Aminoglycoside tolerance in Vibrio cholerae engages translational reprogramming associated to queuosine tRNA modification

  1. Institut Pasteur, Université de Paris Cité, Unité Plasticité du Génome Bactérien, UMR3525, CNRS, 75015, Paris, France
  2. Sorbonne Université, Collège Doctoral, F-75005, Paris, France
  3. Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
  4. Institut Pasteur, Université de Paris, Proteomics Platform, Mass Spectrometry for Biology Unit, UAR2024, CNRS 2000, Paris, France
  5. Institut Pasteur, Université de Paris, Department of Computation Biology, Bioinformatics and Biostatistics Hub, Paris, France
  6. Institut Pasteur, Université Paris cité, Epigenetic Chemical Biology Unit, UMR 3523, CNRS, 75015, Paris, France
  7. Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), F-91198, Gif-sur-Yvette, France
  8. Université de Lorraine, CNRS, Inserm, UAR2008/US40 IBSLor, Epitranscriptomics and RNA Sequencing Core Facility and UMR7365 IMoPA, F-54000, Nancy, France
  9. Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida 32611, United States
  10. University of Florida Genetics Institute, Florida 32611, United States

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Srujana Samhita Yadavalli
    Rutgers, The State University of New Jersey, Piscataway, United States of America
  • Senior Editor
    Wendy Garrett
    Harvard T.H. Chan School of Public Health, Boston, United States of America

Reviewer #1 (Public Review):

Summary of the work: In this work, Fruchard et. al. study the enzyme Tgt and how it modifies guanine in tRNAs to queuosine (Q), essential for Vibrio cholerae's growth under aminoglycoside stress. Q's role in codon decoding efficiency and its proteomic effects during antibiotic exposure is examined, revealing Q modification impacts tyrosine codon decoding and influences RsxA translation, affecting the SoxR oxidative stress response. The research proposes Q modification's regulation under environmental cues reprograms the translation of genes with tyrosine codon bias, including DNA repair factors, crucial for bacterial antibiotic response.

The experiments are well-designed and conducted and the conclusions, for the most part, are well supported by the data. However, a few clarifications will significantly strengthen the manuscript.

Major:
Figure S4 A-D. These growth curves are important data and should be presented in the main figures. Moreover, given that it is not possible to make a rsxA mutant, I wonder if it would be possible to connect rsx and tgt using the following experiment: expression of tgt results in resistance to TOB (in B), while expression of only rsx lower resistance to TOB (in D). Then simultaneous overexpression of both tgt/rsx in the WT strain should have either no effect on TOB resistance or increased resistance, relative to the WT. Perhaps the authors have done this, and if so, the data should be included as it will significantly strengthen their model.

Figure S4 - Is there a rationale for why it is possible to make rsx mutants in E. coli, but not in V. cholerae? For example, does E. coli have a second gene/protein that is redundant in function to rsxA, while V. cholerae does not? I think your data hint at this, since in the right panel growth data, your double mutant does not fully rescue back to rsx single mutant levels, suggesting another factor in tgt mutant also acts to lower resistance to TOB. If so, perhaps a line or two in text will be helpful for readers.

-For growth curves in Figure 2 and relative comparisons like in Figure 5D and Figure S4 (and others in the paper), statistics and error bars, along with replicate information should be provided.

-Figure 6A - Is the transcript fold change in linear or log? If linear, then tgt expression should not be classified as being upregulated in TOB. It is barely up by ~2-fold with TOB- 0.6....which is a mild phenotype, at best.

-Line 779- 780: "This indicates that sub-MIC TOB possibly induces tgt expression through the stringent response activation." To me, the data presented in this figure, do not support this statement. The experiment is indirect.

-Figure 3B and D. - These samples only have tobramycin, correct? The legend says both carbenicillin and tobramycin.

-Figure 5. The color schemes in bars do not match up with the color scheme in cartoons below panels B and C. That makes it confusing to read. Please fix.

-A lot of abbreviations have been used. This makes reading a bit cumbersome. Ideally, less abbreviations will be used.

Reviewer #2 (Public Review):

Fruchard et al. investigate the role of the queuosine (Q) modification of the tRNA (Q-tRNA) in the human pathogen Vibrio cholerae. First, the authors state that the absence of Q-modified tRNAs (tgt mutant) increases the translation of TAT codons and proteins with a high TAT codon bias. Second, the absence of Q increases rsxA translation, because rsxA gene has a high TAT codon bias. Third, increased RsxA in the absence of Q inhibits SoxR response, reducing resistance towards the antibiotic tobramycin (TOB). Authors also predict in silico which genes harbor a higher TAT bias and found that among them are some involved in DNA repair, experimentally observing that a tgt mutant is more resistant to UV than the wt strain. It is worth noting that authors employ a wide variety of techniques, both experimental and bioinformatic. However, some aspects of the work need to be clarified or reevaluated.

(1) The statement that the absence of Q increases the translation of TAT codons and proteins encoded by TAT-enriched genes presents the following problems that should be addressed:

(1.1) The increase in TAT codon translation in the absence of Q is not supported by proteomics, since there was no detected statistical difference for TAT codon usage in proteins differentially expressed. Furthermore, there are some problems regarding the statistics of proteomics. Some proteins shown in Table S1 have adjusted p-values higher than their p-values, which makes no sense. Maybe there is a mistake in the adjusted p-value calculation. In addition, it is not common to assume that proteins that are quantitatively present in one condition and absent in another are differentially abundant proteins. Proteomics data software typically addresses this issue and applies some corrections. It would be advisable to review that.

(1.2) Problems with the interpretation of Ribo-seq data (Figure 4D). On the one hand, the Ribo-seq data should be corrected (normalized) with the RNA-seq data in each of the conditions to obtain ribosome profiling data, since some genes could have more transcription in some of the conditions studied. In other articles in which this technique is used (such as in Tuorto et al., EMBO J. 2018; doi: 10.15252/embj.201899777), it is interpreted that those positions in which the ribosome moves most slowly and therefore less efficiently translated), are the most abundant. Assuming this interpretation, according to the hypothesis proposed in this work, the fragments enriched in TAT codons should have been less abundant in the absence of Q-tRNA (tgt mutant) in the Rib-seq experiment. However, what is observed is that TAT-enriched fragments are more abundant in the tgt mutant, and yet the Ribo-seq results are interpreted as RNA-seq, stating that this is because the genes corresponding to those sequences have greater expression in the absence of Q. On the other hand, it would be interesting to calculate the mean of the protein levels encoded by the transcripts with high and low ribosome profiling data.

(1.3) This statement is contrary to most previously reported studies on this topic in eukaryotes and bacteria, in which ribosome profiling experiments, among others, indicate that translation of TAT codons is slower (or unaffected) than translation of the TAC codons, and the same phenomenon is observed for the rest of the NAC/T codons. This is completely opposed to the results showed in Figure 4. However, the results of these studies are either not mentioned or not discussed in this work. Some examples of articles that should be discussed in this work:
- "Queuosine-modified tRNAs confer nutritional control of protein translation" (Tuorto et al., 2018; 10.15252/embj.201899777)
- "Preferential import of queuosine-modified tRNAs into Trypanosoma brucei mitochondrion is critical for organellar protein synthesis" (Kulkarni et al., 2021; doi:10.1093/nar/gkab567.
- "Queuosine-tRNA promotes sex-dependent learning and memory formation by maintaining codon-biased translation elongation speed" (Cirzi et al., 2023; 10.15252/embj.2022112507)
- "Glycosylated queuosines in tRNAs optimize translational rate and post-embryonic growth" (Zhao et al., 2023; 10.1016/j.cell.2023.10.026)
- "tRNA queuosine modification is involved in biofilm formation and virulence in bacteria" (Diaz-Rullo and Gonzalez-Pastor, 2023; doi: 10.1093/nar/gkad667). In this work, the authors indicate that Q-tRNA increases NAT codon translation in most bacterial species. Could the regulation of TAT codon-enriched proteins by Q-tRNAs in V. cholerae an exception? In addition, authors use a bioinformatic method to identify genes enriched in NAT codons similar to the one used in this work, and to find in which biological process are involved the genes whose expression is affected by Q-tRNAs (as discussed for the phenotype of UV resistance). It will be worth discussing all of this.

(1.4) It is proposed that the stress produced by the TOB antibiotic causes greater translation of genes enriched in TAT codons. On the one hand, it is shown that the GFP-TAT version (gene enriched in TAT codons) and the RsxA-TAT-GFP protein (native gene naturally enriched in TAT) are expressed more, compared to their versions enriched in TAC in a tgt mutant than in a wt, in the presence of TBO (Fig. 5C). However, in the absence of TOB, and in a wt context, although the two versions of GFP have a similar expression level (Fig. 3SD), the same does not occur with RsxA, whose RsxA-TAT form (the native one) is expressed significantly more than the RsxA-TAC version (Fig. 3SA). How can it be explained that in a wt context, in which there are also tRNA Q-modification, a gene naturally enriched in TAT is translated better than the same gene enriched in TAC? It would be expected that in the presence of Q-tRNAs the two versions would be translated equally (as happens with GFP) or even the TAT version would be less translated. On the other hand, in the presence of TOB the fluorescence of WT GFP(TAT) is higher than the fluorescence of WT GFP(TAC) (Figure S3E) (mean fluorescence data for RsxA-GFP version in the presence of TOB is not shown). These results may indicate that the apparent better translation of TAT versions could be due to indirect effects rather from TAT codon translation.

(2) Another problem is related to the already known role of Q in prevention of stop codon readthrough, which is not discuss at all in the work. In the absence of Q, stop codon readthrough is increased. In addition, it is known that aminoglycosides (such as tobramycin) also increase stop codon readthrough ("Stop codon context influences genome-wide stimulation of termination codon readthrough by aminoglycosides"; Wanger and Green, 2023; 10.7554/eLife.52611). Absence of Q and presence of aminoglycosides can be synergic, producing devastating increases in stop codon readthrough and a large alteration of global gene expression. All of these needs to be discussed in the work. Moreover, it is known that stop codon readthrough can alter gene expression and mRNA sequence context all influence the likelihood of stop codon readthrough. Thus, this process could also affect to the expression of recoded GFP and RsxA versions.

(3) The statement about that the TOB resistance depends on RsxA translation, which is related to the presence of Q, also presents some problems:

(3.1) It is observed that the absence of tgt produces a growth defect in V. cholerae when exposed to TOB (Figure 1A), and it is stated that this is mediated by an increase in the translation of RsxA, because its gene is TAT enriched. However, in Figure S4F, it is shown that the same phenotype is observed in E. coli, but its rsxA gene is not enriched in TAT codons. Therefore, the growth defect observed in the tgt mutant in the presence of TOB may not be due to the increase in the translation of TAT codons of the rsxA gene in the absence of Q. This phenotype is very interesting, but it may be related to another molecular process regulated by Q. Maybe the role of Q in preventing stop codon readthrough is important in this process, reducing cellular stress in the presence of TOB and growing better.

(3.2) All experiments related to the effect of Q on the translation of TAT codons have been performed with the tgt mutant strain. Considering that the authors have a pSEVA-tgt plasmid to overexpress this gene, they would have to show whether tgt overexpression in a wt strain produces a decrease in the translation of proteins encoded by TAT-enriched genes such as RsxA. This experiment would allow them to conclude that Q reduces RsxA levels, increasing resistance to TOB.

(3.3) On the other hand, Fig. 1B shows that when the wt and tgt strains compete, both overexpressing tgt, the tgt mutant strain grows better in the presence of TOB. This result is not very well understood, since according to the hypothesis proposed, the absence of modification by Q of the tRNA would increase the translation of genes enriched in TAT, therefore, a strain with a higher proportion of Q-modified tRNAs as in the case of the wt strain overexpressing tgt would express the rsxA gene less than the tgt strain overexpressing tgt and would therefore grow better in the presence of TOB. For all these reasons, it would be necessary to evaluate the effect of tgt overexpression on the translation of RsxA.

(3.4) According to Figure 1I, the overexpression of tRNA-Tyr(GUA) caused a better growth of tgt mutant in comparison to WT. If the growth defect observed in tgt mutant in the presence of TOB is due to a better translation of the TAT codons of rsxA gene, the overexpression of tRNA-Tyr(GUA) in the tgt mutant should have resulted in even better RsxA translation a worse growth, but not the opposite result.

(4) It cannot be stated that DNA repair is more efficient in the tgt mutant of V. cholerae, as indicated in the text of the article and in Fig 7. The authors only observe that the tgt mutant is more resistant to UV radiation and it is suggested that the reason may be TAT bias of DNA repair genes. To validate the hypothesis that UV resistance is increased because DNA repair genes are TAT biased, it would be necessary to check if DNA repair is affected by Q. UV not only produces DNA damage, but also oxidative stress. Therefore, maybe this phenotype is due to the increase in proteins related to oxidative stress controlled by RsxA, such as the superoxide dismutase encoded by sodA. It is also stated that these repair genes were found up for the tgt mutant in the Ribo-seq data, with unchanged transcription levels. Again, it is necessary to clarify this interpretation of the Ribo-seq data, since the fact that they are more represented in a tgt mutant perhaps means that translation is slower in those transcripts. Has it been observed in proteomics (wt vs tgt in the absence of TOB) whether these proteins involved in repair are more expressed in a tgt mutant?

(5) The authors demonstrate that in E. coli the tgt mutant does not show greater resistance to UV radiation (Fig. 7D), unlike what happens in V. cholerae. It should be discussed that in previous works it has been observed that overexpression in E. coli of the tgt gene or the queF gene (Q biosynthesis) is involved in greater resistance to UV radiation (Morgante et al., Environ Microbiol, 2015 doi: 10.1111/1462-2920.12505; and Díaz-Rullo et al., Front Microbiol. 2021 doi: 10.3389/fmicb.2021.723874). As an explanation, it was proposed (Diaz-Rullo and Gonzalez-Pastor, NAR 2023 doi: 10.1093/nar/gkad667) that the observed increase in the capacity to form biofilms in strains that overexpress genes related to Q modification of tRNA would be related to this greater resistance to UV radiation.

Reviewer #3 (Public Review):

Summary:

In this manuscript the authors begin with the interesting phenotype of sub-inhibitory concentrations of the aminoglycoside tobramycin proving toxic to a knockout of the tRNA-guanine transglycosylase (Tgt) of the important human pathogen, Vibrio cholerae. Tgt is important for incorporating queuosine (Q) in place of guanosine at the wobble position of GUN codons. The authors go on to define a mechanism of action where environmental stressors control expression of tgt to control translational decoding of particularly tyrosine codons, skewing the balance from TAC towards TAT decoding in the absence of the enzyme. The authors use advanced proteomics and ribosome profiling to reveal that the loss of tgt results in increased translation of proteins like RsxA and a cohort of DNA repair factors, whose genes harbor an excess of TAT codons in many cases. These findings are bolstered by a series of molecular reporters, mass spectrometry, and tRNA overexpression strains to provide support for a model where Tgt serves as a molecular pivot point to reprogram translational output in response to stress.

Strengths:

The manuscript has many strengths. The authors use a variety of strains, assays, and advanced techniques to discover a mechanism of action for Tgt in mediating tolerance to sub-inhibitory concentrations of tobramycin. They observe a clear phenotype for a tRNA modification in facilitating reprogramming of the translational response, and the manuscript certainly has value in defining how microbes tolerate antibiotics.

Weaknesses:

The conclusions of the manuscript are mostly very well-supported by the data, but in some places control experiments or peripheral findings cloud precise conclusions. Some additional clarification, discussion, or even experimental extension could be useful in strengthening these areas.

(1) The authors have created and used a variety of relevant molecular tools. In some cases, using these tools in additional assays as controls would be helpful. For example, testing for compensation of the observed phenotypes by overexpression of the Tyrosine tRNA(GUA) in Figure 2A with the 6xTAT strain, Figure 5C with the rxsA-GFP fusion, and/or Figure 7B with UV stress would provide additional information of the ability of tRNA overexpression to compensate for the defect in these situations.
(2) The authors present a clear story with a reprogramming towards TAT codons in the knockout strain, particularly regarding tobramycin treatment. The control experiments often hint at other codons also contributing to the observed phenotypes (e.g., His or Asp), yet these effects are mostly ignored in the discussion. It would be helpful to discuss these findings at a minimum in the discussion section, or possibly experimentally address the role of His or Asp by overexpression of these tRNAs together with Tyrosine tRNA(GUA) in an experiment like that of Figure 1I to see if a more "wild type" phenotype would present. In fact, the synergy of Tyr, His, and/or Asp codons likely helps to explain the effects observed with the DNA repair genes in later experiments.
(3) Regarding Figure 6D, the APB northern blot feels like an afterthought. It was loaded with different amounts of RNA as input and some samples are repeated three times, but Δcrp only once. Collectively, it makes this experiment very difficult to assess.

Minor Points:
(4) Fig S2B, do the authors have a hypothesis why the Asp and Phe tRNAs lead to a growth decrease in the untreated samples? It appears like Phe(GAA) partially compensates for the defect.
(5) Lines 655 to 660 seem more appropriate as speculation in the discussion rather than as a conclusion in the results, where no direct experiments are performed. The authors might take advantage of the "Ideas and Speculation" section that eLife allows.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation