Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 627(8004): 620-627, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38448595

RESUMO

The fungus Candida albicans frequently colonizes the human gastrointestinal tract, from which it can disseminate to cause systemic disease. This polymorphic species can transition between growing as single-celled yeast and as multicellular hyphae to adapt to its environment. The current dogma of C. albicans commensalism is that the yeast form is optimal for gut colonization, whereas hyphal cells are detrimental to colonization but critical for virulence1-3. Here, we reveal that this paradigm does not apply to multi-kingdom communities in which a complex interplay between fungal morphology and bacteria dictates C. albicans fitness. Thus, whereas yeast-locked cells outcompete wild-type cells when gut bacteria are absent or depleted by antibiotics, hyphae-competent wild-type cells outcompete yeast-locked cells in hosts with replete bacterial populations. This increased fitness of wild-type cells involves the production of hyphal-specific factors including the toxin candidalysin4,5, which promotes the establishment of colonization. At later time points, adaptive immunity is engaged, and intestinal immunoglobulin A preferentially selects against hyphal cells1,6. Hyphal morphotypes are thus under both positive and negative selective pressures in the gut. Our study further shows that candidalysin has a direct inhibitory effect on bacterial species, including limiting their metabolic output. We therefore propose that C. albicans has evolved hyphal-specific factors, including candidalysin, to better compete with bacterial species in the intestinal niche.


Assuntos
Candida albicans , Proteínas Fúngicas , Microbioma Gastrointestinal , Hifas , Intestinos , Micotoxinas , Simbiose , Animais , Feminino , Humanos , Masculino , Camundongos , Bactérias/crescimento & desenvolvimento , Bactérias/imunologia , Candida albicans/crescimento & desenvolvimento , Candida albicans/imunologia , Candida albicans/metabolismo , Candida albicans/patogenicidade , Proteínas Fúngicas/metabolismo , Microbioma Gastrointestinal/imunologia , Hifas/crescimento & desenvolvimento , Hifas/imunologia , Hifas/metabolismo , Imunoglobulina A/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Micotoxinas/metabolismo , Virulência
2.
Proc Natl Acad Sci U S A ; 120(11): e2218163120, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36893271

RESUMO

Aneuploidy is a frequent occurrence in fungal species where it can alter gene expression and promote adaptation to a variety of environmental cues. Multiple forms of aneuploidy have been observed in the opportunistic fungal pathogen Candida albicans, which is a common component of the human gut mycobiome but can escape this niche and cause life-threatening systemic disease. Using a barcode sequencing (Bar-seq) approach, we evaluated a set of diploid C. albicans strains and found that a strain carrying a third copy of chromosome (Chr) 7 was associated with increased fitness during both gastrointestinal (GI) colonization and systemic infection. Our analysis revealed that the presence of a Chr 7 trisomy resulted in decreased filamentation, both in vitro and during GI colonization, relative to isogenic euploid controls. A target gene approach demonstrated that NRG1, encoding a negative regulator of filamentation located on Chr 7, contributes to increased fitness of the aneuploid strain due to inhibition of filamentation in a gene dosage-dependent fashion. Together, these experiments establish how aneuploidy enables the reversible adaptation of C. albicans to its host via gene dosage-dependent regulation of morphology.


Assuntos
Candida albicans , Trato Gastrointestinal , Humanos , Candida albicans/metabolismo , Trato Gastrointestinal/microbiologia , Dosagem de Genes , Aneuploidia , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica
3.
PLoS Pathog ; 19(12): e1011833, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38091321

RESUMO

The ability of the fungus Candida albicans to filament and form biofilms contributes to its burden as a leading cause of hospital-acquired infections. Biofilm development involves an interconnected transcriptional regulatory network (TRN) consisting of nine transcription factors (TFs) that bind both to their own regulatory regions and to those of the other network TFs. Here, we show that seven of the nine TFs in the C. albicans biofilm network contain prion-like domains (PrLDs) that have been linked to the ability to form phase-separated condensates. Construction of PrLD mutants in four biofilm TFs reveals that these domains are essential for filamentation and biofilm formation in C. albicans. Moreover, biofilm PrLDs promote the formation of phase-separated condensates in the nuclei of live cells, and PrLD mutations that abolish phase separation (such as the removal of aromatic residues) also prevent biofilm formation. Biofilm TF condensates can selectively recruit other TFs through PrLD-PrLD interactions and can co-recruit RNA polymerase II, implicating condensate formation in the assembly of active transcriptional complexes. Finally, we show that PrLD mutations that block the phase separation of biofilm TFs also prevent filamentation in an in vivo model of gastrointestinal colonization. Together, these studies associate transcriptional condensates with the regulation of filamentation and biofilm formation in C. albicans, and highlight how targeting of PrLD-PrLD interactions could prevent pathogenesis by this species.


Assuntos
Candida albicans , Fatores de Transcrição , Candida albicans/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Hifas , Biofilmes , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo
4.
Trends Immunol ; 43(9): 706-717, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35961916

RESUMO

Mucosal tissues are constitutively colonized by a wide assortment of host-adapted microbes. This includes the polymorphic fungus Candida albicans which is a primary target of human adaptive responses. Immunogenicity is replicated after intestinal colonization in preclinical models with a surprising array of protective benefits for most hosts, but harmful consequences for a few. The interaction between fungus and host is complex, and traditionally, the masking of antigenic fungal ligands has been viewed as a tactic for fungal immune evasion during invasive infection. However, we propose that dynamic expression of cell wall moieties, host cell lysins, and other antigenic C. albicans determinants is necessary during the more ubiquitous context of intestinal colonization to prime immunogenicity and optimize mammalian host symbiosis.


Assuntos
Candida albicans , Simbiose , Animais , Parede Celular , Humanos , Evasão da Resposta Imune , Mamíferos
5.
EMBO J ; 38(19): e101597, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31448850

RESUMO

Understanding how cellular activities impact genome stability is critical to multiple biological processes including tumorigenesis and reproductive biology. The fungal pathogen Candida albicans displays striking genome dynamics during its parasexual cycle as tetraploid cells, but not diploid cells, exhibit genome instability and reduce their ploidy when grown on a glucose-rich "pre-sporulation" medium. Here, we reveal that C. albicans tetraploid cells are metabolically hyperactive on this medium with higher rates of fermentation and oxidative respiration relative to diploid cells. This heightened metabolism results in elevated levels of reactive oxygen species (ROS), activation of the ROS-responsive transcription factor Cap1, and the formation of DNA double-strand breaks. Genetic or chemical suppression of ROS levels suppresses each of these phenotypes and also protects against genome instability. These studies reveal how endogenous metabolic processes can generate sufficient ROS to trigger genome instability in polyploid C. albicans cells. We also discuss potential parallels with metabolism-induced instability in cancer cells and speculate that ROS-induced DNA damage could have facilitated ploidy cycling prior to a conventional meiosis in eukaryotes.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Candida albicans/fisiologia , Proteínas de Ciclo Celular/genética , Dano ao DNA , Proteínas Fúngicas/genética , Instabilidade Genômica , Fermentação , Perfilação da Expressão Gênica , Regulação Fúngica da Expressão Gênica , Genoma Fúngico , Metabolômica , Estresse Oxidativo , Poliploidia , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima
6.
PLoS Genet ; 14(5): e1007377, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29734333

RESUMO

Sexual reproduction is a universal mechanism for generating genetic diversity in eukaryotes. Fungi exhibit diverse strategies for sexual reproduction both in nature and in the laboratory. In this study, we report the discovery of same-sex (homothallic) mating in the human fungal pathogen Candida tropicalis. We show that same-sex mating occurs between two cells carrying the same mating type (MTLa/a or α/α) and requires the presence of pheromone from the opposite mating type as well as the receptor for this pheromone. In ménage à trois mating mixes (i.e., "a x a + α helper" or "α x α + a helper" mixes), pheromone secreted by helper strains promotes diploid C. tropicalis cells to undergo same-sex mating and form tetraploid products. Surprisingly, however, the tetraploid mating products can then efficiently mate with cells of the opposite mating type to generate hexaploid products. The unstable hexaploid progeny generated from this coupled process of same- and opposite-sex mating undergo rapid chromosome loss and generate extensive genetic variation. Phenotypic analysis demonstrated that the mating progeny-derived strains exhibit diverse morphologies and phenotypes, including differences in secreted aspartic proteinase (Sap) activity and susceptibility to the antifungal drugs. Thus, the coupling of same- and opposite-sex mating represents a novel mode to generate polyploidy and genetic diversity, which may facilitate the evolution of new traits in C. tropicalis and adaptation to changing environments.


