Skip to main content
Nucleic Acids Research logoLink to Nucleic Acids Research
. 2021 Jul 15;49(14):8309–8323. doi: 10.1093/nar/gkab603

Mutually exclusive substrate selection strategy by human m3C RNA transferases METTL2A and METTL6

Xue-Ling Mao 1, Zi-Han Li 2, Meng-Han Huang 3, Jin-Tao Wang 4, Jing-Bo Zhou 5, Qing-Run Li 6, Hong Xu 7, Xi-Jin Wang 8, Xiao-Long Zhou 9,
PMCID: PMC8373065  PMID: 34268557

Abstract

tRNAs harbor the most diverse posttranscriptional modifications. The 3-methylcytidine (m3C) is widely distributed at position C32 (m3C32) of eukaryotic tRNAThr and tRNASer species. m3C32 is decorated by the single methyltransferase Trm140 in budding yeasts; however, two (Trm140 and Trm141 in fission yeasts) or three enzymes (METTL2A, METTL2B and METTL6 in mammals) are involved in its biogenesis. The rationale for the existence of multiple m3C32 methyltransferases and their substrate discrimination mechanism is hitherto unknown. Here, we revealed that both METTL2A and METTL2B are expressed in vivo. We purified human METTL2A, METTL2B, and METTL6 to high homogeneity. We successfully reconstituted m3C32 modification activity for tRNAThr by METT2A and for tRNASer(GCU) by METTL6, assisted by seryl-tRNA synthetase (SerRS) in vitro. Compared with METTL2A, METTL2B exhibited dramatically lower activity in vitro. Both G35 and t6A at position 37 (t6A37) are necessary but insufficient prerequisites for tRNAThr m3C32 formation, while the anticodon loop and the long variable arm, but not t6A37, are key determinants for tRNASer(GCU) m3C32 biogenesis, likely being recognized synergistically by METTL6 and SerRS, respectively. Finally, we proposed a mutually exclusive substrate selection model to ensure correct discrimination among multiple tRNAs by multiple m3C32 methyltransferases.

INTRODUCTION

Transfer RNA (tRNA) is the most highly and diversely modified RNA species in the cell (1). To date, among 143 currently known modified ribonucleosides, 111 modifications have been identified in tRNAs from all three domains of life (2). tRNA modifications frequently occur in loop regions, such as the D-loop, TψC-loop and anticodon loop, to maintain stability of the tRNA architecture and/or guarantee fidelity and efficiency during ribosomal translation at the decoding site, thereby regulating gene expression and protein homeostasis (3,4).

3-Methylcytidine (m3C) (Supplementary Figure S1A) modification is widely found in eukaryotic cytoplasmic tRNAThr, tRNASer, a subset of tRNAArg species, and mammalian mitochondrial tRNAThr and tRNASer(UCN) at position 32 of the anticodon loop. In addition, it is present at base 20 of mammalian elongator tRNAMet [tRNAMet(e)] and at base 47d (e2) of mammalian tRNALeu(CAG) and all tRNASer species (Supplementary Figure S1B) (5). Only the methyltransferases catalyzing m3C at position 32 (m3C32) of eukaryotic cytoplasmic tRNAThr, tRNASer and tRNAArg have been identified (Supplementary Figure S1B) (Supplementary Table S1) (6–8). In the budding yeast Saccharomyces cerevisiae, only a single enzyme, Trm140 (ScTrm140), introduces m3C32 in both tRNAThr and tRNASer in two different modes (6,7,9). Interestingly, ScTrm140 is expressed in fusion with an upstream actin-binding motif by a programmed +1 frameshift. However, in the fission yeast Schizosaccharomyces pombe, two separate genes encode two m3C32 methyltransferases, Trm140 (SpTrm140) and Trm141 (SpTrm141), without the actin-binding motif (10). Accordingly, SpTrm140 is no longer a dual-specificity enzyme but modifies only tRNAThr, while tRNASer is complementarily modified by SpTrm141 (10). In mammalian cells, mouse Mettl2 and human METTL2A and METTL2B are homologous to ScTrm140/SpTrm140 and have been shown to be responsible for m3C32 formation in tRNAThr species (8). Recently, it has been shown that an additional cofactor, DALRD3, must interact with human METTL2A and/or METTL2B to induce m3C32 formation in human tRNAArg(CCU) and tRNAArg(UCU) species (11), although the interaction pattern and the precise role of each component in modification are still unknown. In addition, SpTrm141-homologous mouse Mettl6 catalyzes m3C32 formation in tRNASer species. Interestingly, both SpTrm141 and Mettl6 interact with seryl-tRNA synthetase (SerRS) (8,9), suggesting that interaction with SerRS is an evolutionarily inherent ability of SpTrm141/Mettl6. The m3C32 modification activity of SpTrm141 is greatly stimulated by the presence of SerRS in vitro (9). However, whether SpTrm141 or Mettl6 alone could mediate m3C32 biogenesis is not yet known, and the precise interaction mode and role of either SpTrm141/Mettl6 or SerRS in tRNA binding are not fully understood. In addition, the biological function of m3C32 is poorly understood. Considering its localization in the anticodon loop, it possibly influences precise pairing between codon and anti-codon and/or biogenesis of ANG-mediated tRNA-derived fragments, as revealed by inhibition effect of m5C formation at C38 (12).

Although genetic data have clearly revealed the above m3C32 methyltransferases, the reconstitution of m3C32 activity using tRNA transcripts in vitro has not been successfully realized (7). Instead, in vitro m3C32 activity was achieved using tRNAs purified from a ScTrm140 gene deletion strain, suggesting that other modifications prior to m3C32 are prerequisites (7). Indeed, genetic studies have clearly demonstrated that t6A at position 37 (t6A37, catalyzed by Sua5 and KEOPS in yeasts) (13,14) or i6A at position 37 (i6A37, catalyzed by MOD5 in yeasts) (15) in specific tRNA substrates significantly triggers m3C32 biogenesis (9,10), giving an exciting example of a tRNA modification circuit (16). Therefore, to determine the tRNA recognition pattern of ScTrm140, different tRNA mutants were expressed in vivo, and the modification status at position 32 was monitored by primer extension assays, which confirmed the importance of t6A37 or i6A37 modification and revealed the key nucleotides in tRNAs (9). However, this assay does not directly determine methylation by ScTrm140, limiting the full understanding of the contributions of other nucleotides of tRNA and of the key amino acids of the methyltransferase or cofactor in m3C biogenesis.

On the other hand, the methylation of nucleotides in mRNA, such as 6-methyladenosine (m6A) and 5-methylcytidine (m5C), plays important roles in gene expression at multiple levels by influencing RNA structure and interactions within the ribosome or by recruiting specific binding proteins that communicate with other signaling pathways in physiological or pathological processes (17–19). Indeed, in addition to its presence in tRNA, m3C is present in mRNA and is suggested to be catalyzed by ScTrm140/SpTrm141/Mettl2a/Mettl6-homologous Mettl8 (8), despite a recent work reporting a dramatically lower abundance of m3C in mRNA than in tRNA (20). However, neither the specific recognition of mRNA and the mechanism of catalysis by Mettl8 nor the potential physiological or pathological role of m3C in mRNA has yet been clearly established. Therefore, studies of the substrate selection mechanism by Mettl8-homologous tRNA m3C32 methyltransferases should help to understand the mRNA m3C modification mechanism.

To understand why mammalian cells need more than one m3C32 methyltransferase and how various homologous enzymes discriminate specific tRNA substrates, in this work, using human tRNA m3C32 methyltransferases (METTL2A, METTL2B and METTL6) as models, we studied their gene expression and cellular localization; we further purified human METTL2A, METTL2B and METTL6 and prepared tRNA transcripts with and without t6A37 modification. We successfully reconstituted the robust m3C32 activities of both METTL2A and METTL6 in vitro, showed that METTL2B exhibited only limited methyltransferase activity in vitro and further provided a detailed tRNA selection mechanism by both enzymes.

MATERIALS AND METHODS

Materials

Anti-FLAG (F7425), anti-Myc (M4439) and anti-GAPDH (G8795) antibodies were purchased from Sigma (St. Louis, MO, USA). Anti-His6 (AE003) was purchased from Abclonal (Shanghai, China). [3H] SAM, [3H] Arg, [14C] Thr and [14C] Ser were obtained from Perkin Elmer Inc. (Waltham, MA, USA). Dynabeads Protein G, MitoTracker and Lipofectamine 2000 transfection reagent were obtained from Thermo Scientific (Waltham, MA, USA). Primers were synthesized in Biosune (Shanghai, China), and DNA sequencing was performed by Tsingke (Shanghai, China).

