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. __________________________________________________________________ C. neoformans has emerged as a valuable fungal model for studying microbial virulence and host–pathogen interactions^[47]1–[48]4. Cryptococcal infections begin in the lungs and subsequently disseminate to other organs, including the brain, which can result in life-threatening meningoencephalitis^[49]4–[50]6. The fungus evades host immune mechanisms via several virulence factors which may have evolved in response to environmental predation^[51]5,[52]6. Notably, melanin is critical for the stress response of C. neoformans during infection as it can scavenge harmful free radicals and protect cells from oxidative bursts during phagocytosis^[53]3,[54]7. Furthermore, cryptococcal meningoencephalitis is associated with fungal-mediated conversion of catecholamines (for example, dopamine) from the central nervous system (CNS) into melanin^[55]2,[56]7–[57]11. Thus, mechanisms underlying the redox-mediated processes for melanin production are crucial for understanding virulence and defence strategies of pathogenic fungi, which are becoming more prevalent due to limited diagnostic and treatment capabilities and a growing at-risk population^[58]12,[59]13. Redox-active thiols, such as glutathione (GSH), are key for the oxidative stress response of fungi and constitute systems that depend on enzymatic and non-enzymatic mechanisms to evade host immunity and cause disease^[60]14–[61]16. Several GSH-dependent enzymes, including glutathione peroxidases (Gpx) and glutathione reductase (GR), are important for C. neoformans stress response pathways and virulence; others (for example, glutaredoxins) execute antioxidant functions that require GSH for oxidoreductase activity^[62]17–[63]19. GSH is also a signalling molecule for pathogens such as Listeria monocytogenes, which requires bacterial and host-derived GSH to activate the master virulence regulator PrfA^[64]14,[65]20. The importance of GSH is therefore not limited to its redox capabilities. This is clearly demonstrated by the inability of γ-Glu-Cys—a potent antioxidant and direct precursor of GSH—to compensate for non-antioxidant functions in Saccharomyces cerevisiae mutants lacking Gsh2, the terminal enzyme in the GSH biosynthetic pathway^[66]21. In fact, GSH is ubiquitous across all kingdoms of life and serves in a vast array of cellular processes; it is arguably the most important intracellular antioxidant for maintaining redox homeostasis^[67]15,[68]22. Perhaps unsurprisingly, GSH has also been linked to melanogenesis in animals—a process that relies heavily on the cellular redox environment^[69]23–[70]27. The involvement of GSH in the growth, survival and virulence of C. neoformans has not been investigated. In addition to maintaining redox homeostasis during infection, GSH might support the cellular conditions needed for fungal melanin production—a phenomenon that has not yet been examined in a human pathogen. We employed genetic and metabolomic approaches to demonstrate that GSH biosynthesis supports the proliferation of C. neoformans, and that mutants lacking GSH2 have attenuated virulence in a murine model of cryptococcosis. Loss of Gsh2 results in an enhanced reducing state that perturbs the specific ‘Goldilocks’ redox conditions required for melanogenesis. Furthermore, we demonstrate that an altered redox environment influences titan cell formation, urease production and interactions with macrophages. These results indicate a critical role for GSH biosynthesis in regulating metabolic and redox-dependent factors that contribute to cryptococcal pathogenesis. Results Mutants lacking GSH2 have attenuated virulence First, we examined the impact of GSH2 deletion (and thus the lack of GSH) on virulence-related phenotypes using two independent gsh2Δ deletion mutants and a gsh2Δ::GSH2 complemented strain, and found that gsh2Δ mutants did not show defects in capsule production or thermotolerance ([71]Extended Data Fig. 1a–[72]c). However, the gsh2Δ mutants were unable to melanize on catecholamine or phenolic substrates ([73]Fig. 1a,[74]b, [75]Extended Data Fig. 1d). Accordingly, we predicted that the gsh2Δ mutant would have impaired virulence in mice because melanin is linked to the dissemination and neurotropism of C. neoformans. We found that mice inoculated with the gsh2Δ mutant had reduced disease symptoms and prolonged survival compared to the wild-type (WT) and complemented strains ([76]Fig. 1c,[77]d). Mice infected with the mutant also had a lower fungal burden in the blood and affected organs at the experimental endpoint ([78]Fig. 1d). Interestingly, fungal burden was particularly diminished in the brain tissue of mutant-infected mice, which is a source of catecholamine precursors used for melanin production during infection^[79]7. Despite multiple attempts, we were unable to isolate melanized cells from tissue homogenates, but confirmed that mutants retrieved from lung homogenates could not melanize ex vivo ([80]Extended Data Fig. 1e). Fig. 1 |. Loss of GSH2 impairs melanin and titan cell formation and attenuates virulence. Fig. 1 | [81]Open in a new tab a, Melanin from cell suspensions incubated for 48 h in l-DOPA medium. Absorbance (OD[490]) was measured before (left) and after (right) digestion ([82]Methods) and normalized to 10^7 cells per ml. CWB, cell wall-bound. Bars represent the mean ± s.d. of n = 3 biological replicates. Significance relative to WT was calculated using one-way ANOVA with Dunnett’s correction for multiple comparisons. b, Melanin production of 10^6 cells on solid l-DOPA medium after 48 h incubation at 30 °C. The image is representative of three independent experiments. c,d, Survival (c) and fungal burden (d) at time of euthanasia in groups of 10 mice after intranasal infection with WT (red), gsh2Δ (blue) and gsh2Δ::GSH2 (blue segmented or dark blue) strains. Statistical analysis was performed using log-rank (Mantel–Cox) and Kruskal–Wallis (with Dunn’s correction for multiple comparisons) tests, respectively (n = 10 mice per group). e, Visualization of cells retrieved from murine lungs (left, in vivo) or cell culture (right, in vitro) with DIC microscopy. In vivo images represent cells from eight murine lungs per strain at each time point, and three independent experiments per strain (n = 50) for in vitro images (scale bars, 10 μm). f, Cell body diameters of cells from e (left, in vivo; right, in vitro). Lines represent mean values of n = 60 cells from eight lungs per strain per time point (in vivo) or n = 50 cells per strain from three independent experiments (in vitro). Significance relative to the WT for in vitro and in vivo measurements was calculated using one-way or two-way ANOVA tests, respectively, with Dunnett’s correction for multiple comparisons. The dashed line indicates the diameter threshold for cryptococcal cells defined as titan cells. g, Phagocytosis, PI and IPR of control or opsonized (+18B7) strains co-incubated with macrophages for 2–24 h. Bars represent mean ± s.d. of n = 6 or n = 12 independent experiments for phagocytosis, or PI and IPR, respectively. Significance of phagocytosis relative to WT was calculated using two-way ANOVA with Šidák’s correction for multiple comparisons. Significance of PI and IPR was calculated using unpaired, two-tailed Student’s t-tests. The onset and dissemination of fungal infection was also markedly delayed for gsh2Δ mutants, which may be partly attributed to an observed inability to form titan cells in vitro and in vivo—as titan cells are strongly linked to cryptococcal persistence and virulence ([83]Fig. 1e,[84]f and [85]Extended Data Fig. 1f,[86]g)^[87]4. Additionally, and compared with the WT, gsh2Δ mutants had reduced phagocytosis, increased intracellular proliferation (IPR) in bone-marrow-derived macrophages (BMDMs), and severely diminished progression to the brain, which was colonized in less than half of mutant-infected mice ([88]Fig. 1g and [89]Extended Data Fig. 1f). By contrast, fungal burden increased in the lungs, liver, spleen and kidneys of mice infected with gsh2Δ mutants 26 days post-infection, suggesting that gsh2Δ-infected mice likely succumbed to cryptococcal pneumonia and/or visceral organ damage rather than brain infection ([90]Extended Data Figs. 1f and [91]2a). This hypothesis is supported by increased lung pro-inflammatory cytokines (IFN-γ, TNF-α, IL-6) observed 26 days post-infection in mutant-infected mice, which were absent 14 days post-infection ([92]Extended Data Fig. 2b). Overall, GSH contributes to key aspects of cryptococcal disease including