ARTICLE

https://doi.org/10.1038/s41467-020-16321-6 OPEN DALRD3 encodes a protein mutated in epileptic encephalopathy that targets arginine tRNAs for 3-methylcytosine modification ✉ Jenna M. Lentini1, Hessa S. Alsaif2, Eissa Faqeih3, Fowzan S. Alkuraya 2,4 & Dragony Fu 1

In mammals, a subset of arginine tRNA isoacceptors are methylated in the anticodon loop by the METTL2 methyltransferase to form the 3-methylcytosine (m3C) modification. However, 1234567890():,; the mechanism by which METTL2 identifies specific tRNA arginine species for m3C forma- tion as well as the biological role of m3C in mammals is unknown. Here, we show that human METTL2 forms a complex with DALR anticodon binding domain containing 3 (DALRD3) protein to recognize particular arginine tRNAs destined for m3C modification. DALRD3- deficient human cells exhibit nearly complete loss of the m3C modification in tRNA-Arg species. Notably, we identify a homozygous nonsense mutation in the DALRD3 that impairs m3C formation in human patients exhibiting developmental delay and early-onset epileptic encephalopathy. These findings uncover an unexpected function for the DALRD3 protein in the targeting of distinct arginine tRNAs for m3C modification and suggest a crucial biological role for DALRD3-dependent tRNA modification in proper neurological development.

1 Department of Biology, Center for RNA Biology, University of Rochester, Rochester, NY, USA. 2 Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia. 3 Section of Medical Genetics, Children’s Specialist Hospital, King Fahad Medical City, Riyadh, Saudi Arabia. ✉ 4 Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia. email: [email protected]

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6

he proper maturation and function of tRNAs has emerged METTL2A and 2B recognizes only a subset of arginine tRNA as a critical modulator of biological processes ranging from substrates as well as the biological roles of m3C modification in T 1–3 gene regulation to development . In particular, tRNAs are mammals are unknown. subject to a diverse range of chemical modifications that play Here, we demonstrate that METTL2A and 2B interact with major roles in their folding, stability and function4–6. The critical DALRD3, a previously uncharacterized protein harboring a role of tRNA modification in organismal physiology and fitness is putative anticodon-binding domain found in arginyl tRNA syn- highlighted by the numerous human diseases that have been thetases. Using gene editing, we show that loss of DARLD3 associated with defects in tRNA modification including neurolo- expression in human cells abolishes m3C formation in arginine gical disorders, mitochondrial pathologies, and cancer (reviewed tRNAs that can be rescued with re-expression of full-length in refs. 7–12). DALRD3. Strikingly, we find that DALRD3 is mutated in human The 3-methylcytosine (m3C) modification at position 32 of the patients exhibiting a severe form of epileptic encephalopathy and anticodon loop in certain tRNAs is a eukaryotic-specific mod- that these patients no longer exhibit m3C modification in their ification conserved from yeast to mammals. The m3C modifica- arginine tRNAs. Altogether, this study uncovers an unanticipated tion is predicted to play a role in anticodon folding and function role for the DALRD3 protein in the recognition of specific argi- since the nucleotide at residue 32 forms a non-canonical inter- nine tRNAs for METTL2-catalyzed m3C formation and impli- action with residue 38 to maintain the conformation of the cates the m3C modification in proper neurological function. anticodon loop13–15. In the yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe, the m3C modification is present in tRNA-Thr and tRNA-Ser isoacceptors16–18. Mammals also pos- Results sess m3C in all tRNA-Ser and tRNA-Thr isoacceptors but have METTL2 interacts with a putative tRNA-binding protein also evolved to harbor m3C in mitochondrial-encoded tRNA-Ser- DALRD3. To identify proteins that interact with human MET- UGA and tRNA-Thr-UGU as well as cytoplasmic tRNA-Arg- TL2A or 2B, we generated 293T human embryonic cell lines UCU and CCU17–22. stably expressing METTL2A or METTL2B fused to the Twin- In S. cerevisiae, the Trm140p methyltransferase is responsible Strep purification tag35. As a comparison, we also generated a for m3C formation in tRNA-Ser and Thr isoacceptors23,24. 293T cell line stably expressing Strep-tagged METTL6, a different Interestingly, the fission yeast S. pombe expresses two Trm140 human Trm140 homolog. Strep-METTL2A, METTL2B and homologs encoded by the Trm140 and Trm141 that are METTL6 were affinity purified from whole cell extracts on separately responsible for catalyzing m3C in tRNA-Thr and streptactin resin, eluted with biotin and analyzed by silver tRNA-Ser, respectively17.InS. cerevisiae, Trm140p recognizes staining to detect interacting proteins. We identified a closely- tRNA-Thr substrates via a sequence element encompassing migrating doublet of bands at ~50 kDa along with a band at ~30 nucleotides 35–37 of the anticodon loop that also includes the kDa that were enriched with METTL2A and METTL2B com- t6A modification at position 3725. The recognition of tRNA-Ser pared to the control purification (Fig. 1a, arrowheads). Immu- isoacceptors by yeast Trm140 homologs is also dependent upon noblotting with anti-Strep tag antibodies revealed that one of the modification of position 37, which can be either t6A or i6A 50 kDa bands was purified Strep-METTL2A/B (Supplementary depending on the tRNA-Ser isoacceptor17,25. Notably however, S. Fig. 1). The METTL6 purification yielded bands at ~60, 40, and cerevisiae Trm140p also requires an interaction with seryl-tRNA 30 kDa (Fig. 1a, arrowheads). The 40 kDa band corresponds to synthetase in order to methylate the corresponding seryl-tRNA25. purified Strep-METTL6 as detected by immunoblotting with anti- This utilization of the seryl-tRNA synthetase ensures the proper Strep tag antibodies (Supplementary Fig. 1). catalysis of m3C on all tRNA serine isotypes since the seryl-tRNA To identify METTL-interacting proteins, the total eluates from synthetase has evolved to recognize the unusually long variable each purification were processed for peptide identification by loop and diverse tertiary structure elements present in the various liquid chromatography-tandem mass spectrometry (LC-MS). As tRNA-Ser species26,27. Recent studies in Trypanosoma brucei expected, peptide sequences corresponding to METTL2A, have also uncovered an unusual mechanism by which TRM140 METTL2B and METTL6 were detected in the respective Strep- interacts with the ADAT2/3 deaminase complex to form m3C at METTL2A, 2B and 6 purifications (Fig. 1b). In addition, both the position 32 as a pre-requisite for subsequent deamination by the METTL2A and 2B purifications contained peptides correspond- ADAT2/3 enzyme to form 3-methyluridine28. Collectively, these ing to an uncharacterized protein encoded by the DALR studies highlight the complex circuitry of modifications in the anticodon-binding protein 3 (DALRD3) gene (Fig. 1b, Supple- anticodon loop (reviewed in refs. 29,30). mentary Data 1). The molecular weights of the two predicted Four human homologs of Trm140p have been identified by DALRD3 isoforms are 55 and 59 kDa, which correspond in size , which are encoded by the METTL2A, to the unidentified bands within the doublet observed by silver METTL2B, METTL6, and METTL8 genes17,31. METTL6 is stain. For the METTL6 purification, we detected numerous responsible for the catalysis of m3C at position 32 in tRNA-Ser peptides for seryl-tRNA synthetase (SARS) (Fig. 1b, Supplemen- while METTL8 has been proposed to play a role in mRNA tary Data 1). The molecular weight of SARS (59 kDa) matches modification32. METTL2A and METTL2B encode paralogous closely in size to the 60 kDa band detected by silver stain in the proteins that share 99% amino acid sequence identity and form METTL6 purification. Moreover, SARS was recently identified as their own vertebrate-specific phylogenetic clade with METTL8 an interactor of METTL632. However, no peptides corresponding homologs within the Trm140p homology tree17,23,24. Like S. to DARLD3 were identified in the METTL6 purification. These cerevisiae Trm140p and S. pombe Trm140p, METTL2A and 2B results suggest that DALRD3 forms a unique interaction with are required for the formation of m3C in threonyl-tRNAs32.In METTL2A and 2B. addition, human METTL2A and METTL2B are required for m3C To confirm the DALRD3 interaction and its specificity, we formation in the arginine tRNA isoacceptors, tRNA-Arg-CCU transiently expressed and purified METTL2A, METTL2B, and and Arg-UCU. The presence of m3C in tRNA-Arg-CCU and METTL6 followed by probing with antibodies against DALRD3. Arg-UCU has been detected in multiple mammalian species but Immunoblot analysis revealed the co-purification of endogenous not in yeast or plants17,19,20,33,34, suggesting that METTL2A/B- DALRD3 with Strep-METTL2A or METTL2B that was not catalyzed modification of arginine tRNAs evolved within the readily apparent with Strep-METTL6 (Fig. 1c). As additional animal kingdom. However, the molecular mechanism by which evidence that METTL2A and B interact with DALRD3, we also

2 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 ARTICLE

Input Purification a b Protein Match Coverage Mascot c Bait ID Peptides (%) score METTL2A 24 46.03 1682.75 control METTL2A METTL2B METTL6 METTL2A kDa DALRD3 9 20.99 161.84 METTL6 METTL2A METTL2B METTL6 vector METTL2A METTL2B Strep tag: vector kDa 200 - * * - 50 150 - METTL2A 22 47.62 2266.59 120 - Strep 100 - METTL2B 85 - DALRD3 9 20.81 283.73 70 - 60 - METTL6 21 47.18 3431.44 - 65 50 - METTL6 * SARS 24 52.53 912.08 DALRD3- - 50 40 - 30 - % DALRD3 purified: 0.0 1.0 0.7 0.0 d Protein Match Coverage Mascot 25 - 20 - Bait ID Peptides (%) score e MTS Catalytic Core DALR domain 15 - RARS * * DALRD3 28 57 3100.08 10 - DALRD3 METTL2B 16 36 328.99 RARS2 METTL2A 14 30 439.37 DALRD3

Fig. 1 Human METTL2A/B interacts with DALR anticodon-binding domain containing 3 (DALRD3). a Silver stain analysis of purified Strep-METTL2A, 2B and 6 from human 293T cells. Arrowheads denote bands that are enriched in the METTL2A, 2B and 6 purifications relative to control. Asterisks denote non-specific contaminants found in all three purifications. b Protein matches detected through LC-MS analysis of peptides present in the indicated protein purifications. c Immunoblot analysis of control, METTL2A, METTL2B and METTL6 purifications. The immunoblot was probed with anti-TwinStrep and anti- DALRD3 antibodies. Input represents 1% of total. Asterisk represents non-specific band. Percentage DALRD3 purified represents the fraction of DALRD3 recovered from the total input in each purification. N = 1. d Coverage of the reciprocal purification of DALRD3 in protein samples analyzed by LC-MS. e Schematic of human cytosolic arginyl-tRNA synthetase (RARS), mitochondrial arginyl-tRNA synthetase (RARS2) and DALRD3. Domains are indicated representing mitochondrial targeting signal (MTS), aminoacyl synthetase catalytic core and DALR anticodon-binding domain. Source data are provided as a Source data file. performed a reciprocal purification of DALRD3 from human corroborate the interaction between DALRD3 and METTL2A/B 293T cells stably expressing Strep-tagged DALRD3. Using LC-MS as shown above. analysis, we detected multiple unique peptide matches to We next examined the RNA species that co-purified with METTL2A and 2B in the DALRD3 purification indicative of an DALRD3 by denaturing PAGE followed by nucleic acid staining. association between DALRD3 and endogenous METTL2A/B While no RNAs were enriched in the control purifications, the (Fig. 1d, Supplementary Data 2). These results identify DALRD3 Strep-DALRD3 purification contained several co-purifying RNA as an interacting partner of METTL2A and 2B. species that correspond in size to 5S and 5.8S rRNA along with DALRD3 is an uncharacterized protein containing a carboxy- tRNAs (Fig. 2b, lane 10). Of note, we observed a considerable terminal sequence homologous to the “DALR” anticodon-binding reduction in co-purification of the 5S and 5.8S rRNAs with domain found in arginyl tRNA synthetases from Archaea to DALRD3 when co-expressed with either METTL2A or 2B Eukaryotes36. The DALR domain is named after characteristic (Fig. 2b, compare lane 10 to lanes 11 and 12). However, the co- conserved amino acids present in the primary protein sequence purification of tRNAs with DALRD3 was maintained upon co- and folds into an all alpha helical structure as observed in S. expression with METTL2A or 2B. These results indicate that cerevisiae arginyl tRNA synthetase36–38. In contrast to the DALRD3 interacts with rRNA and tRNAs when over-expressed carboxy-terminus, the amino-terminal portion of DALRD3 and purified alone but shifts to predominantly binding tRNAs contains no recognizable motifs or domains and is specific only when assembled into a complex with METTL2A/B. to DALRD3 homologs. While no DALRD3 homologs have yet To determine the tRNA-binding specificity of DALRD3, we been identified in single-celled eukaryotes or invertebrates, probed the DALRD3 purifications for distinct tRNAs via DALRD3 homologs can be detected in all sequenced vertebrates Northern blotting. Notably, we found that DALRD3 purifications ranging from primitive jawless fishes to mammals39,40. Within were greatly enriched for arginine tRNAs known to contain m3C vertebrates, the canonical DALR domain is found in cytoplasmic generated by METTL2A/B (Fig. 2c, lanes 10–12, tRNA-Arg-CCU arginyl-tRNA synthetase 1 (RARS1) and mitochondrial arginyl- and UCU). In contrast, arginine tRNA isoacceptors lacking m3C tRNA synthetase 2 (RARS2) in addition to DALRD3 (Fig. 1e). exhibited considerably less co-purification with DALRD3 (Fig. 2c, Unlike RARS1 and RARS2, however, DALRD3 lacks a recogniz- lanes 10–12, tRNA-Arg-UCG, CCG, and ACG). Consistent with able tRNA synthetase catalytic motif, thereby suggesting a distinct the predicted specificity of the DALR domain for arginine tRNAs, function for DALRD3 outside of tRNA aminoacylation. only background levels of tRNA-Thr-AGU or Ser-UGA was present in any of the DALRD3 purifications whether co- expressed with or without METTL2A/B (Fig. 2c, Thr-AGU and The METTL2-DALRD3 complex binds distinct arginine Ser-UGA, lanes 10–12). Collectively, these results uncover a tRNAs. Based upon the structure of S. cerevisiae arginyl-tRNA distinct tRNA-binding specificity for DALRD3 that contrasts with synthetase, the DALR domain forms a nucleic acid binding arginyl-tRNA synthetases, which recognizes all arginine tRNA pocket that recognizes the minor groove side of arginine tRNA isoacceptors regardless of the anticodon. anticodon stems through van der Waals, hydrophobic and elec- trostatic interactions37. Thus, we investigated whether DALRD3 DALRD3 facilitates METTL2A/B binding to tRNA-Arg-CCU interacts with tRNAs and if so, whether there was a particular and UCU. The co-purification of DALRD3 with METTL2A and binding specificity for DALRD3. To elucidate the RNA interac- 2B along with the distinct tRNA-binding specificity of tions mediated by DALRD3, we transiently expressed and pur- DALRD3 suggests that DALRD3 plays a role in targeting MET- ified Strep-DALRD3 from 293T cells either alone or with FLAG- TL2A/B to specific arginine tRNAs for m3C modification. To test METTL2A/B (Fig. 2a). Following binding to streptactin resin, we this hypothesis, we transiently expressed Strep-METTL2A or 2B confirmed the purification of Strep-DALRD3 along with the co- with or without co-expression of FLAG-tagged DALRD3 in purification of METTL2A or 2B (Fig. 2a). These results further 293T cells. Immunoblot analysis of the input extracts confirmed

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 3 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6

a Input Strep Purification c Northern blot

Input Strep Purification % yield FLAG: METTL2B vector METTL2A METTL2B – METTL2A vector METTL2A METTL2B – METTL2A METTL2B Strep-D3 – – –+ + + –––+ ++ kDa Strep-D3: - 65 METTL2A vector METTL2A METTL2B METTL2A METTL2B – METTL2A - 50 FLAG: vector METTL2B – METTL2A METTL2B – METTL2B FLAG: Strep-D3: – – – + + + – – – + + + + + +

Load: - 65 Arg-CCU 10 20 20

b Nucleic acid stain 996 Input Strep Purification Arg-UCU

Arg-CCG 0.8 2 2

FLAG: vector METTL2A METTL2B METTL2A vector – METTL2B METTL2A METTL2B – METTL2A METTL2B Arg-ACG 0.8 0.9 1 Strep-D3: –––+ ++ –––+ ++ Arg-UCG 0.2 0.2 0.2

Thr-AGU 000 5.8S - 5S - Ser-UGA 000

tRNA Lane: 123456 789101112 Lane: 123456 789101112

Fig. 2 DALRD3 forms a complex with METTL2A/B to bind distinct arginine tRNAs. a Immunoblot of Strep-DALRD3 purified from 293T cells expressed alone or in conjunction with FLAG-METTL2A/B. Immunoblot was probed with anti-TwinStrep and anti-FLAG antibodies. Load represents a non-specific protein band detected by the anti-Strep antibody used as load control. b Nucleic acid stain of RNAs extracted from the indicated input or purified samples after denaturing PAGE. The migration pattern of 5.8S rRNA (~150 nt), 5S rRNA (~120 nt) and tRNAs (~70–80 nt) are denoted. c Northern blot analysis of the gel in b using the indicated probes. Input represents 2% of total extracts used for purification. The percentage yield represents the amount of RNA in the Strep purification that was recovered from the total input. The experiment was performed three times with comparable results. Quantification was performed on the blot shown. Source data are provided as a Source data file. the expression of Strep-METTL2A/B either alone or with FLAG- UCU but not the other tRNA-Arg isoacceptors nor tRNA-Ser- DALRD3 (Fig. 3a, lanes 2, 3, 5, and 6). Due to the transfection UGA is consistent with the substrate specificity of METTL2A/ procedure, METTL2A and 2B exhibited reduced levels of B32. Remarkably, the amount of tRNA-Arg-CCU or UCU was expression when co-expressed with DALRD3 compared to greatly increased in the METTL2A or 2B purification when co- METTL2A or 2B expressed alone. Using this approach, we expressed with DALRD3 (Fig. 3c, tRNA-Arg-CCU or UCU, detected the co-purification of FLAG-DALRD3 with Strep- compare lanes 8 and 9 with lanes 11 and 12). In contrast, the METTL2A or 2B, further corroborating our finding that MET- interaction between METTL2A or 2B with tRNA-Thr-AGU was TL2A/B interacts with DALRD3 (Fig. 3a, lanes 11 and 12). We disrupted by co-expression with DALRD3 (Fig. 3c, compare lanes also find that the interaction between DALRD3 and METTL2A/B 8 and 9 with lanes 11 and 12). persists after RNase treatment, suggesting that METTL2 inter- The enhancement of METTL2A/B interaction with arginine action with DALRD3 is not bridged through RNA (Supplemen- tRNAs by DALRD3 co-expression provides evidence that tary Fig. 2). assembly of a METTL2-DALRD3 complex facilitates the Analysis of copurifying RNAs revealed the co-purification of recognition and targeting of specific arginine tRNA substrates tRNA with both METTL2A and 2B (Fig. 3b, lanes 8–9 and for m3C modification. The slight co-purification of tRNA-Arg- 11–12). Moreover, there was an increase in the amount of tRNAs CCU or UCU with METTL2A/B when expressed alone may be co-purifying with METTL2A or 2B when each was co-expressed due to a minor population of METTL2A/B forming a complex with DALRD3 (Fig. 3b, compare lanes 8 and 9 with 11 and 12). with endogenous DALRD3, consistent with our original identi- The increase in co-purifying tRNAs with METTL2A/B when co- fication of DALRD3 with METTL2A/B via mass spectrometry. expressed with DALRD3 is not due to an increase in METTL2A/ Moreover, the enrichment for tRNA-Arg-CCU and -UCU B expression or purification since there was actually less combined with the concomitant reduction in tRNA-Thr-AGU METTL2A/B expressed and purified in the presence of DALRD3 binding by METTL2A/B suggests that interaction with DALRD3 (Fig. 3a, lanes 11 and 12). restricts the tRNA-binding specificity of METTL2A/B towards Using Northern blot hybridization, we detected low levels of particular tRNA-Arg isoacceptors. tRNA-Arg-CCU and UCU copurifying with METTL2A or B The tRNAs stably associated with METTL2 and/or DALRD3 alone but not tRNA-Arg-CCG, ACG or UCG (Fig. 3c, lanes 8 and could represent substrates that have not yet been modified with 9). In addition, we found that tRNA-Thr-AGU also copurified m3C and/or have been modified but not dissociated. To ascertain with METTL2A or 2B when expressed alone (Fig. 3c, lanes 8 and the relative m3C modification state of copurifying tRNAs, we 9, Thr-AGU). As a comparison, we detected only background used an oligonucleotide probe that displays differential hybridiza- levels of tRNA-Ser-UGA, tRNA-Gln-UUG or U6 snRNA in any tion based upon m3C modification status (described in further of the purifications (Fig. 3c, lanes 7–12). The interaction of detail below). Using this probe, we found that DALRD3 co- METTL2A and 2B with tRNA-Thr-AGU, Arg-CCU and Arg- purifies with both modified and unmodified tRNA-Arg-UCU and

4 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 ARTICLE

a c Immunoblot Northern blot Input Strep Purification Input Strep Purification

Strep-tag: Strep-tag: vector METTL2A METTL2B – METTL2A METTL2B vector METTL2A METTL2B – METTL2A METTL2B – – vector METTL2A METTL2B METTL2A METTL2B vector METTL2A METTL2B METTL2A METTL2B m3C METTL FLAG-D3: – – – + + + – – – + + + kDa FLAG-D3: –––+ ++ –––+ ++ - 50 Strep-tag: Arg-CCU + 2A/B FLAG-D3: - 65 Loading: - 80 Arg-UCU + 2A/B Lane: 1 2 3 4 5 6 7 8 9 10 11 12 – – b Nucleic acid stain Arg-CCG Input Strep Purification –– Arg-ACG –– Arg-UCG Strep-tag: vector METTL2A METTL2B – METTL2A METTL2B vector METTL2A METTL2B – METTL2A METTL2B FLAG-D3: –––+ ++ –––+ ++ Thr-AGU + 2A/B

Ser-UGA + 6

5.8S - Gln-UUG – – 5S - U6 – – tRNA Lane: 123456 789101112 Lane: 123456789101112

Fig. 3 DALRD3 mediates the binding of METTL2A/B to tRNA-Arg-CCU and UCU. a Immunoblot analysis of the purification of Strep-METTL2A/B expressed alone or with FLAG-DALRD3 from 293T human embryonic cells. The immunoblot was probed with anti-TwinStrep and anti-FLAG antibodies. Load represents a non-specific protein band detected by the anti-Strep antibody used as load control. Input represents 2% of total. b Nucleic acid stain of RNAs extracted from the indicated input or purified samples after denaturing PAGE. The migration pattern of tRNAs (70–80 nt), 5.8S rRNA (~150 nt), and 5 S rRNA (~120 nt) are denoted. c Northern blot analysis of METTL2A/B-associated RNAs with the indicated probes. The known presence or absence of m3C and the METTL enzyme that generates m3C in a given tRNA is denoted on the right. The purification was repeated three times with similar results. Source data are provided as a Source data file.

Arg-CCU (Supplementary Fig. 3a). In contrast, we found that the the possibility that DALRD3 recognizes specific arginine tRNAs, majority of tRNA-Arg-UCU and Arg-CCU that copurified with in part, through sequence elements in the anticodon loop that METTL2 was modified with m3C (Supplementary Fig. 3b). These include positions 36 and 37 to facilitate METTL2A-dependent results are consistent with DALRD3 protein binding unmodified methylation. tRNA-Arg-UCU or CCU for subsequent methylation upon To test this hypothesis, we probed the methyltransferase interaction with METTL2. activity of METTL2-DALRD3 complexes purified from human cells on in vitro transcribed tRNA-Arg-CCU substrates. For tRNA sequence requirements for methylation by METTL2- monitoring m3C formation, we used a primer extension assay in DALRD3. Mammalian genomes express five different tRNA which the presence of m3C leads to a reverse transcriptase (RT) isoacceptors that decode arginine codons but only tRNA-Arg- block at position 32 while the lack of m3C allows for read- CCU and Arg-UCU are modified to contain m3C at position 32 through and generation of an extended product. As a control to (Fig. 4a)17,20,41. Intriguingly, tRNA-Arg-UCG and Arg-ACG ensure that methylation was occurring at the correct position, we also contain C32 but are not modified by METTL2A/B in either tested a tRNA-Arg-CCU substrate in which C32 was mutated to human or mouse cells. Inspection of the isoacceptor stem A (C32A). To probe the requirement for positions 36 and 37 in loops reveals that tRNA-Arg-CCU and Arg-UCU contain U-A at METTL2-DARLD3 methylation, we generated the tRNA-Arg- positions 36 and 37 while tRNA-Arg-CCG, UCG, and ACG contain CCU substrates U36G and A37G (Fig. 4b). G-G at positions 36 and 37 (Fig. 4a).WealsonotethattRNA-Thr- As a positive control, we performed primer extension on RNA AGU, which is a substrate of METTL2A/B, also contains a U-A harvested from human 293T cells which results in an extension dinucleotide at positions 36 and 37 (Fig. 4a, Thr-AGU). Moreover, stop at position 32 of tRNA-Arg-CCU indicative of the m3C fi A37 is modified to N6-threonylcarbamoyladenosine (t6A) in modi cation that was absent when no RT was added (Fig. 4c, tRNA-Arg-CCU and Arg-UCU along with tRNA-Thr-AGU. Pre- lanes 1 and 2). No RT block at position 32 was detected for vious studies in S. cerevisiae and S. pombe have shown that the in vitro transcribed tRNA-Arg-CCU when pre-incubated with fi identity of residues in the anticodon loop along with the i6A/t6A either water or negative control puri cation (Fig. 4c, lanes 3 and modification at position 37 play key roles in the recognition 4). In contrast, pre-incubation of tRNA-Arg-CCU with either fi and modification of seryl- and threonyl-tRNAs in m3C formation puri ed METTL2A or 2B with or without DALRD3, followed by by the yeast Trm140 enzyme17,25. Combined with the tRNA primer extension, revealed the appearance of a RT block at – interaction studies described above, these observations suggest position 32 indicative of m3C formation (Fig. 4c, lanes 5 8). We

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 5 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6

a human tRNA-Arg tRNA-Thr CCG UCG ACG CCU UCU AGU CG CG CG UA UA CG U A U A U A G C G C U A G C G C G C G C G U G C A U A U A U C G A U U A U32 A C32 A C32 A m3C A m3C A m3C A U m1G U m1G U m1G U32 t6A U32 t6A U32 t6A C C G U C G I C G C C U U C U A G U 36 36 36 36 mcm5s2 36 36 b c Arg-CCU 293T tRNA-Arg-CCU RNA UA UA UA UA G C G C G C G C Purified G C G C G C G C protein: H2O H2O H2O METTL2A METTL2B control C G A C G C G C G METTL2A METTL2B C32A C 32A C 32A C 32 A DALRD3: –– –– –– + + UA UAUA UA RT: –+ + + + + + + C C U C C U C C U C C U G 36 3636 G 36 5’-end> C32 C32A U36G A37G

d tRNA-Arg-CCU

C32C32A U36G A37G m3C>

primer> Purified protein: 123 45 678 Buffer Buffer METTL2A METTL2A METTL2A METTL2A Buffer METTL2A METTL2A METTL2A DALRD3: + – – + – – + – – + e ** 100 ** 5’-end> 80 **

60

40 m3C>

% m3C formation 20 primer> 0 1236710 45 8 9 WT C32A U36G A37G tRNA-Arg-CCU

Fig. 4 The identity of residues 36 and 37 in tRNA-Arg-CCU play a key role in m3C formation by METTL2A/2B in vitro. a Anticodon loops of human tRNA-Arginine isoacceptors and known modifications. b Anticodon loops of in vitro transcribed tRNA-Arg-CCU and variants used in c, d. c, d Primer extension analysis of tRNA-Arg-CCU and variants incubated with either water, vector control eluate, purified METTL2A or METTL2B co-expressed with DALRD3. Primer length, 23 nt. e Quantification of m3C formation in d. Primer extension analysis was performed greater than three times for c and repeated three times for d. Bars represent the standard deviation from the mean. Statistical analysis was performed using one-way ANOVA and significance calculated using Tukey’s multiple comparison test. **P < 0.01. P = 0.0034 for WT versus C32A, 0.0039 for WT versus U36G, 0.0037 for WT versus A37G. Source data are provided as a Source data file. note that METTL2A/B purified from human cells without recognition and interaction of specific arginine tRNAs with overexpression of DALRD3 exhibited m3C modification activity, METTL2A/B. To investigate the requirement for DALRD3 in consistent with the co-purification of endogenous DALRD3 m3C formation in vivo, we engineered human cell lines lacking with the over-expressed METTL2A or 2B to form an active DALRD3 using CRISPR/Cas9 gene editing. Using the HAP1 methyltransferase complex. Thus, the purified METTL2-DALRD3 human haploid cell line42, we generated a cell clone containing a complex is active for m3C formation on an in vitro transcribed 14 base-pair deletion in exon 1 of the DALRD3 gene (Supple- tRNA substrate lacking any other modifications. mentary Fig. 4a). The deletion is predicted to produce a truncated Using the tRNA-Arg-CCU variants, we detected no RT block DALRD3 missing the majority of the polypeptide or a translation when C32 was mutated to A, providing evidence that position 32 frameshift that results in nonsense mediated decay (NMD). was the site of methylation (Fig. 4d, lanes 1–4). Notably, mutation Indeed, immunoblotting revealed the absence of full-length of either U36 or A37 to a G residue in tRNA-Arg-CCU led to a DALRD3 protein in the DALRD3-knockout (D3-KO) cell line major decrease in m3C formation (Fig. 4d, lanes 5–10, quantified compared to the isogenic control wildtype (WT) cell line in 4e). These studies provide evidence that DALRD3 serves as a (Fig. 5a). While expression of DALRD3 was abolished in the D3- discrimination factor to recognize distinct arginine tRNAs based KO cell line, no change in METTL2A levels was detected between upon sequence elements common between tRNA-Arg-CCU and the WT or D3-KO cell lines (Supplementary Fig. 4b). tRNA-Arg-UCU. To monitor m3C formation in tRNA, we used the positive hybridization in the absence of modification (PHA) assay (Fig. 5b)43–46. This Northern blot-based assay relies on m3C formation in cellular arginine tRNAs requires DALRD3. differential probe hybridization to tRNA caused by the presence – The above results suggest that DALRD3 functions in the or absence of m3C, which impairs base-pairing47 49. Thus, a

6 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 ARTICLE

abcNorthern blot - PHA assay HAP1 cell line tRNA: Arg-CCU Arg-UCU Thr-AGU Ser-UGA

DALRD3: WT KO kDa D3: WT KO WT KO WT KO WT KO - 70 * PHA DALRD3 * PHA Control probe probe actin - 40 Control = m3C

d tRNA: Arg-CCU Arg-UCU e Ser-UGA Thr-AGU

Cell 293T HAP1-WT 293T 293T HAP1-D3KO 293T 293T 293T HAP1-WT HAP1-D3KO 293T HAP1-WT HAP1-D3KO 293T HAP1-D3KO line: HAP1-WT RT: – + + + RT: – + + + RT: – + + + RT: – + + + D - D16- 16 D20- D20- m3C m3C32 - 32 - t6A37- m3C - 32 t6A37- m3C32 - > > > >

f h i **** DALR tRNA-Arg-CCU **** WT ns HAP1: WT D3KO ΔDALR **** ΔN-term 100 DALRD3 80 construct: DALR N-term vector g Immunoblot: anti-FLAG vector WT Δ Δ

HAP1: WT D3KO D17- 60

40 DALRD3 D20- DALR N-term construct: % m3C formation vector WT vector WT Δ Δ kDa 20 - 70 WT - m3C - 32 0 ΔDALR - - 50

DALRD3 WT DALR > construct: vector vector N-term ΔN-term - - 30 Δ Δ actin - - 40 1 2 3 4 5 Strain: WT D3-KO

Fig. 5 DALRD3 is required for efficient m3C formation in arginine tRNAs in vivo. a Immunoblot verification for the loss of DALRD3 expression in human DALRD3-knockout (KO) cell lines compared to wild-type human HAP1 cells. Actin was used as a loading control. Asterisk denotes a non-specific band found in both wild-type and D3-KO cell lysates. b Schematic of the Positive Hybridization in the Absence of Modification (PHA) assay. c Northern blot analysis of PHA probes designed to detect m3C at position 32 and a control probe that hybridizes to a different area of the same tRNA. d Primer extension analysis of the tRNA-Arg-UCU and Arg-CCU harvested from the denoted human HAP1 cell lines. Length of primers; tRNA-Arg-UCU, 24 nt; tRNA-Arg- CCU, 23 nt. e Primer extension analysis of the tRNA-Ser-UGA and Thr-AGU harvested from the denoted human HAP1 cell lines. Length of primers; tRNA- Ser-UGA, 23 nt; tRNA-Thr-AGU, 20 nt. f Schematic of DALRD3 variants used for DALRD3 rescue experiments. g Immunoblot analysis confirming expression of DALRD3 variants in the indicated HAP1 cell lines. h Primer extension analysis of tRNA-Arg-CCU from WT or D3KO HAP1 cell lines stable integrated with the indicated DALRD3 expression constructs. (-RT) represents no reverse transcriptase was added; m3C- 3-Methylcytidine; D- dihydrouridine; t6A- threonylcarbamoyladenosine; > labelled probe. i Quantification of m3C formation in tRNA-Arg-CCU by primer extension. n = 3. Error bars represent standard deviation from the mean. Statistical analysis was performed using one-way ANOVA and significance calculated using Tukey’s multiple comparison test. ****P < 0.0001; ns, non-significant. P = 0.7430 for WT strain + vector versus D3-KO strain + WT-DALRD3. a,(c through e), g, h were repeated three times each with similar results. Source data are provided as a Source data file. decrease in m3C modification leads to an increase in PHA probe detected for either tRNA-Ser-UGA or tRNA-Thr-AGU in the D3- signal that can be normalized against the probe signal from a KO cell line, consistent with the substrate specificity of different region of the same tRNA as an internal control. For DALRD3 shown above. We also note that the steady-state levels tRNA-Arg-CCU and UCU, we observed a considerable increase of all tested tRNAs remained similar between the WT and D3-KO in PHA probe signal in the human D3-KO cell line compared to cell lines, including tRNA-Arg-CCU and UCU. WT, indicating the loss of m3C modification in these particular As additional validation, we monitored m3C formation in tRNAs (Fig. 5c). In contrast, no increase in PHA signal was specific cellular tRNAs using the primer extension assay. As

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 7 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 expected, an RT stop indicative of m3C at position 32 of tRNA- to uncharacterized patients with neurodevelopmental disorders50. Arg-CCU and Arg-UCU was detected in wildtype human 293T Parents are healthy first cousins and they have one healthy child and HAP1 cells (Fig. 5d). In contrast, the m3C block in both in addition to the two affected children and one spontaneous tRNA-Arg-CCU and Arg-UCU was greatly reduced in the D3- miscarriage (Fig. 6b). Subsequent exome sequencing guided by KO cell line with subsequent readthrough to the next RT- autozygome analysis51 revealed a single homozygous nucleotide blocking modification (Fig. 5d, HAP1-D3KO lanes). Consistent substitution resulting in a C to A transversion in exon 9 of the with a role for DALRD3 only in arginine tRNA modification, the DALRD3 gene in a shared region of autozygosity between the two m3C modification in tRNA-Ser-UGA and Thr-AGU was affected siblings on 3 (Fig. 6c). Confirmatory Sanger unaffected in the D3-KO cell line (Fig. 5e). These results sequencing validated the full segregation of this variant with the demonstrate that DALRD3 is required for efficient m3C disease in the family in a fully penetrant autosomal-recessive formation in specific arginine tRNAs. model, being homozygous in both patients and heterozygous in To dissect the regions of DALRD3 that play a role in m3C the parents and the healthy sibling (Fig. 6d). The genetic altera- formation, we tested the ability of DALRD3 variants to rescue tion is expected to cause a nonsense mutation due to the intro- m3C formation in the HAP1-D3KO cell line. We generated stable duction of a UAA stop codon within the mRNA transcript cell lines expressing either wildtype DALRD3 or DALRD3 encoding the predominant isoform of DALRD3 (pTyr417*). variants lacking either the DALR domain or the N-terminal Translation of this transcript will result in a truncated protein extension (Fig. 5f, WT, ΔDALR, and ΔNterm). The expression of lacking the DALR tRNA anticodon-binding domain. In addition, each DALRD3 variant in the stable cell lines was confirmed by the variant DALRD3 mRNA could be subject to NMD due to the immunoblotting (Fig. 5g). As expected, HAP1-D3KO cells with presence of a premature stop codon. vector alone exhibited no m3C stop at position 32 in tRNA-Arg- To determine the molecular impact of the DALRD3 mutation CCU when compared to HAP1-WT cells (Fig. 5h, compare lanes on m3C formation, we generated lymphoblastoid cell lines (LCLs) 1 and 2). Re-expression of wildtype DALRD3 in HAP1-D3KO from the two sibling patients (referred to as D3-LCLs generated cells was able to rescue m3C formation in tRNA-Arg-CCU as from patients 1 and 2; P1 and P2). LCLs generated from the evidenced by the re-introduction of the m3C RT block and the affected patients were compared to control lymphoblasts from absence of read-through products (Fig. 5h, lane 3). In contrast, two ethnically matched, healthy, unrelated individuals (LCL-WT1 expression of the DALRD3 mutant lacking the DALR domain or and WT2). Consistent with the nonsense mutation leading to a the N-terminal extension was unable to restore m3C formation in truncation of the DALRD3 protein and/or NMD, the levels of tRNA-Arg-CCU (Fig. 5h, lanes 4 and 5; quantified in 5i). full-length DALRD3 protein were decreased in cell lysates Altogether, these results demonstrate that m3C formation at prepared from either patient LCLs compared to WT-LCLs position 32 in specific arginine tRNAs requires the DALRD3 (Fig. 7a). Using the aforementioned PHA assay, we detected a protein with both the DALR domain and N-terminal region substantial increase in PHA signal for tRNA-Arg-CCU and UCU contributing key roles in methyltransferase activity. in D3-LCLs compared to WT control cells, indicating the loss of m3C modification in these particular tRNAs (Fig. 7b). Follow up investigation using the primer extension assay confirmed the Impact of m3C modification on codon reporter expression. severe reduction of m3C modification in tRNA-Arg-CCU and The interaction between nucleotides 32 and 38 of the tRNA UCU of both D3-LCLs from affected patients (Fig. 7c, d). In anticodon loop is known to impact codon recognition and tRNA contrast, the levels of m3C modification detected by PHA probe binding to the ribosome in Escherichia coli15. Thus, we investi- hybridization or primer extension for tRNA-Ser-UGA and tRNA- gated the possible effects of m3C modification on tRNA-Arg 32 Thr-AGU was similar between D3- and WT control-LCLs function in protein synthesis using codon-dependent reporter (Fig. 7b–d). The deficit in m3C modification in the arginine constructs. We generated lentiviral reporters expressing nanolu- tRNAs of both patient LCLs demonstrates that the DALRD3 ciferase protein fused downstream of ten consecutive AGA or mutation represents a partial loss-of-function allele. AGG codons, which are decoded by m3C-containing tRNA-Arg- Epileptic encephalopathies (EEs) encompass a large, hetero- UCU or CCU (Supplementary Fig. 5a). We also generated genous group of early-onset neurodevelopmental disorders reporter plasmids containing codon runs of CGA or CGC, which characterized by intractable seizures and electroencephalogram are decoded by tRNA-Arg-UCG or ACG lacking m3C. Based abnormalities (reviewed in refs. 52,53). In both patients harboring upon this system, we found no significant difference in protein the DALRD3 mutation, the onset of seizures occurred early in expression between the WT or D3-KO cell lines for any of the infancy at 6 to 7 months of age indicative of early-onset epileptic arginine codon reporters (Supplementary Fig. 5b and c). We also encephalopathy (Table 1). While the epilepsy of patient 2 could be detected no major change in expression of a control GFP con- controlled by anti-epileptic medications, the seizures in patient 1 struct between WT and D3-KO cell lines (Supplementary Fig. 5b were poorly controlled by medication. In addition to develop- and c). These results suggest that the m3C modification has mental delay and epilepsy, both patients exhibited a range of minimal effect on translation efficiency in these codon reporters neurological and physiological symptoms consisting of severe and/or plays a role in arginine tRNAs that cannot be detected motor and speech phenotypes, hypotonia and facial dysmorphia using this experimental system. (Table 1). Based upon the constellation of symptoms characterized by the co-occurrence of developmental delay and frequent epileptic A DALRD3 variant linked to a developmental brain disorder. activity, both patients match clinical conditions now classified as Due to the emerging links between tRNA modification and developmental and epileptic encephalopathy54. Altogether, these neurodevelopmental processes, we next investigated whether findings identify human individuals who lack m3C modification in DALRD3 is associated with any neurological disorders of arginine tRNAs and reveal a biological link between m3C tRNA unknown genetic etiology. We identified a consanguineous family modification and proper neurological function. with two sibling patients exhibiting profound global develop- mental delay and persistent early infantile epilepsy who tested negative for known disease mutations by routine karyotype, Discussion chromosomal microarray, and an epilepsy gene panel (Fig. 6a). Here, we show that human METTL2A/B forms a complex with The family was identified by applying a “genomics first” approach the DALRD3 protein to catalyze the formation of m3C in specific

8 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 ARTICLE

a b d p.(Tyr417*) +NH DALR COO– Father Mother 3 DALRD3 (NM_001009996.3) 5’- -3’ *

P1 P2 P1 P2 19DG0509 19DG0510 19DG0509 19DG0510 P1

chr 22 c chr 21 19DG0509 chr 20 DALRD3

chr 19 chr 1

chr 18 P2 chr 2 chr 17 19DG0510

chr 16

chr 3 Unaffected chr 15 brother

chr 14 chr 4 Mother

chr 13

chr 5 Father chr 12

chr 6

chr 11 Wildtype chr 7

chr 10

chr 8

chr 9

Fig. 6 Identification of a DALRD3 variant linked to developmental delay and epileptic encephalopathy. a Pedigree of the family harboring a nonsense mutation in the DALRD3 gene. b Patients 1 and 2 (P1 and P2) containing the homozygous DALRD3 mutation. c Agile MultiIdeogram output of autozygosity analysis of the study family as a series of block arcs with the two affected individuals represented by the outer two arcs. Autozygous regions from affected individuals are marked as pale blue, those from unaffected individuals as pink, and those homozygous in all affected individuals as dark blue. d Sanger sequencing chromatograms of the indicated individuals from the family in this study along with a wildtype, control individual. arginine tRNAs. Our findings explain how METTL2A/B recog- recognize and modify different seryl-tRNA isoacceptors in nizes particular arginine tRNAs for m3C modification through S. cerevisiae, the tRNA-binding specificity of DALRD3 limits the unique tRNA interaction specificity of DALRD3 protein. METTL2A/B activity to only tRNA-Arg-UCU and CCU. The Notably, we have identified human individuals with autosomal- need to discriminate between the different arginine tRNA iso- recessive nonsense mutations in the DALRD3 gene who lack m3C acceptors could explain why mammals evolved DALRD3 to in their arginine tRNAs. These individuals exhibit a disorder interact with METTL2 rather than arginyl-tRNA synthetase. characterized by severe developmental delay and early-onset Another possibility is that METTL2 contributes primarily to epileptic encephalopathy, thereby linking the m3C modification substrate specificity while DALRD3 serves as an activator of in mammalian arginine tRNAs with proper neurological function. METTL2 methyltransferase activity. The METTL2-DALRD3 complex presents yet another case of a An outstanding question that remains is why the m3C mod- multi-subunit enzyme catalyzing tRNA modification. The emer- ification is present only in a subset of arginine tRNAs. One ging number of multimeric tRNA modification enzymes has been possibility is that tRNA-Arg-UCU and Arg-CCU possess addi- hypothesized to be driven by the need to recognize and modify tional requirements for folding and/or stability that are distinct different substrates while maintaining high specificity55,56. In one from the remaining tRNA-Arg isoacceptors due to differences in pertinent example, the activity of S. cerevisiae Trm140p on seryl- anticodon structure. In addition, the m3C modification in tRNA- tRNA substrates is greatly stimulated by interaction with seryl- Arg-UCU and CCU could be required for their proper aminoa- tRNA synthetase, which can recognize the full cellular repertoire cylation and/or interaction with the ribosome during translation. of seryl-tRNAs even though their anticodons do not have any Interestingly though, DALRD3-deficient human cells exhibited nucleotides in common25. Human METTL6 also interacts with no major change in the expression of reporter proteins fused to a seryl-tRNA synthetase32, providing further evidence that run of AGA or AGG codons, both of which are decoded by Trm140p homologs have evolved to bind additional protein arginine tRNAs containing m3C. However, the arginine codon cofactors in order to efficiently recognize their substrates. The reporter system represents a highly specific context that might interaction between human METTL2A/B and DALRD3 is ana- not reflect the actual physiological impact of the m3C modifica- logous to the interaction of S. cerevisiae Trm140p with seryl- tion on tRNA function in translation in different tissues. Future tRNA synthetase in that a tRNA methyltransferase interacts with studies using ribosome profiling approaches in different a known or predicted tRNA-binding protein in order to catalyze DALRD3-deficient cell types could reveal the mRNA transcripts tRNA modification. However, unlike the interaction of seryl- that are dependent upon m3C modification for proper translation tRNA synthetase with Trm140p, which allows Trm140p to and protein expression.

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 9 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6

a c LCL: WT1 WT2 P1 P2 kDa tRNA: Arg-CCU Arg-UCU * - 70 DALRD3 * LCL: LCL: WT1 WT1 WT2 P1 P2 WT1 WT1 WT2 P1 P2 actin - 40 RT: – + + + + RT: – + + + + D - D16 - b Northern blot - PHA assay 16 D20 - D - tRNA: Arg-CCU Arg-UCU 20 m3C32 - t6A37 - LCL: m3C32 - WT1 WT2 P1 P2 WT1 WT2 P1 P2

PHA > > Control

tRNA: tRNA-Ser-UGA tRNA-Thr-AGU tRNA: Ser-UGA Thr-AGU LCL: WT1 WT1 WT2 P1 P2 WT1 WT1 WT2 P1 P2 LCL: RT: – + + + + – + + + + WT1 WT2 P1 P2 WT1 WT2 P1 P2

PHA m3C32 - - m3C32 - t6A Control > 37 <

d tRNA: Arg-CCU Arg-UCU Ser-UGA Thr-AGU 100 100 100 100

80 80 80 80

60 60 60 60

40 40 40 40 % m3C formation % m3C formation % m3C formation 20 20 20 % m3C formation 20

0 0 0 0 P1 P2 P1 P2 P1 P2 LCL: P1 P2 WT1 WT2 WT1 WT2 WT1 WT2 WT1 WT2 Fig. 7 Lymphoblastoid cell lines derived from patients with the homozygous DALRD3 variant exhibit deficiency in m3C modification in arginine tRNAs. a Immunoblot for the indicated proteins from lysates prepared from LCLs donated from wildtype (WT) individuals and patients P1 and P2 harboring the homozygous DALRD3 mutation. Asterisks (*) represent non-specific bands on either side of the DARLD3 protein. Blot was repeated twice with similar results. b Northern blot analysis using PHA probes designed to detect m3C at position 32 and a control probe that hybridizes to a different area of the same tRNA. Blot was repeated three times with comparable results. c Primer extension analysis of the indicated tRNAs from the denoted human LCLs. (-RT) represents no reverse transcriptase; m3C 3-Methylcytidine; D dihydrouridine; t6A threonylcarbamoyladenosine. > labelled probe. Primer extension analysis of lymphoblastoid cell lines was repeated three times. d Quantification of m3C formation by primer extension for the indicated tRNAs. n = 2. Source data are provided as a Source data file.

Notably, the appearance of DALRD3 in the vertebrate lineage The identification of a DALRD3 mutation that abolishes m3C coincides with the emergence of METTL2 homologs as well as the formation in human patients with developmental delay and epi- m3C modification in vertebrate arginine tRNAs. An intriguing leptic encephalopathy has uncovered a physiological link between possibility is that DALRD3 arose in vertebrates through a duplica- the m3C modification and neurological function. This finding is tion event from an extant arginyl-tRNA synthetase gene that lost intriguing because the majority of tRNA modification defects that catalytic activity but retained tRNA binding through the DALR are known to cause epileptic encephalopathies are due to altera- domain. DALRD3 could also have resulted from the insertion of the tions in mitochondrial function. For example, loss of the i6A59,60 DALR domain coding sequence downstream of a pre-existing or wobble taurine modification61–64 in mitochondrial tRNAs protein-coding gene to generate a vertebrate-specific fusion protein causes abnormal mitochondrial protein synthesis and defects in with a unique domain architecture. In either case, the utilization of mitochondrial respiratory chain complexes that lead to develop- the DALR anticodon-binding domain for tRNA modification mental delay and epileptic encephalopathy. While there are no highlights the modular nature of motifs linked to aminoacyl syn- known mitochondrial tRNAs dependent upon DALRD3 for m3C thetases and the expansion of functions from homologous formation, there could be a possible indirect impact of DALRD3- synthetase predecessors57,58. Future studies will determine if dependent tRNA modification on mitochondrial homeostasis DALRD3 serves additional roles besides tRNA modification in since cytoplasmic tRNA modifications in the anticodon loop have vertebrates. For example, DALRD3 may interact with additional been shown to impact mitochondrial metabolism43. The loss of RNAs besides tRNAs to modulate their modification status or sta- arginine tRNA modification due to the DALRD3 mutation pro- bility. We also note that m3C at position 47:3 of the variable arm is vides an avenue to explore additional biological pathways that are present in mammalian tRNA-Ser isoacceptors but not S. cerevisiae key to proper neurological function and perturbed in neurological tRNAs17, suggesting the possibility of another mammalian-specific disorders. Future experiments will investigate the role of tRNA methyltransferase complex that remains to be identified. DALRD3 and its associated tRNA modifications in protein

10 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 ARTICLE

Table 1 Clinical phenotype and molecular findings for patients with homozygous variants in DALRD3.

Patient 1 Patient 2 ID 19DG0509 19DG0510 Gender Male Female Date of birth 17/04/2009 05/01/2013 Genotype NM_001276405.1:c.1251 C > A: NM_001276405.1:c.1251 C > A: Protein p.(Tyr417*) p.(Tyr417*) Perinatal history Normal spontaneous vaginal delivery with history of placental Full term product of cesarean section due to breech insufficiency and oligohydramnios presentation and oligohydramnios in addition to placental insufficiency Weight at birth (kg) 2.25 (−2.2 SD) 2.5 (4th centile) Developmental delay Severe Severe Motor Immobile Immobile Speech Nonverbal Nonverbal Seizures Seizures started at age 7 months in the form of myoclonic At age 6 months epilepsy ensued, initially as brief episodes of jerks which remains frequent and poorly controlled by flexion tonic spasm of head followed by myoclonic seizures. antiepileptic medications Unlike the sibling brother, the epilepsy of patient 2 is reasonably controlled by antiepileptic medications. EEG Independent multifocal epileptic discharges predominantly Markedly high voltage and slow background for age along over the anterior head region bilaterally as well as over the with slow generalized polyspike and wave activity right temporal and right parietal regions Tone Axial and peripheral hypotonia with dystonic like movement Central and peripheral hypotonia with dystonic like and generalized muscle wasting movements and generalized muscle wasting Microcephaly No Yes Brain finding Mild diffuse brain parenchymal volume loss with diffuse Normal topographical and morphological appearance of the paucity of the myelin within the brain parenchyma. infratentorial and supratentorial structures Audiology assessment Moderate to severe conductive hearing loss in the left ear and N/A mild conductive hearing loss in right ear Dysmorphism Subtle facial dysmorphia and small left ear Microcephaly with subtle facial dysmorphia Other Severe gastroesophageal reflux disease necessitating GT tube Vomiting and chocking on first day of life, mild congenital placement and fundoplication at age 4 years, no visual heart disease that resolved spontaneously, ectopic right tracking or social smile kidney, bilateral optic disc pallor translation to identify cellular pathways that would be affected by by EBV immortalization of primary human lymphocytes obtained from patient the m3C modification and their roles in cell physiology and blood samples. 293T human embryonic kidney cell lines were cultured in Dul- ’ development. An increased understanding of the genetic and becco s Minimal Essential Medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 1X penicillin and streptomycin (ThermoFisher), and 1X Glutamax molecular underpinnings of early-onset epileptic encephalo- (Gibco) at 37 °C with 5% CO2. Cells were passaged every 3 days with 0.25% pathies will be especially vital since many types of these epilepsies Trypsin. Human HAP1 DALRD3 knock-out cell lines were generated by CRISPR/ are resistant to standard conventional epilepsy therapy65. Cas9 mutagenesis (Horizons Discovery Life Sciences). Human HAP1 cell lines were cultured in Iscove’s Modified Dulbecco’s Medium (IMDM) supplemented with 10% FBS and 1X penicillin and streptomycin at 37 °C with 5% CO2. Cells were Methods passaged with 0.05% Trypsin. Human lymphoblastoid cell lines were cultured in Plasmids. The open reading frame (ORF) for METTL2A and METTL2B was RT- Roswell Park Memorial Institute (RPMI) 1640 Medium containing 15% fetal fi PCR ampli ed from HeLa human cervical carcinoma cell cDNA, cloned by bovine serum, 2 mM L-alanyl-L-glutamine (GlutaMax, Gibco) and 1% Penicillin/ restriction digest and verified by Sanger sequencing. The cDNA clones for MET- Streptomycin. TL2A and METTL2B correspond to NM_181725.3 and NM_018396.2, respec- For generation of stable cell lines, 2.5 × 105 293T cells were seeded onto 60 × 15 tively. The ORF for DALRD3 was PCR amplified from cDNA plasmid mm tissue culture dishes. In all, 1.25 μg of pLKO.DEST.puro plasmids containing HsCD00338640 (Plasmid Repository, Harvard Medical School) and cloned into the cloned ORF of METTL2A, 2B, 6, DALRD3, and DALRD3 truncations or empty either pcDNA3.1-TWIN-Strep or pcDNA3.1-3xFLAG. Truncation constructs were vector along with a lentiviral packaging cocktail containing 0.75 μg of psPAX2 PCR amplified and cloned from the ORF of METTL2A and DALRD3 and cloned packaging plasmid and 0.5 μg of pMD2.G envelope plasmid was transfected into into pcDNA3.1-3xFLAG for METTL2A truncation constructs and DALRD3 HEK293T cells using calcium phosphate transfection. In all, 48 hours after truncation constructs. Primers used are listed in Supplementary Table 1. transfection, media containing virus was collected and filtered sterilized through For lentiviral expression constructs, Strep tagged-METTL2A, 2B and DALRD3 0.45 μm filters and flash frozen in 1 ml aliquots. Lentivirus for the pLJM-EGFP or as well as 3x-FLAG-tagged DALRD3 and all truncation constructs of DALRD3 codon reporter plasmids were generated in an identical manner. were first cloned into the pENTR.CMV.ON plasmid using NheI and NotI For lentiviral infection in 293T or HAP1 cell lines, 2.5 × 105 cells were seeded in restriction sites. The entry vectors were recombined via LR clonase reaction six-well plates or 6 × 104 cells in 24-well plates for HAP1 cells infected with the (ThermoFisher) into the pLKO.DEST.puro destination vector to allow for stable NLuc reporter plasmid. 24 hours after initial seeding, 1 ml of virus (or media for integration of the target genes into human cell lines. mock infection) along with 2 ml of media supplemented with 10 μg/ml of For the arginine codon reporter assay, a synthetic gene encoding the polybrene was added to each well. The cells were washed with PBS and fed fresh NanoLuc luciferase protein fused to an N-terminal run of 10X arginine codons media 24 h post-infection. Puromycin selection begun 48 h after infection at a of either AGA, AGG, CGC or CGA was generated by in vitro gene synthesis concentration of 2 μg/ml. Fresh media supplemented with puromycin was added (GenScript). The synthetic codon reporter gene was subcloned into pcDNA3.1 every other day and continued until the mock infection had no observable living and sequence verified. The reporter cassette containing the 10xArg codon- cells. Proper integration and expression of each construct was verified via NanoLuc was then cloned into the pLJM1-EGFP lentiviral plasmid (Sabatini immunoblotting. Lab, Addgene #19319) using NheI-EcoRI to replace the EGFP sequence. The resulting plasmid encodes the luciferase reporter protein expressed from the CMV promoter. Transient transfections and protein-RNA purifications. 293T cells were trans- fected via calcium phosphate transfection method66. Briefly, 2.5 × 106 cells were Tissue cell culture and generation of stable cell lines. The 293T human seeded on 100 × 20 mm tissue culture grade plates (Corning) followed by trans- embryonic cell line was originally obtained from ATCC (CRL-3216). The HAP1 fection with 10–20 μg of plasmid DNA. Cells were harvested 48 h later by trypsin Human Male Chronic Myelogenous Leukemia (CML) cell line was obtained from and neutralization with media, followed by centrifugation of the cells at 700 × g for Horizon Discovery Life Sciences. Human lymphoblastoid cell lines were generated 5 min followed by subsequent PBS wash and a second centrifugation step.

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 11 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6

Protein was extracted by the Hypotonic Lysis protocol immediately after cells human database that was downloaded in December of 2015. Trypsin was selected were harvested post-transfection. Cell pellets were resuspended in 0.5 μlofa as the enzyme allowing up to two missed cleavages, with an MS1 mass tolerance of hypotonic lysis buffer (20 mM HEPES pH 7.9, 2 mM MgCl2, 0.2 mM EGTA, 10% 10 ppm, and an MS2 mass tolerance of 25 mmu. Carbamidomethyl was set as a glycerol, 0.1 mM PMSF, 1 mM DTT) per 100 × 20 mm tissue culture plate. Cells fixed modification, while oxidation of methionine was set as a variable were kept on ice for 5 min and then underwent a freeze-thaw cycle three times to modification. Percolator was used as the FDR calculator, filtering out peptides ensure proper detergent-independent cell lysis. NaCl was then added to the extracts which had a q-value > 0.0169. The Mascot scores of individual peptides were at a concentration of 0.4 M and subsequently incubated on ice for 5 mins and spun calculated as the absolute probability that the observed peptide match is a random down at 14,000 × g for 15 min at 4 °C. In all, 500 μl of Hypotonic Lysis buffer event when matching spectra to all the expected spectra of a given proteome70. The μ − supplemented with 0.2% NP-40 was added to 500 l of the supernatant extract. Mascot score of a given peptide is equal to: 10 × Log10(P), where P is the absolute TWIN-Strep tagged proteins were then purified by incubating whole cell lysates probability. The Mascot scores for individual proteins were then calculated based from the transiently-transfected cell lines with 50 μl of MagStrep “type3” XT beads upon the summation of the individual peptides for all peptides matching a given (IBA Life Sciences) for two hours at 4 °C. Magnetic resin was washed three times in protein. The samples analyzed by LC-MS include purifications from control cell 20 mM HEPES pH 7.9, 2 mM MgCl2, 0.2 mM EGTA, 10% glycerol, 0.1% NP-40, lines, Strep-METTL2A, Strep-METTL2B, and Strep-METTL6. The LC-MS analysis 0.2 M NaCl, 0.1 mM PMSF, and 1 mM DTT. Proteins were eluted with 1X Buffer was performed once with each sample. BX (IBA LifeSciences) which contains 10 mM D-biotin. Purified proteins were visualized on a NuPAGE Bis-Tris polyacrylamide gel (ThermoFisher) and then transferred to Immobilon-FL Hydrophobic PVDF Transfer Membrane (Millipore In vitro methyltransferase assay. tRNA templates were created by in vitro Sigma) with subsequent immunoblotting against either the FLAG tag or TWIN- transcription. A list of single-stranded 4 nmol Ultramer DNA oligos (IDT) used as Strep tag (Anti-FLAG M2, Sigma-Aldrich; THETM NWSHPQFEK antibody, templates are listed in Supplementary Table 1. Each template was designed to ’ harbor a T7 promoter upstream of the tRNA gene sequence. Each template was GenScript). For immunoblots against endogenous DALRD3, Proteintech s fi DALRD3 antibody was used. PCR ampli ed using a T7 Forward Primer (listed in Supplementary Table 1) and a specific reverse primer complementary to the 3′ end of each tRNA species. PCR We also utilized this procedure followed by TRIzol RNA extraction directly on fi the beads in order to identify co-purifying RNA with each protein of interest. Beads ampli cation was done using Herculase II Fusion DNA Polymerase (Agilent first underwent three washes in the Lysis Buffer as mentioned previously and then Technologies) following standard procedures. The PCR parameters are as follows: resuspended in 250 μl of Molecular Biology Grade RNAse-free water (Corning). 95 °C for 2 min followed by 35 cycles of 95 °C for 20 s, 48 °C for 20 s and 72 °C for μ 30 s, ending with 72 °C for 2 min. The PCR products were then resolved on a 2% 10 l of the bead-water mixture was taken for immunoblotting analysis where the fi beads were mixed with 2X Laemmeli Sample Buffer (Bio-Rad) supplemented with agarose gel where bands were excised, and DNA was puri ed using the Qiagen Gel DTT and boiled at 95 °C for five minutes prior to loading onto a BOLT 4–12% Bis- Extraction Kit. In vitro transcription was done using Optizyme T7 RNA Poly- Tris Plus gel (Life Technologies). RNA extraction followed TRIzol LS RNA merase (ThermoFisher) following standard procedures. Reactions were incubated μ at 37 °C for 3 h followed by DNase treatment (RQ1 DNase, Promega) at 37 °C for extraction protocol (Invitrogen). RNA was resuspended in 5 l of RNAse-free water fi and loaded onto a 10% polyacrylamide, 7 M urea gel. The gel was then stained with 30 min. RNA was then puri ed using RNA Clean and Concentrator Zymo-Spin IC SYBR Gold nucleic acid stain (Invitrogen) to visualize RNA. Columns (Zymo Research). tRNA transcripts were visualized on a 15% poly- FLAG- purifications for the DALRD3 truncations followed the same protein acrylamide, 7 M urea gel stained with SYBR Gold nucleic acid stain. purification protocol outlined above, however, we used Anti-DYKDDDDK Proteins to be used in methyltransferase assays were expressed in 293T cells by fi fi transient transfection and purified by above procedures. In all, 5 μl of input and Magnetic Beads (Clontech) as our puri cation resin. These puri cations than fi underwent the TRIzol-RNA extraction procedure. puri ed protein was visualized by immunoblotting to ensure proper protein expression before use in methyltransferase assays. Prior to incubation with purified protein from human cells, the tRNA was first refolded by initial denaturation in 5 mM TRIS pH 7.5 and 0.16 mM EDTA and heated to 95 °C for two minutes Liquid chromatography-tandem mass spectrometry analysis. Each stably- before a two-minute incubation on ice. Refolding was conducted at 37 °C for integrated cell line was plated on two 150 × 25 mm tissue culture dishes plates and fl 20 min in the presence of HEPES pH 7.5, MgCl2, and NaCl. grown until 70% con uency before cells of each line were combined and harvested For each methyltransferase reaction, 100 ng of refolded tRNA was incubated for protein extraction via the aforementioned hypotonic lysis protocol. Proteins with 42 nM of purified protein elution along with 50 mM TRIS pH 7.5, 0.1 mM were again extracted on MagStrep “type 3” XT beads and washed in the same fi EDTA, 1 mM DTT, 0.5 mM S-adenosylmethionine (NEB) for 4 h at 30 °C. RNA buffer as mentioned above. To ensure all protein was ef ciently eluted off of the was purified using RNA Clean and Concentrator Zymo-Spin IC Columns where magnetic resin, the beads were left in 10 mM D-biotin (Buffer BX, IBA Life- the RNA was resuspended in 6 μl of RNase-free water. To observe if methylation at Sciences) overnight at 4°C. Two one-hour elutions were completed the following residue 32 occurred, we utilized primer extension analysis where reverse day and all elutions were pooled together for each individual sample. The total μ transcriptase extension would be impeded if methylation occurred. Extracted tRNA eluate was then placed on a Spin-X UF 500 l Centrifugal Concentrator (Corning) was pre-annealed with 0.625 pmol of a 5′-32P-radiolabelled oligonucleotide that and spun at 15,000 × g for approximately 1 hour and 15 min at 4 °C. hybridizes around 6 nucleotides downstream residue 32 with 2.8 μl5X For protein separation, 30 μl of concentrated eluate was fractionated on a μ – fi hybridization buffer (250 mM TRIS pH 8.5, 300 mM NaCl) to a total of 14 l. NuPAGE 4 12% Bis-Tris Protein gel (ThermoFisher). The gel was xed overnight The mixture was heated at 95 °C for 3 minutes and then allowed to slowly cool in 40% ethanol and 10% acetic acid. The gel was incubated in Sensitizing Solution to either 58 °C. In all, 14 μl of extension mix (0.12 μl of avian myeloblastosis virus (30% ethanol, 0.2% sodium thiosulphate and 6.8% sodium acetate) for 30 minutes reverse transcriptase (Promega), 2.8 μl 5X RT buffer, 1.12 μl1mM before being washed three times with water for 5 min each wash. The gel was then deoxynucleotidetriphosphates, RNase-free water to 14 μl) was added to each stained in 0.25% silver nitrate for 20 min and washed twice more with water for reaction and incubated at 58 °C for 1 h. Samples were then mixed with 2X 1 min each time. The bands were visualized by developing in 2.5% sodium formamide denaturing dye, heated to 95 °C for 3 min and resolved on an 18% carbonate and 0.015% formaldehyde and allowed to incubate until bands appeared. polyacrylamide, 7 M urea denaturing gel. Gels were exposed by Phosphor-Imager The remainder of each eluate (~65 μl) was loaded on a NuPAGE 4–12% Bis-Tris μ fl analysis. Positive controls underwent same treatment but started with 3.2 g protein gel and brie y fractionated to yield a single gel band corresponding to the of RNA. majority of proteins within each purification. Gel bands were excised using a razor blade and subject to in gel reduction, alkylation and trypsin digest by the URMC Mass Spectrometry Resource Lab67,68. Immunoblot and Northern blot assays. To verify loss of DALRD3, cell extracts Peptides were injected onto a homemade 30 cm C18 column with 1.8 μm beads were loaded onto BOLT 4–12% Bis-Tris gels (ThermoFisher) followed by immu- (Sepax), with an Easy nLC-1000 HPLC (Thermo Fisher), connected to a Q Exactive noblotting onto PVDF membrane and probing with DALRD3 antibody (Pro- Plus mass spectrometer (Thermo Fisher). Solvent A: 0.1% formic acid in water, teintech, cat. No. 26294-1-AP, 1:1000 dilution) and anti-Actin C4 (EMD Millipore, Solvent B: 0.1% formic acid in acetonitrile. The gradient began at 3% B and held for cat. No MAB1501, 1:000 dilution). Expression of DALRD3 in stably-infected rescue 2 min, increased to 30% B over 13 min, increased to 70% over 2 minutes and held cell lines were characterized by immunoblotting with the anti-FLAG M2 antibody for 3 mins, then returned to 3% B in 2 min and re-equilibrated for 8 min, for a total (Sigma-Aldrich, F1804, 1:3333 dilution). Transient transfections of TWIN-Strep run time of 30 min. For peptides isolated from the single gel band corresponding to and FLAG-tagged proteins were also loaded onto BOLT 4–12% Bis-Tris gels, the majority of proteins, the gradient began at 3% B and held for 2 min, increased immunoblotted to PVDF membranes and probed with anti-TWIN-Strep (Gen- to 30% B over 41 min, increased to 70% over 3 min and held for 4 min, then script, cat. No. A01732, 1:1000 dilution) and anti-FLAG M2. NLuc immunoblots returned to 3% B in 2 min and re-equilibrated for 8 minutes, for a total run time of were probed with anti-FLAG, anti-GFP (Santa Cruz, cat. No. sc-9996; 1:100 60 minutes. The Q Exactive Plus was operated in data-dependent mode, with a full dilution) and anti-Actin C4 antibodies. Image analysis of immunoblots were per- MS1 scan followed by 8 data-dependent MS2 scans. The full scan was done over a formed using Image Studio software (Li-Cor). range of 400–1400 m/z, with a resolution of 70,000 at m/z of 200, an AGC target of 3-methylcytidine modification status was explored through the PHA assay. To 1e6, and a maximum injection time of 50 ms. The MS2 scans were performed at conduct the PHA assay, probes were designed to hybridize upstream and 17,500 resolution, with an AGC target of 1e5 and a maximum injection time of 250 downstream of residue 32 (oligos listed in Supplementary Table 1). 5 μg of RNA ms. The isolation width was 1.5 m/z, with an offset of 0.3 m/z, and a normalized was loaded onto a 10% polyacrylamide, 1xTBE, 7 M urea gel and transferred onto collision energy of 27 was used. an Amersham Hybond-XL membrane (GE Healthcare) for Northern Blotting Raw data was searched using the SEQUEST search engine within the Proteome analysis. Oligonucleotides used to detect RNAs are listed in Supplementary Table 1. Discoverer software platform, version 1.4 (Thermo Fisher), using the SwissProt The oligos were radiolabeled by T4 polynucleotide kinase (NEB) with adenosine

12 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6 ARTICLE

[γ32P]-triphosphate (6000 Ci/mmol, Amersham Biosciences) following standard 14. Auffinger, P. & Westhof, E. An extended structural signature for the tRNA procedures. Northern blots were visualized by Phosphor-Imager analysis and anticodon loop. Rna 7, 334–341 (2001). stripped via two incubations at 80 °C for 30 min in a buffer containing 0.15 M 15. Olejniczak, M. & Uhlenbeck, O. C. tRNA residues that have coevolved with NaCl, 0.015 m Na-citrate and 0.1% SDS. Image analysis of Phosphorimager scans their anticodon to ensure uniform and accurate codon recognition. Biochimie were performed using ImageJ open source software. 88, 943–950 (2006). 16. Weissenbach, J., Kiraly, I. & Dirheimer, G. [Primary structure of tRNA Thr 1a ’ – Human subjects. Evaluation of affected members by a board-certified clinical and b from brewer s yeast]. Biochimie 59, 381 391 (1977). fi geneticist included obtaining medical and family histories, clinical examination, 17. Arimbasseri, A. G. et al. Evolving speci city of tRNA 3-methyl-cytidine-32 neuroimaging and clinical laboratory investigations. After obtaining a written (m3C32) modification: a subset of tRNAsSer requires N6-isopentenylation of informed consent for enrollment in an IRB-approved project from the Research A37. RNA. 22, 1400–1410 (2016). Advisory Council (RAC) of the King Faisal Specialist Hospital and Research Centre 18. Boccaletto, P. et al. MODOMICS: a database of RNA modification pathways. (KFSHRC RAC# 2121053), venous blood was collected in EDTA and sodium 2017 update. Nucleic Acids Res. 46, D303–D307 (2018). heparin tubes for DNA extraction and establishment of lymphoblastoid cell lines 19. Keith, G. The primary structures of two arginine tRNAs (anticodons C-C-U (patients 19DG0509 and 19DG0510), respectively. All studies abide by the and mcm5a2U-C-psi) and of glutamine tRNA (anticodon C-U-G) from Declaration of Helsinki principles. The authors affirm that the parents of the bovine liver. Nucleic Acids Res. 12, 2543–2547 (1984). human research participants provided informed consent for publication of the 20. Clark, W. C., Evans, M. E., Dominissini, D., Zheng, G. & Pan, T. tRNA base images of the minors in Fig. 6. methylation identification and quantification via high-throughput sequencing. RNA. 22, 1771–1784 (2016). Statistical analyses and reproducibility. All statistics and graphs were performed 21. Suzuki, T. & Suzuki, T. A complete landscape of post-transcriptional fi and generated using GraphPad Prism software. Where applicable, error bars modi cations in mammalian mitochondrial tRNAs. Nucleic Acids Res. 42, – represent the standard deviation. Statistical tests and the number of times each 7346 7357 (2014). fi experiment was repeated are stated in each figure legend. For primer extension 22. de Crecy-Lagard, V. et al. Matching tRNA modi cations in humans to their – assays, statistical analyses of the quantification were performed using one-way known and predicted enzymes. Nucleic Acids Res. 47, 2143 2159 (2019). ANOVA and significance calculated using Tukey’s multiple comparison test. For 23. D’Silva, S., Haider, S. J. & Phizicky, E. M. A domain of the actin binding the codon reporter assays, statistical analyses for significance was calculated a two- protein Abp140 is the yeast methyltransferase responsible for 3- tailed, unpaired students t-test (t = 1.831, df = 4). methylcytidine modification in the tRNA anti-codon loop. RNA 17, 1100–1110 (2011). 24. Noma, A. et al. Actin-binding protein ABP140 is a methyltransferase for 3- Reporting summary. Further information on research design is available in methylcytidine at position 32 of tRNAs in Saccharomyces cerevisiae. RNA 17, the Nature Research Reporting Summary linked to this article. 1111–1119 (2011). 25. Han, L., Marcus, E., D’Silva, S. & Phizicky, E. M. S. cerevisiae Trm140 has two Data availability recognition modes for 3-methylcytidine modification of the anticodon loop of The source data underlying all Figures are provided as a Source data file. The mass tRNA substrates. RNA 23, 406–419 (2017). spectrometry data sets that include peptide spectral matches are included as 26. Lenhard, B., Orellana, O., Ibba, M. & Weygand-Durasevic, I. tRNA Supplementary Data 1 and 2. All data is available from the corresponding author upon recognition and evolution of determinants in seryl-tRNA synthesis. Nucleic reasonable request. Acids Res. 27, 721–729 (1999). 27. Holman, K. M., Puppala, A. K., Lee, J. W., Lee, H. & Simonovic, M. Insights into substrate promiscuity of human seryl-tRNA synthetase. RNA 23, Received: 6 September 2019; Accepted: 24 April 2020; 1685–1699 (2017). 28. Rubio, M. A. et al. Editing and methylation at a single site by functionally interdependent activities. Nature 542, 494–497 (2017). 29. Han, L. & Phizicky, E. M. A rationale for tRNA modification circuits in the anticodon loop. RNA 24, 1277–1284 (2018). 30. Maraia, R. J., & Arimbasseri, A. G. Factors that shape eukaryotic tRNAomes: References processing, modification and anticodon-codon use. Biomolecules 7, 26 (2017). 1. Kapur, M. & Ackerman, S. L. mRNA translation Gone Awry: translation 31. Towns, W. L. & Begley, T. J. Transfer RNA methytransferases and their fidelity and neurological disease. Trends Genet 34, 218–231 (2018). corresponding modifications in budding yeast and humans: activities, predications, 2. Frye, M., Harada, B. T., Behm, M. & He, C. RNA modifications modulate gene and potential roles in human health. DNA Cell Biol. 31, 434–454 (2012). expression during development. Science 361, 1346–1349 (2018). 32. Xu, L. et al. Three distinct 3-methylcytidine (m(3)C) methyltransferases 3. Kirchner, S. & Ignatova, Z. Emerging roles of tRNA in adaptive translation, modify tRNA and mRNA in mice and humans. J. Biol. Chem. 292, signalling dynamics and disease. Nat. Rev. Genet 16,98–112 (2015). 14695–14703 (2017). 4. El Yacoubi, B., Bailly, M. & de Crecy-Lagard, V. Biosynthesis and function of 33. Baum, M. & Beier, H. Wheat cytoplasmic arginine tRNA isoacceptor with a posttranscriptional modifications of transfer RNAs. Annu Rev. Genet 46, U*CG anticodon is an efficient UGA suppressor in vitro. Nucleic Acids Res 26, 69–95 (2012). 1390–1395 (1998). 5. Vare, V. Y., Eruysal, E. R., Narendran, A., Sarachan, K. L. & Agris, P. F. 34. Vandivier, L. E. et al. Chemical modifications mark alternatively spliced and Chemical and conformational diversity of modified nucleosides affects Trna uncapped messenger RNAs in Arabidopsis. Plant Cell 27, 3024–3037 (2015). structure and function. Biomolecules 7, 29 (2017). 35. Schmidt, T. G. et al. Development of the Twin-Strep-tag(R) and its application 6. Jackman, J. E. & Alfonzo, J. D. Transfer RNA modifications: nature’s for purification of recombinant proteins from cell culture supernatants. combinatorial chemistry playground. Wiley Interdiscip. Rev. RNA 4,35–48 Protein Expr. Purif. 92,54–61 (2013). (2013). 36. Wolf, Y. I., Aravind, L., Grishin, N. V. & Koonin, E. V. Evolution of 7. Ramos, J. & Fu, D. The emerging impact of tRNA modifications in the brain aminoacyl-tRNA synthetases—analysis of unique domain architectures and and nervous system. Biochim. Biophys. Acta Gene Regul. Mech. 1862, 412–428 phylogenetic trees reveals a complex history of horizontal gene transfer events. (2019). Genome Res. 9, 689–710 (1999). 8. Bohnsack, M. T. & Sloan, K. E. The mitochondrial epitranscriptome: the roles 37. Delagoutte, B., Moras, D. & Cavarelli, J. tRNA aminoacylation by arginyl- of RNA modifications in mitochondrial translation and human disease. Cell tRNA synthetase: induced conformations during substrates binding. EMBO J. Mol. Life Sci. 75, 241–260 (2018). 19, 5599–5610 (2000). 9. Jonkhout, N. et al. The RNA modification landscape in human disease. Rna 38. Cavarelli, J., Delagoutte, B., Eriani, G., Gangloff, J. & Moras, D. L-arginine 23, 1754–1769 (2017). recognition by yeast arginyl-tRNA synthetase. EMBO J. 17, 5438–5448 (1998). 10. Suzuki, T., Nagao, A. & Suzuki, T. Human mitochondrial tRNAs: biogenesis, 39. Kriventseva, E. V. et al. OrthoDB v10: sampling the diversity of animal, plant, function, structural aspects, and diseases. Annu Rev. Genet 45, 299–329 fungal, protist, bacterial and viral genomes for evolutionary and functional (2011). annotations of orthologs. Nucleic Acids Res. 47, D807–D811 (2019). 11. Santos, M., Fidalgo, A., Varanda, A. S., Oliveira, C. & Santos, M. A. S. tRNA 40. Herrero, J. et al. Ensembl comparative genomics resources. Database (Oxford) deregulation and its consequences in cancer. Trends Mol. Med. 25, 853–865 2016, bav096 (2016). (2019). 41. Chan, P. P. & Lowe, T. M. GtRNAdb 2.0: an expanded database of transfer 12. Torres, A. Batlle, E. & Ribas de Pouplana L. Role of tRNA modifications in RNA genes identified in complete and draft genomes. Nucleic Acids Res. 44, human diseases. Trends Mol. Med. 20, 306–314 (2014). D184–189 (2016). 13. Auffinger, P. & Westhof, E. Singly and bifurcated hydrogen-bonded base-pairs 42. Carette, J. E. et al. Ebola virus entry requires the cholesterol transporter in tRNA anticodon hairpins and ribozymes. J. Mol. Biol. 292, 467–483 (1999). Niemann-Pick C1. Nature 477, 340–343 (2011).

NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications 13 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16321-6

43. Lamichhane, T. N. et al. Lack of tRNA-i6A modification causes 66. Kingston, R. E., Chen, C. A. & Rose, J. K. Calcium phosphate transfection. mitochondrial-like metabolic deficiency in S. pombe by limiting activity of Curr. Protoc. Mol. Biol. Chapter 9, Unit 9 1 (2003). cytosolic tRNATyr, not mito-tRNA. RNA 22, 583–596 (2016). 67. Ramos, J. et al. Formation of tRNA wobble inosine in humans is disrupted by 44. Arimbasseri, A. G. et al. RNA polymerase III output is functionally linked to a millennia-old mutation causing intellectual disability. Mol. Cell. Biol. 39, tRNA dimethyl-G26 modification. PLoS Genet. 11, e1005671 (2015). e00203-19 (2019). 45. Dewe, J. M., Fuller, B. L., Lentini, J. M., Kellner, S. M. & Fu, D. TRMT1- 68. Gu, C. et al. Phosphorylation of human TRM9L integrates multiple stress- Catalyzed tRNA modifications are required for redox homeostasis to ensure signaling pathways for tumor growth suppression. Sci. Adv. 4, eaas9184 proper cellular proliferation and oxidative stress survival. Mol. Cell. Biol. 37, (2018). e00214-17 (2017). 69. The, M., MacCoss, M. J., Noble, W. S. & Kall, L. Fast and accurate protein false 46. Lamichhane, T. N., Blewett, N. H. & Maraia, R. J. Plasticity and diversity of discovery rates on large-scale proteomics data sets with Percolator 3.0. J. Am. tRNA anticodon determinants of substrate recognition by eukaryotic A37 Soc. Mass Spectrom. 27, 1719–1727 (2016). isopentenyltransferases. RNA 17, 1846–1857 (2011). 70. Koenig, T. et al. Robust prediction of the MASCOT score for an improved 47. Hiley, S. L. et al. Detection and discovery of RNA modifications using quality assessment in mass spectrometric proteomics. J. Proteome Res. 7, microarrays. Nucleic Acids Res. 33, e2 (2005). 3708–3717 (2008). 48. Pallan, P. S., Kreutz, C., Bosio, S., Micura, R. & Egli, M. Effects of N2,N2- dimethylguanosine on RNA structure and stability: crystal structure of an RNA duplex with tandem m2 2G:A pairs. RNA 14, 2125–2135 (2008). Acknowledgements 49. Chawla, M., Oliva, R., Bujnicki, J. M. & Cavallo, L. An atlas of RNA base pairs We thank Eric Phizicky, Sina Ghaemmaghami and members of the Fu Lab for comments involving modified nucleobases with optimal geometries and accurate on the paper; Kevin Welle and the URMC Mass Spectrometry Resource Lab for pro- energies. Nucleic Acids Res. 43, 9573 (2015). teomics; and Jon Lueck for assistance in design of the codon reporter constructs. This 50. Anazi, S. et al. Clinical genomics expands the morbid genome of intellectual work was supported by the Saudi Program, King Salman Center for disability and offers a high diagnostic yield. Mol. Psychiatry 22, 615–624 (2017). Disability Research, and King Abdulaziz City for Science and Technology Grant 08- 51. Shamseldin, H. E. et al. Increasing the sensitivity of clinical exome sequencing MED497-20 to F.S.A. and National Science Foundation CAREER Award 1552126 to D.F. through improved filtration strategy. Genet Med. 19, 593–598 (2017). 52. Auvin, S., Cilio, M. R. & Vezzani, A. Current understanding and neurobiology Author contributions of epileptic encephalopathies. Neurobiol. Dis. 92,72–89 (2016). Conceptualization, J.L. and D.F.; methodology, J.L., F.S.A., and D.F.; investigation, J.L., 53. Kalser, J. & Cross, J. H. The epileptic encephalopathy jungle - from Dr West to H.S.A., E.F., F.S.A., and D.F.; writing—original draft, J.L and D.F.; writing—review and the concepts of aetiology-related and developmental encephalopathies. Curr. editing, J.L., E.F., F.S.A., and D.F.; funding acquisition, F.S.A., and D.F.; resources, E.F., Opin. Neurol. 31, 216–222 (2018). F.S.A., and D.F.; supervision, E.F., F.S.A., and D.F. 54. Scheffer, I. E. et al. ILAE classification of the epilepsies: position paper of the ILAE commission for classification and terminology. Epilepsia 58, 512–521 (2017). Competing interests 55. Guy, M. P. & Phizicky, E. M. Two-subunit enzymes involved in eukaryotic The authors declare no competing interests. post-transcriptional tRNA modification. RNA Biol. 11, 1608–1618 (2014). fi 56. Dixit, S., Henderson, J. C. & Alfonzo, J. D. Multi-Substrate Speci city and the Additional information Evolutionary Basis for Interdependence in tRNA Editing and Methylation Supplementary information Enzymes. Front. Genet. 10, 104 (2019). is available for this paper at https://doi.org/10.1038/s41467- 57. Guo, M., Yang, X. L. & Schimmel, P. New functions of aminoacyl-tRNA 020-16321-6. synthetases beyond translation. Nat. Rev. Mol. Cell Biol. 11, 668–674 (2010). Correspondence 58. Gale, A. J. & Schimmel, P. Isolated RNA binding domain of a class I tRNA and requests for materials should be addressed to D.F. synthetase. Biochemistry 34, 8896–8903 (1995). Peer review information 59. Yarham, J. W. et al. Defective i6A37 modification of mitochondrial and Nature Communications thanks the anonymous reviewer(s) for cytosolic tRNAs results from pathogenic mutations in TRIT1 and its substrate their contribution to the peer review of this work. Peer reviewer reports are available. tRNA. PLoS Genet. 10, e1004424 (2014). Reprints and permission information 60. Kernohan, K. D. et al. Matchmaking facilitates the diagnosis of an autosomal- is available at http://www.nature.com/reprints recessive mitochondrial disease caused by biallelic mutation of the tRNA – Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in isopentenyltransferase (TRIT1) gene. Hum. Mutat. 38, 511 516 (2017). fi 61. Yasukawa, T., Suzuki, T., Ishii, N., Ohta, S. & Watanabe, K. Wobble published maps and institutional af liations. modification defect in tRNA disturbs codon-anticodon interaction in a mitochondrial disease. EMBO J. 20, 4794–4802 (2001). 62. Yasukawa, T., Suzuki, T., Ueda, T., Ohta, S. & Watanabe, K. Modification Open Access This article is licensed under a Creative Commons defect at anticodon wobble nucleotide of mitochondrial tRNAs(Leu)(UUR) Attribution 4.0 International License, which permits use, sharing, with pathogenic mutations of mitochondrial myopathy, encephalopathy, lactic adaptation, distribution and reproduction in any medium or format, as long as you give acidosis, and stroke-like episodes. J. Biol. Chem. 275, 4251–4257 (2000). appropriate credit to the original author(s) and the source, provide a link to the Creative 63. Kirino, Y., Goto, Y., Campos, Y., Arenas, J. & Suzuki, T. Specific correlation Commons license, and indicate if changes were made. The images or other third party between the wobble modification deficiency in mutant tRNAs and the clinical material in this article are included in the article’s Creative Commons license, unless features of a human mitochondrial disease. Proc. Natl Acad. Sci. USA 102, indicated otherwise in a credit line to the material. If material is not included in the 7127–7132 (2005). article’s Creative Commons license and your intended use is not permitted by statutory 64. Kirino, Y. et al. Codon-specific translational defect caused by a wobble regulation or exceeds the permitted use, you will need to obtain permission directly from modification deficiency in mutant tRNA from a human mitochondrial disease. the copyright holder. To view a copy of this license, visit http://creativecommons.org/ – Proc. Natl Acad. Sci. USA 101, 15070 15075 (2004). licenses/by/4.0/. 65. Ko, A. & Kang, H.-C. Frequently identified genetic developmental and epileptic encephalopathy: a review focusing on precision medicine. Ann. Child Neurol. 27,2–12 (2019). © The Author(s) 2020

14 NATURE COMMUNICATIONS | (2020) 11:2510 | https://doi.org/10.1038/s41467-020-16321-6 | www.nature.com/naturecommunications