Quick viewing(Text Mode)

Hypomodified Trna in Evolutionarily Distant Yeasts Can Trigger Rapid Trna Decay to Activate

Hypomodified Trna in Evolutionarily Distant Yeasts Can Trigger Rapid Trna Decay to Activate

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1

2

3 Hypomodified tRNA in evolutionarily distant yeasts can trigger rapid tRNA decay to activate

4 the general amino acid control response, but with different consequences

5

6 Thareendra De Zoysa, Eric M. Phizicky1

7 Department of Biochemistry and Biophysics, Center for RNA Biology, University of Rochester School

8 of Medicine, Rochester, NY, USA 14642

9

10 Address correspondence to:

11 Eric M. Phizicky

12 Department of Biochemistry and Biophysics

13 University of Rochester School of Medicine

14 601 Elmwood Avenue

15 Rochester, NY 14642

16 Phone: 585-275-7268

17 Email: [email protected]

18 Running Title: Rapid tRNA decay and general amino acid control in divergent yeasts

19 Key words: S. pombe/S. cerevisiae/Trm8/m7G/rapid tRNA decay/general amino acid control

De Zoysa 1

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 20 Abstract 21

22 All tRNAs are extensively modified, and modification deficiency often results in growth defects

23 in the budding yeast Saccharomyces cerevisiae and neurological or other disorders in humans. In S.

24 cerevisiae, lack of any of several tRNA body modifications results in rapid tRNA decay (RTD) of

25 certain mature tRNAs by the 5’-3’ exonucleases Rat1 and Xrn1. As tRNA quality control decay

26 mechanisms are not extensively studied in other eukaryotes, we studied trm8Δ mutants in the

27 evolutionarily distant fission yeast Schizosaccharomyces pombe, which lack 7-methylguanosine at

28 G46 of tRNAs. We report here that S. pombe trm8Δ mutants are temperature sensitive primarily due to

29 decay of tRNATyr(GUA) and that spontaneous mutations in the RAT1 ortholog dhp1+ restored

30 temperature resistance and prevented tRNA decay, demonstrating conservation of the RTD pathway.

31 We also report for the first time evidence linking the RTD and the general amino acid control (GAAC)

32 pathways, which we show in both S. pombe and S. cerevisiae. In S. pombe trm8Δ mutants,

33 spontaneous GAAC mutations restored temperature resistance and tRNA levels, and the temperature

34 sensitivity of trm8Δ mutants was precisely linked to GAAC activation due to tRNATyr(GUA) decay.

35 Similarly, in the well-studied S. cerevisiae trm8Δ trm4Δ RTD mutant, temperature sensitivity was

36 closely linked to GAAC activation due to tRNAVal(AAC) decay; however, in S. cerevisiae, GAAC

37 mutations increased tRNA decay and enhanced temperature sensitivity. Thus, these results

38 demonstrate a conserved GAAC activation coincident with RTD in S. pombe and S. cerevisiae, but an

39 opposite impact of the GAAC response in the two organisms. We speculate that the RTD pathway

40 and its regulation of the GAAC pathway is widely conserved in eukaryotes, extending to other mutants

41 affecting tRNA body modifications.

42

De Zoysa 2

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 43 Author Summary

44

45 tRNA modifications are highly conserved and their lack frequently results in growth defects in

46 the yeast Saccharomyces cerevisiae and neuorological disorders in humans. S. cerevsiaie has two

47 tRNA quality control decay pathways that sense tRNAs lacking modifications in the main tRNA body.

48 One of these, the rapid tRNA decay (RTD) pathway, targets mature tRNAs for 5'-3' exonucleolytic

49 decay by Rat1 and Xrn1. It is unknown if RTD is conserved in eukaryotes, and if it might explain

50 phenotypes associated with body modification defects. Here we focus on trm8Δ mutants, lacking

7 7 51 m G46, in the evolutionarily distant yeast Schizosaccharomyces pombe. Loss of m G causes

52 temperature sensitivity and RTD in S. cerevisiae, microcephalic primordial dwarfism in humans, and

53 defective stem cell renewal in mice. We show that S. pombe trm8Δ mutants are temperature sensitive

54 due to tY(GUA) decay by Rat1, implying conservation of RTD among divergent eukaryotes. We also

55 show that the onset of RTD triggers activation of the general amino acid control (GAAC) pathway in

56 both S. pombe and S. cerevisiae, resulting in exacerbated decay in S. pombe and reduced decay in

57 S. cerevisiae. We speculate that RTD and its regulation of the GAAC pathway will be widely

58 conserved in eukaryotes including humans.

59

De Zoysa 3

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 60 Introduction

61

62 tRNAs are subject to extensive post-transcriptional modifications that often profoundly affect

63 tRNA function, as lack of modifications often leads to growth defects in the budding yeast

64 Saccharomyces cerevisiae and to neurological or mitochondrial disorders in humans (1-5). Many

65 tRNA modifications in the anticodon loop are important for decoding fidelity, reading frame

66 maintenance, and sometimes charging efficiency (6-15). By contrast, modifications in the tRNA body,

67 the region outside the anticodon loop, are often important for folding and stability (16-18), resulting in

68 substantial growth defects. In S. cerevisiae, deletion of TRM6 or TRM61 is lethal, associated with lack

1 69 of 1-methyladenosine at A58 (m A58) (19), whereas deletion of TAN1, TRM1, or TRM8 (or TRM82)

4 70 results in temperature sensitivity associated with lack of 4-acetylcytidine at C12 (ac C12), N2,N2-

2,2 7 71 dimethylguanosine at G26 (m G26), or 7-methylguanosine at G46 (m G46) respectively (20-22).

72 Similarly, human neurological disorders are linked to mutations in TRMT10A, associated with reduced

1 2,2 7 73 1-methylguanosine at G9 (m G9) (23, 24), TRMT1 (m G26) (25-28), WDR4 (m G46) (29-31) and

5 74 NSUN2, associated with reduced 5-methylcytidine (m C) at C48-50, as well as at C34 and C40 (32-34).

75 In S. cerevisiae, lack of any of several tRNA body modifications leads to decay of a subset of

76 the corresponding hypomodified tRNAs, mediated by either of two tRNA quality control pathways,

77 each acting on different hypomodified tRNAs and at different stages of tRNA biogenesis. First, the

Met 1 78 nuclear surveillance pathway targets pre-tRNAi lacking m A, acting through the the nuclear

79 exosome and the TRAMP complex to degrade the pre-tRNA from the 3' end (17, 35-37). The nuclear

80 surveillance pathway also targets a large portion of wild type (WT) pre-tRNAs shortly after

81 transcription, ascribed to errors in folding of the nascent tRNA or to mutations arising during

82 transcription (38). Second, the rapid tRNA decay (RTD) pathway targets certain mature tRNAs lacking

7 2,2 4 83 m G46, m G26, or ac C12, using the 5'-3' exonucleases Rat1 in the nucleus and Xrn1 in the cytoplasm

84 (18, 21, 22, 39, 40). RTD is inhibited by a met22Δ mutation (22, 39, 41, 42) due to accumulation of the

85 Met22 substrate adenosine 3', 5' bisphosphate (pAp) (43, 44), which binds the active site of Xrn1 and

86 presumably Rat1 (45). The RTD pathway also targets fully modified tRNAs with destabilizing

De Zoysa 4

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 87 mutations in the stems, particularly the acceptor and T-stem, which expose the 5' end (40-42). The

88 hypomodified tRNAs targeted by the RTD pathway also expose the 5' end, ascribed to destabilization

89 of the tertiary fold (40).

90 There is limited evidence documenting tRNA quality control decay pathways that act on

91 hypomodified tRNAs in other eukaryotes. A mouse embryonic stem cell line with a knockout of

92 METTL1 (ortholog of S. cerevisiae TRM8) had undetectable m7G in its tRNA substrates and reduced

93 levels of several METTL1 substrate tRNAs (46). Similarly, knockdown of METTL1 and NSUN2

94 (homolog of S. cerevisiae TRM4) in HeLa cells led to reduced levels of tRNAVal(AAC) (tV(AAC)) at 43°C

95 in the presence of 5-fluorouracil (5-FU) (47), a known inhibitor of pseudouridine synthases and 5-

96 methyluridine methyltransferase (48-50). However, in both of these cases, the underlying mechanism

97 is not known. It was also shown that WT mature tRNAiMet was subject to decay by Xrn1 and Rat1 after

98 43°C heat shock in HeLa cells, although there was no change in the modification pattern in vivo or in

99 physical stability of the tRNA in vitro caused by this temperature shift (51).

100 The goal of the work described here is to determine if and to what extent tRNA quality control

101 decay pathways are linked to hypomodified tRNAs in eukaryotes other than S. cerevisiae. To address

7 102 this issue, we have studied the biology of the tRNA m G46 methyltransferase Trm8 in the fission yeast

103 Schizosaccharomyces pombe, which diverged from S. cerevisiae ~ 600 million years ago (52).

104 We chose to study S.pombe Trm8 because S. cerevisiae trm8Δ mutants were known to trigger

105 decay by the RTD pathway. S. cerevisiae Trm8 forms a complex with Trm82 that is required for

7 106 formation of m G46 in eukaryotic tRNAs (53, 54). S. cerevisiae trm8Δ and trm82Δ mutants are each

107 temperature sensitive (20), and trm8Δ or trm82Δ mutants also lacking any of several other body

108 modifications had enhanced temperature sensitivity (18). Moreover, the temperature sensitivity of

109 trm8Δ mutants was suppressed by a met22Δ mutation and was associated with decay of tV(AAC)

7 5 110 (22), and the temperature sensitivity of trm8Δ trm4Δ mutants (lacking both m G and m C) was shown

111 explicitly to be due to RTD of tV(AAC) (18, 39). Furthermore, Trm8 biology in mammalian cells has

112 additional dimensions of complexity. The human TRM82 ortholog WDR4 was associated with reduced

113 tRNA m7G modification and a distinct form of microcephalic primordial dwarfism (29); METTL1 or

De Zoysa 5

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 114 WDR4 knock out mouse embryonic stem cells showed defects in self renewal and differentiation (46);

115 and METTL1 was also responsible for m7G modification of mammalian miRNAs and mRNAs (55, 56).

116 This evidence emphasizes that Trm8/Trm82 (METTL1/WDR4) and/or its m7G modification product is

117 important in S. cerevisiae and mammals, although the reasons are not yet known beyond S.

118 cerevisiae.

119 We find here that S. pombe trm8Δ mutants have a temperature sensitive growth defect due

120 primarily to decay of tY(GUA) and to some extent tP(AGG) by the Rat1 ortholog Dhp1, demonstrating

121 that a major component of the RTD pathway is conserved between S. pombe and S. cerevisiae. We

122 also find an unexpected connection between the RTD pathway and the general amino acid control

123 (GAAC) pathway in both S. pombe and S. cerevisiae. In both S. pombe trm8Δ mutants and S.

124 cerevisiae trm8Δ trm4Δ mutants, the temperature sensitivity coincides with the onset of tRNA decay,

125 which in turn triggers the GAAC activation, presumably due to the increased stress by the tRNA

126 decay. However, in Sp trm8Δ mutants, GAAC activation is deleterious to growth, as mutations in the

127 GAAC pathway restore growth and tRNA levels, whereas in S. cerevisiae trm8Δ trm4Δ mutants,

128 GAAC pathway activation is beneficial, as GAAC mutations exacerbate the growth defect and

129 accelerate tRNA decay. Thus, our results demonstrate a conserved GAAC response associated with

130 tRNA decay but opposite effects on cell physiology in the two organisms. These findings suggest the

131 widespread conservation of the RTD pathway in eukaryotes, and the widespread conservation of the

132 GAAC pathway in monitoring overall tRNA quality.

133

134 Results

135 The S. pombe trm8Δ mutants lack m7G in tRNAs and are temperature sensitive

136 As Trm8 is the catalytic subunit of the Trm8-Trm82 complex (20), we anticipated that tRNAs

137 from S. pombe trm8Δ mutants would lack m7G. We purified tY(GUA) and tF(GAA), which had each

7 138 been previously shown to have m G46 (57, 58), and then analyzed their nucleosides by HPLC

139 analysis. Purified tY(GUA) from S. pombe trm8Δ mutants had no detectable m7G levels (less than

140 0.03 moles/mole), compared to near stoichiometric levels in tY(GUA) from WT cells (0.93 +/- 0.22

De Zoysa 6

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 141 moles/mole), whereas levels of each of three other analyzed modifications (pseudouridine (Ψ), m5C,

142 and m1A) were very similar in trm8Δ and WT cells (Fig. 1A). Similarly, purified tF(GAA) from trm8Δ

143 mutants had no detectable levels of m7G compared to near stoichiometric levels in WT cells, but

144 otherwise WT levels of Ψ, 2'-O-methylcytidine (Cm) and m2,2G (Fig. 1A). These results suggest

145 strongly that S. pombe trm8+ is the methyltransferase responsible for m7G formation in cytoplasmic

146 tRNAs.

147 To understand the biology of S. pombe trm8Δ mutants, we examined the growth phenotypes

148 of two genetically independent trm8Δ mutants. trm8Δ mutants were temperature sensitive starting at

+ 149 37°C on rich (YES) and minimal (EMM) media, and expression of Ptrm8 trm8 on a plasmid restored

150 WT growth in both media (Fig. 1B). Thus, the temperature sensitivity of trm8Δ mutants was due to

151 lack of trm8+.

152

153 S. pombe trm8Δ mutants have reduced levels of tP(AGG) and tY(GUA) at high temperatures

154 To determine if the temperature sensitivity of S. pombe trm8Δ mutants was associated with

155 tRNA decay, we analyzed tRNA levels of trm8Δ mutants after an 8 hour temperature shift in YES

156 media from 30°C to 36.5°C, 37.5°C, and 38.5°C, which progressively inhibited growth (Fig. S1). We

157 measured tRNA levels of all 21 tRNAs in the Genomic tRNA Database (59) that had a 5-nt variable

158 loop with a central guanosine residue (Table S1), which is the signature for m7G modification (60). We

159 quantified levels of each tRNA at each temperature relative to the levels of that tRNA in WT cells at

160 30°C, after normalization of each to the levels of the non-Trm8 substrate tG(GCC) at the

161 corresponding temperature. We used tG(GCC) as the standard because the usual standards, 5S and

162 5.8S RNA, each had temperature-dependent reduction in their levels in trm8Δ mutants (Fig S2), as

163 determined relative to input RNA levels. Note that with tG(GCC) as the standard, the levels of another

164 non-Trm8 substrate, tL(UAA), were also unaffected.

165 Northern analysis showed that S. pombe trm8Δ mutants had significantly reduced levels of two

166 of the 21 potential Trm8 substrate tRNAs as the temperature was increased. The levels of tP(AGG)

167 were substantially reduced in trm8Δ mutants, from 70% of the levels in WT cells at 30°C, to 50%,

De Zoysa 7

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 168 31%, and 18% after temperature shift to 36.5°C, 37.5°C, and 38.5°C respectively, whereas levels of

169 tP(AGG) in WT cells remained constant as temperature increased (Fig. 2A, 2B). As expected,

170 tP(AGG) is indeed a substrate of Trm8, since purified tP(AGG) from trm8Δ mutants had undetectable

171 levels of m7G, but WT levels of each of three other modifications (Fig. S3). The levels of tY(GUA)

172 were also reduced in trm8Δ mutants as temperature increased, albeit to a lesser extent than tP(AGG)

173 levels. Levels of tY(GUA) in trm8Δ mutants were about the same as those in WT cells at 30°C

174 (119%), remained essentially unchanged at 36.5°C and 37.5°C (124%, and 98%), but were reduced

175 to 67% at 38.5°C. Levels of tY(GUA) in WT cells were relatively constant at all temperatures. In

176 contrast, none of the 19 other predicted Trm8 substrate tRNAs showed a temperature-dependent

177 reduction in levels (Fig. 2A, 2B, S4, S5). Levels of 15 tRNAs were approximately constant in trm8Δ

178 mutants as temperature increased, although the initial levels varied somewhat, and levels of the other

179 four tRNAs (tR(CCU), tMe(CAU), tV(CAC), and tK(UUU)) were modestly increased at 38.5°C. Thus, if

180 the temperature sensitivity of trm8Δ mutants was due to loss of tRNAs, the likely candidates were

181 tP(AGG) and tY(GUA).

182

183 The growth defect of S. pombe trm8Δ mutants is primarily due to loss of tY(GUA)

184 To evaluate the cause of the temperature sensitivity of S. pombe trm8Δ mutants, we analyzed

185 growth after overexpression of tP(AGG) and/or tY(GUA) on plasmids (Fig. 2C). Surprisingly, on both

186 YES media and EMM complete (EMMC) media, trm8Δ mutants expressing tY(GUA) grew almost as

+ 187 well as the trm8Δ [Ptrm8 trm8 ] strain or the WT strain at elevated temperatures, whereas trm8Δ

188 mutants expressing two tP(AGG) on a plasmid had little effect on the temperature sensitivity

189 (Fig. 2D). As expected, northern analysis showed that trm8Δ [leu2+ tY(GUA)] strains had substantially

190 more tY(GUA) than the trm8Δ [leu2+] vector control strain at 30°C and 38.5°C (3.2-fold and 6.8-fold

191 more respectively) (Fig. S6). Similarly, trm8Δ mutants expressing two copies of tP(AGG) had more

192 tP(AGG) at 30°C and 38.5°C than the vector control, and the levels of the two control tRNAs were

193 unchanged in all strains at both temperatures. We conclude that although levels of both tY(GUA) and

De Zoysa 8

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 194 tP(AGG) were reduced in trm8Δ mutants at elevated temperatures in both YES and EMMC media,

195 tY(GUA) is the major physiologically important tRNA for these phenotypes.

196 Although tY(GUA) overexpression almost completely restored growth of S. pombe trm8Δ

197 mutants in YES and EMMC media at 38°C and 39°C, expression of both tY(GUA) and tP(AGG) was

198 required to completely suppress the growth defects in YES + glycerol media (Fig. S7). By contrast,

199 overexpression of tY(GUA) and tP(AGG) had no effect on the known temperature sensitivity of trm8Δ

200 mutants in YES media containing 5-FU (61, 62) (Fig. S7), perhaps due to reduced levels of Ψ and 5-

201 methyluridine modifications.

202

203 dhp1 mutations suppress the S. pombe trm8Δ growth defect and restore tY(GUA) and tP(AGG)

204 levels

205 To identify the mechanisms that restore growth to S. pombe trm8Δ mutants at elevated

206 temperatures, we isolated and analyzed spontaneous suppressors of the temperature sensitivity. One

207 major class of four trm8Δ suppressors were as temperature resistant as WT on YES and EMMC

208 media, and nearly as resistant on YES + 5-FU media (Fig. 3A, S8). Genome sequencing revealed that

209 these mutants each had distinct missense mutations in the RAT1 ortholog dhp1+. The dhp1 mutations

210 each occurred in a highly conserved residue, based on an alignment of 18 RAT1/dhp1+ eukaryotic

211 homologs from multiple phyla (Fig. 3B), and are presumably partial loss of function mutations as S.

212 pombe dhp1+, like S. cerevisiae RAT1, is an essential (63, 64).

213 Because we obtained four genetically independent S. pombe trm8Δ dhp1 mutants and very

214 few other mutations in the whole genome sequencing, it was highly likely that the dhp1 mutations

215 were responsible for the restoration of growth of trm8Δ dhp1 mutants. Consistent with this, a plasmid

216 expressing dhp1+ complemented the S. pombe trm8Δ dhp1-1 suppressor, resulting in temperature

217 sensitivity, but had no effect on WT or trm8Δ mutants (Fig. S9). Thus, we conclude that the dhp1

218 mutations were responsible for the rescue of growth at high temperature.

219 As Dhp1 encodes a 5'-3' exonuclease (65), it seemed highly likely that the S. pombe trm8Δ

220 dhp1 mutants prevented decay of tY(GUA) and tP(AGG) at non-permissive temperature. Indeed, we

De Zoysa 9

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 221 found that for each of two dhp1 suppressors, tY(GUA) levels were almost completely restored at

222 38.5°C, from 29% in the trm8Δ mutant to 85% and 82% in the trm8Δ dhp1-1 and trm8Δ dhp1-2 strains

223 respectively (Fig. 3C, 3D). Similarly, tP(AGG) levels were virtually completely restored at 38.5°C, from

224 16% in the trm8Δ mutant to 74% and 66% in the trm8Δ dhp1 suppressors, and the levels of the

225 control tRNA (tT(AGU)) was unaffected. Similar restoration of tY(GUA) and tP(AGG) levels was also

226 observed in the two other trm8Δ dhp1 suppressors at 38.5°C (Fig. S10). We conclude that, as for

227 RTD in S. cerevisiae modification mutants (22, 39, 40), decay of tY(GUA) and tP(AGG) in S. pombe

228 trm8Δ mutants occurs by 5'-3' exonucleolytic degradation of tRNA, providing strong evidence for

229 conservation of the RTD pathway in S. pombe.

230

231 Mutations in the GAAC pathway suppress the S. pombe trm8Δ growth defect and restore tRNA

232 levels

233 A second major group of six S. pombe trm8Δ suppressors was temperature resistant on YES

234 and EMMC media, but sensitive on YES + 5-FU media, and genome sequencing showed that these

235 suppressors each had distinct mutations in elements of the GAAC pathway (Fig. 4A, S11). Among

236 these, we found three trm8Δ suppressors with gcn2 mutations, one with a gcn1 mutation, and two

237 with tif221 mutations, encoding the translation initiation factor eIF2Bα (Table S2). Each of these genes

238 is known to be critical in the S. cerevisiae GAAC pathway (66-68), which is widely conserved in

239 eukaryotes, including S. pombe and mammals (69-74). In this pathway, amino acid starvation leads to

240 uncharged tRNAs that bind Gcn2 to activate its kinase domain, phosphorylation of eIF2α by Gcn2,

241 and derepression of translation of the transcription factor Gcn4, resulting in increased transcription of

242 nearly one tenth of the S. cerevisiae genes (66, 75-78). A similar massive transcription program

243 change occurs in S. pombe after amino acid starvation (79).

244 As expected, all S. pombe trm8Δ mutants with suppressing mutations in gcn2, gcn1, or tif221,

245 grew poorly on media containing 3-Amino-1,2,4-triazole (3-AT) (Fig. S11), the classical inducer of the

246 GAAC pathway (79-81). Furthermore, each of two S. pombe trm8Δ suppressors tested (with gcn2-1

De Zoysa 10

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 247 and tif221-2 mutations) was complemented by re-introduction of the WT gene (Fig. 4B), and a re-

248 constructed trm8Δ gcn2Δ strain was temperature resistant and sensitive to 3-AT (Fig. S12).

249 Consistent with their role as S. pombe trm8Δ suppressors, all six of the trm8Δ GAAC mutants

250 had increased levels of tY(GUA) and tP(AGG) at high temperature. After growth in YES media at

251 38.5°C, all trm8Δ GAAC mutants showed a 1.7-fold to 2.1-fold increase in tY(GUA) levels compared

252 to the parent trm8Δ mutant, and tP(AGG) levels were increased ~ 3 fold, whereas the controls

253 tT(AGU) and tV(AAC) did not have increased levels (Fig. 4C, 4D, S13). These results provided strong

254 evidence that the effect of the GAAC mutants was to increase tRNA levels to restore growth.

255

256 The temperature sensitivity of S. pombe trm8Δ mutants coincides precisely with the onset of

257 GAAC activation and tY(GUA) decay.

258 Since S. pombe trm8Δ suppressors in different components of the GAAC pathway all restored

259 tY(GUA) and tP(AAG) levels, we speculated that trm8Δ mutants activated the GAAC pathway at non-

260 permissive temperatures to somehow promote further loss of the tRNA. To establish the precise

261 connection between growth, tRNA levels, and GAAC activation in trm8Δ mutants, we measured each

262 parameter during liquid growth in rich media at a permissive temperature (30°C), and at three

263 elevated temperatures: 36.5°C, 37.5°C, and 38.5°C. In this experiment, trm8Δ mutants grew virtually

264 identically to WT control strains at 36.5°C and 37.5°C, and the growth defect was only obvious at

265 38.5°C (Fig. S14).

266 Strikingly, S. pombe trm8Δ mutants activated the GAAC pathway only at 38.5°C, the lowest

267 temperature at which the growth defect was obvious. We measured GAAC activation by measuring

268 mRNA levels of the known GAAC targets lys4+ and aro8+ (SPAC56E4.03) (79), which we had

269 previously used (82). At 38.5°C in trm8Δ mutants, we observed a 7.1-fold increase in lys4+ mRNA

270 levels (relative to the standard act1+), compared to that from WT or trm8Δ mutants at 30°C (5.3 vs

271 0.74) (Fig. 5A). By contrast, we found no measurable change in lys4+ mRNA levels in trm8Δ mutants

272 grown at 36.5°C and 37.5°C, relative to that observed in trm8Δ mutants at 30°C, or in WT cells at any

273 temperature. Moreover, the increase in relative lys4+ mRNA levels in trm8Δ mutants in YES media at

De Zoysa 11

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 274 38.5°C was almost as high as that observed in WT cells induced with 3-AT, and was completely

275 eliminated in trm8Δ gcn2-1 mutants. Examination of relative aro8+ mRNA levels gave a similar result

276 (Fig. S15): a substantial Gcn2-dependent increase in relative aro8+ mRNA levels at 38.5°C in trm8Δ

277 mutants, relative to 37.5°C (1.5 vs 0.45), and no change in relative aro8+ mRNA levels at 37.5°C in

278 trm8Δ mutants compared to WT (0.45 vs 0.44).

279 Consistent with the appearance of the S. pombe trm8Δ growth defect and the GAAC activation

280 only at 38.5°C, tY(GUA) decay was only significant at 38.5°C (Fig. 5B, 5C), and the gcn2-1 mutation

281 significantly prevented that tY(GUA) decay. Moreover, the gcn2-1 mutation appeared to exert its effect

282 on tY(GUA) levels by preventing decay. Using intron probes for the four tY(GUA) genes, we found

283 that levels of the primary transcript, corresponding to the largest species, were essentially unchanged

284 in trm8Δ gcn2-1 mutants, relative to trm8Δ mutants (Fig. 5B). As there was also no substantial

285 reduction of any pre-tY(GUA) species in trm8Δ mutants at 38.5°C, relative to WT levels, we infer that

286 tY(GUA) decay in Sp trm8Δ mutants occurred at the level of mature tRNA, as for RTD in S. cerevisiae

287 (39). Indeed, the relative increase in several pre-tY(GUA) species in trm8Δ mutants at elevated

288 temperatures, relative to WT, might imply a processing defect due to lack of m7G.

289 As tY(GUA) was the major physiologically relevant substrate in YES media (Fig. 2D), we

290 speculated that at 38.5°C, tY(GUA) decay might be driving the GAAC activation associated with the

291 trm8Δ growth defect. Alternatively, GAAC activation could be a consequence of both tY(GUA) and

292 tP(AAG) decay, reduced tRNA charging associated with trm8Δ mutants at high temperature, or partly

293 as a consequence of temperature stress itself, as a number of different stress conditions are known to

294 activate the GAAC pathway (72, 74, 83, 84).

295 To determine the extent to which reduced tRNA levels activated the GAAC pathway, we

296 examined GAAC induction of S. pombe trm8Δ strains after overproduction of tY(GUA) and tP(AGG),

297 using the same samples we used to show that overproduction of tY(GUA) suppressed the trm8Δ

298 temperature sensitivity (Fig. 2D, S6). As expected, relative lys4+ mRNA levels were increased in

299 trm8Δ [vector] strains grown at 38.5°C, compared to this strain at 30°C (4.17 vs 0.57, 7.3-fold) or to

300 WT at 38.5°C (7.0 fold), indicating GAAC activation (Fig. 5D). Notably, lys4+ mRNA levels were

De Zoysa 12

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 301 reduced 4.0-fold in the trm8Δ [tY(GUA)] strain compared to the corresponding trm8Δ [vector]

302 strain(from 4.2 to 1.04), and were not reduced in the trm8Δ [tP(AGG)] strain. Based on these results,

303 we conclude that reduced function of tY(GUA) is the primary cause of GAAC activation and

304 temperature sensitivity of trm8Δ mutants at 38.5°C. As charging of tY(GUA) was distinctly but

305 marginally reduced at 38.5°C relative to WT (Fig.S16), we cannot determine if the GAAC activation

306 was due to reduced levels of tY(GUA), or to a combination of reduced levels and charging (69, 76, 80,

307 82, 85).

308

309 In S. cerevisiae, mutation of the GAAC pathway exacerbates the effects of the RTD pathway

310 To investigate the evolutionary implications of the GAAC pathway on RTD, we examined the

311 consequences of deletion of GAAC components in S. cerevisiae trm8Δ trm4Δ mutants, which are

312 known to be temperature sensitive due to decay of tV(AAC) by the RTD pathway (18, 39). In contrast

313 to our results in S. pombe trm8Δ GAAC mutants, deletion of GCN1 or GCN2 exacerbated the

314 temperature sensitivity of S. cerevisiae trm8Δ trm4Δ mutants in both rich (YPD) and minimal complete

315 (SDC) media, and this phenotype was also observed upon deletion of the GAAC transcription factor

316 GCN4 (Fig. 6A). A similar exacerbated temperature sensitivity due to mutation of the GAAC pathway

317 was also observed in other S. cerevisiae modification mutants known to be subject to RTD (22, 39),

318 including trm8Δ, trm1Δ, tan1Δ, and tan1Δ trm44Δ mutants (Fig. S17).

319 To determine if the exacerbated growth defect of S. cerevisiae trm8Δ trm4Δ GAAC mutants

320 was due to exacerbated decay of tV(AAC), we analyzed tRNA levels after a four-hour temperature

321 shift from permissive to non-permissive temperature (27°C to 32°C). Consistent with the exacerbated

322 temperature sensitivity caused by the gcn1Δ and gcn2Δ mutations, tV(AAC) levels were further

323 reduced in both trm8Δ trm4Δ gcn1Δ and trm8Δ trm4Δ gcn2Δ mutants at 32°C, compared to the trm8Δ

324 trm4Δ mutant (11.8% and 18.5% vs 43.4%, relative to the values at 27°C) (Fig. 6B, 6C). As pre-

325 tV(AAC) levels were unchanged at 32°C in the trm8Δ trm4Δ gcn1Δ mutants (Fig S18), we infer that

326 tV(AAC) transcription was not affected by the gcn1Δ mutation, and that the enhanced loss of tV(AAC)

327 was due to enhanced decay of mature tV(AAC). If so, then the enhanced temperature sensitivity of

De Zoysa 13

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 328 trm8Δ trm4Δ mutants due to a gcn2Δ or a gcn4Δ mutation was likely also due to enhanced decay of

329 tV(AAC). Consistent with the conclusion that GAAC mutations enhanced RTD in S. cerevisiae, we

330 found that a met22Δ mutation, known to prevent RTD, reversed the enhanced temperature sensitivity

331 of a trm8Δ trm4Δ gcn2Δ strain relative to a trm8Δ trm4Δ mutant (Fig. S19).

332

333 The temperature sensitivity and tV(AAC) decay of S. cerevisiae trm8Δ trm4Δ mutants coincides

334 with GAAC activation.

335 Since GAAC mutations exacerbated the RTD growth defect and enhanced the decay of an S.

336 cerevisiae trm8Δ trm4Δ mutant at 32°C, it seemed likely that the GAAC pathway was activated in the

337 trm8Δ trm4Δ mutant. To evaluate GAAC activation, we measured mRNA levels of the Gcn4 target

338 genes LYS1 and HIS5 after the 4 hour temperature shift of trm8Δ trm4Δ mutants to 32°C, using the

339 same RNA as in the Northern analysis of decay (Fig. 6B, 6C, S18). RT-qPCR analysis showed that

340 trm8Δ trm4Δ mutants had a large increase in relative LYS1 mRNA levels at 32°C, compared to 27°C

341 (22.2 vs 0.81, 27.4-fold), or to WT cells at either 27°C or 32°C (Fig. 6D), showing that the GAAC

342 activation was specific to the trm8Δ trm4Δ mutant at 32°C. We observed a similar activation of the

343 GCN4 target HIS5 in the trm8Δ trm4Δ mutant at 32°C. This GAAC activation was correlated with a

344 modest but distinct increase in uncharged tV(AAC) commensurate with the reduced tV(AAC) levels

345 (Fig. S20). Furthermore, we found that overproduction of tV(AAC) in trm8Δ trm4Δ mutants

346 suppressed induction of the GAAC pathway (Fig. 6E), showing that GAAC activation in trm8Δ trm4Δ

347 mutants was due to the reduced function of tV(AAC). Thus, in both S. cerevisiae trm8Δ trm4Δ mutants

348 and S pombe trm8Δ mutants, degradation of a single biologically relevant tRNA is the cause of GAAC

349 induction, which then either promotes further decay in S. pombe, or prevents further decay in S.

350 cerevisiae (Fig. 7).

351 352 Discussion

353 The results described here provide strong evidence that the RTD pathway is conserved

354 between the distantly related species S. cerevisiae and S. pombe. We have shown that the

355 temperature sensitivity of S. pombe trm8Δ mutants is due to reduced levels of tY(GUA) and to some

De Zoysa 14

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 356 extent tP(AGG), and is efficiently suppressed by mutations in the 5'-3' exonuclease Dhp1 that

357 concomitantly restore the levels of these tRNAs, strongly suggesting decay of tY(GUA) and tP(AGG)

2,2 4 358 by the RTD pathway. As RTD is triggered in S. cerevisiae strains lacking m G26 or ac C12, as well as

7 359 in strains lacking m G46, (22, 39), we speculate that the RTD pathway will also act in S. pombe strains

360 lacking other body modifications. Furthermore, given the large evolutionary distance between S.

361 cerevisiae and S. pombe, we speculate that the RTD pathway is conserved throughout eukaryotes.

362 The existence of a mammalian RTD pathway could explain the reduced levels of specific tRNA

363 species in mouse strains lacking m5C in their tRNAs (33, 86) and in mouse embryonic stem cells

364 lacking m7G in their tRNAs (46), and might explain other phenotypes associated with mutations in

365 METTL1 or WDR4 (29, 30, 46, 87).

366 It is puzzling that although tP(AGG) is substantially more degraded than tY(GUA) in S. pombe

367 trm8Δ mutants at elevated temperatures, the temperature sensitivity of the mutants in both rich and

368 minimal media is primarily due to decay of tY(GUA). One possible explanation of this result is that

369 tY(GUA) levels might be more limiting in the cell than tP(AGG) levels at elevated temperature, relative

370 to the number of their respective cognate codons requiring decoding. This type of argument was

371 advanced as a possible explanation for why the growth defects of i6A-lacking S. pombe tit1Δ mutants

372 were rescued by increased expression of tY(GUA), but not by any of the other four Tit1 tRNA

373 substrates (88). A second, and less likely, interpretation is that tP(AGG) decoding might be

374 compensated by other tRNAPro isodecoders specific for the CCN codon box. This explanation is based

375 on the finding that in S. cerevisiae, deletion of the two tP(AGG) genes is viable, implying that the 10

376 tP(UGG) isodecoders can decode all four proline CCN codons (89). However, as S. pombe has six

377 tP(AGG) genes and only two tP(UGG) genes (as well as one tP(CGG)), it seems unlikely that the loss

378 of almost all of the tP(AGG) in trm8Δ mutants can be efficiently compensated by the small number of

379 tP(UGG) species (assuming that tRNA expression from each gene is comparable).

380 It is not immediately clear why tP(AGG) and tY(GUA) are the specific tRNAs subject to RTD in

381 S. pombe trm8Δ mutants. Based on current understanding of RTD determinants in S. cerevisiae, RTD

382 substrate specificity is determined by stability of the stacked acceptor and T-stem, with contributions

De Zoysa 15

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 383 to stability from the tertiary fold that are reduced in modification mutants, and some contributions from

384 the other two stems, but not the anticodon loop (18, 22, 39-42). tP(AGG) may be an RTD substrate

385 because it is predicted to have a less stable acceptor and T-stem than most Trm8 substrates (Table

386 S1) (90). Furthermore, the destabilizing C4-A69 mismatch in the middle of the tP(AGG) acceptor stem

387 might be expected to lead to increased local breathing at the 5' end, which is likely important for

388 recognition by the 5'-3' exonucleases Xrn1 and Rat1, as the Xrn1 active site binds the three most 5'

389 nucleotides (91). However, it is more difficult to rationalize why tY(GUA) is a substrate for RTD in

390 trm8Δ mutants, as its acceptor and T-stem are predicted to be moderately stable among Trm8

391 substrates. However, tY(GUA) does have a destabilizing N27-N43 pair (C27-U43 for 3 isodecoders, and

392 U27-U43 for one isodecoder), which might reduce the stability of the tertiary fold by affecting stability of

393 the adjacent tertiary 26-44 interaction (92-94).

394 It seems likely by analogy with RTD in S. cerevisiae that tRNA decay by Dhp1 in S. pombe

395 involves retrograde transport of mature tRNA from the cytoplasm back to the nucleus. In S.

396 cerevisiae, Rat1 is nuclear (95) and catalyzes a substantial amount of the decay of mature tV(AAC) in

397 S. cerevisiae trm8Δ trm4Δ mutants (39), implying that the retrograde transport pathway is required to

398 deliver the tV(AAC) substrate to the nucleus (5, 96-99). We argued above that mature tY(GUA) was

399 the actual substrate for decay in S. pombe trm8Δ mutants. As pre-tRNA splicing in S. pombe is known

400 to initiate on the surface of mitochondria (100), mature tY(GUA) is almost certainly transported back to

401 the nucleus by the tRNA retrograde pathway to be degraded by Dhp1(64, 101). Retrograde transport

402 could occur in S. pombe trm8Δ mutants driven in part by the hypomodification, as speculated

403 previously in S. cerevisiae and shown for S. cerevisiae trm1Δ mutants (99).

404 It seems likely that the 5-FU sensitivity of S. pombe trm8Δ mutants is due to decay of multiple

405 tRNA species in the presence of the drug, caused by the reduced levels of Ψ and m5U (48-50), in

406 addition to the lack of m7G. This interpretation is consistent with the lack of suppression of the 5-FU

407 sensitivity of trm8Δ mutants by tY(GUA) and tP(AGG), and its almost complete suppression by dhp1

408 mutations, and is consistent with the enhanced 5-FU sensitivity of a number of tRNA body

409 modification mutants in S. cerevisiae (61).

De Zoysa 16

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 410 Our finding that loss of function of tRNA due to tRNA decay is itself the trigger for induction of

411 the GAAC response in both S. pombe trm8Δ mutants and S. cerevisiae trm8Δ trm4Δ mutants

412 suggests an intimate relationship between reduced tRNA function and GAAC activation. The loss of

413 functional tRNA is the proximal cause of GAAC induction, because in each organism the GAAC

414 pathway is activated at the lowest temperature at which tRNA decay and a growth defect is observed,

415 and in each organism overproduction of the physiologically relevant tRNA represses GAAC induction.

416 The GAAC pathway has previously been implicated in the biology of a number of anticodon loop

417 modifications (82, 102, 103). Robust constitutive GAAC induction is observed in S. cerevisiae and S.

418 pombe trm7Δ mutants (lacking Nm32 and Nm34) and S. cerevisiae pus3Δ mutants (lacking Ψ38 and

419 Ψ39) (82, 103), each of which has a constitutive growth defect (104, 105), and GAAC induction is

420 known to be Gcn2-dependent in S. cerevisiae trm7Δ mutants (82). By contrast, S. cerevisiae mutants

421 lacking either the mcm5U or the s2U moiety of mcm5s2U induce the GAAC pathway independently of

422 Gcn2 at 30°C (102) and are temperature sensitive at 37°C (106). Our finding that S. pombe trm8Δ

423 and S. cerevisiae trm8Δ trm4Δ mutants each trigger Gcn2-dependent GAAC induction only at the

424 temperature that the growth defect is observed is consistent with Gcn2-dependent GAAC induction in

425 S. cerevisiae anticodon loop modification mutants with a constitutive growth defect.

426 It is striking that the induction of the GAAC response due to tRNA decay in S. pombe trm8Δ

427 mutants and S. cerevisiae trm8Δ trm4Δ mutants has opposite consequences in each organism. In S.

428 pombe trm8Δ mutants, activation of the GAAC response exacerbates the growth defect, as mutation

429 of any of three components (gcn1, gcn2, or tif221) protects against decay. Activation of the GAAC

430 pathway is also part of the reason that S. cerevisiae trm7Δ mutants grow poorly (82), and defects in

431 the integrated stress response pathway (ISR) in humans are implicated in disease phenotypes (107-

432 109). By contrast, in S. cerevisiae trm8Δ trm4Δ mutants, activation of the GAAC response rescues the

433 growth defect, as deletion of any of three GAAC components (gcn1Δ, gcn2Δ, or gcn4Δ) exacerbates

434 the growth defect. Although this rescue of RTD in S. cerevisiae by GAAC activation is opposite to that

435 in S. pombe, it is consistent with a concerted stress response. Moreover the finding that S. cerevisiae

436 gcn4Δ mutations exacerbated RTD nearly as much as gcn1Δ or gcn2Δ mutations has mechanistic

De Zoysa 17

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 437 implications. Deletion of GCN1 or GCN2 each prevent sensing of tRNA status, and the consequent

438 eIF2α phosphorylation, reduced translation initiation, and reduced global translation. However, as

439 GCN4 is downstream of the sensing machinery, we infer that the gcn4Δ-mediated GAAC effects on

440 RTD in S. cerevisiae are mostly due to lack of transcription activation by Gcn4, rather than the

441 increased eIF2α phosphorylation leading to the reduction in translation initiation and overall

442 translation.

443 Our evidence that GAAC activation has opposite consequences in S. pombe and S. cerevisiae

444 is likely due to the differential GAAC response in genes known to regulate RTD. Although in each

445 organism the GAAC pathway is known to regulate the transcription of more than 500 genes (75, 77,

446 79, 110), there are distinct differences in the GAAC response in the two species. For example, it is

447 known that the GAAC response to amino acid starvation results in repression of methionine synthesis

448 genes in S. pombe but induction of these genes in S. cerevisiae (79). As a met22Δ mutation is known

449 to inhibit RTD in S. cerevisiae, this opposite GAAC activation effect on methionine genes in the two

450 organisms is in the wrong direction to explain the opposite RTD effects of the GAAC pathway. Other

451 possible explanations for the differential effects of the GAAC pathway on RTD include differential

452 regulation of the synthesis or biochemical activity of RTD regulators such as EF1A, aminoacyl tRNA

453 synthetases, pol III transcription (22, 111), or 5'-3' exonucleases (39, 42), as well as changes in any

454 number of indirect effectors affecting overall levels or availability of tRNA and/or nucleases. Moreover,

455 the overall stress response pathways are substantially different between S. cerevisiae and S. pombe.

456 In S. cerevisiae, Gcn2 is the sole eIF2 kinase regulating stress responses (78, 112, 113), whereas in

457 S. pombe three different eIF2 kinases (Gcn2, Hri1 and Hri2) (114) each respond to a diverse set of

458 stress treatments (72, 74, 83, 84, 107). The differences in kinases affecting eIF2α phosphorylation

459 implies substantial differences between the two species in regulation of all sorts of combinations of

460 stress response, which might be occurring at elevated temperature when tRNA decay is occurring

461 (115).

462 The results outlined here underscore that GAAC activation occurs in S. cerevisiae and S.

463 pombe trm8Δ modification mutants precisely at the point of observed growth stress due to tRNA

De Zoysa 18

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 464 decay, albeit with different effects in S. pombe trm8Δ mutants and S. cerevisiae trm8Δ trm4Δ mutants.

465 These results, coupled with the constitutive GAAC activation in S. pombe and S. cerevisiae trm7Δ

466 mutants and S. cerevisiae pus3Δ mutants (82, 103), fuel speculation that the GAAC response will also

467 be activated in mammals and other eukaryotes with tRNA modification mutants or mutations that

468 result in growth phenotypes due to reduced tRNA function.

469 As Trm8 is phosphorylated and likely inactivated by treatment of HEK293 cells with insulin-like

470 growth factor-1 (116), it seems plausible that if RTD is conserved in mammals, such m7G modification

471 dynamics could be used to regulate tRNA levels physiologically. There is growing evidence that levels

472 of a number of modifications are under dynamic control in different conditions (117, 118). There is

473 also evidence that regulation of tRNA expression plays an important role in differentiation and

474 proliferation, and is also a characteristic of breast cancer (119-123). For example, tRNAArg iso-

475 acceptors have different expression in differentiated vs proliferating cells (122) and tR(UCU) iso-

476 decoders show tissue specific regulation of expression (124). It remains to be determined if regulated

477 changes in expression, phosphorylation, or biochemical activity of METTL1 or WDR4 result in altered

478 m7G levels and consequent changes in tRNA levels that physiologically regulate expression.

479

De Zoysa 19

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 480 481 Materials and Methods

482 Yeast Strains

483 S. pombe haploid WT and two independent S. pombe trm8Δ::kanMX strains were derived from

484 SP286 (ade6-M210/ade6-M216, leu1-32/leu1-32, ura4-D18/ura4-D18 h+/h+)(125), and were obtained

485 from Dr. Jeffrey Pleiss. S. pombe trm8Δ::kanMX strains were also generated from haploid WT strains

486 by PCR amplification of trm8Δ::kanMX DNA, followed by linear transformation using lithium acetate

487 (126). S. cerevisiae deletion strains are shown in Table S3, and were constructed by linear

488 transformation with PCR amplified DNA from the appropriate knockout strain (127). All strains were

489 confirmed before use by PCR amplification.

490

491 Plasmids

492 Plasmids used in this study are listed in Table S4. AB553-1 was constructed by insertion of a

493 NotI restriction site between the Pnmt1 promoter and the PstI site of the pREP3X plasmid. The S.

+ 494 pombe plasmid expressing S. pombe Ptrm8+ trm8 (ETD 67-1) was constructed by inserting PCR

495 amplified DNA genomic DNA (including 1000 bp upstream and 1000 bp downstream) into the NotI

+ 496 and XhoI sites of AB 553-1, removing the Pnmt1 promoter. Plasmids expressing S. pombe Pdhp1 dhp1

497 or S. pombe tRNA genes were constructed using the same approach, including ~ 300 bp upstream

+ 498 and 300 bp downstream for the tRNA genes. S. pombe plasmids expressing Pnmt1** gcn2 (low

+ 499 strength, no message in thiamine) or Pnmt1* tif221 (media strength, no message in thiamine) were

500 constructed by PCR amplification of the respective coding sequence from S. pombe WT genomic

501 DNA (including introns), and insertion into the XhoI and BamHI sites of the pREP81X or pRE41X

502 vectors respectively.

503

504 Yeast media and growth conditions

505 S. pombe strains were grown at desired temperatures in rich (YES) media (containing 0.5% yeast

506 extract, 3% glucose, and supplements of 225 mg/l of adenine, uracil, leucine, histidine and lysine), or

507 Edinburgh minimal media (EMM) containing glucose and the same supplements, as well as similar

De Zoysa 20

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 508 amounts of relevant auxotrophic requirements. Minimal complete (EMM-C) media was supplemented

509 with 225 mg/l of all amino acids, adenine, and uracil, as well as 100 mg/l of para-amino benzoic acid

510 and inositol, and 1125 mg/l of leucine for Leu- auxotrophs (79). For temperature shift experiments,

511 cells were grown in YES or EMMC media at 30°C to OD600 ~ 0.5, diluted to ~ 0.1 OD in pre-warmed

512 media at the desired temperature, grown to OD ~ 0.5, harvested at 4°C, washed with ice cold water,

513 frozen on dry ice, and stored at -80°C. To select spontaneous suppressors of S. pombe trm8Δ

514 mutants, cells were grown overnight in YES media at 30°C and ~107 cells were plated on YES media

515 plates at 38°C and 39°C. S. cerevisiae strains were grown in rich (YPD) media (containing 1% yeast

516 extract, 2% peptone, 2% dextrose, and 80 mg/L adenine hemisulfate), or minimal complete (SDC)

517 media (128) as indicated, and temperature shift experiments were performed as described for S.

518 pombe. All experiments with measurements were performed in biological triplicate, unless otherwise

519 noted.

520

521 Bulk RNA preparation and northern blot analysis

522 For northern analysis, 2 or 3 biological replicates were grown in parallel, and then bulk RNA

523 was isolated from ~ 3-5 OD pellets using glass beads and phenol (129) (for S. pombe) or hot phenol

524 (for S. cerevisiae), resolved on a 10% polyacrylamide (19:1), 7M urea, 1X TBE gel, transferred to

525 Amersham Hybond-N+ membrane, and analyzed by hybridization to 5’ 32P-labeled DNA probes (Table

526 S5) as described (18). For analyzing tRNA charging levels of both S. pombe and S. cerevisiae, RNA

527 was prepared under acidic conditions (pH 4.5), resolved on a 6.5 % polyacrylamide (19:1), 8 M urea,

528 0.1 M sodium acetate (pH 5.0) gel at 4°C, and analyzed as described. (18).

529

530 Quantitative RT-PCR analysis

531 Strains were grown in triplicate to log phase and bulk RNA was prepared from 2-5 OD pellets

532 using acid washed glass beads and phenol. Then, RNA was treated with RQ1 RNase-free DNase

533 (Promega), reverse transcribed with Superscript II Reverse Transcriptase, and quantitative PCR was

534 performed on the cDNA as previously described (130).

De Zoysa 21

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 535

536 Isolation and purification of tRNA

537 S. pombe WT and trm8Δ mutant strains were grown to ~ 0.5 OD in YES media at 30°C. Then

538 bulk low molecular weight RNA was extracted from ~ 300 OD of pellets by using hot phenol, and

539 tRNAs were purified using 5'-biotinylated oligonucleotides complementary to the corresponding tRNAs

540 (Table S6) as previously described (131).

541

542 HPLC analysis of nucleosides of purified tRNA

543 Purified tRNAs (~ 1.25 µg) were digested to nucleosides by treatment with P1 nuclease,

544 followed by phosphotase, as previously described (131), and nucleosides were analyzed by HPLC at

545 pH 7.0 as previously described (132).

546

547 Whole genome sequencing

548 Whole genome sequencing was performed by the University of Rochester Genomics Center at

549 a read depth of 20-110 per genome nucleotide.

550

551 Acknowledgements

552 We are grateful to Dr. Jeffrey Pleiss for the gift of S. pombe strains and invaluable help in

553 preparing samples for whole genome sequencing. We also thank Dr. Elizabeth Grayhack, and the

554 members of the Phizicky lab and Grayhack lab for valuable discussions and comments during the

555 course of this work. This research was supported by NIH Grant GM052347 to E.M.P.

556 .

De Zoysa 22

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 557 References

558

559 1. Phizicky EM, Hopper AK. tRNA biology charges to the front. Genes Dev. 2010;24(17):1832- 560 60.

561 2. Ramos J, Fu D. The emerging impact of tRNA modifications in the brain and nervous system. 562 Biochim Biophys Acta Gene Regul Mech. 2019;1862(3):412-28.

563 3. Boccaletto P, Machnicka MA, Purta E, Piatkowski P, Baginski B, Wirecki TK, et al. 564 MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res. 565 2018;46(D1):D303-D7.

566 4. Pereira M, Francisco S, Varanda AS, Santos M, Santos MAS, Soares AR. Impact of tRNA 567 Modifications and tRNA-Modifying Enzymes on Proteostasis and Human Disease. International 568 journal of molecular sciences. 2018;19(12).

569 5. Hopper AK. Transfer RNA post-transcriptional processing, turnover, and subcellular dynamics 570 in the yeast Saccharomyces cerevisiae. Genetics. 2013;194(1):43-67.

571 6. Urbonavicius J, Qian O, Durand JMB, Hagervall TG, Bjork GR. Improvement of reading frame 572 maintenance is a common function for several tRNA modifications. Embo Journal. 2001;20(17):4863- 573 73.

574 7. Lecointe F, Namy O, Hatin I, Simos G, Rousset JP, Grosjean H. Lack of pseudouridine 38/39 575 in the anticodon arm of yeast cytoplasmic tRNA decreases in vivo recoding efficiency. J Biol Chem. 576 2002;277(34):30445-53.

577 8. Waas WF, de Crecy-Lagard V, Schimmel P. Discovery of a gene family critical to wyosine 578 base formation in a subset of phenylalanine-specific transfer RNAs. J Biol Chem. 579 2005;280(45):37616-22.

580 9. El Yacoubi B, Hatin I, Deutsch C, Kahveci T, Rousset JP, Iwata-Reuyl D, et al. A role for the 581 universal Kae1/Qri7/YgjD (COG0533) family in tRNA modification. EMBO J. 2011;30(5):882-93.

582 10. Gerber AP, Keller W. An adenosine deaminase that generates inosine at the wobble position 583 of tRNAs. Science. 1999;286(5442):1146-9.

584 11. Murphy FVt, Ramakrishnan V. Structure of a purine-purine wobble in the decoding 585 center of the ribosome. Nat Struct Mol Biol. 2004;11(12):1251-2.

586 12. Bjork GR, Huang B, Persson OP, Bystrom AS. A conserved modified wobble nucleoside 587 (mcm5s2U) in lysyl-tRNA is required for viability in yeast. RNA. 2007;13(8):1245-55.

588 13. Weixlbaumer A, Murphy FVt, Dziergowska A, Malkiewicz A, Vendeix FA, Agris PF, et al. 589 Mechanism for expanding the decoding capacity of transfer RNAs by modification of uridines. Nat 590 Struct Mol Biol. 2007;14(6):498-502.

591 14. Muramatsu T, Nishikawa K, Nemoto F, Kuchino Y, Nishimura S, Miyazawa T, et al. Codon and 592 amino-acid specificities of a transfer RNA are both converted by a single post-transcriptional 593 modification. Nature. 1988;336(6195):179-81.

594 15. Putz J, Florentz C, Benseler F, Giege R. A single methyl group prevents the mischarging of a 595 tRNA. Nat Struct Biol. 1994;1(9):580-2. De Zoysa 23

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 596 16. Helm M, Giege R, Florentz C. A Watson-Crick base-pair-disrupting methyl group (m1A9) is 597 sufficient for cloverleaf folding of human mitochondrial tRNALys. Biochemistry. 1999;38(40):13338-46.

598 17. Kadaba S, Krueger A, Trice T, Krecic AM, Hinnebusch AG, Anderson J. Nuclear surveillance 599 and degradation of hypomodified initiator tRNAMet in S. cerevisiae. Genes Dev. 2004;18(11):1227- 600 40.

601 18. Alexandrov A, Chernyakov I, Gu W, Hiley SL, Hughes TR, Grayhack EJ, et al. Rapid tRNA 602 decay can result from lack of nonessential modifications. Mol Cell. 2006;21(1):87-96.

603 19. Anderson J, Phan L, Cuesta R, Carlson BA, Pak M, Asano K, et al. The essential Gcd10p- 604 Gcd14p nuclear complex is required for 1-methyladenosine modification and maturation of initiator 605 methionyl-tRNA. Gene Dev. 1998;12(23):3650-62.

606 20. Alexandrov A, Grayhack EJ, Phizicky EM. tRNA m7G methyltransferase Trm8p/Trm82p: 607 evidence linking activity to a growth phenotype and implicating Trm82p in maintaining levels of active 608 Trm8p. RNA. 2005;11(5):821-30.

609 21. Kotelawala L, Grayhack EJ, Phizicky EM. Identification of yeast tRNA Um(44) 2'-O- 610 methyltransferase (Trm44) and demonstration of a Trm44 role in sustaining levels of specific 611 tRNA(Ser) species. RNA. 2008;14(1):158-69.

612 22. Dewe JM, Whipple JM, Chernyakov I, Jaramillo LN, Phizicky EM. The yeast rapid tRNA decay 613 pathway competes with elongation factor 1A for substrate tRNAs and acts on tRNAs lacking one or 614 more of several modifications. RNA. 2012;18(10):1886-96.

615 23. Gillis D, Krishnamohan A, Yaacov B, Shaag A, Jackman JE, Elpeleg O. TRMT10A dysfunction 616 is associated with abnormalities in glucose homeostasis, short stature and microcephaly. J Med 617 Genet. 2014;51(9):581-6.

618 24. Cosentino C, Toivonen S, Diaz Villamil E, Atta M, Ravanat JL, Demine S, et al. Pancreatic 619 beta-cell tRNA hypomethylation and fragmentation link TRMT10A deficiency with diabetes. Nucleic 620 Acids Res. 2018;46(19):10302-18.

621 25. Najmabadi H, Hu H, Garshasbi M, Zemojtel T, Abedini SS, Chen W, et al. Deep sequencing 622 reveals 50 novel genes for recessive cognitive disorders. Nature. 2011;478(7367):57-63.

623 26. Davarniya B, Hu H, Kahrizi K, Musante L, Fattahi Z, Hosseini M, et al. The Role of a Novel 624 TRMT1 Gene Mutation and Rare GRM1 Gene Defect in Intellectual Disability in Two Azeri Families. 625 PloS one. 2015;10(8):e0129631.

626 27. Zhang K, Lentini JM, Prevost CT, Hashem MO, Alkuraya FS, Fu D. An intellectual disability- 627 associated missense variant in TRMT1 impairs tRNA modification and reconstitution of enzymatic 628 activity. Human mutation. 2020;41(3):600-7.

629 28. Dewe JM, Fuller BL, Lentini JM, Kellner SM, Fu D. TRMT1-Catalyzed tRNA Modifications Are 630 Required for Redox Homeostasis To Ensure Proper Cellular Proliferation and Oxidative Stress 631 Survival. Mol Cell Biol. 2017;37(21).

632 29. Shaheen R, Abdel-Salam GM, Guy MP, Alomar R, Abdel-Hamid MS, Afifi HH, et al. Mutation 633 in WDR4 impairs tRNA m(7)G46 methylation and causes a distinct form of microcephalic primordial 634 dwarfism. Genome biology. 2015;16(1):210.

635 30. Chen X, Gao Y, Yang L, Wu B, Dong X, Liu B, et al. Speech and language delay in a patient 636 with WDR4 mutations. Eur J Med Genet. 2018;61(8):468-72. De Zoysa 24

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 637 31. Trimouille A, Lasseaux E, Barat P, Deiller C, Drunat S, Rooryck C, et al. Further delineation of 638 the phenotype caused by biallelic variants in the WDR4 gene. Clinical genetics. 2018;93(2):374-7.

639 32. Martinez FJ, Lee JH, Lee JE, Blanco S, Nickerson E, Gabriel S, et al. Whole exome 640 sequencing identifies a splicing mutation in NSUN2 as a cause of a Dubowitz-like syndrome. J Med 641 Genet. 2012;49(6):380-5.

642 33. Tuorto F, Liebers R, Musch T, Schaefer M, Hofmann S, Kellner S, et al. RNA cytosine 643 methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nat Struct Mol Biol. 644 2012;19(9):900-5.

645 34. Abbasi-Moheb L, Mertel S, Gonsior M, Nouri-Vahid L, Kahrizi K, Cirak S, et al. Mutations in 646 NSUN2 cause autosomal-recessive intellectual disability. Am J Hum Genet. 2012;90(5):847-55.

647 35. Kadaba S, Wang X, Anderson JT. Nuclear RNA surveillance in Saccharomyces cerevisiae: 648 Trf4p-dependent polyadenylation of nascent hypomethylated tRNA and an aberrant form of 5S rRNA. 649 RNA. 2006;12(3):508-21.

650 36. LaCava J, Houseley J, Saveanu C, Petfalski E, Thompson E, Jacquier A, et al. RNA 651 degradation by the exosome is promoted by a nuclear polyadenylation complex. Cell. 652 2005;121(5):713-24.

653 37. Vanacova S, Wolf J, Martin G, Blank D, Dettwiler S, Friedlein A, et al. A new yeast poly(A) 654 polymerase complex involved in RNA quality control. PLoS Biol. 2005;3(6):e189.

655 38. Gudipati RK, Xu Z, Lebreton A, Seraphin B, Steinmetz LM, Jacquier A, et al. Extensive 656 degradation of RNA precursors by the exosome in wild-type cells. Mol Cell. 2012;48(3):409-21.

657 39. Chernyakov I, Whipple JM, Kotelawala L, Grayhack EJ, Phizicky EM. Degradation of several 658 hypomodified mature tRNA species in Saccharomyces cerevisiae is mediated by Met22 and the 5'-3' 659 exonucleases Rat1 and Xrn1. Genes Dev. 2008;22(10):1369-80.

660 40. Whipple JM, Lane EA, Chernyakov I, D'Silva S, Phizicky EM. The yeast rapid tRNA decay 661 pathway primarily monitors the structural integrity of the acceptor and T-stems of mature tRNA. Genes 662 Dev. 2011;25(11):1173-84.

663 41. Guy MP, Young DL, Payea MJ, Zhang X, Kon Y, Dean KM, et al. Identification of the 664 determinants of tRNA function and susceptibility to rapid tRNA decay by high-throughput in vivo 665 analysis. Genes Dev. 2014;28(15):1721-32.

666 42. Payea MJ, Sloma MF, Kon Y, Young DL, Guy MP, Zhang X, et al. Widespread temperature 667 sensitivity and tRNA decay due to mutations in a yeast tRNA. RNA. 2018;24(3):410-22.

668 43. Murguia JR, Belles JM, Serrano R. The yeast HAL2 nucleotidase is an in vivo target of salt 669 toxicity. Journal of Biological Chemistry. 1996;271(46):29029-33.

670 44. Dichtl B, Stevens A, Tollervey D. Lithium toxicity in yeast is due to the inhibition of RNA 671 processing enzymes. Embo Journal. 1997;16(23):7184-95.

672 45. Yun JS, Yoon JH, Choi YJ, Son YJ, Kim S, Tong L, et al. Molecular mechanism for the 673 inhibition of DXO by adenosine 3',5'-bisphosphate. Biochemical and biophysical research 674 communications. 2018;504(1):89-95.

De Zoysa 25

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 675 46. Lin S, Liu Q, Lelyveld VS, Choe J, Szostak JW, Gregory RI. Mettl1/Wdr4-Mediated m(7)G 676 tRNA Methylome Is Required for Normal mRNA Translation and Embryonic Stem Cell Self-Renewal 677 and Differentiation. Mol Cell. 2018;71(2):244-55 e5.

678 47. Okamoto M, Fujiwara M, Hori M, Okada K, Yazama F, Konishi H, et al. tRNA Modifying 679 Enzymes, NSUN2 and METTL1, Determine Sensitivity to 5-Fluorouracil in HeLa Cells. PLoS genetics. 680 2014;10(9):e1004639.

681 48. Frendewey DA, Kladianos DM, Moore VG, Kaiser, II. Loss of tRNA 5-methyluridine 682 methyltransferase and pseudouridine synthetase activities in 5-fluorouracil and 1-(tetrahydro-2- 683 furanyl)-5-fluorouracil (ftorafur)-treated Escherichia coli. Biochimica et biophysica acta. 684 1982;697(1):31-40.

685 49. Santi DV, Hardy LW. Catalytic mechanism and inhibition of tRNA (uracil-5-)methyltransferase: 686 evidence for covalent catalysis. Biochemistry. 1987;26(26):8599-606.

687 50. Huang L, Pookanjanatavip M, Gu X, Santi DV. A conserved aspartate of tRNA pseudouridine 688 synthase is essential for activity and a probable nucleophilic catalyst. Biochemistry. 1998;37(1):344- 689 51.

690 51. Watanabe K, Miyagawa R, Tomikawa C, Mizuno R, Takahashi A, Hori H, et al. Degradation of 691 initiator tRNAMet by Xrn1/2 via its accumulation in the nucleus of heat-treated HeLa cells. Nucleic 692 Acids Res. 2013;41(8):4671-85.

693 52. Parfrey LW, Lahr DJ, Knoll AH, Katz LA. Estimating the timing of early eukaryotic 694 diversification with multigene molecular clocks. Proc Natl Acad Sci U S A. 2011;108(33):13624-9.

695 53. Alexandrov A, Martzen MR, Phizicky EM. Two proteins that form a complex are required for 7- 696 methylguanosine modification of yeast tRNA. Rna. 2002;8(10):1253-66.

697 54. Leulliot N, Chaillet M, Durand D, Ulryck N, Blondeau K, van Tilbeurgh H. Structure of the yeast 698 tRNA m7G methylation complex. Structure. 2008;16(1):52-61.

699 55. Pandolfini L, Barbieri I, Bannister AJ, Hendrick A, Andrews B, Webster N, et al. METTL1 700 Promotes let-7 MicroRNA Processing via m7G Methylation. Mol Cell. 2019;74(6):1278-90 e9.

701 56. Zhang LS, Liu C, Ma H, Dai Q, Sun HL, Luo G, et al. Transcriptome-wide Mapping of Internal 702 N(7)-Methylguanosine Methylome in Mammalian mRNA. Mol Cell. 2019;74(6):1304-16 e8.

703 57. McCutchan T, Silverman S, Kohli J, Soll D. Nucleotide sequence of phenylalanine transfer 704 RNA from Schizosaccharomyces pombe: implications for transfer RNA recognition by yeast 705 phenylalanyl-tRNA synthetase. Biochemistry. 1978;17(9):1622-8.

706 58. Vogeli G. The nucleotide sequence of tRNA tyrosine from the fission yeast 707 Schizosaccharomyces pombe. Nucleic Acids Res. 1979;7(4):1059-65.

708 59. Chan PP, Lowe TM. GtRNAdb 2.0: an expanded database of transfer RNA genes identified in 709 complete and draft genomes. Nucleic Acids Res. 2016;44(D1):D184-9.

710 60. Matsumoto K, Toyooka T, Tomikawa C, Ochi A, Takano Y, Takayanagi N, et al. RNA 711 recognition mechanism of eukaryote tRNA (m7G46) methyltransferase (Trm8-Trm82 complex). FEBS 712 Lett. 2007;581(8):1599-604.

713 61. Gustavsson M, Ronne H. Evidence that tRNA modifying enzymes are important in vivo targets 714 for 5-fluorouracil in yeast. RNA. 2008;14(4):666-74. De Zoysa 26

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 715 62. Mojardin L, Botet J, Moreno S, Salas M. segregation and organization are 716 targets of 5'-Fluorouracil in eukaryotic cells. . 2015;14(2):206-18.

717 63. Sugano S, Shobuike T, Takeda T, Sugino A, Ikeda H. Molecular analysis of the dhp1+ gene of 718 Schizosaccharomyces pombe: an essential gene that has homology to the DST2 and RAT1 genes of 719 Saccharomyces cerevisiae. Molecular & general genetics : MGG. 1994;243(1):1-8.

720 64. Shobuike T, Tatebayashi K, Tani T, Sugano S, Ikeda H. The dhp1(+) gene, encoding a 721 putative nuclear 5'-->3' exoribonuclease, is required for proper chromosome segregation in fission 722 yeast. Nucleic Acids Res. 2001;29(6):1326-33.

723 65. Xiang S, Cooper-Morgan A, Jiao X, Kiledjian M, Manley JL, Tong L. Structure and function of 724 the 5'-->3' exoribonuclease Rat1 and its activating partner Rai1. Nature. 2009;458(7239):784-8.

725 66. Dever TE, Feng L, Wek RC, Cigan AM, Donahue TF, Hinnebusch AG. Phosphorylation of 726 initiation factor 2 alpha by protein kinase GCN2 mediates gene-specific translational control of GCN4 727 in yeast. Cell. 1992;68(3):585-96.

728 67. Sattlegger E, Hinnebusch AG. Separate domains in GCN1 for binding protein kinase GCN2 729 and ribosomes are required for GCN2 activation in amino acid-starved cells. EMBO J. 730 2000;19(23):6622-33.

731 68. Pavitt GD, Yang W, Hinnebusch AG. Homologous segments in three subunits of the guanine 732 nucleotide exchange factor eIF2B mediate translational regulation by phosphorylation of eIF2. Mol 733 Cell Biol. 1997;17(3):1298-313.

734 69. Castilho BA, Shanmugam R, Silva RC, Ramesh R, Himme BM, Sattlegger E. Keeping the 735 eIF2 alpha kinase Gcn2 in check. Biochimica et biophysica acta. 2014;1843(9):1948-68.

736 70. Marton MJ, Vazquez de Aldana CR, Qiu H, Chakraburtty K, Hinnebusch AG. Evidence that 737 GCN1 and GCN20, translational regulators of GCN4, function on elongating ribosomes in activation of 738 eIF2alpha kinase GCN2. Mol Cell Biol. 1997;17(8):4474-89.

739 71. Sood R, Porter AC, Olsen DA, Cavener DR, Wek RC. A mammalian homologue of GCN2 740 protein kinase important for translational control by phosphorylation of eukaryotic initiation factor- 741 2alpha. Genetics. 2000;154(2):787-801.

742 72. Zhan K, Vattem KM, Bauer BN, Dever TE, Chen JJ, Wek RC. Phosphorylation of eukaryotic 743 initiation factor 2 by heme-regulated inhibitor kinase-related protein kinases in Schizosaccharomyces 744 pombe is important for fesistance to environmental stresses. Mol Cell Biol. 2002;22(20):7134-46.

745 73. Elsby R, Heiber JF, Reid P, Kimball SR, Pavitt GD, Barber GN. The alpha subunit of 746 eukaryotic initiation factor 2B (eIF2B) is required for eIF2-mediated translational suppression of 747 vesicular stomatitis virus. Journal of virology. 2011;85(19):9716-25.

748 74. Anda S, Zach R, Grallert B. Activation of Gcn2 in response to different stresses. PloS one. 749 2017;12(8):e0182143.

750 75. Hinnebusch AG. Evidence for translational regulation of the activator of general amino acid 751 control in yeast. Proc Natl Acad Sci U S A. 1984;81(20):6442-46.

752 76. Dong J, Qiu H, Garcia-Barrio M, Anderson J, Hinnebusch AG. Uncharged tRNA activates 753 GCN2 by displacing the protein kinase moiety from a bipartite tRNA-binding domain. Mol Cell. 754 2000;6(2):269-79.

De Zoysa 27

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 755 77. Natarajan K, Meyer MR, Jackson BM, Slade D, Roberts C, Hinnebusch AG, et al. 756 Transcriptional profiling shows that Gcn4p is a master regulator of gene expression during amino acid 757 starvation in yeast. Mol Cell Biol. 2001;21(13):4347-68.

758 78. Hinnebusch AG, Natarajan K. Gcn4p, a master regulator of gene expression, is controlled at 759 multiple levels by diverse signals of starvation and stress. Eukaryot Cell. 2002;1(1):22-32.

760 79. Udagawa T, Nemoto N, Wilkinson CR, Narashimhan J, Jiang L, Watt S, et al. Int6/eIF3e 761 promotes general translation and Atf1 abundance to modulate Sty1 MAPK-dependent stress 762 response in fission yeast. J Biol Chem. 2008;283(32):22063-75.

763 80. Hinnebusch AG. Translational regulation of GCN4 and the general amino acid control of yeast. 764 Annu Rev Microbiol. 2005;59:407-50.

765 81. Moehle CM, Hinnebusch AG. Association of RAP1 binding sites with stringent control of 766 ribosomal protein gene transcription in Saccharomyces cerevisiae. Mol Cell Biol. 1991;11(5):2723-35.

767 82. Han L, Guy MP, Kon Y, Phizicky EM. Lack of 2'-O-methylation in the tRNA anticodon loop of 768 two phylogenetically distant yeast species activates the general amino acid control pathway. PLoS 769 genetics. 2018;14(3):e1007288.

770 83. Dunand-Sauthier I, Walker CA, Narasimhan J, Pearce AK, Wek RC, Humphrey TC. Stress- 771 activated protein kinase pathway functions to support protein synthesis and translational adaptation in 772 response to environmental stress in fission yeast. Eukaryot Cell. 2005;4(11):1785-93.

773 84. Martin R, Berlanga JJ, de Haro C. New roles of the fission yeast eIF2alpha kinases Hri1 and 774 Gcn2 in response to nutritional stress. J Cell Sci. 2013;126(Pt 14):3010-20.

775 85. Ishimura R, Nagy G, Dotu I, Chuang JH, Ackerman SL. Activation of GCN2 kinase by 776 ribosome stalling links translation elongation with translation initiation. Elife. 2016;5.

777 86. Hussain S, Tuorto F, Menon S, Blanco S, Cox C, Flores JV, et al. The mouse cytosine-5 RNA 778 methyltransferase NSun2 is a component of the chromatoid body and required for testis 779 differentiation. Mol Cell Biol. 2013;33(8):1561-70.

780 87. Braun DA, Shril S, Sinha A, Schneider R, Tan W, Ashraf S, et al. Mutations in WDR4 as a new 781 cause of Galloway-Mowat syndrome. American journal of medical genetics Part A. 782 2018;176(11):2460-5.

783 88. Lamichhane TN, Arimbasseri AG, Rijal K, Iben JR, Wei FY, Tomizawa K, et al. Lack of tRNA- 784 i6A modification causes mitochondrial-like metabolic deficiency in S. pombe by limiting activity of 785 cytosolic tRNATyr, not mito-tRNA. RNA. 2016;22(4):583-96.

786 89. Johansson MJ, Esberg A, Huang B, Bjork GR, Bystrom AS. Eukaryotic wobble uridine 787 modifications promote a functionally redundant decoding system. Mol Cell Biol. 2008;28(10):3301-12.

788 90. Reuter JS, Mathews DH. RNAstructure: software for RNA secondary structure prediction and 789 analysis. BMC Bioinformatics. 2010;11:129.

790 91. Jinek M, Coyle SM, Doudna JA. Coupled 5' nucleotide recognition and processivity in Xrn1- 791 mediated mRNA decay. Mol Cell. 2011;41(5):600-8.

792 92. Robertus JD, Ladner JE, Finch JT, Rhodes D, Brown RS, Clark BF, et al. Structure of yeast 793 phenylalanine tRNA at 3 A resolution. Nature. 1974;250(467):546-51.

De Zoysa 28

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 794 93. Kim SH, Suddath FL, Quigley GJ, McPherson A, Sussman JL, Wang AH, et al. Three- 795 dimensional tertiary structure of yeast phenylalanine transfer RNA. Science. 1974;185(4149):435-40.

796 94. Westhof E, Dumas P, Moras D. Crystallographic refinement of yeast aspartic acid transfer 797 RNA. J Mol Biol. 1985;184(1):119-45.

798 95. Johnson AW. Rat1p and Xrn1p are functionally interchangeable exoribonucleases that are 799 restricted to and required in the nucleus and cytoplasm, respectively. Mol Cell Biol. 1997;17(10):6122- 800 30.

801 96. Shaheen HH, Hopper AK. Retrograde movement of tRNAs from the cytoplasm to the nucleus 802 in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 2005;102(32):11290-5.

803 97. Shaheen HH, Horetsky RL, Kimball SR, Murthi A, Jefferson LS, Hopper AK. Retrograde 804 nuclear accumulation of cytoplasmic tRNA in rat hepatoma cells in response to amino acid 805 deprivation. Proc Natl Acad Sci U S A. 2007;104(21):8845-50.

806 98. Takano A, Endo T, Yoshihisa T. tRNA Actively Shuttles Between the Nucleus and Cytosol in 807 Yeast. Science. 2005;309:140-2.

808 99. Kramer EB, Hopper AK. Retrograde transfer RNA nuclear import provides a new level of tRNA 809 quality control in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. 2013;110(52):21042-7.

810 100. Wan Y, Hopper AK. From powerhouse to processing plant: conserved roles of mitochondrial 811 outer membrane proteins in tRNA splicing. Genes Dev. 2018;32(19-20):1309-14.

812 101. Chalamcharla VR, Folco HD, Dhakshnamoorthy J, Grewal SI. Conserved factor 813 Dhp1/Rat1/Xrn2 triggers premature transcription termination and nucleates heterochromatin to 814 promote gene silencing. Proc Natl Acad Sci U S A. 2015;112(51):15548-55.

815 102. Zinshteyn B, Gilbert WV. Loss of a conserved tRNA anticodon modification perturbs cellular 816 signaling. PLoS genetics. 2013;9(8):e1003675.

817 103. Chou HJ, Donnard E, Gustafsson HT, Garber M, Rando OJ. Transcriptome-wide Analysis of 818 Roles for tRNA Modifications in Translational Regulation. Mol Cell. 2017;68(5):978-92 e4.

819 104. Pintard L, Lecointe F, Bujnicki JM, Bonnerot C, Grosjean H, Lapeyre B. Trm7p catalyses the 820 formation of two 2'-O-methylriboses in yeast tRNA anticodon loop. EMBO J. 2002;21(7):1811-20.

821 105. Han L, Kon Y, Phizicky EM. Functional importance of Psi38 and Psi39 in distinct tRNAs, 822 amplified for tRNAGln(UUG) by unexpected temperature sensitivity of the s2U modification in yeast. 823 RNA. 2015;21(2):188-201.

824 106. Xu F, Bystrom AS, Johansson MJO. SSD1 suppresses phenotypes induced by the lack of 825 Elongator-dependent tRNA modifications. PLoS genetics. 2019;15(8):e1008117.

826 107. Wek RC. Role of eIF2alpha Kinases in Translational Control and Adaptation to Cellular Stress. 827 Cold Spring Harb Perspect Biol. 2018;10(7).

828 108. Rashidi A, Miska J, Lee-Chang C, Kanojia D, Panek WK, Lopez-Rosas A, et al. GCN2 is 829 essential for CD8(+) T cell survival and function in murine models of malignant glioma. Cancer 830 Immunol Immunother. 2020;69(1):81-94.

831 109. Manaud G, Nossent EJ, Lambert M, Ghigna MR, Boet A, Vinhas MC, et al. Comparison of 832 Human and Experimental Pulmonary Veno-Occlusive Disease. Am J Respir Cell Mol Biol. 2020. De Zoysa 29

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 833 110. Duncan CDS, Rodriguez-Lopez M, Ruis P, Bahler J, Mata J. General amino acid control in 834 fission yeast is regulated by a nonconserved transcription factor, with functions analogous to 835 Gcn4/Atf4. Proc Natl Acad Sci U S A. 2018;115(8):E1829-E38.

836 111. Turowski TW, Karkusiewicz I, Kowal J, Boguta M. Maf1-mediated repression of RNA 837 polymerase III transcription inhibits tRNA degradation via RTD pathway. RNA. 2012;18(10):1823-32.

838 112. Yang R, Wek SA, Wek RC. Glucose limitation induces GCN4 translation by activation of Gcn2 839 protein kinase. Mol Cell Biol. 2000;20(8):2706-17.

840 113. Narasimhan J, Staschke KA, Wek RC. Dimerization is required for activation of eIF2 kinase 841 Gcn2 in response to diverse environmental stress conditions. J Biol Chem. 2004;279(22):22820-32.

842 114. Zhan K, Narasimhan J, Wek RC. Differential activation of eIF2 kinases in response to cellular 843 stresses in Schizosaccharomyces pombe. Genetics. 2004;168(4):1867-75.

844 115. Chen D, Toone WM, Mata J, Lyne R, Burns G, Kivinen K, et al. Global transcriptional 845 responses of fission yeast to environmental stress. Mol Biol Cell. 2003;14(1):214-29.

846 116. Cartlidge RA, Knebel A, Peggie M, Alexandrov A, Phizicky EM, Cohen P. The tRNA methylase 847 METTL1 is phosphorylated and inactivated by PKB and RSK in vitro and in cells. EMBO J. 848 2005;24(9):1696-705.

849 117. Huber SM, Leonardi A, Dedon PC, Begley TJ. The Versatile Roles of the tRNA 850 Epitranscriptome during Cellular Responses to Toxic Exposures and Environmental Stress. Toxics. 851 2019;7(1).

852 118. Gu C, Begley TJ, Dedon PC. tRNA modifications regulate translation during cellular stress. 853 FEBS Lett. 2014;588(23):4287-96.

854 119. Torres AG, Reina O, Stephan-Otto Attolini C, Ribas de Pouplana L. Differential expression of 855 human tRNA genes drives the abundance of tRNA-derived fragments. Proc Natl Acad Sci U S A. 856 2019;116(17):8451-6.

857 120. Dittmar KA, Goodenbour JM, Pan T. Tissue-specific differences in human transfer RNA 858 expression. PLoS genetics. 2006;2(12):e221.

859 121. Pavon-Eternod M, Gomes S, Geslain R, Dai Q, Rosner MR, Pan T. tRNA over-expression in 860 breast cancer and functional consequences. Nucleic Acids Res. 2009;37(21):7268-80.

861 122. Gingold H, Tehler D, Christoffersen NR, Nielsen MM, Asmar F, Kooistra SM, et al. A dual 862 program for translation regulation in cellular proliferation and differentiation. Cell. 2014;158(6):1281- 863 92.

864 123. Pavon-Eternod M, Gomes S, Rosner MR, Pan T. Overexpression of initiator methionine tRNA 865 leads to global reprogramming of tRNA expression and increased proliferation in human epithelial 866 cells. RNA. 2013;19(4):461-6.

867 124. Ishimura R, Nagy G, Dotu I, Zhou H, Yang XL, Schimmel P, et al. RNA function. Ribosome 868 stalling induced by mutation of a CNS-specific tRNA causes neurodegeneration. Science. 869 2014;345(6195):455-9.

870 125. Kim DU, Hayles J, Kim D, Wood V, Park HO, Won M, et al. Analysis of a genome-wide set of 871 gene deletions in the fission yeast Schizosaccharomyces pombe. Nature biotechnology. 872 2010;28(6):617-23. De Zoysa 30

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 873 126. Bahler J, Wu JQ, Longtine MS, Shah NG, McKenzie A, 3rd, Steever AB, et al. Heterologous 874 modules for efficient and versatile PCR-based gene targeting in Schizosaccharomyces pombe. Yeast. 875 1998;14(10):943-51.

876 127. Giaever G, Chu AM, Ni L, Connelly C, Riles L, Veronneau S, et al. Functional profiling of the 877 Saccharomyces cerevisiae genome. Nature. 2002;418(6896):387-91.

878 128. Sherman F. Getting started with yeast. Methods in enzymology. 1991;194:3-21.

879 129. Elder RT, Loh EY, Davis RW. RNA from the yeast transposable element Ty1 has both ends in 880 the direct repeats, a structure similar to retrovirus RNA. Proc Natl Acad Sci U S A. 1983;80(9):2432-6.

881 130. Preston MA, D'Silva S, Kon Y, Phizicky EM. tRNAHis 5-methylcytidine levels increase in 882 response to several growth arrest conditions in Saccharomyces cerevisiae. RNA. 2013;19(2):243-56.

883 131. Jackman JE, Montange RK, Malik HS, Phizicky EM. Identification of the yeast gene encoding 884 the tRNA m1G methyltransferase responsible for modification at position 9. RNA. 2003;9(5):574-85.

885 132. Guy MP, Podyma BM, Preston MA, Shaheen HH, Krivos KL, Limbach PA, et al. Yeast Trm7 886 interacts with distinct proteins for critical modifications of the tRNAPhe anticodon loop. RNA. 887 2012;18(10):1921-33.

888 133. Corpet F. Multiple sequence alignment with hierarchical clustering. Nucl Acids Res. 889 1988;16:10881-90.

890 134. Forsburg SL. Comparison of Schizosaccharomyces pombe expression systems. Nucleic Acids 891 Res. 1993;21(12):2955-6.

892

893

De Zoysa 31

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 894 Figure Legends

895 Fig. 1. S. pombe trm8Δ mutants lacked m7G and were temperature sensitive.

896 (A) trm8Δ mutants had no detectable m7G in their tY(GUA) and tF(GAA). S. pombe trm8Δ

897 mutants and WT cells were grown in biological triplicate in YES media at 30°C and tRNAs were

898 purified, digested to nucleosides, and analyzed for modifications by HPLC as described in Materials

899 and Methods. The bar chart depicts average moles/mol values of nucleosides with associated

900 standard deviation; WT, green; S. pombe (Sp) trm8Δ, brown. (B) trm8Δ mutants were temperature

901 sensitive due to lack of trm8+. Strains with plasmids as indicated were grown overnight in EMM-Leu

902 media at 30°C, diluted to OD600 ~ 0.5, serially diluted 10-fold in EMM-Leu, and 2 μL was spotted onto

903 plates containing EMM-Leu or YES media and incubated at 33°C, 37°C, and 38°C. The two

904 independent trm8Δ mutants were labeled as Sp trm8Δ V1 and V2.

905

906 Fig. 2. S. pombe trm8Δ mutants had reduced levels of tP(AGG) and tY(GUA) at elevated

907 temperatures.

908 (A) Northern analysis of Trm8 substrates tP(AGG), tY(GUA), and tT(AGU) in trm8Δ and WT

909 cells after shift from 30°C to 36.5°C, 37.5°C, and 38.5°C. Strains were grown in YES media at

910 30°C, shifted to the indicated temperatures for 8 hours as described in Materials and Methods, and

911 RNA was isolated and analyzed by northern blotting. n=2 for WT cells, n=3 for S. pombe trm8Δ

912 mutants. (B) Quantification of tP(AGG), tY(GUA), and tT(AGU) levels in WT and trm8Δ mutants

913 at different temperatures. The bar chart depicts relative levels of tRNA species at each temperature,

914 relative to their levels in the WT strain at 30°C (each value itself first normalized to levels of the control

915 non-Trm8 substrate tG(GCC)). For each tRNA, lighter shades indicate progressively higher

916 temperatures (30°C, 36.5°C, 37.5°C to 38.5°C) for tT(AGU), brown; tP(AGG), purple; tY(GUA), gray.

917 Standard deviations for each tRNA measurement are indicated. The statistical significance of tRNA

918 levels was evaluated using a two-tailed Student’s t-test assuming equal variance. ns, not significant (p

919 > 0.05); *, p < 0.05; **, p < 0.01; ***, p < 0.001. (C) Schematic of the secondary structure of

920 tY(GUA)-1 and tP(AGG). Modifications of tY(GUA) are as annotated. WC base pairs, black lines; GU

De Zoysa 32

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

7 921 base pairs, black dots; mismatch C-A or C-U base pairs, red diamonds; presumed m G46, red circle.

922 (D) Overproduction of tY(GUA), but not tP(AGG), suppressed the temperature sensitive growth

923 defect of trm8Δ mutants. Strains with plasmids as indicated were grown overnight in EMMC-Leu

924 media at 30°C and analyzed for growth as in Fig. 1B on the indicated plates.

925

926 Fig. 3. Mutations in dhp1 suppress the temperature sensitivity of S. pombe trm8Δ mutants and

927 restore tP(AGG) and tY(GUA) levels.

928 (A) dhp1 mutations restored growth of trm8Δ mutants at high temperature. Strains as indicated

929 were grown overnight in YES media at 30°C and analyzed for growth as in Fig. 1B. (B) Mutations in

930 dhp1 that restored growth of S. pombe trm8Δ mutants reside in evolutionarily conserved

931 residues. The amino acid sequence of Sp Dhp1 was aligned with putative Rat1/Dhp1 orthologs from

932 17 evolutionarily distant eukaryotes, using MultAlin (http://multalin.toulouse.inra.fr/multalin/) (133). red,

933 > 90% conservation; blue, 50%-90% conservation. Alleles of dhp1 mutations are indicated at the top.

934 (C) Each of two trm8Δ dhp1 mutants had restored tRNA levels in YES media at 38.5°C. Strains

935 were grown in YES media at 30°C and shifted to 38.5°C for 8 hours, and RNA was isolated and

936 analyzed by Northern blotting as in Fig. 2A. (D) Quantification of tRNA levels of trm8Δ dhp1

937 mutants shown in Figure 3C. tRNA levels were quantified as in Fig. 2B. tT(AGU), brown; tP(AGG),

938 purple; tY(GUA), gray; dark shades, 30°C; light shades, 38.5°C

939

940 Fig. 4. Mutations in the GAAC pathway suppress the temperature sensitivity of S. pombe

941 trm8Δ mutants and restore tP(AGG) and tY(GUA) levels.

942 (A) gcn1, gcn2, and tif221 mutations each restored growth of trm8Δ mutants at high

943 temperature. Strains as indicated were grown overnight in YES media at 30°C and analyzed for

944 growth as in Fig. 1B. (B) Expression of gcn2+ or tif221+ complemented the suppression

945 phenotype of trm8Δ gcn2-1 or trm8Δ tif221-2 mutants. S. pombe trm8Δ gcn2-1 and trm8Δ tif221-2

946 mutants expressing gcn2+ and tif221+ respectively, or a vector, were grown overnight in EMMC-Leu

947 media at 30°C, and analyzed for growth as in Fig. 1B. Note that expression of gcn2+ was kept to

De Zoysa 33

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

948 modest levels by adding thiamine to the media to partially suppress overexpression from the Pnmt1**

949 promoter. (C) gcn1, gcn2, and tif221 mutations each partially restored tY(GUA) and tP(AGG)

950 levels of trm8Δ mutants. Strains were grown in YES media at 30°C and shifted to 38.5°C for 8 hours

951 as described in Materials and Methods, and RNA was isolated and analyzed by northern blotting. (D)

952 Quantification of tRNA levels of trm8Δ GAAC mutants shown in Figure 4C. tRNA levels were

953 quantified as in Fig. 2B. tT(AGU), brown; tP(AGA), purple; tY(GUA), gray.

954

955 Fig. 5. S. pombe trm8Δ mutant temperature sensitivity is associated with induction of the

956 GAAC pathway due to Y(GUA) decay.

957 (A) trm8Δ mutants induced lys4+ mRNA expression at 38.5°C but not at 36.5°C or 37.5°C.

958 Strains as indicated were grown in YES media at 30°C and shifted to 36.5°C, 37.5°C, or 38.5°C for 8

959 hours (Fig. S14), and bulk RNA was isolated and analyzed by RT-qPCR as described in Materials and

960 Methods. The mRNA levels of lys4+ were normalized to levels of act1+, a non-regulated control

961 mRNA. WT, green; Sp trm8Δ, brown; Sp trm8Δ gcn2-1, light blue. Right side: GAAC induction of WT

962 cells grown at 30°C in EMMC-His and treated with 10 mM 3-AT for 4 hours, evaluated in parallel. (B)

963 A trm8Δ gcn2-1 mutant had restored levels of tY(GUA) and tP(AGG) at 38.5°C. Bulk RNA from

964 the growth done for Fig. 5A was used for the northern analysis. pre-tY(GUA) levels were analyzed

965 using appropriate gene-specific probes (Table S5) for defined tY(GUA) species (59). Cartoons at the

966 right indicate exons, heavy bars; 5' leaders, 3' trailers, and introns, light bars. (C) Quantification of

967 tY(GUA) and tP(AGG) levels in WT, trm8Δ and trm8Δ gcn2-1 mutants at different temperatures.

968 tRNA levels were quantified as described in Fig. 2. (D) tY(GUA) overproduction repressed the

969 GAAC induction of trm8Δ mutants at 38.5°C. Strains as indicated with plasmids expressing

970 tY(GUA) and/or tP(AGG) were grown in EMMC-Leu media at 30°C and shifted to 38.5°C for 8 hours,

971 and then RNA was isolated and lys4+ mRNA levels were analyzed by RT-qPCR as described in Fig.

972 5A. Right side: GAAC induction of WT cells grown at 30°C in EMMC-His media, and treated with 10

973 mM of 3-AT for 4 hours, evaluated in parallel to other samples.

974

De Zoysa 34

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 975 Fig. 6. Mutations in the GAAC pathway exacerbate the temperature sensitivity of S. cerevisiae

976 trm8Δ trm4Δ mutants as well as tV(ACC) decay.

977 (A) Deletion of GCN1, GCN2, or GCN4 exacerbated the temperature sensitivity of trm8Δ trm4Δ

978 mutants in YPD and SDC media. Strains were grown overnight in YPD media at 27°C and analyzed

979 for growth on YPD and SDC plates at different temperatures. (B) Deletion of GCN1 or GCN2

980 exacerbated tV(AAC) decay of trm8Δ trm4Δ mutants at 32°C. Strains were grown in YPD media

981 27°C, shifted to 32°C and harvested after 4 hours, and then bulk RNA was isolated and analyzed by

982 Northern blotting. (C) Quantification of tRNA tV(AAC), tR(UCU) and tW(CCA) levels of the

983 northern in Figure 6B. The bar chart depicts levels of tRNA species at 27°C or 32°C, relative to

984 levels of that tRNA in the WT strain at 27°C (each value itself first normalized to levels of the control

985 5S rRNA). tRNA levels are indicated by diagonal hatching for WT (green); S. cerevisiae (Sc) trm8Δ

986 trm4Δ (brown); Sc trm8Δ trm4Δ gcn2Δ (light blue); and Sc trm8Δ trm4Δ gcn1Δ (dark blue). (D) trm8Δ

987 trm4Δ mutants induced the GAAC pathway at 32°C. Bulk RNA from the growth done for Fig. 6B

988 was used for RT-qPCR analysis of levels of LYS1 and HIS5 mRNA, normalized to ACT1. mRNA

989 levels are indicated by horizontal lines for LYS1 and hatching for HIS5, WT (green); Sc trm8Δ trm4Δ

990 (brown). Right side: Relative levels of LYS1 mRNA of WT cells grown at 30°C in SD-His media, and

991 then treated with 10 mM 3-AT for 1 hour, evaluated in parallel to other samples. (E) tV(AAC)

992 overproduction repressed the GAAC induction of trm8Δ trm4Δ mutants at 36°C. Strains with

993 plasmids as indicated were grown in SD-Ura media 27°C and shifted to 1 hour at 36°C and RNA was

994 isolated and relative LYS1 mRNA levels were analyzed by RT-qPCR as described in Fig. 6D. Right

995 side: GAAC induction of WT cells grown and induced with 3-AT as described in Fig. 6D.

996

997 Fig. 7. A model illustrating the interplay of the RTD pathway and GAAC induction in S. pombe

998 and S. cerevisiae. Left: S. pombe trm8Δ mutants (red) trigger RTD of tY(GUA), leading to GAAC

999 induction and further decay of tY(GUA) and tP(AGG). The increased tRNA decay resulting from

1000 GAAC induction could be due to the global reduction in translation and/or the transcription

1001 upregulation of Fil1 target genes. Right: S. cerevisiae trm8Δ trm4Δ mutants (green) trigger RTD of

De Zoysa 35

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1002 tV(AAC), leading to GAAC induction and inhibition of decay of tV(AAC). The reduced tRNA decay due

1003 to GAAC induction could be due to the reduced global translation and/or the transcription upregulation

1004 of Gcn4 target genes.

De Zoysa 36

Fig. 1.

A tY(GUA) tF(GAA) 4.10 3.66 4.05 4.0bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083WT; this version posted May 29, 2020.4.0 The copyright holder for this preprint (which WT was3.29 not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made availableSp under atrm8CC-BY 4.0Δ International license. Sp trm8Δ 3.0 3.0

2.0 2.0 1.08 1.03 0.96 0.88 0.93 0.94 1.00 0.80 0.82

1.0 moles/mole 1.0 0.63 moles/mole

0.0 <0.03 0.0 <0.03 Ψ m5C m1A m7G Ψ Cm m2,2G m7G

B YES EMM-Leu LEU2 33 37 38 33 37 38 strain plasmid + WT 1 Ptrm8 trm8 WT 1 vector + Sp trm8Δ V1 Ptrm8 trm8 Sp trm8Δ V1 vector + Sp trm8Δ V2 Ptrm8 trm8 Sp trm8Δ V2 vector 2.0 Fig. 2. A CC A C G A C U A 1.6 WT 1 WT 2 Sp trm8Δ Sp trm8Δ Sp trm8Δ C G V2-1 V2-2 C G V1-1 U A 36.5° 38.5° 36.5° 38.5° 36.5° 38.5° 36.5° 38.5° 36.5° 38.5° G C Um1A 30° 37.5° 30° 37.5° 30° 37.5° 30° 37.5° 30° 37.5° A U U G A U C GG U C G *** D 1 ● tP(AGG) D Ψ m G ns bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (whichG U G GC U A G m5U C was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.G It is made m5C Ψ available under aCC-BY 4.0 International license. C A C 1.2 ** G U A U U D A tY(GUA) D C♦U Gm7G ns C G 0.80 C G tRNA 0.68 tY(GUA)-1 G C tT(AGU) 0.73 0.53G C 0.76 C A 0.71 6 0.50 0.80 U i A 0.8 tG(GCC) 0.60 0.56G Ψ A 2.02.0 0.56 0.60 2.0

2.51.61.6

Rel. 0.42

1.6 1.16 1.22 (AGG) tP 1.22 A (GUA) 1.16 tY 1.22 1.01 1.24 2.0 1.20C 1.19

B 1.16 2.5 1.00 1.01 0.41 1.24 1.20 1.19C 1.15 1.04

1.01 1.22 1.24 2.01.00 1.20 1.191.01 1.15C 1.15 1.04 1.00 1.00 1.00 1.04 gcn2 Sp - + + - + 1.2 1.2 1.00 1.00 1.01 + 0.82

1.00 1.002 1.01 0.84 1.6 G C 0.90 0.85 0.89

2.5 0.84 0.90 0.89 0.74 0.82 0.89 0.82

2.0 *** ns 0.84 0.74 0.710.85 0.90 0.85 0.68 0.65 trm8

1.6 G Sp 1.50.8 1.241.24 C 0.74 0.65 0.66 + 1.2 + Δ ** Δ 0.71 Δ

0.35 1.5 0.68 Δ 0.65

2.0 ns 0.98 0.71 0.68

0.8 tT(AGU) 0.80 1.19 0.66

0.68 tP(AGG) tY(GUA)0.98 C tRNA G 1.6 0.53 0.73 0.8 0.0 0.76 0.66 0.80 1.19 0.71 *** 0.0 0.4 1.160.56 1.22 0.60 1 0.50 ns 0.8 0.67 1.24 Rel. tRNA 0.60 1.20.70 0.56 ** C♦A 1.01 1.19 0.29Rel. 0.42 Rel. tRNA 1.201.6 30° 0.50

1.00 0.41 0.41 30° 301.150.29° 1.04ns

1.2 1.00 1.000.4 1.01 1.220.31 0.16 0.16 0.5 1.16 0.35 0.80 G C

tRNA 0.16 0.67 0.4

2 0.84 ** 1.010.900.70 1.24 0.89 1.200.680.820.29 1.19 0.73 0.53 0.18 36.5° 0.7436.5°0.4 0.8536.50.18° 0.76 1.15 0.18 1.04 0.29 0.4 1.00 0.18

0.71 1.2 0.50 0.71 0.65 A 0.80

0.8 ns 0.81.00 0.681.00 1.01 0.660.16 0.50U 0.60 0.56 U 0.4

0.0 0.00 37.5° 0.840.56 0.0 0.600.90 0.89 0.82 37.5° 37.5° 0.35 0.74 0.85A

0.5 Rel. 0.31 U A 0.42

Δ Δ + ns Δ Δ + 0.71 trm8 Sp 0.68U 0.41 0.65

Rel. Rel. tRNA 0.290.8 C A C C C 0.66

0.4 38.5° 0.41 U

38.5°0.8 0.1838.5° A

0.66 - + 0.16+ - + + 0.35G gcn2 Sp G

0.42 0.0

0.4 Rel.

0.68

0.71

0.65 U G

0.60 U

0.56 0.18

(AGG) tP Rel. tRNA (GUA) tY

0.74 0.29 0.85 1.24

0.84 0.90

0.56 C 0.8

0.0 0.4 0.60 G U G G G U 0.89 1.5 0

C U 0.50

0.82 0.16 G U

0.80

G ● 0.71

0.76 1.00

1.01 C 1.00

0.98 1.2 0.73

0.53 0.0

0.68

1.00

1.04 0.0 G tRNA

1.15

U G A U 0.80 U 1.20

1.19 *** + Δ Δ + Δ Δ

1.19 1.01

1.24 Sp trm8 U U A A ns

1.16

1.22 WT Sp trm8Δ V1 Sp trm8Δ V1 ** Sp trm8Δ V1 0.0 G G 1.2

*** Sp gcn2WT + Sp trm8Δ+ V1 - C G Sp trm8Δ+ V1 + Sp -trm8Δ V1 1.6 ns

WT 1 Sp trm8Δ* V1 Sp trm8Δ V1 Sp trm8ΔtYdhp1(GUA) -V11 C G dhp1*** -2 tP(AGG) Rel. Rel. tRNA dhp1-1 dhp1-2

0.70 0.67 dhp1-1 dhp1-2 A U

Δ Δ 1.6 0.50 Δ 2.0 Δ Sp trm8 + + WT Sp trm8Δt PV1(AGG) SpU trm8ΔA V1 Sp trm8Δ V1 Cdhp1G -1 dhp1-2

0.5 0.31 U U 0.18 U G 2.0 + + - + + -A G G Sp gcn2 0 tT(AGU) tP(AGG) tY(GUA) tT(AGU) tP(AGG) tY(GUA) tY(GUA) Sp trm8Δ tP(AGG)WT

D EMMC-Leu YES 33 37 38 33 37 38 strain LEU2 plasmid 2X tP(AGG) tY(GUA) tP(AGG) tY(GUA) WT 1 vector + Ptrm8 trm8 vector 2X tP(AGG) tY(GUA) tP(AGG) tY(GUA) Sp trm8Δ V1 vector + Ptrm8 trm8 vector Fig. 3. YES EMMC-His A 33 37 38 33 36 38

WT 1 WT 2 Sp trm8Δ V1 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which Spwas nottrm8 certifiedΔ byV2 peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made Sp trm8Δ dhp1-1 available under aCC-BY 4.0 International license. Sp trm8Δ dhp1-2 Sp trm8Δ dhp1-3 Sp trm8Δ dhp1-4

B C dhp1-3 dhp1-2 dhp1-1 dhp1-4 (R94L) (R218Q) (W326L) (C632F) Sp trm8Δ V1 Sp trm8Δ V1 WT 1 Sp trm8Δ V1 dhp1-2 S.pombe LLFAIIRPRK QRVKPEYD DDWVFFIFFV GCPSWTWYYPY dhp1-1 D.discoide RVLNMARPRK QRAQPDYD DDFIFLCFFV GCSSWGWYYPY 30° 38.5° 30° 38.5° 30° 38.5° 30° 38.5° S.cerevisiae RILAMVRPRQ QRADPEYN DDWVFMCFFC GCASWNWFYPY A.thaliana RLFVMVRPRK QRNLPGFD DDFIFICFFV GVCSWQWFYPY O.sativa RLFVMVRPRK QRNLPGFN DDFIFMCFFV GVCSWQWFYPY tY(GUA) O.tauri RLFLMIRPRK QRGLPGAN DDFIFMCFFV GCCSWGWFYPY D.melanogaster RLFGIVRPRK QRAQPDHD DDWVFMCFFV GCASWNWYFPY H.sapiens RLFSIVRPRR QRAQPNHD DDWVFMCFFV GCASWKWYYPF tP(AGG) M.musculus RLFNIVRPRR QRAQPNHD DDWVFMCFFV GCASWKWYYPF X.tropicalis RLFNIVRPRR QRAQPHHD DDWVFMCFFV GCASWKWFFPF P.infestans RLVAAVRPRR QRSQPGYD DDFVFLCFFV GCASWQWFYPF G.intestinalis RVVSICRPRR QRSQPGYD DDFVFLTFLV KVEDWGWYYPY tT(AGU) D.rerio RLFNIVRPRR QRGQPNHD DDWVFMCFFV C.merolae RIVALVRPRR QRAQPGYD DDFVFLCFFV RATSLRDWKQL N.gruberi QLFQLIKPSK RKMEAGYQ DDFVFFIFFT GVDADEDLEDE tG2.0(GCC2.0) C.elegans NLYNLIRPQK ERAGSSYD DDWILLGLLV RLTSEENARNS T.brucei K2.0LFNAIQPRK RKMQPDYG DDFVLMCFFI RVTPEERKNNR C.parameci YLFRIVKPKK QKVYFQYK DDFIFMCMLL - CPSWNWFYPE 1.61.6 1.6 1.161.16 1.221.22 2.0 1.16 1.22 1.01 1.241.24 1.20 1.19 1.15 1.01 1.001.241.01 1.20 1.19 1.20 1.19 1.15 1.04

1.00 1.2 2.0 1.00 1.00 1.00 1.01 1.15 1.04 1.04

1.6 1.2 1.00 1.2 1.011.00 1.00 1.01 0.82 D 1.00 0.0 0.84 0.90 0.89 1.16 1.22 0.89 0.74 0.820.85 0.82 1.01 1.24 1.20 1.19 0.84 1.15 0.840.90 0.74 0.710.85 0.90 0.68 0.89 0.65

1.2 1.00 2.4 0.8 1.6 1.04 0.74 0.85 0.65 0.66

1.00 1.00 1.01 1.16 1.220.71 0.68 0.65 0.16 0.8 tT(AGU) tP(AGG) 0.710.82 0.68 0.66

0.84 1.010.90 0.74 0.85tY(GUA1.24) 0.89 1.20 1.19 0.8 1.00 0.4 1.15 1.04 0.66 0.29 0.71 0.65 0.8 1.2 1.00 0.681.00 1.01 0.66 Rel. Rel. tRNA 30° 30° 30° 0.29 0.89 0.82 Rel. Rel. tRNA 0.4 0.84 0.90 0.74 0.85

0.4 0.29 0.71 0.68 0.65

Rel. Rel. tRNA 1.8 0.16

0.4 0.290.8 38.5° 38.50.16° 38.5° 0.66

0.8

0.66 0.16 0.4 0.29 0.68

0.71

0.65 1.01 1.22

1.16 Rel. tRNA 0.16

0.74 0.0 0.29 0.85 0.4 0.84 1.24 0.90 0.0 0.0

0.89 1.20 1.19 1.15

0.82 1.2 1.00 1.00 1.00 1.01 1.04 0.82 0.16 1.00 1.01

1.00

0.74 1.2 0.89

0.90 1.00

1.04 0.84 0.00.0 1.15 0.85

1.20 1.19

1.01

1.24 0.71 0.68 0.66 0.65

1.16 1.22 Rel. Rel. tRNA 0.6 1.6 WT Sp trm8Δ V10.16 0.29 Sp trm8Δ V1 Sp trm8Δ V1

WT Spdhp1 trm8Δ-1 V1 Spdhp1 trm8Δ-2 V1 Sp trm8Δ V1 0.0 WTWT 2.0 SpSp trm8Δ trm8Δ V1 V1Sp trm8ΔSp V1 trm8ΔSp V1 trm8Δ V1 Sp trm8Δ V1 dhp1-1 dhp1-1 dhp1dhp1--21 dhp1-2 dhp1-2

WT Sp trm8Δ V1 Sp trm8Δ V1 Sp trm8Δ V1 dhp1-1 dhp1-2 Fig. 4.

C A YES EMMC-His 33 38 39 33 37 38 38.5 Sp trm8 trm8+ trm8Δ WT 1 WT 2 GAAC + + gcn2-1 gcn1-1 tif221-2

bioRxiv preprintV1 doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made V2 available under aCC-BY 4.0 International license. dhp1-1 tY(GUA) gcn1-1 Sp trm8Δ gcn2-1 gcn2-2 tP(AGG) gcn2-3 tif221-1 tif221-2 tT(AGU)

tG(GCC)

2.0

B EMMC1.6-Leu+Thiamine D 2.0 1.16 1.22 0.0 1.01 1.24 1.20 1.19

LEU2 1.2 1.00 1.6 1.15 1.04

33 37 381.00 1.00 1.01 1.16 1.22 0.16 0.82

0.84 0.901.010.74 0.85 1.24 0.89 1.20 1.19 1.00 0.4 1.15 1.04 0.29 0.71 0.65 P 0.8 1.2 1.000.681.00 1.01 0.66 strain nmt1** 1.6 0.82 0.84 0.90 0.74 0.85 0.89

0.71 0.68 0.65

+ Rel. tRNA 0.4 0.290.8 0.66 0.8

0.66 gcn2 0.16 tT(AGU) tP(AGG) tY(GUA) 0.68 0.71

0.65 Rel. tRNA

0.74 0.29

0.85 0.4 0.84

WT 1 0.90 0.0 0.89

0.82 vector 0.16

1.00 1.01 1.00

1.141.2 1.15 1.00

1.04 0.0

1.15 1.2

1.20 1.10 1.19 1.01

gcn2+ 1.24

1.00 1.16

Sp trm8Δ V1 1.00 1.22 1.00 vector WT Sp trm8Δ V1 0.89 Sp trm8Δ V11.6 Sp trm8Δ V1

+ dhp1-1 dhp1-2 Sp trm8Δ V1 gcn2 0.8 WT2.0 Sp trm8Δ V1 Sp trm8Δ V1 Sp trm8Δ V1 dhp1-1 dhp1-2 gcn2-1 vector 0.40

Rel. tRNA Rel. 0.30 0.30 0.37 0.35 0.36 EMMC-Leu 0.4 0.11 LEU2 33 37 38 0.20 strain Pnmt1* 0.0 tif221+ WT 1 GAAC + + gcn2-1 gcn1-1 tif221-2 vector trm8+ trm8Δ tif221+ Sp trm8 Sp trm8Δ V1 vector Sp trm8Δ V1 tif221 + tif221-2 vector 12.0 8.6 10.0

8.0 mRNA 6.0

+ 5.3

6.0 5.6 5.5 lys1 4.0 6.0 5.6 4.2 5.5 4.2

mRNA 4.0 mRNA 4.0 Rel. 2.0Fig.0.74 5. 12.0 0.71 0.64 8.6 lys1+ lys1+ 2.0 WT 0.74 0.59 A 0.401.04 0.43 0.35 10.00.49 0.50Sp trm8Δ 0.63 lys1+ lys1+ 0.34 2.0 Rel. 0.52 0.49 0.50 0.60 0.53 0.57 0.49 1.04 Sp trm8Δ gcn2-1 0.0 0.00.63 8.0 Rel. 0.52 0.49 0.50 0.60 30 0.5336.50.57 0.4937.5 38.5 30 mRNA 6.0

0.0 + 5.3 WT + 3-AT YESlys1 4.0 WT 38.5 Sp trm8Δ 38.5 Sp trm8(EMMCΔ 30- HisWT) 30 Rel. 2.0 0.74 0.71 0.64 0.74 0.59 0.35 0.34 0.49 0.50 0.40 0.43 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.1230830.0 ; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made 30available under36.5 aCC-BY 4.0 37.5International license38.5. 30 WT 38.5 Sp trm8Δ 38.5 Sp trm8Δ 30 WT 30 WT + 3-AT YES (EMMC-His) 2.0 B 30 36.5 37.5 38.5 Sp trm8 + Δ Δ Δ Δ + Δ Δ + Δ Δ Sp gcn2 + + - + - + + - + + - 1.6 tY(GUA) tP(AGG) *** tG(GCC) 1.2 ns ns ** Pre-tY(GUA) 0.80 1-1, 1-2 tRNA 0.68 0.76 Pre-tY(GUA) 0.73 0.53 0.71 0.501-3 0.80 0.8 2.0 0.60 0.56

0.56 0.60 2.0

Pre-tY(GUA) 1.6

(AGG) Rel. tP (GUA) 1.6 1.22 0.42 tY 1.16

2 1.01 1.22 1.24 2.0 1.20 1.19

1.00 1.162.0 1.24 1.15 1.04 1.2 1.01 0.41 1.20 1.19 gcn2 Sp 1.00 1.01 1.15 - + + - + 1.00 1.00 1.04 +

1.2 1.00 1.00 1.01 1.6 0.89 0.82

0.84 0.90 0.74 0.85 0.82

2.0 *** 0.84 0.90 0.85 0.89 0.65

trm8 1.6 ns 0.71 0.74 0.68 Sp + + Δ 2.0 Δ Δ 0.8 ** 0.71 1.2 Δ 0.65 0.66

0.35 ns 0.68 0.8 0.80 0.66

1.60.53 0.73 2.0 0.68 tY(GUA) tP(AGG) tRNA 0.80 0.76 0.71 *** 0.0 0.56 0.60 1.16 1.22 0.50 ns 0.8

0.4 Rel. tRNA 0.60 0.56 1.24 1.2 0.42 1.01 1.20 1.19 ** Rel. 0.29 Rel. Rel. tRNA 30° 30° 1.15 1.2 1.00 0.41 0.4 0.4 1.6 ns 0.291.04

1.00 1.00 1.01 1.16 0.35 1.22 0.800.16

1.6 tRNA 0.16 0.680.4 0.82 0.18 0.841.01 36.51.24°0.90 0.740.7636.5° 0.85 1.20 1.190.89 0.731.15 0.53

0.18 0.18 C 1.00 1.04 0.71 0.71 0.65 0.80

0.8 1.2 0.81.00 1.00 1.01 0.68 0.50 0.66 0.60 0.56

0.0 0.0 37.5° 0.8437.50.56° 0.0 0.600.90 0.85 0.89 0.82 0.4 0.74 0.35 Rel. 0.42

Δ Δ ***+ Δ Δ + 0.71 trm8 Sp 0.68 0.41 0.65

Rel. Rel. tRNA 0.4 ns 0.8 0.29 0.66

- 1.2 + + - 0.16 + 38.5° 0.41 38.5+ 0.35° gcn2 Sp

** 0.4 0.42

Rel. Rel.

(AGG) tP ns Rel. tRNA (GUA) tY 0.18

0.60

0.0 0.4 0.29 0.56

0.56 0.8

0.80 0.60 0.16

tRNA

0.68 0.50

0.80

0.73 0.53 0.71

0.76 0.0 0.76

0.0 0.73

0.71 0.53 0.80

0.50 0.68 + Δ Δ + Δ Δ tRNA

0.8 0.56 Sp0.60trm8 0.60 0.56 0.80

0.0 WT Sp trm8Δ V1 Sp trm8Δ V1 Sp trm8Δns V1

Rel. WT Sp trm8Δ0.42 V1 Sp trm8Δ V1 Sp trm8Δ V1 + + - + ** + -

Sp gcn2 0.41 1.2 WT Sp trm8Δ V1 Sp trm8Δ V1 Sp trm8Δdhp1 V1-1 dhp1ns -2 0.35 tY(GUA) dhp1-1tP (AGG) *** dhp1-2

+ Δ Δ 0.4 + dhp1Δ-1 dhp1-2 Δ WT Sp0.18 trm8Δ V1 Sp trm8Δ V1 Sp trm8Δ V1 Sp trm8 1.6 dhp1-1 dhp1-2 6.0 0.0

5.6 5.5 + Δ Δ + Δ Δ Sp trm8 2.0 + + 6.0 - + + - Sp gcn2 4.2 Sp gcn25.6 + + - + + 5.5 -

tY(GUA) 4.2 tP(AGG) mRNA mRNA 4.0 tY(GUA)4.0 tP(AGG) lys1+ lys1+ 2.0 WT 6.0 1.04 5.6

Sp trm8Δ 0.63 5.5 lys1+ lys1+ 2.0 Rel. 0.52 0.49 D0.50 0.60 0.53 0.574.2 0.49 1.04 0.00.63

mRNA 4.0 Rel. 0.52 0.49 0.50 0.60 0.53 0.57 + 0.49

0.0 lys1 2.0 1.04 0.63 Rel. 0.52 0.60 0.53 0.57 WT 38.5 Sp trm8Δ 38.50.49 0.50Sp trm8Δ 30 WT 30 0.49 0.0 - [tY(GUA)] - + + - - + + - + - 3-AT [tP(AGG)] +,+ - + - +,+ - + - - - Sp trm8 trm8+, 38.5 trm8Δ, 38.5 trm8Δ, 30 trm8+, 30 WT 38.5 Sp trm8Δ 38.5 Sp trm8Δ 30 WT 30 Fig. 6.

YPD SDC 28 30 32 28 30 31284 A 300 250 gcn1Δ

gcn2Δ Sc trm8Δ gcn4Δ bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083trm4Δ ; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available200 under aCC-BY 4.0 International license.

50 gcn1Δ gcn2Δ WT 284 300 gcn4Δ 250 100

B 200 D 22.4 22.2 50 22.2 Sc trm8Δ Sc trm8Δ 22.4 Sc trm8Δ WT Sc trm8Δ 284trm4Δ 284 WT Δ trm40.88Δ 1.50trm4Δ1.03 1.05 0.811.23 trm4 22.4 300 LYS1 LYS1 250 0300gcn2Δ 22.2gcn1Δ 250 100 HIS5 HIS5 0.881.50 1.031.05 0.81 1.23 50 0 27° 32° 27° 32° 27° 32° 27° 32° 200 tV(AAC) 22.222.4 0.88 1.50 0.881.031.501.05 0.811.031.231.05 0.81 1.23 2.0 tW(CCA) 0 2.0 0 200 22.4 WT Sc trm8 Rel. mRNA Δ trm4Δ WT Sc trm8Δ trm4100 Δ 22.2 Sc tRtrm8(UCU) Δ trm4Δ gcn2Δ Sc trm8Δ trm4Δ gcn1Δ Sc trm8Δ trm4Δ gcn2Δ Sc trm8Δ trm4Δ gcn1Δ 22.2 1.5 5S 284 22.4 1.5 0.88 1.50 1.03 1.05 0.811.23 300 0 250 1.00 1.00 1.121.12 0.881.50 1.031.05 0.81 1.23 100 0 0.77 2.0 27 32 27 32 3-AT 30

0.77 2.0 WT Sc trm8Δ trm4Δ tRNA tRNA 1.0 1.0 WTSc trm8Δ trm4Δ gcn2Δ Sc trm8Sc trm8Δ trm4Δ Δtrm4 gcn1ΔΔ WT, YPD Sc trm8Δ trm4Δ, YPD WT, SD 0.64 1.5 Sc trm8Δ trm4Δ gcn2Δ Sc trm8Δ trm4Δ gcn1Δ 0.64 C1.5 1.00 2001.12 E 0.77 1.12 tRNA 1.00 0.65 1.0 0.64 0.77 0.65 22.2 0.65 tRNA 1.0 0.33 0.33 150 22.4

2.0 Rel. Rel. 0.64 131 0.330.5 0.11 WT Sc trm8Δ trm4Δ

WT0.65 0.33 Sc trm8Δ trm4Δ gcn2Δ Rel. 0.5 Rel. 0.08 0.88 1.50 1.03 1.05 1250.811.23LYS1 LYS1 0.50.0 0.11Sc trm8Δ trm40.11Δ Sc trm8Δ trm4Δ gcn1Δ 0.5 1.5 27 32 270 32 27 32 0.111.12 100 0.08 100 0.01.00 tV(AAC) tR(UCU) tW(CCA) 0.77 270.08 32 27 32 27 32

tRNA 75 1.0 0.64 0.08tV(AAC) tR(UCU) 25 tW(CCA) 0.0 0.65 0.33 50 22.2mRNA Rel. 22.4 0.5 0.11 18.0 27 32 0.88271.50 1.03 1.0532 0.811.23 27 25 32 18.0 0.0 0.08 0 LYS1 1.4 2.5 0.65 0.99 0.83 0.91 5.3 0.0 0 27 3227 32 27 27 32 27 3232 27 32 tV(AAC) tR(UCU) Rel. tW(CCA) tV(AAC) tR(UCU) tW(CCA) 5.3 2.5 tV(AAC) tR(UCU) 1.4 0.65tW0.99(CCA)0.83 0.91 0 28 37 30 Vec tV(AAC) Vec tV(AAC) Vec tV(AAC) Vec tV(AAC) 3-AT Sc trm8Δ WT WT Sc trm8Δ WT trm4Δ trm4Δ Fig. 7. Sp trm8Δ Sc trm8Δ trm4Δ 38.5° 32°

Dhp1 CCA 3' Rat1/Xrn1 CCA 3'

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

tY(GUA) decay tV(AAC) decay

Gcn2

eIF2α-P Fil1/Gcn4 translation Transcription Global translation

Decay of tY(GUA) tV(AAC) and tP(AGG) decay bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1005 Supplementary Figure Legends

1006

1007 Fig. S1. S. pombe trm8Δ mutants showed a temperature sensitive growth defect in liquid YES

1008 media. Strains were grown in YES media at 30°C, shifted to the indicated temperatures, and then

1009 growth was monitored for 8 hours before harvest as described in Materials and Methods, and tRNA

1010 analysis as done in Fig. 2A, 2B. WT, green; Sp trm8Δ, brown.

1011

1012 Fig. S2. S. pombe trm8Δ mutants had reduced 5S rRNA and 5.8S rRNA levels at higher

1013 temperatures. The northern blot shown in Fig. 2A was used to analyze the non-Trm8 substrate

1014 tL(UAA), 5S rRNA, and 5.8S rRNA. The bar chart depicts levels of RNA species at each temperature,

1015 relative to their levels in the WT strain at 30°C (each value itself first normalized to levels of the control

1016 non-Trm8 substrate tG(GCC)). 30°C, green; 36.5°C, yellow; 37.5°C, orange; 38.5°C. red.

1017

1018 Fig. S3. tP(AGG) of S. pombe trm8Δ mutants had no detectable m7G. trm8Δ mutants and WT

1019 cells were grown in YES media at 30°C and tP(AGG) was purified and analyzed for modifications as

1020 in Fig. 1A.

1021

1022 Fig. S4. Northern analysis of all Trm8 substrates (except tP(AGG), tY(GUA), and tT(AGU)) in S.

1023 pombe trm8Δ and WT cells after shift from 30°C to 36.5°C, 37.5°C, and 38.5°C. The northern blot

1024 shown in Fig. 2A was continued to analyze levels of all other predicted Trm8 substrate tRNAs, as well

1025 as the non-Trm8 substrate tL(UAA), 5S RNA, and 5.8S RNA, in WT and trm8Δ mutants at different

1026 temperatures.

1027

1028 Fig. S5. Among 21 predicted Trm8 substrate tRNAs, only tP(AGG) and tY(GUA) had reduced

1029 levels in S. pombe trm8Δ mutants at elevated temperatures. tRNA levels were quantified, relative

1030 to tG(GCC), in trm8Δ mutants and WT cells at different temperatures, for all predicted Trm8

De Zoysa 37

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1031 substrates, as described in Fig. 2B. Note that data from Fig. 2B is also included here for

1032 completeness.

1033

1034 Fig. S6. A. Overproduction of tY(GUA) and tP(AGG) resulted in increased levels of the

1035 corresponding tRNAs in S. pombe trm8Δ mutants and WT cells. Strains with plasmids as

1036 indicated were grown in EMMC-Leu media at 30°C and shifted to 38.5°C for 8 hours, and then RNA

1037 was isolated and analyzed by Northern blotting as in Fig. 2A. B. Quantification of tRNA levels in S.

1038 pombe trm8Δ mutants and WT cells overproducing tY(GUA) or tP(AGG). Quantification was done

1039 as in Fig. 2B.

1040

1041 Fig. S7. Overproduction of both tY(GUA) and tP(AGG) fully restored growth of S. pombe trm8Δ

1042 mutants in YES + glycerol media, but not in YES media containing 5-FU. Strains grown for Fig.

1043 2D were analyzed for growth on plates containing YES media with 3% glycerol (instead of 3%

1044 glucose) and YES media with 5-FU (30 µg/ml).

1045

1046 Fig. S8. dhp1 mutations restored growth of S. pombe trm8Δ mutants in YES + 5-FU media.

1047 Strains grown for Fig. 3A were analyzed for growth on YES + 5-FU (30 µg/ml) plates.

1048

+ 1049 Fig. S9. Expression of Pdhp1 dhp1 restored temperature sensitive growth in the S. pombe trm8

+ 1050 dhp1-1 mutant. WT, trm8Δ, and trm8Δ dhp1-1 cells expressing Pdhp1 dhp1 or a vector were grown

1051 overnight in EMMC-Leu media at 30°C, and analyzed for growth.

1052

1053 Fig. S10. A. S. pombe trm8Δ dhp1-3 and trm8Δ dhp1-4 mutants also restored tY(GUA) and

1054 tP(AGG) tRNA levels at 38.5 °C. Strains were grown in YES media at 30°C and shifted to 38.5°C for

1055 8 hours, and RNA was isolated and analyzed by Northern blotting as in Fig. 2A. B. Quantification of

1056 tRNA levels in different S. pombe trm8Δ dhp1 mutants. tRNA levels were quantified as in Fig. 2B.

1057 tT(AGU), brown; tP(AGG), purple; tY(GUA), gray.

De Zoysa 38

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1058

1059 Fig. S11. Mutations in the GAAC pathway did not rescue the 5-FU sensitivity of S. pombe

1060 trm8Δ mutants, and conferred enhanced 3-AT sensitivity. Strains grown for Fig. 4A were analyzed

1061 for growth on plates containing YES media + 5-FU (30 µg/ml) and EMMC-His media + 10 mM 3-AT.

1062

1063 Fig. S12. Reconstructed S. pombe trm8Δ gcn2Δ mutants had the same growth properties as

1064 the original trm8Δ gcn2-1 strain. Strains were analyzed for growth on YES media, EMMC-His or

1065 EMMC-His media containing 10 mM 3-AT, as described in Fig. 1B. Reconstructed S. pombe trm8Δ

1066 mutant was labeled as V3.

1067

1068 Fig. S13. A. Northern analysis of WT, Sp trm8Δ, Sp trm8Δ gcn2-2, trm8Δ tif221-1, and trm8Δ

1069 gcn2-3 cells. Strains were grown in YES media at 30°C and shifted to 38.5°C for 8 hours, and RNA

1070 was isolated and analyzed by Northern blotting as in Fig. 2A. B. Quantification of tRNA levels. tRNA

1071 levels were quantified as in Fig. 2B. n=2 for all strains.

1072

1073 Fig. S14. The temperature sensitivity of S. pombe trm8Δ mutants observed at 38.5°C in YES

1074 liquid media was suppressed in a trm8Δ gcn2-1 mutant. Strains were grown in YES media at

1075 30°C, shifted to the indicated temperatures, and then growth was monitored for 8 hours before

1076 harvest as described in Materials and Methods, and analysis of mRNAs and tRNAs in Fig. 5A, 5B, 5C,

1077 and S15.

1078

1079 Fig. S15. S. pombe trm8Δ mutants induced aro8+ mRNA expression at 38.5°C but not at 37.5°C.

1080 Bulk RNA from the growth in Fig. S14 was used for the RT-qPCR analysis of aro8+(SPAC56E4.03)

1081 mRNA levels, as in Fig. 5A.

1082

1083 Fig. S16. A. Analysis of charging levels of tY(GUA) or tP(AGG) in S. pombe trm8Δ mutants at

1084 38.5°C. Strains were grown in YES media at 30°C and shifted to 38.5°C, and samples were harvested

De Zoysa 39

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1085 after 8 hours. Then bulk RNA was isolated and resolved by denaturing PAGE under acidic conditions

1086 (to preserve tRNA charging), transferred, and then analyzed by hybridization as described in Materials

1087 and Methods. Control samples (WT and S. pombe trm8Δ mutants) were treated with 1 mM EDTA and

1088 0.1 M Tris-HCl (pH 9.0) for 30 min at 37°C to de-acylate the tRNA. b, base treated bulk RNA; Upper

1089 arrows, charged tRNA species; lower arrows with dashed lines, uncharged tRNA species. B.

1090 Quantification of tY(GUA) or tP(AGG) charging and tRNA levels. The percent charging was

1091 calculated as the ratio of aminoacylated species to the total for each tRNA. Relative levels of tP(AGG)

1092 and tY(GUA) were quantified as in Fig. 2B, relative to the non-Trm8 substrate tL(UAA).

1093

1094 Fig. S17. A. Deletion of GCN2 exacerbated the temperature sensitivity of S. cerevisiae trm8Δ

1095 and trm1Δ mutants in SDC media. Strains were grown overnight in YPD media at 28°C and

1096 analyzed for growth on SDC plates. B. Deletion of GCN2 exacerbated the temperature sensitivity

1097 of S. cerevisiae tan1Δ and tan1Δ trm44Δ mutants in SDC media. Strains were grown overnight in

1098 YPD media 28°C and analyzed for growth on SDC plates.

1099

1100 Fig. S18. Analysis of pre-tV(AAC) levels in WT, S. cerevisiae trm8Δ trm4Δ, and trm8Δ trm4Δ

1101 gcn1Δ cells after shift from 28°C to 32°C. Bulk RNA from the growth done for Fig. 6B, 6C was used

1102 for the northern analysis.

1103

1104 Fig. S19. Deletion of MET22 restored growth of S. cerevisiae trm8Δ trm4Δ gcn2Δ mutants at

1105 elevated temperatures. Strains were grown overnight in YPD media 28°C and analyzed for growth

1106 on YPD plates.

1107

1108 Figure S20. A. Analysis of tRNA charging levels in S. cerevisiae trm8Δ trm4Δ mutants after

1109 shift to 32°C. Cell pellets from the growth for Fig. 6B were used to isolate acidic RNA and analyzed

1110 by acidic northern as described in Fig. S16. B. Quantification of tRNA charging and tRNA levels.

De Zoysa 40

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1111 The percent aminoacylation of tV(AAC) and tK(UUU) was calculated as described in Fig. S16, and

1112 relative tRNA levels were quantified as in Fig. 6C.

De Zoysa 41

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S1

WT Sp trm8Δ 36.5° 37.5° 38.5°

1.00 1.00 1.00 ) ) ) 600 600 600 (OD (OD (OD 10 10 10 log log log

0.10 0.10 0.10 0 5 10 (h) 0 5 10 (h) 0 5 10 (h) bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S2

Sp trm8Δ WT 2.0 2.0 30°C 36.5°C 37.5°C 38.5°C 30°C 36.5°C 37.5°C 38.5°C 1.18 1.5 1.06 1.5 0.92 1.00 0.85 1.05 0.81 0.89 1.03 0.82 1.00 1.01 0.77 0.96 0.67 0.68 1.05 1.00 0.78 1.0 1.0 0.86 0.70 0.63 0.56 0.50 Rel. RNA Rel. RNA 0.5 0.5

0.0 0.0 tL(UAA) 5.8S rRNA 5S rRNA tL(UAA) 5.8S rRNA 5S rRNA Fig. bioRxiv preprint was notcertifiedbypeerreview)istheauthor/funder,whohasgrantedbioRxivalicensetodisplaypreprintinperpetuity.Itmade S3 moles/mole doi: 0.0 2.0 4.0 6.0 https://doi.org/10.1101/2020.05.29.123083 2.54 tP Ψ (AGG) 2.35 available undera 1.87 m 2 1.94 ; G CC-BY 4.0Internationallicense this versionpostedMay29,2020. 1.01 Sp trm8∆ WT I 1.18 . m 0.87 The copyrightholderforthispreprint(which 7 G < 0.03 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S4 WT 1 WT 2 Sp trm8Δ Sp trm8Δ Sp trm8Δ V1 V2 V2 36.5° 38.5° 36.5° 38.5° 36.5° 38.5° 36.5° 38.5° 36.5° 38.5° 30° 37.5° 30° 37.5° 30° 37.5° 30° 37.5° 30° 37.5°

tT(CGU)

tK(UUU)

tMe(CAU) tN(GUU) tMi(CAU)

tV(CAC)

tV(UAC) tK(CUU) tI(AAU) tW(CCA) tRCCU)

tA(CGC) tV(AAC) tF(GAA) tP(UGG) tA(AGC) tA(UGC)

tV(AAC) tL(UAA) 5.8S rRNA

5S rRNA bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S5 A. Sp trm8Δ

30° 36.5° 37.5° 38.5° 2 tRNA 1 Rel. 0

B. WT

30° 36.5° 37.5° 38.5° 2

tRNA 1

Rel. 0 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S6 A. WT, 30° Sp trm8Δ, 30° Sp trm8Δ, 38.5° [tP(AGG)] +,+ - + - +,+ - + - +,+ - + - [tY(GUA)] - + + - - + + - - + + - tY(GUA)

tP(AGG)

tT(AGU)

tG(GCC) B. tY(GUA) tP(AGG) tT(AGU) 6.0 6.0 6.0 Vec Vec Vec tY(GUA) 3.82 tY(GUA) tY(GUA) 3.54 3.40 tY(GUA), tP(AGG) tY(GUA), tP(AGG) tY(GUA), tP(AGG) 4.0 2X tP(AGG) 4.0 4.0 2X tP(AGG) 2.55 2X tP(AGG) 2.80 2.24 2.21 1.36 1.49 1.54 Rel. tRNA 2.0 1.51 Rel. tRNA 2.0 0.96 1.00 Rel. tRNA 2.0 0.94 0.98 1.06 1.00 0.73 1.03 0.95 0.47 1.001.00 0.99 0.94 0.69 0.58 0.99 0.59 0.20 0.43 0.81 0.81 0.22 0.26 0.09 0.0 0.0 0.0 WT Sp trm8Δ Sp trm8Δ WT Sp trm8Δ Sp trm8Δ WT Sp trm8Δ Sp trm8Δ 30° 30° 38.5° 30° 30° 38.5° 30° 30° 38.5° bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S7 YES + Glycerol YES + 5-FU

30° 33° 34° 33° 36° 37° strain LEU2 plasmid 2X tP(AGG) tY(GUA) WT tP(AGG) tY(GUA) vector

+ Ptrm8 trm8 vector 2X tP(AGG) tY(GUA) tP(AGG) tY(GUA) Sp trm8Δ V1 vector + Ptrm8 trm8 vector bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S8

YES + 5-FU 30° 34° 36° 38°

WT-1 WT-2 Sp trm8Δ V1 Sp trm8Δ V2 Sp trm8Δ dhp1-1 Sp trm8Δ dhp1-2 Sp trm8Δ dhp1-3 Sp trm8Δ dhp1-4 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S9 EMMC-Leu 33° 37° 38° 39°C strain LEU2 plasmid

+ Pdhp1 dhp1 WT vector

+ Pdhp1 dhp1 Sp trm8Δ vector

+ Pdhp1 dhp1 Sp trm8Δ dhp1-1 vector bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S10 A. 38.5° Sp trm8Δ Sp trm8Δ Sp trm8Δ WT Sp trm8Δ dhp1-1 dhp1-3 dhp1-4 tY(GUA)

tP(AGG)

tT(AGU)

tL(UAA)

tG(GCC)

tT(AGU) tP(AGG) tY(GUA) B. 1.60

1.00 1.20 1.00 1.00 0.87 0.94 0.73 0.87 0.67 0.74 0.70 0.80 0.75 0.73 0.56 Rel. tRNA 0.40 0.11 0.19

0.00 WT Sp trm8Δ Sp trm8Δ Sp trm8Δ Sp trm8Δ dhp1-1 dhp1-3 dhp1-4 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S11 YES + 5FU (EMMC-His) + 3-AT 33° 36° 37° 33° 37° 38°

WT-1 WT-2 V1 V2 dhp1 gcn1-1

Sp trm8Δ gcn2-1 gcn2-2 gcn2-3 tif221-1 tif221-2 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S12

YES EMMC-His (EMMC-His) + 3-AT 33° 37° 38° 33° 37° 38° 33° 37°

WT-1 WT-2 Sp trm8Δ V3 Sp trm8Δ V3 gcn2Δ-1 Sp trm8Δ V3 gcn2Δ-2 Sp trm8Δ V3 gcn2Δ-3 Sp trm8Δ gcn2-1 Sp gcn2Δ bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S13

A. 38.5° Sp trm8 trm8+ trm8Δ GAAC + + gcn2-2 tif221-1 gcn2-3 tY(GUA)

tP(AGG)

tV(AAC)

tG(GCC)

B. 1.6 tV(AAC) tP(AGG) tY(GUA) 1.2 1.00 1.00 1.00 0.75 0.71 0.81 0.70 0.8 0.31 0.29 tRNA 0.31 0.57 0.58 0.53 0.09

Rel. 0.4 0.28

0.0 GAAC + + gcn2-2 tif221-1 gcn2-3 Sp trm8 trm8+ trm8Δ bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S14.

WT Sp trm8Δ Sp trm8Δ gcn2-1 36.5° 37.5° 38.5° 1 1 1 ) ) ) 600 600 600 (OD (OD (OD 10 10 10 log log log

0.1 0.1 0.1 0 5 10 (h) 0 5 10 (h) 0 5 10 (h) bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S15

8.0 WT Sp trm8Δ 6.3 Sp trm8Δ gcn2-1 mRNA mRNA

4.0 aro8+

1.5 Rel. 0.440.45 0.23 0.35 0.22 0.0 37.5° 38.5° 30° WT + 3-AT (EMMC-his) bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S16

A. WT Sp trm8Δ V1 b 30° 38.5° b 30° 38.5°

tY(GUA)

tP(AGG)

tL(UAA)

tW(CCA)

B. 120.0 WT Sp trm8Δ 1.6 WT Sp trm8Δ 1.39 * *** 72.2 80.8 80.3 1.06 1.20 80.0 77.8 1.2 1.00 69.8 59.0 1.00 0.85 50.2 56.0 tRNA 0.8 0.70 Charging

40.0 Rel.

% 0.4 0.12 0.0 0.0 30° 38.5° 30° 38.5° 30° 38.5° 30° 38.5° tY(GUA) tP(AGG) tY(GUA) tP(AGG) bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S17.

A. B. SDC SDC 30° 37° 39.5° 28° 34° 37°

gcn2Δ gcn2Δ Sc trm8Δ Sc tan1Δ

met22Δ WT gcn2Δ WT gcn2Δ gcn2Δ Sc tan1Δ trm44Δ Sc trm1Δ

met22Δ WT gcn2Δ WT gcn2Δ bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S18

Sc trm8Δ WT Sc trm8Δ trm4Δ trm4Δ gcn1Δ 27° 32° 27° 32° 27° 32°

Pre-tV(AAC)

tV(AAC)

tF(GAA)

5S bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S19 YPD 28° 30° 32° 37°

gcn2Δ met22Δ

gcn2Δ Sc trm8Δ trm4Δ

met22Δ

gcn2Δ WT bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license.

Fig. S20

A. Sc trm8Δ WT trm4Δ b 27° 32° b 27° 32°

tV(AAC)

tK(UUU)

tL(CAA)

5S B. 125 WT Sc trm8Δ trm4Δ 1.6 WT Sc trm8Δ trm4Δ 1.22 100 1.34 87** 88*** 77 1.00 1.03 80 75 73 75 1.2 0.92 75 71 1.00 tRNA 0.8 0.58 50 0.29 25 Rel. 0.4 % Charging 0 0.0 27° 32° 27° 32° 27° 32° 27° 32° tV(AAC) tK(UUU) tV(AAC) tK(UUU) bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1113 Supplementary Tables

1114 Table S1. Variable loop sequences and the folding free energies of the acceptor stem/Tstem

1115 loop of predicted Trm8 substrate tRNAs of S. pombe trm8Δ mutants

1116

o Variable loop ΔG 37 (Kcal/mole) tRNA sequence Acceptor/T stem loop tC(GCA-1) AGGUC -16.0 tF(GAA-1) UGGUC -16.8 tT(CGU) AGGUC -16.8 tT(AGU) AGGCC -16.9 tK(UUU) AGGUU -17.0 tP (AGG) UGGUC -17.7 tA(AGC) AGGUC -17.7 tR(CCU) AGGUU -18.2 tC(GCA-2) AGGUC -18.3 tF(GAA-2) UGGUC -18.5 tV(AAC) CGGUC -19.9 tMe (CAU) AGGUU -20.2 tY(GUA) UGGUC -20.5 tP(UGG-1) UGGUC -20.6 tW(CCA) AAGUC -20.7 tV(UAC) CGGUC -20.9 tA(UGC) UCGUC -22.0 tK(CUU) AGGUU -22.2 tP(UGG-1) UGGUC -23.7 tI(AAU) AUGUC -24.0 tA(CGC) AGGUC -24.2 tN(GUU) AGGUC -25.2 tMi(CAU) AGGUU -25.5 tV(CAC) AGGUC -25.5 1117 1118 1119

De Zoysa 42

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1120 Table S2. GAAC mutations identified in S. pombe trm8Δ suppressors 1121 Strain Mutation Sp trm8 gcn1-1 inframe deletion in V2193-A2195 Sp trm8 gcn2-1 start lost, G3A Sp trm8 gcn2-2 Frameshift at R71 Sp trm8 gcn2-3 Stop at W982 Sp trm8 tif221-1 Stop at Q257 Sp trm8 tif221-2 Frameshift at L43 1122 1123

De Zoysa 43

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1124 Table S3. S. cerevisiae Strains used in this study 1125 Strain name Parent strain Genotype Source YTD 386 AA 527 trm8Δ::natMX trm4Δ::kanMX (18) YTD 558 YTD 386 trm8Δ::natMX trm4Δ::BleMX This study YTD 559 YTD 558 trm8Δ::natMX trm4Δ::BleMX gcn2Δ::KanMX This study YTD 561 YTD 566 gcn2Δ::KanMX This study YTD 562 AA 413 trm8Δ::NatMX This study YTD 563 YLN 5 trm1Δ::BleMX This study YTD 564 ISC 188 tan1Δ::KanMX This study YTD 565 ISC 834 tan1Δ::KanMX trm44Δ::natMX (21) YTD 566 ISC 534 WT MATa This study YTD 567 YTD 558 trm8Δ::natMX trm4Δ::BleMX gcn1Δ::KanMX This study YTD 568 YTD 558 trm8Δ::natMX trm4Δ::BleMX gcn4Δ::KanMX This study YTD 569 YTD 562 trm8Δ::NatMX gcn2Δ::KanMX This study YTD 570 YTD 563 trm1Δ::NatMX gcn2Δ::KanMX This study YTD 571 YTD 564 tan1Δ::BleMX This study YTD 572 YTD 565 tan1Δ::BleMX trm44Δ::natMX This study YTD 573 YTD 566 gcn1Δ::KanMX This study YTD 574 YTD 566 gcn4Δ::HygMX This study YTD 575 YTD 559 trm8Δ trm4Δ gcn2Δ met22Δ::HygMX This study YTD 576 YTD 571 tan1Δ::BleMX gcn2Δ::KanMX This study YTD 577 YTD 572 tan1Δ::BleMX trm44Δ::natMX gcn2Δ::KanMX This study 1126 1127

De Zoysa 44

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1128 1129 1130 Table S4. Plasmids used in this study 1131 Plasmid Parent Description Source

pREP3X LEU2 Pnmt1 (134)

pREP41X LEU2 Pnmt1* (134)

pREP81X LEU2 Pnmt1** (134)

AB 553-1 pREP3X LEU2 with a NotI between PstI and Pnmt1 This study + ETD 67-1 AB 553-1 LEU2 Ptrm8 trm8 This study ETD 70-1 AB 553-1 LEU2 SPATRNAPRO.05 SPBTRNAPRO.05 This study ETD 113-1 AB 553-1 LEU2 SPATRNATYR.01 This study ETD 115-1 AB 553-1 LEU2 SPATRNATYR.01 SPBTRNAPRO.05 This study + ETD 104-1 AB 553-1 LEU2 Pdhp1 dhp1 This study

ETD 109-1 pREP81X LEU2 Pnmt1*** gcn2+ This study

ETD 119-1 pREP41X LEU2 Pnmt1** tif221+ This study ETD 126 EIC-17 tV(AAC)E2 URA3 2u (39) 1132

De Zoysa 45

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1133 Table S5. Oligomers used for northern analysis 1134 Name Target RNAa Probe (5'-3') Sequence TDZ 114 tV(AAC) 73-54 TGATCGTGCCAAGACTCGAA TDZ 115 tMi(CAU) 73-55 TTGCGCGGCCAGGTTTCGA TDZ 123 tMe(CAU) 20-1 ACCGACTGAGCTACAGAAGC TDZ 124 tK(CUU) 73-55 CTCCCAAGGCGAGACTCGA TDZ 125 tK(UUU) 73-54 CTCCCACTGCGAGATTCGAA TDZ 126 tP(AGG) 72-53 GGGCTGTTGTGGGAATCGAA TDZ 128 tY(GUA) 73-52 TCTCCTGAGCCAGAATCGAACT TDZ 129 tV(CAC) 23-1 TCATACCACTAGAACACGGGACT TDZ 130 tV(UAC) 73-52 TGCATCTACCAGGGATCGAAC TDZ 131 tI(AAU) 73-55 TGGTCACAGCCGGGTTCGA TDZ 132 tN(GUU) 73-57 CGGTCAGGGAGGGACTC TDZ 137 tA(AGC) 73-50 GGACAAGCCAGAACTCGAATCT TDZ 138 tA(CGC) 73-54 TGGAGATGCCGGGAATCGAA TDZ 140 tT(AGU) 72-54 GCTCCAGCAGTGACTCGAA TDZ 141 tT(CGU) 54-34 ACACTGGACCTCCTGTTTACG TDZ 143 tP(UGG) 27-4 AGTATGCTACCACTACACCACGAA TDZ 155 5.8S rRNA 60-37 GCTGCGTTCTTCATCGATGCG TDZ 177 tP(AGG) 27-5 GGTATCATACCACTAGACCAAAC TDZ 182 tR(CCU) 39-17 CTCGTTAGGAATGAGATGCGCTA TDZ 184 tC(GCA) 22-1 CTAACCACTGAGCTATGACCCC TDZ 185 tW(CCA) 22-1 CTACCAACTGAGTTAAGGGGCC TDZ 186 tA(UGC) 73-54 TGGACACGCTGGGGATTGAA TDZ 364 5S rRNA 58-33 TAACTGCAGTGATCGGACGGG TDZ 395 pre-tY(GUA):2 In10-26 ATCGTTTTAATTACAGCCGGATG TDZ 396 pre-tY(GUA):1-1, 1-2 41-In-29 CGGTATCTTTATAATTACAGCCGG TDZ 397 pre-tY(GUA):1-3 41-In-29 CGGTATCATCGAAATTACAGCCGG LH 735 tL(UAA) 73-54 TGCGGCCAGAGAGGTTCGAA LH 737 tG(GCC) 73-53 TGCTTTGGCCGGGAATCGAAC Val(CCA) tV(AAC) 76-60 TGGTGATTTCGCCCAGGA tRNAPheloop tF(GAA) 64-45 TGTGGATCGAACACAGGACC TrpP1 tW(CCA) 33-15 AGT CGA AAG CTC TAC CAT 5S 5S 20-1 TGGTAGATATGGCCGCAACC 5'-tRNALys(UUU) tK(UUU) 18-1 CAACTGAGCTAACAAGGA LeuCAAP1 tL(CAA) 62-e3 AGATTCGAACTCTTGCATCTT 5'-tRNAArg(UCU) tR(UCU) 24-5 CGTTGCCATTACGCCACGC TDZ 415 pre-tV(AAC) +9-60 AAAAAAAATTGATTTCGCCCAGG 1135 1136 aNote that S. cerevisiae oligomers are indicated in italics 1137

De Zoysa 46

bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123083; this version posted May 29, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1138 1139 Table S6. Oligomers used for tRNA purifications 1140 Target Name RNA Probe (5'-3') Sequence TDZ 342 tF(GAA) 76-53 biotin-TGGTGTCACAAACCGGGATCGAAC TDZ 358 tY(GUA) 76-64 biotin-TGGTCTCCTGAGCCAGAATCGAACTA TDZ 359 tP(AGG) 76-54 biotin-TGGGGGCTGTTGTGGGAATCGAA 1141

De Zoysa 47