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:

Glutathione-mediated redox regulation in Cryptococcus neoformans impacts virulence

Abstract

The fungal pathogen Cryptococcus neoformans is well adapted to its host environment. It has several defence mechanisms to evade oxidative and nitrosative agents released by phagocytic host cells during infection. Among them, melanin production is linked to both fungal virulence and defence against harmful free radicals that facilitate host innate immunity. How C.neoformans manipulates its redox environment to facilitate melanin formation and virulence is unclear. Here we show that the antioxidant glutathione is inextricably linked to redox-active processes that facilitate melanin and titan cell production, as well as survival in macrophages and virulence in a murine model of cryptococcosis. Comparative metabolomics revealed that disruption of glutathione biosynthesis leads to accumulation of reducing and acidic compounds in the extracellular environment of mutant cells. Overall, these findings highlight the importance of redox homeostasis and metabolic compensation in pathogen adaptation to the host environment and suggest new avenues for antifungal drug development.

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: Loss of GSH2 impairs melanin and titan cell formation and attenuates virulence.
Fig. 2: Gsh2 is required for growth upon nutrient depletion and WT cells secrete extracellular GSH.
Fig. 3: Deletion of GSH2 reduces susceptibility to H2O2 stress and alters non-GSH antioxidant functions.
Fig. 4: Dysregulation of GSH biosynthesis affects cellular metabolism.
Fig. 5: Extracellular acidity and reducing pontential inhibits melanin formation.
Fig. 6: GSH modulates redox homeostasis to influence melanin production.

Similar content being viewed by others

Data availability

The authors declare that all data supporting the findings of this study are available within the main text and Supplementary Information. The complete dataset used for LC-HRMS/MS analysis is publicly available on figshare (https://doi.org/10.6084/m9.figshare.24011169)32 and raw experimental data with their associated metadata are available on MetaboLights (MTBLS9984, https://www.ebi.ac.uk/metabolights/MTBLS9984). Source data are provided with this paper.

Code availability

All analyses were performed using publicly available software as described in the Methods. No custom code was generated for data analyses in this study.

References

  1. Nathan, C. & Shiloh, M. U. Reactive oxygen and nitrogen intermediates in the relationship between mammalian hosts and microbial pathogens. Proc. Natl Acad. Sci. USA 97, 8841–8848 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Nosanchuk, J. D. & Casadevall, A. The contribution of melanin to microbial pathogenesis. Cell. Microbiol. 5, 203–223 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Missall, T. A., Moran, J. M., Corbett, J. A. & Lodge, J. K. Distinct stress responses of two functional laccases in Cryptococcus neoformans are revealed in the absence of the thiol-specific antioxidant Tsa1. Eukaryot. Cell 4, 202–208 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zaragoza, O. Basic principles of the virulence of Cryptococcus. Virulence 10, 490–501 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ma, H. & May, R. C. in Advances in Applied Microbiology Vol. 67 (eds Laskin, A. I. et al.) Ch. 5, 131–190 (Elsevier, 2009).

  6. Maziarz, E. K. & Perfect, J. R. Cryptococcosis. Infect. Dis. Clin. North Am. 30, 179–206 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Casadevall, A., Rosas, A. L. & Nosanchuk, J. D. Melanin and virulence in Cryptococcus neoformans. Curr. Opin. Microbiol. 3, 354–358 (2000).

    Article  CAS  PubMed  Google Scholar 

  8. Tran-Ly, A. N., Reyes, C., Schwarze, F. W. M. R. & Ribera, J. Microbial production of melanin and its various applications. World J. Microbiol. Biotechnol. 36, 170 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cordero, R. J. B. & Casadevall, A. Functions of fungal melanin beyond virulence. Fungal Biol. Rev. 31, 99–112 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Cordero, R. J. B., Camacho, E. & Casadevall, A. Melanization in Cryptococcus neoformans requires complex regulation. mBio 11, e03313-19 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Slominski, R. M. et al. Melanoma, melanin, and melanogenesis: the Yin and Yang relationship. Front. Oncol. 12, 842496 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Fisher, M. C. & Denning, D. W. The WHO fungal priority pathogens list as a game-changer. Nat. Rev. Microbiol. 21, 211–212 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. WHO Fungal Priority Pathogens List to Guide Research, Development and Public Health Action (World Health Organization, 2022); https://www.who.int/publications/i/item/9789240060241

  14. Hider, R. C. & Kong, X. L. Glutathione: a key component of the cytoplasmic labile iron pool. BioMetals 24, 1179–1187 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Walker, E. A., Port, G. C., Caparon, M. G. & Janowiak, B. E. Glutathione synthesis contributes to virulence of streptococcus agalactiae in a murine model of sepsis. J. Bacteriol. 201, e00367-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Gutiérrez-Escobedo, G., Orta-Zavalza, E., Castaño, I. & De Las Peñas, A. Role of glutathione in the oxidative stress response in the fungal pathogen Candida glabrata. Curr. Genet. 59, 91–106 (2013).

    Article  PubMed  Google Scholar 

  17. Missall, T. A., Cherry-Harris, J. F. & Lodge, J. K. Two glutathione peroxidases in the fungal pathogen Cryptococcus neoformans are expressed in the presence of specific substrates. Microbiology 151, 2573–2581 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Missall, T. A. et al. Posttranslational, translational, and transcriptional responses to nitric oxide stress in Cryptococcus neoformans: implications for virulence. Eukaryot. Cell 5, 518–529 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Attarian, R. et al. The monothiol glutaredoxin Grx4 regulates iron homeostasis and virulence in Cryptococcus neoformans. mBio 9, e02377-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Reniere, M. L. et al. Glutathione activates virulence gene expression of an intracellular pathogen. Nature 517, 170–173 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Grant, C. M., MacIver, F. H. & Dawes, I. W. Glutathione synthetase is dispensable for growth under both normal and oxidative stress conditions in the yeast Saccharomyces cerevisiae due to an accumulation of the dipeptide γ-glutamylcysteine. Mol. Biol. Cell 8, 1699–1707 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lu, S. C. Glutathione synthesis. Biochim. Biophys. Acta 1830, 3143–3153 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Villarama, C. D. & Maibach, H. I. Glutathione as a depigmenting agent: an overview. Int. J. Cosmet. Sci. 27, 147–153 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Galván, I. & Alonso-Alvarez, C. An intracellular antioxidant determines the expression of a melanin-based signal in a bird. PLoS One 3, e3335 (2008).

  25. Benathan, M. & Labidi, F. Cysteine-dependent 5-S-cysteinyldopa formation and its regulation by glutathione in normal epidermal melanocytes. Arch. Dermatol. Res. 288, 697–702 (1996).

    Article  CAS  PubMed  Google Scholar 

  26. Jara, J. R., Aroca, P., Solano, F., Martinez, J. H. & Lozano, J. A. The role of sulfhydryl compounds in mammalian melanogenesis: the effect of cysteine and glutathione upon tyrosinase and the intermediates of the pathway. Biochim. Biophys. Acta 967, 296–303 (1988).

    Article  CAS  PubMed  Google Scholar 

  27. Spencer, J. P. E. et al. Conjugates of catecholamines with cysteine and GSH in Parkinson’s disease: possible mechanisms of formation involving reactive oxygen species. J. Neurochem. 71, 2112–2122 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Ku, J. W. & Gan, Y. H. Modulation of bacterial virulence and fitness by host glutathione. Curr. Opin. Microbiol. 47, 8–13 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Laman Trip, D. S. & Youk, H. Yeasts collectively extend the limits of habitable temperatures by secreting glutathione. Nat. Microbiol. 5, 943–954 (2020).

    Article  PubMed  Google Scholar 

  30. Worthington, D. J. & Rosemeyer, M. A. Glutathione reductase from human erythrocytes: catalytic properties and aggregation. Eur. J. Biochem. 67, 231–238 (1976).

    Article  CAS  PubMed  Google Scholar 

  31. Eisenman, H. C., Chow, S. K., Tsé, K. K., McClelland, E. E. & Casadevall, A. The effect of L-DOPA on Cryptococcus neoformans growth and gene expression. Virulence 2, 329–336 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Black, B. et al. LC-HRMS/MS-based untargeted metabolomic profiling of Cryptococcus neoformans mutants lacking the gene encoding glutathione synthetase, GSH2. figshare https://doi.org/10.6084/m9.figshare.24011169 (2024).

  33. Turner, E., Hager, L. J. & Shapiro, B. M. Ovothiol replaces glutathione peroxidase as a hydrogen peroxide scavenger in sea urchin eggs. Science 242, 939–941 (1988).

    Article  CAS  PubMed  Google Scholar 

  34. Narainsamy, K. et al. Oxidative-stress detoxification and signalling in cyanobacteria: the crucial glutathione synthesis pathway supports the production of ergothioneine and ophthalmate. Mol. Microbiol. 100, 15–24 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Baker, R. P. & Casadevall, A. Reciprocal modulation of ammonia and melanin production has implications for cryptococcal virulence. Nat. Commun. 14, 849 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Waterman, S. R. et al. Cell wall targeting of laccase of Cryptococcus neoformans during infection of mice. Infect. Immun. 75, 714–722 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Quintana-Cabrera, R. et al. γ-Glutamylcysteine detoxifies reactive oxygen species by acting as glutathione peroxidase-1 cofactor. Nat. Commun. 3, 718 (2012).

  38. Schafer, F. Q. & Buettner, G. R. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic. Biol. Med. 30, 1191–1212 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Reuwsaat, J. C. V. et al. The transcription factor Pdr802 regulates titan cell formation and pathogenicity of Cryptococcus neoformans. mBio 12, e03457-20 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Casadevall, A. Immunity to invasive fungal diseases. Annu. Rev. Immunol. 40, 121–141 (2022).

    Article  CAS  PubMed  Google Scholar 

  41. García-Barbazán, I. et al. The formation of titan cells in Cryptococcus neoformans depends on the mouse strain and correlates with induction of Th2-type responses. Cell Microbiol. 18, 111–124 (2016).

    Article  PubMed  Google Scholar 

  42. Ku, J. W. K. & Gan, Y. H. New roles for glutathione: modulators of bacterial virulence and pathogenesis. Redox Biol. 44, 102012 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Piacenza, L., Trujillo, M. & Radi, R. Reactive species and pathogen antioxidant networks during phagocytosis. J. Exp. Med. 216, 501–516 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chang, J. et al. Health-promoting phytochemicals and antioxidant capacity in different organs from six varieties of Chinese kale. Sci. Rep. 9, 20344 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Xu, Q. N. et al. Phenolic glycosides and flavonoids with antioxidant and anticancer activities from Desmodium caudatum. Nat. Prod. Res. 35, 4534–4541 (2021).

    Article  CAS  PubMed  Google Scholar 

  46. Johnson, J. B. et al. Hitting the sweet spot: a systematic review of the bioactivity and health benefits of phenolic glycosides from medicinally used plants. Phyther. Res. 35, 3484–3508 (2021).

    Article  CAS  Google Scholar 

  47. Shi, Z.-Z. et al. Mutations in the glutathione synthetase gene cause 5–oxoprolinuria. Nat. Genet. 14, 361–365 (1996).

    Article  CAS  PubMed  Google Scholar 

  48. Desideri, E., Ciccarone, F. & Ciriolo, M. R. Targeting glutathione metabolism: partner in crime in anticancer therapy. Nutrients 11, 1926 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ristoff, E. & Larsson, A. Inborn errors in the metabolism of glutathione. Orphanet J. Rare Dis. 2, 16 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Uchida, R., Ishikawa, S. & Tomoda, H. Inhibition of tyrosinase activity and melanine pigmentation by 2-hydroxytyrosol. Acta Pharm. Sin. B 4, 141–145 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Zhou, Y. Z., Alany, R. G., Chuang, V. & Wen, J. Studies of the rate constant of l-DOPA oxidation and decarboxylation by HPLC. Chromatographia 75, 597–606 (2012).

    Article  CAS  Google Scholar 

  52. Morris, G. et al. The glutathione system: a new drug target in neuroimmune disorders. Mol. Neurobiol. 50, 1059–1084 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Nielson, J. A., Jezewski, A. J., Wellington, M. & Davis, J. M. Survival in macrophages induces enhanced virulence in Cryptococcus. mSphere 9, e00504-23 (2024).

    Article  PubMed  Google Scholar 

  54. Gerstein, A. C. et al. Polyploid titan cells produce haploid and aneuploid progeny to promote stress adaptation. mBio 6, 01340-15 (2015).

    Article  Google Scholar 

  55. Toffaletti, D. L., Rude, T. H., Johnston, S. A., Durack, D. T. & Perfect, J. R. Gene transfer in Cryptococcus neoformans by use of biolistic delivery of DNA. J. Bacteriol. 175, 1405–1411 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Horianopoulos, L. C., Lee, C. W. J., Hu, G., Caza, M. & Kronstad, J. W. Dnj1 promotes virulence in Cryptococcus neoformans by maintaining robust endoplasmic reticulum homeostasis under temperature stress. Front. Microbiol. 12, 727039 (2021).

  57. Hommel, B. et al. Titan cells formation in Cryptococcus neoformans is finely tuned by environmental conditions and modulated by positive and negative genetic regulators. PLOS Pathog. 14, e1006982 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Won Hee, J. et al. Iron source preference and regulation of iron uptake in Cryptococcus neoformans. PLoS Pathog. 4, e45 (2008).

    Article  Google Scholar 

  59. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Re, R. et al. Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radic. Biol. Med. 26, 1231–1237 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Aldana, J., Romero-otero, A. & Cala, M. P. Exploring the lipidome: current lipid extraction techniques for mass spectrometry analysis. Metabolites 10, 231 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Matyash, V., Liebisch, G., Kurzchalia, T. V., Shevchenko, A. & Schwudke, D. Lipid extraction by methyl-tert-butyl ether for high-throughput lipidomics. J. Lipid Res. 49, 1137–1146 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Alcazar Magana, A. et al. Integration of mass spectral fingerprinting analysis with precursor ion (MS1) quantification for the characterisation of botanical extracts: application to extracts of Centella asiatica (L.) Urban. Phytochem. Anal. 31, 722–738 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Alcazar Magana, A. et al. Vitamin C activates the folate-mediated one-carbon cycle in C2C12 myoblasts. Antioxidants 9, 217 (2020).

    Article  PubMed Central  Google Scholar 

  65. Guijas, C. et al. METLIN: a technology platform for identifying knowns and unknowns. Anal. Chem. 90, 3156–3164 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Horai, H. et al. MassBank: a public repository for sharing mass spectral data for life sciences. J. Mass Spectrom. 45, 703–714 (2010).

    Article  CAS  PubMed  Google Scholar 

  67. Sumner, L. W. et al. Proposed minimum reporting standards for chemical analysis. Metabolomics 3, 211–221 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Viant, M. R., Kurland, I. J., Jones, M. R. & Dunn, W. B. How close are we to complete annotation of metabolomes? Curr. Opin. Chem. Biol. 36, 64–69 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Eberly, A. R. et al. Data highlighting phenotypic diversity of urine-associated Escherichia coli isolates. Data Brief 31, 105811 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  70. How does normalisation work in Progenesis QI? Nonlinear Dynamics https://www.nonlinear.com/progenesis/qi/v2.0/faq/how-normalisation-works.aspx (accessed 9 November 2022).

  71. Li, S. et al. Predicting network activity from high throughput metabolomics. PLoS Comput. Biol. 9, e1003123 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank T. Huan for useful discussions. Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health (NIH) under award number R01AI053721 (to J.W.K.). A.C. was supported in part by NIH grants AI052733, AI15207, AI171093-01 and HL059842. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Additional support came from a University of British Columbia (UBC) Four Year Doctoral Fellowship (to B.B.), and a Natural Sciences and Engineering Research Council of Canada (NSERC) Postgraduate Scholarship – Doctoral (to B.B.). L.C.H. is an NSERC postdoctoral fellow. J.W.K. is a Burroughs Wellcome Fund Scholar in Molecular Pathogenic Mycology, and the Power Corporation Fellow in the Canadian Institute for Advanced Research Program: Fungal Kingdom, Threats and Opportunities. Untargeted metabolomic analysis was performed in the Life Sciences Institute (LSI) Metabolomics Core Facility of the LSI at UBC, supported by the Canada Foundation for Innovation, the BC Knowledge Development Fund, the LSI, and the UBC GREx Biological Resilience Initiative. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.B. and J.W.K. conceptualized this project. B.B. developed methodology with guidance from J.W.K., A.C. and D.F.Q.S. and performed all growth experiments, flow cytometry, melanin/ABTS and colorimetric/fluorometric kit assays and data analysis. B.B. wrote the manuscript and J.W.K. provided edits. All authors participated in the review and editing of the manuscript. G.H., L.B.R.d.S., X.Q., B.B., L.C.H. and M.C. performed animal work and X.Q. imaged cells. Macrophage experiments were conducted by L.B.R.d.S., X.Q. and B.B., and L.B.R.d.S. performed all cytokine profiling experiments. G.H., L.C.H. and R.A. designed and constructed the mutant and complemented strains used in this study. A.A.M. conducted all LC-HRMS/MS experiments with guidance from L.J.F. and assisted B.B. with sample preparation and metabolomics data analysis. D.F.Q.S. performed all urease and cell wall laccase experiments and advised on ABTS antioxidant assays. J.W.K. and A.C. acquired funding for the project and L.J.F. funds the LSI Metabolomics Core Facility at UBC.

Corresponding author

Correspondence to James W. Kronstad.

Ethics declarations

Competing interests

The authors declare no competing interests

Peer review

Peer review information

Nature Microbiology thanks Jesus Pla, Jacob Walsh, Oscar Zaragoza and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Loss of GSH2 does not affect capsule size but delays virulence and impairs dissemination to the brain.

a, Polysaccharide capsule (left) and cell body diameter (right) of cells grown for 48 h. Measurements represent mean values ± s.d. of n = 50 cells from three independent experiments per strain. Significance was calculated relative to the WT using one-way ANOVA with Dunnett’s correction for multiple comparisons. b, Visualization of polysaccharide capsule with DIC microscopy. Images represent three independent experiments (scale bars = 10 μm). c, Spot assay of growth on solid YPD medium starting at 106 cells ml-1 with 10-fold serial dilutions. Plates were incubated at 30 ˚C, 37 ˚C, 39 ˚C. d, Melanin production of 106 cells on epinephrine (Epi, 0.1 g L-1), dopamine (Dm, 0.1 g L-1), and Niger seed (NS)-containing minimal media at 30 C. NS medium was prepared from 70 g 0.1 g L-1 seed extract supplemented with 0.1 g L-1 glucose, 20 g L-1 Bacto Agar, and 0.05% Tween 20. e, Melanin production of 106 cells per strain retrieved from murine lungs and spotted on solid l-DOPA medium. Agar plates in c, d, and e were grown for 48 h before imaging, and images are representative of three biological replicates. f, Time-course of fungal burden in mice intranasally infected with WT (red), gsh2∆ (light blue), and gsh2∆::GSH2 (dark blue) strains. Significance was calculated using two-way ANOVA with Dunnett’s correction for multiple comparisons. Solid bars indicate mean fungal burden (n = 8 mice per group) and segmented bars represent interquartile range. g, Visualization of fungal cells retrieved from murine lungs with DIC microscopy (top). Images represent fungal cells retrieved from 8 murine lungs per strain (n = 60 cells per sample) per timepoint (bars = 10 μm). Cell body diameter of gsh2∆ mutant cells retrieved from murine lungs (bottom). Lines represent mean values of n = 60 cells from 8 lungs per strain per time point.

Source data

Extended Data Fig. 2 Mutants lacking GSH2 have reduced lung proliferation and cell size in vivo.

a, Representative images of H & E stained lung tissue sections from four mice infected with WT, gsh2∆, or gsh2∆::GSH2 strains at each of the time points indicated (arrows = cryptococcal cells; bars = 10 μm). Segmented squares indicate enhanced zoom sections shown in the lower-right corner of each image. b, Cytokine levels in murine tissue homogenate of lungs infected with the WT (red) or gsh2∆ mutant (blue) strains at the indicated timepoints (n = 8 lungs for each strain at each time point). Uninfected mice (Naïve, gray, inoculated with PBS) were used as a control. Significance was calculated relative to treatment naïve mice using two-way ANOVA with either Tukey’s (7 & 14 dpi) or Šidák’s (21 & 26 dpi) correction for multiple comparisons. Solid bars indicate mean cytokine level and segmented bars represent interquartile range.

Source data

Extended Data Fig. 3 GSH is required for growth in minimal medium and is not restored with ascorbate or sulfur-containing amino acids.

a, Growth curve analysis of WT (light blue circles), gsh2∆ (blue squares or circles), and gsh2∆::GSH2 (light blue squares) strains grown in rich or minimal medium with and without methionine (Met), cysteine (Cys), or ascorbic acid (AA) supplementation at the indicated concentrations. b, Spot assays on minimal media. Each strain was spotted starting at 106 cells with 10-fold serial dilutions on agar minimal medium with or without Met, Cys, or AA supplementation at the indicated concentrations. Images are representative of three biological replicates. c, Growth of WT, gsh2∆, and gsh2∆::GSH2 strains in minimal medium with or without GSH supplementation at the indicated concentrations. Data points for a and c indicate mean OD600 values ± s.d., and the initial inoculum for each strain was 2 × 104 cells ml-1. Growth was monitored for 72 h with OD600 values measured every 24 h. d–e, Growth of WT, gsh2∆, and gsh2∆::GSH2 cells in 5 ml liquid cultures of l-DOPA medium (with or without GSH supplementation in e). Initial inoculum for each strain was 106 cells ml-1 and CFUs were counted every 24 h for 72 h (d) or at 48 h (e). Points (d) or bars (e) represent mean CFUs ml-1 ± s.d. at each time point or GSH concentration, respectively. f, ABTS antioxidant assay (see Methods) for the proportion of ABTS radical quenched by supernatant isolated from the indicated strains after 72 h incubation in l-asparagine minimal medium (lacking l-DOPA). Pigmentation (blue colouration) indicates presence of the ABTS radical. Images represent three independent experiments. Data are representative of three biological replicates for each experiment.

Source data

Extended Data Fig. 4 Metabolic profiling of the gsh2∆ mutant relative to WT reveals differences in extracellular and intracellular fractions.

a, Volcano plots showing QC-normalized LC-HRMS/MS feature data and differentially abundant metabolites between WT and gsh2∆ mutant cells in supernatant (left) and cell extract (right) fractions. The horizontal axis represents the directional intensity of the metabolite peak abundance fold change (FC) and the vertical axis represents statistical significance. A P-value threshold of P < 0.05 and FC threshold of > 1.5 or < 0.667 (segmented lines) were used to identify differences between the WT and mutant strains, which were determined using an unpaired, two-tailed Student’s t-test with Benjamini-Hochberg FDR correction for multiple comparisons in MetaboAnalyst. b, Heatmap comparing relative intensity of metabolite abundances in the supernatant (sup) and cell extract (ext) of WT and gsh2∆ mutants (KO); n = 3 biological replicates were analyzed for each fragment. Higher and lower intensity values are coloured red and blue, respectively. c, PCA score plot of the first two principal components from WT and gsh2∆ mutant supernatant and cellular extract fractions with QC samples for data normalization. Each data set represents n = 3 biological replicates. Purple = WT; red = gsh2∆ mutant; green = QC.

Source data

Extended Data Fig. 5 Mutants lacking GSH2 show distinct changes in relative abundance of key energy metabolites, antioxidants, and extracellular acids.

Relative abundance of select compounds identified via LC-HRMS/MS between WT (blue) and gsh2∆ mutant (red) strains. Both cell extract (ext) and supernatant (sup) fractions were analyzed for relative peak intensity. Bars represent mean relative abundances ± s.d. for n = 3 biological replicates per strain. Significance was calculated relative to the WT supernatant (sup) fraction using multiple unpaired, two-tailed Student’s t-tests with Benjamini-Hochberg FDR corrections for multiple comparisons.

Source data

Extended Data Fig. 6 Extracellular acidification of gsh2∆ mutants is independent of l-DOPA and GSH pathway metabolites influence melanin formation.

a, pH values of supernatant isolates from WT and gsh2∆ mutant cells grown in l-asparagine minimal medium and normalized to 107 cells ml-1. Statistical significance was calculated using an unpaired, two-tailed Student’s t-test. Bars represent mean pH values ± s.d. b–c, ABTS antioxidant assay indicates the reducing power of compounds tested at the indicated concentrations. Decreased pigmentation (light blue & clear in image) in c indicates increased ABTS radical scavenging activity. GSH = glutathione; AA = ascorbic acid; Cys = cysteine.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

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

Black, B., da Silva, L.B.R., Hu, G. et al. Glutathione-mediated redox regulation in Cryptococcus neoformans impacts virulence. Nat Microbiol (2024). https://doi.org/10.1038/s41564-024-01721-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41564-024-01721-x

Search

Quick links

Nature Briefing Microbiology

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

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