Plasmid construction, expression and protein purification

Genes encoding METTL2A (UniProt No. Q96IZ6), METTL2B (UniProt No. Q6P1Q9), METTL6 (UniProt No. Q8TCB7) and SerRS (UniProt No. P49591) were amplified from cDNA obtained by reverse transcription of total RNA from human HEK293T cells. For gene expression in HEK293T cells, METTL2A and METTL2B were inserted between the Hind III and Xho I sites of pCMV-3Tag-3A and pCMV-3Tag-4A, and METTL6 was inserted between the Hind III and Xho I sites of pcDNA3.1. For gene expression in E. coli, METTL2A and METTL2B were inserted between the Sac I and Not I sites of pRSFDuet1 with an N-terminal His6 tag, respectively. METTL6 was inserted between the SacI and NotI sites of pRSFDuet1 with an N-terminal His6 tag. SerRS was inserted between the Nde I and Xho I sites of pET22b with a C-terminal His6 tag. The primers used for cloning are listed in Supplementary Table S2. The METTL2A, METTL2B, METTL6 and SerRS genes were expressed in Escherichia coli BL21 (DE3) cells and induced with 200 μM isopropyl β-d-1-thiogalactopyranoside (IPTG) when the initial cell culture reached an absorbance at 600 nm (A600) of 0.6, and transformants were cultured overnight at 18°C. Protein purification from Escherichia. coli transformants was performed with a procedure described in a previous report (21). Protein concentration was determined using a Protein Quantification Kit (BCA Assay, Beyotime, Shanghai, China) according to the manufacturer's instructions.

tRNA gene cloning and transcription

Genes encoding human cytoplasmic (hc) tRNAThr(AGU, CGU, UGU), tRNASer(GCU), tRNAArg(CCU, UCU), tRNAAsn(GUU), tRNAMet(e) and E. coli tRNAThr(UGU) (EctRNAThr) were incorporated into the pTrc99b plasmid. tRNA transcripts were obtained by in vitro T7 RNA polymerase transcription as described previously (22,23). The overexpression and purification of EctRNAThr from E. coli have been described in previous reports (24,25). tRNA gene mutagenesis was performed according to the protocol provided with the KOD-plus mutagenesis kit. The primers used for template preparation are listed in Supplementary Table S2.

Determination of amino acid accepting activities

The amino acid accepting activities of various tRNAs were determined in the following reactions. A 40 μl reaction mixture containing 50 mM Tris–HCl, pH 7.5, 20 mM KCl, 10 mM MgCl2, 2 mM DTT, 4 mM ATP, 20 μM [14C] Thr and 2.5 μM hctRNAThr was incubated with 2 μM mThrRS; a 40 μl reaction mixture containing 50 mM Tris-HCl, pH 7.5, 20 mM KCl, 10 mM MgCl2, 2 mM DTT, 4 mM ATP, 20 μM [14C] Ser and 2.5 μM hctRNASer(GCU) was incubated with 2 μM SerRS; a 40 μl reaction mixture containing 50 mM Tris–HCl, pH 7.5, 80 mM KCl, 12 mM MgCl2, 2 mM DTT, 4 mM ATP, 20 μM [3H] Arg and 2.5 μM hctRNAArg was incubated with 2 μM ArgRS. At time intervals ranging between 15 and 60 min, aliquots of the reaction solution were added to Whatman filter pads and subsequently processed in a similar procedure with aminoacylation assays (26).

t6A activity assay and t6A modification of tRNAs

The t6A modification reaction was performed at 37°C in a 40 μl reaction mixture containing 50 mM Tris–HCl (pH 8.0), 200 mM NaCl, 15 mM MgCl2, 5 mM MnCl2, 50 mM NaHCO3, 5 mM DTT, 4 mM ATP, 100 μM [14C] Thr, 10 μM hctRNAs or variants and 2 μM Sua5 and KEOPS.

Modification of tRNAs or variants with t6A was performed as follows: a 200 μl reaction mixture containing 50 mM Tris–HCl (pH 8.0), 200 mM NaCl, 15 mM MgCl2, 5 mM MnCl2, 50 mM NaHCO3, 5 mM DTT, 4 mM ATP, 1 mM Thr, 50–100 μg of tRNAs or variants and 5 μM Sua5 and KEOPS. The reaction was incubated at 37°C for 1 h, and the t6A-modified tRNA was purified by phenol and chloroform and precipitated by EtOH with NaAc overnight at –20°C. The t6A-modified tRNA concentration was determined by denaturing UREA-PAGE based on linear curves from tRNA transcript samples with known concentrations.

tRNA methylation assay

The methylation reactions were performed at 37°C in a reaction mixture containing 50 mM Tris–HCl (pH 7.5), 20 mM KCl, 10 mM MgCl2, 10 mM spermidine, 10 mM DTT, 20 μM [3H] SAM, 5 μM transcribed, t6A-modified or overexpressed tRNAs, and 1 μM METTL2A, METTL2B or METTL6. At time intervals ranging between 5 and 15 min, aliquots were removed to Whatman filter pads and processed as described above.

LC-MS/MS analysis of t6A and m3C modified tRNA

One microgram of hctRNAThr(AGU), t6A-hctRNAThr(AGU), or m3C-t6A-hctRNAThr(AGU) was completely hydrolyzed by benzonase, phosphodiesterase I, and bacterial alkaline phosphatase in a 60 μl reaction containing 20 mM NH4Ac (pH 5.2) at 37°C for 24 h. One microliter of the solution was then applied to LC–MS/MS analysis. The nucleosides were separated by HPLC on a C18 column (Agilent Zorbax Eclipse Plus C18, 2.1 50 mm, 1.8 mm) and then detected by a triple-quadrupole mass spectrometer (Agilent 6495 QQQ) in positive ion multiple reaction-monitoring mode. Mass transitions from m/z 413.1 to 281.1 (t6A) and m/z 258.1 to 126.1 (m3C) were monitored and recorded.

Cell culture, transfection and co-immunoprecipitation (Co-IP)

HEK293T cells were cultured in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum in a 37°C incubator with 5% CO2 at a confluence of 70% before transfection using Lipofectamine 2000 transfection reagent according to the manufacturer's protocol. At 24 h after transfection, cells were harvested, washed with ice-cold phosphate-buffered saline (PBS) three times, and lysed with 1 ml of ice-cold lysis buffer (50 mM Tris–HCl, pH 7.5, 150 mM NaCl, 5 mM ethylenediaminetetraacetic acid, 1% Triton X-100) supplemented with a protease inhibitor cocktail for 15 min at 4°C with rotation. Co-IP was performed as described previously (27,28).

Western blotting

Protein samples were separated on a 10% separating gel by SDS-PAGE and transferred to a methanol-activated polyvinylidene fluoride (PVDF) membrane, which was then blocked with 5% milk in PBST for 1 h at room temperature. Immunoblotting was performed using anti-FLAG, anti-Myc or anti-HA antibodies overnight, followed by incubation with secondary antibodies, and detected as described previously (27,28)

Immunofluorescence

HEK293T cells were transfected with specific plasmids. After 24 h, cells were stained with MitoTracker for 30 min and then fixed in 4% paraformaldehyde containing PBS for 30 min at room temperature. Fixed cells were blocked in PBS plus 0.1% Triton X-100 buffer containing 5% BSA and incubated with the primary antibody overnight at 4°C. The cells were immunostained with Alexa Fluor 488-conjugated secondary antibody in PBS for 2 h and the nuclear counterstain DAPI for 5 min at room temperature. Fluorescence images were captured with a Leica TCS SP8 STED confocal microscope.

RESULTS

Both METTL2A and METTL2B are expressed in vivo and located in the cytoplasm

In mammalian cells, METTL2A, METTL2B and METTL6 have been shown to participate in tRNAThr and tRNASer m3C32 modification (8). Primary sequence analysis showed that the three human enzymes, together with ScTrm140 and SpTrm141, share a conserved C-terminal S-adenosyl methionine (SAM) binding domain (CTD), while a striking difference exists in the N-terminal domain (NTD) with unassigned function (Supplementary Figure S2). Compared with ScTrm140, human METTL2A and METTL2B have a large insertion (approximately 68 aa in length) in the NTD, while ScTrm141 and human METTL6 display a truncated NTD in the N-terminus. However, all the enzymes retain a conserved ‘FFKDR’ motif with an unknown role in the NTD (Supplementary Figure S2; Supplementary Figure S3A).

In humans, METTL2A (gene ID 339175) and METTL2B (gene ID 55798) are encoded by two separate genes located on chromosomes 17 and 7, respectively. However, only one gene, Mettl2 (gene ID 52686) on chromosome 11, encodes a single Mettl2 for m3C32 modification in mice. Due to the nearly identical genomic and protein sequences of human METTL2A and METTL2B (see text below), the detailed evolutionary path between the two human genes is unclear. Considering protein sequences (both 378 residues in length), only six different sites are present among the two enzymes. Three positions harbor amino acids with similar side chain properties (Val12, Ile266 and Met288 in METTL2A vs. Ile12, Val266 and Val288 in METTL2B), while the other three positions have completely different residues (Arg26, Pro124 and Leu155 in METTL2A vs. Ser26, Cys124 and Pro155 in METTL2B) (Supplementary Figure S3A).

We initially explored whether one or two genes are expressed in human cells by using liquid chromatography-tandem mass spectrometry (LC-MS) analysis of the whole cell lysis (WCL) of HEK293T cells to capture METTL2A- or METTL2B-specific peptides. Indeed, peptides spanning the same region in both METTL2A (AGSYPEGAPAVLADKR) and METTL2B (AGSYPEGAPAILDKR) were clearly detected (Supplementary Figure S3B). These data definitely showed that both METTL2A and METTL2B genes are expressed in vivo.

Furthermore, we introduced a gene encoding a C-terminal FLAG-tagged METTL2A (METTL2A-FLAG) or METTL2B (METTL2B-FLAG) separately into HEK293T cells. Immunofluorescence (IF) assays showed that METTL2A-FLAG and METTL2B-FLAG were both distributed in the cytoplasm (Supplementary Figure S3C). No clear fluorescence signal was observed in the mitochondria (Supplementary Figure S3C).

Subsequently, we purified METTL2A from E. coli cells to high homogeneity (Supplementary Figure S4A). The calculated molecular mass of purified METTL2A together with the His6-tag should be 45.4 kDa. Its molecular mass was determined to be 34.3 kDa by gel filtration analysis with Superdex S200 based on the elution volumes of three standard proteins, apoferritin (443 kDa), yeast alcohol dehydrogenase (150 kDa) and bovine serum albumin (BSA, 66 kDa) (Supplementary Figure S4B, C). Considering that the elution volume of METTL2A was even larger than that of BSA with the smallest molecular mass among the three standards, to more accurately determine its molecular mass, a similar analysis was also performed using Superdex S75, and the molecular mass was determined to be 48.5 kDa (Supplementary Figure S4D, E) based on four standard proteins, conalbumin (75 kDa), ovalbumin (44 kDa), ribonuclease A (RNase A, 13.7 kDa) and aprotinin (6.5 kDa). These results suggested that purified METTL2A was a monomer in solution.

The above data collectively revealed that both the METTL2A and METTL2B genes are readily expressed and that METTL2A and METTL2B are located in the cytoplasm.

t6A37 is essential for m3C32 biogenesis of tRNAThr by METTL2A

m3C32 is present in tRNAThr, tRNASer and tRNAArg in human cells (5). These tRNAs, including hctRNAThr(AGU, CGU, UGU), hctRNASer(GCU) and hctRNAArg(CCU, UCU), were prepared by in vitro T7 run-off transcription (Figure 1A). To validate correct tRNA folding and quality, their corresponding aminoacyl-tRNA synthetases (aaRSs), including mouse cytoplasmic threonyl-tRNA synthetase (ThrRS) (26), human cytoplasmic SerRS and arginyl-tRNA synthetase (ArgRS) (29), were purified. The amino acid accepting activities of these tRNAs were approximately 1000–1500 pmol/A260 (Figure 1A), indicating that the tRNAs were correctly folded and of high quality.

Figure 1.

Figure 1.

t6A37 is essential for the m3C32 modification of hctRNAThr. (A) Urea gel separation of six tRNA transcripts. Their lengths and amino acid accepting activities are indicated below. (B) Time-course curves of the m3C modification of six hctRNA transcripts by METTL2A. (C) LC–MS/MS analysis of the digested products of the hctRNAThr(AGU) transcript, modification products by Sua5/KEOPS and subsequently by METTL2A. (D) Time-course curves of the m3C modification of the six t6A37-modified hctRNAs by METTL2A. Data represent averages of two independent experiments (A) or three independent experiments (B) and the corresponding standard deviation.

Subsequently, we used METTL2A and these tRNAs to reconstitute m3C32 modification activity in vitro. None of these tRNAs were modified by METTL2A (Figure 1B) [modification of hctRNAThr(CGU) was negligible, if any], indicating that purified METTL2A is inactive in vitro, or is active but tRNA transcripts are not suitable substrates or needs a cofactor for catalysis. ScTrm140 recognizes tRNAThr substrates via a sequence element including t6A37 (9). To understand whether t6A37 is a prerequisite for m3C32 formation in humans, the S. cerevisiae t6A modification machinery, including Sua5 and KEOPS, was purified (22). Sua5/KEOPS was able to efficiently modify all six human cytoplasmic tRNA transcripts in vitro (see text below). The tRNAThr(CGU) modified by Sua5/KEOPS was collected, digested with benzonase and analyzed by liquid chromatography coupled with electrospray ionization tandem mass spectrometry (LC-MS/MS). t6A37 was readily detected (Figure 1C), suggesting efficient modification of t6A37 by Sua5/KEOPS. Then, methylation assays clearly showed that METTL2A was able to introduce m3C32 only into the three t6A-hctRNAThr species but not into t6A-hctRNAArg(CCU), -hctRNAArg(UCU) or -hctRNASer(GCU) (Figure 1D). However, the modification levels of the three tRNAThr species were different, with tRNAThr(CGU) having the highest efficiency. In addition, LC-MS/MS analysis confirmed that the m3C32 moiety was readily decorated in the modified hctRNAThr(CGU) products (Figure 1C).

Thus, we successfully reconstituted m3C32 modification activity by METTL2A and revealed that METTL2A alone could modify only tRNAThr but not tRNAArg and tRNASer, which requires t6A37 as a prerequisite.

G35 is a determinant of METTL2A for m3C32 activity reconstitution

We further explored how METTL2A discriminates among different tRNA substrates. Due to the localization of position 32 in the anticodon loop, the various anticodon loops of three hctRNAThrs, two hctRNAArgs and hctRNASer were checked. Each of the hctRNAThrs, with C34, A34 or U34, could be modified, suggesting that position 34 is not a key site for modification. Among other bases in the loop, only position 35 is divergent among these tRNAs, with G35 in all tRNAThrs (Figure 2A). To understand its potential importance, G35 of hctRNAThr(CGU) (with the highest m3C32 modification efficiency) was then mutated to A35, C35 or U35. The tRNA mutants were t6A-modified by Sua5/KEOPS to comparable levels, suggesting that G35 is not an identity element in t6A modification (Supplementary Figure S5A), which was consistent with observations with the human mitochondrial t6A modification enzyme OSGEPL1 (22). After the preparation of t6A-modified tRNAThr(CGU) mutants, methylation determination clearly revealed that m3C32 was no longer formed in the mutants (Figure 2B), suggesting that G35 was a determinant in m3C32 biogenesis by METTL2A.

Figure 2.

Figure 2.

G35 is a determinant of m3C biogenesis. (A) A schema showing the sequences of anticodon loops of six hctRNAs with t6A and m3C32 modifications (indicated in red) and two hctRNAs [hctRNAAsn(GUU) and hctRNAMet(e)] with only t6A modifications. The m3C modification levels of t6A-hctRNAThr(CGU) (black filled circles) and t6A-hctRNAThr(CGU)-G35A (blue filled squares), -G35C (green filled triangles), and -G35U (orange filled inverted triangles) (B); of t6A-hctRNAAsn(GUU) (green filled circles) and t6A-hctRNAAsn(GUU)-U35G mutant (blue filled squares) (C); of t6A-hctRNAMet(e) (black filled circles) and t6A-hctRNAMet(e)-U35G (blue filled squares) (D); and of t6A-hctRNAArg(UCU) (black filled circles) and t6A-hctRNAArg(UCU)-U35G (blue filled squares) (E) by METTL2A. Data represent averages of three independent experiments and the corresponding standard deviation.

Subsequently, in addition to hctRNAArg(UCU) (with C35), we also transcribed hctRNAAsn(GUU) (with U35) and hctRNAMet(e) (with A35), which are used to decode codons starting with A (ANN codons) and are supposed to be modified with t6A (note that hctRNAAsn(GUU) and hctRNAMet(e) do not contain m3C32 in human cells) (30). The nucleotides at position 35 of these tRNAs were also changed to G35. No impairment (instead an increase in hctRNAAsn(GUU) and hctRNAMet(e)) was observed in t6A modification by Sua5/KEOPS with wild-type and mutant tRNAs (Supplementary Figure S5B–D). However, different effects of the presence of G35 in various tRNA species were monitored; both t6A-modified hctRNAAsn(GUU)-U35G and hctRNAMet(e)-A35G clearly gained an m3C32 modification (Figure 2C, D), while m3C32 was only negligibly (if at all) introduced into hctRNAArg(UCU)-C35G (Figure 2E), implying that other elements in addition to t6A37 and G35 also critically control m3C32 formation by METTL2A.

Therefore, the above evidence showed that G35 is a critical element in m3C32 formation in tRNAThr species; introducing only a single G35 into a non-m3C tRNA [hctRNAAsn(GUU) or hctRNAMet(e)] could confer the capacity to be modified by METTL2A.

G35 and t6A37 are insufficient for m3C modification

The above data from hctRNAArg(UCU)-C35G showed that t6A37 and G35 alone are insufficient to confer m3C32 modification in specific tRNAs. t6A37 and G35 are present in E. coli tRNAThr species; however, m3C32 is absent in bacterial tRNAs due to the lack of m3C32 methyltransferase. To study whether METTL2A has the ability to introduce m3C32 to bacterial tRNA species, we prepared a t6A-modified E. coli tRNAThr(UGU) (EctRNAThr) transcript using Sua5/KEOPS. However, we found that t6A-containing EctRNAThr was not modified by METTL2A (Figure 3A). To understand whether other modifications of EctRNAThr were required for efficient reconstitution, we overexpressed and purified EctRNAThr from the E. coli MT102 strain. However, native EctRNAThr was likewise not a substrate of METTL2A. Notably, the t6A-containing hctRNAThr(UGU) transcript was clearly modified by METTL2A (Figure 3A). These results suggested that the tRNA sequence is the primary element leading to EctRNAThr hypomodification.

Figure 3.

Figure 3.

Anticodon stems harbor critical elements for m3C32 modification by METTL2A. (A) m3C modification levels of t6A-hctRNAThr(UGU) (black filled circles), t6A-EctRNAThr(UGU) (blue filled squares) and overexpressed (O.E.) EctRNAThr(UGU) (orange filled inverted triangles) by METTL2A. (B) Secondary structures of EctRNAThr(UGU) (left) and hctRNAThr(AGU) (right) showing the construction of five EctRNAThr mutants and one hctRNAThr(AGU) mutant. m3C modification levels of t6A-EctRNAThr(UGU) (black filled circles) and t6A-EctRNAThr:hc-1 (blue filled squares), :hc-3 (green filled triangles), :hc-4 (orange filled inverted triangles) and :hc-5 (purple filled diamond) (C); of t6A-EctRNAThr(UGU) (black filled circles) and t6A-EctRNAThr:hc-4(blue filled squares), -27(C-G) (green filled triangles) and -31(U-A) (orange filled inverted triangles) (D); and of t6A-hctRNAThr(AGU) (black filled circles) and t6A-hctRNAThr(AGU):Ec (blue filled squares) (E) by METTL2A. EctRNAThr:hc-2 could not be modified with t6A37 and was thus not included in the methylation assay. Data represent averages of three independent experiments (A, C) or two independent experiments (D, E) and the corresponding standard deviation.

Based on the sequence of EctRNAThr and hctRNAThr(AGU) (the two tRNAs were compared because they display the highest sequence identity), we designed five EctRNAThr mutants by replacing some elements with their counterparts in hctRNAThr(AGU), including EctRNAThr:hc-1 (with A73U), :hc-2 (amino acid acceptor stem swapping), :hc-3 (17AGG19 replaced by 17GGU19 in the D-loop), :hc-4 (anticodon stem swapping) [note that the two stems differ only in two base pairs, A27-U43/A31-U39 in EctRNAThr versus C27-G43/U31-A39 in hctRNAThr(AGU)], and :hc-5 (C51-G63/C62 replaced by G51-C63/U62 in the TψC-stem) (Figure 3B). However, EctRNAThr:hc-2 was defective in t6A37 modification by Sua5/KEOPS, while the other mutants were modified with t6A37 to comparable levels (Supplementary Figure S5E). The determination of m3C32 activity showed that METTL2A indeed gained the ability to modify only EctRNAThr:hc-4 (Figure 3C), highlighting the determinant role of the anticodon stem. To further verify which base pairs play a key role in EctRNAThr:hc-4, we obtained two EctRNAThr mutants based on EctRNAThr:hc-4 [EctRNAThr-27(C–G) and EctRNAThr-31(U–A)]. m3C32 activity determination showed that both the C27–G43 and U31–A39 base pairs are needed to confer modification on EctRNAThr (Supplementary Figure S5F, Figure 3D). Conversely, we replaced the anticodon stem of hctRNAThr(AGU) with that of EctRNAThr (Figure 3B), and the resultant hctRNAThr(AGU):Ec displayed impaired but obvious t6A37 modification by Sua5/KEOPS (Supplementary Figure S5G) but was hypomodified by METTL2A (Figure 3E).

The above results clearly showed that in addition to G35 and t6A37, sequences in the anticodon stem are critical elements in determining the m3C32 activity of METTL2A. However, we found that the anticodon stem sequences are not completely conserved among human tRNAThr species, suggesting that they likely work collaboratively with anticodon loops and/or other elements to determine m3C32 levels in specific tRNAs, which is likely why the three human tRNAThr species displayed different m3C32 modification levels in vitro (Figure 1D).

The rationale of hctRNASer(GCU) not modified by METTL2A

The above data clarified the key role of G35 and t6A37 in determining the m3C32 modification status by METTL2A. We have shown that t6A-hctRNASer(GCU) was not modified by METTL2A in vitro. Therefore, we changed C35 of hctRNASer(GCU) to G35, and the mutant hctRNASer(GCU)-C35G exhibited a similar level of t6A modification to wild-type tRNA (Supplementary Figure S5H). Strikingly, the data showed that METTL2A could indeed introduce m3C32 into hctRNASer(GCU)-C35G (Figure 4A), confirming the critical role of G35 in determining the m3C32 modification specificity of METTL2A.

Figure 4.

Figure 4.

C35 and the long variable arm prevent hctRNASer(GCU) from being modified by METTL2A. m3C32 modification levels of t6A-tRNASer(GCU) (black filled circles), t6A-tRNASer(GCU)-C35G (blue filled squares), -v-Thr (orange filled triangles) and -C35G-v-Thr (green filled inverted triangles) (A) and of t6A-hctRNAThr(CGU) (black filled circles) and t6A-hctRNAThr(CGU)-v-Ser (blue filled squares) by METTL2A (C). (B) Schematic diagram showing the construction of various hctRNASer(GCU) and hctRNAThr(CGU) variable arm mutants. Data represent averages of three independent experiments (A) or two independent experiments (C) and the corresponding standard deviation.

hctRNASer(GCU) differs from hctRNAThr predominantly in the long variable arm. To understand whether the long variable arm was also a negative element in m3C32 activity by METTL2A, we replaced hctRNASer(GCU) (44UGUGCUCUGCACGC48) with hctRNAThr(CGU) (44AGAUC48) (Figure 4B). Despite the lack of impairment in t6A modification (Supplementary Figure S5H), however, the resultant hctRNASer(GCU)-v-Thr was still hypomodified by METTL2A (Figure 4A). In sharp contrast, in the C35G context, hctRNASer(GCU)-C35G-v-Thr showed a clearly and robustly greater m3C32 modification level than hctRNASer(GCU)-C35G (Figure 4A). In parallel, we changed the variable loop of hctRNAThr(CGU) to that of hctRNASer(GCU) (Figure 4B). No impairment in t6A modification level was observed (Supplementary Figure S5I); however, m3C32 modification of the mutant hctRNAThr(CGU)-v-Ser was significantly decreased to only slightly higher than the basal level (Figure 4C).

Taken together, these data elucidated that the absence of G35 and the presence of the long variable arm of hctRNASer(GCU) precluded its modification by METTL2A.

METTL2B exhibited little m3C32 modification activity in vitro

Both the METTL2A and METTL2B genes are well expressed in vivo and have the same cytoplasmic distribution, eliciting the question of whether they display similar and redundant m3C32 modification activity. Thus, we purified METTL2B to high homogeneity (Supplementary Figure S6A). Unexpectedly, methyltransferase activity determination showed that the activity of METTL2B was only approximately 1/10 of that of METTL2A (Supplementary Figure S6B). METTL2A contains three sites (Arg26, Pro124 and Leu155) that exhibit totally different side chain properties from their counterparts in METTL2B (Ser26, Cys124 and Pro155). Thus, three single-point mutants, METTL2A-R26S, -P124C and -L155P, were constructed and purified (Supplementary Figure S6A). Subsequent methylation measurement showed that the activities of both METTL2A-R26S and -L155P were reduced to approximately half that of the wild-type enzyme, and the activity of METTL2A-P124C was as low as that of METTL2B (Supplementary Figure S6B). These results suggested that several natural amino acids in METTL2B, especially Cys124, likely determined its low m3C32 methylation activity in comparison with METTL2A.

METTL6 is located in the cytoplasm and nucleus and interacts with SerRS

A previous report has shown that mouse tRNASer species are modified by Mettl6 (8). We overexpressed a gene encoding C-terminal FLAG-tagged human METTL6 (METTL6-FLAG) in HEK293T cells, and IF analysis showed that METTL6-FLAG was distributed in both the cytoplasm and nucleus. Its possible mitochondrial localization was not observed (Supplementary Figure S7A). Furthermore, previous studies have shown that yeast SerRS stimulates the activity of ScTrm140 (9). METTL6-FLAG and a C-terminal Myc-tagged SerRS (SerRS-Myc) were coexpressed in HEK293T cells. By using anti-FLAG antibodies to perform Co-IP, SerRS-Myc could be precipitated with METTL6-FLAG (Supplementary Figure S7B). To understand whether the interaction is direct or indirect by relying on the presence of RNA or DNA, we then digested the DNA or RNA of whole cell lysates by DNase I or RNase A prior to immunoprecipitation. The results suggested that the interaction between METTL6 and SerRS was disrupted by RNase treatment; however, DNase I treatment was unable to abolish the interaction (Supplementary Figure S7B), suggesting that the interaction of METTL6 with SerRS depends on RNA (likely tRNA substrates).

SerRS is essential for the m3C32 biogenesis of hctRNASer(GCU) by METTL6

Subsequently, we overexpressed and purified METTL6 from E. coli (Supplementary Figure S7C). We initially incubated METTL6 with t6A-modified hctRNASer(GCU) based on the above data showing that METTL2A requires t6A37 for efficient m3C32 methylation. However, no methylation product was observed (Figure 5A), indicating that purified METTL6 alone is inactive or is active but requires other cofactors for modification. Considering that yeast SerRS stimulates the activity of ScTrm140 (9) and that METTL6 interacts with SerRS (Supplementary Figure S7B), we further purified human cytoplasmic SerRS (encoded by SARS1) (Supplementary Figure S7C). The inclusion of increasing concentrations of SerRS in the activity assay reaction of METTL6 (SerRS/METTL6 ranging from 1:1 to 5:1) triggered robust m3C modification; however, further elevation of SerRS from 5:1 to 10:1 decreased the m3C modification activity of METTL6 (Figure 5A). Thus, all subsequent activity determination of METTL6 was performed with SerRS at a 5:1 ratio (SerRS/METTL6). These results clearly showed that the m3C modification activity of METTL6 for tRNA, at least for hctRNASer(GCU), requires the presence of SerRS. To explore whether aminoacylation or tRNA binding capacity of SerRS contributes to m3C32 modification of hctRNASer(GCU), we mutated Arg317, which is absolutely conserved in SerRSs from three domains of life and directly interacts with γ-phosphate of AMPPNP in human SerRS-AMPPNP structure (PDB No. 4RQE) but not tRNA (Supplementary Figure S8A) (31), to Ala. SerRS-R317A exhibited an abolished aminoacylation activity (Supplementary Figure S8B); however, it stimulated m3C32 activity of METTL6 to comparable levels (Supplementary Figure S8C), indicating that tRNA binding but not aminoacylation by SerRS contributed to m3C32 modification by METTL6. Subsequently, to understand whether t6A37 is a determinant in METTL6-catalyzed methylation, we used hctRNASer(GCU) transcript as a substrate. Again, METTL6 showed no activity in the absence of SerRS (Figure 5B). In contrast to the modification of t6A-hctRNAThr(CGU) by METTL2A, the modification of the hctRNASer(GCU) transcript was as robust as that of t6A-modified hctRNASer(GCU), with similar trends concerning the relative ratio of SerRS/METTL6 (5:1 ratio with the highest efficiency) (Figure 5B). These results highlighted that the t6A37 moiety was nonessential and contributed little to m3C biogenesis in hctRNASer(GCU) by METTL6.

Figure 5.

Figure 5.

m3C modification activity of METTL6 for hctRNASer(GCU) requires the presence of SerRS and the long variable arm. m3C modification levels of t6A-hctRNASer(GCU) (A) and hctRNASer(GCU) transcripts (B) by METTL2A without SerRS (red filled circles) and with increasing amounts of SerRS as indicated. (C) m3C modification levels of hctRNASer(GCU) transcript (black filled circles) and hctRNASer(GCU)-C32G (blue filled squares) by METTL6-SerRS. (D) m3C modification levels of hctRNASer(GCU) (black filled circles), hctRNAThr(CGU) (blue filled squares) and hctRNAArg(UCU) (green filled triangles) transcripts by METTL6-SerRS. (E) m3C modification levels of hctRNASer(GCU) transcript (black filled circles) and hctRNASer(GCU)-v-Thr (blue filled squares), -v-Leu (green filled triangles) and -v-Sec (orange filled inverted triangles) by METTL6-SerRS; (F) Schematic diagram of constructing various variable stem and loop replacement mutants and (G) various base pair or base substitutions based on hctRNASer(GCU). m3C modification levels of hctRNASer(GCU) transcript (black filled circles) and hctRNASer(GCU)-e11 (C-G) (blue filled squares), -e12 (U-G) (green filled triangles), -e13 (C-G) (orange filled inverted triangles), -e14 (U-G) (purple filled diamond) and -ΔUe3 (red hollow circle) (H); of hctRNASer(GCU) transcript (black filled circles) and hctRNASer(GCU)-Ue1G (blue filled squares), -Ce2A (green filled triangles), -Ue3G (orange filled inverted triangles), -e12 (A-U) (purple filled diamond) and -e12 (G-C) (red hollow circle) (I); of hctRNAThr(CGU) transcript (black filled circles) and hctRNAThr(CGU)-v-Ser (blue filled squares) (J) by METTL6-SerRS. Data represent averages of two independent experiments except three independent experiments (C) and the corresponding standard deviation.

Both positions 32 and 47d (e2) of hctRNASer(GCU) contain m3C modification. We then determined which position was m3C-modified by METTL6-SerRS. To this end, we prepared a hctRNASer(GCU)-C32G mutant, which was subsequently found to be hypomodified, suggesting that METTL6-SerRS is responsible for m3C32 but not m3C47d biogenesis (Figure 5C).

Rationale of the lack of modification of tRNAThr by METTL6-SerRS due to the lack of the variable region of tRNASer as a modification determinant

Subsequently, we found no modification of hctRNAThr(CGU) and hctRNAArg(UCU) transcripts by METTL6-SerRS (Figure 5D), suggesting that METTL6-SerRS has high specificity for only tRNASer.

With a typical long variable arm, hctRNASer, together with hctRNALeu and hctRNASec, constitute all the class II tRNAs in human cells. To determine any role of the long variable arm, hctRNASer-v-Thr (Figure 4A) was used as a substrate in the METTL6-SerRS modification assay (Figure 5E). The results showed that the replacement of the variable arm abolished m3C32, indicating a critical role of the long variable arm. Furthermore, the variable arm of hctRNASer(GCU) was changed to that of class II hctRNASec or hctRNALeu(UAG) (Figure 5F). Similarly, the data showed that neither hctRNASer(GCU)-v-Sec nor hctRNASer(GCU)-v-Leu was m3C32-modified by METTL6-SerRS (Figure 5E). The above data collectively revealed that METTL6-SerRS modified hctRNASer(GCU) depending on the sequence of the long variable arm.

Subsequently, we performed a sequence comparison among the variable arms of hctRNASer(GCU), hctRNASec and hctRNALeu(UAG). The variable arm of hctRNASer(GCU) most resembles that of hctRNALeu(UAG) (Figure 5F), sharing U44, Ue1, Ce2, G47 and C48, but with a reduced size of the variable loop and a different variable stem. Targeting the different base pairs, we initially replaced each base of hctRNASer(GCU) with that of hctRNALeu(UAG), including e11 (C–G), e12 (U-G), e13 (C-G) and e14 (U-G). Additionally, to obtain different loop sizes, we deleted Ue3 (ΔUe3) (Figure 5G). Methylation determination using the above hctRNASer(GCU) mutants showed that, in comparison to wild-type hctRNASer(GCU), the modification of hctRNASer(GCU)-ΔUe3 and -e12 (U–G) was completely abolished; that of hctRNASer(GCU)-e11 (C-G) was dramatically reduced, while that of hctRNASer(GCU)-e13 (C–G) or -e14 (U–G) was not affected at all (Figure 5H). These data suggested that the e12 base pair and loop size and/or sequence were critical for m3C32 modification. We further replaced e12 (U-A) with e12 (A–U) or e12 (G–C) and constructed hctRNASer(GCU)-Ue1G, -Ce2A and -Ue3G (Figure 5G). We found that a single point mutation at position Ue1 or Ce2 decreased m3C32 modification and that mutation at Ue3 abolished methylation, while the modification of hctRNASer(GCU)-e12 (A–U) or -e12 (G–C) decreased to different extents (Figure 5I). It is worth noting that the Ce2A mutant from Ce2 (namely, C47d) could still be m3C-modified, again confirming that METTL6 introduces methylation at position C32 but not Ce2 (C47d).

In addition, in contrast to hctRNAThr(CGU), after transplanting the variable arm of tRNASer, hctRNAThr(CGU)-v-Ser (Figure 4A) clearly gained m3C32 modification by METTL6-SerRS, although with clearly lower efficiency than hctRNASer(GCU) (Figure 5J).

Altogether, these results revealed that hctRNAThr was not a substrate of METTL6-SerRS due to lacking the long variable arm of hctRNASer; the base pairs e11 and e12 and residues e1 and e3 are among the critical nucleotide elements for the m3C32 modification of hctRNASer(GCU) by METTL6-SerRS.

Anticodon loop harbors key determinants for m3C32 modification by METTL6-SerRS

METTL6 requires SerRS for m3C32 modification (Figure 5A, B) but does not directly bind it (Supplementary Figure S7B). It is well established that the long variable arm of tRNASer is bound and recognized by SerRS (32). In addition, G35 is a critical determinant of methylation by METTL2A. All this evidence prompted us to explore whether the anticodon loop of hctRNASer(GCU) plays a potentially important role in tRNA recognition by METTL6. To this end, we changed each base of the anticodon loop of hctRNASer(GCU), except the C32 modification site (Figure 6A), resulting in the hctRNASer(GCU)-U33G, -G34A, -C35G, -U36A, -A37C, and -A38C mutants. Methylation determination using the above mutants showed that, in comparison to that of wild-type hctRNASer(GCU), modification of hctRNASer(GCU)-U33G, -G34A, -U36A and -A37C modification was completely abolished, and hctRNASer(GCU)-C35G modification was dramatically reduced, while that of hctRNASer(GCU)-A38C was unexpectedly significantly increased (Figure 6B). Among the above single-point mutants, only hctRNASer(GCU)-G34A and -C35G exhibited comparable levels of t6A modification by Sua5/KEOPS; therefore, we prepared t6A-modified hctRNASer(GCU)-G34A and -C35G. The results showed that t6A-hctRNASer(GCU)-G34A was still hypomodified, while the modification efficiency for t6A-hctRNASer(GCU)-C35G was slightly elevated compared with its transcript (Figure 6B, C), suggesting that the presence of t6A37 compensates for the loss of the optimal anticodon sequence.

Figure 6.

Figure 6.

The anticodon loop critically determines m3C32 formation in hctRNASer (GCU) by METTL6-SerRS. (A) Schematic diagram showing the construction of six hctRNASer(GCU) mutants. m3C modification levels of hctRNASer(GCU) transcript (black filled circles) and hctRNASer(GCU)-U33G (blue filled squares), -G34A (green filled triangles), -C35G (orange filled inverted triangles), -U36A (purple filled diamonds), -A37C (red hollow circles) and -A38C (brown forks) (B); of t6A-hctRNASer(GCU) (black filled circles) and t6A-hctRNASer(GCU)-G34A (green filled triangles) and -C35G (orange filled inverted triangles) (C); of hctRNASer(GCU) transcript (black filled circles) and hctRNASer(AGA) transcript (blue filled squares) (D) by METTL6-SerRS. Data represent averages of three independent experiments (B) or two independent experiments (C, D) and the corresponding standard deviation.

hctRNASer(AGA) also harbors m3C32 modification in vivo (5); however, it has a different anticodon from that of hctRNASer(GCU). Subsequent modification assays showed that, in contrast to hctRNASer(GCU), hctRNASer(AGA) transcript was unable to be modified by METTL6-SerRS (Figure 6D). This result, together with the stimulatory role of t6A37 in the hctRNASer(GCU)-C35G mutant, suggested that the presence of i6A37 modification in hctRNASer(AGA) is likely a key determinant of its m3C32 modification.

In summary, in addition to the long variable arm, the anticodon loop of hctRNASer(GCU) contains key elements that determine m3C32 biogenesis by METTL6-SerRS.

The NTD and CTD domains of METTL2A and METTL6 are mutually incompatible

METTL2A and METTL6 share highly similar CTD domains. However, the NTD domain of METTL6 is sharply truncated or degenerated compared with that of METTL2A. Which domain or element in enzymes determines their totally distinct substrate specificity is unclear. Therefore, we switched the corresponding NTD domains to understand whether the substrate preference could be artificially altered. We fused the NTD of METTL6 with the CTD of METTL2A to obtain N6-METTL2A; similarly, N2-METTL6 was also constructed (Supplementary Figure S9A).

We purified both N6-METTL2A and N2-METTL6 from E. coli. In vitro methylation assays showed that neither enzyme was able to introduce methylation at t6A-hctRNAThr(CGU) (Supplementary Figure S9B), suggesting that both the METTL2A NTD and CTD domains are critical for its m3C32 modification activity. In the presence of SerRS, we also determined the modification of t6A37-hypomodified or t6A37-modified hctRNASer(GCU) by the two chimeric enzymes. Similarly, no modification was observed (Supplementary Figure S9C, D). In addition, considering that METTL2A requires G35 as a positive determinant, we also modified t6A-hctRNASer(GCU)-C35G; again, the two chimeric enzymes generated no m3C32 (Supplementary Figure S9E).

Furthermore, we expressed the genes encoding METTL2A, METTL6, N6-METTL2A and N2-METTL6 in HEK293T cells. However, the two chimeric mutants were not detected in the WCL, probably due to inefficient expression or rapid degradation after synthesis (Supplementary Figure S9F).

Above all, these data collectively suggested that the NTD and CTD domains of METTL2A and METTL6 were mutually incompatible in expression/stability in vivo and for m3C32 modification in vitro.

DISCUSSION

Only a single Mettl2 is present in some eukaryotes, such as mice; however, the simultaneous existence of two nearly identical m3C32 methyltransferases with the same cellular distribution, METTL2A and METTL2B, in others, such as human, is puzzling (8). We showed here that the activity of METTL2B is far lower than that of METTL2A. Notably, among the three different variations between the two enzymes, alternative rigid Pro residue is frequently observed. Indeed, the P124C mutation alone in METTL2A is sufficient to dramatically reduce its activity to levels comparable to those of METTL2B. Considering that DALRD3 is required for m3C32 formation in tRNAArg(CCU) and tRNAArg(UCU) isoacceptors in human cells (11), it is likely that these amino acid variations between two enzymes, especially the presence or absence of rigid Pro, frequently function in determining local/global protein conformation and fine-tune protein conformation and/or structure, which may influence their interaction with DARLD3 to control the tRNAArg methylation level. Alternatively, METTL2B possibly needs other unknown co-factors in catalyzing tRNA modification. On the other hand, tRNA modification has been shown to be highly dynamic in response to various stimuli or stresses (33,34). It is possible that the m3C32 modification level is precisely balanced based on the expression level or ratio of METTL2A and METTL2B, which is regulated in a tissue- or cell-specific manner.

The deficiency of METTL2A in modifying tRNAArg is intriguing, considering that tRNAArg and tRNAThr, both class I tRNAs, have similar secondary and tertiary structures, in contrast to class II tRNASer. The presence of a tRNA binding motif in DALRD3 implies that METTL2A is inefficient in binding tRNAArg and is probably assisted by DALRD3. However, the exact functional assignment of METTL2A-DALRD3 remains unclear.

We also observed that once G35 is introduced into hctRNASer(GCU), in the presence of t6A37, the tRNA mutant gains the ability to be modified by METTL2A, although further deletion of the long variable arm significantly elevates the modification level. However, in human cells, most hctRNASer species, including hctRNASer(AGA), hctRNASer(UGA) and hctRNASer(CGA), naturally contain a G35. Sure, in these hctRNASer species with G35, i6A37 is present instead of t6A37. In addition to the long variable arm, whether C35 and t6A37 in hctRNASer(GCU) or G35 and i6A37 in other hctRNASer species synergistically determine substrate specificity by METTL6-SerRS but not METTL2A is unknown and needs further exploration.

One remarkable difference revealed here is that METTL6 does not require modification at residue 37 as a prerequisite, at least in modification for hctRNASer(GCU). On the other hand, m3C32 formation by METTL6 is dependent on the presence of SerRS. METTL6 did not directly interact with SerRS. Increasing ratio of SerRS to METTL6 from 5:1 to 10:1 decreased methylation activity of METTL6; we suggested that the affinity between components of METTL6-tRNASer-SerRS ternary complex is not strong and too much SerRS might form SerRS-tRNASer binary complex and thus competes with formation of ternary complex. On the other hand, the possibility of presence of trace amount of nuclease in the SerRS sample cannot be absolutely excluded. Perplexingly, a recent report shows that purified GST-tagged METTL6 alone is able to introduce m3C32 modification into total cellular RNA (35). hctRNASer(GCU) transcript was used in this work. Although the possibility that other modifications in tRNASer species eliminate the requirement of SerRS cannot be absolutely excluded, we suggest that the modification of total RNA may not be derived from tRNA modification. Alternatively, modifications other than hctRNASer(GCU) used here, including hctRNASer(AGA), hctRNASer(UGA) or hctRNASer(CGA), could be performed by METTL6 alone. However, when the hctRNASer(UGA) transcript was used in an in vitro methylation assay, only a basal level of modification (with CPM 100–300) was observed (35). Therefore, we hypothesize that the m3C32 activity of METTL6 for hctRNASer is critically dependent on SerRS. The biological function of m3C32 modification, despite being currently unidentified, should be of high significance considering that multiple tRNA methyltransferases and cofactors have evolved in human cells. Thus, SerRS is indeed a key multifaceted regulator in protein synthesis, vascular development and other functions, such as tRNA modification (36).

m3C modification has been shown to be present in mRNA (8). The independence of the t6A modification of METTL6 elicits another interesting question: is METTL6 able to introduce m3C to RNA species other than tRNA? To date, t6A modification has been detected only in tRNAs and not found in mRNAs. Therefore, in combination with our findings that METTL2A is dependent on t6A modification prior to m3C biogenesis, METTL6 is more likely to form m3C in RNAs other than METTL2A. Previous work has shown that yeast Trm140 recognizes SctRNAThrs and SctRNASer(GCU) depending on t6A37 and SctRNASer(AGA), (CGA) and (UGA) depending on i6A37. In addition, XGU and t6A37 are sufficient for m3C32 modification of yeast tRNAThrs (9). Our work shows that the modification of tRNAThrs by human METTL2A also requires an anticodon stem and that human METTL6 recognizes hctRNASer(GCU) independently of t6A, suggesting that humans and yeasts have different mechanisms for recognizing tRNA substrates. Indeed, cellular mRNAs frequently form anticodon stem and loop-like architectures, which are responsible for recruiting interacting protein partners, such as aminoacyl-tRNA synthetases (37). In addition, some noncoding RNAs contain tRNA-like structures (38). Although a previous report indicated no obvious change in m3C abundance in the mRNA fraction in Mettl6 knockout mice (8), the frequency of m3C modification by METTL6 may be low to be accurately captured. Indeed, a recent m3C mapping analysis revealed little m3C abundance in mRNA (20).

Mettl8 has been shown to participate mRNA m3C biogenesis (8). However, nothing is known about the mRNA substrate selection mechanism of Mettl8. Mettl8 resembles METTL2A most closely in primary sequence. Our clarification of the tRNA selection recognition pattern by METTL2A, especially the key role of the anticodon stem and loop region of tRNA, provides valuable insights into how METTL8 recognizes mRNA substrates. Considering that t6A modification has not been detected in mRNA, whether Mettl8 does not rely on t6A modification is an open question. Moreover, our work showed that neither METTL6 nor METTL2A was significantly localized in mitochondria, while a recent study showed that METTL8 was localized in mitochondria (39). Therefore, we reasonably infer that METTL8 is highly likely to be responsible for the m3C32 modification of mitochondrial tRNAs.

The CTDs for binding SAM are highly conserved, while the most striking sequence difference is observed in the NTDs between METTL2A and METTL6. The NTD of METTL6 is significantly truncated only with a conserved ‘FFKDR’ motif; however, that of METTL2A is a much longer domain even than those of ScTrm140 and SpTrm140 (Supplementary Figure S2). In combination with our revealed tRNA sequence requirement for both hctRNAThrs and hctRNASer(GCU), we proposed a model for multiple substrate selection and modification by multiple m3C32 modification enzymes (Figure 7). The CTD of both enzymes recognizes the anticodon loop region, relying on distinct sets of determinants in hctRNAThrs (G35 and t6A37) and hctRNASer(GCU) (positions 33–37); the long NTD of METTL2A recognizes other elements, such as the anticodon stem, of hctRNAThrs. However, the degenerated NTD of METTL6 is unable to bind hctRNASer(GCU) efficiently, which is augmented by SerRS for recognizing the long variable arm (including Ue3 and e12 base pairs), but they do not interact directly. METTL2A fails to modify hctRNASer(GCU) due to the presence of C35 in the anticodon and the long variable arm, possibly leading to spatial conflict and/or electrostatic repulsion between the variable arm and the long NTD of METTL2A. On the other hand, hctRNAThrs cannot be decorated by METTL6 due to an unfavorable anticodon, the lack of the long variable arm of hctRNAThrs (leading to inability to be captured by METTL6-SerRS) and the truncated NTD of METTL6 (making the efficient recognition of hctRNAThrs by METTL6 alone impossible). Notably, hctRNASer(AGA) transcripts cannot be modified by METTL6-SerRS, suggesting a distinct recognition pattern between t6A37- and i6A37-harboring hctRNASer species, which should be further explored.

Figure 7.

Figure 7.

Model of mutually exclusive substrate selection strategy by human m3C RNA transferases METTL2A and METTL6. When hctRNAThrs are modified by METTL2A, the key elements in the anticodon loop (t6A37 and G35) are recognized by the CTD, and other key elements, including the anticodon stem, are recognized by the long NTD (lower left quadrant). hctRNASer (GCU) is modified by METTL6 with the assistance of SerRS, which recognizes the essential elements on the anticodon loop (bases 33–37) and variable arm (including base pair e12 and Ue3). The truncated NTD of METTL6 is unable to bind tRNA as efficiently as that of METTL2A (upper right quadrant). hctRNAThrs are not modified by METTL6-SerRS due to the unfavorable anticodon loop and lack of the long variable arm (upper left quadrant); in parallel, hctRNASer(GCU) is not modified by METTL2A due to the lack of G35 and presence of a long variable arm (lower right quadrant). The variable arms are indicated in pink.

Supplementary Material

gkab603_Supplemental_File

ACKNOWLEDGEMENTS

We thank Prof. En-Duo Wang in our institute for valuable advices. We are grateful to Dr. Luang Xu in our institute for assistance in LC–MS/MS analysis. We also thank Dr Herman van Tilbeurgh (Institute for Integrative Biology of the Cell, CNRS) and Dr Jin-Qiu Zhou in our institute for providing the plasmid expressing KEOPS and Dr Xi-Peng Liu (Shanghai Jiao Tong University) for providing the plasmid expressing DNA polymerase.

Contributor Information

Xue-Ling Mao, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

Zi-Han Li, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

Meng-Han Huang, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

Jin-Tao Wang, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

Jing-Bo Zhou, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

Qing-Run Li, CAS Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

Hong Xu, Shanghai Key Laboratory of Embryo Original Diseases, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910 Heng Shan Road, Shanghai 200030, China.

Xi-Jin Wang, Department of Neurology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kong Jiang Road, Shanghai 200092, China.

Xiao-Long Zhou, State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.

SUPPLEMENTARY DATA

Supplementary Data are available at NAR Online.

FUNDING

Natural Science Foundation of China [31670801, 31822015, 81870896]; Shanghai Key Laboratory of Embryo Original Diseases [Shelab201904]; Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University [ZDFY2020-RG-0003]. Funding for open access charge: Natural Science Foundation of China [31670801, 31822015, 81870896].

Conflict of interest statement. None declared.

REFERENCES

  • 1.El Yacoubi B., Bailly M., de Crecy-Lagard V.. Biosynthesis and function of posttranscriptional modifications of transfer RNAs. Annu. Rev. Genet. 2012; 46:69–95. [DOI] [PubMed] [Google Scholar]
  • 2.McCown P.J., Ruszkowska A., Kunkler C.N., Breger K., Hulewicz J.P., Wang M.C., Springer N.A., Brown J.A.. Naturally occurring modified ribonucleosides. Wiley Interdiscip Rev RNA. 2020; 11:e1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Suzuki T.The expanding world of tRNA modifications and their disease relevance. Nat. Rev. Mol. Cell Biol. 2021; 22:375–392. [DOI] [PubMed] [Google Scholar]
  • 4.Agris P.F., Narendran A., Sarachan K., Väre V.Y.P., Eruysal E.. The importance of being modified: The role of RNA modifications in translational fidelity. Enzymes. 2017; 41:1–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Clark W.C., Evans M.E., Dominissini D., Zheng G., Pan T.. tRNA base methylation identification and quantification via high-throughput sequencing. RNA. 2016; 22:1771–1784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.D'Silva S., Haider S.J., Phizicky E.M.. A domain of the actin binding protein Abp140 is the yeast methyltransferase responsible for 3-methylcytidine modification in the tRNA anti-codon loop. RNA. 2011; 17:1100–1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Noma A., Yi S., Katoh T., Takai Y., Suzuki T., Suzuki T.. Actin-binding protein ABP140 is a methyltransferase for 3-methylcytidine at position 32 of tRNAs in Saccharomyces cerevisiae. RNA. 2011; 17:1111–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Xu L., Liu X., Sheng N., Oo K.S., Liang J., Chionh Y.H., Xu J., Ye F., Gao Y.G., Dedon P.C.et al.. Three distinct 3-methylcytidine (m3C) methyltransferases modify tRNA and mRNA in mice and humans. J. Biol. Chem. 2017; 292:14695–14703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Han L., Marcus E., D'Silva S., Phizicky E.M.. S. cerevisiae Trm140 has two recognition modes for 3-methylcytidine modification of the anticodon loop of tRNA substrates. RNA. 2017; 23:406–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Arimbasseri A.G., Iben J., Wei F.Y., Rijal K., Tomizawa K., Hafner M., Maraia R.J.. Evolving specificity of tRNA 3-methyl-cytidine-32 (m3C32) modification: a subset of tRNAsSer requires N6-isopentenylation of A37. RNA. 2016; 22:1400–1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lentini J.M., Alsaif H.S., Faqeih E., Alkuraya F.S., Fu D.. DALRD3 encodes a protein mutated in epileptic encephalopathy that targets arginine tRNAs for 3-methylcytosine modification. Nat. Commun. 2020; 11:2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Schaefer M., Pollex T., Hanna K., Tuorto F., Meusburger M., Helm M., Lyko F.. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev. 2010; 24:1590–1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.El Yacoubi B., Hatin I., Deutsch C., Kahveci T., Rousset J.P., Iwata-Reuyl D., Murzin A.G., de Crecy-Lagard V.. A role for the universal Kae1/Qri7/YgjD (COG0533) family in tRNA modification. EMBO J. 2011; 30:882–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Srinivasan M., Mehta P., Yu Y., Prugar E., Koonin E.V., Karzai A.W., Sternglanz R.. The highly conserved KEOPS/EKC complex is essential for a universal tRNA modification, t6A. EMBO J. 2011; 30:873–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dihanich M.E., Najarian D., Clark R., Gillman E.C., Martin N.C., Hopper A.K.. Isolation and characterization of MOD5, a gene required for isopentenylation of cytoplasmic and mitochondrial tRNAs of Saccharomyces cerevisiae. Mol. Cell. Biol. 1987; 7:177–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Han L., Phizicky E.M.. A rationale for tRNA modification circuits in the anticodon loop. RNA. 2018; 24:1277–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhao B.S., Roundtree I.A., He C.. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 2017; 18:31–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chen Y.S., Yang W.L., Zhao Y.L., Yang Y.G.. Dynamic transcriptomic m5C and its regulatory role in RNA processing. Wiley Interdiscip Rev. RNA. 2021; 12:e1639. [DOI] [PubMed] [Google Scholar]
  • 19.Gu C., Shi X., Dai C., Shen F., Rocco G., Chen J., Huang Z., Chen C., He C., Huang T.et al.. RNA m6A modification in cancers: Molecular mechanisms and potential clinical applications. The Innovation. 2020; 1: 10.1016/j.xinn.2020.100066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cui J., Liu Q., Sendinc E., Shi Y., Gregory R.I.. Nucleotide resolution profiling of m3C RNA modification by HAC-seq. Nucleic Acids Res. 2021; 49:e27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zeng Q.Y., Peng G.X., Li G., Zhou J.B., Zheng W.Q., Xue M.Q., Wang E.D., Zhou X.L.. The G3-U70-independent tRNA recognition by human mitochondrial alanyl-tRNA synthetase. Nucleic Acids Res. 2019; 47:3072–3085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhou J.B., Wang Y., Zeng Q.Y., Meng S.X., Wang E.D., Zhou X.L.. Molecular basis for t6A modification in human mitochondria. Nucleic Acids Res. 2020; 48:3181–3194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wang Y., Zhou J.B., Zeng Q.Y., Wu S., Xue M.Q., Fang P., Wang E.D., Zhou X.L.. Hearing impairment-associated KARS mutations lead to defects in aminoacylation of both cytoplasmic and mitochondrial tRNALys. Sci. China Life Sci. 2020; 63:1227–1239. [DOI] [PubMed] [Google Scholar]
  • 24.Zhou X.L., Chen Y., Fang Z.P., Ruan Z.R., Wang Y., Liu R.J., Xue M.Q., Wang E.D.. Translational quality control by bacterial threonyl-tRNA Synthetases. J. Biol. Chem. 2016; 291:21208–21221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zheng W.Q., Zhang Y., Yao Q., Chen Y., Qiao X., Wang E.D., Chen C., Zhou X.L.. Nitrosative stress inhibits aminoacylation and editing activities of mitochondrial threonyl-tRNA synthetase by S-nitrosation. Nucleic Acids Res. 2020; 48:6799–6810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chen Y., Ruan Z.R., Wang Y., Huang Q., Xue M.Q., Zhou X.L., Wang E.D.. A threonyl-tRNA synthetase-like protein has tRNA aminoacylation and editing activities. Nucleic Acids Res. 2018; 46:3643–3656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Peng G.X., Zhang Y., Wang Q.Q., Li Q.R., Xu H., Wang E.D., Zhou X.L.. The human tRNA taurine modification enzyme GTPBP3 is an active GTPase linked to mitochondrial diseases. Nucleic Acids Res. 2021; 49:2816–2834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhou X.L., Chen Y., Zeng Q.Y., Ruan Z.R., Fang P., Wang E.D.. Newly acquired N-terminal extension targets threonyl-tRNA synthetase-like protein into the multiple tRNA synthetase complex. Nucleic Acids Res. 2019; 47:8662–8674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Li G., Eriani G., Wang E.D., Zhou X.L.. Distinct pathogenic mechanisms of various RARS1 mutations in Pelizaeus-Merzbacher-like disease. Sci. China Life Sci. 2021; 10.1007/s11427-020-1838-2. [DOI] [PubMed] [Google Scholar]
  • 30.Thiaville P.C., Iwata-Reuyl D., de Crecy-Lagard V.. Diversity of the biosynthesis pathway for threonylcarbamoyladenosine (t6A), a universal modification of tRNA. RNA Biol. 2014; 11:1529–1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang C., Guo Y., Tian Q., Jia Q., Gao Y., Zhang Q., Zhou C., Xie W.. SerRS-tRNASec complex structures reveal mechanism of the first step in selenocysteine biosynthesis. Nucleic Acids Res. 2015; 43:10534–10545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Biou V., Yaremchuk A., Tukalo M., Cusack S.. The 2.9 A crystal structure of T. thermophilus seryl-tRNA synthetase complexed with tRNASer. Science. 1994; 263:1404–1410. [DOI] [PubMed] [Google Scholar]
  • 33.Preston M.A., D'Silva S., Kon Y., Phizicky E.M.. tRNAHis 5-methylcytidine levels increase in response to several growth arrest conditions in Saccharomyces cerevisiae. RNA. 2013; 19:243–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Galvanin A., Vogt L.M., Grober A., Freund I., Ayadi L., Bourguignon-Igel V., Bessler L., Jacob D., Eigenbrod T., Marchand V.et al.. Bacterial tRNA 2′-O-methylation is dynamically regulated under stress conditions and modulates innate immune response. Nucleic Acids Res. 2020; 48:12833–12844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ignatova V.V., Kaiser S., Ho J.S.Y., Bing X., Stolz P., Tan Y.X., Lee C.L., Gay F.P.H., Lastres P.R., Gerlini R.et al.. METTL6 is a tRNA m3C methyltransferase that regulates pluripotency and tumor cell growth. Sci. Adv. 2020; 6:eaaz4551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Xu X., Shi Y., Zhang H.M., Swindell E.C., Marshall A.G., Guo M., Kishi S., Yang X.L.. Unique domain appended to vertebrate tRNA synthetase is essential for vascular development. Nat. Commun. 2012; 3:681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Levi O., Arava Y.. mRNA association by aminoacyl tRNA synthetase occurs at a putative anticodon mimic and autoregulates translation in response to tRNA levels. PLoS Biol. 2019; 17:e3000274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wilusz J.E., Freier S.M., Spector D.L.. 3′ end processing of a long nuclear-retained noncoding RNA yields a tRNA-like cytoplasmic RNA. Cell. 2008; 135:919–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rath S., Sharma R., Gupta R., Ast T., Chan C., Durham T.J., Goodman R.P., Grabarek Z., Haas M.E., Hung W.H.W.et al.. MitoCarta3.0: an updated mitochondrial proteome now with sub-organelle localization and pathway annotations. Nucleic Acids Res. 2021; 49:D1541–D1547. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

gkab603_Supplemental_File

Articles from Nucleic Acids Research are provided here courtesy of Oxford University Press

RESOURCES