Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Science ; 381(6657): 502-508, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37535745

RESUMO

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.


Assuntos
Antifúngicos , Peptídeos Antimicrobianos , Candida , Celulas de Paneth , Fragmentos de Peptídeos , Peptídeo YY , Animais , Antifúngicos/metabolismo , Peptídeos Antimicrobianos/metabolismo , Candida/efeitos dos fármacos , Candida/fisiologia , Celulas de Paneth/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Simbiose , Humanos , Camundongos
2.
Nat Commun ; 14(1): 4528, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37500616

RESUMO

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.


Assuntos
Oxirredutases , Fatores de Transcrição , Humanos , Animais , Camundongos , Virulência/genética , Oxirredutases/genética , Oxirredutases/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Candida albicans/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo
3.
mBio ; 14(2): e0343422, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36809010

RESUMO

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.


Assuntos
Candida albicans , Proteínas Fúngicas , Humanos , Camundongos , Animais , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Virulência , Transdução de Sinais/genética , Hifas , Morfogênese , Regulação Fúngica da Expressão Gênica
4.
Cell Host Microbe ; 30(11): 1589-1601.e5, 2022 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-36323314

RESUMO

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.


Assuntos
Candida albicans , Interleucina-17 , Humanos , Camundongos , Animais , Candida albicans/genética , Antifúngicos/uso terapêutico , Lipase/genética , Lipídeos , Proteínas Fúngicas
5.
Cell Rep ; 39(7): 110837, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35584674

RESUMO

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.


Assuntos
Candida albicans , Células Th17 , Animais , Candida albicans/genética , Parede Celular , Intestinos , Camundongos , Simbiose
6.
Nature ; 596(7870): 114-118, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34262174

RESUMO

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.


Assuntos
Imunidade Adaptativa , Candida albicans/imunologia , Candida albicans/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Simbiose/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos de Fungos/imunologia , Candida albicans/patogenicidade , Colite/imunologia , Colite/microbiologia , Colite/patologia , Feminino , Vacinas Fúngicas/imunologia , Microbioma Gastrointestinal/imunologia , Humanos , Hifas/imunologia , Imunoglobulina A/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem
7.
Cell Host Microbe ; 29(6): 1002-1013.e9, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-33915113

RESUMO

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.


Assuntos
Candida albicans/genética , Candida albicans/metabolismo , Proteínas de Ligação a DNA/fisiologia , Trato Gastrointestinal/microbiologia , Regulação Fúngica da Expressão Gênica , Simbiose , Fatores de Transcrição/fisiologia , Animais , Feminino , Proteínas Fúngicas/fisiologia , Genes Fúngicos Tipo Acasalamento , Genes de Troca , Ensaios de Triagem em Larga Escala , Interações entre Hospedeiro e Microrganismos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Mutação , Transcriptoma
8.
J Vis Exp ; (153)2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31762460

RESUMO

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.


Assuntos
Candida albicans/isolamento & purificação , Trato Gastrointestinal/microbiologia , Hibridização in Situ Fluorescente/métodos , Animais , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Microbioma Gastrointestinal , Camundongos , Mucosa , Simbiose
9.
PLoS Biol ; 17(6): e3000331, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31226107

RESUMO

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.


Assuntos
Proteína Centromérica A/metabolismo , Segregação de Cromossomos/fisiologia , Histonas/metabolismo , Aneugênicos/metabolismo , Aneuploidia , Candida albicans/genética , Candida albicans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrômero/metabolismo , Proteína Centromérica A/fisiologia , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Histonas/fisiologia , Cinetocoros/metabolismo , Meiose , Mitose , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo
10.
Cell Host Microbe ; 25(3): 432-443.e6, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30870623

RESUMO

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.


Assuntos
Candida albicans/crescimento & desenvolvimento , Candida albicans/patogenicidade , Trato Gastrointestinal/microbiologia , Regulação Fúngica da Expressão Gênica , Simbiose , Animais , Candida albicans/citologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Hifas/citologia , Hifas/crescimento & desenvolvimento , Camundongos , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Virulência
11.
mBio ; 9(4)2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30131363

RESUMO

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.


Assuntos
Candida albicans/genética , Genes Fúngicos , Interações Hospedeiro-Patógeno , Macrófagos/microbiologia , Piroptose , Animais , Candida albicans/patogenicidade , Feminino , Ensaios de Triagem em Larga Escala , Evasão da Resposta Imune , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose
12.
Nat Commun ; 8(1): 599, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28928442

RESUMO

Infectious diseases have a profound impact on our health and many studies suggest that host genetics play a major role in the pathogenesis of most of them. We perform 23 genome-wide association studies for common infections and infection-associated procedures, including chickenpox, shingles, cold sores, mononucleosis, mumps, hepatitis B, plantar warts, positive tuberculosis test results, strep throat, scarlet fever, pneumonia, bacterial meningitis, yeast infections, urinary tract infections, tonsillectomy, childhood ear infections, myringotomy, measles, hepatitis A, rheumatic fever, common colds, rubella and chronic sinus infection, in over 200,000 individuals of European ancestry. We detect 59 genome-wide significant (P < 5 × 10-8) associations in genes with key roles in immunity and embryonic development. We apply fine-mapping analysis to dissect associations in the human leukocyte antigen region, which suggests important roles of specific amino acid polymorphisms in the antigen-binding clefts. Our findings provide an important step toward dissecting the host genetic architecture of response to common infections.Susceptibility to infectious diseases is, among others, influenced by the genetic landscape of the host. Here, Tian and colleagues perform genome-wide association studies for 23 common infections and find 59 risk loci for 17 of these, both within the HLA region and non-HLA loci.


Assuntos
Antígenos HLA/genética , Infecções/genética , População Branca/genética , Candidíase Vulvovaginal/genética , Estudos de Casos e Controles , Varicela/genética , Doença Crônica , Resfriado Comum/genética , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Hepatite A/genética , Hepatite B/genética , Herpes Labial/genética , Herpes Zoster/genética , Humanos , Mononucleose Infecciosa/genética , Masculino , Sarampo/genética , Meningites Bacterianas/genética , Ventilação da Orelha Média , Caxumba/genética , Otite Média/genética , Otite Média/cirurgia , Faringite/genética , Pneumonia/genética , Febre Reumática/genética , Rubéola (Sarampo Alemão)/genética , Escarlatina/genética , Sinusite/genética , Infecções Estreptocócicas/genética , Tonsilectomia , Tonsilite/genética , Tonsilite/cirurgia , Teste Tuberculínico , Tuberculose/diagnóstico , Tuberculose/genética , Infecções Urinárias/genética , Verrugas/genética
13.
Nat Rev Microbiol ; 15(2): 96-108, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27867199

RESUMO

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.


Assuntos
Candida albicans/patogenicidade , Plasticidade Celular/fisiologia , Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno/fisiologia , Hifas/fisiologia , Regulação Fúngica da Expressão Gênica , Humanos , Transdução de Sinais , Simbiose , Virulência
14.
J Bacteriol ; 196(9): 1683-93, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24532776

RESUMO

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.


Assuntos
Proteínas de Bactérias/genética , Elementos de DNA Transponíveis , Mutagênese Insercional , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Proteínas de Bactérias/metabolismo , Cromatografia Líquida , Fenazinas/metabolismo , Pseudomonas aeruginosa/química , Espectrometria de Massas em Tandem
15.
Curr Opin Microbiol ; 16(6): 708-15, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24121029

RESUMO

Candida albicans is a fungal commensal-pathogen that persistently associates with its mammalian hosts. Between the commensal and pathogenic lifestyles, this microorganism inhabits host niches that differ markedly in the levels of bioavailable iron. A number of recent studies have exposed C. albicans specializations for acquiring iron from specific host molecules in regions where iron is scarce, while also defending against iron-related toxicity in regions where iron occurs in surfeit. Together, these results point to a central role for iron homeostasis in the evolution of this important human pathogen.


Assuntos
Candida albicans/metabolismo , Homeostase , Ferro/metabolismo , Simbiose , Virulência
16.
Nat Genet ; 45(9): 1088-91, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23892606

RESUMO

Among ∼5,000,000 fungal species, C. albicans is exceptional in its lifelong association with humans, either within the gastrointestinal microbiome or as an invasive pathogen. Opportunistic infections are generally ascribed to defective host immunity but may require specific microbial programs. Here we report that exposure of C. albicans to the mammalian gut triggers a developmental switch, driven by the Wor1 transcription factor, to a commensal cell type. Wor1 expression was previously observed only in rare genetic backgrounds, where it controls a white-opaque switch in mating. We show that passage of wild-type cells through the mouse gastrointestinal tract triggers WOR1 expression and a novel phenotypic switch. The resulting GUT (gastrointestinally induced transition) cells differ morphologically and functionally from previously defined cell types, including opaque cells, and express a transcriptome that is optimized for the digestive tract. The white-GUT switch illuminates how a microorganism can use distinct genetic programs to transition between commensalism and invasive pathogenesis.


Assuntos
Candida albicans/genética , Candida albicans/metabolismo , Trato Gastrointestinal/microbiologia , Fenótipo , Animais , Candida albicans/ultraestrutura , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Perfilação da Expressão Gênica , Regulação Fúngica da Expressão Gênica , Aptidão Genética , Humanos , Camundongos , Mutação
17.
PLoS Pathog ; 8(11): e1002956, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133381

RESUMO

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.


Assuntos
Candida albicans/metabolismo , Candida albicans/patogenicidade , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica/fisiologia , Candida albicans/genética , Candidíase/genética , Candidíase/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Proteínas Fúngicas/genética , Humanos , Ferro/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mutação
18.
Cell Host Microbe ; 10(2): 118-35, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21843869

RESUMO

The mammalian gastrointestinal tract and bloodstream are highly disparate biological niches that differ in concentrations of nutrients such as iron. However, some commensal-pathogenic microorganisms, such as the yeast Candida albicans, thrive in both environments. We report the evolution of a transcription circuit in C. albicans that controls iron uptake and determines its fitness in both niches. Our analysis of DNA-binding proteins that regulate iron uptake by this organism suggests the evolutionary intercalation of a transcriptional activator called Sef1 between two broadly conserved iron-responsive transcriptional repressors, Sfu1 and Hap43. Sef1 activates iron-uptake genes and promotes virulence in a mouse model of bloodstream infection, whereas Sfu1 represses iron-uptake genes and is dispensable for virulence but promotes gastrointestinal commensalism. Thus, C. albicans can alternate between genetic programs conferring resistance to iron depletion in the bloodstream versus iron toxicity in the gut, and this may represent a fundamental attribute of gastrointestinal commensal-pathogens.


Assuntos
Candida albicans/patogenicidade , Proteínas Fúngicas/metabolismo , Homeostase , Ferro/metabolismo , Simbiose , Alelos , Animais , Candida albicans/genética , Candida albicans/crescimento & desenvolvimento , Imunoprecipitação da Cromatina/métodos , Feminino , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Filogenia , Plasma/metabolismo , Plasma/microbiologia , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional , Virulência
19.
Methods Enzymol ; 470: 737-58, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20946834

RESUMO

Candida albicans is an opportunistic fungal pathogen of humans. Although a normal part of our gastrointestinal flora, C. albicans has the ability to colonize nearly every human tissue and organ, causing serious, invasive infections. In this chapter we describe current methodologies used in molecular genetic studies of this organism. These techniques include rapid sequential gene disruption, DNA transformation, RNA isolation, epitope tagging, and chromatin immunoprecipitation. The ease of these techniques, combined with the high-quality C. albicans genome sequences now available, have greatly facilitated research into this important pathogen. Candida albicans is a normal resident of the human gastrointestinal tract; it is also the most common fungal pathogen of humans, causing both mucosal and systemic infections, particularly in immune compromised patients. C. albicans and Saccharomyces cerevisiae last shared a common ancestor more than 900 million years ago; in terms of conserved coding sequences, the two species are approximately as divergent as fish and humans. Although C. albicans and S. cerevisiae share certain core features, they also exhibit many significant differences. This is not surprising as C. albicans has the ability to survive in nearly every niche of a mammalian host, a property not shared by S. cerevisiae. Research into C. albicans is important in its own right, particularly with regards to its ability to cause disease in humans; in addition, comparison with S. cerevisiae can reveal important insights into evolutionary processes. Many of the methodologies developed for use in S. cerevisiae have been adapted for C. albicans, and we describe some of the most common. Although alternative procedures are described in the literature, we have found those described below to be the most convenient. Because the C. albicans parasexual cycle is cumbersome to use in the laboratory, genetics in this organism has been based almost entirely on directed mutations. Because the organism is diploid, creating a deletion mutant requires two rounds of gene disruption. We describe a rapid method for creating sequential disruptions, one which can be scaled up to create large collections of C. albicans deletion mutants. We also describe a series of additional techniques including DNA transformation, mRNA isolation, epitope tagging, and chromatin immunoprecipitation (ChIP). The ease of these techniques, combined with the high-quality C. albicans genome sequences now available, has greatly increased the quality and pace of research into this important pathogen.


Assuntos
Candida albicans/metabolismo , Candida albicans/genética , Imunoprecipitação da Cromatina , Regulação Fúngica da Expressão Gênica/genética , Regulação Fúngica da Expressão Gênica/fisiologia , Reação em Cadeia da Polimerase
20.
Nat Genet ; 42(7): 590-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20543849

RESUMO

Candida albicans is the most common cause of serious fungal disease in humans. Creation of isogenic null mutants of this diploid organism, which requires sequential gene targeting, allows dissection of virulence mechanisms. Published analyses of such mutants show a near-perfect correlation between C. albicans pathogenicity and the ability to undergo a yeast-to-hypha morphological switch in vitro. However, most studies have used mutants constructed with a marker that is itself a virulence determinant and therefore complicates their interpretation. Using alternative markers, we created approximately 3,000 homozygous deletion strains affecting 674 genes, or roughly 11% of the C. albicans genome. Screening for infectivity in a mouse model and for morphological switching and cell proliferation in vitro, we identified 115 infectivity-attenuated mutants, of which nearly half demonstrated normal morphological switching and proliferation. Analysis of such mutants revealed that virulence requires the glycolipid glucosylceramide. To our knowledge, this is the first C. albicans small molecule that has been found to be required specifically for virulence.


Assuntos
Candida albicans/genética , Candidíase/microbiologia , Deleção de Genes , Biblioteca Gênica , Animais , Candida albicans/crescimento & desenvolvimento , Candida albicans/patogenicidade , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glucosilceramidas/biossíntese , Glucosilceramidas/química , Homozigoto , Humanos , Hifas/genética , Hifas/crescimento & desenvolvimento , Hifas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Morfogênese , Mutação , Análise de Sobrevida , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA