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:

Proteasome-dependent truncation of the negative heterochromatin regulator Epe1 mediates antifungal resistance

Abstract

Epe1 histone demethylase restricts H3K9-methylation-dependent heterochromatin, preventing it from spreading over, and silencing, gene-containing regions in fission yeast. External stress induces an adaptive response allowing heterochromatin island formation that confers resistance on surviving wild-type lineages. Here we investigate the mechanism by which Epe1 is regulated in response to stress. Exposure to caffeine or antifungals results in Epe1 ubiquitylation and proteasome-dependent removal of the N-terminal 150 residues from Epe1, generating truncated Epe1 (tEpe1) which accumulates in the cytoplasm. Constitutive tEpe1 expression increases H3K9 methylation over several chromosomal regions, reducing expression of underlying genes and enhancing resistance. Reciprocally, constitutive non-cleavable Epe1 expression decreases resistance. tEpe1-mediated resistance requires a functional JmjC demethylase domain. Moreover, caffeine-induced Epe1-to-tEpe1 cleavage is dependent on an intact cell integrity MAP kinase stress signaling pathway, mutations in which alter resistance. Thus, environmental changes elicit a mechanism that curtails the function of this key epigenetic modifier, allowing heterochromatin to reprogram gene expression, thereby bestowing resistance to some cells within a population. H3K9me-heterochromatin components are conserved in human and crop-plant fungal pathogens for which a limited number of antifungals exist. Our findings reveal how transient heterochromatin-dependent antifungal resistant epimutations develop and thus inform on how they might be countered.

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: Epe1 migrates faster after caffeine stress and requires protein synthesis for recovery.
Fig. 2: Epe1 altered mobility results from cleavage and release of the N-terminal region.
Fig. 3: The Epe1 N-terminal region is sufficient for caffeine-induced cleavage that is dependent on proteasome function.
Fig. 4: N-terminally-truncated Epe1 and caffeine-processed Epe1 lose association with chromatin.
Fig. 5: Loss of Epe1 nuclear foci coincides with reduced heterochromatin association, increased H3K9 methylation and associated gene expression changes.
Fig. 6: Clr4-dependent H3K9 methylation mediates caffeine and antifungal resistance and is enhanced by Epe1 truncation.
Fig. 7: The cell integrity stress pathway regulates Epe1 processing through Pek1/MAPKK, Pmk1/MAPK and Pmp1/MAPK phosphatase.
Fig. 8: Model for induction of heterochromatin-mediated gene silencing and resulting resistance.

Similar content being viewed by others

Data availability

The publicly accessible PomBase database (https://www.pombase.org) was used to obtain genome sequence and annotation for the Schizosaccharomyces pombe (fission yeast) genome.

All raw and processed reads from sequencing experiments (ChIP–seq and RNA-Seq) are available at the GEO under accession number GSE190267.

All raw mass spectrometry data are available at PRIDE using project accession number PXD030205. Source data are provided with this paper.

References

  1. Fisher, M. C., Hawkins, N. J., Sanglard, D. & Gurr, S. J. Worldwide emergence of resistance to antifungal drugs challenges human health and food security. Science 360, 739–742 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Perlin, D. S., Rautemaa-Richardson, R. & Alastruey-Izquierdo, A. The global problem of antifungal resistance: prevalence, mechanisms, and management. Lancet Infect. Dis. 17, e383–e392 (2017).

    Article  PubMed  Google Scholar 

  3. Robbins, N., Caplan, T. & Cowen, L. E. Molecular evolution of antifungal drug resistance. Annu. Rev. Microbiol. 71, 753–775 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Stop neglecting fungi. Nat. Microbiol. 2, 17120 (2017).

  5. Godfray, H. C. J. et al. Food security: the challenge of feeding 9 billion people. Science 327, 812–818 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Fisher, M. C. et al. Threats posed by the fungal kingdom to humans, wildlife, and agriculture. mBio 11, e00449-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Audergon, P. N. C. B. et al. Restricted epigenetic inheritance of H3K9 methylation. Science 348, 132–135 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ragunathan, K., Jih, G. & Moazed, D. Epigenetic inheritance uncoupled from sequence-specific recruitment. Science 348, 1258699 (2015).

    Article  PubMed  CAS  Google Scholar 

  9. Cerulus, B., New, A. M., Pougach, K. & Verstrepen, K. J. Noise and epigenetic inheritance of single-cell division times influence population fitness. Curr. Biol. 26, 1138–1147 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hall, I. M. et al. Establishment and maintenance of a heterochromatin domain. Science 297, 2232–2237 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Calo, S. et al. Antifungal drug resistance evoked via RNAi-dependent epimutations. Nature 513, 555–558 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bheda, P. et al. Single-cell tracing dissects regulation of maintenance and inheritance of transcriptional reinduction memory. Mol. Cell 78, 915–925 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Torres-Garcia, S. et al. Epigenetic gene silencing by heterochromatin primes fungal resistance. Nature 585, 453–458 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Heard, E. & Martienssen, R. A. Transgenerational epigenetic inheritance: myths and mechanisms. Cell 157, 95–109 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Whittaker, C. & Dean, C. The FLC locus: a platform for discoveries in epigenetics and adaptation. Annu. Rev. Cell Dev. Biol. 33, 555–575 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Miska, E. A. & Ferguson-Smith, A. C. Transgenerational inheritance: models and mechanisms of non-DNA sequence-based inheritance. Science 354, 59–63 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Duempelmann, L., Skribbe, M. & Bühler, M. Small RNAs in the transgenerational inheritance of epigenetic information. Trends Genet. 36, 203–214 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Allshire, R. C., Nimmo, E. R., Ekwall, K., Javerzat, J. P. & Cranston, G. Mutations derepressing silent centromeric domains in fission yeast disrupt chromosome segregation. Genes Dev. 9, 218–233 (1995).

    Article  CAS  PubMed  Google Scholar 

  19. Nakayama, J., Rice, J. C., Strahl, B. D., Allis, C. D. & Grewal, S. I. S. Role of histone H3 lysine 9 methylation in epigenetic control of heterochromatin assembly. Science 292, 110–113 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Wang, J. et al. The proper connection between shelterin components is required for telomeric heterochromatin assembly. Genes Dev. 30, 827–839 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kanoh, J., Sadaie, M., Urano, T. & Ishikawa, F. Telomere binding protein Taz1 establishes Swi6 heterochromatin independently of RNAi at telomeres. Curr. Biol. 15, 1808–1819 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Zofall, M. et al. RNA elimination machinery targeting meiotic mRNAs promotes facultative heterochromatin formation. Science 335, 96–100 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Sugiyama, T. et al. Enhancer of rudimentary cooperates with conserved RNA-processing factors to promote meiotic mRNA decay and facultative heterochromatin assembly. Mol. Cell 61, 747–759 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yamanaka, S. et al. RNAi triggered by specialized machinery silences developmental genes and retrotransposons. Nature 493, 557–560 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Zofall, M., Smith, D. R., Mizuguchi, T., Dhakshnamoorthy, J. & Grewal, S. I. S. Taz1-shelterin promotes facultative heterochromatin assembly at chromosome-internal sites containing late replication origins. Mol. Cell 62, 862–874 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gallagher, P. S. et al. Iron homeostasis regulates facultative heterochromatin assembly in adaptive genome control. Nat. Struct. Mol. Biol. 25, 372–383 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, J., Reddy, B. D. & Jia, S. Rapid epigenetic adaptation to uncontrolled heterochromatin spreading. eLife 2015, e06179 (2015).

    Article  CAS  Google Scholar 

  28. Trewick, S. C., Minc, E., Antonelli, R., Urano, T. & Allshire, R. C. The JmjC domain protein Epe1 prevents unregulated assembly and disassembly of heterochromatin. EMBO J. 26, 4670–4682 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Roguev, A. et al. Conservation and rewiring of functional modules revealed by an epistasis map in fission yeast. Science 322, 405–410 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang, J., Lawry, S. T., Cohen, A. L. & Jia, S. Chromosome boundary elements and regulation of heterochromatin spreading. Cell. Mol. Life Sci. 71, 4841–4852 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Antequera, F., Tamame, M., Villanueva, J. R. & Santos, T. DNA methylation in the fungi. J. Biol. Chem. 259, 8033–8036 (1984).

