1887

Abstract

Cryptic links between apparently unrelated metabolic systems represent potential new drug targets in fungi. Evidence of such a link between zinc and gliotoxin (GT) biosynthesis in is emerging. Expression of some genes of the GT biosynthetic gene cluster is influenced by the zinc-dependent transcription activator ZafA, zinc may relieve GT-mediated fungal growth inhibition and, surprisingly, GT biosynthesis is influenced by zinc availability. In , dithiol gliotoxin (DTG), which has zinc-chelating properties, is converted to either GT or -dethiobis(methylthio)gliotoxin (BmGT) by oxidoreductase GliT and methyltransferase GtmA, respectively. A double deletion mutant lacking both GliT and GtmA was previously observed to be hypersensitive to exogenous GT exposure. Here we show that compared to wild-type exposure, exogenous GT and the zinc chelator ,,′,′-tetrakis(2-pyridinylmethyl)−1,2-ethanediamine (TPEN) inhibit ΔΔ growth, specifically under zinc-limiting conditions, which can be reversed by zinc addition. While GT biosynthesis is evident in zinc-depleted medium, addition of zinc (1 µM) suppressed GT and activated BmGT production. In addition, secretion of the unferrated siderophore, triacetylfusarinine C (TAFC), was evident by wild-type (at >5 µM zinc) and Δ (at >1 µM zinc) in a low-iron medium. TAFC secretion suggests that differential zinc-sensing between both strains may influence fungal Fe requirement. Label-free quantitative proteomic analysis of both strains under equivalent differential zinc conditions revealed protein abundance alterations in accordance with altered metabolomic observations, in addition to increased GliT abundance in Δ at 5 µM zinc, compared to wild-type, supporting a zinc-sensing deficiency in the mutant strain. The relative abundance of a range of oxidoreductase- and secondary metabolism-related enzymes was also evident in a zinc- and strain-dependent manner. Overall, we elaborate new linkages between zinc availability, natural product biosynthesis and oxidative stress homeostasis in .

Funding
This study was supported by the:
  • Science Foundation Ireland (Award Infrastructure Award 12/RI/2346(3))
    • Principle Award Recipient: SeanDoyle
  • National University of Ireland, Maynooth (Award John and Pat Hume Scholarship)
    • Principle Award Recipient: AimeeM. Traynor
  • Ministerio de Ciencia e Innovación (Award PID2019-110994RB-I00)
    • Principle Award Recipient: JoséA. Calera
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/micro/10.1099/mic.0.001106
2021-11-05
2024-04-26
Loading full text...

Full text loading...

/deliver/fulltext/micro/167/11/mic001106.html?itemId=/content/journal/micro/10.1099/mic.0.001106&mimeType=html&fmt=ahah

References

  1. Dagenais TRT, Keller NP. Pathogenesis of Aspergillus fumigatus in invasive aspergillosis. Clin Microbiol Rev 2009; 22:447–465 [View Article] [PubMed]
    [Google Scholar]
  2. Bernardo PH, Brasch N, Chai CLL, Waring P. A novel redox mechanism for the glutathione-dependent reversible uptake of a fungal toxin in cells. J Biol Chem 2003; 278:46549–46555 [View Article] [PubMed]
    [Google Scholar]
  3. Saleh AA, Jones GW, Tinley FC, Delaney SF, Alabbadi SH et al. Systems impact of zinc chelation by the epipolythiodioxopiperazine dithiol gliotoxin in Aspergillus fumigatus: a new direction in natural product functionality. 854 |. Met 2018; 10:854
    [Google Scholar]
  4. Traynor AM, Sheridan KJ, Jones GW, Calera JA, Doyle S. Involvement of sulfur in the biosynthesis of essential metabolites in pathogenic fungi of animals, particularly Aspergillus spp.: molecular and therapeutic implications. Front Microbiol 2019; 10:2859 [View Article] [PubMed]
    [Google Scholar]
  5. Oliva B, O’Neill A, Wilson JM, O’Hanlon PJ, Chopra I. Antimicrobial properties and mode of action of the pyrrothine holomycin. Antimicrob Agents Chemother 2001; 45:532–539 [View Article] [PubMed]
    [Google Scholar]
  6. Li B, Ry RF, Albert AB, Frank CS, Christopher TW. A backup plan for self-protection: S-Methylation of holomycin biosynthetic intermediates in Streptomyces clavuligerus. Chembiochem 2013; 13:2521–2526
    [Google Scholar]
  7. Chan AN, Shiver AL, Wever WJ, Razvi SZA, Traxler MF et al. Role for dithiolopyrrolones in disrupting bacterial metal homeostasis. Proc Natl Acad Sci U S A 2017; 114:2717–2722 [View Article] [PubMed]
    [Google Scholar]
  8. Vicentefranqueira R, Amich J, Marín L, Sánchez CI, Leal F et al. The transcription factor ZafA regulates the homeostatic and adaptive response to Zinc starvation in aspergillus fumigatus. Genes (Basel) 2018; 9:E318 [View Article] [PubMed]
    [Google Scholar]
  9. Scharf DH, Habel A, Heinekamp T, Brakhage AA, Hertweck C. Opposed effects of enzymatic gliotoxin N - And S -methylations. J Am Chem Soc 2014; 136:11674–11679 [View Article] [PubMed]
    [Google Scholar]
  10. Liu H, Xu W, Solis N, Woolford C, Mitchell AP et al. Functional convergence of glip and aspf1 in aspergillus fumigatus pathogenicity. Virulence 2018; 9:1062–1073 [View Article] [PubMed]
    [Google Scholar]
  11. Chen J, Lou Q, He L, Wen C, Lin M et al. Reduced-gliotoxin induces ROS-mediated anoikis in human colorectal cancer cells. Int J Oncol 2018; 52:1023–1032 [View Article] [PubMed]
    [Google Scholar]
  12. König S, Pace S, Pein H, Heinekamp T, Kramer J et al. Gliotoxin from Aspergillus fumigatus abrogates leukotriene B 4 formation through inhibition of leukotriene A 4 hydrolase. Cell Chem Biol 2019; 26:524–534 [View Article] [PubMed]
    [Google Scholar]
  13. Steiger MG, Patzschke A, Holz C, Lang C, Causon T et al. Impact of glutathione metabolism on zinc homeostasis in Saccharomyces cerevisiae. FEMS Yeast Res 2017; 17: [View Article] [PubMed]
    [Google Scholar]
  14. Marreiro D do N, Cruz KJC, Morais JBS, Beserra JB, Severo JS et al. Zinc and oxidative stress: Current mechanisms. Antioxidants 2017; 6:
    [Google Scholar]
  15. Paris S, Wysong D, Debeaupuis JP, Shibuya K, Philippe B et al. Catalases of Aspergillus fumigatus. Infect Immun 2003; 71:3551–3562 [View Article] [PubMed]
    [Google Scholar]
  16. Frealle E, Aliouat-Denis C-M, Delhaes L, Hot D, Dei-Cas E. Transcriptomic insights into the oxidative response of stress-exposed Aspergillus fumigatus. Curr Pharm Des 2013; 19:3713–3737 [View Article] [PubMed]
    [Google Scholar]
  17. Kurucz V, Krüger T, Antal K, Dietl AM, Haas H et al. Additional oxidative stress reroutes the global response of Aspergillus fumigatus to iron depletion. BMC Genomics 2018; 19:357 [View Article] [PubMed]
    [Google Scholar]
  18. Forest KT, Langford PR, Kroll JS, Getzoff ED. Cu,Zn superoxide dismutase structure from a microbial pathogen establishes a class with a conserved dimer interface. J Mol Biol 2000; 296:145–153 [View Article] [PubMed]
    [Google Scholar]
  19. Lambou K, Lamarre C, Beau R, Dufour N, Latge JP. Functional analysis of the superoxide dismutase family in Aspergillus fumigatus. Mol Microbiol 2010; 75:910–923 [View Article] [PubMed]
    [Google Scholar]
  20. Fréalle E, Noël C, Nolard N, Symoens F, Felipe MS et al. Manganese superoxide dismutase based phylogeny of pathogenic fungi. Mol Phylogenet Evol 2006; 41:28–39 [View Article] [PubMed]
    [Google Scholar]
  21. Owens RA, Hammel S, Sheridan KJ, Jones GW, Doyle S. A proteomic approach to investigating gene cluster expression and secondary metabolite functionality in Aspergillus fumigatus. PLoS One 2014; 9:106942 [View Article] [PubMed]
    [Google Scholar]
  22. Doyle S, Jones GW, Dolan SK. Dysregulated gliotoxin biosynthesis attenuates the production of unrelated biosynthetic gene cluster-encoded metabolites in Aspergillus fumigatus. Fungal Biol 2018; 122:214–221 [View Article] [PubMed]
    [Google Scholar]
  23. Dolan SK, Owens RA, O’Keeffe G, Hammel S, Fitzpatrick DA et al. Regulation of nonribosomal peptide synthesis: bis-thiomethylation attenuates gliotoxin biosynthesis in Aspergillus fumigatus. Chem Biol 2014; 21:999–1012 [View Article] [PubMed]
    [Google Scholar]
  24. Dolan SK, Bock T, Hering V, Owens RA, Jones GW et al. Structural, mechanistic and functional insight into gliotoxin bis-thiomethylation in Aspergillus fumigatus. Open Biol 2017; 7:160292 [View Article] [PubMed]
    [Google Scholar]
  25. Hein KZ, Takahashi H, Tsumori T, Yasui Y, Nanjoh Y et al. Disulphide-reduced psoriasin is a human apoptosis inducing broad-spectrum fungicide. Proc Natl Acad Sci U S A 2015; 112:13039–13044 [View Article] [PubMed]
    [Google Scholar]
  26. O’Keeffe G, Hammel S, Owens RA, Keane TM, Fitzpatrick DA et al. RNA-seq reveals the pan-transcriptomic impact of attenuating the gliotoxin self-protection mechanism in Aspergillus fumigatus. BMC Genomics 2014; 15:894 [View Article] [PubMed]
    [Google Scholar]
  27. Haas H. Molecular genetics of fungal siderophore biosynthesis and uptake: The role of siderophores in iron uptake and storage. Appl Microbiol Biotechnol 2003; 62:316–330 [View Article] [PubMed]
    [Google Scholar]
  28. Amich J, Vicentefranqueira R, Leal F, Calera JA. Aspergillus fumigatus Survival in Alkaline and Extreme Zinc-Limiting Environments Relies on the Induction of a Zinc Homeostasis System Encoded by the zrfC and aspf2 Genes. Eukaryot Cell 2010; 9:424–437 [View Article] [PubMed]
    [Google Scholar]
  29. Vodisch M, Scherlach K, Winkler R, Hertweck C, Braun H-P et al. Analysis of the Aspergillus fumigatus proteome reveals metabolic changes and the activation of the Pseurotin A biosynthesis gene cluster in response to hypoxia. J Proteome Res 2011; 10:2508–2524 [View Article] [PubMed]
    [Google Scholar]
  30. Lim FY, Won TH, Raffa N, Baccile JA, Wisecaver J et al. Fungal isocyanide synthases and xanthocillin biosynthesis in Aspergillus fumigatus. mBio 2018; 9:2020 [View Article] [PubMed]
    [Google Scholar]
  31. Schrettl M, Carberry S, Kavanagh K, Haas H, Jones GW et al. Self-protection against gliotoxin-a component of the gliotoxin biosynthetic cluster, gliT, completely protects Aspergillus fumigatus against exogenous gliotoxin. PLoS Pathog 2010; 6:e1000952 [View Article] [PubMed]
    [Google Scholar]
  32. Belkacemi L, Barton RC, Hopwood V, Evans EGV. Determination of optimum growth conditions for gliotoxin production by Aspergillus fumigatus and development of a novel method for gliotoxin detection. Med Mycol 1999; 37:227–233 [PubMed]
    [Google Scholar]
  33. Lulloff SJ, Hahn BL, Sohnle PG. Fungal susceptibility to zinc deprivation. J Lab Clin Med 2004; 144:208–214 [View Article] [PubMed]
    [Google Scholar]
  34. Laskaris P, Atrouni A, Calera JA, D’Enfert C, Munier-Lehmann H et al. Administration of zinc chelators improves survival of mice infected with Aspergillus fumigatus both in monotherapy and in combination with caspofungin. Antimicrob Agents Chemother 2016; 60:5631–5639 [View Article] [PubMed]
    [Google Scholar]
  35. O’Hanlon Cohrt KA, Marín L, Kjellerup L, Clausen JD, Dalby-Brown W et al. Novel zinc-attenuating compounds as potent broad-spectrum antifungal agents with in vitro and in vivo efficacy. Antimicrob Agents Chemother 2018; 62:
    [Google Scholar]
  36. Seo H, Kang S, Park YS, Yun CW. The role of zinc in gliotoxin biosynthesis of Aspergillus fumigatus. Int J Mol Sci 2019; 20:6192 [View Article]
    [Google Scholar]
  37. Bok JW, Chung DW, Balajee SA, Marr KA, Andes D et al. GliZ, a transcriptional regulator of gliotoxin biosynthesis, contributes to Aspergillus fumigatus virulence. Infect Immun 2006; 74:6761–6768 [View Article] [PubMed]
    [Google Scholar]
  38. Vicentefranqueira R, Leal F, Marín L, Sánchez CI, Calera JA. The interplay between zinc and iron homeostasis in Aspergillus fumigatus under zinc-replete conditions relies on the iron-mediated regulation of alternative transcription units of zafA and the basal amount of the ZafA zinc-responsiveness transcription fac. Environ Microbiol 2019; 21:2787–2808 [View Article] [PubMed]
    [Google Scholar]
  39. Danchin A. Zinc, an unexpected integrator of metabolism?. Microb Biotechnol 2020; 13:895–898 [View Article] [PubMed]
    [Google Scholar]
  40. Yasmin S, Abt B, Schrettl M, Moussa TAA, Werner ER et al. The interplay between iron and zinc metabolism in Aspergillus fumigatus. Fungal Genet Biol 2009; 46:707–713 [View Article] [PubMed]
    [Google Scholar]
  41. Banerjee B, Greenberger PA, Fink JN, Kurup VP. Immunological characterization of Asp f 2, a major allergen from Aspergillus fumigatus associated with allergic bronchopulmonary aspergillosis. Infect Immun 1998; 66:5175–5182 [View Article] [PubMed]
    [Google Scholar]
  42. Dasari P, Shopova IA, Stroe M, Wartenberg D, Martin-Dahse H et al. Aspf2 from Aspergillus fumigatus recruits human immune regulators for immune evasion and cell damage. Front Immunol 2018; 9:3 [View Article]
    [Google Scholar]
  43. Sugui JA, Kim HS, Zarember KA, Chang YC, Gallin JI et al. Genes differentially expressed in conidia and hyphae of Aspergillus fumigatus upon exposure to human neutrophils. PLoS One 2008; 3:e2655 [View Article] [PubMed]
    [Google Scholar]
  44. Gardiner DM, Howlett BJ. Bioinformatic and expression analysis of the putative gliotoxin biosynthetic gene cluster of Aspergillus fumigatus. FEMS Microbiol Lett 2005; 248:241–248 [View Article] [PubMed]
    [Google Scholar]
  45. Albrecht D, Guthke R, Brakhage AA, Kniemeyer O. Integrative analysis of the heat shock response in Aspergillus fumigatus. BMC Genomics 2010; 11:32 [View Article] [PubMed]
    [Google Scholar]
  46. Owens RA, O’keeffe G, Smith EB, Dolan SK, Hammel S et al. Interplay between gliotoxin resistance, secretion, and the methyl/ methionine cycle in Aspergillus fumigatus. Eukaryot Cell 2015; 14:941–957 [View Article] [PubMed]
    [Google Scholar]
  47. Scott J, Sueiro-Olivares M, Thornton B, Owens R, Muhamadali H et al. Targeting methionine synthase in a fungal pathogen causes a metabolic imbalance that impacts cell energetics, growth and virulence. mBio 2020; 11:
    [Google Scholar]
  48. Maiya S, Grundmann A, Li SM, Turner G. The fumitremorgin gene cluster of Aspergillus fumigatus: Identification of a gene encoding brevianamide F synthetase. ChemBioChem 2006; 7:1062–1069 [View Article] [PubMed]
    [Google Scholar]
  49. Wiemann P, Guo CJ, Palmer JM, Sekonyela R, Wang CCC et al. Prototype of an intertwined secondary metabolite supercluster. Proc Natl Acad Sci U S A 2013; 110:17065–17070 [View Article] [PubMed]
    [Google Scholar]
  50. Dolan SK, O’Keeffe G, Jones GW, Doyle S. Resistance is not futile: Gliotoxin biosynthesis, functionality and utility. Trends in Microbiology 2015; 23:419–428 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/micro/10.1099/mic.0.001106
Loading
/content/journal/micro/10.1099/mic.0.001106
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error