melanogenesis, titan cell formation, interactions with phagocytes and dissemination to the brain. GSH is critical for growth upon nutrient deprivation Next, we characterized the impact of GSH deficiency on growth upon nutrient depletion, which mimics the nutrient-sequestered host environment. We found that gsh2Δ mutants had markedly impaired growth in minimal medium but no growth defect in nutrient-rich medium ([93]Fig. 2a,[94]b). Exogenous GSH is also important for combating external stressors in bacterial pathogens, and the secretion and extracellular accumulation of GSH in S. cerevisiae is crucial for survival and replication under high temperature stress^[95]28,[96]29. We therefore investigated whether C. neoformans secreted GSH extracellularly and if loss of this extracellular GSH pool impacted gsh2Δ mutant function. First, we quantified extracellular total GSH (reduced plus oxidized GSH) for strains grown in minimal medium. We found that the WT and gsh2Δ::GSH2 strains secreted and accumulated GSH extracellularly ([97]Fig. 2c). By contrast, gsh2Δ mutants had no detectable extracellular GSH, consistent with the inability of these mutants to synthesize GSH endogenously ([98]Fig. 2c). Fig. 2 |. Gsh2 is required for growth upon nutrient depletion and WT cells secrete extracellular GSH. Fig. 2 | [99]Open in a new tab a, Growth curve analysis of WT, gsh2Δ and gsh2Δ::GSH2 strains grown in minimal medium (YNB with amino acids plus 2% glucose) with and without GSH supplementation. Data points indicate mean OD[600] values ± s.d. of n = 3 biological replicates at each time point. The initial inoculum for each strain was 2 × 10^4 cells ml^−1 in minimal medium and growth was monitored for 72 h, with OD[600] values measured every 24 h. b, Spot assays of growth on solid agar medium starting at 10^6 cells with 10-fold serial dilutions with or without GSH or GSSG supplementation at the indicated concentrations. Images are representative of three biological replicates. c, Quantification of extracellular total GSH (reduced and oxidized GSH) for the indicated strains normalized to OD[600]. Bars represent mean ± s.d. of n = 3 independent experiments. Significance was calculated relative to WT using one-way ANOVA with Dunnett’s correction for multiple comparisons. d, Schematic for the setup of growth curves in SM. e, Growth curve analysis of strains growth in SM. SM was isolated from WT, gsh2Δ and gsh2Δ::GSH2 log-phase cells grown in minimal medium for 16 h and was used to monitor growth of fresh cells for each of the strains. Points indicate mean OD[600] values ± s.d. of n = 3 biological replicates at each time point. Because WT C. neoformans secreted and accumulated GSH, we posited that transferring gsh2Δ mutants to WT-conditioned medium could help elucidate the function of extracellular GSH. Although mutants transferred from rich to minimal medium no longer grew ([100]Fig. 2a), transferring mutants into spent minimal medium (SM), conditioned only with WT cells, fully rescued gsh2Δ mutant growth ([101]Fig. 2d,[102]e). This outcome suggests that an extracellular factor (likely GSH) supports mutant growth in nutrient-limited conditions. Growth of gsh2Δ mutants was only partially restored in SM conditioned with the gsh2Δ::GSH2 strain ([103]Fig. 2e), which may be due to partial complementation and/or lower accumulation of GSH in this strain. By contrast, gsh2Δ mutants grown in SM conditioned with the gsh2Δ mutant did not recover, confirming that mutants are unable to secrete and accumulate extracellular GSH ([104]Fig. 2e). To eliminate the possibility that other components in the WT SM influenced mutant growth, we tested the impact of exogenous GSH on the gsh2Δ mutant. Indeed, mutant growth in minimal medium supplemented with GSH was restored to WT levels ([105]Fig. 2a,[106]b). Liquid-grown cultures were particularly sensitive to GSH supplementation and were fully rescued with concentrations as low as 50 μM GSH—a concentration similar to that found in WT supernatant ([107]Fig. 2c and [108]Extended Data Fig. 3a). Addition of the oxidized disulfide GSSG also rescued gsh2Δ mutant growth, though to a lesser extent than GSH ([109]Fig. 2a,[110]b). This effect may be attributed to the delay in regenerating GSH via reduction of GSSG by GR^[111]30, resulting in the initial lag in growth for mutants supplemented with GSSG. Given the overall positive effect of GSH on gsh2Δ mutant growth compared to other compounds tested (cysteine, methionine and ascorbic acid; [112]Fig. 2 and [113]Extended Data Fig. 3b,[114]c), we conclude that GSH is critical for the growth of C. neoformans and could impact survival and proliferation in vertebrate hosts. Loss of GSH2 enhances antioxidant defense S. cerevisiae gsh2Δ mutants accumulate γ-Glu-Cys^[115]21, an antioxidant and direct precursor of GSH, and we hypothesized that a similar accumulation of upstream GSH constituents would occur in C. neoformans gsh2Δ mutants. Disruption of GSH biosynthesis and loss of extracellular GSH could also impact the external redox environment and response to stress. Consistent with observations in S. cerevisiae, we found that mutants treated with H[2]O[2] had lower ROS accumulation than the WT and complemented strains^[116]21 ([117]Fig. 3a,[118]b). These observed differences led us to investigate changes in other antioxidant functions that could potentially compensate for the loss of GSH in mutants. Indeed, gsh2Δ mutants had enhanced activity of superoxide dismutase and catalase antioxidant enzymes, which are crucial for the fungal oxidative stress response ([119]Fig. 3c). We further speculated that loss of the extracellular GSH pool could affect extracellular redox homeostasis. Such changes were of particular interest because melanin assembly occurs at the cell wall and is highly dependent on redox-active processes that drive radical polymerization^[120]7,[121]10. Furthermore, accumulation of l-DOPA (l-3,4-dihydroxyphenylalanine) in the cell supernatant can cause toxicity and impaired growth^[122]31—a trend observed in gsh2Δ mutants grown in l-DOPA medium, which was rescued with ≥50 μM GSH supplementation ([123]Extended Data Fig. 3d,[124]e). We therefore examined the antioxidant/reducing power of the mutant supernatant using a radical-scavenging assay with 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS), which positively correlates reducing capacity with decolorization of a blue/green ABTS radical chromophore. The supernatant isolated from gsh2Δ mutants grown in l-DOPA medium fully quenched the ABTS radical suggesting a highly reductive extracellular environment consistent with the reduced accumulation of ROS in this strain ([125]Fig. 3d,[126]e). However, because l-DOPA is known to scavenge ABTS radicals, we also tested the scavenging potential of cells grown in minimal medium lacking l-DOPA. Both WT and mutant supernatant from cells grown in this medium were unable to quench the ABTS radical, suggesting that accumulated l-DOPA in the gsh2Δ mutant supernatant contributed to the ABTS radical scavenging effect ([127]Extended Data Fig. 3f). Because the gsh2Δ mutants had less ROS accumulation than the WT and higher IPR in BMDMs ([128]Figs. 1g and [129]3a,[130]b), we suspected that other mechanisms contributed to the observed changes in redox homeostasis. We postulated that loss of the extracellular GSH pool altered the composition of the mutant supernatant, likely due to accumulation and/or secretion of metabolites upstream of GSH—some of which (for example, γ-Glu-Cys and cysteine) could compensate for the antioxidant and/or reducing abilities of GSH. We did find modestly elevated thiol content in gsh2Δ mutant supernatant relative to the WT or complemented strains ([131]Fig. 3f), and thiols are known to interfere with oxidation of l-DOPA^[132]25–[133]27. Thus, GSH deficiency may induce dysregulation of the cellular redox environment and impair formation of virulence-related traits (for example, melanin and titan cell formation) of gsh2Δ mutants. Fig. 3 |. Deletion of GSH2 reduces susceptibility to H[2]O[2] stress and alters non-GSH antioxidant functions. Fig. 3 | [134]Open in a new tab a, 2D density plots of the indicated strains stained with 16 μM DCFDA (untreated, +H[2]O[2]) or without (control), and analysed via flow cytometry. Each measurement represents 30,000 gated single cells with or without 1 mM H[2]O[2] treatment 1 h prior to harvesting from minimal medium. Graphs represent results from three independent experiments. b, The percentage of DCFDA-stained (DCFDA^+) cells after H[2]O[2] treatment relative to untreated cells was measured for each strain from a. Statistical significance relative to untreated cells was calculated using a two-way ANOVA with Dunnett’s correction for multiple comparisons. Bars represent mean percentage of DCFDA^+ cells ± s.d. NS, not significant. c, Superoxide dismutase (Sod) and catalase (Cat) enzyme activity (U mg^−1 protein) of WT (red) and gsh2Δ mutant (blue) cell lysate normalized by protein concentration. Bars represent the mean ± s.d. and significance was calculated relative to WT using unpaired, two-tailed Student’s t-tests. d,e, ABTS antioxidant assay for the proportion of ABTS radical (ABTS^•, blue colouration in e) quenched by supernatant isolated from the strains indicated after 72 h incubation in l-DOPA medium. Measurements were taken every 24 h for 72 h and were quantified as a percentage of total quenched ABTS radical (d). Decreased pigmentation (light blue and clear in e) indicates radical scavenging activity. Data points in d represent the mean ± s.d. from three biological replicates for each strain, and images in e represent three independent experiments. f, Fluorescence quantification of supernatant thiol content after 48 h growth in l-DOPA medium for the indicated strains (OD[535]). Bars represent the mean thiol concentration ± s.d. per 10^7 cells per ml^−1 and significance was calculated relative to WT using a one-way ANOVA with Dunnett’s correction for multiple comparisons. Data are representative of three biological replicates for each experiment. Mutants lacking Gsh2 have a dysregulated metabolome To explain the metabolic changes incited by GSH2 deletion, including changes to regulatory machinery for redox control, we next compared the metabolomes of WT and gsh2Δ mutant cells grown under melanizing conditions, as melanin synthesis depends on oxidative processes to facilitate polymerization. In particular, we searched for changes in the relative abundance of reducing compounds that could explain the altered extracellular redox potential of gsh2Δ mutants. This included quantification of thiol and/or antioxidant compounds directly upstream of Gsh2 (including sulfur-containing compounds and other reductants) with reducing power. Analysis of WT and gsh2Δ mutant supernatants and cellular extracts via liquid chromatography–high-resolution tandem mass spectrometry (LC-HRMS/MS) untargeted metabolomics revealed peak areas of more than 1,500 distinct deconvoluted molecular features (positive ionization mode), of which 439 extracellular and 229 intracellular metabolites were notably different (fold change (FC) > 1.5 or < 0.667, P < 0.05) between strains ([135]Extended Data Fig. 4a). Multivariate heat map and principal component analysis (PCA) revealed a high degree of similarity between WT and mutant cell extracts ([136]Extended Data Fig. 4b,[137]c), but substantial differences in extracellular metabolites between the two strains ([138]Extended Data Fig. 4b,[139]c). Of the 439 differentially abundant metabolites in the supernatant, 294 had a more-than 1.5-fold increased abundance in the gsh2Δ mutant relative to the WT and 80 had more-than a 50-fold increase. However, features with a maximum FC ≥ 50 were interpreted with caution, as excessively high FC values could be attributed to abundances below the limit of quantification in the WT strain. To evaluate the biological relevance of these metabolic changes, we performed mummichog pathway enrichment analysis (based on KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway data) and identified notable differences in several key metabolic pathways including amino acid and secondary metabolite metabolism, as well as carbohydrate/energy metabolism ([140]Fig. 4a). We further annotated specific metabolites using a robust scoring technique ([141]Methods) and confidently identified more than 100 features that contributed to the observed pathway changes. We used these annotated metabolites for further analysis^[142]32, and found substantial extracellular accumulation of several aromatic amino acids and weak acids (for example, dicarboxylic acids, ketones/keto-acids and hydroxy fatty acids) in the gsh2Δ mutant and a depleted intracellular amino acid content ([143]Fig. 4b,[144]c). The mutant also had substantially depleted intracellular levels of key energy pathway intermediates including adenosine and adenosine monophosphate (AMP), l-carnitine and propionylcarnitine, pantothenate, α-ketoglutaric acid and β-nicotinamide adenine dinucleotide (NAD) and nicotinamide, as well as the urea cycle intermediates ornithine and argininosuccinic acid ([145]Supplementary Table 1). Consistent with the melanin defect and enhanced ABTS radical scavenging, gsh2Δ mutants had an increased relative abundance of the melanin precursor l-DOPA ([146]Extended Data Fig. 5), suggesting an inability to utilize l-DOPA for melanin production. Finally, the mutant accumulated substantial amounts of extracellular glucosinolates (1,4-dimethoxyglucobrassicin and indolylmethyl-desulfoglucosinolate), sulfur-containing compounds and cysteine derivatives, and other antioxidants that could account for some of the characterized redox changes ([147]Supplementary Table 1). For instance, the antioxidants caffeic acid, salvianolic acid and pyrogallol-2-O-glucuronide were more abundant in the gsh2Δ mutant than the WT ([148]Supplementary Table 1). Anti-melanogenic compounds were also found, including myo-inositol and hydroxytyrosol, the latter of which was highly abundant in the gsh2Δ mutant supernatant ([149]Extended Data Fig. 5). We also detected an approximately 10-fold increase of the cysteine derivative S-(5-histidyl)cysteine sulfoxide, a precursor to the potent antioxidant ovothiol which has Gpx-like activity but has not yet been described in C. neoformans ([150]Supplementary Table 1)^[151]33. Of note, the WT strain had a high intracellular concentration of the antioxidant ergothioneine (EGT), an integral redox buffer in several non-yeast fungi, cyanobacteria and certain Gram-positive bacteria including Mycobacterium tuberculosis ([152]Supplementary Table 1)^[153]34. Although EGT has not been described in C. neoformans, our findings further support the proposed interdependency of EGT synthesis on GSH and/or the enzymes involved in GSH biosynthesis^[154]34. Fig. 4 |. Dysregulation of GSH biosynthesis affects cellular metabolism. Fig. 4 | [155]Open in a new tab a, Peak list profile of enriched metabolic pathways using mummichog analysis (v.2) from MetaboAnalyst 5.0. Metabolic pathways are grouped by S. cerevisiae KEGG pathway module and class datasets. b, Classification of differentially abundant metabolites (P < 0.05, measured using unpaired, two-tailed Student’s t-tests with Benjamini–Hochberg FDR corrections for multiple comparisons) between gsh2Δ mutant (FC < 0.667) and WT (H99; FC > 1.5) strains using ClassyFire batch compound classification (v.2.0, [156]https://cfb.fiehnlab.ucdavis.edu/). Only metabolites with high confidence annotations (Progenesis QI score ≥40) from either positive or negative ionization modes were included in the classification analysis. c, Relative abundances of amino acids and derivatives identified via LC-HRMS/MS in both the cell extract (top) and supernatant (bottom) fractions. The horizontal axis represents directional intensity of the metabolite peak abundance FC, and the vertical axis represents statistical significance. Dashed lines indicate significance thresholds for −log[10] FDR-corrected P values (FDR < 0.05, horizontal) and log[2] FC (FC > 1.5 or < 0.667, vertical) were used to determine significance, which was calculated using an unpaired, two-tailed Student’s t-test with Benjamini–Hochberg FDR corrections for multiple comparisons in MetaboAnalyst (a, c). Blue dots indicate metabolites with higher abundance in gsh2Δ mutants relative to WT; red dots indicate metabolites with higher abundance in WT relative to gsh2Δ mutants. Acidification and metabolite changes impair melanization We further characterized melanin formation as a readily assayable phenotype for examining the impact of metabolite differences between WT and mutant cells. Disparities in the abundance of extracellular acids predicted pH differences, and we found that the supernatant of the gsh2Δ mutant culture was more acidic than that of the WT ([157]Fig. 5a). This was true of mutant cells grown in either l-DOPA or l-asparagine minimal media, which demonstrates that accumulated l-DOPA in the mutant supernatant was not responsible for acidification ([158]Extended Data Fig. 6a). We also investigated urease activity because melanin formation is influenced by extracellular pH via the urease-dependent production of ammonia^[159]35, and mutant growth in l-DOPA medium resulted in a low extracellular pH. Consistent with an acidic extracellular pH, gsh2Δ mutants had impaired urease activity ([160]Fig. 5b). Cell wall laccase activity was also diminished in mutants relative to the WT, suggesting that reduced expression and/or improper localization of the enzyme contributed to the melanin defect, potentially due to pH-dependent regulation of laccase activity^[161]36 ([162]Fig. 5c). Buffering the gsh2Δ culture medium with 3-(N-morpholino)propanesulfonic acid (MOPS) restored melanin production, validating an inhibitory role for extracellular acidification ([163]Fig. 5d,[164]e). Fig. 5 |. Extracellular acidity and reducing pontential inhibits melanin formation. Fig. 5 | [165]Open in a new tab a, pH values of supernatant isolates from cells grown in l-DOPA medium and normalized to 10^7 cells ml^−1. b, Spot assays of 10^6 cells ml^−1 on solid agar urea broth following incubation at 30 °C for 24 h (top). The diffusion radius of ammonia into the medium (pink colouration, measured in the bottom panel) indicates urease activity of cells. For a and b (bottom), statistical significance was calculated using unpaired, two-tailed Student’s t-tests and bars show mean ± s.d. c, Laccase activity of the cell wall fraction of cell lysate measured by conversion of the ABTS substrate, which can be detected at OD[734]. Bars represent mean OD[734] values ± s.d. of n = 3 biological replicates for each strain. d, Melanin formation of cells grown in liquid l-DOPA medium with or without 1 M MOPS pH-buffering. e, Extracellular (left) and CWB (right) melanin were quantified by measuring absorbance of cell supernatant and digests at OD[490] ([166]Methods) and normalizing by CFU to 10^7 cells ml^−1. Significance in e was calculated using two-way ANOVA with Tukey’s correction for multiple comparisons. Bars represent mean OD[490] values ± s.d. of n = 3 biological replicates. f,g, Melanin formation of cultures grown in liquid (f) or on solid agar (g) l-DOPA medium for 72 h with or without supplementation as indicated. f, Extracellular (top panel) and CWB (bottom) melanin of cells grown with or without 5 mM GSH, 2.5 mM GSSG, or 5 mM γ-Glu-Cys supplementation and quantified by absorbance of cell supernatant and digests at OD[490] ([167]Methods). Bars represent mean OD[490] values ± s.d. of n = 3 biological replicates for each strain and condition. Significance was calculated relative to the l-DOPA condition for each strain using two-way ANOVA with Dunnett’s correction for multiple comparisons. g, Spot assays of 10^6 cells plated on solid l-DOPA medium with or without supplementation (GSH (5 mM), GSSG (2.5 mM), ascorbic acid (AA, 10 mM), and 5 mM or 10 mM cysteine (+Cys or ++Cys, respectively)). Images representative of three independent experiments. Metabolomic analyses also predicted changes in antioxidants, which could inhibit redox-dependent radical polymerization for melanization^[168]7. Such changes corroborated the potent reducing power of the gsh2Δ mutant supernatant ([169]Fig. 3d). We tested other GSH pathway intermediates and ascorbic acid, a well-documented antioxidant, to determine whether the melanin defect of gsh2Δ mutants was specific to loss of GSH. GSH, cysteine and ascorbic acid had comparable radical scavenging activities to the gsh2Δ mutant supernatant ([170]Extended Data Fig. 6b,[171]c), and high concentrations of these compounds blocked melanogenesis of all strains including the WT and complement ([172]Fig. 5f,[173]g). We also found that the GSH pathway intermediate γ-Glu-Cys inhibited melanin production in all strains ([174]Fig. 5f), commensurate with the ability of this compound to compensate for antioxidant functions in gsh2Δ mutants of S. cerevisiae^[175]21. This intermediate also fully quenched the ABTS radical chromophore at high concentrations but was less potent than GSH ([176]Extended Data Fig. 6c). We note that γ-Glu-Cys was not detected via LC-HRMS/MS, perhaps due to chemical conjugation or use in alternate biochemical pathways. For instance, γ-Glu-Cys can directly interact with and neutralize ROS and serves as a cofactor for antioxidant enzymes^[177]37. By contrast, GSSG fully restored melanin production in gsh2Δ mutants and even enhanced melanin production of the WT and complemented strains ([178]Fig. 5f,[179]g). Because gsh2Δ mutants retain GR activity, we questioned whether trace amounts of GSH derived from GSSG were responsible for restoring melanin production in GSSG-treated mutants. We performed a GSH titration of mutant cells grown in l-DOPA medium, and found that low levels of GSH (50–500 μM) restored extracellular melanin in gsh2Δ mutants and further enhanced melanin in the WT ([180]Fig. 6a,[181]b). Notably, the GSH concentrations required to restore melanin production in gsh2Δ mutants were similar to the concentrations in the WT supernatant ([182]Fig. 2c). More intriguingly, extracellular melanin production was blocked in both the mutant and WT when treated with GSH concentrations of 750 μM or higher ([183]Fig. 6a,[184]b), suggesting a ‘tipping point’ for redox-dependent melanogenesis. GSH at all concentrations impaired melanin deposition at the cell wall in the WT and at high concentrations in the gsh2Δ mutant ([185]Fig. 6a). However, GSSG addition (which is oxidizing) and MOPS buffering restored gsh2Δ mutant cell wall-bound melanin—an effect not seen with low-concentration GSH treatment ([186]Figs. 5d–[187]g and [188]6a). Fig. 6 |. GSH modulates redox homeostasis to influence melanin production. Fig. 6 | [189]Open in a new tab a, Melanin formation of cells grown in liquid l-DOPA medium with or without GSH supplementation at the indicated concentrations. Extracellular (a, left) and CWB (a, right) melanin were quantified by measuring absorbance of cell supernatant and digests at OD[490] (see [190]Methods) and normalizing by CFU to 10^7 cells ml^−1. Significance was calculated relative to cells grown without GSH supplementation using one-way ANOVA with Dunnett’s correction for multiple comparisons. Bars represent mean OD[490] values ± s.d. from n = 3 biological replicates per strain for each condition. b, Images representing three independent experiments of extracellular melanin from WT and gsh2Δ mutant cells grown in l-DOPA medium with or without GSH supplementation. c, Schematic for the setup of melanization assays in l-DOPA SM. d, WT and gsh2Δ mutant cells melanized for 72 h on agar-supplemented with l-DOPA or l-DOPA SM isolated from log-phase cells grown in l-DOPA medium. Images from the upper panel show individual wells of a 48-well plate and the lower panel shows enhanced-zoom images of spots from the upper panel and represent three independent experiments. e, Extracellular and CWB melanin of WT, gsh2Δ and gsh2Δ::GSH2 cells grown for 48 h in SM isolated from log-phase cells grown in l-DOPA medium. Melanin content was quantified after normalizing by CFU to 10^7 cells ml^−1 ([191]Methods). Bars indicate mean OD[490] values ± s.d. of n = 3 biological replicates for each condition. Significance for each condition was calculated using two-way ANOVA with a two-stage setup method of Benjamini, Krieger and Yekutieli FDR correction for multiple comparisons. Since supernatant isolated from the WT had a higher pH, contained GSH at low concentrations, and restored gsh2Δ mutant growth, we reasoned that l-DOPA medium conditioned with WT cells could restore gsh2Δ melanin production. We tested this hypothesis and found that mutants transferred to WT SM had increased extracellular melanin and modestly elevated cell wall melanin ([192]Fig. 6d,[193]e). By contrast, WT cells transferred to gsh2Δ SM had less released melanin (P = 0.077) and less cell-wall-bound melanin ([194]Fig. 6d,[195]e). Overall, we conclude that changes to extracellular pH and antioxidant composition in gsh2Δ mutants lead to a redox environment that precludes melanin formation. Discussion GSH is a potent antioxidant and a key player in redox homeostasis for many organisms^[196]1,[197]38. For C. neoformans. a pathogen of global health importance^[198]12,[199]13, we found that manipulation of GSH metabolism by deletion of the GSH2 gene attenuated both proliferation in host tissue and dissemination to the brain in a murine model of cryptococcosis. The diminished virulence of gsh2Δ mutants, which are non-melanized and have reduced uptake by phagocytes, affirms the critical role for melanin in dissemination via survival in macrophages and traversal of the blood–brain barrier^[200]39. Melanin is well established as an antioxidant that protects cells against oxidative bursts in the phagosome of phagocytic cells and promotes dissemination to the brain^[201]3,[202]7. The gsh2Δ mutant showed enhanced proliferation in phagocytes, consistent with greater extracellular antioxidant activity observed in our analysis. This phenotype may not be sufficient to overcome impaired dissemination to the brain, but may support persistence in the lung that contributes to the delayed onset of disease. The gsh2Δ mutant also failed to produce titan cells in vitro and during infection, and this phenotype has a complex influence on dissemination^[203]40. A previous study found that higher proportions of titan cells in the mouse lung were associated with greater dissemination^[204]41. In contrast, Reuwsaat et al. found that a regulatory mutant that overproduced titan cells had poor proliferation and reduced dissemination^[205]39. Titan cells can also induce Th2 immunity, which is non-protective and skews adaptive immunity to favour cryptococcal dissemination to the CNS, thereby enhancing disease^[206]4,[207]40. We hypothesize that this contribution of titan cells is particularly relevant for the gsh2Δ mutant because we observed an increased abundance of the Th2 cytokine IL-4 in the lungs of WT-infected mice at the time of death, but not in mutant-infected mice. Conversely, the surge of Th1-associated cytokines (IFN-γ and TNF-α) in gsh2Δ-infected mice at the experimental endpoint suggests enhanced inflammation that may damage lungs and exacerbate disease^[208]42. These findings suggest that changes to redox homeostasis due to loss of GSH2 impacted virulence both through influencing interactions with phagocytic cells and host adaptive immunity. Consistent with our study, GSH influences virulence in several fungal and bacterial species and plays a critical role in mitigating the impact of host-derived ROS released during infection^[209]15,[210]16,[211]20,[212]28,[213]41. Our global metabolomic analysis revealed that loss of GSH dramatically altered the metabolome and resulted in dysregulation of the cellular redox environment—a key determinant of survival in the host^[214]43. For example, the mutant supernatant had elevated thiol content and substantial quantities of glucosinolates and phenolic glycosides with antioxidant activity^[215]44–[216]46. Of note, we found a twofold increase in pyroglutamic acid, a γ-glutamyl cycle intermediate formed from the GSH precursor γ-Glu-Cys. This finding is intriguing because glutathione synthetase deficiency, a heritable disorder in humans, also leads to hyperaccumulation of pyroglutamic acid resulting in high anion gap metabolic acidosis—a form of acidosis defined by acid accumulation and low pH of the blood^[217]47,[218]48. Consistent with this phenotype, our mutants accumulated and secreted high quantities of amino acids and other weak acids (for example, lactic acid, dicarboxylic acids and keto acids) leading to lower extracellular pH and urease activity compared to the WT. Mutant supernatant fractions also contained several amino acid derivatives with known antioxidant and anti-melanogenic properties including hydroxytyrosol^[219]49. We propose that acidification of mutant supernatant (along with accumulation of antioxidants) precludes the chemical oxidation of the catecholamine precursor l-DOPA needed for melanization^[220]2,[221]9, as this process is supported by an alkaline pH^[222]35,[223]50,[224]51 and was reversible by MOPS-buffering of gsh2Δ mutant media. Consistent with this mechanism, treatment with exogenous reducing compounds (for example, cysteine, GSH, ascorbic acid and γ-Glu-Cys) also inhibited melanization. In summary, we have shown that perturbation of the cellular redox environment via loss of GSH upsets the ‘Goldilocks’ conditions needed for melanin formation, as well as other virulence traits including titan cell formation, urease activity and dissemination to the brain. These findings are consistent with the roles of GSH across fungal and bacterial species^[225]20,[226]52 and provide a framework for understanding how microorganisms not only mitigate harmful radicals released by host phagocytic cells but also manipulate the extracellular metabolite environment to condition virulence. These findings collectively support GSH metabolism as a potential therapeutic target for cryptococcosis, and we note that glutathione metabolism has been targeted in human diseases and cancer^[227]53,[228]54 but not for antifungal drug discovery. Methods Ethics statement All animal experiments performed with mice in this study were conducted in accordance with the guidelines of the Canadian Council on Animal Care and approved by the University of British Columbia’s Committee on Animal Care (protocol A21–0105). Strains and growth media A detailed list of strains and oligonucleotides used in this study are provided in [229]Supplementary Tables 2 and [230]3, respectively. C. neoformans var. grubii (serotype A) strain H99 was used for generation of gsh2Δ deletion mutants and as the WT strain ([231]Supplementary Table 2). WT and mutant strains were maintained on yeast peptone dextrose medium (YPD, Difco; 1% yeast extract, 2% peptone, 2% dextrose, 2% agar). Cells for growth and phenotypic assays were grown overnight at 30 °C (with shaking at 220 r.p.m.) in liquid YPD and were then either harvested or transferred to experiment-specific media for further experimentation. Yeast nitrogen base minimal medium (YNB, Difco; with amino acids, supplemented with 2% glucose, pH 5.6), which lacks GSH, was used as minimal medium. Solid YPD medium containing 200 μg ml^−1 hygromycin was used for selection of gsh2Δ deletion mutants, and medium containing 100 μg ml^−1 nourseothricin was used to select for the gsh2Δ::GSH2 reconstituted strain in the gsh2Δ mutant background. Strain construction The gsh2Δ mutants were constructed with deletion cassette prepared via three-step overlapping PCR using primers listed in [232]Supplementary Table 3 with WT genomic DNA and the pJAF15 plasmid as templates. The resulting construct was biolistically transformed into the WT strain as described previously^[233]55, and positive transformants were selected on hygromycin (200 μg ml^−1) and confirmed via PCR and Southern blot analysis. The gsh2Δ::GSH2 complemented strain was generated by insertion of a gene encoding a C-terminal Gsh2-HA fusion protein at the native locus. GSH2-HA was constructed using three-step overlapping PCR with WT and Dnj1-HA genomic DNA, as described previously^[234]56. The resulting construct was biolistically transformed into the gsh2Δ mutant background and positive transformants were selected on nourseothricin (100 μg ml^−1). Primers used for the complement construct are listed in [235]Supplementary Table 3. Murine infection and virulence assays Virulence of WT, gsh2Δ mutant and gsh2Δ::GSH2 strains was tested in a murine inhalation model of cryptococcosis using 4–6 week-old female BALB/c mice (Mus musculus) from Charles River Laboratories. Mice were housed in a facility with 14 h light/10 h dark cycles, 40–60% humidity and an ambient temperature range of 21–23 °C. Feed (PicoLab Rodent Diet 20, LabDiet, 0007688) and water were provided ad libitum for the duration of the experiments. Briefly, fungal cells were grown in YPD at 30 °C overnight, washed twice with phosphate-buffered saline (PBS) and resuspended in PBS. For survival and endpoint fungal burden, groups of 10 BALB/c mice were intranasally inoculated with a suspension of 2 × 10^5 cells in 50 μl for each strain and mice reaching the humane endpoint were euthanized by CO[2] anoxia. For time-course measurements of fungal burden, groups of eight BALB/c mice were intranasally inoculated with a suspension of 2 × 10^5 cells in 30 μl for each strain and timepoint. Mice infected for 7, 14, 21 or 26 days were euthanized by CO[2] anoxia. Mouse organs were harvested by dissection at humane or pre-determined endpoints, weighed and homogenized in sterile PBS and plated on YPD agar containing chloramphenicol (100 μg ml^−1) to quantify colony-forming units (CFUs) for each organ. No statistical methods were used to pre-determine sample sizes, but sample sizes were similar to those reported in previous publications^[236]17–[237]19,[238]35. To enhance experimental rigour and minimize bias for data collection, mice from different litters were mixed and randomly assigned to one of three experimental groups (WT-infected, gsh2Δ-infected or gsh2Δ::GSH2-infected). Cages from each group were chosen at random for euthanasia at pre-determined experimental endpoints. The health status of mice for all experiments was monitored daily post-inoculation. Microscopy of C. neoformans and lung tissue sections For in vitro titan cell assays, cells were grown on Sabouraud dextrose agar (SDA; 4% glucose, 1% peptone, 1.5% agar) for 2–5 days at room temperature (~20–22 °C), as described previously^[239]57. After incubation, 10^7 cells were resuspended in a T25 flask with 10 ml YPD and cultured for 22 h at 30 °C with shaking at 150 r.p.m. until stationary phase (approximately 2 × 10^8 cells ml^−1). Cells were washed twice with minimal medium, adjusted to 10^6 cells ml^−1, and incubated at 30 °C for 48 h with shaking at 800 r.p.m. In vivo titan cell formation was evaluated using WT and gsh2Δ cells isolated from murine lung homogenate and washed twice with minimal medium. For polysaccharide capsule images, cells were incubated for 48 h in capsule-inducing medium (CIM) prepared as previously described^[240]19,[241]58. All cells were stained with India ink prior to imaging. Titan cell and polysaccharide capsule formation were evaluated by differential interference contrast (DIC) microscopy using a Zeiss Axiopolan 2 microscope equipped with a Plan-Apochromat 100×/1.46 objective lens and an ORCA-Flash4.0 LT CMOS camera (Hamamatsu Photonics). Zeiss Zen 2 Blue edition (v.2.3) and ImageJ^[242]59 (v.2.14.0) were used for collection and analysis of microscopy images, respectively. Cells with a body size >10 μm were considered titan cells, as described previously^[243]57. Histological samples were obtained from lung tissue of four infected mice for each strain and timepoint. Lung tissue was stored in 10% formalin and submitted to Wax-It Histology Services for processing. Resin-embedded tissues were sectioned, mounted and stained with hematoxylin and eosin prior to imaging. In vitro macrophage infections with C. neoformans BMDMs were isolated from BALB/c mice and differentiated at 37 °C with 5% CO[2] for 7 days in media consisting of DMEM, 10% FBS, 1% nonessential amino acids, 1% penicillin-streptomycin, 1% GlutaMAX (Gibco, 35050061), 1% HEPES buffer, 20% L-929 cell-conditioned supernatant and 0.1% 2-mercaptoethanol^[244]35. Differentiated cells were detached from culture dishes using a cell scraper (Falcon, 353085) and washed into DMEM. Cells were seeded at a density of 2 × 10^5 cells per well in 24-well plates and incubated at 37 °C in 5% CO[2] for 24 h in DMEM supplemented with 100 U ml^−1 of IFN-γ (Gibco, PMC4031) to activate. Overnight cultures of WT and gsh2Δ C. neoformans grown in YPD were washed twice with PBS, counted and resuspended in DMEM at a density of 2 × 10^7 cells ml^−1. Fungal cells were opsonized by incubation with 5 μg ml^−1 18B7 monoclonal antibody (mAb) for 1 h at room temperature. BMDMs were then infected with 2 × 10^6 cells ml^−1 of opsonized or non-opsonized (−18B7, control) C. neoformans and incubated for 2–24 h at 37 °C. After infection, BMDMs were washed twice with PBS to remove extracellular yeast cells and lysed with 1 ml sterile water. Serial dilutions of BMDM lysate were plated on YPD and grown for 2–3 days at 30 °C. Colonies were then counted to determine CFUs. Phagocytosis of control and opsonized cells was calculated as the number of BMDMs containing cryptococcal cells after 2 h incubation divided by the total number of BMDMs. Phagocytic index (PI) was determined by dividing the CFUs from lysed cells 2 h post-infection (hpi) by the total number of BMDMs. IPR was calculated by dividing the CFUs at 24 hpi by the CFUs at 2 hpi. Lung cytokine isolation and analysis Lung cytokines were quantified from the supernatant of thawed organ homogenates (stored at −80 °C) from each experimental timepoint using the BD Mouse Th1/Th2/Th17 Cytometric Bead Array (CBA) Kit (BD Biosciences, 560485). Briefly, supernatants were incubated for 2 h at room temperature in equal proportions with anti-cytokine mAb-coated beads and a cytokine phycoerythrin (PE) detection reagent. After incubation, samples were washed once and measured on a Northern Lights full spectrum flow cytometer (Cytek Biosciences) with SpectroFlo software (v.3.2.1). Data were analysed using FlowJo (BD Biosciences, v.10.10) with the CBA Plug-in (v.5.2.2). Serial dilution spot assays and growth curves In a typical growth experiment, single colonies of the WT, gsh2Δ mutants or gsh2Δ::GSH2 strains were selected from an agar plate and incubated at 30 °C in YPD medium overnight with shaking at 220 r.p.m. Cells were harvested and washed twice with sterile water. For spot assays, 10-fold serial dilutions were prepared starting at 10^6 cells and 5 μl of cell suspensions were spotted onto solid agar medium. Plates were then incubated at 30 °C, 37 °C or 39 °C for 2–4 days and imaged. For growth curves, overnight cultures were first counted to determine CFUs. Cells were then diluted in the appropriate medium and grown in either a 96-well plate or 5 ml liquid culture at 30 °C for 72 h with shaking at 220 r.p.m. For YPD and YNB medium growth curves, absorbance at an optical density of 600 nm (OD[600]) was measured every 24 h using a Tecan Infinite M200 PRO microplate reader with Tecan i-control (v.1.7.1.12). For growth in l-DOPA medium, cells were inoculated in chemically defined l-DOPA medium containing 0.1% l-asparagine, 0.1% dextrose, 3 mg ml^−1 KH[2]PO[4], 0.25 mg ml^−1 MgSO[4]·7H[2]O, 1 μg ml^−1 thiamine, 5 ng ml^−1 biotin and 0.2 mg ml^−1 l-DOPA (Sigma-Aldrich, D9628) and counted every 24 h to determine CFUs. Quantification of melanin production in liquid culture Cells from overnight cultures were harvested and counted to determine CFUs. Each strain was then diluted and transferred at 1 × 10^6 cells ml^−1 to l-DOPA medium with or without supplementation at the concentrations indicated. Cultures were grown for 48 h at 30 °C with shaking at 220 r.p.m., then harvested and counted to determine CFUs. Absorbance of the supernatant was measured at OD[490] using a Tecan Infinite M200 PRO microplate reader to determine extracellular melanin content. The remaining cell pellet was washed twice with water, diluted to 10^7 cells ml^−1, and digested in 100 μl of 1 M NaOH with 10% dimethyl sulfoxide (DMSO) for 1 h at 95 °C. Cell digests were centrifuged and the OD[490] was measured to determine cell wall melanin content. Extracellular melanin content was normalized to growth as determined by CFUs for each strain. Media transfer and quantification of extracellular GSH Following overnight incubation, WT, gsh2Δ mutant and gsh2Δ::GSH2 cells were washed twice with sterile water, resuspended and transferred into 25 ml YPD broth at an OD[600] of 0.3 for an 8 h growth period. Log-phase cells were then harvested, counted to determine CFUs and diluted in 5 ml of either YNB or l-DOPA medium at concentrations of 3 × 10^4 or 1 × 10^6 cells ml^−1, respectively. After 16 h of growth, cells were harvested via centrifugation and the resulting supernatant was harvested and sterilized using 0.22 μm syringe filter sterilizing units (VWR, 76479–016) to remove residual cells. For YNB medium transfer experiments, fresh overnight cells of each strain were then inoculated into the filtered spent medium (SM) and aliquoted into a 96-well plate at an OD[600] of 0.0001 and grown for 72 h at 30 °C with shaking at 220 r.p.m. and OD[600] measurements taken every 24 h. For liquid l-DOPA medium transfer experiments, fresh cells were aliquoted into 3 ml spent l-DOPA medium at an initial density of 1 × 10^6 cells ml^−1 and grown for 48 h (with shaking at 220 r.p.m.) and then harvested and counted to determine CFUs. Melanin content of liquid cultures was then quantified (as described above). In parallel, fresh cells of each strain were grown in YNB or l-DOPA media to compare cell growth in fresh versus filtered SM. For solid medium experiments, spent l-DOPA medium was supplemented with 2% sterile agar and dispensed into a 48-well plate. After the medium solidified, fresh cells were spotted onto solid SM at 1 × 10^6 cells ml^−1 and incubated for 48 h at 30 °C prior to imaging. Extracellular GSH was quantified by centrifugation and filter sterilization (VWR, 76479–016) of culture supernatants to remove cells. The concentration of GSH was measured using a glutathione assay kit according to the manufacturer’s instructions (Cayman Chemical, 703002). Absorbance at OD[415] was measured using a Tecan Infinite M200 PRO microplate reader and GSH concentration was determined by constructing a standard curve using GSH solutions provided by the manufacturer. Measurement of ROS content using flow cytometry Cells were grown for 24 h in liquid YPD medium, washed twice with water and the absorbance at OD[600] was measured. Resuspended cells were inoculated in fresh YNB (pH 5.6) at an OD[600] of 0.3 and grown for 16 h with shaking at 30 °C. Following a 16 h incubation period, cells were harvested via centrifugation, washed twice using sterile PBS and resuspended in PBS at a cell density of OD[600] = 1.0. Cells exposed to oxidative stress were treated with 1 mM H[2]O[2] for 1 h with shaking at 30 °C. After treatment with H[2]O[2], the treated cells were centrifuged, washed twice and resuspended in PBS. Cells were then treated with 16 μM of the ROS-sensitive fluorogenic probe 2′,7′-dichlorodihydrofluorescein diacetate (DCFDA, Sigma-Aldrich) for 1 h in the dark at 30 °C. Stained cells were then washed once with PBS and ROS levels were analysed using a CytoFLEX S flow cytometer (Beckman Coulter) with lasers at wavelengths of 405 nm (violet), 488 nm (blue), 561 nm (yellow) and 633 nm (red). Results were gated to single yeast cells and PBS was used as a blank control. Fluorescence of DCFDA was measured using the FITC-GFP channel, and data were acquired and analysed using the CytExpert cytometry analysis software (Beckman Coulter). ABTS free-radical scavenging assays Preparation of the radical ABTS solution was performed as previously described^[245]60. To examine the radical scavenging activity in WT and gsh2Δ mutant culture supernatants, cells were first grown at 1 × 10^6 cells ml^−1 in l-DOPA medium, centrifuged at 20,627g and 1 ml of the supernatant was collected and dried overnight at room temperature via rotary evaporation. Samples were then resuspended in 200 μl of nanopure water and 10 μl of each sample was transferred to a 96-well plate. For testing specific chemicals, 10 μl of each compound was serially diluted to the indicated concentrations. Then 190 μl of radical ABTS solution was added to each sample, as described previously^[246]60. Following a 5 min incubation in the dark, absorbance at OD[734] was measured using a Tecan Infinite M200 PRO microplate reader to quantify decolourization and values were normalized to 10^7 CFUs per strain. Thiol quantification and antioxidant enzyme activity Thiol concentration of culture supernatants was measured using a fluorometric thiol assay kit according to the manufacturer’s instructions (Sigma-Aldrich, MAK151). Fluorescence was measured using a BioTek Synergy H1 microplate reader (Agilent) with BioTek Gen5 software (v.3.15.15) at an excitation wavelength of 490 nm and an emission wavelength of 525 nm. Thiol concentration was determined by constructing a standard curve using serial dilutions of a GSH standard provided by the manufacturer and normalized by CFU. The activities of superoxide dismutase and catalase were measured using enzyme-specific colorimetric assay kits according to the manufacturer’s instructions (Cayman Chemical, 706002 and 707002, respectively). Absorbance was measured at 440 nm (superoxide dismutase) or 540 nm (catalase) using a Tecan Infinite M200 PRO microplate reader, and enzyme activity was normalized to protein concentration as determined by Pierce BCA protein quantification (Thermo Fisher Scientific, 23225) relative to a BSA standard. Measurement of urease and cell wall laccase activity Overnight cultures were harvested and washed twice with PBS at 4,000g for 4 min. For urease assays, cells were diluted 1:40 in PBS and 25 μl of diluted culture was spotted onto urease detection agar medium (2% urea, 1.5% agar, 0.2% monopotassium phosphate, 0.1% peptone, 0.1% dextrose, 0.5% sodium chloride, 0.0012% pH 6.8 phenol red). Plates were incubated for 24 h at 30 °C and imaged. Pink halos indicative of urease activity were measured using ImageJ software^[247]59. For cell wall laccase activity, washed cultures were added 1:100 in 100 ml of l-asparagine medium (0.1% l-asparagine, 0.1% dextrose, 3 mg ml^−1 KH[2]PO[4], 0.25 mg ml^−1 MgSO[4]·7H[2]O, 1 mg ml^−1 thiamine, 5 ng ml^−1 biotin) and grown for 48 h, shaking at 120 r.p.m. Following incubation, cultures were harvested and washed twice in PBS at 14,000g for 15 min. Cells were then resuspended in 10 ml of cold PBS and incubated with cOmplete mini EDTA-free protease inhibitor (Roche, 11836170001) for 30 min at 4 °C. Cells were kept on ice and lysed using a French pressure cell press (Glen Mills) with five passes at 25,000 psi. The resulting lysate was then centrifuged at 3,000g for 10 min and the supernatant was removed. The resulting cell wall pellet was washed twice in PBS at 3,000g for 10 min and resuspended in 10 ml PBS. Then, 180 μl aliquots of resuspended cell pellet were added to a 96-well plate in triplicate and 20 μl of 10 mM ABTS was added to each sample. Plates were incubated for 2 h at 30 °C with constant shaking. Absorbance at 734 nm was measured after 2 h using a SpectraMax iD5 spectrophotometer with SoftMax Pro software (v.7.1). Cell preparation for metabolome profiling Samples for metabolite analysis were incubated for 48 h in l-DOPA medium starting at 1 × 10^6 cells ml^−1. WT and gsh2Δ mutant cells were then harvested via centrifugation at 15,493g for 10 min at 4 °C and separated into supernatant and cell pellet fractions. Both fractions were kept on ice. Next, 2.5 volumes of 100% MeOH + 0.05% butylated hydroxytoluene (BHT; Cayman Chemical) were added to the supernatant fraction to limit oxidation and/or isomerization of reactive compounds^[248]61. The resulting mixture was incubated for 1 h at −20 °C to precipitate residual salts from the chemically defined medium. Supernatant fractions were then centrifuged (20,627g for 10 min, 4 °C) and a 1 ml aliquot was removed and stored on ice. Two-phase extraction was then performed as described previously, with slight modifications, to isolate intracellular contents of fungal cells^[249]62. First, the remaining cell pellets were washed twice with pre-cooled nanopure water and 150 μl 100% MeOH + 0.05% BHT was added. The resulting solution was mixed by vortexing and then shaken gently for 2 min at 4 °C. Pre-cooled nanopure water (200 μl) was added with ~150 μl glass beads. Samples were homogenized using a Mini-BeadBeater-8 (BioSpec) for three rounds of 20 s each, with 30 s resting on ice between each round. A 500 μl aliquot of methyl-tert-butyl-ether (MTBE) was then added to each sample followed by vortexing to mix and gentle shaking for 10 min at 4 °C. After centrifugation (20,627g for 10 min, 4 °C) to induce phase separation, the aqueous layer was extracted and placed in a separate microfuge tube. Both the aqueous cell extract and cell supernatant were then dried using a rotary evaporator overnight at room temperature. Fractions were stored at −80 °C until LC-HRMS/MS analysis. Liquid chromatography–mass spectrometry For untargeted metabolomics analysis, samples were analysed using an Impact II high-resolution mass spectrometer (Bruker Daltonics) coupled with an Elute UHPLC system (Bruker). Separation of compounds was achieved using a multigradient method on an Inertsil Ph-3 UHPLC column (2 μm, 150 × 2.1 mm; GL Sciences) equipped with a Ph-3 guard column (2 μm, 2.1 × 10 mm). The mobile phase consisted of water (A) with 0.1% v/v formic acid and methanol (B) with 0.1% v/v formic acid. The separation was conducted using a multi-step gradient ranging from 5% to 99% mobile phase B over 18 min as follows: 0 min 5% B; 0–1 min 5% B; 1–8 min 35% B; 8–10.5 min 99% B; 10.5–14 min 99% B; 14–14.5 min 5% B; 14.5–18 min 5% B. The column temperature was set to 55 °C, while the autosampler was maintained at 4 °C, and the flow rate was 0.3 ml min^−1. Aliquots of 100 μl from each sample were pooled to generate a quality control sample (QC) used for evaluating instrument performance. A QC sample was injected every six samples. Data-dependent acquisitions were conducted in positive (ESI+) and negative (ESI−) electrospray ionization modes to obtain precursor and fragment ion information for annotating compounds. For ESI+, the mass spectrometer settings were as follows: capillary voltage of 4,500 V, nebulizer gas pressure of 2.0 bar, dry gas flow rate of 10 l min^−1, dry gas temperature of 220 °C, mass scan range of 50–1300 m/z, spectra acquisition rate of 10 Hz and cycle time of 0.4 s. Collision energy of 20 V was ramped through each MS/MS scan from 100 to 250%. For ESI−, the capillary voltage was set at −3,500 V. The mass spectrometer was calibrated with sodium formate at the beginning of each run to ensure accuracy. Average mass error in annotation was below 2.0 ppm (ref. [250]32). Data processing and annotation of metabolites Raw data processing was performed using Progenesis QI software (v.3.0.7600.27622) with METLIN plugin v.1.0.7642.33805 (NonLinear Dynamics) and entailed peak picking, alignment, normalization, and database searching^[251]63. Annotations were performed as previously described^[252]63,[253]64. However, to increase confidence in annotations, only experimental MS/MS data were used for querying and matching against the in-house library (Mass Spectrometry Metabolite Library of Standards, MSMLS, supplied by IROA Technologies), METLIN and MassBank of North America^[254]65,[255]66. A Progenesis QI score ≥40 was considered to select a candidate for annotation in accordance with reporting criteria for chemical analysis suggested by the Metabolomics Standards Initiative (MSI)^[256]67,[257]68. Ions generated from QC samples were retained for annotation and included in the dataset if the coefficient of variation (CV) for abundance did not exceed 25%. When compounds were detected in both ion modes, the one with the highest signal-to-noise ratio was retained. Relative quantities of metabolites were determined by calculating their corresponding peak areas. Finally, putative metabolites with high confidence annotations were categorized using the ontology-based ClassyFire Batch Compound Classification web platform (v.2.0, [258]https://cfb.fiehnlab.ucdavis.edu/) and visualized with the ggplot2 package (v.3.5.0) in R. Data was normalized using a robust Progenesis QI built-in approach designed explicitly for untargeted metabolomics^[259]63,[260]64,[261]69. Progenesis QI employed median and means absolute deviation analysis based on all detected abundances to reduce bias and noise in the data^[262]70. A unique gain factor is then calculated for each sample (represented by a scalar multiplier αk) and compound ion abundances were adjusted to a normalization reference run^[263]69. In brief, all compounds were included to normalize the data, resulting in an aggregate matrix with measurements for every compound ion in each run. This aggregate matrix was used to generate a ratio for comparison of compound ion abundance in a particular run with the corresponding value in the normalization reference (typically a QC). Ratios were log transformed to create a normal distribution for all ratio data within each run for all samples. Finally, scalar estimations were employed to align log distributions with that of the normalization reference. The tight clustering exhibited by QC samples at the center of the PCA analysis indicated excellent system performance and stability ([264]Fig. 4c). The resulting dataset and parameters used in statistical analyses were made publicly available^[265]32. Statistical analysis Statistical analyses were performed using GraphPad Prism 8 (v.8.2.1), unless otherwise specified. Statistical significance between two groups was calculated using unpaired, two-tailed Student’s t-tests; significance of three or more groups was calculated using one- or two-way analysis of variance (ANOVA) and corrected for multiple comparisons using the methods indicated. The threshold for statistical significance was defined as α = 0.05 or q = 0.05 for false discovery rate (FDR)-adjusted P values. Exact P values are not provided when P < 0.0001, but information on statistical analyses can be found in figure legends and relevant Source Data files. For parametric tests (t-tests, ANOVA), data distribution was assumed to be normal, but this was not formally tested (individual data points are shown in figures). Sample sizes were determined based on those reported in previous publications^[266]17,[267]19,[268]35. No data were excluded from the analyses. The investigators were not blinded to allocation during the experiments and outcome assessment. Metabolomics data were analysed using the web-based platform Metaboanalyst 5.0 ([269]https://www.metaboanalyst.ca/faces/home.xhtml). Univariate analysis of peak intensity values was performed by volcano plot analysis wherein P values were determined with a two-sided Wilcoxon rank-sum test and then adjusted for multiple testing by FDR correction using an adjusted P value threshold of q = 0.05. Adjusted P values were plotted on a negative log[10] scale. Volcano plot FC values were calculated as the ratio of peak intensity means between samples for each feature and plotted on a log[2] scale with cut-off values of FC > 1.5 or < 0.667. Multivariate analysis was performed to identify interactions and changes in the overall metabolome profile of WT and gsh2Δ mutant strains, as well as supernatant and cellular extract fractions. Metabo-Analyst was used to conduct a PCA and a heatmap analysis to visualize distribution of samples and relative intensity of LC-HRMS/MS features, respectively. Metabolic pathway enrichment analysis was performed using MetaboAnalyst peak list profiling which utilizes the mummichog v.2 algorithm, based on KEGG pathway data, to quantify enrichment of putatively annotated peaks at the network level, as described previously^[270]71. The S. cerevisiae KEGG pathway data (accessed 9 November 2022) was used for enrichment analysis as this dataset most closely resembles C. neoformans pathway information of datasets available on the MetaboAnalyst platform. Enrichment data were visualized using the ggplot2 package (v.3.5.0) in R. Extended Data Extended Data Fig. 1 |. Loss of GSH2 does not affect capsule size but delays virulence and impairs dissemination to the brain. Extended Data Fig. 1 | [271]Open in a new tab 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 10^6 cells ml^−1 with 10-fold serial dilutions. Plates were incubated at 30 °C, 37 °C, 39 °C. d, Melanin production of 10^6 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 10^6 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), gsh2Δ (light blue), and gsh2Δ::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 gsh2Δ mutant cells retrieved from murine lungs (bottom). Lines represent mean values of n = 60 cells from 8 lungs per strain per time point. Extended Data Fig. 2 |. Mutants lacking GSH2 have reduced lung proliferation and cell size in vivo. Extended Data Fig. 2 | [272]Open in a new tab a, Representative images of H & E stained lung tissue sections from four mice infected with WT, gsh2Δ, or gsh2Δ::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 gsh2Δ 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. Extended Data Fig. 3 |. GSH is required for growth in minimal medium and is not restored with ascorbate or sulfur-containing amino acids. Extended Data Fig. 3 | [273]Open in a new tab a, Growth curve analysis of WT (light blue circles), gsh2Δ (blue squares or circles), and gsh2Δ::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 10^6 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, gsh2Δ, and gsh2Δ::GSH2 strains in minimal medium with or without GSH supplementation at the indicated concentrations. Data points for a and c indicate mean OD[600] values ± s.d., and the initial inoculum for each strain was 2 × 10^4 cells ml^−1. Growth was monitored for 72 h with OD[600] values measured every 24 h. d–e, Growth of WT, gsh2Δ, and gsh2Δ::GSH2 cells in 5 ml liquid cultures of l-DOPA medium (with or without GSH supplementation in e). Initial inoculum for each strain was 10^6 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 [274]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. Extended Data Fig. 4 |. Metabolic profiling of the gsh2Δ mutant relative to WT reveals differences in extracellular and intracellular fractions. Extended Data Fig. 4 | [275]Open in a new tab a, Volcano plots showing QC-normalized LC-HRMS/MS feature data and differentially abundant metabolites between WT and gsh2Δ 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 gsh2Δ 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 gsh2Δ mutant supernatant and cellular extract fractions with QC samples for data normalization. Each data set represents n = 3 biological replicates. Purple = WT; red = gsh2Δ mutant; green = QC. Extended Data Fig. 5 |. Mutants lacking GSH2 show distinct changes in relative abundance of key energy metabolites, antioxidants, and extracellular acids. Extended Data Fig. 5 | [276]Open in a new tab Relative abundance of select compounds identified via LC-HRMS/MS between WT (blue) and gsh2Δ 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. Extended Data Fig. 6 |. Extracellular acidification of gsh2Δ mutants is independent of l-DOPA and GSH pathway metabolites influence melanin formation. Extended Data Fig. 6 | [277]Open in a new tab a, pH values of supernatant isolates from WT and gsh2Δ mutant cells grown in l-asparagine minimal medium and normalized to 10^7 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. Supplementary Material Supplementary Figures [278]NIHMS2063451-supplement-Supplementary_Figures.pdf^ (1.1MB, pdf) Acknowledgements