    Article  CAS  PubMed  Google Scholar 

  32. Capuano, F., Mülleder, M., Kok, R., Blom, H. J. & Ralser, M. Cytosine DNA methylation is found in Drosophila melanogaster but absent in saccharomyces cerevisiae, schizosaccharomyces pombe, and other yeast species. Anal. Chem. 86, 3697–3702 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rape, M. & Jentsch, S. Productive RUPture: activation of transcription factors by proteasomal processing. Biochim. Biophys. Acta 1695, 209–213 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Tian, L. & Matouschek, A. Where to start and when to stop. Nat. Struct. Mol. Biol. 13, 668–670 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Li, F. et al. Lid2 is required for coordinating H3K4 and H3K9 methylation of heterochromatin and euchromatin. Cell 135, 272–283 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Thodberg, M. et al. Comprehensive profiling of the fission yeast transcription start site activity during stress and media response. Nucleic Acids Res. 47, 1671–1691 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Bohn, S. et al. Structure of the 26S proteasome from Schizosaccharomyces pombe at subnanometer resolution. Proc. Natl Acad. Sci. USA 107, 20992–20997 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gordon, C., McGurk, G., Dillon, P., Rosen, C. & Hastie, N. D. Defective mitosis due to a mutation in the gene for a fission yeast 26S protease subunit. Nature 366, 355–357 (1993).

    Article  CAS  PubMed  Google Scholar 

  39. Manasanch, E. E. & Orlowski, R. Z. Proteasome inhibitors in cancer therapy. Nat. Rev. Clin. Oncol. 14, 417–433 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zofall, M. & Grewal, S. I. S. Swi6/HP1 Recruits a JmjC domain protein to facilitate transcription of heterochromatic repeats. Mol. Cell 22, 681–692 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Braun, S. et al. The Cul4-Ddb1Cdt2 ubiquitin ligase inhibits invasion of a boundary-associated antisilencing factor into heterochromatin. Cell 144, 41–54 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Nicholas Laribee, R. & Weisman, R. Nuclear functions of TOR: impact on transcription and the epigenome. Genes 11, 1–24 (2020).

    Google Scholar 

  43. Weisman, R. Target of rapamycin (TOR) regulates growth in response to nutritional signals. Microbiol. Spectr. https://doi.org/10.1128/microbiolspec.FUNK-0006-2016 (2016).

  44. Cohen, A. et al. TOR complex 2 in fission yeast is required for chromatin-mediated gene silencing and assembly of heterochromatic domains at subtelomeres. J. Biol. Chem. 293, 8138–8150 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Oya, E. et al. Leo1 is essential for the dynamic regulation of heterochromatin and gene expression during cellular quiescence. Epigenetics Chromatin 12, 45 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Papadakis, M. A. & Workman, C. T. Oxidative stress response pathways: fission yeast as archetype. Crit. Rev. Microbiol. 41, 520–535 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Perez, P. & Cansado, J. Cell integrity signaling and response to stress in fission yeast. Curr. Protein Pept. Sci. 11, 680–692 (2011).

    Article  Google Scholar 

  48. Pérez, P., Cortés, J. C. G., Cansado, J. & Ribas, J. C. Fission yeast cell wall biosynthesis and cell integrity signalling. Cell Surf. 4, 1–9 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Heusch, M., Lin, L., Geleziunas, R. & Greene, W. C. The generation of nfkb2 p52: mechanism and efficiency. Oncogene 18, 6201–6208 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Palombella, V. J., Rando, O. J., Goldberg, A. L. & Maniatis, T. The ubiquitinproteasome pathway is required for processing the NF-κB1 precursor protein and the activation of NF-κB. Cell 78, 773–785 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Rape, M. & Jentsch, S. Taking a bite: proteasomal protein processing. Nat. Cell Biol. 4, E113–E116 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Jiang, J. & Struhl, G. Regulation of the Hedgehog and Wingless signalling pathways by the F- box/WD40-repeat protein Slimb. Nature 391, 493–496 (1998).

    Article  CAS  PubMed  Google Scholar 

  53. Pan, Y., Bai, C. B., Joyner, A. L. & Wang, B. Sonic hedgehog signaling regulates gli2 transcriptional activity by suppressing its processing and degradation. Mol. Cell. Biol. 26, 3365–3377 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sato, K., Ito, H., Yokoyama, A., Toba, G. & Yamamoto, D. Partial proteasomal degradation of Lola triggers the male-to-female switch of a dimorphic courtship circuit. Nat. Commun. 10, 166 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tian, L., Holmgren, R. A. & Matouschek, A. A conserved processing mechanism regulates the activity of transcription factors Cubitus interruptus and NF-κB. Nat. Struct. Mol. Biol. 12, 1045–1053 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Sha, Z. & Goldberg, A. L. Proteasome-mediated processing of Nrf1 is essential for coordinate induction of all proteasome subunits and p97. Curr. Biol. 24, 1573–1583 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wilson, M. D. et al. Proteasome-mediated processing of Def1, a critical step in the cellular response to transcription stress. Cell 154, 983–995 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hoppe, T. et al. Activation of a membrane-bound transcription factor by regulated ubiquitin/proteasome-dependent processing. Cell 102, 577–586 (2000).

