Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Selenocysteine tRNA methylation promotes oxidative stress resistance in melanoma metastasis

Abstract

Selenocysteine-containing proteins play a central role in redox homeostasis. Their translation is a highly regulated process and is dependent on two tRNASec isodecoders differing by a single 2′-O-ribose methylation called Um34. Here we characterized FTSJ1 as the Um34 methyltransferase and show that its activity is required for efficient selenocysteine insertion at the UGA stop codon during translation. Specifically, loss of Um34 leads to ribosomal stalling and decreased UGA recoding. FTSJ1-deficient cells are more sensitive to oxidative stress and show decreased metastatic colonization in xenograft models of melanoma metastasis. We found that FTSJ1 mediates efficient translation of selenoproteins essential for the cellular antioxidant response. Our findings uncover a role for tRNASec Um34 modification in oxidative stress resistance and highlight FTSJ1 as a potential therapeutic target specific for metastatic disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Selenoprotein translation and tRNASec Um34 levels increase in response to oxidative stress.
Fig. 2: FTSJ1 is the Um34 methyltransferase for tRNASec.
Fig. 3: Um34 regulates UGA recoding efficiency in a subset of selenoproteins involved in oxidative stress.
Fig. 4: Um34 modification protects cancer cells from oxidative stress.
Fig. 5: Micrometastases experience high levels of stress and increase selenocysteine biosynthesis.
Fig. 6: tRNASec Um34 modification is necessary and sufficient for metastasis in vivo.

Similar content being viewed by others

Data availability

Ribosomal-sequencing and RNA-sequencing data that support the findings of this study have been deposited in the Gene Expression Omnibus under accession codes GSE270971 and GSE270976. MS proteomics data and nucleoside modification analysis data have been provided as Supplementary Tables. All other data supporting the findings of this study are available from the corresponding author on reasonable request by email. Requests will be processed within 30 days. Source data are provided with this paper.

Code availability

All custom scripts used in this study are deposited at https://github.com/abcwcm/piskounova_ribo.

References

  1. Chan, C., Pham, P., Dedon, P. C. & Begley, T. J. Lifestyle modifications: coordinating the tRNA epitranscriptome with codon bias to adapt translation during stress responses. Genome Biol. 19, 228 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Suzuki, T. The expanding world of tRNA modifications and their disease relevance. Nat. Rev. Mol. Cell Biol. 22, 375–392 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Agris, P. F., Narendran, A., Sarachan, K., Vare, V. Y. P. & Eruysal, E. The importance of being modified: the role of RNA modifications in translational fidelity. Enzymes 41, 1–50 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Endres, L., Dedon, P. C. & Begley, T. J. Codon-biased translation can be regulated by wobble-base tRNA modification systems during cellular stress responses. RNA Biol. 12, 603–614 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Hatfield, D. L., Tsuji, P. A., Carlson, B. A. & Gladyshev, V. N. Selenium and selenocysteine: roles in cancer, health, and development. Trends Biochem. Sci. 39, 112–120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fradejas-Villar, N. et al. The RNA-binding protein SECISBP2 differentially modulates UGA codon reassignment and RNA decay. Nucleic Acids Res. 45, 4094–4107 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Small-Howard, A. et al. Supramolecular complexes mediate selenocysteine incorporation in vivo. Mol. Cell. Biol. 26, 2337–2346 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xu, X. M. et al. Evidence for direct roles of two additional factors, SECp43 and soluble liver antigen, in the selenoprotein synthesis machinery. J. Biol. Chem. 280, 41568–41575 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Hatfield, D. L., Carlson, B. A., Xu, X. M., Mix, H. & Gladyshev, V. N. Selenocysteine incorporation machinery and the role of selenoproteins in development and health. Prog. Nucleic Acid Res. Mol. Biol. 81, 97–142 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Kim, L. K. et al. Methylation of the ribosyl moiety at position 34 of selenocysteine tRNA[Ser]Sec is governed by both primary and tertiary structure. RNA 6, 1306–1315 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. van den Born, E. et al. ALKBH8-mediated formation of a novel diastereomeric pair of wobble nucleosides in mammalian tRNA. Nat. Commun. 2, 172 (2011).

    Article  PubMed  Google Scholar 

  12. Songe-Moller, L. et al. Mammalian ALKBH8 possesses tRNA methyltransferase activity required for the biogenesis of multiple wobble uridine modifications implicated in translational decoding. Mol. Cell. Biol. 30, 1814–1827 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Diamond, A. M. et al. Dietary selenium affects methylation of the wobble nucleoside in the anticodon of selenocysteine tRNA[Ser]Sec. J. Biol. Chem. 268, 14215–14223 (1993).

    Article  CAS  PubMed  Google Scholar 

  14. Howard, M. T., Carlson, B. A., Anderson, C. B. & Hatfield, D. L. Translational redefinition of UGA codons is regulated by selenium availability. J. Biol. Chem. 288, 19401–19413 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Carlson, B. A. et al. Selective restoration of the selenoprotein population in a mouse hepatocyte selenoproteinless background with different mutant selenocysteine tRNAs lacking Um34. J. Biol. Chem. 282, 32591–32602 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Li, Z. et al. Ribosome stalling during selenoprotein translation exposes a ferroptosis vulnerability. Nat. Chem. Biol. 18, 751–761 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yant, L. J. et al. The selenoprotein GPX4 is essential for mouse development and protects from radiation and oxidative damage insults. Free Radic. Biol. Med. 34, 496–502 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Ingold, I. et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Jakupoglu, C. et al. Cytoplasmic thioredoxin reductase is essential for embryogenesis but dispensable for cardiac development. Mol. Cell. Biol. 25, 1980–1988 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Conrad, M. et al. Essential role for mitochondrial thioredoxin reductase in hematopoiesis, heart development, and heart function. Mol. Cell. Biol. 24, 9414–9423 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tarrago, L. et al. The selenoprotein methionine sulfoxide reductase B1 (MSRB1). Free Radic. Biol. Med. 191, 228–240 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Cox, A. G. et al. Selenoprotein H is an essential regulator of redox homeostasis that cooperates with p53 in development and tumorigenesis. Proc. Natl Acad. Sci. USA 113, E5562–E5571 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sreelatha, A. et al. Protein AMPylation by an evolutionarily conserved pseudokinase. Cell 175, 809–821 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Addinsall, A. B., Wright, C. R., Andrikopoulos, S., van der Poel, C. & Stupka, N. Emerging roles of endoplasmic reticulum-resident selenoproteins in the regulation of cellular stress responses and the implications for metabolic disease. Biochem. J. 475, 1037–1057 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Kohrle, J. Thyroid hormone deiodinases—a selenoenzyme family acting as gate keepers to thyroid hormone action. Acta Med. Austriaca 23, 17–30 (1996).

    CAS  PubMed  Google Scholar 

  26. Shetty, S. P. & Copeland, P. R. The selenium transport protein, selenoprotein P, requires coding sequence determinants to promote efficient selenocysteine incorporation. J. Mol. Biol. 430, 5217–5232 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Reich, H. J. & Hondal, R. J. Why nature chose selenium. ACS Chem. Biol. 11, 821–841 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Brigelius-Flohe, R. & Flohe, L. Selenium and redox signaling. Arch. Biochem. Biophys. 617, 48–59 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Piskounova, E. et al. Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Endres, L. et al. ALKBH8 regulates selenocysteine-protein expression to protect against reactive oxygen species damage. PLoS ONE 10, e0131335 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Touat-Hamici, Z., Legrain, Y., Bulteau, A. L. & Chavatte, L. Selective up-regulation of human selenoproteins in response to oxidative stress. J. Biol. Chem. 289, 14750–14761 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hall, S. et al. Cellular effects of pyocyanin, a secreted virulence factor of Pseudomonas aeruginosa. Toxins 8, 236 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zhu, Z., Guo, R., Li, Y., Li, S. & Tu, P. Comparison of three analytical methods for superoxide produced by activated immune cells. J. Pharmacol. Toxicol. Methods 101, 106637 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Mistry, H. D. et al. Differential expression and distribution of placental glutathione peroxidases 1, 3 and 4 in normal and preeclamptic pregnancy. Placenta 31, 401–408 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Li, J. et al. Intellectual disability-associated gene FTSJ1 is responsible for 2′-O-methylation of specific tRNAs. EMBO Rep. 21, e50095 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Nagayoshi, Y. et al. Loss of Ftsj1 perturbs codon-specific translation efficiency in the brain and is associated with X-linked intellectual disability. Sci. Adv. 7, eabf3072 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ingolia, N. T. Ribosome footprint profiling of translation throughout the genome. Cell 165, 22–33 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. McGlincy, N. J. & Ingolia, N. T. Transcriptome-wide measurement of translation by ribosome profiling. Methods 126, 112–129 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Karlenius, T. C. et al. The selenium content of cell culture serum influences redox-regulated gene expression. Biotechniques 50, 295–301 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Carlson, B. A., Xu, X. M., Gladyshev, V. N. & Hatfield, D. L. Selective rescue of selenoprotein expression in mice lacking a highly specialized methyl group in selenocysteine tRNA. J. Biol. Chem. 280, 5542–5548 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. Guo, L. et al. Selenocysteine-specific mass spectrometry reveals tissue-distinct selenoproteomes and candidate selenoproteins. Cell Chem. Biol. 25, 1380–1388 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Bak, D. W., Gao, J., Wang, C. & Weerapana, E. A quantitative chemoproteomic platform to monitor selenocysteine reactivity within a complex proteome. Cell Chem. Biol. 25, 1157–1167 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Schoenmakers, E. et al. Mutation in human selenocysteine transfer RNA selectively disrupts selenoprotein synthesis. J. Clin. Invest. 126, 992–996 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Vindry, C. et al. A homozygous mutation in the human selenocysteine tRNA gene impairs UGA recoding activity and selenoproteome regulation by selenium. Nucleic Acids Res. 51, 7580–7601 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Quintana, E. et al. Human melanoma metastasis in NSG mice correlates with clinical outcome in patients. Sci. Transl. Med. 4, 159ra149 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Moustafa, M. E. et al. Selective inhibition of selenocysteine tRNA maturation and selenoprotein synthesis in transgenic mice expressing isopentenyladenosine-deficient selenocysteine tRNA. Mol. Cell. Biol. 21, 3840–3852 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Fradejas-Villar, N. et al. The effect of tRNA[Ser]Sec isopentenylation on selenoprotein expression. Int. J. Mol. Sci. 22, 11454 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Guy, M. P. et al. Defects in tRNA anticodon loop 2′-O-methylation are implicated in nonsyndromic X-linked intellectual disability due to mutations in FTSJ1. Hum. Mutat. 36, 1176–1187 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Takano, K. et al. A loss-of-function mutation in the FTSJ1 gene causes nonsyndromic X-linked mental retardation in a Japanese family. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147B, 479–484 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Goodarzi, H. et al. Modulated expression of specific tRNAs drives gene expression and cancer progression. Cell 165, 1416–1427 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Crain, P. F. Preparation and enzymatic hydrolysis of DNA and RNA for mass spectrometry. Methods Enzymol. 193, 782–790 (1990).

    Article  CAS  PubMed  Google Scholar 

  52. Corish, P. & Tyler-Smith, C. Attenuation of green fluorescent protein half-life in mammalian cells. Protein Eng. 12, 1035–1040 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Shah, A., Qian, Y., Weyn-Vanhentenryck, S. M. & Zhang, C. CLIP Tool Kit (CTK): a flexible and robust pipeline to analyze CLIP sequencing data. Bioinformatics 33, 566–567 (2017).

    Article  CAS  PubMed  Google Scholar 

  54. Volders, P. J. et al. LNCipedia: a database for annotated human lncRNA transcript sequences and structures. Nucleic Acids Res. 41, D246–D251 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Quintana, E. et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell 18, 510–523 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Quintana, E. et al. Efficient tumour formation by single human melanoma cells. Nature 456, 593–598 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the American Cancer Society (E.P.), an Elsa U. Pardee Foundation Grant (E.P.) and a Feldstein Medical Research Foundation Award (E.P.). L.A.N. is supported by a Ruth L. Kirschstein Predoctoral Individual National Research Service Award.

Author information

Authors and Affiliations

Authors

Contributions

L.A.N. designed and performed experiments, analyzed data and wrote the paper. K.P.C., I.D., M.Z., G.C., R.O.H. and K.N.A. performed experiments and analyzed data. L.E.D. supervised CRISPR–Cas9 experiments and provided reagents. S.M. and S.J. designed, performed and supervised the ribosomal-sequencing experiments. M.A., P.Z. and D.B. designed, performed and supervised ribosomal-sequencing and RNA-sequencing data analysis. E.P. designed, performed and supervised experiments, analyzed data, supervised the data analysis and wrote the paper.

Corresponding author

Correspondence to Elena Piskounova.

Ethics declarations

Competing interests

L.E.D. is an advisor and holds equity in Mirimus, Inc., and is a consultant for Volastra Therapeutics and Frazier Healthcare Partners. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Extracellular selenium drives translation of selenoproteins.

(A) Levels and purity of tRNASec analyzed by SDS-PAGE gel after biotinylated pull-down from increasing amounts of small RNA input fraction. Representative image of 3 independent experiments shown. (B) Western blot analysis of selenocysteine proteins in DMEM/F12 media (0 nM Se) with increasing amounts of selenium in A375 cells. Representative image of 2 independent experiments shown. (C) Transcript levels of selenocysteine proteins in DMEM/F12 media (0 nM Se) with increasing amounts of selenium in A375 cells measured by qRT-PCR. Mean +/- SD. n = 3 independent replicates. Statistical significance assessed by two-way analysis of variance (ANOVA) with each condition compared to the zero selenium condition.

Source data

Extended Data Fig. 2 FTSJ1 is necessary and sufficient for Um34 modification on tRNASec.

(A) Co-immunoprecipitation assay of the interaction between HA-tagged FTSJ1 and FLAG-tagged component of selenocysteine machinery, SECp43, compared to empty vector control co-expressed in A375 melanoma cells. Representative experiment of 3 independent experiments shown. (B) Levels of Phe tRNA bound to FLAG-tagged Renilla, wild type or A26P-mutant FTSJ1, ALKBH8 and EEFSEC measured by qRT-PCR. n = 3. Mean +/- SD of 3 independent experiments. Two-sided unpaired t-test. (C) Lys UUU and Glu CUC tRNA levels bound to FLAG-tagged Renilla, wild type and A26P FTSJ1, ALKBH8 and EEFSEC by qRT-PCR. n = 3. Mean +/- SD of 3 independent experiments. (D) Quantification of Um34 levels on tRNASec in FTSJ1-KO SK-MEL-28 melanoma cells measured by LC-MS/MS. n = 6 from two independent experiments. Mean +/- SD. (E) Quantification of Um34 levels on tRNASec in FTSJ1-KO SK-MEL-2 melanoma cells measured by LC-MS/MS. n = 4 from two independent experiments. Mean +/- SD. (F) Quantification of Um34 levels in SK-MEL-28 melanoma cells overexpressing wild-type FTSJ1 or A26P-mutant. n = 3 independent experiments. Mean +/- SD. One-way analysis of variance (ANOVA) followed by Dunnett’s tests for multiple comparisons. (G) Quantification of Um34 modification levels on tRNASec in Control and FTSJ1-KO A375 melanoma cells overexpressing either Renilla luciferase, WT FTSJ1 or mutant A26P FTSJ1 measured by LC-MS/MS. n = 3-6 from two independent experiments. Mean +/- SD. Two-tailed one sample t-test. (H) Quantification of Um34 modification levels on tRNASec in Control and FTSJ1-KO HeLa cells overexpressing Renilla luciferase, WT FTSJ1 or mutant A26P FTSJ1 measured by LC-MS/MS. n = 2 independent experiments. Mean +/- SD. (I) Levels of recombinant FTSJ1 from a His- tag purification assessed by Coomassie staining. Representative of 2 independent experiments shown. (J) Levels of recombinant WDR6 from a His-tag purification assessed by Western blotting for WDR6. Representative of 2 independent experiments shown.

Source data

Extended Data Fig. 3 FTSJ1 regulates recoding of UGA as a selenocysteine.

(A) Global analysis of codon readthrough in FTSJ1-KO A375 melanoma cells compared to control A375 melanoma cells in the presence of selenium (30 nM). Pearson Correlation was used for statistical analysis, gray band is 95% confidence interval. (B) Levels of selenoprotein transcripts in Control and FTSJ1-KO A375 and SK-MEL-2 melanoma cell lines as measured by qRT-PCR. Mean +/- SD from 3 independent replicates. Mean +/- SD. Two-way ANOVA followed by Fisher LSD test for multiple comparisons. (C) Western blot analysis of selenocysteine proteins TXNRD1, GPX1 and GPX4 in normal (10% FBS), selenium-depleted (2%) and supplemented conditions (10% + 30 nM NaSe and 2% + 30 nM NaSe) in A375 melanoma cell line, HEK293Ts and SK-MEL-2 melanoma cell line. Representative of 3 independent experiments shown. (D) Transcript levels of TXNRD1, GPX4 and GPX1 in normal (10% FBS), selenium-depleted (2%) and supplemented conditions (10% + 30 nM NaSe and 2% + 30 nM NaSe) in A375 melanoma cell line by qRT-PCR. Mean +/- SD of 3 independent replicates. Two-sided unpaired t-test. (E) Western blot analysis of selenocysteine-containing proteins immunoprecipitated with selenocysteine-specific pulldown. SECISBP2 was used as a non-selenocysteine control protein. 1% of total protein input was run. Representative of 2 independent experiments shown. (F) Western blot analysis of selenocysteine-containing proteins immunoprecipitated with selenocysteine-specific pulldown from Control and FTSJ1-KO A375 melanoma cells. 1% of total protein input was run. (* = longer exposure of film) Representative of 3 independent experiments shown. (G) Western blot analysis of selenocysteine-containing proteins immunoprecipitated with selenocysteine-specific pulldown from A375 melanoma cells overexpressing Renilla or FTSJ1. 1% of total protein input was run. Representative of 2 independent experiments shown.

Source data

Extended Data Fig. 4 Loss of FTSJ1 causes ribosomal stalling around Sec UGA in a subset of selenoproteins.

(A) Cumulative RPF plots for affected selenoproteins in WT A375 melanoma cells in the absence (red) and presence (blue) of selenium (30 nM NaSe). (B) Cumulative RPF plots for affected selenoproteins in Control (blue) and FTSJ1-KO (red) melanoma cells in the presence of selenium (30 nM NaSe).

Extended Data Fig. 5 Loss of FTSJ1 has no additional effect on selenoprotein translation in the absence of selenium.

(A) Cumulative RPF plots for unaffected selenoproteins in WT A375 melanoma cells in the absence (red) and presence (blue) of selenium (30 nM NaSe). (B) Cumulative RPF plots for unaffected selenoproteins in Control (blue) and FTSJ1-KO (red) melanoma cells in the presence of selenium (30 nM NaSe).

Extended Data Fig. 6 Loss of FTSJ1 causes ribosomal stalling upstream of the Sec UGA in selenoproteins.

Non-cumulative RPF plots for all detected selenoproteins in Control or FTSJ1-KO A375 melanoma cells in the presence of selenium (30 nM NaSe).

Extended Data Fig. 7 Sensitivity to oxidative stress can not be rescued by overexpression of other FTSJ1 tRNA targets.

(A) Mitochondrial ROS levels measured by flow cytometry using CellROXGreen dye in Control and A375 melanoma cells in the presence of selenium (30 nM). n = 3 independent experiments. (B) Cytoplasmic ROS levels measured by flow cytometry using CellROX DeepRed in Control and A375 melanoma cells in the presence of selenium (30 nM). n = 3 independent experiments. (C) Lipid peroxidation levels measured by flow cytometry using BODIPY665/676 in Control and A375 melanoma cells in the presence of selenium (30 nM). Mean +/- SD. Representative of 2 independent experiments. (D-E) Cell survival of Control vs FTSJ1-KO A375 melanoma cells under increasing concentrations of RSL3 in the presence (D) or absence (E) of selenium. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). (F-G) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without FTSJ1 overexpression under increasing concentrations of RSL3 in the presence (F) or absence (G) of selenium. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). (H-I) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without FTSJ1 overexpression under increasing concentrations of RSL3 in the absence (H) or presence (I) of selenium. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). (J-K) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression of Phe tRNA with 25uM of pyocyanin in the presence (J) or absence (K) of selenium (30 nM NaSe). n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). (L) Quantification of Um34 modification levels on tRNASec in Control, WT tRNASec or C65G mutant tRNASec overexpressing A375 melanoma cells. n = 3 independent experiments. Mean +/- SD. Ordinary one-way ANOVA followed by Uncorrected Fisher’s LSD test for multiple comparisons. (M-N) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression of Phe tRNA with 25uM of pyocyanin in the presence (M) or absence (N) of selenium (30nM NaSe). n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). (O-P) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression of Trp tRNA with 25uM of pyocyanin in the presence (O) or absence (P) of selenium (30nM NaSe). n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided).

Source data

Extended Data Fig. 8 FTSJ1 overexpression can partially rescue sensitivity to oxidative stress while other tRNA targets of FTSJ1 can not.

(A) Cell survival of Control and FTSJ1-KO A375 melanoma cells with and without FTSJ1-overexpression under increasing concentrations of pyocyanin in the presence of selenium (30 nM NaSe) as measured by Cell Titer Glo. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). Representative of 3 independent experiments. (B) Cell survival of Control and FTSJ1-KO A375 melanoma cells with and without FTSJ1-overexpression under increasing concentrations of pyocyanin in the absence of selenium as measured by Cell Titer Glo. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). Representative of 3 independent experiments. (C) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression Phe tRNA under increasing concentrations of pyocyanin in the presence of selenium (30 nM NaSe). n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). Representative of 3 independent experiments. (D) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression Phe tRNA under increasing concentrations of pyocyanin in the absence of selenium. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). Representative of 3 independent experiments. (E) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression Trp tRNA under increasing concentrations of pyocyanin in the presence of selenium (30 nM NaSe). n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). Representative of 3 independent experiments. (F) Cell survival of Control or FTSJ1-KO A375 melanoma cells with or without overexpression Trp tRNA under increasing concentrations of pyocyanin in the absence of selenium. n = 3 independent culture experiments. Mean +/- SD. Representative of 3 independent experiments. Statistical difference between IC50 values analyzed by Extra-Sum-of-Squares F-Test (two-sided). Representative of 3 independent experiments.

Source data

Extended Data Fig. 9 Metastases show translational upregulation of selenoproteins.

(A) Transcript levels of selenoproteins in primary tumors compared to macro and micro metastatic nodules, isolated from n = 3 NSG mice transplanted with patient-derived melanomas M481 or M405, as measured by RNA-sequencing, quantified as a z-score. Table of p-values and log2 Fold Change listed in Appendix A. p-values were generated using DESeq2 algorithm (Wald test). Multiple comparisons adjustment was calculated using False Discovery Rate (FDR)/Benjamini-Hochberg test. (B) Levels of precursor tRNASec in Primary, macro and micrometastatic nodules from patient-derived M405 melanoma measured by qRT-PCR. Mean +/- SD from 3 independent replicates. One-way analysis of variance (ANOVAs) followed by Dunnett’s tests for multiple comparisons. (C) Levels of mature tRNASec in Primary, macro and micrometastatic nodules from patient-derived M405 melanoma measured by qRT-PCR. Mean +/- SD from 3 independent replicates. One-way analysis of variance (ANOVAs) followed by Dunnett’s tests for multiple comparisons. (D) Levels of gROSI variant levels with No UTR, GPX1 SECIS or GPX4 SECIS in primary and metastatic nodules, formed by A375 melanoma cells transplanted into NSG mice, as measured by immunofluorescence staining for GFP expression. n = number of mice analyzed, indicated in the figure. (E) Quantification of the GFP signal from gROSI variants shown in (E). Mean +/- SD. Two-way ANOVA followed by two-tailed Fisher’s LSD test.

Source data

Extended Data Fig. 10 Loss of FTSJ1 affects metastatic colonization but has no effect on lipid peroxidation levels.

(A) Western blot analysis of control and FTSJ1-KO A375 melanoma primary tumors. Representative from 3 independent experiments. (B) qRT-PCR analysis of FTSJ1-targeting shRNAs in A375 melanoma cells. Mean +/- SD from 3 independent replicates. One-way analysis of variance (ANOVAs) followed by Dunnett’s tests for multiple comparisons. (C) Western blot analysis of FTSJ1-targeting shRNAs in MeWo melanoma cells. Representative from 3 independent experiments. (D) Subcutaneous tumor growth of MeWo cells transplanted into mice expressing Control or shRNA5-targeting FTSJ1. n = 5 mice per group. Representative of 3 independent experiments. Mean +/- SD. (E-F) Western blot analysis of (E) M405 or (F) M481 tumors with either control or FTSJ1-targeting shRNAs. Representative from 4 independent experiments. (G-H) Western blot analysis of (G) A375 or (H) M481 melanoma tumors overexpressing Control Renilla luciferase or FTSJ1. Representative from 3 independent experiments. (I) Growth of subcutaneous tumors in mice transplanted from patient-derived melanoma, M405, overexpressing either Renilla luciferase or FTSJ1. n = 5 mice per group. Mean +/- SD. Representative of 2 independent experiments. (J) Total metastatic burden in visceral organs of mice transplanted with patient-derived melanoma, M405, overexpressing either Renilla luciferase or FTSJ1. n = number of mice analyzed, indicated in the figure. Mean +/- SD. Statistical significance assessed by one-way analysis of variance (ANOVAs) followed by Dunnett’s tests for multiple comparisons. (K) Total metastatic burden in visceral organs of mice transplanted intravenously with either Control or FTSJ1 KO A375 melanoma cells. n = number of mice analyzed, indicated in the figure. Mean +/- SD. Unpaired two-tailed t-test. (L) Total metastatic burden in visceral organs of mice transplanted intravenously with either Control or FTSJ1 overexpressing A375 melanoma cells. n = number of mice analyzed, indicated in the figure. Mean +/- SD. (M) Quantification of 4-HNE immunofluorescent staining of subcutaneous tumors expressing either control shRNA or FTSJ1-targeting shRNAs. n = 3 mice, each replicate is a tumor. Mean +/- SD. Ordinary one-way ANOVA, multiple comparisons calculated using uncorrected Fisher’s LSD test. (N) Flow cytometry analysis of lipid peroxidation levels in subcutaneous tumors overexpressing Control Renilla luciferase or FTSJ1, using BODIPY dye. n = 10 mice per group. Unpaired two-tailed t-test.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–3: gating strategies.

Reporting Summary

Supplementary Tables 1–3

Supplementary Tables 1–3.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nease, L.A., Church, K.P., Delclaux, I. et al. Selenocysteine tRNA methylation promotes oxidative stress resistance in melanoma metastasis. Nat Cancer 5, 1868–1884 (2024). https://doi.org/10.1038/s43018-024-00844-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s43018-024-00844-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer