205 research outputs found

    Surviving the heat of the moment : a fungal pathogens perspective

    Get PDF
    Peer reviewedPublisher PD

    Translation inhibition by rocaglates activates a species-specific cell death program in the emerging fungal pathogen Candida auris

    Get PDF
    Fungal infections are a major contributor to infectious disease-related deaths worldwide. Recently, global emergence of the fungal pathogen Candida auris has caused considerable concern because most C. auris isolates are resistant to fluconazole, the most commonly administered antifungal, and some isolates are resistant to drugs from all three major antifungal classes. To identify novel agents with bioactivity against C. auris, we screened 2,454 compounds from a diversity-oriented synthesis collection. Of the five hits identified, most shared a common rocaglate core structure and displayed fungicidal activity against C. auris These rocaglate hits inhibited translation in C. auris but not in its pathogenic relative Candida albicans Species specificity was contingent on variation at a single amino acid residue in Tif1, a fungal member of the eukaryotic initiation factor 4A (eIF4A) family of translation initiation factors known to be targeted by rocaglates. Rocaglate-mediated inhibition of translation in C. auris activated a cell death program characterized by loss of mitochondrial membrane potential, increased caspase-like activity, and disrupted vacuolar homeostasis. In a rocaglate-sensitized C. albicans mutant engineered to express translation initiation factor 1 (Tif1) with the variant amino acid that we had identified in C. auris, translation was inhibited but no programmed cell death phenotypes were observed. This surprising finding suggests divergence between these related fungal pathogens in their pathways of cellular responses to translation inhibition. From a therapeutic perspective, the chemical biology that we have uncovered reveals species-specific vulnerability in C. auris and identifies a promising target for development of new, mechanistically distinct antifungals in the battle against this emerging pathogen. IMPORTANCE Emergence of the fungal pathogen Candida auris has ignited intrigue and alarm within the medical community and the public at large. This pathogen is unusually resistant to antifungals, threatening to overwhelm current management options. By screening a library of structurally diverse molecules, we found that C. auris is surprisingly sensitive to translation inhibition by a class of compounds known as rocaglates (also known as flavaglines). Despite the high level of conservation across fungi in their protein synthesis machinery, these compounds inhibited translation initiation and activated a cell death program in C. auris but not in its relative Candida albicans Our findings highlight a surprising divergence across the cell death programs operating in Candida species and underscore the need to understand the specific biology of a pathogen in attempting to develop more-effective treatments against it.Published versio

    Insights into the host-pathogen interaction: C. albicans manipulation of macrophage pyroptosis

    Get PDF
    The innate immune system is the first defense against invasive fungal infections, including those caused by Candida albicans. Although C. albicans can exist as a commensal, it can also cause systemic or mucosal infections, especially when the innate immune system is impaired. A key aspect of the interaction between C. albicans and innate immune cells is the ability of C. albicans to induce macrophage pyroptosis, an inflammatory cell death program. The induction of pyroptosis is temporally coupled to a morphological transition between yeast and filamentous growth. However, the relationship between fungal morphogenesis and activation of macrophage pyroptosis is complex. Although most C. albicans mutants with defects in filamentation are also unable to induce macrophage pyroptosis, filamentation is neither necessary nor sufficient for activation of pyroptosis. In our study [O’Meara et al., 2018 mBio], we set out to map the genetic circuitry in both the fungus and the host macrophage that leads to pyroptosis, and determine the impact of altered pyroptosis on infection. We identified 98 C. albicans genes that were dispensable for filamentation in the macrophage but important for enabling the fungus to activate macrophage pyroptosis. Using these mutants, we demonstrated that pyroptosis is required for robust neutrophil accumulation at the site of C. albicans infection. We also showed that, in contrast to previous work, inflammasome priming and activation can be decoupled in the response to C. albicans infection, and that phagolysosomal rupture is not the inflammasome activating signal. Our work provides the most comprehensive analysis of C. albicans interactions with host cells to date, and reveals new factors governing the outcomes of this interaction

    Ydj1 governs fungal morphogenesis and stress response, and facilitates mitochondrial protein import via Mas1 and Mas2

    Get PDF
    We thank Zhen-Yuan Lin for help in the preparation of the AP-MS samples, and Cathy Collins for technical assistance. MDL is supported by a Sir Henry Wellcome Postdoctoral Fellowship (Wellcome Trust 096072), LEC is supported by a Canada Research Chair in Microbial Genomics and Infectious Disease and by Cana-dian Institutes of Health Research (CIHR) Grants MOP-119520 and MOP-86452. OK is supported by National Insti-tutes of Health grant 5R01GM108975. A-CG is supported by a CIHR Foundation Grant (FDN143301), Genome Cana-da Genomics Innovation Network (GIN) Node and Tech-nical Development Grants, and a Canada Research Chair in Functional Proteomics. J-PL was supported by a TD Bank Health Research Fellowship at the Lunenfeld-Tanenbaum Research Institute and by a Scholarship for the Next Gen-eration of Scientists from the Cancer Research Society. JLX is supported by a CIHR – Frederick Banting and Charles Best Canada Graduate Scholarship. The funding agencies had no role in the study design, data collection and inter-pretation, or the decision to submit the work for publication.Peer reviewedPublisher PD

    Candida albicans Is Resistant to Polyglutamine Aggregation and Toxicity

    Get PDF
    Acknowledgments We thank the Donnelly Sequencing Centre for sequencing, and Jonathan Krieger at the SikKids Proteomics, Analytics, Robotics & Chemical Biology Centre at The Hospital for Sick Children for mass spectrometry analysis. M.D.L. is supported by a Sir Henry Wellcome Postdoctoral Fellowship (Wellcome Trust grant 096072), T.K. is supported by a Queen Elizabeth II Graduate Scholarship in Science and Technology (University of Toronto), M.L.D. is supported by a Canadian Institutes of Health Research (CIHR) Operating grant 325538, L.E.C. is supported by a Canada Research Chair in Microbial Genomics and Infectious Disease, by CIHR grants MOP-119520 and MOP-86452, and by the Natural Sciences and Engineering Research Council (NSERC) of Canada (grants 06261 and 462167).Peer reviewedPublisher PD

    PKC Signaling Regulates Drug Resistance of the Fungal Pathogen Candida albicans via Circuitry Comprised of Mkc1, Calcineurin, and Hsp90

    Get PDF
    Fungal pathogens exploit diverse mechanisms to survive exposure to antifungal drugs. This poses concern given the limited number of clinically useful antifungals and the growing population of immunocompromised individuals vulnerable to life-threatening fungal infection. To identify molecules that abrogate resistance to the most widely deployed class of antifungals, the azoles, we conducted a screen of 1,280 pharmacologically active compounds. Three out of seven hits that abolished azole resistance of a resistant mutant of the model yeast Saccharomyces cerevisiae and a clinical isolate of the leading human fungal pathogen Candida albicans were inhibitors of protein kinase C (PKC), which regulates cell wall integrity during growth, morphogenesis, and response to cell wall stress. Pharmacological or genetic impairment of Pkc1 conferred hypersensitivity to multiple drugs that target synthesis of the key cell membrane sterol ergosterol, including azoles, allylamines, and morpholines. Pkc1 enabled survival of cell membrane stress at least in part via the mitogen activated protein kinase (MAPK) cascade in both species, though through distinct downstream effectors. Strikingly, inhibition of Pkc1 phenocopied inhibition of the molecular chaperone Hsp90 or its client protein calcineurin. PKC signaling was required for calcineurin activation in response to drug exposure in S. cerevisiae. In contrast, Pkc1 and calcineurin independently regulate drug resistance via a common target in C. albicans. We identified an additional level of regulatory control in the C. albicans circuitry linking PKC signaling, Hsp90, and calcineurin as genetic reduction of Hsp90 led to depletion of the terminal MAPK, Mkc1. Deletion of C. albicans PKC1 rendered fungistatic ergosterol biosynthesis inhibitors fungicidal and attenuated virulence in a murine model of systemic candidiasis. This work establishes a new role for PKC signaling in drug resistance, novel circuitry through which Hsp90 regulates drug resistance, and that targeting stress response signaling provides a promising strategy for treating life-threatening fungal infections

    Mapping the Hsp90 Genetic Network Reveals Ergosterol Biosynthesis and Phosphatidylinositol-4-Kinase Signaling as Core Circuitry Governing Cellular Stress

    Get PDF
    <div><p><i>Candida albicans</i> is a leading human fungal pathogen that causes life-threatening systemic infections. A key regulator of <i>C</i>. <i>albicans</i> stress response, drug resistance, morphogenesis, and virulence is the molecular chaperone Hsp90. Targeting Hsp90 provides a powerful strategy to treat fungal infections, however, the therapeutic utility of current inhibitors is compromised by toxicity due to inhibition of host Hsp90. To identify components of the Hsp90-dependent circuitry governing virulence and drug resistance that are sufficiently divergent for selective targeting in the pathogen, we pioneered chemical genomic profiling of the Hsp90 genetic network in <i>C</i>. <i>albicans</i>. Here, we screen mutant collections covering ~10% of the genome for hypersensitivity to Hsp90 inhibition in multiple environmental conditions. We identify 158 <i>HSP90</i> chemical genetic interactors, most of which are important for growth only in specific environments. We discovered that the sterol C-22 desaturase gene <i>ERG5</i> and the phosphatidylinositol-4-kinase (PI4K) gene <i>STT4</i> are <i>HSP90</i> genetic interactors under multiple conditions, suggesting a function upstream of Hsp90. By systematic analysis of the ergosterol biosynthetic cascade, we demonstrate that defects in ergosterol biosynthesis induce cellular stress that overwhelms Hsp90’s functional capacity. By analysis of the phosphatidylinositol pathway, we demonstrate that there is a genetic interaction between the PI4K Stt4 and Hsp90. We also establish that Stt4 is required for normal actin polarization through regulation of Wal1, and suggest a model in which defects in actin remodeling induces stress that creates a cellular demand for Hsp90 that exceeds its functional capacity. Consistent with this model, actin inhibitors are synergistic with Hsp90 inhibitors. We highlight new connections between Hsp90 and virulence traits, demonstrating that Erg5 and Stt4 enable activation of macrophage pyroptosis. This work uncovers novel circuitry regulating Hsp90 functional capacity and new effectors governing drug resistance, morphogenesis and virulence, revealing new targets for antifungal drug development.</p></div
    • …
    corecore