    Article  CAS  PubMed  Google Scholar 

  59. Kravtsova-Ivantsiv, Y., Cohen, S. & Ciechanover, A. Modification by single ubiquitin moieties rather than polyubiquitination is sufficient for proteasomal processing of the p105 NF-κB precursor. Mol. Cell 33, 496–504 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Piwko, W. & Jentsch, S. Proteasome-mediated protein processing by bidirectional degradation initiated from an internal site. Nat. Struct. Mol. Biol. 13, 691–697 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Seo, H. D. et al. The 19S proteasome is directly involved in the regulation of heterochromatin spreading in fission yeast. J. Biol. Chem. 292, 17144–17155 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Rousseau, A. & Bertolotti, A. An evolutionarily conserved pathway controls proteasome homeostasis. Nature 536, 184–189 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Liu, Q. et al. Dysfunction of prohibitin 2 results in reduced susceptibility to multiple antifungal drugs via activation of the oxidative stress-responsive transcription factor Pap1 in fission yeast. Antimicrob. Agents Chemother. 62, e00860-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Fang, Y. et al. A genomewide screen in Schizosaccharomyces pombe for genes affecting the sensitivity of antifungal drugs that target ergosterol biosynthesis. Antimicrob. Agents Chemother. 56, 1949–1959 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Calvo, I. A., García, P., Ayté, J. & Hidalgo, E. The transcription factors Pap1 and Prr1 collaborate to activate antioxidant, but not drug tolerance, genes in response to H2O2. Nucleic Acids Res. 40, 4816–4824 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Dhar, R., Missarova, A. M., Lehner, B. & Carey, L. B. Single cell functional genomics reveals the importance of mitochondria in cell-to-cell phenotypic variation. eLife 8, e38904 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Kaelin, W. G. & McKnight, S. L. Influence of metabolism on epigenetics and disease. Cell 153, 56–69 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Dumesic, P. A. et al. Product binding enforces the genomic specificity of a yeast Polycomb repressive complex. Cell 160, 204–218 (2015).

    Article  CAS  PubMed  Google Scholar 

  69. Palmer, J. M., Perrin, R. M., Dagenais, T. R. T. & Keller, N. P. H3K9 methylation regulates growth and development in Aspergillus fumigatus. Eukaryot. Cell 7, 2052–2060 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Möller, M. et al. Destabilization of chromosome structure by histone H3 lysine 27 methylation. PLoS Genet. 15, e1008093 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Elías-Villalobos, A., Barrales, R. R. & Ibeas, J. I. Chromatin modification factors in plant pathogenic fungi: insights from Ustilago maydis. Fungal Genet. Biol. 129, 52–64 (2019).

    Article  PubMed  CAS  Google Scholar 

  72. Torres-Garcia, S. et al. SpEDIT: a fast and efficient CRISPR/Cas9 method for fission yeast. Wellcome Open Res. 5, 274 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Sato, M., Dhut, S. & Toda, T. New drug-resistant cassettes for gene disruption and epitope tagging in Schizosaccharomyces pombe. Yeast 22, 583–591 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Wilson, M. D., Saponaro, M., Leidl, M. A. & Svejstrup, J. Q. MultiDsk: a ubiquitin-specific affinity resin. PLoS ONE 7, e46398 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Singh, P. P. et al. Hap2-Ino80-facilitated transcription promotes de novo establishment of CENP-A chromatin. Genes Dev. 34, 226–238 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

    Article  CAS  PubMed  Google Scholar 

  77. Olsen, J. V. et al. Higher-energy C-trap dissociation for peptide modification analysis. Nat. Methods 4, 709–712 (2007).

    Article  CAS  PubMed  Google Scholar 

  78. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

  79. Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).

    Article  CAS  PubMed  Google Scholar 

  80. Cox, J. et al. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell. Proteom. 13, 2513–2526 (2014).

    Article  CAS  Google Scholar 

  81. Schwanhüusser, B. et al. Global quantification of mammalian gene expression control. Nature 473, 337–342 (2011).

    Article  CAS  Google Scholar 

  82. Shukla, M. et al. Centromere DNA destabilizes H3 nucleosomes to promote CENP-A deposition during the cell cycle. Curr. Biol. 28, 3924–3936 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Tong, P. et al. Interspecies conservation of organisation and function between nonhomologous regional centromeres. Nat. Commun. 10, 2343 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Nerusheva, O. O., Galander, S., Fernius, J., Kelly, D. & Marston, A. L. Tension-dependent removal of pericentromeric shugoshin is an indicator of sister chromosome biorientation. Genes Dev. 28, 1291–1309 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Kelly (WCB, Edinburgh) for microscopy and instrumentation support; members of the Allshire Lab for valuable discussions and input; T. Urano for the 5.1.1 (H3K9me2) antibody; A. Fellas for GFP expressing and clr4∆ strains; K. Gull for α-tubulin antibody; K. Sawin for Mto2 antibody; A. L. Marston for the Sgo1-GFP S. cerevisiae strain; J. Svejstrup for provision of the MultiDsk2 expression construct; M. D. Wilson for comments on the manuscript; and colleagues at WCB for support and encouragement during a difficult 2020–21. This research was supported by award of an EMBO Long Term Fellowship to I. Y. (EMBO ALTF 130–2018), Darwin Trust of Edinburgh PhD studentships to S. T.-G. and R. Y., a Wellcome 4 Year iCM program PhD studentship to E. G. (218470), a Wellcome Instrument grant to J. R. (108504), a Wellcome Investigator award to M. E. K. (205008), a Wellcome Principal Research Fellowship to R. C. A. (200885; 224358) and core funding for the Wellcome Centre for Cell Biology (203149). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. For the purpose of Open Access, the author has applied a CC-BY public copyright licence to any author-accepted manuscript version arising from this submission.

Author information

Authors and Affiliations

Authors

Contributions

I. Y. and S. A. W. designed and performed experiments. S. T.-G. contributed to ChIP, ChIP–seq and RNA-seq experiments and CRISPR design. M. L. carried out ChIP–seq and RNA-seq analyses and data visualization. E. G. performed Halo-tag experiments with advice on microscopy and experimental design from M.E.K. R. Y. contributed to antifungal-resistance experiments. C. S. ran samples and performed mass spectrometry analysis with J. R. A. L. P. advised on experimental design, performed ChIP and constructed strains. I. Y., S. A. W., A. L. P. and R. C. A. prepared figures and wrote the manuscript.

Corresponding author

Correspondence to Robin C. Allshire.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural and Molecular Biology thanks to Gary Karpen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Carolina Perdigoto, in collaboration with the Nature Structural & Molecular Biology team.

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 Caffeine-induced alteration in Epe1-GFP mobility does not result from transcriptional changes in 5’ region of epe1+ gene or Epe1-Myc phosphorylation.

a. Epe1-GFP or α-tubulin western from cells untreated (-)/treated (+) with 14 mM caffeine for 16 h. b. qRT-PCR measurement of steady-state Epe1 transcript levels in cells untreated (-)/treated (+) with 14 mM caffeine for 16 h. Locations of primers used indicated. Data shown represent 3 technical replicates + /− SEM. c. Scheme to detect caffeine-induced Epe1-GFP phosphorylation by mass spectrometry (top). Representation of phosphorylated residues detected on Epe1-GFP from cells untreated or treated with 14 mM caffeine for 16 h (middle). Spectra showing detection of phosphorylated serine S791, S795 and S803 residues after caffeine treatment (bottom). Full data, Supplementary Table 4. d. Epe1-Myc and Mto2 westerns from cells treated with 14 mM caffeine for 16 h before (-)/after (+) lambda protein phosphatase addition. Mto2 mobility is known to be Lambda Phosphatase sensitive1.

Source data

Extended Data Fig. 2 Deletions beyond residue 150 in Epe1-GFP do not hinder caffeine-induced processing and peptides within the first 100 residues are not detected following caffeine treatment.

a. Schematic of indicated 20 residue deletion mutants in the N-terminal coding region of the endogenous epe1 gene expressed as GFP fusions (Epe1∆150–170-GFP, Epe1∆171–190-GFP, Epe1∆191–210-GFP; left). Western detecting indicated mutant Epe1-GFP fusion proteins or α-tubulin from cells untreated (-) or treated (+) with 14 mM caffeine; right). b. Epe1-Myc peptides detected following immunoprecipitation from cells untreated (-) or treated (+) with 14 mM caffeine and analysis by mass spectrometry. Top: schematic showing position of peptides detected relative to Epe1 (residues 1–948). Bottom: Epe1 peptides detected in Epe1-Myc immunoprecipitates from treated (+) or untreated (-) with 14 mM caffeine. Of the eighteen peptides detected from Epe1-Myc in untreated (-) samples three were not detected (n.d.; red) in the caffeine-treated sample. The three peptides not detected in the presence of caffeine are derived from within the first 100 residues on the N-terminus. Analysis was performed on three independent immunoprecipitates. Full data, Supplementary Table 5.

Source data

Extended Data Fig. 3 A caffeine-induced N-terminal processing product is not detectable but Epe1-associated ubiquitination increases and Epe1 processing is sensitive to loss of specific E3 ligases which influence resistance.

a. Anti-Myc western of cells untreated (-)/treated (+) with 14 mM caffeine showing that a predicted N-terminal 20 kDa Myc-Epe11–150 processing product is not detectable. b. Western showing detection of both isoforms of the histone acetyltransferase Mst2-Myc or α-tubulin from wild-type or mst2–1 proteasome defective cells untreated (-)/treated (+) with 14 mM caffeine for 16 h. This control demonstrates that other known stress-induced changes still occur in mst2–1 cells. c. Western showing detection of both isoforms of the histone acetyltransferase Mst2-Myc or α-tubulin from wild-type cells incubated without (-)/with (+) with the 26 S proteasome inhibitor Bortezomib (BTZ) prior to no caffeine (-)/14 mM caffeine (+) treatment for 16 h. This control demonstrates that other known stress-induced changes still occur in the presence of BTZ. d. Immunolocalization of Epe1-GFP in wild-type or mts2–1 cells untreated (-)/ treated(+) with 14 mM/16 h caffeine. No tag, negative control. e. Quantification of anti-GFP/Epe1-GFP nuclear signals of cells in d. Data are represented as individual measurements from ≥ 20 cells per sample. Bars represent mean, with error bars + /− SD. The significance of the difference between samples was evaluated using a two-tailed Student’s t-test. (*) P < 0.033, (**) P < 0.002; (***) P < 0.0002; (****) P < 0.0001; (n.s). not significant. f. Western detecting Epe1-GFP or α-tubulin in wild-type or ddb1Δ, cells untreated (-)/treated (+) with 14 mM caffeine for 16 h. Numbers below tracks: levels of full-length FL-Epe1 and truncated tEpe1 normalised to no treatment and adjusted relative to α-tubulin loading control, measurement average of 2 biological replicates. g. Number of resistant colonies formed/1×104 viable cells by wild-type or ddb1Δ cells plated on caffeine or fluconazole plates. Error bars are from independent measurements from 3 biological replicates. Data are presented mean values +/− SD, with dots representing independent plate counts. The significance of the difference between samples was evaluated using a two-tailed Student’s t-test. (*) P < 0.033, (**) P < 0.002; (***) P < 0.0002; (****) P < 0.0001; (n.s). not significant.

Source data

Extended Data Fig. 4 Chromatin-associated nuclear proteins are enriched with full length Epe1 whereas more cytoplasmic proteins are enriched with constitutively truncated Epe1ΔN150-GFP.

a. Volcano plot of proteins enriched with full-length Epe1-GFP extracted from cells detected by proteomic analysis (top), specific proteins labelled (red dots). Table with names and functions of proteins enriched (bottom). Full data, Supplementary Table 6. b. Volcano plot of proteins enriched with constitutively processed Epe1ΔN150-GFP extracted from cells and detected by proteomic analysis (top), specific proteins labelled (red dots). Table with names and functions of proteins enriched (bottom). Full data, Supplementary Table 7.

Extended Data Fig. 5 Altered silencing and gene expression in epe1∆ and Epe1-∆N150-GFP cells.

a. Wild-type, epe1∆, Epe1-∆N150, Epe1-GFP/wt and Epe1-∆N150-GFP cells harbouring ura4+ marker gene insertions at either site 1 or site 2 as indicated (top) were serially diluted and plated on non-selective (N/S), selective (-URA) or counter-selective (FOA) plates. b. Heat-map generated from processed data of RNA-seq two biological replicates from Epe1∆N150-GFP or epe1Δ cells compared with that of wild-type Epe1-GFP cells. Genes with higher (turquoise) or lower (brown) expression relative to wild-type Epe1-GFP cells are shown as a Log2 scale. The position of specific affected genes along S. pombe chromosomes Chr I, II, III are shown with arrowheads indicating telomeres on Chr I and ChrII. The annotated S. pombe genome shows only rDNA arrays (rectangles) at both ends of Chr III. c. Heat-map showing that RNA-seq data (used in b.) resulting from two Epe1∆N150-GFP and epe1Δ biological replicates are more similar to each other than they are to wild-type Epe1-GFP cells.

Extended Data Fig. 6 Cells expressing predicted catalytically inactive Epe1-GFP and Epe1-∆N150-GFP exhibit decreased resistance.

a. Schematic showing position of H297A and K314A mutations predicted to reduce association of the essential iron/Fe(2) and 2-Oxoglutarate (α-ketoglutarate) cofactors, respectively, with the JmjC demethylase domain of Epe1 (Top). Westerns detecting Epe1-GFP, Epe1-cd-GFP, Epe1∆N150-GFP and Epe1∆N150-cd-GFP (cd; catalytically dead) or α-tubulin (bottom). b. Number of resistant colonies formed/1×104 viable cells by Epe1-GFP, Epe1-cd-GFP, Epe1∆N150-GFP and Epe1∆N150-cd-GFP cells plated on caffeine or fluconazole plates Error bars are from independent measurements from 3 biological replicates. Data are presented mean values+/−SD, with dots representing independent plate counts. The significance of the difference between samples was evaluated using a two-tailed Student’s t-test. (*) P < 0.033, (**) P < 0.002; (***) P < 0.0002; (****) P < 0.0001; (n.s). not significant.

Source data

Extended Data Fig. 7 The cell integrity/Pmk1 but not the TORC2/Gad8 stress signalling pathway regulates Epe1 processing and resistance.

a. Diagram of part the S. pombe TORC2/Tor1-dependent signalling pathway. b. Western detecting Epe1-GFP or α-tubulin from wild-type or gad8Δ cells untreated (-)/ treated (+) with 14 mM/16 h caffeine. c. Western detecting Epe1-Myc or α-tubulin from wild-type, pek1Δ, pmk1Δ, atf1Δ or pmp1Δ cells untreated (-)/treated (+) with 0.5 mM/16 h fluconazole (FLC). d. Number of resistant colonies formed/1×104 viable cells plated by wild-type, epe1Δ, sty1Δ, atf1Δ and gad8Δ on plates containing 16 mM caffeine (CAF). e. Number of resistant colonies formed/1×104 viable cells plated by wild-type, epe1Δ, sty1Δ, atf1Δ and gad8Δ on plates containing 0.5 mM fluconazole (FLC). Plots d and e; Error bars are from independent measurements from 3 biological replicates. Data are presented mean values + /− SD, with dots representing independent plate counts. The significance of the difference between samples was evaluated using a two-tailed Student’s t-test. (*) P < 0.033, (**) P < 0.002; (***) P < 0.0002; (****) P < 0.0001; (n.s). not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Figures 1–7, Supplementary Methods, Source Data for Extended Figures

Reporting Summary

Supplementary Tables

Supplementary Tables 1–7

Source data

Source Data Fig. 1

Uncropped blots

Source Data Fig. 1

Statistical source data

Source Data Fig. 2

Uncropped blots

Source Data Fig. 3

Uncropped blots and original micrographs

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Uncropped blots and original micrographs

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Fig. 6

Uncropped blots

Source Data Fig. 6

Statistical source data

Source Data Fig. 7

Uncropped blots

Source Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 1

Uncropped blots

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 2

Uncropped blots

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 3

Uncropped blots

Source Data Extended Data Fig. 6

Uncropped blots

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 7

Uncropped blots

Source Data Fig. 7

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yaseen, I., White, S.A., Torres-Garcia, S. et al. Proteasome-dependent truncation of the negative heterochromatin regulator Epe1 mediates antifungal resistance. Nat Struct Mol Biol 29, 745–758 (2022). https://doi.org/10.1038/s41594-022-00801-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00801-y

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing