and Technological Innovation Initiative (K20241057); Natural science foundation of Nantong (JC2024019).

Data Availability Statement

All data generated or analyzed during this study are included in this published article.

Acknowledgments

The author sincerely thanks the anonymous editor and reviewers for their insightful comments and constructive suggestions, which significantly improved the clarity and quality of this manuscript. Their expertise and critical feedback were instrumental in refining the scientific content and presentation. The author also acknowledges the valuable support and guidance from colleagues and mentors during the preparation of this work.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

m6AN6-Methyladenosine
ABAAbscisic Acid
MeRIP-seqMethylated RNA Immunoprecipitation Sequencing
miCLIPMethylation Individual-Nucleotide-Resolution Crosslinking and Immunoprecipitation
YTHYT521-B Homology Domain
METTL3Methyltransferase-Like 3
METTL14Methyltransferase-Like 14
3′UTR3′ Untranslated Region
FIP37FKBP12 Interacting Protein 37 (WTAP homolog in plants)
TFTranscription Factor

References

  1. Birla, D.S.; Malik, K.; Sainger, M.; Chaudhary, D.; Jaiwal, R.; Jaiwal, P.K. Progress and challenges in improving the nutritional quality of rice (Oryza sativa L.). Crit. Rev. Food Sci. Nutr. 2017, 57, 2455–2481. [Google Scholar] [CrossRef] [PubMed]
  2. Mohidem, N.A.; Hashim, N.; Shamsudin, R.; Che Man, H. Rice for food security: Revisiting its production, diversity, rice milling process and nutrient content. Agriculture 2022, 12, 741. [Google Scholar] [CrossRef]
  3. Kwon, Y.; Kabange, N.R.; Lee, J.-Y.; Seo, B.Y.; Shin, D.; Lee, S.-M.; Cha, J.-K.; Cho, J.-H.; Kang, J.-W.; Park, D.-S. RNA-Seq and electrical penetration graph revealed the role of Grh1-mediated activation of defense mechanisms towards green rice leafhopper (Nephotettix cincticeps Uhler) resistance in rice (Oryza sativa L.). Int. J. Mol. Sci. 2021, 22, 10696. [Google Scholar] [CrossRef] [PubMed]
  4. Yin, M.; Wang, S.; Wang, Y.; Wei, R.; Liang, Y.; Zuo, L.; Huo, M.; Huang, Z.; Lang, J.; Zhao, X. Impact of abiotic stress on rice and the role of DNA methylation in stress response mechanisms. Plants 2024, 13, 2700. [Google Scholar] [CrossRef]
  5. Edwards, G.I.; Nelson, K.M.; Le Clec’h, S.; Luu, T.; Coast, O.; Futakuchi, K.; Kok, K. Twenty-five rice research priorities for sustainable rice systems by 2050. Glob. Sustain. 2024, 7, e23. [Google Scholar] [CrossRef]
  6. Ramakrishnan, M.; Satish, L.; Kalendar, R.; Narayanan, M.; Kandasamy, S.; Sharma, A.; Emamverdian, A.; Wei, Q.; Zhou, M. The dynamism of transposon methylation for plant development and stress adaptation. Int. J. Mol. Sci. 2021, 22, 11387. [Google Scholar] [CrossRef]
  7. Cai, J.; Shen, L.; Kang, H.; Xu, T. RNA modifications in plant adaptation to abiotic stresses. Plant Commun. 2025, 6, 101229. [Google Scholar] [CrossRef]
  8. Su, T.; Chen, J.; Huo, X.; Kuang, L.; Yan, T.; Gao, F.; Wu, D. Transcriptome-wide m6A methylation and metabolomic analysis reveal regulatory networks in rice roots under manganese stress. Environ. Exp. Bot. 2024, 226, 105906. [Google Scholar] [CrossRef]
  9. Tang, L.; Risalat, H.; Cao, R.; Hu, Q.; Pan, X.; Hu, Y.; Zhang, G. Food security in China: A brief view of rice production in recent 20 years. Foods 2022, 11, 3324. [Google Scholar] [CrossRef]
  10. Barbosa Amorim, L.L.; Bezerra-Neto, J.P.; da Fonseca do Santos, R.; Ferreira Neto, J.R.C.; Kido, E.A.; Matos, M.; Benko-Iseppon, A.M. Transcription Factors Involved in Plant Drought Tolerance Regulation. In Drought Stress Tolerance in Plants, Vol 2: Molecular and Genetic Perspectives; Hossain, M.A., Wani, S.H., Bhattacharjee, S., Burritt, D.J., Tran, L.-S.P., Eds.; Springer International Publishing: Cham, Switzerland, 2016; pp. 315–358. [Google Scholar]
  11. Yue, Y.; Liu, J.; He, C. RNA N6-methyladenosine methylation in post-transcriptional gene expression regulation. Genes Dev. 2015, 29, 1343–1355. [Google Scholar] [CrossRef]
  12. Hou, N.; Li, C. MdMTA-mediated m(6) A modification enhances drought tolerance by promoting mRNA stability and translation efficiency of genes involved in lignin deposition and oxidative stress. New Phytol. 2022, 234, 1294–1314. [Google Scholar] [CrossRef] [PubMed]
  13. Tang, J.; Lei, D.; Yang, J.; Chen, S.; Wang, X.; Huang, X.; Zhang, S.; Cai, Z.; Zhu, S.; Wan, J. OsALKBH9-mediated m6A demethylation regulates tapetal PCD and pollen exine accumulation in rice. Plant Biotechnol. J. 2024, 22, 2410–2423. [Google Scholar] [CrossRef] [PubMed]
  14. Boulias, K.; Greer, E.L. Biological roles of adenine methylation in RNA. Nat. Rev. Genet. 2023, 24, 143–160. [Google Scholar] [CrossRef] [PubMed]
  15. Sreepathi, N.; Puttegowda, D.; Firdose, N.; Gangadharappa, B.S.; Chandana Kumari, V.; Ramu, R. GABA as a Signaling Molecule in Plants. In GABA in Plants: Biosynthesis, Plant Development, and Food Security; John Wiley & Sons: Hoboken, NJ, USA, 2025; pp. 65–96. [Google Scholar]
  16. Muhammad, Z.M.; Muhammad, S.; Ghulam, A.; Javaid, A.; Zia, U.-Q. Drought stress impairs grain yield and quality of rice genotypes by impaired photosynthetic attributes and K nutrition. Rice Sci. 2020, 27, 5. [Google Scholar]
  17. Wilhelm, C.; Selmar, D. Energy dissipation is an essential mechanism to sustain the viability of plants: The physiological limits of improved photosynthesis. J. Plant Physiol. 2011, 168, 79–87. [Google Scholar] [CrossRef]
  18. Sandhu, D.; Coleman, Z.; Atkinson, T.; Rai, K.M.; Mendu, V. Genetics and physiology of the nuclearly inherited yellow foliar mutants in soybean. Front. Plant Sci. 2018, 9, 471. [Google Scholar] [CrossRef]
  19. Kerchev, P.I.; Van Breusegem, F. Improving oxidative stress resilience in plants. Plant J. 2022, 109, 359–372. [Google Scholar] [CrossRef]
  20. Ouyang, W.; Struik, P.C.; Yin, X.; Yang, J. Stomatal conductance, mesophyll conductance, and transpiration efficiency in relation to leaf anatomy in rice and wheat genotypes under drought. J. Exp. Bot. 2017, 68, 5191–5205. [Google Scholar] [CrossRef]
  21. Aslam, M.M.; Rashid, M.A.R.; Siddiqui, M.A.; Khan, M.T.; Farhat, F.; Yasmeen, S.; Khan, I.A.; Raja, S.; Rasool, F.; Sial, M.A. Recent insights into signaling responses to cope drought stress in rice. Rice Sci. 2022, 29, 105–117. [Google Scholar] [CrossRef]
  22. Liang, Z.; Riaz, A.; Chachar, S.; Ding, Y.; Du, H.; Gu, X. Epigenetic modifications of mRNA and DNA in plants. Mol. Plant 2020, 13, 14–30. [Google Scholar] [CrossRef]
  23. Růžička, K.; Zhang, M.; Campilho, A.; Bodi, Z.; Kashif, M.; Saleh, M.; Eeckhout, D.; El-Showk, S.; Li, H.; Zhong, S. 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] [PubMed]
  24. Niu, Q.; Song, Z.; Tang, K.; Chen, L.; Wang, L.; Ban, T.; Guo, Z.; Kim, C.; Zhang, H.; Duan, C.-G. A histone H3K4me1-specific binding protein is required for siRNA accumulation and DNA methylation at a subset of loci targeted by RNA-directed DNA methylation. Nat. Commun. 2021, 12, 3367. [Google Scholar] [CrossRef] [PubMed]
  25. Kuluev, B.; Mikhaylova, E.; Ermoshin, A.; Veselova, S.; Tugbaeva, A.; Gumerova, G.; Gainullina, K.; Zaikina, E. The ARGOS-LIKE genes of Arabidopsis and tobacco as targets for improving plant productivity and stress tolerance. J. Plant Physiol. 2019, 242, 153033. [Google Scholar] [CrossRef] [PubMed]
  26. Zhao, B.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]
  27. 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, 201–206. [Google Scholar] [CrossRef]
  28. Madsen, J.G.S.; Rauch, A.; Van Hauwaert, E.L.; Schmidt, S.F.; Winnefeld, M.; Mandrup, S. Integrated analysis of motif activity and gene expression changes of transcription factors. Genome Res. 2018, 28, 243–255. [Google Scholar] [CrossRef]
  29. 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. N6-Methyladenosine Inhibits Local Ribonucleolytic Cleavage to Stabilize mRNAs in Arabidopsis. Cell Rep. 2018, 25, 1146–1157.e3. [Google Scholar] [CrossRef]
  30. Hu, J.; Xu, T.; Kang, H. Crosstalk between RNA m6A modification and epigenetic factors for gene regulation in plants. Plant Commun. 2024, 5, 101037. [Google Scholar] [CrossRef]
  31. Erdmann, R.M.; Picard, C.L. RNA-directed DNA methylation. PLoS Genet. 2020, 16, e1009034. [Google Scholar] [CrossRef]
  32. Xue, F.; Zhang, J. m(6)A demethylase OsALKBH5 is required for double-strand break formation and repair by affecting mRNA stability in rice meiosis. New Phytol. 2024, 244, 2326–2342. [Google Scholar] [CrossRef]
  33. Shen, L.; Liang, Z.; Gu, X.; Chen, Y.; Teo, Z.W.N.; Hou, X.; Cai, W.M.; Dedon, P.C.; Liu, L.; Yu, H. N6-methyladenosine RNA modification regulates shoot stem cell fate in Arabidopsis. Dev. Cell 2016, 38, 186–200. [Google Scholar] [CrossRef] [PubMed]
  34. Nguyen, T.K.H.; Kang, H. Reading m6A marks in mRNA: A potent mechanism of gene regulation in plants. J. Integr. Plant Biol. 2024, 66, 2586–2599. [Google Scholar] [CrossRef] [PubMed]
  35. 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] [PubMed]
  36. Cai, Z.; Tang, Q.; Song, P.; Tian, E.; Yang, J.; Jia, G. The m6A reader ECT8 is an abiotic stress sensor that accelerates mRNA decay in Arabidopsis. Plant Cell 2024, 36, 2908–2926. [Google Scholar] [CrossRef]
  37. Ma, W.; Cui, S.; Lu, Z.; Yan, X.; Cai, L.; Lu, Y.; Cai, K.; Zhou, H.; Ma, R.; Zhou, S.; et al. YTH Domain Proteins Play an Essential Role in Rice Growth and Stress Response. Plants 2022, 11, 2206. [Google Scholar] [CrossRef]
  38. Yu, F.; Qi, H.; Gao, L.; Luo, S.; Njeri Damaris, R.; Ke, Y.; Wu, W.; Yang, P. Identifying RNA modifications by direct RNA sequencing reveals complexity of epitranscriptomic dynamics in rice. Genom. Proteom. Bioinform. 2023, 21, 788–804. [Google Scholar] [CrossRef]
  39. Li, Y.; Yin, M.; Wang, J.; Zhao, X.; Xu, J.; Wang, W.; Fu, B. Epitranscriptome profiles reveal participation of the RNA methyltransferase gene OsMTA1 in rice seed germination and salt stress response. BMC Plant Biol. 2025, 25, 115. [Google Scholar] [CrossRef]
  40. Fang, Y.; Chen, L.; Lin, K.; Feng, Y.; Zhang, P.; Pan, X.; Sanders, J.; Wu, Y.; Wang, X.-e.; Su, Z. Characterization of functional relationships of R-loops with gene transcription and epigenetic modifications in rice. Genome Res. 2019, 29, 1287–1297. [Google Scholar] [CrossRef]
  41. Satam, H.; Joshi, K. Next-Generation Sequencing Technology: Current Trends and Advancements. Biology 2023, 12, 997. [Google Scholar] [CrossRef]
  42. Tang, Y.; Gao, C.-C.; Gao, Y.; Yang, Y.; Shi, B.; Yu, J.-L.; Lyu, C.; Sun, B.-F.; Wang, H.-L.; Xu, Y.; et al. OsNSUN2-Mediated 5-Methylcytosine mRNA Modification Enhances Rice Adaptation to High Temperature. Dev. Cell 2020, 53, 272–286.e7. [Google Scholar] [CrossRef]
  43. Wang, L.; Zhuang, H.; Fan, W.; Zhang, X.; Dong, H.; Yang, H.; Cho, J. m6A RNA methylation impairs gene expression variability and reproductive thermotolerance in Arabidopsis. Genome Biol. 2022, 23, 244. [Google Scholar] [CrossRef] [PubMed]
  44. Song, P.; Cai, Z. Principles, functions, and biological implications of m(6)A in plants. RNA 2024, 30, 491–499. [Google Scholar] [CrossRef] [PubMed]
  45. Arribas-Hernández, 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]
  46. Pan, Y.; Jia, Y.; Liu, W.; Zhao, Q.; Pan, W.; Jia, Y.; Lv, S.; Liu, X.; Nie, X. Transcriptome-wide m6A methylation profile reveals its potential role underlying drought response in wheat (Triticum aestivum L.). Planta 2024, 260, 65. [Google Scholar] [CrossRef] [PubMed]
  47. Boughanem, H.; Pilo, J.; Rego, A.; Garcia-Flores, L.A.; de Vera, T.D.; Tinahones, F.J.; Martin-Nuñez, G.M.; González, M.M. A multi-omic integrative approach combining m6A-epitranscriptomic, transcriptomic, and splicing alternative events reveals potential candidates for colorectal cancer diagnosis. Genes Dis. 2025, 101537. [Google Scholar] [CrossRef]
  48. Yang, Y.; Huang, Y.; Wang, T.; Li, S.; Jiang, J.; Chen, S.; Chen, F.; Wang, L. mRNA m6A regulates gene expression via H3K4me3 shift in 5’UTR. Genome Biol. 2025, 26, 54. [Google Scholar] [CrossRef]
  49. Zheng, Y.; Liu, Y.; Yang, J.; Dong, L.; Zhang, R.; Tian, S.; Yu, Y.; Ren, L.; Hou, W.; Zhu, F.; et al. Multi-omics data integration using ratio-based quantitative profiling with Quartet reference materials. Nat. Biotechnol. 2024, 42, 1133–1149. [Google Scholar] [CrossRef]
  50. Shen, L.; Yu, H. RNA m6A modification meets plant hormones. Nat. Plants 2025, 11, 686–695. [Google Scholar] [CrossRef]
  51. Bhat SS, Bielewicz D, Gulanicz T, Bodi Z, Yu X, Anderson SJ, Szewc L, Bajczyk M, Dolata J, Grzelak N, Smolinski DJ, Gregory BD, Fray RG, Jarmolowski A, Szweykowska-Kulinska Z. mRNA adenosine methylase (MTA) deposits m6A on pri-miRNAs to modulate miRNA biogenesis in Arabidopsis thaliana. Proc. Natl. Acad. Sci. USA, 2020; 17, 21785–21795. [CrossRef]
  52. Wei, L.H.; Song, P.; Wang, Y.; Lu, Z.; Tang, Q.; Yu, Q.; Xiao, Y.; Zhang, X.; Duan, H.C.; Jia, G. The m(6)A Reader ECT2 Controls Trichome Morphology by Affecting mRNA Stability in Arabidopsis. Plant Cell 2018, 30, 968–985. [Google Scholar] [CrossRef]
  53. Quadrana, L.; Bortolini Silveira, A.; Mayhew, G.F.; LeBlanc, C.; Martienssen, R.A.; Jeddeloh, J.A.; Colot, V. The Arabidopsis thaliana mobilome and its impact at the species level. elife 2016, 5, e15716. [Google Scholar] [CrossRef]
  54. Govindan, G.; Sharma, B.; Li, Y.F.; Armstrong, C.D.; Merum, P.; Rohila, J.S.; Gregory, B.D. mRNA N(6)-methyladenosine is critical for cold tolerance in Arabidopsis. Plant J. 2022, 111, 1052–1068. [Google Scholar] [CrossRef] [PubMed]
  55. Das, S.; Shil, S.; Rime, J.; Alice, A.K.; Yumkhaibam, T.; Mounika, V.; Singh, A.P.; Kundu, M.; Lalhmangaihzuali, H.; Hazarika, T.K. Phytohormonal signaling in plant resilience: Advances and strategies for enhancing abiotic stress tolerance. Plant Growth Regul. 2025, 105, 329–360. [Google Scholar] [CrossRef]
  56. Ganguly, D.R.; Li, Y.; Bhat, S.S.; Tiwari, S.; Ng, P.J.; Gregory, B.D.; Sunkar, R. mRNA ADENOSINE METHYLASE promotes drought tolerance through N6-methyladenosine-dependent and independent impacts on mRNA regulation in Arabidopsis. New Phytol. 2025, 245, 183–199. [Google Scholar] [CrossRef] [PubMed]
  57. Fidler, J.; Graska, J.; Gietler, M. PYR/PYL/RCAR Receptors Play a Vital Role in the Abscisic-Acid-Dependent Responses of Plants to External or Internal Stimuli. Cells 2022, 11, 1352. [Google Scholar] [CrossRef]
  58. Bimpong, D.; Zhao, L.; Ran, M.; Zhao, X.; Wu, C.; Li, Z.; Wang, X.; Cheng, L.; Fang, Z.; Hu, Z. Transcriptomic analysis reveals the regulatory mechanisms of messenger RNA (mRNA) and long non-coding RNA (lncRNA) in response to waterlogging stress in rye (Secale cereale L.). BMC Plant Biol. 2024; 24, 534. [Google Scholar] [CrossRef]
  59. Yan, Y.; Liu, W.; Wei, Y.; Shi, H. MeCIPK23 interacts with Whirly transcription factors to activate abscisic acid biosynthesis and regulate drought resistance in cassava. Plant Biotechnol. J. 2020, 18, 1504. [Google Scholar] [CrossRef]
  60. Zhou, J.; Wan, J.; Shu, X.E.; Mao, Y.; Liu, X.-M.; Yuan, X.; Zhang, X.; Hess, M.E.; Brüning, J.C.; Qian, S.-B. N6-methyladenosine guides mRNA alternative translation during integrated stress response. Mol. Cell 2018, 69, 636–647.e7. [Google Scholar] [CrossRef]
  61. Wang, Y.N.; Yu, C.Y.; Jin, H.Z. RNA N(6)-Methyladenosine Modifications and the Immune Response. J. Immunol. Res. 2020, 2020, 6327614. [Google Scholar] [CrossRef]
  62. Cui, X.; Liang, Z.; Shen, L.; Zhang, Q.; Bao, S.; Geng, Y.; Zhang, B.; Leo, V.; Vardy, L.A.; Lu, T. 5-Methylcytosine RNA methylation in Arabidopsis thaliana. Mol. Plant 2017, 10, 1387–1399. [Google Scholar] [CrossRef]
  63. Engreitz, J.M.; Ollikainen, N.; Guttman, M. Long non-coding RNAs: Spatial amplifiers that control nuclear structure and gene expression. Nat. Rev. Mol. Cell Biol. 2016, 17, 756–770. [Google Scholar] [CrossRef]
  64. Wu, Q.; Bazzini, A.A. Translation and mRNA Stability Control. Annu. Rev. Biochem. 2023, 92, 227–245. [Google Scholar] [CrossRef]
  65. Hanson, G.; Alhusaini, N.; Morris, N.; Sweet, T.; Coller, J. Translation elongation and mRNA stability are coupled through the ribosomal A-site. RNA 2018, 24, 1377–1389. [Google Scholar] [CrossRef] [PubMed]
  66. Shoaib, Y.; Usman, B.; Kang, H.; Jung, K.-H. Epitranscriptomics: An additional regulatory layer in plants’ development and stress response. Plants 2022, 11, 1033. [Google Scholar] [CrossRef] [PubMed]
  67. Yang, X.; Patil, S.; Joshi, S.; Jamla, M.; Kumar, V. Exploring epitranscriptomics for crop improvement and environmental stress tolerance. Plant Physiol. Biochem. 2022, 183, 56–71. [Google Scholar] [CrossRef] [PubMed]
  68. Engel, M.; Eggert, C.; Kaplick, P.M.; Eder, M.; Röh, S.; Tietze, L.; Namendorf, C.; Arloth, J.; Weber, P.; Rex-Haffner, M. The role of m6A/m-RNA methylation in stress response regulation. Neuron 2018, 99, 389–403.e9. [Google Scholar] [CrossRef]
  69. Sun, C.; Ali, K.; Yan, K.; Fiaz, S.; Dormatey, R.; Bi, Z.; Bai, J. Exploration of epigenetics for improvement of drought and other stress resistance in crops: A review. Plants 2021, 10, 1226. [Google Scholar] [CrossRef]
  70. Nasr, A.; Copeland, N.; Munir, M. Structural Analysis of Virus Regulatory N6-Methyladenosine (m6A) Machinery of the Black Flying Fox (Pteropus alecto) and the Egyptian Fruit Bat (Rousettus aegyptiacus) Shows Evolutionary Conservation Amongst Mammals. Genes 2024, 15, 1361. [Google Scholar] [CrossRef]
  71. Fan, Y.; Sun, C.; Yan, K.; Li, P.; Hein, I.; Gilroy, E.M.; Kear, P.; Bi, Z.; Yao, P.; Liu, Z. Recent advances in studies of genomic DNA methylation and its involvement in regulating drought stress response in crops. Plants 2024, 13, 1400. [Google Scholar] [CrossRef]
  72. Wang, Y.; Huan, Q.; Li, K.; Qian, W. Single-cell transcriptome atlas of the leaf and root of rice seedlings. J. Genet. Genom. 2021, 48, 881–898. [Google Scholar] [CrossRef]
  73. Luo, W.; Tang, Y.; Li, S.; Zhang, L.; Liu, Y.; Zhang, R.; Diao, X.; Yu, J. The m6A reader SiYTH1 enhances drought tolerance by affecting the messenger RNA stability of genes related to stomatal closure and reactive oxygen species scavenging in Setaria italica. J. Integr. Plant Biol. 2023, 65, 2569–2586. [Google Scholar] [CrossRef]
  74. Wang, W.; Qin, Q.; Sun, F.; Wang, Y.; Xu, D.; Li, Z.; Fu, B. Genome-wide differences in DNA methylation changes in two contrasting rice genotypes in response to drought conditions. Front. Plant Sci. 2016, 7, 1675. [Google Scholar] [CrossRef]
  75. Hasan, M.; Nishat, Z.S.; Hasan, M.S.; Hossain, T.; Ghosh, A. Identification of m6A RNA methylation genes in Oryza sativa and expression profiling in response to different developmental and environmental stimuli. Biochem. Biophys. Rep. 2024, 38, 101677. [Google Scholar] [CrossRef] [PubMed]
  76. Lephatsi, M.M.; Meyer, V.; Piater, L.A.; Dubery, I.A.; Tugizimana, F. Plant responses to abiotic stresses and rhizobacterial biostimulants: Metabolomics and epigenetics perspectives. Metabolites 2021, 11, 457. [Google Scholar] [CrossRef] [PubMed]
  77. Li, P.; Wang, Y.; Sun, Y.; Jiang, S.; Li, J. N 6-methyladenosine RNA methylation: From regulatory mechanisms to potential clinical applications. Front. Cell Dev. Biol. 2022, 10, 1055808. [Google Scholar] [CrossRef] [PubMed]
  78. Vaschetto, L.M. DNA Methylation, Histone Modifications, and Non-coding RNA Pathways. In Epigenetics in Crop Improvement: Safeguarding Food Security in an Ever-Changing Climate; Springer: Berlin/Heidelberg, Germany, 2024; pp. 15–27. [Google Scholar]
  79. Zhang, W.; Wu, Y.; Schnable, J.C.; Zeng, Z.; Freeling, M.; Crawford, G.E.; Jiang, J. High-resolution mapping of open chromatin in the rice genome. Genome Res. 2012, 22, 151–162. [Google Scholar] [CrossRef]
  80. Zhao, L.; Wang, S.; Cao, Z.; Ouyang, W.; Zhang, Q.; Xie, L.; Zheng, R.; Guo, M.; Ma, M.; Hu, Z. Chromatin loops associated with active genes and heterochromatin shape rice genome architecture for transcriptional regulation. Nat. Commun. 2019, 10, 3640. [Google Scholar] [CrossRef]
  81. Fanata, W.I.D.; Lee, S.Y.; Lee, K.O. The unfolded protein response in plants: A fundamental adaptive cellular response to internal and external stresses. J. Proteom. 2013, 93, 356–368. [Google Scholar] [CrossRef]
  82. Yang, Y.; Hsu, P.J.; Chen, Y.-S.; Yang, Y.-G. Dynamic transcriptomic m6A decoration: Writers, erasers, readers and functions in RNA metabolism. Cell Res. 2018, 28, 616–624. [Google Scholar] [CrossRef]
  83. Jain, M. Gene regulatory networks in abiotic stress responses via single-cell sequencing and spatial technologies: Advances and opportunities. Curr. Opin. Plant Biol. 2024, 82, 102662. [Google Scholar] [CrossRef]
  84. Zhou, L.; Gao, G.; Tang, R.; Wang, W.; Wang, Y.; Tian, S. m(6) A-mediated regulation of crop development and stress responses. Plant Biotechnol. J. 2022, 20, 1447–1455. [Google Scholar] [CrossRef]
  85. Zhu, T.; Xia, C.; Yu, R.; Zhou, X.; Xu, X.; Wang, L.; Zong, Z.; Yang, J.; Liu, Y.; Ming, L. Comprehensive mapping and modelling of the rice regulome landscape unveils the regulatory architecture underlying complex traits. Nat. Commun. 2024, 15, 6562. [Google Scholar] [CrossRef]
  86. Singh, A.; Jain, D.; Pandey, J.; Yadav, M.; Bansal, K.C.; Singh, I.K. Deciphering the role of miRNA in reprogramming plant responses to drought stress. Crit. Rev. Biotechnol. 2023, 43, 613–627. [Google Scholar] [CrossRef] [PubMed]
  87. Zargar, S.M.; Mir, R.A.; Ebinezer, L.B.; Masi, A.; Hami, A.; Manzoor, M.; Salgotra, R.K.; Sofi, N.R.; Mushtaq, R.; Rohila, J.S. Physiological and multi-omics approaches for explaining drought stress tolerance and supporting sustainable production of rice. Front. Plant Sci. 2022, 12, 803603. [Google Scholar] [CrossRef] [PubMed]
  88. Nakamura, M.; Gao, Y.; Dominguez, A.A.; Qi, L.S. CRISPR technologies for precise epigenome editing. Nat. Cell Biol. 2021, 23, 11–22. [Google Scholar] [CrossRef] [PubMed]
  89. Schaefer, M.; Kapoor, U.; Jantsch, M.F. Understanding RNA modifications: The promises and technological bottlenecks of the ‘epitranscriptome’. Open Biol. 2017, 7, 170077. [Google Scholar] [CrossRef]
Figure 1. The Role of RNA methylation in epitranscriptomic regulation during rice drought stress adaptation.
Figure 1. The Role of RNA methylation in epitranscriptomic regulation during rice drought stress adaptation.
Plants 14 02002 g001
Figure 2. Overview of m6A RNA methylation machinery in plants.
Figure 2. Overview of m6A RNA methylation machinery in plants.
Plants 14 02002 g002
Figure 3. Cross-talk between m6A methylation and other regulatory pathways.
Figure 3. Cross-talk between m6A methylation and other regulatory pathways.
Plants 14 02002 g003
Table 2. Roles of m6A RNA methylation in drought adaptation pathways in plants.
Table 2. Roles of m6A RNA methylation in drought adaptation pathways in plants.
Regulatory LevelExample Targets or PathwaysImpact of m6A ModificationEvidence (Species)Identified Gap in RiceReference
Transcription factor regulationDREB2A, NAC, bZIP TFsm6A increases mRNA stability or enhances translationArabidopsis thaliana, tomatoFunctional validation of TFs under m6A control is lacking[56]
ABA signaling pathwayPYR/PYL/RCAR, PP2Cs, SnRK2sFine-tunes ABA response by modulating key mRNA levelsA. thaliana, maizeABA components under m6A control in rice not fully mapped[57]
Gene networksLEA proteins, HSPs, RD29AStabilizes stress-inducible transcriptsA. thalianaTranscriptome-wide validation in rice not yet available[58]
Epigenetic–epitranscriptomic interfaceHistone-modifying enzymes, chromatin remodelersCo-regulation of gene expression with histone marks and RNA methylationA. thalianaEpigenomic integration with m6A data is absent in rice[54]
Oxidative stress responseSOD, CAT, APX (antioxidant enzymes)Modulates ROS-scavenging enzyme transcriptsWheat, A. thalianaRole of m6A in rice oxidative stress management remains unexplored[59]
Protein translation efficiencyeIFs (eukaryotic initiation factors), ribosomal proteinsRegulates translation under stressHuman cells, A. thalianaNo direct evidence in rice under drought[60]
Circadian clock regulationCCA1, TOC1 (circadian clock genes)Alters mRNA turnover of clock-related genesA. thalianaPotential link between m6A and drought-responsive circadian shifts in rice is unknown[61]
Long non-coding RNAs (lncRNAs)Drought-responsive lncRNAsm6A marks influence lncRNA stability and functionMaize, A. thalianaNo studies on m6A-modified lncRNAs in rice drought response[62]
Alternative polyadenylationPoly(A) site selection regulatorsAffects mRNA stability and processing efficiencyMammalian cells, A. thalianaWhether m6A influences polyadenylation in rice under drought is unclear[63]
This table summarizes the core components of the m6A RNA methylation machinery identified in rice (Oryza sativa), including homologs characterized in A. thaliana, along with their putative functions and potential involvement in drought stress responses. methyltransferase enzymes that install the m6A mark, demethylases that remove it, and RNA-binding proteins that interpret it affect RNA fate. While these components have been bioinformatically or functionally inferred from A. thaliana and mammalian studies, their precise regulatory mechanisms under drought stress in rice remain largely uncharacterized and require further functional validation.
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

Fan, X.; Zhang, Y.; Gu, P.; Naz, M. Epitranscriptomic Control of Drought Tolerance in Rice: The Role of RNA Methylation. Plants 2025, 14, 2002. https://doi.org/10.3390/plants14132002

AMA Style

Fan X, Zhang Y, Gu P, Naz M. Epitranscriptomic Control of Drought Tolerance in Rice: The Role of RNA Methylation. Plants. 2025; 14(13):2002. https://doi.org/10.3390/plants14132002

Chicago/Turabian Style

Fan, Xiaoru, Yong Zhang, Pengyuan Gu, and Misbah Naz. 2025. "Epitranscriptomic Control of Drought Tolerance in Rice: The Role of RNA Methylation" Plants 14, no. 13: 2002. https://doi.org/10.3390/plants14132002

APA Style

Fan, X., Zhang, Y., Gu, P., & Naz, M. (2025). Epitranscriptomic Control of Drought Tolerance in Rice: The Role of RNA Methylation. Plants, 14(13), 2002. https://doi.org/10.3390/plants14132002

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