Abstract
Chromatin and transcription states are crucial for establishing and maintaining gene regulation and genome integrity, governed by a complex network of epigenetic modifications. Despite significant advancements, the interplay between epitranscriptomic and epigenetic modifications in transcriptional regulation and chromatin state remains elusive in plants. Here we profile a comprehensive spectrum of N6-methyladenosine (m6A) on chromatin-associated RNAs, especially retrotransposon transcripts, in Arabidopsis. We show that m6A writer subunits FIP37 and VIR mediate the methylation of retrotransposon RNAs, which are recognized by the nuclear m6A reader proteins CPSF30-L and ECT12, leading to a closed heterochromatin state and transcription suppression of m6A retrotransposon RNAs. Through monitoring nuclear RNA stability and transcription rate upon the depletion of either writer or reader, we provide evidence consistent with a model in which CPSF30-L associates with SUVH4/5/6 and ATXR5/6 and contributes to histone H3 K9 dimethylation and H3 K27 monomethylation at m6A-marked retrotransposons. Our findings elucidate a regulatory mechanism involving RNA m6A formation and recognition, coupled with histone modifications, thereby highlighting the intricate epitranscriptomic and epigenetic interplay in plant physiological and biological processes.
Similar content being viewed by others
Main
Over 170 types of RNA modifications have been identified across all kingdoms of life. N6-methyladenosine (m6A) is the most widespread and highly conserved modification in mRNAs and noncoding RNAs (ncRNAs)1. In Arabidopsis, m6A is co-transcriptionally incorporated into RNA transcripts by the MTA–MTB complex and several accessory subunits2,3,4, erased by demethylases ALKBH9B/10B5,6 and recognized by readers such as YTH-domain-containing proteins7,8,9,10,11,12,13. The m6A modification regulates many critical post-transcriptional processes in the cytoplasm and nucleus in both plants and mammals, such as mRNA stability, RNA–protein granule formation, alternative polyadenylation and the resolution of R-loops8,9,10,11,13,14. Additionally, chromatin states and transcription can be modulated by m6A methylation in critical nuclear RNAs, especially chromatin-associated regulatory RNAs, in mammalian cells15,16,17,18,19. These studies suggest that m6A modification in different RNA types plays distinctive roles in the epitranscriptomic regulation of gene expression.
Transposons are DNA molecules capable of moving from one location to another and are widespread in most eukaryotic genomes20. Retrotransposons, classified as class I transposons, proliferate through reverse transcription of RNA intermediates and comprise over 40% and 70% of the human and maize genomes, respectively21,22,23. Due to their potentially adverse effects on host genomes, most transposons are extensively repressed by epigenetic marks, including DNA methylation and histone modifications. Plants and mammals exhibit striking divergence in heterochromatin-associated histone modifications: histone H3 K9 dimethylation and H3 K27 monomethylation serve as heterochromatin marks in plants, whereas mammals primarily use H3 K9 trimethylation for heterochromatin assembly, with H3 K9 dimethylation predominantly marking euchromatic regions24,25. Recent independent studies have revealed that METTL3-mediated m6A methylation on retrotransposons orchestrates with H3 K9 trimethylation to maintain heterochromatin formation, thereby regulating retrotransposon repression and preserving embryonic stem cell identity18,26. FTO-mediated m6A demethylation on LINE1 RNA regulates its abundance and the local chromatin state during mouse oocyte and embryonic development16. Moreover, the expression of human FTO in rice demethylates m6A and promotes chromatin accessibility and transcriptional activation27, suggesting that plant endogenous m6A modification might play a role in regulating chromatin state and transcription. However, it remains unknown whether and how m6A regulates chromatin state and transcription in plants.
Here we optimized m6A antibody immunoprecipitation sequencing (m6A-Seq) for plant chromatin-associated RNA (caRNA) and profiled the methylome landscape of retrotransposons in Arabidopsis. m6A methylation on retrotransposon RNAs, deposited by m6A writers, is recognized by the nuclear m6A reader protein CPSF30-L. CPSF30-L physically interacts with histone methylation writers SUVH4/5/6 and ATXR5/6 to increase the repressive histone marks H3 K9 dimethylation and H3 K27 monomethylation in the local heterochromatin region, leading to closed chromatin accessibility and transcription suppression. Moreover, ECT12 is characterized as another nuclear m6A reader protein, which interacts with CPSF30-L and redundantly regulates retrotransposon RNA transcription and the local heterochromatic histone levels. Our results uncover a layer of epigenetic regulation in which RNA methylation couples with histone modifications to ensure silenced transcription and heterochromatin integrity in plants.
Results
Landscape of m6A retrotransposon RNAs revealed via caRNA m6A-Seq in Arabidopsis
To unveil the transcriptional spectrum of m6A-methylated RNA molecules, we optimized m6A-Seq suitable for plant caRNA, referred to as caRNA m6A-Seq (Fig. 1a). This method was developed on the basis of the efficient isolation of non-DNA-contaminated and non-ribosomal caRNAs followed by m6A immunoprecipitation (m6A-IP) (Fig. 1b and Supplementary Fig. 1a,b). We also incorporated m6A spike-in and External RNA Controls Consortium (ERCC) spike-ins to calibrate the efficiency of m6A-IP and the variability of expression levels, respectively.
a, The caRNA m6A-Seq method suitable for plant research. NGS, next-generation sequencing. b, Cell fractionation validation shown via immunoblot analysis. UDP-glucose pyrophosphorylase (UGPase) and H3 were used as cytoplasmic and nuclear protein markers, respectively. c, Ratio of reads in caRNA-Seq versus RNA-Seq libraries mapped to exons, introns, exon/intron junctions or intergenic regions. The data are presented as mean ± s.e.m. (n = 2 independent experiments). d, Distribution of m6A peaks at distinct genomic regions including 3′ UTRs, exons, introns, 5′ UTRs, retrotransposon RNAs, DNA transposon RNAs and other ncRNAs annotated from TAIR (https://www.arabidopsis.org/) in caRNA m6A-Seq and poly(A)+ m6A-Seq (m6A-Seq). m6A peaks were categorized into three groups: m6A peaks detected only in caRNA m6A-Seq, m6A peaks detected in both caRNA and m6A-Seq, and m6A peaks detected only in m6A-Seq. e, Box plot showing m6A levels of m6A-marked pre-mRNAs, transposon RNAs and other ncRNAs. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 10,438, 201 and 38 annotated pre-mRNAs, transposon RNAs and ncRNAs, respectively). P values were determined using paired two-sided Wilcoxon tests. f, Metagene plot of caRNA m6A-Seq read density over m6A retrotransposon RNAs. g, Pie chart depicting the types of m6A retrotransposon RNAs. h, Genomic visualization showing the normalized read densities of m6A methylation and input from caRNA m6A-Seq on the representative loci COPIA62 and COPIA66. Blue and grey transcripts indicate transposon and protein-coding RNAs, respectively. i, m6A-IP–qPCR validation of the m6A peaks in COPIA62 and COPIA66. The data are presented as mean ± s.e.m. (n = 3 independent experiments). The P values are from two-tailed Student’s t-tests. NS, not significant.
To compare the m6A profiles of transcriptional transcriptomes and steady-state transcriptomes, we conducted parallel extractions of caRNAs and processed mRNAs (poly(A)+ RNAs) from two biological replicates of six-day-old wild-type (WT) seedlings and performed caRNA m6A-Seq and canonical m6A-Seq. Correlation analysis of paired input and m6A-IP sets between caRNAs and poly(A)+ RNAs was conducted independently for each set (Supplementary Fig. 1c). Considering that newly transcribed transcripts contain processing intermediates, we classified the reads into exonic, intronic, exon/intron junction and intergenic categories. Notably, caRNA sequencing (caRNA-Seq) revealed a more than fivefold higher occupancy of reads in intronic, exon/intron junction and intergenic regions compared with poly(A)+ RNA (Fig. 1c and Supplementary Fig. 1d), suggesting that caRNA m6A-Seq can specifically capture nascent transcripts. On the basis of the analysis criteria (false discovery rate < 0.05), 11,466 and 11,227 m6A peaks were exclusively detected in caRNAs and poly(A)+ RNAs, respectively (Supplementary Fig. 1e and Supplementary Tables 1 and 2).
Comparing identified m6A sites between caRNAs and poly(A)+ RNAs, we observed an increased number of m6A peaks in exonic regions in caRNA m6A-Seq (Fig. 1d). This difference may be attributed to variations in transcript composition, particularly the enrichment of nascent RNAs in which RNA Polymerase II (Pol II) has not yet completed transcription of the full 3′ untranslated region (UTR), as well as insufficient m6A demethylation during transcription. Moreover, we observed that a high number of m6A peaks were enriched in transposon RNAs and other noncoding RNA regions (Fig. 1d and Supplementary Fig. 1f). We computationally interrogated the m6A levels of these three caRNA types with spike-ins for calibration. Intriguingly, the m6A levels of transposon RNAs are higher than those of pre-mRNAs and ncRNAs (Fig. 1e).
Retrotransposons and DNA transposons are the two primary classes of transposable elements. Through caRNA m6A-Seq analysis, we identified 242 m6A peaks on retrotransposon RNAs and 67 on DNA transposon RNAs (Supplementary Fig. 1g). We also observed that retrotransposons are transcribed more actively (Supplementary Fig. 1h), underscoring the preferential involvement of m6A in retrotransposon RNA regulation. Interrogating a comprehensive methylome profiling of m6A in poly(A)+ RNAs and caRNAs, we identified a total of 139 annotated m6A-modified retrotransposon RNAs (referred to as m6A retrotransposon RNAs) due to the low expression of retrotransposon RNAs (Supplementary Table 3). These m6A retrotransposon RNAs are predominantly localized at pericentromeric heterochromatin regions (Supplementary Fig. 1i). Unlike the typical accumulation of m6A in the 3′ UTR of mRNA, m6A was distributed across the entire transcripts, with particularly high enrichment at the 5′ and 3′ ends of retrotransposon transcripts (Fig. 1f and Supplementary Fig. 1j,k). Motif analysis clustering all m6A peaks in retrotransposon RNA identified the WAC (W indicates A or U) motif, consistent with the canonical RAC (R indicates A or G) motif found in mRNAs (Supplementary Fig. 1l). Additionally, m6A retrotransposon RNAs exhibited GAG or SAU (S indicates G or C) motifs (Supplementary Fig. 1l), resembling the GAAGG and GGAUU non-canonical motifs found in mammalian retrotransposon RNAs26.
Retrotransposons include long-terminal-repeat (for example, COPIA and GYPSY) and non-long-terminal-repeat retrotransposons (for example, LINE and SINE)28. We observed that COPIA, GYPSY and LINE retrotransposons harbour 108, 88 and 43 m6A peaks, corresponding to 48, 61 and 24 transcripts, respectively, whereas SINE displays fewer m6A peaks (Fig. 1g, Supplementary Fig. 1m,n and Supplementary Table 3). m6A-IP followed by quantitative PCR (m6A-IP–qPCR) using COPIA62 and COPIA66 as representative examples confirmed authentic m6A enrichment in these identified peak regions but not elsewhere (Fig. 1h,i). The high detectability of methylation on nascent retrotransposon RNAs enables us to explore the role and mechanism of m6A in transcriptional regulation.
m6A is deposited on retrotransposon RNA transcribed by Pol II and Pol V
Since m6A is deposited co-transcriptionally during RNA polymerase transcription, we first investigated which polymerase is responsible for retrotransposon transcription. Unlike mammals, which possess only Pol I, Pol II and Pol III, plants have uniquely evolved two additional RNA polymerases, Pol IV and Pol V29. Pol I is involved in rRNA transcription; Pol II transcribes mRNAs and certain ncRNAs; Pol III synthesizes 5S rRNA, tRNA and other small RNAs; Pol IV produces ~30–40-nucleotide transcripts, primarily from repetitive regions; and Pol V generates noncoding transcripts, mainly from heterochromatic regions30,31,32. We performed caRNA-Seq using six-day-old seedlings of the WT line, a line carrying the weak Pol II mutant allele nrpb2-3 (ref. 33) and the Pol V mutant nrpe1-11 (ref. 34) to examine whether the expression levels of retrotransposon RNAs are controlled by Pol II and Pol V. We found that the expression levels of certain retrotransposon RNAs, regardless of m6A modification, were decreased in both nrpb2-3 and nrpe1-11 mutants, while some expression levels were specifically reduced in nrpb2-3 or nrpe1-11 (Supplementary Fig. 2a–d and Supplementary Table 4). Our results revealed that retrotransposon transcription is finely regulated by Pol II and Pol V occupancy, with both polymerases coordinately and specifically modulating the transcription of these RNAs.
Considering the distinct RNA products transcribed by each polymerase, we analysed Pol II and Pol V occupancy levels across m6A and non-m6A retrotransposon RNAs using published data from chromatin immunoprecipitation followed by sequencing (ChIP–Seq)35,36. We observed that both Pol II and Pol V exhibit higher binding abundance on m6A retrotransposons than on non-m6A retrotransposons, which aligns with the higher expression levels of m6A retrotransposon RNAs than those of non-m6A retrotransposons (Supplementary Fig. 2e,f), indicating that m6A is deposited on highly expressed transcripts. Furthermore, to examine whether m6A is deposited on transcripts transcribed by Pol II and Pol V, we performed formaldehyde-crosslinking-assisted RNA immunoprecipitation (FA-RIP) and quantified m6A levels on Pol-II- and Pol-V-bound transcripts using Pol II antibody in six-day-old WT seedlings and for Pol V using anti-GFP beads in six-day-old NRPE1::NRPE1-GFP nrpe1-11 seedlings. The results showed that m6A was significantly enriched in Pol II- and Pol V-bound transcripts compared with the input and immunoglobulin G (IgG) control portions, providing direct evidence that m6A is deposited on both Pol-II and Pol-V-transcribed transcripts (Supplementary Fig. 2g).
The m6A modification on retrotransposon RNAs deposited by the MTA–MTB writer complex represses the expression of retrotransposon RNAs
To investigate the m6A-mediated regulatory functions of retrotransposons in Arabidopsis, we employed the subunit mutants of the m6A MTA–MTB writer complex: fip37-4 LEC1::FIP37 and vir37, which cause a nearly 90% reduction in m6A levels in poly(A)+ RNA (Supplementary Fig. 3a). We performed caRNA m6A-Seq on six-day-old fip37-4 LEC1::FIP37 and vir mutants with two biological replicates, ensuring high replicability between the replicates (Supplementary Fig. 3b and Supplementary Tables 5 and 6). Compared with WT plants, the disruption of m6A writer function in either fip37-4 LEC1::FIP37 or vir globally reduced total m6A levels and m6A peaks in pre-mRNAs, retrotransposon RNAs and other ncRNAs (Fig. 2a and Supplementary Fig. 3c–e), suggesting that the MTA–MTB writer complex is responsible for m6A installation on these RNAs. Almost all m6A modifications on retrotransposon RNAs were dramatically decreased in both fip37-4 LEC1::FIP37 and vir (Supplementary Fig. 3f). Furthermore, expression level analysis revealed that over 80% of m6A retrotransposon RNAs were upregulated in fip37-4 LEC1::FIP37 and vir (Supplementary Fig. 3g). In contrast to the minimal changes in m6A DNA transposon RNA expression, the abundance of m6A-modified retrotransposon RNAs, but not non-m6A ones, was significantly increased upon the depletion of either FIP37 or VIR (Fig. 2b,c, Supplementary Fig. 3h–k and Supplementary Table 7). To confirm the reliability of m6A-writer-mediated regulation of retrotransposon RNAs, we independently conducted caRNA-Seq on six-day-old WT, fip37-4 LEC1::FIP37 and vir seedlings with two biological replicates, which showed consistent results (Supplementary Fig. 3l–n).
a, Box plot showing the m6A levels of retrotransposon RNAs in WT, fip37-4 LEC1::FIP37 and vir seedlings. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 139 m6A retrotransposon RNAs). P values were determined using paired two-sided Wilcoxon tests. b, Box plot showing the expression levels of m6A retrotransposon RNAs in WT, fip37-4 LEC1::FIP37 and vir seedlings. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 139 m6A retrotransposon RNAs). P values were determined using paired two-sided Wilcoxon tests. c, Volcano plots showing expression level changes of m6A retrotransposon RNAs upon either FIP37 (left) or VIR (right) depletion. Light green, dark green, blue and purple denote significantly dysregulated m6A retrotransposon RNAs belonging to COPIA, GYPSY, LINE and SINE, respectively. Grey denotes non-differentially expressed m6A retrotransposon RNAs. FC, fold change. P values were calculated via a two-sided negative binomial generalized linear model from the count data. The threshold is log2FC > 0.58 with P < 0.05. d, Heat map showing changes in relative m6A and expression levels of the indicated m6A retrotransposon subfamilies in WT, fip37-4 LEC1::FIP37 and vir seedlings. e, Scatter plots showing the positive correlation between decreased m6A levels and expression change of m6A retrotransposon RNAs upon either FIP37 (top) or VIR (bottom) depletion. The light-blue region indicates the 95% confidence interval. PCC, Pearson’s correlation coefficient. f, Genomic visualization showing the normalized read densities of m6A methylation and input from the WT, fip37-4 LEC1::FIP37 and vir on the representative loci COPIA22 and ATRE1. Blue and grey transcripts indicate transposon and protein-coding RNAs, respectively. g, m6A-IP–qPCR analysis of m6A levels on the COPIA22 and ATRE1 loci in the indicated 6-day-old seedlings. m6A levels are shown relative to those in the WT. The data are presented as mean ± s.e.m. (n = 3 independent experiments). The P values are from two-tailed Student’s t-tests. h, Relative expression levels of COPIA22 and ATRE1 in the caRNAs of the indicated 6-day-old seedlings. Expression levels are shown relative to those in the WT. ACTIN2 is used as the internal control gene. The data are shown as mean ± s.e.m. (n = 4 independent experiments). The P values are from two-tailed Student’s t-tests.
We further examined the correlation between m6A and expression levels across the classified types of retrotransposon RNAs. Retrotransposon RNAs in the COPIA, GYPSY and LINE families, as well as their transcriptionally active subfamilies, displayed lower m6A methylation levels and higher expression levels in either fip37-4 LEC1::FIP37 or vir than in WT plants (Fig. 2d). The elevated expression levels of these retrotransposon RNAs were positively correlated with the decreased m6A methylation level caused by either FIP37 or VIR depletion (Fig. 2e). Moreover, m6A-IP–qPCR and qPCR assays were performed to validate the m6A-mediated negative regulation of COPIA22 and ATRE1 transcripts (Fig. 2f–h). Taken together, these results suggest that the MTA–MTB writer complex is required for retrotransposon m6A deposition, and m6A methylation suppresses retrotransposon RNA abundance.
CPSF30-L binds and suppresses the expression of m6A retrotransposon RNAs
The mechanism by which m6A guides and decides RNA fate is determined by its reader proteins38,39. In Arabidopsis, CPSF30-L has been characterized as the nuclear m6A reader9,10. Its co-expression patterns with FIP37 and VIR, along with its nuclear localization, suggest a potential role in binding m6A retrotransposon RNAs (Supplementary Fig. 4a). To determine whether the m6A-binding function of CPSF30-L regulates the expression of these RNAs, we performed two biological replicates of caRNA-Seq using six-day-old seedlings from WT, cpsf30-l and two complementation lines respectively expressing WT CPSF30-L (CPSF30::CPSF30-L cpsf30-l, termed CPSF30-L) and the CPSF30-L(W259A/W310A) mutant with disrupted m6A-binding capability (CPSF30::CPSF30-Lm cpsf30-l, termed CPSF30-Lm) in the cpsf30-l background10. The pairwise correlation between the WT and CPSF30-L, as well as between cpsf30-l and CPSF30-Lm, showed high reproducibility (Supplementary Fig. 4b). In line with the results obtained when depleting m6A methyltransferase subunits (Fig. 2b,c), we observed a substantial increase in the abundance of m6A retrotransposon RNAs and typical retrotransposon subfamilies in the cpsf30-l mutant (Fig. 3a–c). Importantly, the alterations observed in cpsf30-l were rescued by expressing CPSF30-L but not the m6A-binding-function-abolished CPSF30-Lm (Fig. 3a–c, Supplementary Fig. 4c,d and Supplementary Table 8). We also compared the expression levels of m6A and non-m6A retrotransposon RNAs, revealing that the expression levels of m6A-modified transcripts, but not non-m6A RNAs, were significantly higher in both cpsf30-l and CPSF30-Lm than in WT and CPSF30-L plants (Supplementary Fig. 4e). Furthermore, the increased expression levels of m6A retrotransposon RNAs in cpsf30-l and CPSF30-Lm plants exhibited a positive correlation with their m6A methylation levels (Fig. 3d). These results suggest that the m6A-binding function of CPSF30-L suppresses the expression levels of m6A retrotransposon RNAs.
a, Box plot showing the expression level of m6A retrotransposon RNAs in WT, cpsf30-l, CPSF30-L and CPSF30-Lm seedlings. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 139 m6A retrotransposon RNAs). P values were determined using paired two-sided Wilcoxon tests. b, Volcano plots illustrating the expression level changes of m6A-marked retrotransposon RNAs in the paired comparisons of cpsf30-l/WT and CPSF30-Lm/CPSF30-L. Light green, dark green, blue and purple denote significantly dysregulated m6A retrotransposon RNAs belonging to COPIA, GYPSY, LINE and SINE, respectively. Grey denotes non-differentially expressed m6A retrotransposon RNAs. P values were calculated using a two-sided negative binomial generalized linear model from the count data. The threshold is log2FC > 0.58 with P < 0.05. c, Heat map showing changes of relative expression levels of the indicated m6A retrotransposon subfamilies in WT, cpsf30-l, CPSF30-L and CPSF30-Lm seedlings. d, Scatter plots showing the positive correlation between m6A level and expression change of m6A retrotransposon RNAs in the paired comparisons of cpsf30-l/WT and CPSF30-Lm/CPSF30-L. The light-blue region indicates the 95% confidence interval. e, Box plot showing the CPSF30-L binding levels of m6A and non-m6A retrotransposon RNAs. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 57 detected m6A retrotransposon RNAs and 373 detected non-m6A retrotransposon RNAs). The P value was determined using a two-sided Wilcoxon test. f, Genomic visualization showing the normalized read densities of m6A modifications, CPSF30-L binding levels and caRNA-Seq from the WT, cpsf30-l, CPSF30-L and CPSF30-Lm on the representative loci COPIA22 and ATRE1. g, FA-RIP-qPCR showing the binding affinity of CPSF30-L to the COPIA22 and ATRE1 loci in 6-day-old CPSF30-L and CPSF30-Lm seedlings. AT2G07689 was used as the internal control gene. The data are shown as mean ± s.e.m. (n = 4 independent experiments). The P values are from two-tailed Student’s t-tests. h, Relative expression levels of the COPIA22 and ATRE1 loci in the caRNAs of the indicated 6-day-old seedlings. Expression levels are shown relative to those in the WT. ACTIN2 was used as the internal control gene. The data are shown as mean ± s.e.m. (n = 4 independent experiments). The P values are from two-tailed Student’s t-tests. i, Venn diagram of upregulated m6A retrotransposon RNAs after loss of function of CPSF30-L or depletion of m6A writers.
Given the nuclear subcellular localization of CPSF30-L, we exploited nuclear enrichment in combination with formaldehyde cross-linking and immunoprecipitation (nuclear FA-CLIP) in six-day-old CPSF30-L and CPSF30-Lm seedlings (Supplementary Fig. 4f). Compared with standard FA-CLIP, nuclear FA-CLIP demonstrated more effective enrichment of m6A-modified RNA in the CPSF30-L-IP fraction (Supplementary Fig. 4g). Clustering analysis of two biological replicates confirmed the reproducibility of our nuclear FA-CLIP sequencing data (Supplementary Fig. 4h). CPSF30-L displayed significantly higher binding intensity to m6A-modified retrotransposon RNAs than to non-m6A RNAs (Fig. 3e). As illustrated in nuclear FA-CLIP and caRNA-Seq, CPSF30-L specifically bound to the m6A modification on COPIA22 and ATRE1 and mediated their expression (Fig. 3f). FA-RIP-qPCR and qPCR assays further confirmed that CPSF30-L directly binds to retrotransposon RNAs and suppresses their abundance in an m6A-dependent manner (Fig. 3g,h).
We also overlapped the upregulated m6A retrotransposon RNAs in m6A writer mutants (fip37-4 LEC1::FIP37 and vir) with those in m6A reader mutants (cpsf30-l and CPSF30-Lm). The results showed that the depletion of m6A writer subunits FIP37 and VIR, and m6A reader CPSF30-L, increases the expression of the majority of m6A retrotransposon RNAs (Fig. 3i). This further supports the notion that m6A methylation on retrotransposon RNAs, recognized by CPSF30-L, contributes to suppressing the expression of these RNAs.
m6A has less effect on the stability of retrotransposon RNAs
The increased abundance of m6A retrotransposon RNAs may result from m6A-meditated transcriptional and/or post-transcriptional regulation. Given its major effect on the lifetime of chromatin-associated regulatory RNA in mammals15,19, we sought to determine whether m6A could destabilize retrotransposon RNAs. We performed nuclear RNA decay assays using six-day-old WT, vir and cpsf30-l seedlings to sequence retrotransposon RNA levels after transcription inhibition with cordycepin, and then calculated the half-life (Supplementary Tables 9 and 10). The half-life of m6A retrotransposon RNAs was comparable to that of non-m6A retrotransposon RNAs in both vir and cpsf30-l (Fig. 4a and Supplementary Fig. 5a). We also conducted nuclear RNA decay assays and measured the half-life of COPIA22 following transcriptional inhibition with either cordycepin or actinomycin D. The results showed that neither m6A removal nor the loss of m6A recognition affected the half-life of COPIA22 (Fig. 4b and Supplementary Fig. 5b). Given that the mammalian m6A reader YTHDC1 associates with components of the nuclear exosome targeting complex and is involved in retrotransposon RNA degradation15, we performed yeast two-hybrid (Y2H) assays and found that CPSF30-L does not directly interact with the nuclear exosome targeting components ZCCHC8A, ZCCHC8B and RBM7 (Supplementary Fig. 5c). These results suggest that m6A on retrotransposon RNAs barely affects their stability and probably functions at the chromatin level.
a, Box plots showing the half-life ratio (vir/WT and cpsf30-l/WT) of m6A and non-m6A retrotransposon RNAs. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 81 detected m6A retrotransposon RNAs and 189 detected non-m6A retrotransposon RNAs for vir/WT; n = 109 detected m6A retrotransposon RNAs and 194 detected non-m6A retrotransposon RNAs for cpsf30-l/WT). P values were determined using two-sided Wilcoxon tests. b, RT-qPCR showing the lifetime of COPIA22 in the indicated seedlings treated with cordycepin at different time points. Relative RNA levels (normalized to spike-ins) are normalized to t = 0. t1/2, half-life. The data are shown as mean ± s.e.m. (n = 3 independent experiments). c, Density plot and box plot showing the transcription rate difference between cpsf30-l and WT of m6A and non-m6A retrotransposon RNAs. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 25 detected m6A retrotransposon RNAs and 699 detected non-m6A retrotransposon RNAs). The P value was determined using a two-sided Wilcoxon test. d,e, Nuclear run-on assay showing that the transcription of COPIA22 and ATRE1 increases under either the depletion of m6A writers (d) or the loss of function of an m6A reader (e). Transcript accumulation levels are shown relative to that in the WT. ACTIN2 was used as a negative control. The data are shown as mean ± s.e.m. (n = 3 independent experiments). The P values are from two-tailed Student’s t-tests.
m6A silences the transcription of retrotransposon RNAs
The m6A modification has been shown to affect chromatin-based transcriptional regulation15,16,18. To explore this further, we performed time-course RNA sequencing of nascent transcripts in six-day-old WT and cpsf30-l seedlings (Supplementary Table 11). Global analysis revealed that the transcription rates of m6A retrotransposon RNAs were significantly upregulated in the cpsf30-l mutant relative to non-m6A retrotransposon RNAs (Fig. 4c). Additionally, nuclear run-on assays in six-day-old WT, fip37-4 LEC1::FIP37 and vir seedlings showed that the loss of m6A caused a significant upregulation in the transcription rate of retrotransposon RNAs, such as COPIA22 and ATRE1 (Fig. 4d). The increased transcription rates of COPIA22 and ATRE1 were consistently observed in the cpsf30-l mutant, which was restored upon expressing CPSF30-L but not CPSF30-Lm (Fig. 4e). Together, these findings suggest that m6A recognized by CPSF30-L suppresses the transcriptional activity of m6A-modified retrotransposon RNAs in Arabidopsis.
m6A on retrotransposon RNAs maintains local closed heterochromatin by enhancing the histone repressive modifications H3 K9 dimethylation and H3 K27 monomethylation
Transcriptional regulation is a complex and dynamic process in which histone modifications are a key component40. We first investigated the association of m6A retrotransposon RNAs with heterochromatin and euchromatin histone modifications41,42. The calculated overlapping ratios (Jaccard statistics) showed that most m6A retrotransposon RNAs were associated with two repressive heterochromatin modifications: dimethylation at H3 K9 and monomethylation at H3 K27, with rare associations with other repressive marks or euchromatin modifications (Supplementary Fig. 6a). Moreover, both dimethylated H3 K9 (H3K9me2) and monomethylated H3 K27 (H3K27me1) were highly enriched at the m6A position on m6A-modified retrotransposons (Fig. 5a). The methylation levels of H3 K9 and H3 K27 are significantly higher at the loci of m6A retrotransposon RNAs than at non-m6A ones (Supplementary Fig. 6b). The average m6A peak densities are positively correlated with H3K9me2 and H3K27me1 levels on m6A retrotransposon RNAs (Fig. 5b). These observations suggest that m6A may play a role in heterochromatin in Arabidopsis. Immunostaining H3K9me2 and H3K27me1 in isolated nuclei from six-day-old seedlings revealed a slight but clear reduction in their levels in fip37-4 LEC1::FIP37, vir and cpsf30-l (Fig. 5c,d). The reduced H3K9me2 and H3K27me1 signals in cpsf30-l could be restored by expressing CPSF30-L but not the m6A-binding-function-abolished CPSF30-Lm (Fig. 5c,d), indicating that the m6A binding function of CPSF30-L regulates H3K9me2 and H3K27me1 levels.
a, Read-count tag density pile-ups of m6A levels, H3K9me2 levels and H3K27me1 levels on m6A retrotransposon RNAs. b, Scatter plots illustrating the positive correlation between m6A levels and H3K9me2, as well as H3K27me1, levels in m6A retrotransposon RNAs. The light-blue region indicates the 95% confidence interval. c, H3K9me2 and H3K27me1 immunostaining from 6-day-old WT, fip37-4 LEC1::FIP37, vir, cpsf30-l, CPSF30-L and CPSF30-Lm seedlings. Scale bars, 5 μm. The images are representative of three independent experiments. d, Quantification of H3K9me2 and H3K27me1 immunostaining in c; the intensity is normalized to the nuclear area. The data are shown as mean ± s.e.m. (n = 40, 40, 40, 40, 48 and 53 nuclei for H3K9me2 immunostaining and n = 43, 40, 40, 40, 40 and 40 nuclei for H3K27me1 immunostaining for each genotype from left to right). The P values are from two-tailed Student’s t-tests. e, Read-count tag density pile-ups of H3K9me2 and H3K27me1 levels on m6A retrotransposon RNAs in WT, vir and cpsf30-l seedlings. TSS, transcription start site; TES, transcription end site. f, ChIP-qPCR analysis of H3K9me2 and H3K27me1 levels at the COPIA22 and ATRE1 loci upon the loss of either m6A writers or an m6A reader. H3K9me2 and H3K27me1 levels are shown relative to that in the WT. The data are shown as mean ± s.e.m. (n = 3 independent experiments). The P values are from two-tailed Student’s t-tests. g, Read-count tag density pile-ups of H3K9me2 levels on SUVH4/5/6-dependent peaks in WT, suvh4 suvh5 suvh6, vir and cpsf30-l seedlings, and H3K27me1 levels on ATXR5/6-dependent peaks in WT, atxr5 atxr6, vir and cpsf30-l seedlings. There were 3,897 SUVH4/5/6-dependent peaks and 8,961 ATXR5/6-dependent peaks. h, Y2H assay showing the interaction of CPSF30-L with SUVH4, SUVH5, SUVH6, ATXR5 and ATXR6 in yeast cells. The images are representative of two independent experiments. i, BiFC assay showing the physical interaction between CPSF30-L and SUVH4, SUVH5, SUVH6, ATXR5 and ATXR6 in N. benthamiana leaf cells. Scale bars, 5 μm. The images are representative of three independent experiments. j, Semi-in vivo pull-down assay showing the interaction of CPSF30-L with SUVH4, SUVH5, SUVH6, ATXR5 and ATXR6. The images are representative of two independent experiments.
In plants, constitutive heterochromatin forms over repetitive elements, including retrotransposons, which are heavily decorated by H3K9me2 and H3K27me1. These repressive histone modifications prominently maintain heterochromatin integrity and silence transposons in Arabidopsis24. To further investigate whether and how m6A contributes to the specialized establishment of heterochromatin states, we conducted genome-wide H3K9me2 with two biological replicates and H3K27me1 ChIP–Seq in six-day-old WT, vir and cpsf30-l seedlings (Supplementary Fig. 6c and Supplementary Tables 12 and 13). Compared with the reduced levels of H3K9me2 and H3K27me1 in their respective writer mutants, the loss of m6A writer subunit VIR or m6A reader CPSF30-L partially decreased these two histone modification levels (Supplementary Fig. 6d,e), consistent with the immunostaining results (Fig. 5c,d). Notably, H3K9me2 and H3K27me1 levels were significantly reduced on m6A-modified retrotransposons and their subfamilies (Fig. 5e and Supplementary Fig. 6f), whereas no significant differences were observed in non-m6A retrotransposons between vir and WT or cpsf30-l and WT plants (Supplementary Fig. 6g). To confirm these findings, we also performed ChIP-qPCR assays to examine the H3K9me2 and H3K27me1 levels on COPIA22 and ATRE1 in the genotypic plants. The results demonstrated that m6A removal in m6A writer mutants or the loss of m6A recognition in cpsf30-l decreases H3K9me2 and H3K27me1 levels on COPIA22 and ATRE1 (Fig. 5f and Supplementary Fig. 6h). These results thus suggest that m6A modulates H3K9me2 and H3K27me1 deposition at specific m6A-modified retrotransposon regions.
Given that both repressive histone modifications, H3K9me2 and H3K27me1, are concurrently present on transposons43, we speculated that they might synergistically contribute to the transcriptional silencing of m6A retrotransposon RNAs. To address this, we analysed ChIP–Seq data in the WT and observed substantial overlap between H3K9me2 and H3K27me1 peaks (Supplementary Fig. 6i). Over 60% of m6A retrotransposon RNAs harboured both modifications (termed H3K9me2 & H3K27me1 & m6A retrotransposon RNAs; Supplementary Fig. 6j). Strikingly, vir and cpsf30-l mutants exhibited pronounced reductions in H3K9me2 and H3K27me1 levels at H3K9me2 & H3K27me1 & m6A retrotransposon RNAs (Supplementary Fig. 6k). Moreover, lacking functions of either m6A methylation or m6A recognition leads to a greater increase in the expression of m6A retrotransposon RNAs carrying both histone modifications compared with those marked by individual histone modifications (Supplementary Fig. 6l,m).
Considering that the decrease in repressive histone modifications potentially opens the chromatin state and influences nearby gene expression44, we categorized the distance of genes to the nearest m6A retrotransposon RNAs, ranging from 0 to 1 Mb, to explore the effect of m6A regulation on neighbouring gene expression. The results showed that the expression of genes adjacent to m6A retrotransposon RNAs tends to get lower as the distance increases in both vir and cpsf30-l mutants (Supplementary Fig. 7a), accompanied by gradually increased H3K9me2 and H3K27me1 levels (Supplementary Fig. 7b,c). However, these trends remained largely unchanged for non-m6A retrotransposon RNAs (Supplementary Fig. 7d–f). These results indicate that the loss of deposition and recognition of m6A compromises local and adjacent heterochromatin integrity and transcription.
The m6A reader protein CPSF30-L associates with SUVH4/5/6 and ATXR5/6 and is required for H3K9me2/H3K27me1 levels at m6A retrotransposons
We next investigated the mechanism underlying histone modifications and heterochromatin repression mediated by m6A. Since H3K9me2 is methylated by SUVH4 (also known as KYP), SUVH5 and SUVH6 methyltransferases45,46,47,48, and H3K27me1 is methylated by ATXR5 and ATXR6 (ref. 49), we analysed publicly available ChIP–Seq data for H3K9me2 and H3K27me1 in suvh4 suvh5 suvh6 and atxr5 atxr6 mutants50,51. Our analysis revealed a substantial reduction in H3K9me2 and H3K27me1 levels at the regions of m6A-modified retrotransposons in suvh4 suvh5 suvh6 and atxr5 atxr6 (Supplementary Fig. 8a,b), confirming the roles of these methyltransferases in depositing H3K9me2 and H3K27me1 at the local regions of m6A retrotransposon RNAs. Accordingly, we classified H3K9me2 peaks into two categories: SUVH4/5/6-dependent (decreased in suvh4 suvh5 suvh6 mutants) and SUVH4/5/6-independent (unaltered in suvh4 suvh5 suvh6 mutants). Similarly, H3K27me1 peaks were classified into ATXR5/6-dependent (decreased in atxr5 atxr6 mutants) and ATXR5/6-independent (unaltered in atxr5 atxr6 mutants). The depletion of VIR or CPSF30-L partially decreases SUVH4/5/6-dependent H3K9me2 and ATXR5/6-dependent H3K27me1 levels but has little effect on SUVH4/5/6-independent or ATXR5/6-independent peaks (Fig. 5g and Supplementary Fig. 8c), indicating that m6A partially modulates H3K9me2 and H3K27me1 deposition via their respective methyltransferases.
As the deposition of H3K9me2 and H3K27me1 on retrotransposons requires heterochromatin targeting, we proceeded to investigate how m6A spatially associates with heterochromatin to facilitate specialized methylation. Although no obvious differences in the expression levels of SUVH4/5/6 and ATXR5/6 were observed across WT, fip37-4 LEC1::FIP37, vir and cpsf30-l seedlings (Supplementary Fig. 8d), we observed that the expression levels of CPSF30-L were positively correlated with those of SUVH4/5 and ATXR5/6 (Supplementary Fig. 8e). Given that the depletion of m6A writers and the m6A reader CPSF30-L reduces H3K9me2 and H3K27me1 levels and enhances the transcription of m6A-modified retrotransposons (Figs. 4c and 5e), we speculated that CPSF30-L, as an m6A reader, might be responsible for m6A-mediated transcriptional regulation through associating with SUVH4/5/6 and ATXR5/6 at specific loci for H3K9me2 and H3K27me1 deposition. To verify this hypothesis, we performed Y2H assays, revealing that CPSF30-L independently interacts with SUVH4/5/6 and ATXR5/6 (Fig. 5h). Bimolecular fluorescence complementation (BiFC) assays conducted in Nicotiana benthamiana leaves showed strong reconstituted YFP signal in the nucleus or nuclear foci among five protein pairs (Fig. 5i and Supplementary Fig. 8f). Moreover, semi-in vivo pull-down assays revealed that CPSF30-L–FLAG specifically binds glutathione S-transferase (GST)-targeted SUVH4/5/6 and ATXR5/6, but not GST alone (Fig. 5j). These interactions were not observed with m6A writers (Supplementary Fig. 8g). Altogether, our findings provide evidence consistent with CPSF30-L acting as an intermediary linking m6A to H3K9me2 and H3K27me1 deposition, thereby maintaining heterochromatin integrity in m6A-modified retrotransposon RNA regions.
ECT12, as a nuclear m6A reader, interacts with CPSF30-L and enhances the m6A-binding function
Although disturbing either the deposition or recognition of m6A impacts retrotransposon transcription, we noticed that partial m6A hypomethylation on retrotransposons in fip37-4 LEC1::FIP37 and vir mutants led to more pronounced expression increases in m6A retrotransposon RNAs than in the cpsf30-l mutant (Supplementary Fig. 9a). These imbalanced effects caused by m6A writers and an m6A reader imply that other m6A reader proteins may redundantly participate in this regulation. To identify the potential m6A reader responsible for retrotransposon transcription in Arabidopsis, we conducted phylogenetic analysis comparing the identified metazoan YTHDC1 with its plant counterparts. This analysis revealed two putative homologues of YTHDC1 in Arabidopsis: CPSF30-L and ECT12 (AT4G11970; Supplementary Fig. 9b). Unlike other YTH-containing proteins, both CPSF30-L and ECT12 are exclusively localized in the nucleus (Supplementary Fig. 9c). Moreover, the mRNA expression level pattern of ECT12 strongly correlated with that of CPSF30-L (Supplementary Fig. 9d), indicating that they might have synergistic roles in biological and molecular process.
Given that ECT12 has been identified as a potential m6A reader52, we proceeded to elucidate its m6A-binding function by performing an electrophoretic mobility shift assay with recombinant GST-tagged ECT12 (GST–ECT12) and a synthetic fluorescein-amidite-labelled RNA probe containing either m6A or A. The electrophoretic mobility shift assay results revealed that ECT12 specifically binds m6A-modified RNA but not unmethylated RNA (Supplementary Fig. 9e). Structural analysis of ECT12 identified conserved tryptophan residues at positions 94 and 144 as critical for m6A recognition in the YTH domain. These residues form a hydrophobic aromatic cage, similar to those observed in other YTH domain proteins, such as YTHDC1 and CPSF30-L (Supplementary Fig. 9f). To verify that ECT12 depends on its YTH domain to recognize m6A, we performed in vitro RIP followed by liquid chromatography–tandem mass spectrometry (LC–MS/MS) detection using the recombinant GST–ECT12, GST–ECT12/YTH and GST–ECT12/YTHm harbouring a putative non-m6A binding alteration (W144A) with isolated Arabidopsis poly(A)+ RNA. The LC–MS/MS results showed that m6A was highly enriched in the immunoprecipitated fraction of GST–ECT12 and GST–ECT12/YTH, but not in the flow-through or the bound fraction of GST–ECT12/YTHm (Supplementary Fig. 9g), suggesting that the YTH domain is responsible for m6A binding in ECT12. To further confirm that ECT12 indeed serves as an m6A-binding protein in planta, we generated ECT12::gECT12 and ECT12::gECT12m transgenic plants (Supplementary Fig. 9h), where ECT12m harbours the m6A-binding-function-abolished alteration W144A, and then performed an in vivo RIP–LC–MS/MS assay. The results showed that ECT12 pulled down significantly more m6A-modified RNAs than the control IgG-IP and ECT12m-IP (Fig. 6a). These results establish ECT12 as an m6A reader protein in Arabidopsis. To investigate the potential spatial linkage between ECT12 and CPSF30-L, we examined transgenic Arabidopsis plants (ECT12::gECT12-eGFP) and found that ECT12 is localized in the nucleus (Supplementary Fig. 9h,i). We also performed Y2H and BiFC assays, confirming that ECT12 physically interacts with CPSF30-L, but not with m6A writers (Fig. 6b,c and Supplementary Fig. 9j). Semi-in vivo pull-down assays also validated the interaction between CPSF30-L and ECT12 (Fig. 6d). To assess the regulatory role of the interaction between CPSF30-L and ECT12 in m6A-modified RNAs, we conducted an in vitro RIP assay using purified recombinant CPSF30-L and ECT12 proteins along with poly(A)+ RNAs. The results showed that CPSF30-L and ECT12 together bind significantly more m6A-modified poly(A)+ RNAs than the same amount of single CPSF30-L-IP and ECT12-IP (Fig. 6e). This indicates that the interaction between CPSF30-L and ECT12 enhances the m6A-binding ability more effectively than the individual actions of CPSF30-L or ECT12 alone.
a, In vivo nuclear FA-RIP–LC–MS/MS showing that m6A is enriched in the ECT12-IP fraction compared with IgG-IP and the ECT12m-IP portion. The data are shown as mean ± s.e.m. (n = 3 independent experiments). The P values are from two-tailed Student’s t-tests. b, Y2H assay showing the interaction of CPSF30-L with ECT12 in yeast cells. The images are representative of two independent experiments. c, BiFC assay showing the physical interaction between CPSF30-L and ECT12 in N. benthamiana leaf cells. Scale bars, 5 μm. The images are representative of three independent experiments. d, Semi-in vivo pull-down assay showing the interaction of CPSF30-L with ECT12. The images are representative of two independent experiments. e, In vitro RIP–LC–MS/MS showing m6A levels in the input, CPSF30-L-IP, ECT12-IP and CPSF30-L–ECT12-IP RNA portions. m6A levels are shown relative to those in the input. The data are shown as mean ± s.e.m. (n = 3 independent experiments). The P values are from two-tailed Student’s t-tests. f, H3K9me2 and H3K27me1 immunostaining from 6-day-old WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings. Scale bars, 5 μm. The images are representative of three independent experiments. g, Quantification of H3K9me2 immunostaining in f; the intensity is normalized to the nuclear area. The data are shown as mean ± s.e.m. (n = 30, 31, 29, 31 and 31 nuclei for H3K9me2 immunostaining and n = 24, 21, 23, 20 and 20 nuclei for H3K27me1 immunostaining for each genotype from left to right). The P values are from two-tailed Student’s t-tests. h, Read-count tag density pile-ups of H3K9me2 and H3K27me1 levels on m6A retrotransposon RNAs in WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings. i, Read-count tag density pile-ups of H3K9me2 levels on SUVH4/5/6-dependent peaks and H3K27me1 levels on SUVH4/5/6-dependent peaks in WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings. There were 3,897 SUVH4/5/6-dependent peaks and 8,961 ATXR5/6-dependent peaks.
CPSF30-L and ECT12 redundantly mediate H3K9me2 and H3K27me1 formation and suppress the transcription of m6A retrotransposon RNAs
Considering that CPSF30-L and ECT12 interact with each other as nuclear m6A reader proteins, we proceeded to assess whether they redundantly regulate heterochromatic histone modifications and transcription of m6A retrotransposon RNAs. To address this, we first characterized a homozygous transfer-DNA insertion mutant, ect12-3 (SALK_071944), which almost completely disrupts the expression of full-length ECT12 transcript (Supplementary Fig. 10a). We also generated cpsf30-l ect12 double mutants using the CRISPR–Cas9 genome editing system, incorporating two guide RNAs targeting the second exonic region of ECT12 in the cpsf30-l background (Supplementary Fig. 10a). We performed H3K9me2 and H3K27me1 immunostaining on these mutants to investigate whether ECT12 and CPSF30-L redundantly affect the methylation levels of these histone modifications. The results showed that the decrease in these two histone modification levels was less pronounced in ect12-3 than in cpsf30-l, while the cpsf30-l ect12 double mutants showed a more substantial reduction in these histone marks than either single mutant (Fig. 6f,g), consistent with genetic redundancy.
Consistent with these observations, ChIP–Seq analysis for H3K9me2 and H3K27me1 in six-day-old WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings also indicated redundant regulation of heterochromatic histone modifications (Supplementary Tables 12 and 13), particularly on m6A retrotransposon RNAs, but not on non-m6A retrotransposon RNAs (Fig. 6h and Supplementary Fig. 10b–f). These findings were corroborated by ChIP-qPCR targeting selected loci (Supplementary Fig. 10g). Consistently, the reduction in the levels of SUVH4/5/6-dependent H3K9me2 and ATXR5/6-dependent H3K27me1 in cpsf30-l was more pronounced in the cpsf30-l ect12 double mutants (Fig. 6i and Supplementary Fig. 10h). Furthermore, no significant differences in the expression levels of SUVH4/5/6 and ATXR5/6 were detected across WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings, and no physical interaction was observed between ECT12 and SUVH4/5/6 or ATXR5/6 (Supplementary Fig. 10i,j). These results confirm that CPSF30-L and ECT12 form a complex to redundantly mediate H3K9me2 and H3K27me1 deposition.
To further elucidate how CPSF30-L and ECT12 redundantly regulate the expression of m6A retrotransposon RNAs, we performed parallel caRNA-Seq on six-day-old WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings with two biological replicates (Supplementary Fig. 11a and Supplementary Table 14). Reflecting their shared involvement in regulating heterochromatic histone modifications, CPSF30-L and ECT12 collectively modulate the abundance of m6A retrotransposon RNAs and specific retrotransposon subfamilies, displaying genetic redundancy (Fig. 7a and Supplementary Fig. 11b,c). The ect12-3 and cpsf30-l mutants exhibited significant increases in m6A retrotransposon RNA expression, and cpsf30-l ect12 double mutants showed more pronounced upregulation than either single mutant (Fig. 7a and Supplementary Fig. 11b,c). Overlapping the upregulated m6A retrotransposon RNAs in cpsf30-l, ect12-3 and cpsf30-l ect12 demonstrated that virtually all of them were coordinately modulated by CPSF30-L and ECT12 (Fig. 7b). Further supporting this, we confirmed the significant elevation in the transcription and expression levels of COPIA22 and ATRE1 in cpsf30-l ect12 mutants, with a twofold to tenfold increase compared with cpsf30-l or ect12-3 mutants (Fig. 7c,d and Supplementary Fig. 11d). We also found that the changes in expression levels primarily followed a redundant trend, accompanied by a concurrent decrease in both H3K9me2 and H3K27me1 levels at H3K9me2 & H3K27me1 & m6A retrotransposon RNA regions (Supplementary Fig. 11e,f). The disruption of m6A nuclear readers led to a greater increase in the expression of m6A retrotransposon RNAs carrying both histone modifications compared with those marked by a single modification (Supplementary Fig. 11f). Altogether, these results emphasize that CPSF30-L and ECT12 redundantly regulate H3K9me2 and H3K27me1 deposition, thereby suppressing the transcription of m6A-modified retrotransposon RNAs.
a, Box plot showing the expression levels of m6A retrotransposon RNAs in WT, cpsf30-l, ect12-3 and cpsf30-l ect12 seedlings. The medians (horizontal lines), interquartile ranges (boxes), and ±1.5× the interquartile range (whiskers) of the data are shown (n = 139 m6A retrotransposon RNAs). P values were determined using paired two-sided Wilcoxon tests. b, Venn diagram of upregulated m6A retrotransposon RNAs upon the depletion of individual m6A readers and double m6A readers. c,d, Relative transcriptional (c) and expression levels (d) of COPIA22 and ATRE1 in the indicated 6-day-old seedlings. ACTIN2 is used as a negative and internal control. The data are shown relative to that in the WT. The data are shown as mean ± s.e.m. (n = 3 biological replicates). The P values are from two-tailed Student’s t-tests. e, Hypothetical model depicting the mechanism of m6A-mediated heterochromatin silencing of retrotransposons in Arabidopsis. m6A on retrotransposon RNAs, deposited by m6A writers, is recognized by CPSF30-L/ECT12 reader proteins, of which CPSF30-L recruits SUVH4/5/6 and ATXR5/6 to achieve repressive H3 K9 dimethylation and H3 K27 monomethylation, leading to closed heterochromatin and reduced transcription. Panel e created with BioRender.com.
Discussion
Our study unveils a comprehensive landscape of the RNA modification m6A on caRNAs, especially in retrotransposon transcripts in Arabidopsis, revealing their transcriptional regulation via m6A deposition and recognition with m6A writer and reader proteins. Specifically, CPSF30-L and ECT12 form a complex to enhance m6A binding affinity. CPSF30-L interacts with SUVH4/5/6 and ATXR5/6, together with ECT12, to facilitate H3K9me2 and H3K27me1 deposition at designated retrotransposons, achieving largely closed chromatin and repressed retrotransposons (Fig. 7e). These results highlight a new layer of epitranscriptomic regulation that modulates retrotransposon RNA transcription and maintains heterochromatin integrity.
It has been shown that m6A promotes the decay of a subset of chromatin-associated regulatory RNAs, such as LINE1 in the nucleus of mouse embryonic stem cells15. Unlike the regulatory mechanism in mammals, our data show that m6A modifications do not affect the stability of retrotransposon RNAs (Fig. 4a,b). Additionally, the lack of interaction between nuclear m6A readers and nuclear exosome targeting components suggests that the m6A-mediated regulatory mechanism primarily operates at the transcriptional level in plants (Supplementary Fig. 5c). This was further supported by increased transcription rates of m6A retrotransposon RNAs in m6A writer and reader mutants (Fig. 4c–e), implicating both evolutionarily conserved and divergent mechanisms of m6A in transcriptional regulation.
We discovered that m6A modifications are prevalent across various retrotransposon families and are particularly enriched at the 5′ and 3′ ends of transcripts (Fig. 1f). Disruption of the m6A writer subunit in fip37-4 LEC1::FIP37 and vir mutants significantly reduced m6A levels on retrotransposon RNAs, leading to their increased abundance (Fig. 2a,b). This inverse relationship between m6A methylation and RNA levels suggests that m6A modification is crucial for suppressing retrotransposon expression. The investigation into the m6A reader CPSF30-L revealed its essential role in binding m6A-marked retrotransposons and suppressing their expression through m6A recognition (Fig. 3).
We noticed that partial m6A hypomethylation on retrotransposons in fip37-4 LEC1::FIP37 and vir mutants led to more pronounced expression increases in m6A retrotransposon RNAs than in the cpsf30-l mutant (Supplementary Fig. 9a). This disparity in effects, attributed to the mechanisms of m6A writer and reader molecules, hints at the participation of additional m6A reader proteins in a redundant regulatory capacity. Through phylogenetic analysis and molecular assays, we identified ECT12 as a nuclear m6A reader that interacts with CPSF30-L and enhances its m6A-binding function (Fig. 6a–e). The synergistic action between CPSF30-L and ECT12 underscores the evolutionary significance of m6A-mediated regulation, where multiple m6A readers ensure robust transcriptional repression of retrotransposons.
Heterochromatic histone marks differ between mammals and plants. Although m6A has been reported to regulate H3K9me3 deposition and heterochromatin states in mouse embryonic stem cells16,18,26, it is necessary to investigate whether and how m6A regulates heterochromatin state in plants. We observed that m6A-modified retrotransposons were associated with heterochromatic marks H3K9me2 and H3K27me1 (Fig. 5a). Immunostaining results showed that the reduced H3K9me2 and H3K27me1 signals in cpsf30-l can be restored by expressing CPSF30-L but not by the m6A-binding-function-abolished CPSF30-Lm (Fig. 5c,d), indicating that the m6A binding function of CPSF30-L regulates H3K9me2 and H3K27me1 levels. In m6A writer and reader mutants, we noted a notable reduction in H3K9me2 and H3K27me1 levels at m6A retrotransposon loci (Fig. 5e). Compared with the reduced levels of H3K9me2 and H3K27me1 in their respective writer mutants (suvh4 suvh5 suvh6 and atxr5 atxr6), the loss of m6A writer subunit VIR or m6A reader CPSF30-L only partially decreased these histone modifications (Supplementary Fig. 6d,e). This suggests that m6A acts as a regulatory layer in modulating H3K9me2 and H3K27me1 deposition, specifically at m6A-modified retrotransposon RNA regions. Mechanistically, CPSF30-L interacts with SUVH4/5/6 and ATXR5/6, the enzymes responsible for H3K9me2 and H3K27me1 deposition (Fig. 5h–j). This interaction links m6A methylation with histone modification, maintaining a closed chromatin state and transcriptional repression of m6A retrotransposon RNAs.
Besides histone modification, DNA methylation serves as another gene-silencing epigenetic mark. Heterochromatic non-CG methylation and H3 K9 dimethylation are interconnected through a cyclic reinforcement loop between the writers of non-CG DNA methylation and H3 K9 dimethylation53,54. In addition, DNA methylation is guided by small RNAs that target specific genomic sequences in a process known as RNA-directed DNA methylation, where Pol IV is required for short interfering RNA (siRNA) biogenesis and Pol V provides scaffold RNAs that guide siRNA-directed DNA methylation55. ChIP–Seq and caRNA-Seq analyses reveal that retrotransposon transcription is tightly regulated by the occupancy of Pol II and Pol V (Supplementary Fig. 2), suggesting a potential role for siRNAs and the involvement of Pol IV and Pol V in retrotransposon regulation. Furthermore, recent mammalian studies found that the m6A reader FXR1 recruits DNA 5mC dioxygenase TET1 for 5mC demethylation in nearby genomic loci in normal and cancer cells, and the m6A writer METTL3–METTL14 recruits DNA methyltransferase DNMT1 to chromatin for gene-body methylation56,57, suggesting that m6A could directly regulate DNA 5mC deposition. We therefore hypothesize that m6A modification may also influence DNA methylation directly and siRNA-directed DNA methylation at retrotransposons apart from the H3K9me2-mediated DNA methylation pathway.
Retrotransposons account for over 40% of the human and mouse genomes, and their transcriptional regulation is essential for mammalian zygotic genome activation, embryonic stem cell identity and embryonic development16,18,58. Similarly, retrotransposons comprise an even more significant portion of crop plant genomes and can be transcriptionally activated by environmental challenges, promoting genetic diversity and adaptive evolutionary changes59,60. Here we found that the RNA modification m6A is installed on retrotransposon RNAs to suppress their transcription, thereby securing genome integrity. It is worth investigating whether environmental challenges affect the installation of RNA modifications on retrotransposon RNAs. Besides m6A, it is also important to explore whether other RNA modifications regulate retrotransposon RNAs in plants. Recently, the expression of FTO in rice was shown to increase field yield and biomass, accompanied by m6A demethylation on repeat RNAs, enhanced chromatin accessibility and transcriptional activation27. This suggests that epitranscriptomic regulation in transcription represents a promising new avenue for plant breeding. Understanding how endogenous m6A in plants modulates retrotransposon transcription and heterochromatin states could provide valuable insights for future crop improvement strategies.
Overall, our work demonstrates that m6A co-transcriptionally directs heterochromatic histone modifications through its reader complexes, enabling a direct flow of RNA modification information from RNA to chromatin to ensure silenced transcription and heterochromatin integrity at m6A-modified retrotransposon RNA regions. This regulatory interplay between the epitranscriptome and the epigenome provides deeper insights into m6A functions as well as the mechanisms of retrotransposon regulation and heterochromatin maintenance in plant evolution and adaptation.
Methods
Plant materials and growth conditions
The WT Arabidopsis thaliana plants used in this study were of the Col-0 ecotype. nrpe1-11, nrpb2-3, fip37-4 LEC1::FIP37, vir, cpsf30-l, CPSF30::CPSF30-L cpsf30-l (CPSF30-L) and CPSF30::CPSF30-Lm cpsf30-l (CPSF30-Lm) have been described previously3,4,10,33,34. The ect12-3 (SALK_071944) mutant lines were obtained from the Arabidopsis Biological Resource Center. Loss-of-function mutants of cpsf30-l ect12 were generated using the CRISPR–Cas9 system. Specifically, two single guide RNA sequences targeting the ECT12 gene were amplified via PCR, using the pDT1T2 vector as a template. The resulting products were purified, digested with BsaI and then ligated into the binary vector pHEE401E. Subsequently, the constructed plasmid was introduced into the cpsf30-l background through GV3101-mediated floral dipping. Positive seedlings were selected from 1/2× Murashige and Skoog plates containing 50 mg l−1 hygromycin B and confirmed via direct Sanger sequencing. All seeds of the WT and mutants were sterilized in 75% ethanol for 10 min followed by immersion in 20% bleaching solution for an additional 10 min, and immediately rinsed at least four times with sterile water. The sterilized seeds were stratified at 4 °C in darkness for three days and grown on 1/2× Murashige and Skoog nutrient agar plates. All plant germination and growth were under long-day conditions (16 h light/8 h dark at 22 °C with a light intensity of 90 to 120 μmol m−2 s−1).
RT-qPCR
Isolated total RNAs and caRNAs were reverse-transcribed into first-strand cDNA using SuperScript III (Thermo Fisher Scientific) with random primers. The transcribed cDNAs were then appropriately diluted and used as templates for PCR reactions, employing Hieff qPCR SYBR Green Master Mix (Low Rox) from Yeasen. These reaction systems were subsequently analysed on a ViiA 7 instrument (Applied Biosystems) following the provided instructions. To ensure the accuracy of the results, each independent sample comprised at least three biological replicates and two technical replicates. A complete list of the primers used is provided in Supplementary Table 15.
Extraction of caRNAs
caRNAs were extracted using a previously reported method61. Briefly, 2 g of six-day-old seedlings were ground into a fine powder using liquid nitrogen. The powdered material was transferred into a 50-ml tube containing 10 ml of ice-cold Honda buffer (0.44 M sucrose, 1.25% (w/v) Ficoll, 2.5% (w/v) dextran T40, 20 mM HEPES-KOH pH 7.4, 10 mM MgCl2, 0.5% (w/v) Triton X-100, 1 mM dithiothreitol (DTT), 1× protease inhibitor (Roche), 0.4 U μl−1 Ribolock RNase inhibitor (Thermo Fisher Scientific) and 100 ng μl−1 yeast tRNA (Invitrogen)). After homogenization via rotation, the homogenate was filtered through two layers of Miracloth (Merck). After centrifugation at 4 °C and 2,000 g for 5 min, the pellet was resuspended and washed with 5 ml of Honda buffer three or four times until the pellet appeared white. The washed pellet was resuspended in 1 ml of Honda buffer and centrifuged at 4 °C and 8,000 g for 1 min. After the supernatant was removed, the nucleus pellet was weighed. One volume of nucleus resuspension buffer (50% (v/v) glycerol, 0.5 mM EDTA, 1 mM DTT, 25 mM Tris-HCl pH 7.5, 100 mM NaCl and 0.4 U μl−1 Ribolock RNase inhibitor (Thermo Fisher Scientific)) was added, and the pellet was stirred to mix with a pipette tip. Two volumes of washing buffer (25 mM Tris-HCl pH 7.5, 300 mM NaCl, 1 M urea, 0.5 mM EDTA, 1 mM DTT, 1% (v/v) Tween-20 and 0.4 U μl−1 Ribolock RNase inhibitor (Thermo Fisher Scientific)) were then added, and the pellet was washed by pipetting up and down 30 times and centrifuged at 4 °C and 8,000 g for 1 min. This washing procedure was repeated three times, and the pellet was retained as the chromatin-associated fraction. For RNA extraction, the pellet was resuspended in 1 ml of TRIzol (Invitrogen), and the extraction procedures were performed using the Direct-zol RNA Miniprep Plus kit with DNase digestion (ZYMO).
caRNA m6A-Seq and m6A-Seq
The caRNAs were concentrated with a ZYMO RNA Clean & Concentrator-5 kit (ZYMO) and processed for rRNA depletion according to the manufacturer’s instructions (RiboPOOL). Then, 1 μl of 1:1,000 diluted m6A spike-in from an EpiMark N6-Methyladenosine Enrichment Kit (NEB) and 1:100 diluted ERCC spike-ins (Ambion) were added to 1 μg of rRNA-depleted caRNA, followed by fragmentation into 100–150 nucleotides using the Magnesium RNA Fragmentation Module (NEB). m6A-IP was performed following the manufacturer’s protocols. Library preparation was performed using a NEBNext Ultra II Directional RNA Library Prep Kit (NEB) according to the manufacturer’s protocols. Sequencing was performed on an Illumina Novaseq machine in paired-end mode with 150 bp per read (Genewiz).
Poly(A)+ RNA from six-day-old WT seedlings was applied to profile the m6A methylome. 1 μg of poly(A)+ RNA was fragmented into 100–150 nucleotides using the Magnesium RNA Fragmentation Module (NEB). m6A-IP and library preparation were performed using the EpiMark N6-Methyladenosine Enrichment Kit (NEB). Input and RNA eluted from m6A-IP were used to prepare libraries with a NEBNext Ultra II Directional RNA Library Prep Kit (NEB). Sequencing was performed on an Illumina Novaseq machine in paired-end mode with 150 bp per read (Genewiz).
m6A-IP–qPCR
For each biological replicate, 20 µg of caRNAs extracted from six-day-old Arabidopsis seedlings were supplemented with 1 µl of 1:100 diluted m6A and non-m6A spike-ins from the EpiMark N6-Methyladenosine Enrichment Kit (NEB). The RNA samples were then fragmented using the Magnesium RNA Fragmentation Module (NEB), and the resulting fragments were subjected to IP with m6A-conjugated antibodies bound to Protein G Dynabeads (Thermo Fisher Scientific), following the manufacturer’s instructions. RNA was extracted from both the IP and input samples and analysed via RT-qPCR. The spike-ins served as normalization controls for m6A level quantification. The primers used for qPCR are listed in Supplementary Table 15.
Nuclear FA-CLIP
For each biological replicate, 2 g of six-day-old seedlings were harvested and crosslinked with 1% (v/v) formaldehyde under vacuum. The crosslinking reaction was stopped after 15 min by adding glycine to a final concentration of 0.125 M, followed by an additional 5 min under vacuum. Nuclei were isolated from frozen, ground plant tissue using ice-cold Honda buffer (0.44 M sucrose, 1.25% (w/v) Ficoll, 2.5% (w/v) dextran T40, 20 mM HEPES-KOH pH 7.4, 10 mM MgCl2, 0.5% (w/v) Triton X-100, 1 mM DTT, 1× protease inhibitor (Roche) and 0.4 U μl−1 Ribolock RNase inhibitor (Thermo Fisher Scientific)) and collected via centrifugation at 2,000 g for 5 min at 4 °C. The nuclei were washed twice with Honda buffer and resuspended in 1 ml of nuclear lysis buffer (150 mM KCl, 50 mM HEPES, pH 7.5, 2 mM EDTA, 1% (v/v) NP-40, 0.5 mM DTT, 1× protease inhibitor (Roche) and 40 U ml−1 RNase inhibitor (Thermo Fisher Scientific)). Sonication was performed using a Bioruptor (Diagenode) for four cycles (low power, 30 s pulse-on, 60 s pulse-off). After centrifugation, the supernatant was treated with 1 U μl−1 RNase T1 (Thermo Fisher Scientific) at 22 °C for 10 min and incubated with Anti-Flag M2 Magnetic beads (Sigma) with rotation at 4 °C for 3 h. The beads were washed three times with low-salt washing buffer (150 mM KCl, 50 mM HEPES, pH 7.5, 0.05% (w/v) NP-40, 0.5 mM DTT and 1× protease inhibitor (Roche)), followed by RNase T1 treatment and a second incubation at 22 °C for 10 min. After three additional washes with high-salt washing buffer (500 mM KCl, 50 mM HEPES, pH 7.5, 0.05% (w/v) NP-40, 0.5 mM DTT and 1× protease inhibitor (Roche)), the RNA underwent end repair and proteinase K digestion on beads. RNA was then recovered using the RNA Clean & Concentrator-5 Kit (Zymo), and libraries were prepared using the NEBNext Small RNA Library Prep Set (NEB). Sequencing was performed on the MGISEQ-2000 platform using a paired-end model (PE100).
In vivo FA-RIP-qPCR
Six-day-old CPSF30::CPSF30-L-Flag cpsf30-l and CPSF30::CPSF30-Lm-Flag cpsf30-l seedlings were separately fixed with 1% (v/v) formaldehyde solution. The fixed plant materials were ground into powder and incubated with lysis buffer (150 mM KCl, 50 mM HEPES, pH 7.5, 2 mM EDTA, 1% (v/v) NP-40, 1× protease inhibitor (Roche) and 40 U ml−1 Ribolock RNase inhibitor (Thermo Fisher Scientific)) in a head-over-tail rotation for 30 min at 4 °C. After full lysis and centrifugation, the lysates were collected and then immunoprecipitated with Anti-Flag M2 beads (Sigma-Aldrich) or a control IgG (CST) conjugated with protein G Dynabeads (Thermo Fisher Scientific) on a rotating wheel for 2 h at 4 °C. After washing, proteinase K digestion (NEB) and ethanol precipitation, the recovered RNA fractions were reverse-transcribed into cDNA to calculate the relative enrichment fold via RT-qPCR. AT2G07689 was used as the internal control.
Nuclear RNA lifetime sequencing
Six-day-old WT and mutant seedlings were placed in 3 ml of incubation buffer (15 mM sucrose, 1 mM KCl, 1 mM PIPES, pH 6.25 and 1 mM sodium citrate) in six-well plates (Corning Costar) and rotated at 600 g for 15 min. The initial (T0) samples were promptly frozen, while the remaining samples underwent transcriptional inhibition by adding 3 ml of fresh buffer with 1 mM cordycepin (Macklin). Subsequently, the samples were subjected to three additional rounds of 1-min vacuum infiltration with 1-min decompression intervals. Samples were collected at various time points (15, 30, 60 and 120 min) after the third vacuum infiltration and frozen in liquid nitrogen. Approximately 0.2 g of seedlings per sample at each time point was used. The nuclear fraction was isolated, and total RNA was purified using Trizol (Invitrogen). Prior to purifying non-ribosomal RNA with the rRNA Depletion Kit (RiboPOOL), an equal amount of ERCC RNA spike-in control (Ambion) was added to the total nuclear RNA samples as internal controls. Library preparation was carried out using the NEBNext Ultra II Directional RNA Library Prep Kit (NEB). Sequencing was conducted on an Illumina Novaseq machine in paired-end mode with 150 bp per read (Genewiz).
Nuclear RNA lifetime measurement via qPCR
Six-day-old WT and mutant seedlings were placed in six-well plates containing 1 mM cordycepin (Macklin) or 200 μM Actinomycin D (Selleck). After three rounds of 1-min vacuum infiltration, samples were collected at 15, 30, 60 and 120 min. The nuclear fraction was isolated, and total RNA was extracted using Trizol (Invitrogen). Before RNA quantification via RT-qPCR, 1 μl of 1:100 diluted m6A and non-m6A spike-in from the EpiMark N6-Methyladenosine Enrichment Kit (NEB) was introduced. The RNA degradation rate (k) was determined by plotting Nt/N0 against time and fitting the equation Nt/N0 = e−kt, where t represents transcription inhibition time, and Nt and N0 indicate RNA quantities at time t and time 0. The RNA lifetime (t1/2) was calculated as t1/2 = (ln2)/k.
Time-course RNA sequencing for transcription rate measurement
Six-day-old WT and cpsf30-l seedlings were placed in six-well plates containing 0.5 mM 5-ethynyl uridine (Jena Bioscience) and were collected at 30 min, 60 min and 120 min. Total RNA was purified using Trizol followed by rRNA depletion; nascent rRNA-depleted RNA was captured using a Click-iT Nascent RNA Capture Kit (Invitrogen). Before constructing libraries with a NEBNext Ultra II Directional RNA Library Prep Kit (NEB), an equal amount of ERCC RNA spike-in control (Ambion) was added to each sample. Sequencing was conducted on an Illumina Novaseq machine in paired-end mode with 150 bp per read (Genewiz).
Nuclear run-on assay
Six-day-old WT and mutant seedlings were ground into a fine powder in liquid N2 and resuspended in 5 ml of pre-cooled nuclease-free Honda buffer. After filtering through two layers of Miracloth and centrifuging at 2,000 g for 10 min at 4 °C, the nuclei were washed two or three times with Honda buffer and resuspended in 50 µl of nucleus storage buffer (50 mM Tris-HCl, pH 7.8, 1 mM DTT, 20% (v/v) glycerol, 5 mM MgCl2 and 0.44 M sucrose). The run-on assay was performed in a reaction containing 10 µl of 10× transcription assay buffer (500 mM Tris-HCl, pH 7.5, 50 mM MgCl2, 1.5 M KCl, 1% (v/v) sarkosyl, 20 U ml−1 Ribolock RNase inhibitor (Thermo Fisher Scientific) and 100 mM DTT), 50 µl of nuclei in storage buffer, 5 µl of NTP mixture with BrUTP (100 mM ATP, 100 mM CTP, 100 mM GTP and 100 mM BrUTP (Sigma)) and 35 µl of H2O. The run-on reaction was performed at 30 °C for 30 min. The reaction was stopped by adding 900 µl of TRIzol reagent (Invitrogen), and RNA was extracted using the Direct-zol RNA Miniprep Plus kit (ZYMO). The purified RNA was diluted in 500 µl of incubation buffer (20 mM Tris-HCl pH 7.5, 4 mM MgCl2 and 0.2% (v/v) NP-40) and incubated with 60 μl of anti-BrdU beads (Santa Cruz) at 4 °C for 2 h. The precipitated RNA was extracted with TRIzol reagent (Invitrogen) and used for RT-qPCR analysis.
ChIP–Seq and ChIP-qPCR
The protocol for ChIP–Seq was adapted from a previously published method62. Briefly, 2 g of six-day-old WT, cpsf30-l, ect12-3, cpsf30-l ect12-1, cpsf30-l ect12-2 and vir seedlings were ground with liquid nitrogen into fine powder and immediately fixed with 1% (v/v) formaldehyde. Nuclei were extracted and mononucleosomes were generated with micrococcal nuclease (MNase, NEB). Subsequently, MNase digestion was carried out and stopped with the addition of EDTA and EGTA to a final concentration of 10 mM. After centrifugation, the supernatant was diluted for IP. IP was conducted with anti-H3K9me2 (Abcam) and anti-H3K27me1 (Millipore) antibodies. After incubation overnight, Protein G Dynabeads (Thermo Fisher Scientific) were added to collect the immunocomplexes. After washing, elution, reverse crosslinking and DNA purification, recovered DNA and input were subjected to qPCR and library preparation with the NEXTflex Rapid DNA-Seq Kit for Illumina (PerkinElmer) according to the manufacturer’s instructions and sequenced with the MGISEQ-2000 platform using a paired-end model (PE150).
Immunostaining
For immunostaining, 0.5 g of six-day-old seedlings were fixed with 4% paraformaldehyde for 20 min under vacuum. The fixed seedlings were then chopped in a petri dish containing 3 ml of ice-cold NEB1 buffer (10 mM Tris-HCl pH 9.5, 10 mM KCl, 500 mM sucrose and 0.1% Triton-X 100) to obtain a homogenate. The homogenate was filtered with a 40-μm cell strainer and centrifuged at 600 g for 3 min at 4 °C. The pellet was gently resuspended in 300 μl of NEB2 buffer (10 mM Tris-HCl, pH 9.5, 10 mM KCl, 125 mM sucrose and 0.1% Triton-X 100) and layered onto 600 μl of NEB3 buffer (10 mM Tris-HCl, pH 9.5, 10 mM KCl, 850 mM sucrose and 0.1% Triton-X 100). Nuclei were pelleted via centrifugation at maximum speed for 30 min at 4 °C, resuspended in 40 μl of NEB1 buffer and spread onto slides. The slides were air-dried, fixed with 4% paraformaldehyde for 30 min at room temperature and washed with PBST. Blocking was performed with 5% BSA for 1 h at 37 °C, followed by overnight incubation at room temperature with primary antibodies (1:100 anti-H3K27me1 (Abcam) and anti-H3K9me2 (Abcam)). After the PBST washes, the slides were incubated with secondary antibodies (1:100 Goat Anti-Mouse IgG H&L (Alexa Fluor 488, Abcam) and 1:100 Goat Anti-Rabbit IgG H&L (Alexa Fluor 555, Abcam)) for 1 h at 37 °C. Finally, the slides were washed with PBST, mounted with DAPI Fluoromount-G and imaged using a Zeiss LSM900 confocal microscope.
Y2H assay
The full-length coding sequences of ZCCHC8A, ZCCHC8B, RBM7, SUVH4, SUVH5, SUVH6, ATXR5, ATXR6, CPSF30-L and ECT12 were fused in frame with the GAL4 activation domain in pGADT7 as prey. The full-length coding sequences of CPSF30-L, ECT12, MTA, FIP37 and VIR with amino acid residues 1 to 1,470 (VIRN) and 1,456 to 2,138 (VIRC) were fused in frame with pGBKT7 as bait. Yeast strain Y2HGold cells were co-transformed with the bait and prey constructs. The empty vectors pGADT7 and pGBKT7 were co-transformed as the negative controls. Co-transformed yeast strain was spotted onto selective medium (minus His, Ade, Leu and Trp) and supplemented with the inhibitor of histidine synthesis 3-amino-1,2,4-triazole (Sigma). Images were taken after incubation for two to three days at 28 °C.
BiFC assay
The full-length coding sequences of SUVH4, SUVH5, SUVH6, ATXR5, ATXR6, CPSF30-L and ECT12 were cloned into the binary pBI121–YFPC or pBI121–YFPN vector to generate SUVH4–YFPC, SUVH5–YFPC, SUVH6–YFPC, ATXR5–YFPC, ATXR6–YFPC, ECT12–YFPC and CPSF30-L–YFPN constructs. The constructs with the indicated combinations were transiently co-expressed in N. benthamiana leaves via GV3101-mediated infiltration. The plants were grown in dark conditions for another two or three days, and the YFP fluorescence signal for each combination was detected using a confocal laser scanning microscope (ZESIS). Specifically, a 488-nm-wavelength laser was used to excite the YFP, with the emission signal collected from 500 to 550 nm.
Semi-in vivo pull-down assay
Total proteins were extracted from Arabidopsis seedlings expressing CPSF30-L–FLAG with IP buffer. 2 mg of total protein extracts were mixed with 20 μg of purified proteins infused with GST tag or GST alone. The mixture was incubated at 4 °C with gentle rotation for 2 h. After incubation, pre-washed Pierce Glutathione Magnetic Agarose Beads (Thermo Fisher Scientific) were added and incubated at 4 °C with gentle rotating for another 2 h. Proteins bound to the beads were eluted in elution buffer (125 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 mM DTT and 50 mM reduced glutathione) and were analysed via protein immunoblot with GST antibody (CST) and FLAG antibody (MCE).
Protein expression and purification
Plasmids containing GST–SUVH4, GST–SUVH5, GST–SUVH6, GST–ATXR5, GST–ATXR6, GST–ECT12, GST–ECT12/YTH, GST-ECT12/YTHm and MBP–CPSF30-L were transfected into Escherichia coli strain BL-21 Rosetta competent cells. Protein expression was induced at 18 °C with 1 mM isopropyl β-D-1-thiogalactopyranoside for 16 h. Cells were collected and resuspended in lysis buffer (10 mM Tris-HCl, pH 8.0, 500 mM NaCl, 1 mM PMSF, 3 mM DTT and 5% (v/v) glycerol), then lysed via sonication and centrifuged. The soluble proteins were first purified with either a GST-affinity column (GE Healthcare) or Amylose Resin (NEB) and eluted using the corresponding elution buffer. The crude eluted fractions were further purified on a Superdex 75 exclusion column (GE Healthcare). The purified proteins were stored in storage buffer (10 mM Tris-HCl, pH 7.5, 200 mM NaCl, 3 mM DTT and 20% (v/v) glycerol) at −80 °C.
Electrophoretic mobility shift assay
The recombinant proteins GST–ECT12 were separately diluted to a series of concentrations ranging from 0 to 2 μM in binding buffer (10 mM HEPES-KOH, pH 8.0, 50 mM KCl, 1 mM EDTA, 0.05% (v/v) Triton X-100, 5% (v/v) glycerol, 10 μg ml−1 salmon DNA, 1 mM DTT and 40 U ml−1 Ribolock RNase inhibitor (Thermo Fisher Scientific)). Fluorescein-amidite-labelled RNA probe (4 nM final concentration) and 1 μl of protein were mixed in 10 μl of binding buffer and incubated on ice for 30 min. The entire RNA–protein mixture was loaded into a Novex 4–20% TBE gel (Thermo Fisher Scientific) for electrophoresis separation at 4 °C. The gel was visualized using ChemiDoc (Bio-Rad).
In vivo nuclear FA-RIP–LC–MS/MS
Nuclear RIP was conducted following a protocol similar to that for nuclear FA-CLIP, omitting the second RNase T1 digestion. Crosslinked six-day-old seedlings of CPSF30::CPSF30-L cpsf30-l, CPSF30::CPSF30-Lm cpsf30-l, ECT12::gECT12 ect12-3 and ECT12::gECT12m ect12-3 transgenic lines were subjected to IP using FLAG M2 Magnetic Beads (Sigma) or normal rabbit IgG (CST) bound to Protein G (Invitrogen). Crosslinked six-day-old WT seedlings were subjected to RIP using anti-Pol II antibody (Abcam) or IgG (CST) as a control. For GFP-tagged material, crosslinked NRPE1::NRPE1-GFP/nrpe1 seedlings were immunoprecipitated using GFP-Trap Dynabeads (ChromoTek) or IgG (CST) as a control. After washing and RNA recovery, the m6A levels in the RNA fractions were quantified via LC–MS/MS.
In vitro RIP–LC–MS/MS
For RIP–LC–MS/MS, 0.8 μg of poly(A)+ RNA and the recombinant protein GST–ECT12 or MBP–CPSF30-L (final concentration 500 nM) were incubated in 200 μl of IPP buffer (150 mM NaCl, 0.1% (v/v) NP-40, 10 mM Tris-HCl, pH 7.5, 40 U ml−1 Ribolock RNase inhibitor (Thermo Fisher Scientific) and 0.5 mM DTT) with rotation at 4 °C for 2 h. Pre-equilibrated 10 μl of GST-affinity magnetic beads (Thermo Fisher Scientific) or amylose magnetic beads (NEB) with IPP buffer were added to each solution and incubated for an additional 2 h at 4 °C. The aqueous phase was harvested, recovered via ethanol precipitation and termed the flow-through. GST beads were washed four times with 200 μl of IPP buffer. The RNA on the beads was extracted with TRIzol reagent (Invitrogen) and saved as the recombinant protein-IP sample. LC–MS/MS was used to measure the ratio of m6A/C in input, flow-through and recombinant protein-IP samples.
Quantification analysis via LC–MS/MS
LC–MS/MS analysis was performed as follows: RNA (around 100 to 200 ng) was digested with Nucleoside Digestion Mix (NEB) in 20 μl at 37 °C for 2 h. After centrifugation at maximum speed for 30 min, the aqueous phase containing nucleosides was separated using a LC pump (Shimadzu) equipped with a ZORBAX SB-Aq column (Agilent) and analysed via MS/MS on a Triple Quad 5500 mass spectrometer (AB SCIEX) in positive ion mode with multiple reaction monitoring. Nucleosides were quantified using the following mass transitions: 282.0 to 150.1 (m6A), 244.0 to 112.0 (C). Quantification was based on a standard curve generated from pure nucleoside standards run alongside the samples. The ratios of m6A/C were calculated using the calibrated concentrations.
Data analysis for caRNA-Seq and RNA-Seq
The sequencing data from the caRNA-Seq and RNA-Seq libraries were processed by trimming adapters with cutadapt (v.4.4)63, followed by alignment to the TAIR10 reference genome via hisat2 (v.2.2.1)64 with the following parameters: rna-strandness, FR; k, 10; pen-noncansplice, 1000000. We eliminated PCR duplicates using SAMtools in conjunction with Picard (v.21.0.1; https://broadinstitute.github.io/picard/)65. To enhance the authenticity and the repeatability of the results, we eliminated the transcript components of rRNA and tRNA, as well as those belonging to chloroplasts and mitochondria. Gene expression quantification was conducted using featureCounts (v.2.0.1)66 and edgeR (v.3.36.0)67 to compute counts, counts per million and differential gene expression. Therein, external ERCC spike-ins were also used to normalize the counts of each gene by using the R package RUVseq (v.1.28.0)68. The Pearson correlation was calculated using the R package cor (v.4.3.2)69.
Data analysis for caRNA m6A-Seq and m6A-Seq
The analysis for the input group in caRNA m6A-Seq or m6A-Seq was conducted following the pipeline of RNA-Seq above. We employed cutadapt (v4.4)63 for adapter trimming and size selection, retaining fragments that were 30 nucleotides or longer. These sequences were subsequently mapped to the TAIR10 reference genome using hisat2 (v.2.2.1)64 with the default parameters. PCR duplicates were eliminated using SAMtools (v.1.13)65 and Picard (v.21.0.1; https://broadinstitute.github.io/picard/). After obtaining the BAM files avoiding the reads classified to rRNA, tRNA, chloroplasts and mitochondria, we conducted a m6A region enrichment analysis using MACS2 callpeak (v.2.2.7.1)70 with the following settings: nomodel; shift, 25; extsize, 50; slocal, 200; llocal, 500; f, BAM; q, 0.05; g, 1.3e + 8. We considered regions exhibiting a false discovery rate below 0.05 for the analysis of m6A peaks. Referring to the strategy and parameters used for m6A peak calibration with IVT RNAs71, we applied the same elimination rate criteria. If m6A peaks appeared on retrotransposon RNAs in caRNA m6A-Seq and did not overlap by more than 50% with those in m6A-Seq, we considered these peaks as belonging to retrotransposon RNAs.
The m6A level was calculated as previously described16. Specifically, to determine the m6A level for each transcript in the WT, we first calculated the CPM using edgeR (v.3.36.0)67. The m6A level was then derived using the formula (CPM_IP + 0.01)/(CPM_Input + 0.01). To account for differences in IP efficiency between WT and mutant samples, an equal amount of m6A-modified spike-in RNA from the EpiMark N6-Methyladenosine Enrichment Kit (NEB) was added to the rRNA-depleted caRNA samples of both the WT and mutants as an external control. For normalization across samples, the m6A ratio of the spike-in (r_spike-in) was calculated as (CPM_IP-spike-in + 0.01)/(CPM_Input-spike-in + 0.01). The m6A normalization factor (n.f.) for each mutant was defined as the ratio of r_spike-in in each mutant to the average r_spike-in across all WT samples. This factor corrects for differences in IP efficiency, and the final m6A level for each mutant was determined using the formula (CPM_IP + 0.01)/(CPM_Input + 0.01)/n.f. The decrease in m6A level was determined using the formula (m6A level in WT − m6A level in mutant)/(m6A level in WT).
Data analysis for nuclear RNA stability assay
The data processing procedure to generate the BAM files mirrors the procedure we described above. Subsequent to this, gene expression quantification was performed using featureCounts (v.2.0.1)66 and edgeR (v.3.36.0)67 to compute counts normalized by ERCC and CPM at each time point. Setting the expression level at the initial time to 1, we fitted all degradation curves using the equation y = exp(−A × x), where y is the relative expression level of a certain gene and x is the processing time. Subsequently, we eliminated genes with a value of A less than 0, derived from the fitting equation, from all sets of sequencing data. The half-lives of other genes were computed using the formula t1/2 = (ln2)/A.
Data analysis for time-course RNA sequencing
The procedure to generate the BAM files for each sample at different time points aligns with the pipeline we outlined above. Following this, gene expression was finished using featureCounts (v.2.0.1)66 and edgeR (v.3.36.0)67 to calculate gene counts, normalized by ERCC and CPM at each respective time point. We fit all gene expression levels using a linear model represented as y = A × x + B, where y signifies the expression level of a specific gene and x indicates the processing time. We then excluded genes with an A value less than 0, derived from the fitting equation, from all sequencing datasets. The transcription rate of a gene was represented by A.
Data analysis for ChIP–Seq
The alignment process was similar to that for other sequencing data including input groups and H3K9me2/H3K27me1 IP groups. H3K9me2/H3K27me1 peaks were identified using broad-peak mode in MACS2 (v.2.2.7.1)70 with false discovery rate < 0.05. To assess the H3K9me2/H3K27me1 levels, heat maps and profile plots were generated using deepTools (v.3.5.1)72. The enrichment levels were normalized using reads per kilobase per million mapped reads to account for the differences between the IP and input datasets.
Statistics and reproducibility
The statistical tests performed on experimental data and the sample sizes are noted in the figure legends. All data points are derived from biological replicates.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
The raw sequence data from caRNA m6A-Seq, m6A-Seq, caRNA-Seq, nuclear RNA lifetime sequencing, time-course RNA sequencing and ChIP–Seq reported in this paper have been deposited in the Genome Sequence Archive at the National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, under accession number PRJCA037507; they are publicly accessible at https://bigd.big.ac.cn/gsa. All remaining data are provided in the main paper or the supplementary materials. Source data are provided with this paper.
References
Deng, X. et al. RNA N6-methyladenosine modification in cancers: current status and perspectives. Cell Res. 28, 507–517 (2018).
Zhong, S. et al. MTA is an Arabidopsis messenger RNA adenosine methylase and interacts with a homolog of a sex-specific splicing factor. Plant Cell 20, 1278–1288 (2008).
Shen, L. et al. N6-Methyladenosine RNA modification regulates shoot stem cell fate in Arabidopsis. Dev. Cell 38, 186–200 (2016).
Růžička, K. et al. Identification of factors required for m6A mRNA methylation in Arabidopsis reveals a role for the conserved E3 ubiquitin ligase HAKAI. N. Phytol. 215, 157–172 (2017).
Martínez-Pérez, M. et al. Arabidopsis m6A demethylase activity modulates viral infection of a plant virus and the m6A abundance in its genomic RNAs. Proc. Natl Acad. Sci. USA 114, 10755–10760 (2017).
Duan, H. C. et al. ALKBH10B is an RNA N6-methyladenosine demethylase affecting Arabidopsis floral transition. Plant Cell 29, 2995–3011 (2017).
Arribas-Hernández, L. et al. An m6A–YTH module controls developmental timing and morphogenesis in Arabidopsis. Plant Cell 30, 952–967 (2018).
Wei, L. H. et al. The m6A reader ECT2 controls trichome morphology by affecting mRNA stability in Arabidopsis. Plant Cell 30, 968–985 (2018).
Hou, Y. et al. CPSF30-L-mediated recognition of mRNA m6A modification controls alternative polyadenylation of nitrate signaling-related gene transcripts in Arabidopsis. Mol. Plant 14, 688–699 (2021).
Song, P. et al. Arabidopsis N6-methyladenosine reader CPSF30-L recognizes FUE signals to control polyadenylation site choice in liquid-like nuclear bodies. Mol. Plant 14, 571–587 (2021).
Wu, X. et al. N6-Methyladenosine-mediated feedback regulation of abscisic acid perception via phase-separated ECT8 condensates in Arabidopsis. Nat. Plants 10, 469–482 (2024).
Cai, Z. et al. The m6A reader ECT8 is an abiotic stress sensor that accelerates mRNA decay in Arabidopsis. Plant Cell 36, 2908–2926 (2024).
Song, P. et al. m6A readers ECT2/ECT3/ECT4 enhance mRNA stability through direct recruitment of the poly(A) binding proteins in Arabidopsis. Genome Biol. 24, 103 (2023).
Xu, C. et al. R-loop resolution promotes co-transcriptional chromatin silencing. Nat. Commun. 12, 1790 (2021).
Liu, J. et al. N6-Methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586 (2020).
Wei, J. et al. FTO mediates LINE1 m6A demethylation and chromatin regulation in mESCs and mouse development. Science 376, 968–973 (2022).
Li, Y. et al. N6-Methyladenosine co-transcriptionally directs the demethylation of histone H3K9me2. Nat. Genet. 52, 870–877 (2020).
Liu, J. et al. The RNA m6A reader YTHDC1 silences retrotransposons and guards ES cell identity. Nature 591, 322–326 (2021).
Chelmicki, T. et al. m6A RNA methylation regulates the fate of endogenous retroviruses. Nature 591, 312–316 (2021).
Feschotte, C. Transposable elements and the evolution of regulatory networks. Nat. Rev. Genet. 9, 397–405 (2008).
Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).
Lanciano, S. & Cristofari, G. Measuring and interpreting transposable element expression. Nat. Rev. Genet. 21, 721–736 (2020).
Jiao, Y. et al. Improved maize reference genome with single-molecule technologies. Nature 546, 524–527 (2017).
Liu, C., Lu, F., Cui, X. & Cao, X. Histone methylation in higher plants. Annu. Rev. Plant Biol. 61, 395–420 (2010).
Jambhekar, A., Dhall, A. & Shi, Y. Roles and regulation of histone methylation in animal development. Nat. Rev. Mol. Cell Biol. 20, 625–641 (2019).
Xu, W. et al. METTL3 regulates heterochromatin in mouse embryonic stem cells. Nature 591, 317–321 (2021).
Yu, Q. et al. RNA demethylation increases the yield and biomass of rice and potato plants in field trials. Nat. Biotechnol. 39, 1581–1588 (2021).
Quesneville, H. Twenty years of transposable element analysis in the Arabidopsis thaliana genome. Mob. DNA 11, 28 (2020).
Ream, T. S. et al. Subunit compositions of the RNA-silencing enzymes Pol IV and Pol V reveal their origins as specialized forms of RNA polymerase II. Mol. Cell 33, 192–203 (2009).
Wierzbicki, A. T., Haag, J. R. & Pikaard, C. S. Noncoding transcription by RNA polymerase Pol IVb/Pol V mediates transcriptional silencing of overlapping and adjacent genes. Cell 135, 635–648 (2008).
Böhmdorfer, G. et al. Long non-coding RNA produced by RNA polymerase V determines boundaries of heterochromatin. eLife 5, e19092 (2016).
Zhai, J. et al. A one precursor one siRNA model for Pol IV-dependent siRNA biogenesis. Cell 163, 445–455 (2015).
Zheng, B. et al. Intergenic transcription by RNA polymerase II coordinates Pol IV and Pol V in siRNA-directed transcriptional gene silencing in Arabidopsis. Genes Dev. 23, 2850–2860 (2009).
Pontes, O. et al. The Arabidopsis chromatin-modifying nuclear siRNA pathway involves a nucleolar RNA processing center. Cell 126, 79–92 (2006).
Liu, C. et al. Arabidopsis ARGONAUTE 1 binds chromatin to promote gene transcription in response to hormones and stresses. Dev. Cell 44, 348–361.e347 (2018).
Liu, Y. et al. A conserved Pol II elongator SPT6L mediates Pol V transcription to regulate RNA-directed DNA methylation in Arabidopsis. Nat. Commun. 15, 4460 (2024).
Shen, L. Functional interdependence of N6-methyladenosine methyltransferase complex subunits in Arabidopsis. Plant Cell 35, 1901–1916 (2023).
Wang, X. et al. N6-Methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).
Wang, X. et al. N6-Methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015).
Zaib, S., Rana, N. & Khan, I. Histone modifications and their role in epigenetics of cancer. Curr. Med. Chem. 29, 2399–2411 (2022).
Luo, C., Durgin, B. G., Watanabe, N. & Lam, E. Defining the functional network of epigenetic regulators in Arabidopsis thaliana. Mol. Plant 2, 661–674 (2009).
Antunez-Sanchez, J. et al. A new role for histone demethylases in the maintenance of plant genome integrity. eLife 9, e58533 (2020).
Roudier, F. et al. Integrative epigenomic mapping defines four main chromatin states in Arabidopsis. EMBO J. 30, 1928–1938 (2011).
Allshire, R. C. & Madhani, H. D. Ten principles of heterochromatin formation and function. Nat. Rev. Mol. Cell Biol. 19, 229–244 (2018).
Du, J. et al. Mechanism of DNA methylation-directed histone methylation by KRYPTONITE. Mol. Cell 55, 495–504 (2014).
Ebbs, M. L. & Bender, J. Locus-specific control of DNA methylation by the Arabidopsis SUVH5 histone methyltransferase. Plant Cell 18, 1166–1176 (2006).
Ebbs, M. L., Bartee, L. & Bender, J. H3 lysine 9 methylation is maintained on a transcribed inverted repeat by combined action of SUVH6 and SUVH4 methyltransferases. Mol. Cell. Biol. 25, 10507–10515 (2005).
Jackson, J. P., Lindroth, A. M., Cao, X. & Jacobsen, S. E. Control of CpNpG DNA methylation by the KRYPTONITE histone H3 methyltransferase. Nature 416, 556–560 (2002).
Jacob, Y. et al. ATXR5 and ATXR6 are H3K27 monomethyltransferases required for chromatin structure and gene silencing. Nat. Struct. Mol. Biol. 16, 763–768 (2009).
Li, X. et al. Mechanistic insights into plant SUVH family H3K9 methyltransferases and their binding to context-biased non-CG DNA methylation. Proc. Natl Acad. Sci. USA 115, E8793–E8802 (2018).
Ma, Z. et al. Arabidopsis Serrate coordinates histone methyltransferases ATXR5/6 and RNA processing factor RDR6 to regulate transposon expression. Dev. Cell 45, 769–784 (2018).
Amara, U., Hu, J., Park, S. J. & Kang, H. ECT12, an YTH-domain protein, is a potential mRNA m6A reader that affects abiotic stress responses by modulating mRNA stability in Arabidopsis. Plant Physiol. Biochem. 206, 108255 (2024).
Stroud, H. et al. Non-CG methylation patterns shape the epigenetic landscape in Arabidopsis. Nat. Struct. Mol. Biol. 21, 64–72 (2014).
Law, J. A. & Jacobsen, S. E. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 11, 204–220 (2010).
Matzke, M. A. & Mosher, R. A. RNA-directed DNA methylation: an epigenetic pathway of increasing complexity. Nat. Rev. Genet. 15, 394–408 (2014).
Deng, S. et al. RNA m6A regulates transcription via DNA demethylation and chromatin accessibility. Nat. Genet. 54, 1427–1437 (2022).
Quarto, G. et al. Fine-tuning of gene expression through the Mettl3–Mettl14–Dnmt1 axis controls ESC differentiation. Cell 188, 998–1018.e1026 (2025).
Li, X. et al. LINE-1 transcription activates long-range gene expression. Nat. Genet. 56, 1494–1502 (2024).
Lisch, D. How important are transposons for plant evolution? Nat. Rev. Genet. 14, 49–61 (2013).
Ito, H. et al. An siRNA pathway prevents transgenerational retrotransposition in plants subjected to stress. Nature 472, 115–119 (2011).
Long, Y., Jia, J., Mo, W., Jin, X. & Zhai, J. FLEP-seq: simultaneous detection of RNA polymerase II position, splicing status, polyadenylation site and poly(A) tail length at genome-wide scale by single-molecule nascent RNA sequencing. Nat. Protoc. 16, 4355–4381 (2021).
Xue, M. et al. The INO80 chromatin remodeling complex promotes thermomorphogenesis by connecting H2A.Z eviction and active transcription in Arabidopsis. Mol. Plant 14, 1799–1813 (2021).
Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011).
Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).
Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).
Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).
Chen, Y., Lun, A. T. & Smyth, G. K. From reads to genes to pathways: differential expression analysis of RNA-Seq experiments using Rsubread and the edgeR quasi-likelihood pipeline. F1000Res. 5, 1438 (2016).
Risso, D., Ngai, J., Speed, T. P. & Dudoit, S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat. Biotechnol. 32, 896–902 (2014).
R Core Team. R: a language and environment for statistical computing. R package version 4.3.2. https://www.R-project.org/ (2025).
Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).
Zhang, Z. et al. Systematic calibration of epitranscriptomic maps using a synthetic modification-free RNA library. Nat. Methods 18, 1213–1222 (2021).
Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. DeepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).
Acknowledgements
We thank H. Yu (National University of Singapore), Y. Qi (Tsinghua University), B. Zheng (Fudan University) and C. Chen (Chinese Academy of Sciences) for providing the fip37-4 LEC1::FIP37, nrpe1-11, nrpb2-3 and NRPE1::NRPE1-GFP nrpe1 lines. We also thank the National Center for Protein Sciences at Peking University in Beijing, China, for assistance with sequencing, and G. Li for help with the MGI2000 experiment. This work was supported by the Biological Breeding-National Science and Technology Major Project of China (grant no. 2023ZD04073, G.J.), the National Key R&D Program of China (grant no. 2024YFF1000302, G.J.) and the National Natural Science Foundation of China (grant nos. 22225704, 22477005 and 22321005, G.J.).
Author information
Authors and Affiliations
Contributions
G.J. and P.S. conceived of the project. P.S. performed the experiments with the help of S.T., E.T., K.Y., L.L. and Z. Cai. Z. Cai, Z. Chen and P.S. analysed the sequencing data. G.J. and P.S. designed the experiments, interpreted the results and wrote the paper. All authors commented on the paper.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Plants thanks Leandro Quadrana, Jixian Zhai and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Supplementary Information
Supplementary Figs. 1–12.
Supplementary Table 1
Statistics of m6A peaks from caRNA m6A-Seq in 6-day-old WT seedlings.
Supplementary Table 2
Statistics of m6A peaks from m6A-Seq in 6-day-old WT seedlings.
Supplementary Table 3
Statistics of m6A peaks on retrotransposon RNAs in 6-day-old WT seedlings.
Supplementary Table 4
Statistics of expression levels of retrotransposon RNAs in 6-day-old WT, nrpb2-3 and nrpe1-11 seedlings.
Supplementary Table 5
Statistics of m6A peaks from caRNA m6A-Seq in 6-day-old fip37-4 LEC1::FIP37 seedlings.
Supplementary Table 6
Statistics of m6A peaks from caRNA m6A-Seq in 6-day-old vir seedlings.
Supplementary Table 7
Statistics of relative expression levels of m6A retrotransposon RNAs in 6-day-old WT, fip37-4 LEC1::FIP37 and vir seedlings.
Supplementary Table 8
Statistics of relative expression levels of m6A retrotransposon RNAs in 6-day-old WT, cpsf30-l, CPSF30-L and CPSF30-Lm seedlings.
Supplementary Table 9
Statistics of half-lives of m6A retrotransposon RNAs in 6-day-old WT and vir seedlings.
Supplementary Table 10
Statistics of half-lives of m6A retrotransposon RNAs in 6-day-old WT and cpsf30-l seedlings.
Supplementary Table 11
Statistics of transcription rates of m6A retrotransposon RNAs in 6-day-old WT and cpsf30-l seedlings.
Supplementary Table 12
Statistics of H3K9me2 levels of m6A retrotransposons in 6-day-old WT, vir, cpsf30-l, ect12-3 and cpsf30-l/ect12 seedlings.
Supplementary Table 13
Statistics of H3K27me1 levels of m6A retrotransposons in 6-day-old WT, vir, cpsf30-l, ect12-3 and cpsf30-l/ect12 seedlings.
Supplementary Table 14
Statistics of relative expression levels of m6A retrotransposons in 6-day-old WT, cpsf30-l, ect12-3 and cpsf30-l/ect12 seedlings.
Supplementary Table 15
List of primers used in this study.
Supplementary Data 1
Source data for Supplementary Figs. 1–5 and 8–10.
Source data
Source Data Fig. 1
Unprocessed western blots.
Source Data Fig. 5
Unprocessed western blots.
Source Data Fig. 6
Unprocessed western blots.
Source Data Figs. 1–7
Statistical source data.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Song, P., Cai, Z., Tayier, S. et al. RNA m6A regulates the transcription and heterochromatin state of retrotransposons in Arabidopsis. Nat. Plants 11, 2300–2318 (2025). https://doi.org/10.1038/s41477-025-02137-z
Received:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s41477-025-02137-z