Assuntos
Candida tropicalis/genética , Genes Fúngicos Tipo Acasalamento/genética , Variação Genética , Poliploidia , Candida tropicalis/metabolismo , Candida tropicalis/fisiologia , Candidíase/microbiologia , Cruzamentos Genéticos , Diploide , Genótipo , Humanos , Fenótipo , Feromônios/metabolismo , Tetraploidia
7.
Proc Natl Acad Sci U S A ; 115(37): E8688-E8697, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30150418

RESUMO

Candida albicans is a heterozygous diploid yeast that is a commensal of the human gastrointestinal tract and a prevalent opportunistic pathogen. Here, whole-genome sequencing was performed on multiple C. albicans isolates passaged both in vitro and in vivo to characterize the complete spectrum of mutations arising in laboratory culture and in the mammalian host. We establish that, independent of culture niche, microevolution is primarily driven by de novo base substitutions and frequent short-tract loss-of-heterozygosity events. An average base-substitution rate of ∼1.2 × 10-10 per base pair per generation was observed in vitro, with higher rates inferred during host infection. Large-scale chromosomal changes were relatively rare, although chromosome 7 trisomies frequently emerged during passaging in a gastrointestinal model and was associated with increased fitness for this niche. Multiple chromosomal features impacted mutational patterns, with mutation rates elevated in repetitive regions, subtelomeric regions, and in gene families encoding cell surface proteins involved in host adhesion. Strikingly, de novo mutation rates were more than 800-fold higher in regions immediately adjacent to emergent loss-of-heterozygosity tracts, indicative of recombination-induced mutagenesis. Furthermore, genomes showed biased patterns of mutations suggestive of extensive purifying selection during passaging. These results reveal how both cell-intrinsic and cell-extrinsic factors influence C. albicans microevolution, and provide a quantitative picture of genome dynamics in this heterozygous diploid species.


Assuntos
Candida albicans/genética , Diploide , Evolução Molecular , Mutação , Candida albicans/crescimento & desenvolvimento , Candidíase/microbiologia , Cromossomos Fúngicos , Genoma Fúngico/genética , Heterozigoto , Humanos , Perda de Heterozigosidade , Seleção Genética
8.
Nature ; 506(7488): 387-390, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24390351

RESUMO

Sexual reproduction is restricted to eukaryotic species and involves the fusion of haploid gametes to form a diploid cell that subsequently undergoes meiosis to generate recombinant haploid forms. This process has been extensively studied in the unicellular yeast Saccharomyces cerevisiae, which exhibits separate regulatory control over mating and meiosis. Here we address the mechanism of sexual reproduction in the related hemiascomycete species Candida lusitaniae. We demonstrate that, in contrast to S. cerevisiae, C. lusitaniae exhibits a highly integrated sexual program in which the programs regulating mating and meiosis have fused. Profiling of the C. lusitaniae sexual cycle revealed that gene expression patterns during mating and meiosis were overlapping, indicative of co-regulation. This was particularly evident for genes involved in pheromone MAPK signalling, which were highly induced throughout the sexual cycle of C. lusitaniae. Furthermore, genetic analysis showed that the orthologue of IME2, a 'diploid-specific' factor in S. cerevisiae, and STE12, the master regulator of S. cerevisiae mating, were each required for progression through both mating and meiosis in C. lusitaniae. Together, our results establish that sexual reproduction has undergone significant rewiring between S. cerevisiae and C. lusitaniae, and that a concerted sexual cycle operates in C. lusitaniae that is more reminiscent of the distantly related ascomycete, Schizosaccharomyces pombe. We discuss these results in light of the evolution of sexual reproduction in yeast, and propose that regulatory coupling of mating and meiosis has evolved multiple times as an adaptation to promote the haploid lifestyle.


Assuntos
Evolução Biológica , Candida/genética , Candida/fisiologia , Haploidia , Meiose/genética , Meiose/fisiologia , Candida/citologia , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Genes Essenciais/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Feromônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Reprodução/genética , Reprodução/fisiologia , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/citologia , Schizosaccharomyces/fisiologia , Sexo , Fatores de Transcrição/metabolismo
9.
Proc Natl Acad Sci U S A ; 114(52): 13780-13785, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29255038

RESUMO

Several pathogenic Candida species are capable of heritable and reversible switching between two epigenetic states, "white" and "opaque." In Candida albicans, white cells are essentially sterile, whereas opaque cells are mating-proficient. Here, we interrogate the mechanism by which the white-opaque switch regulates sexual fecundity and identify four genes in the pheromone MAPK pathway that are expressed at significantly higher levels in opaque cells than in white cells. These genes encode the ß subunit of the G-protein complex (STE4), the pheromone MAPK scaffold (CST5), and the two terminal MAP kinases (CEK1/CEK2). To define the contribution of each factor to mating, C. albicans white cells were reverse-engineered to express elevated, opaque-like levels of these factors, either singly or in combination. We show that white cells co-overexpressing STE4, CST5, and CEK2 undergo mating four orders of magnitude more efficiently than control white cells and at a frequency approaching that of opaque cells. Moreover, engineered white cells recapitulate the transcriptional and morphological responses of opaque cells to pheromone. These results therefore reveal multiple bottlenecks in pheromone MAPK signaling in white cells and that alleviation of these bottlenecks enables efficient mating by these "sterile" cell types. Taken together, our findings establish that differential expression of several MAPK factors underlies the epigenetic control of mating in C. albicans We also discuss how fitness advantages could have driven the evolution of a toggle switch to regulate sexual reproduction in pathogenic Candida species.


Assuntos
Candida albicans/metabolismo , Epigênese Genética/fisiologia , Regulação Fúngica da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Feromônios/metabolismo , Candida albicans/genética , Feromônios/genética
11.
Nature ; 494(7435): 55-9, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23364695

RESUMO

Candida albicans, the most prevalent human fungal pathogen, is considered to be an obligate diploid that carries recessive lethal mutations throughout the genome. Here we demonstrate that C. albicans has a viable haploid state that can be derived from diploid cells under in vitro and in vivo conditions, and that seems to arise through a concerted chromosome loss mechanism. Haploids undergo morphogenetic changes like those of diploids, including the yeast-hyphal transition, chlamydospore formation and a white-opaque switch that facilitates mating. Haploid opaque cells of opposite mating type mate efficiently to regenerate the diploid form, restoring heterozygosity and fitness. Homozygous diploids arise spontaneously by auto-diploidization, and both haploids and auto-diploids show a similar reduction in fitness, in vitro and in vivo, relative to heterozygous diploids, indicating that homozygous cell types are transient in mixed populations. Finally, we constructed stable haploid strains with multiple auxotrophies that will facilitate molecular and genetic analyses of this important pathogen.


Assuntos
Candida albicans/citologia , Candida albicans/genética , Diploide , Haploidia , Sexo , Animais , Candida albicans/crescimento & desenvolvimento , Candida albicans/patogenicidade , Separação Celular , Citometria de Fluxo , Deleção de Genes , Aptidão Genética , Técnicas Genéticas , Haplótipos , Heterozigoto , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos ICR , Inoculações Seriadas , Estresse Fisiológico , Virulência/genética
12.
PLoS Genet ; 12(10): e1006353, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27711197

RESUMO

Heritable epigenetic changes underlie the ability of cells to differentiate into distinct cell types. Here, we demonstrate that the fungal pathogen Candida tropicalis exhibits multipotency, undergoing stochastic and reversible switching between three cellular states. The three cell states exhibit unique cellular morphologies, growth rates, and global gene expression profiles. Genetic analysis identified six transcription factors that play key roles in regulating cell differentiation. In particular, we show that forced expression of Wor1 or Efg1 transcription factors can be used to manipulate transitions between all three cell states. A model for tristability is proposed in which Wor1 and Efg1 are self-activating but mutually antagonistic transcription factors, thereby forming a symmetrical self-activating toggle switch. We explicitly test this model and show that ectopic expression of WOR1 can induce white-to-hybrid-to-opaque switching, whereas ectopic expression of EFG1 drives switching in the opposite direction, from opaque-to-hybrid-to-white cell states. We also address the stability of induced cell states and demonstrate that stable differentiation events require ectopic gene expression in combination with chromatin-based cues. These studies therefore experimentally test a model of multistate stability and demonstrate that transcriptional circuits act synergistically with chromatin-based changes to drive cell state transitions. We also establish close mechanistic parallels between phenotypic switching in unicellular fungi and cell fate decisions during stem cell reprogramming.


Assuntos
Proteínas de Ligação a DNA/genética , Epigênese Genética , Proteínas Fúngicas/biossíntese , Genes de Troca/genética , Fatores de Transcrição/genética , Candida albicans/genética , Candida albicans/crescimento & desenvolvimento , Candida tropicalis/genética , Candida tropicalis/crescimento & desenvolvimento , Diferenciação Celular/genética , Linhagem da Célula/genética , Cromatina/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas Fúngicas/genética , Regulação da Expressão Gênica no Desenvolvimento , Regulação Fúngica da Expressão Gênica , Fatores de Transcrição/biossíntese
13.
Mol Microbiol ; 105(5): 810-824, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28657681

RESUMO

Transcriptional regulation involves both positive and negative regulatory elements. The Dig1 negative regulators are part of a fungal-specific module that includes a transcription factor (a Ste12 family member) and a Dig1 family member. In Saccharomyces cerevisiae, the post-genome-duplication Dig1/Dig2 proteins regulate MAP kinase controlled signalling pathways involved in mating and filamentous growth. We have identified the single Dig1 orthologue in the fungal pathogen Candida albicans. Genetic studies and transcriptional profiling experiments show that this single protein is implicated in the regulation of MAP kinase-controlled processes involved in mating, filamentous growth and biofilm formation, and also influences cAMP-regulated processes. This suggests that the multiple cellular roles of the Dig1 protein are ancestral and predate the sub-functionalization apparent in S. cerevisiae after the genome duplication. Intriguingly, even though loss of Dig1 function in C. albicans enhances filamentous growth and biofilm formation, colonization of the murine gastrointestinal tract is reduced in the mutant. The complexity of the processes influenced by Dig1 in C. albicans, and the observation that Dig1 is one of the few regulatory proteins that were retained in the duplicated state after the whole genome duplication event in yeast, emphasizes the important role of these negative regulators in fungal transcriptional control.


Assuntos
Candida albicans/genética , Candida albicans/metabolismo , Animais , Biofilmes/crescimento & desenvolvimento , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica/genética , Camundongos/microbiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Regiões Promotoras Genéticas/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética
14.
Genome Res ; 25(3): 413-25, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25504520

RESUMO

Candida albicans is a commensal fungus of the human gastrointestinal tract and a prevalent opportunistic pathogen. To examine diversity within this species, extensive genomic and phenotypic analyses were performed on 21 clinical C. albicans isolates. Genomic variation was evident in the form of polymorphisms, copy number variations, chromosomal inversions, subtelomeric hypervariation, loss of heterozygosity (LOH), and whole or partial chromosome aneuploidies. All 21 strains were diploid, although karyotypic changes were present in eight of the 21 isolates, with multiple strains being trisomic for Chromosome 4 or Chromosome 7. Aneuploid strains exhibited a general fitness defect relative to euploid strains when grown under replete conditions. All strains were also heterozygous, yet multiple, distinct LOH tracts were present in each isolate. Higher overall levels of genome heterozygosity correlated with faster growth rates, consistent with increased overall fitness. Genes with the highest rates of amino acid substitutions included many cell wall proteins, implicating fast evolving changes in cell adhesion and host interactions. One clinical isolate, P94015, presented several striking properties including a novel cellular phenotype, an inability to filament, drug resistance, and decreased virulence. Several of these properties were shown to be due to a homozygous nonsense mutation in the EFG1 gene. Furthermore, loss of EFG1 function resulted in increased fitness of P94015 in a commensal model of infection. Our analysis therefore reveals intra-species genetic and phenotypic differences in C. albicans and delineates a natural mutation that alters the balance between commensalism and pathogenicity.


Assuntos
Candida albicans/genética , Variação Genética , Fenótipo , Aneuploidia , Candida albicans/classificação , Candidíase/microbiologia , Cromossomos Fúngicos , Variações do Número de Cópias de DNA , Evolução Molecular , Genoma Fúngico , Genótipo , Humanos , Filogenia , Polimorfismo de Nucleotídeo Único , Seleção Genética , Análise de Sequência de DNA
15.
Biol Chem ; 399(12): 1375-1388, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30367778

RESUMO

Pseudogymnoascus destructans is a pathogenic fungus responsible for White-nose Syndrome (WNS), a disease afflicting multiple species of North American bats. Pseudogymnoascus destructans infects susceptible bats during hibernation, invading dermal tissue and causing extensive tissue damage. In contrast, other Pseudogymnoascus species are non-pathogenic and cross-species comparisons may therefore reveal factors that contribute to virulence. In this study, we compared the secretome of P. destructans with that from several closely related Pseudogymnoascus species. A diverse set of hydrolytic enzymes were identified, including a putative serine peptidase, PdCP1, that was unique to the P. destructans secretome. A recombinant form of PdCP1 was purified and substrate preference determined using a multiplexed-substrate profiling method based on enzymatic degradation of a synthetic peptide library and analysis by mass spectrometry. Most peptide substrates were sequentially truncated from the carboxyl-terminus revealing that this enzyme is a bona fide carboxypeptidase. Peptides with arginine located close to the carboxyl-terminus were rapidly cleaved, and a fluorescent substrate containing arginine was therefore used to characterize PdCP1 activity and to screen a selection of peptidase inhibitors. Antipain and leupeptin were found to be the most potent inhibitors of PdCP1 activity.


Assuntos
Ascomicetos/enzimologia , Carboxipeptidases/metabolismo , Quirópteros/microbiologia , Micoses/metabolismo , Animais , Antipaína/farmacologia , Carboxipeptidases/antagonistas & inibidores , Carboxipeptidases/isolamento & purificação , Leupeptinas/farmacologia , Micoses/microbiologia , Síndrome
16.
Proc Natl Acad Sci U S A ; 112(24): 7478-83, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-25944934

RESUMO

Pseudogymnoascus destructans is the causative agent of white-nose syndrome, a disease that has caused the deaths of millions of bats in North America. This psychrophilic fungus proliferates at low temperatures and targets hibernating bats, resulting in their premature arousal from stupor with catastrophic consequences. Despite the impact of white-nose syndrome, little is known about the fungus itself or how it infects its mammalian host. P. destructans is not amenable to genetic manipulation, and therefore understanding the proteins involved in infection requires alternative approaches. Here, we identify hydrolytic enzymes secreted by P. destructans, and use a novel and unbiased substrate profiling technique to define active peptidases. These experiments revealed that endopeptidases are the major proteolytic activities secreted by P. destructans, and that collagen, the major structural protein in mammals, is actively degraded by the secretome. A serine endopeptidase, hereby-named Destructin-1, was subsequently identified, and a recombinant form overexpressed and purified. Biochemical analysis of Destructin-1 showed that it mediated collagen degradation, and a potent inhibitor of peptidase activity was identified. Treatment of P. destructans-conditioned media with this antagonist blocked collagen degradation and facilitated the detection of additional secreted proteolytic activities, including aminopeptidases and carboxypeptidases. These results provide molecular insights into the secretome of P. destructans, and identify serine endopeptidases that have the clear potential to facilitate tissue invasion and pathogenesis in the mammalian host.


Assuntos
Ascomicetos/enzimologia , Ascomicetos/patogenicidade , Quirópteros/microbiologia , Colagenases/metabolismo , Proteínas Fúngicas/metabolismo , Micoses/veterinária , Sequência de Aminoácidos , Animais , Ascomicetos/genética , Sequência de Bases , Domínio Catalítico , Colagenases/química , Colagenases/genética , DNA Fúngico/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Modelos Moleculares , Dados de Sequência Molecular , Micoses/microbiologia , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Virulência
17.
Microbiology (Reading) ; 163(6): 856-865, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28640746

RESUMO

Candida albicans is an important opportunistic fungal pathogen capable of causing both mucosal and disseminated disease. Infections are often treated with fluconazole, a front-line antifungal drug that targets the biosynthesis of ergosterol, a major component of the fungal cell membrane. Resistance to fluconazole can arise through a variety of mechanisms, including gain-of-function mutations, loss of heterozygosity events and aneuploidy. The clinical isolate P60002 was found to be highly resistant to azole-class drugs, yet lacked mutations or chromosomal rearrangements known to be associated with azole resistance. Transcription profiling suggested that increased expression of two putative drug efflux pumps, CDR11 and QDR1, might confer azole resistance. However, ectopic expression of the P60002 alleles of these genes in a drug-susceptible strain did not increase fluconazole resistance. We next examined whether the presence of three copies of chromosome 4 (Chr4) or chromosome 6 (Chr6) contributed to azole resistance in P60002. We established that Chr4 trisomy contributes significantly to fluconazole resistance, whereas Chr6 trisomy has no discernible effect on resistance. In contrast, a Chr4 trisomy did not increase fluconazole resistance when present in the standard SC5314 strain background. These results establish a link between Chr4 trisomy and elevated fluconazole resistance, and demonstrate the impact of genetic background on drug resistance phenotypes in C. albicans.


Assuntos
Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Candida albicans/genética , Candidíase/microbiologia , Cromossomos Fúngicos/genética , Farmacorresistência Fúngica , Fluconazol/farmacologia , Trissomia , Candida albicans/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Testes de Sensibilidade Microbiana
19.
Eukaryot Cell ; 14(8): 755-74, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26024903

RESUMO

Candida albicans is a major fungal pathogen whose virulence is associated with its ability to transition from a budding yeast form to invasive hyphal filaments. The kinesin-14 family member CaKar3 is required for transition between these morphological states, as well as for mitotic progression and karyogamy. While kinesin-14 proteins are ubiquitous, CaKar3 homologs in hemiascomycete fungi are unique because they form heterodimers with noncatalytic kinesin-like proteins. Thus, CaKar3-based motors may represent a novel antifungal drug target. We have identified and examined the roles of a kinesin-like regulator of CaKar3. We show that orf19.306 (dubbed CaCIK1) encodes a protein that forms a heterodimer with CaKar3, localizes CaKar3 to spindle pole bodies, and can bind microtubules and influence CaKar3 mechanochemistry despite lacking an ATPase activity of its own. Similar to CaKar3 depletion, loss of CaCik1 results in cell cycle arrest, filamentation defects, and an inability to undergo karyogamy. Furthermore, an examination of the spindle structure in cells lacking either of these proteins shows that a large proportion have a monopolar spindle or two dissociated half-spindles, a phenotype unique to the C. albicans kinesin-14 homolog. These findings provide new insights into mitotic spindle structure and kinesin motor function in C. albicans and identify a potentially vulnerable target for antifungal drug development.


Assuntos
Candida albicans/metabolismo , Proteínas Fúngicas/metabolismo , Cinesinas/metabolismo , Morfogênese/fisiologia , Fuso Acromático/metabolismo , Adenosina Trifosfatases/metabolismo , Antifúngicos/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitose/fisiologia
20.
PLoS Genet ; 9(3): e1003369, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23555286

RESUMO

Phenotypic switching allows for rapid transitions between alternative cell states and is important in pathogenic fungi for colonization and infection of different host niches. In Candida albicans, the white-opaque phenotypic switch plays a central role in regulating the program of sexual mating as well as interactions with the mammalian host. White-opaque switching is controlled by genes encoded at the MTL (mating-type-like) locus that ensures that only a or α cells can switch from the white state to the mating-competent opaque state, while a/α cells are refractory to switching. Here, we show that the related pathogen C. tropicalis undergoes white-opaque switching in all three cell types (a, α, and a/α), and thus switching is independent of MTL control. We also demonstrate that C. tropicalis white cells are themselves mating-competent, albeit at a lower efficiency than opaque cells. Transcriptional profiling of C. tropicalis white and opaque cells reveals significant overlap between switch-regulated genes in MTL homozygous and MTL heterozygous cells, although twice as many genes are white-opaque regulated in a/α cells as in a cells. In C. albicans, the transcription factor Wor1 is the master regulator of the white-opaque switch, and we show that Wor1 also regulates switching in C. tropicalis; deletion of WOR1 locks a, α, and a/α cells in the white state, while WOR1 overexpression induces these cells to adopt the opaque state. Furthermore, we show that WOR1 overexpression promotes both filamentous growth and biofilm formation in C. tropicalis, independent of the white-opaque switch. These results demonstrate an expanded role for C. tropicalis Wor1, including the regulation of processes necessary for infection of the mammalian host. We discuss these findings in light of the ancestral role of Wor1 as a transcriptional regulator of the transition between yeast form and filamentous growth.


Assuntos
Candida tropicalis , Proteínas Fúngicas/genética , Genes Fúngicos Tipo Acasalamento , Genes de Troca , Reprodução , Fatores de Transcrição/genética , Biofilmes/crescimento & desenvolvimento , Candida albicans/metabolismo , Candida tropicalis/genética , Candida tropicalis/crescimento & desenvolvimento , Citoesqueleto/metabolismo , Citoesqueleto/fisiologia , Regulação Fúngica da Expressão Gênica , Homozigoto , Fenótipo , Reprodução/genética , Reprodução/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA