Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nature ; 596(7870): 114-118, 2021 08.
Article in English | MEDLINE | ID: mdl-34262174

ABSTRACT

Pathogenic fungi reside in the intestinal microbiota but rarely cause disease. Little is known about the interactions between fungi and the immune system that promote commensalism. Here we investigate the role of adaptive immunity in promoting mutual interactions between fungi and host. We find that potentially pathogenic Candida species induce and are targeted by intestinal immunoglobulin A (IgA) responses. Focused studies on Candida albicans reveal that the pathogenic hyphal morphotype, which is specialized for adhesion and invasion, is preferentially targeted and suppressed by intestinal IgA responses. IgA from mice and humans directly targets hyphal-enriched cell-surface adhesins. Although typically required for pathogenesis, C. albicans hyphae are less fit for gut colonization1,2 and we show that immune selection against hyphae improves the competitive fitness of C. albicans. C. albicans exacerbates intestinal colitis3 and we demonstrate that hyphae and an IgA-targeted adhesin exacerbate intestinal damage. Finally, using a clinically relevant vaccine to induce an adhesin-specific immune response protects mice from C. albicans-associated damage during colitis. Together, our findings show that adaptive immunity suppresses harmful fungal effectors, with benefits to both C. albicans and its host. Thus, IgA uniquely uncouples colonization from pathogenesis in commensal fungi to promote homeostasis.


Subject(s)
Adaptive Immunity , Candida albicans/immunology , Candida albicans/physiology , Host-Pathogen Interactions/immunology , Symbiosis/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antigens, Fungal/immunology , Candida albicans/pathogenicity , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Female , Fungal Vaccines/immunology , Gastrointestinal Microbiome/immunology , Humans , Hyphae/immunology , Immunoglobulin A/immunology , Male , Mice , Middle Aged , Young Adult
2.
Cell ; 135(1): 174-88, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18854164

ABSTRACT

The fungus Cryptococcus neoformans is a leading cause of mortality and morbidity among HIV-infected individuals. We utilized the completed genome sequence and optimized methods for homologous DNA replacement using high-velocity particle bombardment to engineer 1201 gene knockout mutants. We screened this resource in vivo for proliferation in murine lung tissue and in vitro for three well-recognized virulence attributes-polysaccharide capsule formation, melanization, and growth at body temperature. We identified dozens of previously uncharacterized genes that affect these known attributes as well as 40 infectivity mutants without obvious defects in these traits. The latter mutants affect predicted regulatory factors, secreted proteins, and immune-related factors, and represent powerful tools for elucidating novel virulence mechanisms. In particular, we describe a GATA family transcription factor that inhibits phagocytosis by murine macrophages independently of the capsule, indicating a previously unknown mechanism of innate immune modulation.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Animals , Bacterial Capsules/genetics , Gene Deletion , Humans , Lung/microbiology , Melanins/genetics , Mice , Mutagenesis, Insertional , Virulence
3.
PLoS Biol ; 17(6): e3000331, 2019 06.
Article in English | MEDLINE | ID: mdl-31226107

ABSTRACT

Eukaryotes have evolved elaborate mechanisms to ensure that chromosomes segregate with high fidelity during mitosis and meiosis, and yet specific aneuploidies can be adaptive during environmental stress. Here, we identify a chromatin-based system required for inducible aneuploidy in a human pathogen. Candida albicans utilizes chromosome missegregation to acquire tolerance to antifungal drugs and for nonmeiotic ploidy reduction after mating. We discovered that the ancestor of C. albicans and 2 related pathogens evolved a variant of histone 2A (H2A) that lacks the conserved phosphorylation site for kinetochore-associated Bub1 kinase, a key regulator of chromosome segregation. Using engineered strains, we show that the relative gene dosage of this variant versus canonical H2A controls the fidelity of chromosome segregation and the rate of acquisition of tolerance to antifungal drugs via aneuploidy. Furthermore, whole-genome chromatin precipitation analysis reveals that Centromere Protein A/ Centromeric Histone H3-like Protein (CENP-A/Cse4), a centromeric histone H3 variant that forms the platform of the eukaryotic kinetochore, is depleted from tetraploid-mating products relative to diploid parents and is virtually eliminated from cells exposed to aneuploidy-promoting cues. We conclude that genetically programmed and environmentally induced changes in chromatin can confer the capacity for enhanced evolvability via chromosome missegregation.


