A drug-sensitive genetic network masks fungi from the immune system

Robert T Wheeler, Gerald R Fink, Robert T Wheeler, Gerald R Fink

Abstract

Fungal pathogens can be recognized by the immune system via their beta-glucan, a potent proinflammatory molecule that is present at high levels but is predominantly buried beneath a mannoprotein coat and invisible to the host. To investigate the nature and significance of "masking" this molecule, we characterized the mechanism of masking and consequences of unmasking for immune recognition. We found that the underlying beta-glucan in the cell wall of Candida albicans is unmasked by subinhibitory doses of the antifungal drug caspofungin, causing the exposed fungi to elicit a stronger immune response. Using a library of bakers' yeast (Saccharomyces cerevisiae) mutants, we uncovered a conserved genetic network that is required for concealing beta-glucan from the immune system and limiting the host response. Perturbation of parts of this network in the pathogen C. albicans caused unmasking of its beta-glucan, leading to increased beta-glucan receptor-dependent elicitation of key proinflammatory cytokines from primary mouse macrophages. By creating an anti-inflammatory barrier to mask beta-glucan, opportunistic fungi may promote commensal colonization and have an increased propensity for causing disease. Targeting the widely conserved gene network required for creating and maintaining this barrier may lead to novel broad-spectrum antimycotics.

Conflict of interest statement

Competing interests. The authors have declared that no competing interests exist.

Figures

Figure 1. Fungal β-Glucan Is Buried in…
Figure 1. Fungal β-Glucan Is Buried in the Cell Wall and Largely Inaccessible
(A) Transmission electron micrograph demonstrating layer structure of fungal cell wall (courtesy of C. Rondeau). The plasma membrane is tightly connected to a thick layer of β-glucan network. Mannoproteins are linked to β-glucan and protrude outside of this layer to make up a dense coat. Schematic adapted from [5]. (B–D) There is little β-glucan on live C. albicans or S. cerevisiae that is exposed and accessible to the anti-β-glucan antibody (B), the Dectin-CRD (C), or the Dectin-CRD-anti-Myc probe (D). The staining with anti-β-glucan and Dectin-CRD-anti-Myc is nearly indistinguishable, and is more specific than that with the directly labeled Dectin-CRD. The Dectin-CRD-anti-Myc has the same size as an antibody and the same specificity as Dectin-1. The difference in staining between the β-glucan-binding reagents (B and D versus C) is likely due to the size of the reagents (IgG has dimensions of approximately 95 Å × 171 Å [33], while the CRD of CD69, which is similar to the Dectin-1 CRD, has dimensions of 44 Å × 32 Å × 30 Å [34]) relative to the estimated pore size of the S. cerevisiae cell wall (58 Å [23]), and thus the monomeric Dectin-CRD likely has more access to smaller areas of exposed β-glucan.
Figure 2. Subinhibitory Concentrations of the Antifungal…
Figure 2. Subinhibitory Concentrations of the Antifungal Drug CF Cause Exposure of β-Glucan
Wild-type C. albicans (CAF2) was grown overnight for ten generations in YPD medium, favoring yeast-form growth (A, B, C, and E) or RPMI medium, favoring hyphal growth (D). Cultures grown at one-quarter and one-eighth of the CF MIC50 were stained with anti-β-glucan antibody and Cy3-labeled secondary antibody (A and D) for visualization by microscopy or with PE-labeled secondary antibody (B) for FACS quantification. Mean fluorescence intensity (MFI) values were 9, 65, and 161, respectively, for no treatment, one-eighth the CF MIC50, and one-quarter the CF MIC50. Concurrently, cells were labeled briefly with propidium iodide to assess viability and visualized by epifluorescence microscopy or quantified by FACS (C). Cells grown overnight in YPD with or without CF were UV-inactivated and then exposed to BMDMs at a yeast:macrophage ratio of 10:1. Supernatants were taken at 6 h and assayed for TNFα (E).
Figure 3. Automated Genome-Wide Screen for Increased…
Figure 3. Automated Genome-Wide Screen for Increased β-Glucan Exposure
We screened the genome-wide set of S. cerevisiae knockout mutants by staining with anti-β-glucan antibody and quantifying β-glucan exposure using the Cellomics system. (A) Images of yeast were taken at 20×, and object-finding software used the ConA fluorescence signal to identify cells (green outline); the software then applied a mask (blue outline) and quantified the average level of fluorescence from the anti-β-glucan channel for each cell. Wild-type yeast (left photomicrographs) show little or no fluorescence from the anti-β-glucan channel, whereas the unmasked vrp1Δ mutant shows high levels of fluorescence from this channel. (B) Most of the mutants found with increased β-glucan exposure also showed greater binding to Dectin-CRD and increased TNFα elicitation from RAW 264.7 macrophages. Of the 76 mutants identified with increased anti-β-glucan binding (encircled with the black line), 48 (encircled with the dotted line) hyperelicited TNFα from macrophages and 65 (encircled with the red line) showed increased binding to the labeled Dectin-CRD. Numbers within the black oval show the intersections of each of these groups (e.g., 44 had increased anti-β-glucan binding and increased Dectin-CRD binding and increased TNFα elicitation from macrophages). (C–E) To exemplify the methodology used, we chose the gas1Δ mutant, which has intermediate β-glucan exposure and TNFα elicitation. (C) Live wild-type (upper left image) or gas1Δ mutant (lower left image) S. cerevisiae were stained with anti-β-glucan antibody. To show specificity of the binding, gas1Δ cells were stained omitting primary antibody (upper right image) or antibody was preincubated in 100 μg/ml soluble glucan (laminarin) before and during the staining (lower right image). (D) Live wild-type or gas1Δ mutant cells were stained with Alexa Fluor-labeled Dectin-CRD. (E) Live wild-type or gas1Δ mutant cells were exposed to RAW264.7 macrophages at a ratio of 5:1 (yeast:macrophage), and supernatants were collected after 6 h and measured for TNFα levels.
Figure 4. The Cell Wall Remodeling Network…
Figure 4. The Cell Wall Remodeling Network Required for β-Glucan Masking
A map of physical and genetic interactions among β-glucan masking genes was made using the Osprey v1.2.0 visualization program (available at: http://biodata.mshri.on.ca/osprey/servlet/Index). All genes of hypereliciting mutants identified in the screen are represented. Lines connecting genes are colored to represent the nature of known interaction(s) between a pair of genes (i.e., synthetic lethality, two-hybrid, or coimmunoprecipitation). Vertices are colored to represent the cellular processes directed by the gene product, as annotated by the Yeast Proteome Database (available at http://www.proteome.com). Vertices of genes required for caspofungin resistance [15,16] are circled in black.
Figure 5. C. albicans Mutants of Masking…
Figure 5. C. albicans Mutants of Masking Genes Have More Exposed β-Glucan
Wild-type or mutant C. albicans strains were grown overnight in YPD, then stained with anti-β-glucan antibody and Cy3-labeled (A–C) or PE-labeled (D–F) secondary antibody. (A–C) Upper photomicrographs show overlay of brightfield and anti-β-glucan staining of Cy3-labeled cells; lower photomicrographs show anti-β-glucan staining alone. (D–F) Overlay histograms of FACS analysis of PE-labeled cells; data on 20,000 cells are shown. MFI values for wild-type and mutants are shown in insets. In parallel experiments, strains that were complemented with a wild-type copy of the gene showed full reversal of β-glucan exposure (for KRE5) or partial reduction in exposure (for PHR2) (Figure S5). This correlates with other phenotypes observed for these complemented strains [17,20,35].
Figure 6. Unmasked C. albicans Mutants Hyperelicit…
Figure 6. Unmasked C. albicans Mutants Hyperelicit TNFα from Macrophages through the β-Glucan Receptor
(A) C. albicans strains were exposed to RAW264.7 macrophages at a ratio of 2:1 (yeast:macrophage), and supernatants were collected after 6 h. (B) Different numbers of C. albicans (wild-type or kre5Δ/Δ mutant) were exposed to BMDMs, and supernatants were collected after 6 h. (C) BMDMs were pretreated for 20 min on ice with media or soluble β-glucan (laminarin); they were then exposed to different C. albicans strains at a ratio of 10:1 (yeast:macrophage). After unbound fungi were washed off, macrophages were incubated for 6 h at 37 °C, and supernatants were collected for TNFα quantitation.

References

    1. Underhill DM, Ozinsky A. Phagocytosis of microbes: Complexity in action. Annu Rev Immunol. 2002;20:825–852.
    1. Hajjeh RA, Sofair AN, Harrison LH, Lyon GM, Arthington-Skaggs BA, et al. Incidence of bloodstream infections due to Candida species and in vitro susceptibilities of isolates collected from 1998 to 2000 in a population-based active surveillance program. J Clin Microbiol. 2004;42:1519–1527.
    1. Singh N, Paterson DL. Aspergillus infections in transplant recipients. Clin Microbiol Rev. 2005;18:44–69.
    1. Poulain D, Jouault T. Candida albicans cell wall glycans, host receptors and responses: Elements for a decisive crosstalk. Curr Opin Microbiol. 2004;7:342–349.
    1. Chaffin WL, Lopez-Ribot JL, Casanova M, Gozalbo D, Martinez JP. Cell wall and secreted proteins of Candida albicans: Identification, function, and expression. Microbiol Mol Biol Rev. 1998;62:130–180.
    1. Torosantucci A, Bromuro C, Chiani P, De Bernardis F, Berti F, et al. A novel glyco-conjugate vaccine against fungal pathogens. J Exp Med. 2005;202:597–606.
    1. Brown GD, Gordon S. Immune recognition of fungal beta-glucans. Cell Microbiol. 2005;7:471–479.
    1. Denning DW. Echinocandin antifungal drugs. Lancet. 2003;362:1142–1151.
    1. Orlean P. Biogenesis of yeast wall and surface components. In: Pringle JR, Broach JR, Jones EW, editors. The molecular and cellular biology of the yeast Saccharomyces cerevisiae. Plainview (New York): Cold Spring Harbor Laboratory Press; 1997. 1131 p.
    1. Meikle PJ, Bonig I, Hoogenraad NJ, Clarke AE, Stone BA. The location of (1-]3)-beta-glucans in the walls of pollen tubes of Nicotiana alata using a (1-]3)-beta-glucan-specific monoclonal-antibody. Planta. 1991;185:1–8.
    1. Gantner BN, Simmons RM, Underhill DM. Dectin-1 mediates macrophage recognition of Candida albicans yeast but not filaments. EMBO J. 2005;24:1277–1286.
    1. Angiolella L, Maras B, Stringaro A, Arancia G, Mondello F, et al. Glucan-associated protein modulations and ultrastructural changes of the cell wall in Candida albicans treated with micafungin, a water-soluble, lipopeptide antimycotic. J Chemother. 2005;17:409–416.
    1. Drees BL, Sundin B, Brazeau E, Caviston JP, Chen GC, et al. A protein interaction map for cell polarity development. J Cell Biol. 2001;154:549–571.
    1. Pruyne D, Legesse-Miller A, Gao L, Dong Y, Bretscher A. Mechanisms of polarized growth and organelle segregation in yeast. Annu Rev Cell Dev Biol. 2004;20:559–591.
    1. Lesage G, Sdicu AM, Menard P, Shapiro J, Hussein S, et al. Analysis of beta-1,3-glucan assembly in Saccharomyces cerevisiae using a synthetic interaction network and altered sensitivity to caspofungin. Genetics. 2004;167:35–49.
    1. Reinoso-Martin C, Schuller C, Schuetzer-Muehlbauer M, Kuchler K. The yeast protein kinase C cell integrity pathway mediates tolerance to the antifungal drug caspofungin through activation of Slt2p mitogen-activated protein kinase signaling. Eukaryot Cell. 2003;2:1200–1210.
    1. De Bernardis F, Muhlschlegel FA, Cassone A, Fonzi WA. The pH of the host niche controls gene expression in and virulence of Candida albicans . Infect Immun. 1998;66:3317–3325.
    1. Fonzi WA. PHR1 and PHR2 of Candida albicans encode putative glycosidases required for proper cross-linking of beta-1,3- and beta-1,6-glucans. J Bacteriol. 1999;181:7070–7079.
    1. Kollar R, Reinhold BB, Petrakova E, Yeh HJ, Ashwell G, et al. Architecture of the yeast cell wall. Beta(1—>6)-glucan interconnects mannoprotein, beta(1—>)3-glucan, and chitin. J Biol Chem. 1997;272:17762–17775.
    1. Herrero AB, Magnelli P, Mansour MK, Levitz SM, Bussey H, et al. KRE5 gene null mutant strains of Candida albicans are avirulent and have altered cell wall composition and hypha formation properties. Eukaryot Cell. 2004;3:1423–1432.
    1. Zlotnik H, Fernandez MP, Bowers B, Cabib E. Saccharomyces cerevisiae mannoproteins form an external cell wall layer that determines wall porosity. J Bacteriol. 1984;159:1018–1026.
    1. Romani L. Immunity to Candida albicans: Th1, Th2 cells and beyond. Curr Opin Microbiol. 1999;2:363–367.
    1. De Nobel JG, Barnett JA. Passage of molecules through yeast cell walls: A brief essay-review. Yeast. 1991;7:313–323.
    1. Romani L. Immunity to fungal infections. Nat Rev Immunol. 2004;4:1–23.
    1. Mathews HL, Witek-Janusek L. Host defense against oral, esophageal, and gastrointestinal candidiasis. In: Calderone RA, editor. Candida and candidiasis. Washington (D. C.): ASM Press; 2002. pp. 179–192.
    1. Tuite A, Mullick A, Gros P. Genetic analysis of innate immunity in resistance to Candida albicans . Genes Immun. 2004;5:576–587.
    1. Ishibashi K, Yoshida M, Nakabayashi I, Shinohara H, Miura NN, et al. Role of anti-beta-glucan antibody in host defense against fungi. FEMS Immunol Med Microbiol. 2005;44:99–109.
    1. Viriyakosol S, Fierer J, Brown GD, Kirkland TN. Innate immunity to the pathogenic fungus Coccidioides posadasii is dependent on Toll-like receptor 2 and Dectin-1. Infect Immun. 2005;73:1553–1560.
    1. Steele C, Marrero L, Swain S, Harmsen AG, Zheng M, et al. Alveolar macrophage-mediated killing of Pneumocystis carinii f. sp. muris involves molecular recognition by the Dectin-1 beta-glucan receptor. J Exp Med. 2003;198:1677–1688.
    1. Winzeler EA, Shoemaker DD, Astromoff A, Liang H, Anderson K, et al. Functional characterization of the S. cerevisiae genome by gene deletion and parallel analysis. Science. 1999;285:901–906.
    1. Fonzi WA, Irwin MY. Isogenic strain construction and gene mapping in Candida albicans . Genetics. 1993;134:717–728.
    1. Brown GD, Herre J, Williams DL, Willment JA, Marshall AS, et al. Dectin-1 mediates the biological effects of beta-glucans. J Exp Med. 2003;197:1119–1124.
    1. Saphire EO, Parren PW, Pantophlet R, Zwick MB, Morris GM, et al. Crystal structure of a neutralizing human IGG against HIV-1: A template for vaccine design. Science. 2001;293:1155–1159.
    1. Llera AS, Viedma F, Sanchez-Madrid F, Tormo J. Crystal structure of the C-type lectin-like domain from the human hematopoietic cell receptor CD69. J Biol Chem. 2001;276:7312–7319.
    1. Muhlschlegel FA, Fonzi WA. PHR2 of Candida albicans encodes a functional homolog of the pH-regulated gene PHR1 with an inverted pattern of pH-dependent expression. Mol Cell Biol. 1997;17:5960–5967.
    1. Page N, Gerard-Vincent M, Menard P, Beaulieu M, Azuma M, et al. A Saccharomyces cerevisiae genome-wide mutant screen for altered sensitivity to K1 killer toxin. Genetics. 2003;163:875–894.
    1. el-Sherbeini M, Clemas JA. Cloning and characterization of GNS1: A Saccharomyces cerevisiae gene involved in synthesis of 1,3-beta-glucan in vitro. J Bacteriol. 1995;177:3227–3234.
    1. Shahinian S, Dijkgraaf GJ, Sdicu AM, Thomas DY, Jakob CA, et al. Involvement of protein N-glycosyl chain glucosylation and processing in the biosynthesis of cell wall beta-1,6-glucan of Saccharomyces cerevisiae . Genetics. 1998;149:843–856.
    1. Dijkgraaf GJ, Abe M, Ohya Y, Bussey H. Mutations in Fks1p affect the cell wall content of beta-1,3- and beta-1,6-glucan in Saccharomyces cerevisiae . Yeast. 2002;19:671–690.

Source: PubMed

3
Abonnieren