Subject(s)
Centromere Protein A/metabolism , Chromosome Segregation/physiology , Histones/metabolism , Aneugens/metabolism , Aneuploidy , Candida albicans/genetics , Candida albicans/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , Centromere Protein A/physiology , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Evolution, Molecular , Histones/physiology , Kinetochores/metabolism , Meiosis , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
4.
J Bacteriol ; 196(9): 1683-93, 2014 May.
Article in English | MEDLINE | ID: mdl-24532776

ABSTRACT

In microbiology, gene disruption and subsequent experiments often center on phenotypic changes caused by one class of specialized metabolites (quorum sensors, virulence factors, or natural products), disregarding global downstream metabolic effects. With the recent development of mass spectrometry-based methods and technologies for microbial metabolomics investigations, it is now possible to visualize global production of diverse classes of microbial specialized metabolites simultaneously. Using imaging mass spectrometry (IMS) applied to the analysis of microbiology experiments, we can observe the effects of mutations, knockouts, insertions, and complementation on the interactive metabolome. In this study, a combination of IMS and liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to visualize the impact on specialized metabolite production of a transposon insertion into a Pseudomonas aeruginosa phenazine biosynthetic gene, phzF2. The disruption of phenazine biosynthesis led to broad changes in specialized metabolite production, including loss of pyoverdine production. This shift in specialized metabolite production significantly alters the metabolic outcome of an interaction with Aspergillus fumigatus by influencing triacetylfusarinine production.


Subject(s)
Bacterial Proteins/genetics , DNA Transposable Elements , Mutagenesis, Insertional , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/metabolism , Chromatography, Liquid , Phenazines/metabolism , Pseudomonas aeruginosa/chemistry , Tandem Mass Spectrometry
5.
PLoS Pathog ; 8(11): e1002956, 2012.
Article in English | MEDLINE | ID: mdl-23133381

ABSTRACT

The yeast Candida albicans transitions between distinct lifestyles as a normal component of the human gastrointestinal microbiome and the most common agent of disseminated fungal disease. We previously identified Sef1 as a novel Cys(6)Zn(2) DNA binding protein that plays an essential role in C. albicans virulence by activating the transcription of iron uptake genes in iron-poor environments such as the host bloodstream and internal organs. Conversely, in the iron-replete gastrointestinal tract, persistence as a commensal requires the transcriptional repressor Sfu1, which represses SEF1 and genes for iron uptake. Here, we describe an unexpected, transcription-independent role for Sfu1 in the direct inhibition of Sef1 function through protein complex formation and localization in the cytoplasm, where Sef1 is destabilized. Under iron-limiting conditions, Sef1 forms an alternative complex with the putative kinase, Ssn3, resulting in its phosphorylation, nuclear localization, and transcriptional activity. Analysis of sfu1 and ssn3 mutants in a mammalian model of disseminated candidiasis indicates that these post-transcriptional regulatory mechanisms serve as a means for precise titration of C. albicans virulence.


Subject(s)
Candida albicans/metabolism , Candida albicans/pathogenicity , Core Binding Factor Alpha 1 Subunit/biosynthesis , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal/physiology , Candida albicans/genetics , Candidiasis/genetics , Candidiasis/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Fungal Proteins/genetics , Humans , Iron/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation
6.
mBio ; 14(2): e0343422, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36809010

ABSTRACT

The ability to transition between yeast and filamentous growth states is critical for virulence of the leading human fungal pathogen Candida albicans. Large-scale genetic screens have identified hundreds of genes required for this morphological switch, but the mechanisms by which many of these genes orchestrate this developmental transition remain largely elusive. In this study, we characterized the role of Ent2 in governing morphogenesis in C. albicans. We showed that Ent2 is required for filamentous growth under a wide range of inducing conditions and is also required for virulence in a mouse model of systemic candidiasis. We found that the epsin N-terminal homology (ENTH) domain of Ent2 enables morphogenesis and virulence and does so via a physical interaction with the Cdc42 GTPase-activating protein (GAP) Rga2 and regulation of its localization. Further analyses revealed that overexpression of the Cdc42 effector protein Cla4 can overcome the requirement for the ENTH-Rga2 physical interaction, indicating that Ent2 functions, at least in part, to enable proper activation of the Cdc42-Cla4 signaling pathway in the presence of a filament-inducing cue. Overall, this work characterizes the mechanism by which Ent2 regulates hyphal morphogenesis in C. albicans, unveils the importance of this factor in enabling virulence in an in vivo model of systemic candidiasis and adds to the growing understanding of the genetic control of a key virulence trait. IMPORTANCE Candida albicans is a leading human fungal pathogen that can cause life-threatening infections in immunocompromised individuals, with mortality rates of ~40%. The ability of this organism to grow in both yeast and filamentous forms is critical for the establishment of systemic infection. Genomic screens have identified many genes required for this morphological transition, yet our understanding of the mechanisms that regulate this key virulence trait remains incomplete. In this study, we characterized Ent2 as a core regulator of C. albicans morphogenesis. We show that Ent2 regulates hyphal morphogenesis through an interaction between its ENTH domain and the Cdc42 GAP, Rga2, which signals through the Cdc42-Cla4 signaling pathway. Finally, we show that the Ent2 protein, and specifically its ENTH domain, is required for virulence in a mouse model of systemic candidiasis. Overall, this work identifies Ent2 as a key regulator of filamentation and virulence in C. albicans.


Subject(s)
Candida albicans , Fungal Proteins , Humans , Mice , Animals , Fungal Proteins/genetics , Fungal Proteins/metabolism , Virulence , Signal Transduction/genetics , Hyphae , Morphogenesis , Gene Expression Regulation, Fungal
7.
Nat Commun ; 14(1): 4528, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37500616

ABSTRACT

Metabolic flexibility enables fungi to invade challenging host environments. In Candida albicans, a common cause of life-threatening infections in humans, an important contributor to flexibility is alternative oxidase (Aox) activity. Dramatic induction of this activity occurs under respiratory-stress conditions, which impair the classical electron transport chain (ETC). Here, we show that deletion of the inducible AOX2 gene cripples C. albicans virulence in mice by increasing immune recognition. To investigate further, we examined transcriptional regulation of AOX2 in molecular detail under host-relevant, ETC-inhibitory conditions. We found that multiple transcription factors, including Rtg1/Rtg3, Cwt1/Zcf11, and Zcf2, bind and regulate the AOX2 promoter, conferring thousand-fold levels of inducibility to AOX2 in response to distinct environmental stressors. Further dissection of this complex promoter revealed how integration of stimuli ranging from reactive species of oxygen, nitrogen, and sulfur to reduced copper availability is achieved at the transcriptional level to regulate AOX2 induction and enable pathogenesis.


Subject(s)
Oxidoreductases , Transcription Factors , Humans , Animals , Mice , Virulence/genetics , Oxidoreductases/genetics , Oxidoreductases/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Candida albicans/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism
8.
Science ; 381(6657): 502-508, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37535745

ABSTRACT

The mammalian gut secretes a family of multifunctional peptides that affect appetite, intestinal secretions, and motility whereas others regulate the microbiota. We have found that peptide YY (PYY1-36), but not endocrine PYY3-36, acts as an antimicrobial peptide (AMP) expressed by gut epithelial paneth cells (PC). PC-PYY is packaged into secretory granules and is secreted into and retained by surface mucus, which optimizes PC-PYY activity. Although PC-PYY shows some antibacterial activity, it displays selective antifungal activity against virulent Candida albicans hyphae-but not the yeast form. PC-PYY is a cationic molecule that interacts with the anionic surfaces of fungal hyphae to cause membrane disruption and transcriptional reprogramming that selects for the yeast phenotype. Hence, PC-PYY is an antifungal AMP that contributes to the maintenance of gut fungal commensalism.


Subject(s)
Antifungal Agents , Antimicrobial Peptides , Candida , Paneth Cells , Peptide Fragments , Peptide YY , Animals , Antifungal Agents/metabolism , Antimicrobial Peptides/metabolism , Candida/drug effects , Candida/physiology , Paneth Cells/metabolism , Peptide Fragments/metabolism , Peptide YY/metabolism , Symbiosis , Humans , Mice
9.
PLoS Genet ; 5(12): e1000783, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20041210

ABSTRACT

Candida albicans is a normal resident of the gastrointestinal tract and also the most prevalent fungal pathogen of humans. It last shared a common ancestor with the model yeast Saccharomyces cerevisiae over 300 million years ago. We describe a collection of 143 genetically matched strains of C. albicans, each of which has been deleted for a specific transcriptional regulator. This collection represents a large fraction of the non-essential transcription circuitry. A phenotypic profile for each mutant was developed using a screen of 55 growth conditions. The results identify the biological roles of many individual transcriptional regulators; for many, this work represents the first description of their functions. For example, a quarter of the strains showed altered colony formation, a phenotype reflecting transitions among yeast, pseudohyphal, and hyphal cell forms. These transitions, which have been closely linked to pathogenesis, have been extensively studied, yet our work nearly doubles the number of transcriptional regulators known to influence them. As a second example, nearly a quarter of the knockout strains affected sensitivity to commonly used antifungal drugs; although a few transcriptional regulators have previously been implicated in susceptibility to these drugs, our work indicates many additional mechanisms of sensitivity and resistance. Finally, our results inform how transcriptional networks evolve. Comparison with the existing S. cerevisiae data (supplemented by additional S. cerevisiae experiments reported here) allows the first systematic analysis of phenotypic conservation by orthologous transcriptional regulators over a large evolutionary distance. We find that, despite the many specific wiring changes documented between these species, the general phenotypes of orthologous transcriptional regulator knockouts are largely conserved. These observations support the idea that many wiring changes affect the detailed architecture of the circuit, but not its overall output.


Subject(s)
Candida albicans/cytology , Candida albicans/genetics , Gene Regulatory Networks/genetics , Candida albicans/growth & development , Colony Count, Microbial , Databases, Genetic , Gene Knockout Techniques , Gene Library , Homeostasis/genetics , Models, Biological , Phenotype , Saccharomyces cerevisiae/genetics , Transcription, Genetic
10.
PLoS Genet ; 5(9): e1000664, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19779551

ABSTRACT

Localization of specific mRNAs is an important mechanism through which cells achieve polarity and direct asymmetric growth. Based on a framework established in Saccharomyces cerevisiae, we describe a She3-dependent RNA transport system in Candida albicans, a fungal pathogen of humans that grows as both budding (yeast) and filamentous (hyphal and pseudohyphal) forms. We identify a set of 40 mRNAs that are selectively transported to the buds of yeast-form cells and to the tips of hyphae, and we show that many of the genes encoded by these mRNAs contribute to hyphal development, as does the transport system itself. Although the basic system of mRNA transport is conserved between S. cerevisiae and C. albicans, we find that the cargo mRNAs have diverged considerably, implying that specific mRNAs can easily move in and out of transport control over evolutionary timescales. The differences in mRNA cargos likely reflect the distinct selective pressures acting on the two species.


Subject(s)
Candida albicans/growth & development , Candida albicans/metabolism , Hyphae/cytology , Hyphae/growth & development , RNA Transport , RNA, Fungal/metabolism , Actins/metabolism , Candida albicans/cytology , Candida albicans/genetics , Epithelial Cells/microbiology , Epithelial Cells/pathology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Genes, Fungal , Humans , Hyphae/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics
11.
Science ; 377(6610): 1049, 2022 09 02.
Article in English | MEDLINE | ID: mdl-36048934

ABSTRACT

RNA trailblazer who illuminated splicing mechanics.


Subject(s)
Genetics , RNA Splicing , Genetics/history , History, 20th Century , History, 21st Century , United States
12.
Cell Host Microbe ; 30(11): 1589-1601.e5, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36323314

ABSTRACT

Candida albicans is the most common cause of fungal infection in humans. IL-17 is critical for defense against superficial fungal infections, but the role of this response in invasive disease is less understood. We show that C. albicans secretes a lipase, Lip2, that facilitates invasive disease via lipid-based suppression of the IL-17 response. Lip2 was identified as an essential virulence factor in a forward genetic screen in a mouse model of bloodstream infection. Murine infection with C. albicans strains lacking Lip2 display exaggerated IL-17 responses that lead to fungal clearance from solid organs and host survival. Both IL-17 signaling and lipase activity are required for Lip2-mediated suppression. Lip2 inhibits IL-17 production indirectly by suppressing IL-23 production by tissue-resident dendritic cells. The lipase hydrolysis product, palmitic acid, similarly suppresses dendritic cell activation in vitro. Thus, C. albicans suppresses antifungal IL-17 defense in solid organs by altering the tissue lipid milieu.


Subject(s)
Candida albicans , Interleukin-17 , Humans , Mice , Animals , Candida albicans/genetics , Antifungal Agents/therapeutic use , Lipase/genetics , Lipids , Fungal Proteins
13.
Cell Rep ; 39(7): 110837, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584674

ABSTRACT

Systemic immunity is stringently regulated by commensal intestinal microbes, including the pathobiont Candida albicans. This fungus utilizes various transcriptional and morphological programs for host adaptation, but how this heterogeneity affects immunogenicity remains uncertain. We show that UME6, a transcriptional regulator of filamentation, is essential for intestinal C. albicans-primed systemic Th17 immunity. UME6 deletion and constitutive overexpression strains are non-immunogenic during commensal colonization, whereas immunogenicity is restored by C. albicans undergoing oscillating UME6 expression linked with ß-glucan and mannan production. In turn, intestinal reconstitution with these fungal cell wall components restores protective Th17 immunity to mice colonized with UME6-locked variants. These fungal cell wall ligands and commensal C. albicans stimulate Th17 immunity through multiple host pattern recognition receptors, including Toll-like receptor 2 (TLR2), TLR4, Dectin-1, and Dectin-2, which work synergistically for colonization-induced protection. Thus, dynamic gene expression fluctuations by C. albicans during symbiotic colonization are essential for priming host immunity against disseminated infection.


Subject(s)
Candida albicans , Th17 Cells , Animals , Candida albicans/genetics , Cell Wall , Intestines , Mice , Symbiosis
14.
Cell Host Microbe ; 29(6): 1002-1013.e9, 2021 06 09.
Article in English | MEDLINE | ID: mdl-33915113

ABSTRACT

Candida albicans is a fungal component of the human gut microbiota and an opportunistic pathogen. C. albicans transcription factors (TFs), Wor1 and Efg1, are master regulators of an epigenetic switch required for fungal mating that also control colonization of the mammalian gut. We show that additional mating regulators, WOR2, WOR3, WOR4, AHR1, CZF1, and SSN6, also influence gut commensalism. Using Calling Card-seq to record Candida TF DNA-binding events in the host, we examine the role and relationships of these regulators during murine gut colonization. By comparing in-host transcriptomes of regulatory mutants with enhanced versus diminished commensal fitness, we also identify a set of candidate commensalism effectors. These include Cht2, a GPI-linked chitinase whose gene is bound by Wor1, Czf1, and Efg1 in vivo, that we show promotes commensalism. Thus, the network required for a C. albicans sexual switch is biochemically active in the host intestine and repurposed to direct commensalism.


Subject(s)
Candida albicans/genetics , Candida albicans/metabolism , DNA-Binding Proteins/physiology , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Fungal , Symbiosis , Transcription Factors/physiology , Animals , Female , Fungal Proteins/physiology , Genes, Mating Type, Fungal , Genes, Switch , High-Throughput Screening Assays , Host Microbial Interactions , Mice , Mice, Inbred BALB C , Models, Animal , Mutation , Transcriptome
15.
Nat Commun ; 12(1): 6497, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764269

ABSTRACT

Fungal pathogens pose a global threat to human health, with Candida albicans among the leading killers. Systematic analysis of essential genes provides a powerful strategy to discover potential antifungal targets. Here, we build a machine learning model to generate genome-wide gene essentiality predictions for C. albicans and expand the largest functional genomics resource in this pathogen (the GRACE collection) by 866 genes. Using this model and chemogenomic analyses, we define the function of three uncharacterized essential genes with roles in kinetochore function, mitochondrial integrity, and translation, and identify the glutaminyl-tRNA synthetase Gln4 as the target of N-pyrimidinyl-ß-thiophenylacrylamide (NP-BTA), an antifungal compound.


Subject(s)
Machine Learning , Antifungal Agents/pharmacology , Candida albicans/drug effects , Genome-Wide Association Study , Kinetochores/metabolism , Systems Biology/methods
16.
J Vis Exp ; (153)2019 11 05.
Article in English | MEDLINE | ID: mdl-31762460

ABSTRACT

Candida albicans is a fungal component of the gut microbiota in humans and many other mammals. Although C. albicans does not cause symptoms in most colonized hosts, the commensal reservoir does serve as a repository for infectious disease, and the presence of high fungal titers in the gut is associated with inflammatory bowel disease. Here, we describe a method to visualize C. albicans cell morphology and localization in a mouse model of stable gastrointestinal colonization. Colonization is established using a single dose of C. albicans in animals that have been treated with oral antibiotics. Segments of gut tissue are fixed in a manner that preserves the architecture of luminal contents (microorganisms and mucus) as well as the host mucosa. Finally, fluorescent in situ hybridization is performed using probes against fungal rRNA to stain for C. albicans and hyphae. A key advantage of this protocol is that it allows for simultaneous observation of C. albicans cell morphology and its spatial association with host structures during gastrointestinal colonization.


Subject(s)
Candida albicans/isolation & purification , Gastrointestinal Tract/microbiology , In Situ Hybridization, Fluorescence/methods , Animals , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Gastrointestinal Microbiome , Mice , Mucous Membrane , Symbiosis
17.
Cell Host Microbe ; 25(3): 432-443.e6, 2019 Mar 13.
Article in English | MEDLINE | ID: mdl-30870623

ABSTRACT

Candida albicans is a gut commensal and opportunistic pathogen. The transition between yeast and invasive hyphae is central to virulence but has unknown functions during commensal growth. In a mouse model of colonization, yeast and hyphae co-occur throughout the gastrointestinal tract. However, competitive infections of C. albicans homozygous gene disruption mutants revealed an unanticipated, inhibitory role for the yeast-to-hypha morphogenesis program on commensalism. We show that the transcription factor Ume6, a master regulator of filamentation, inhibits gut colonization, not by effects on cell shape, but by activating the expression of a hypha-specific pro-inflammatory secreted protease, Sap6, and a hyphal cell surface adhesin, Hyr1. Like a ume6 mutant, strains lacking SAP6 exhibit enhanced colonization fitness, whereas SAP6-overexpression strains are attenuated in the gut. These results reveal a tradeoff between fungal programs supporting commensalism and virulence in which selection against hypha-specific markers limits the disease-causing potential of this ubiquitous commensal-pathogen.


Subject(s)
Candida albicans/growth & development , Candida albicans/pathogenicity , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Fungal , Symbiosis , Animals , Candida albicans/cytology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Hyphae/cytology , Hyphae/growth & development , Mice , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence
18.
Mol Biol Cell ; 16(10): 4814-26, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16030247

ABSTRACT

Here, we investigate how Candida albicans, the most prevalent human fungal pathogen, protects itself from nitric oxide (*NO), an antimicrobial compound produced by the innate immune system. We show that exposure of C. albicans to *NO elicits a reproducible and specific transcriptional response as determined by genome-wide microarray analysis. Many genes are transiently induced or repressed by *NO, whereas a set of nine genes remain at elevated levels during *NO exposure. The most highly induced gene in this latter category is YHB1, a flavohemoglobin that detoxifies *NO in C. albicans and other microbes. We show that C. albicans strains deleted for YHB1 have two phenotypes in vitro; they are hypersensitive to *NO and they are hyperfilamentous. In a mouse model of disseminated candidiasis, a YHB1 deleted C. albicans strain shows moderately attenuated virulence, but the virulence defect is not suppressed by deletion of the host NOS2 gene. These results suggest that *NO production is not a prime determinant of virulence in the mouse tail vein model of candidiasis and that the attenuated virulence of a yhb1delta/yhb1delta strain is attributable to a defect other than its reduced ability to detoxify *NO.


Subject(s)
Candida albicans/metabolism , Fungal Proteins/metabolism , Nitric Oxide/physiology , Alkenes/pharmacology , Animals , Candida albicans/genetics , Candida albicans/pathogenicity , Candidiasis/metabolism , Fungal Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal , Genome, Fungal , Male , Mice , Mice, Knockout , Mutation , Nitric Oxide Donors/pharmacology , Oligonucleotide Array Sequence Analysis , Transcription, Genetic , Virulence
19.
mBio ; 9(4)2018 08 21.
Article in English | MEDLINE | ID: mdl-30131363

ABSTRACT

The innate immune system is the first line of defense against invasive fungal infections. As a consequence, many successful fungal pathogens have evolved elegant strategies to interact with host immune cells. For example, Candida albicans undergoes a morphogenetic switch coupled to cell wall remodeling upon phagocytosis by macrophages and then induces macrophage pyroptosis, an inflammatory cell death program. To elucidate the genetic circuitry through which C. albicans orchestrates this host response, we performed the first large-scale analysis of C. albicans interactions with mammalian immune cells. We identified 98 C. albicans genes that enable macrophage pyroptosis without influencing fungal cell morphology in the macrophage, including specific determinants of cell wall biogenesis and the Hog1 signaling cascade. Using these mutated genes, we discovered that defects in the activation of pyroptosis affect immune cell recruitment during infection. Examining host circuitry required for pyroptosis in response to C. albicans infection, we discovered that inflammasome priming and activation can be decoupled. Finally, we observed that apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization can occur prior to phagolysosomal rupture by C. albicans hyphae, demonstrating that phagolysosomal rupture is not the inflammasome activating signal. Taking the data together, this work defines genes that enable fungal cell wall remodeling and activation of macrophage pyroptosis independently of effects on morphogenesis and identifies macrophage signaling components that are required for pyroptosis in response to C. albicans infection.IMPORTANCECandida albicans is a natural member of the human mucosal microbiota that can also cause superficial infections and life-threatening systemic infections, both of which are characterized by inflammation. Host defense relies mainly on the ingestion and destruction of C. albicans by innate immune cells, such as macrophages and neutrophils. Although some C. albicans cells are killed by macrophages, most undergo a morphological change and escape by inducing macrophage pyroptosis. Here, we investigated the C. albicans genes and host factors that promote macrophage pyroptosis in response to intracellular fungi. This work provides a foundation for understanding how host immune cells interact with C. albicans and may lead to effective strategies to modulate inflammation induced by fungal infections.


Subject(s)
Candida albicans/genetics , Genes, Fungal , Host-Pathogen Interactions , Macrophages/microbiology , Pyroptosis , Animals , Candida albicans/pathogenicity , Female , High-Throughput Screening Assays , Immune Evasion , Macrophages/pathology , Mice , Mice, Inbred C57BL , Phagocytosis
20.
Nat Rev Microbiol ; 15(2): 96-108, 2017 02.
Article in English | MEDLINE | ID: mdl-27867199

ABSTRACT

Candida albicans is a ubiquitous commensal of the mammalian microbiome and the most prevalent fungal pathogen of humans. A cell-type transition between yeast and hyphal morphologies in C. albicans was thought to underlie much of the variation in virulence observed in different host tissues. However, novel yeast-like cell morphotypes, including opaque(a/α), grey and gastrointestinally induced transition (GUT) cell types, were recently reported that exhibit marked differences in vitro and in animal models of commensalism and disease. In this Review, we explore the characteristics of the classic cell types - yeast, hyphae, pseudohyphae and chlamydospores - as well as the newly identified yeast-like morphotypes. We highlight emerging knowledge about the associations of these different morphotypes with different host niches and virulence potential, as well as the environmental cues and signalling pathways that are involved in the morphological transitions.


Subject(s)
Candida albicans/pathogenicity , Cell Plasticity/physiology , Gastrointestinal Microbiome , Host-Pathogen Interactions/physiology , Hyphae/physiology , Gene Expression Regulation, Fungal , Humans , Signal Transduction , Symbiosis , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL