Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
PLoS Pathog ; 17(3): e1009235, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33780518

RESUMEN

To gain a better understanding of the transcriptional response of Aspergillus fumigatus during invasive pulmonary infection, we used a NanoString nCounter to assess the transcript levels of 467 A. fumigatus genes during growth in the lungs of immunosuppressed mice. These genes included ones known to respond to diverse environmental conditions and those encoding most transcription factors in the A. fumigatus genome. We found that invasive growth in vivo induces a unique transcriptional profile as the organism responds to nutrient limitation and attack by host phagocytes. This in vivo transcriptional response is largely mimicked by in vitro growth in Aspergillus minimal medium that is deficient in nitrogen, iron, and/or zinc. From the transcriptional profiling data, we selected 9 transcription factor genes that were either highly expressed or strongly up-regulated during in vivo growth. Deletion mutants were constructed for each of these genes and assessed for virulence in mice. Two transcription factor genes were found to be required for maximal virulence. One was rlmA, which is required for the organism to achieve maximal fungal burden in the lung. The other was sltA, which regulates of the expression of multiple secondary metabolite gene clusters and mycotoxin genes independently of laeA. Using deletion and overexpression mutants, we determined that the attenuated virulence of the ΔsltA mutant is due in part to decreased expression aspf1, which specifies a ribotoxin, but is not mediated by reduced expression of the fumigaclavine gene cluster or the fumagillin-pseruotin supercluster. Thus, in vivo transcriptional profiling focused on transcription factors genes provides a facile approach to identifying novel virulence regulators.


Asunto(s)
Aspergillus fumigatus/genética , Regulación Fúngica de la Expresión Génica/genética , Pulmón/virología , Factores de Transcripción/metabolismo , Animales , Aspergilosis/microbiología , Aspergillus fumigatus/patogenicidad , Proteínas Fúngicas/metabolismo , Perfilación de la Expresión Génica/métodos , Hierro/metabolismo , Pulmón/metabolismo , Ratones , Virulencia/genética
2.
PLoS Genet ; 16(1): e1008582, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961865

RESUMEN

Metabolic adaptation is linked to the ability of the opportunistic pathogen Candida albicans to colonize and cause infection in diverse host tissues. One way that C. albicans controls its metabolism is through the glucose repression pathway, where expression of alternative carbon source utilization genes is repressed in the presence of its preferred carbon source, glucose. Here we carry out genetic and gene expression studies that identify transcription factors Mig1 and Mig2 as mediators of glucose repression in C. albicans. The well-studied Mig1/2 orthologs ScMig1/2 mediate glucose repression in the yeast Saccharomyces cerevisiae; our data argue that C. albicans Mig1/2 function similarly as repressors of alternative carbon source utilization genes. However, Mig1/2 functions have several distinctive features in C. albicans. First, Mig1 and Mig2 have more co-equal roles in gene regulation than their S. cerevisiae orthologs. Second, Mig1 is regulated at the level of protein accumulation, more akin to ScMig2 than ScMig1. Third, Mig1 and Mig2 are together required for a unique aspect of C. albicans biology, the expression of several pathogenicity traits. Such Mig1/2-dependent traits include the abilities to form hyphae and biofilm, tolerance of cell wall inhibitors, and ability to damage macrophage-like cells and human endothelial cells. Finally, Mig1 is required for a puzzling feature of C. albicans biology that is not shared with S. cerevisiae: the essentiality of the Snf1 protein kinase, a central eukaryotic carbon metabolism regulator. Our results integrate Mig1 and Mig2 into the C. albicans glucose repression pathway and illuminate connections among carbon control, pathogenicity, and Snf1 essentiality.


Asunto(s)
Candida albicans/genética , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Glucosa/metabolismo , Factores de Transcripción/metabolismo , Animales , Biopelículas , Candida albicans/efectos de los fármacos , Candida albicans/patogenicidad , Línea Celular , Farmacorresistencia Fúngica , Células Endoteliales/microbiología , Proteínas Fúngicas/genética , Humanos , Macrófagos/microbiología , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética
3.
PLoS Pathog ; 15(5): e1007787, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31116789

RESUMEN

Genotype-phenotype relationships can vary extensively among members of a species. One cause of this variation is circuit diversification, the alteration of gene regulatory relationships among members of a species. Circuit diversification is thought to be a starting point for the circuit divergence or rewiring that occurs during speciation. How widespread is circuit diversification? Here we address this question with the fungal pathogen Candida albicans, which forms biofilms rich in distinctive hyphal cells as a prelude to infection. Our understanding of the biofilm/hyphal regulatory network comes primarily from studies of one clinical isolate, strain SC5314, and its marked derivatives. We used CRISPR-based methods to create mutations of four key biofilm transcription factor genes-BCR1, UME6, BRG1, and EFG1 -in SC5314 and four additional clinical isolates. Phenotypic analysis revealed that mutations in BCR1 or UME6 have variable impact across strains, while mutations in BRG1 or EFG1 had uniformly severe impact. Gene expression, sampled with Nanostring probes and examined comprehensively for EFG1 via RNA-Seq, indicates that regulatory relationships are highly variable among isolates. Our results suggest that genotype-phenotype relationships vary in this strain panel in part because of differences in control of BRG1 by BCR1, a hypothesis that is supported through engineered constitutive expression of BRG1. Overall, the data show that circuit diversification is the rule, not the exception, in this biofilm/hyphal regulatory network.


Asunto(s)
Biopelículas/clasificación , Biopelículas/crecimiento & desarrollo , Candida albicans/clasificación , Candidiasis/genética , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Hifa/genética , Candida albicans/genética , Candidiasis/virología , Estudios de Asociación Genética , Especiación Genética , Humanos , Hifa/crecimiento & desarrollo , Transducción de Señal , Factores de Transcripción
4.
PLoS Pathog ; 13(5): e1006339, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28542565

RESUMEN

Streptococcus pneumoniae (pneumococcus) is a leading cause of death and disease in children and elderly. Genetic variability among isolates from this species is high. These differences, often the product of gene loss or gene acquisition via horizontal gene transfer, can endow strains with new molecular pathways, diverse phenotypes, and ecological advantages. PMEN1 is a widespread and multidrug-resistant pneumococcal lineage. Using comparative genomics we have determined that a regulator-peptide signal transduction system, TprA2/PhrA2, was acquired by a PMEN1 ancestor and is encoded by the vast majority of strains in this lineage. We show that TprA2 is a negative regulator of a PMEN1-specific gene encoding a lanthionine-containing peptide (lcpA). The activity of TprA2 is modulated by its cognate peptide, PhrA2. Expression of phrA2 is density-dependent and its C-terminus relieves TprA2-mediated inhibition leading to expression of lcpA. In the pneumococcal mouse model with intranasal inoculation, TprA2 had no effect on nasopharyngeal colonization but was associated with decreased lung disease via its control of lcpA levels. Furthermore, the TprA2/PhrA2 system has integrated into the pneumococcal regulatory circuitry, as PhrA2 activates TprA/PhrA, a second regulator-peptide signal transduction system widespread among pneumococci. Extracellular PhrA2 can release TprA-mediated inhibition, activating expression of TprA-repressed genes in both PMEN1 cells as well as another pneumococcal lineage. Acquisition of TprA2/PhrA2 has provided PMEN1 isolates with a mechanism to promote commensalism over dissemination and control inter-strain gene regulation.


Asunto(s)
Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Pandemias , Infecciones Neumocócicas/microbiología , Transducción de Señal , Streptococcus pneumoniae/genética , Anciano , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Transferencia de Gen Horizontal , Genómica , Humanos , Ratones , Modelos Biológicos , Mutación , Nasofaringe/microbiología , Filogenia , Infecciones Neumocócicas/epidemiología , Regulón/genética , Alineación de Secuencia , Streptococcus pneumoniae/fisiología
5.
PLoS Genet ; 12(12): e1006487, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27935965

RESUMEN

Biofilm formation on implanted medical devices is a major source of lethal invasive infection by Candida albicans. Filamentous growth of this fungus is tied to biofilm formation because many filamentation-associated genes are required for surface adherence. Cell cycle or cell growth defects can induce filamentation, but we have limited information about the coupling between filamentation and filamentation-associated gene expression after cell cycle/cell growth inhibition. Here we identified the CDK activating protein kinase Cak1 as a determinant of filamentation and filamentation-associated gene expression through a screen of mutations that diminish expression of protein kinase-related genes implicated in cell cycle/cell growth control. A cak1 diminished expression (DX) strain displays filamentous growth and expresses filamentation-associated genes in the absence of typical inducing signals. In a wild-type background, expression of filamentation-associated genes depends upon the transcription factors Bcr1, Brg1, Efg1, Tec1, and Ume6. In the cak1 DX background, the dependence of filamentation-associated gene expression on each transcription factor is substantially relieved. The unexpected bypass of filamentation-associated gene expression activators has the functional consequence of enabling biofilm formation in the absence of Bcr1, Brg1, Tec1, Ume6, or in the absence of both Brg1 and Ume6. It also enables filamentous cell morphogenesis, though not biofilm formation, in the absence of Efg1. Because these transcription factors are known to have shared target genes, we suggest that cell cycle/cell growth limitation leads to activation of several transcription factors, thus relieving dependence on any one.


Asunto(s)
Candida albicans/genética , Quinasas Ciclina-Dependientes/genética , Proteínas Fúngicas/genética , Morfogénesis/genética , Proteínas Quinasas/genética , Biopelículas/crecimiento & desarrollo , Candida albicans/crecimiento & desarrollo , Candidiasis/genética , Candidiasis/microbiología , Ciclo Celular/genética , Citoesqueleto/genética , Proteínas Fúngicas/biosíntesis , Regulación Fúngica de la Expresión Génica , Humanos , Hifa/genética , Hifa/crecimiento & desarrollo , Hifa/patogenicidad , Proteínas Quinasas/biosíntesis , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Quinasa Activadora de Quinasas Ciclina-Dependientes
6.
Mol Microbiol ; 105(4): 554-571, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28557053

RESUMEN

Streptococcus pneumoniae (pneumococcus) is a major human pathogen. It is a common colonizer of the human respiratory track, where it utilizes cell-cell communication systems to coordinate population-level behaviors. We reasoned that secreted peptides that are highly expressed during infection are pivotal for virulence. Thus, we used in silico pattern searches to define a pneumococcal secretome and analyzed the transcriptome of the clinically important PMEN1 lineage to identify which peptide-encoding genes are highly expressed in vivo. In this study, we characterized virulence peptide 1 (vp1), a highly expressed Gly-Gly peptide-encoding gene in chinchilla middle ear effusions. The vp1 gene is widely distributed across pneumococcus as well as encoded in related species. Studies in the chinchilla model of middle ear infection demonstrated that VP1 is a virulence determinant. The vp1 gene is positively regulated by a transcription factor from the Rgg family and its cognate SHP (short hydrophobic peptide). In vitro data indicated that VP1 promotes increased thickness and biomass for biofilms grown on chinchilla middle ear epithelial cells. Furthermore, the wild-type biofilm is restored with the exogenous addition of synthetic VP1. We conclude that VP1 is a novel streptococcal regulatory peptide that controls biofilm development and pneumococcal pathogenesis.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Streptococcus pneumoniae/metabolismo , Virulencia/genética , Animales , Proteínas Bacterianas/metabolismo , Comunicación Celular/fisiología , Chinchilla , Bases de Datos de Ácidos Nucleicos , Oído Medio/microbiología , Regulación Bacteriana de la Expresión Génica/genética , Otitis Media/microbiología , Péptidos/metabolismo , Infecciones Neumocócicas/metabolismo , Análisis de Secuencia de ADN/métodos , Streptococcus/metabolismo , Streptococcus pneumoniae/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
PLoS Biol ; 13(2): e1002076, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25693184

RESUMEN

Gene expression dynamics have provided foundational insight into almost all biological processes. Here, we analyze expression of environmentally responsive genes and transcription factor genes to infer signals and pathways that drive pathogen gene regulation during invasive Candida albicans infection of a mammalian host. Environmentally responsive gene expression shows that there are early and late phases of infection. The early phase includes induction of zinc and iron limitation genes, genes that respond to transcription factor Rim101, and genes characteristic of invasive hyphal cells. The late phase includes responses related to phagocytosis by macrophages. Transcription factor gene expression also reflects early and late phases. Transcription factor genes that are required for virulence or proliferation in vivo are enriched among highly expressed transcription factor genes. Mutants defective in six transcription factor genes, three previously studied in detail (Rim101, Efg1, Zap1) and three less extensively studied (Rob1, Rpn4, Sut1), are profiled during infection. Most of these mutants have distinct gene expression profiles during infection as compared to in vitro growth. Infection profiles suggest that Sut1 acts in the same pathway as Zap1, and we verify that functional relationship with the finding that overexpression of either ZAP1 or the Zap1-dependent zinc transporter gene ZRT2 restores pathogenicity to a sut1 mutant. Perturbation with the cell wall inhibitor caspofungin also has distinct gene expression impact in vivo and in vitro. Unexpectedly, caspofungin induces many of the same genes that are repressed early during infection, a phenomenon that we suggest may contribute to drug efficacy. The pathogen response circuitry is tailored uniquely during infection, with many relevant regulatory relationships that are not evident during growth in vitro. Our findings support the principle that virulence is a property that is manifested only in the distinct environment in which host-pathogen interaction occurs.


Asunto(s)
Candida albicans/metabolismo , Candida albicans/patogenicidad , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Redes Reguladoras de Genes , Animales , Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candidiasis/tratamiento farmacológico , Candidiasis/metabolismo , Candidiasis/microbiología , Candidiasis/patología , Caspofungina , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Equinocandinas/farmacología , Proteínas Fúngicas/metabolismo , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Inyecciones Intravenosas , Riñón/metabolismo , Riñón/microbiología , Riñón/patología , Lipopéptidos , Masculino , Ratones , Ratones Endogámicos BALB C , Anotación de Secuencia Molecular , Mutación , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Virulencia
8.
Infect Immun ; 83(9): 3684-92, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26150537

RESUMEN

Candida albicans is normally a commensal fungus of the human mucosae and skin, but it causes life-threatening systemic infections in hospital settings in the face of predisposing conditions, such as indwelling catheters, abdominal surgery, or antibiotic use. Immunity to C. albicans involves various immune parameters, but the cytokine interleukin-17A (IL-17A) (also known as IL-17) has emerged as a centrally important mediator of immune defense against both mucosal and systemic candidiasis. Conversely, IL-17A has been suggested to enhance the virulence of C. albicans, indicating that it may exert detrimental effects on pathogenesis. In this study, we hypothesized that a C. albicans strain expressing IL-17A would exhibit reduced virulence in vivo. To that end, we created a Candida-optimized expression cassette encoding murine IL-17A, which was transformed into the DAY286 strain of C. albicans. Candida-derived IL-17A was indistinguishable from murine IL-17A in terms of biological activity and detection in standard enzyme-linked immunosorbent assays (ELISAs). Expression of IL-17A did not negatively impact the growth of these strains in vitro. Moreover, the IL-17A-expressing C. albicans strains showed significantly reduced pathogenicity in a systemic model of Candida infection, mainly evident during the early stages of disease. Collectively, these findings suggest that IL-17A mitigates the virulence of C. albicans.


Asunto(s)
Candida albicans/patogenicidad , Candidiasis/inmunología , Ingeniería Genética/métodos , Interleucina-17/inmunología , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Virulencia/inmunología
9.
Eukaryot Cell ; 13(11): 1403-10, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25217462

RESUMEN

Septin proteins are conserved structural proteins that often demarcate regions of cell division. The essential nature of the septin ring, composed of several septin proteins, complicates investigation of the functions of the ring, although careful analysis in the model yeast Saccharomyces cerevisiae has elucidated the role that septins play in the cell cycle. Mutation analysis of nonessential septins in the pathogenic fungus Candida albicans has shown that septins also have vital roles in cell wall regulation (CWR), hyphal formation, and pathogenesis. While mutations in nonessential septins have been useful in establishing phenotypes, the septin defect is so slight that identifying causative associations between septins and downstream effectors has been difficult. In this work, we describe decreased abundance by mRNA perturbation (DAmP) alleles of essential septins, which display a septin defect more severe than the defect observed in deletions of nonessential septins. The septin DAmP alleles have allowed us to genetically separate the roles of septins in hyphal growth and CWR and to identify the cyclic AMP pathway as a pathway that likely acts in a parallel manner with septins in hyphal morphogenesis.


Asunto(s)
Candida albicans/genética , Hifa/crecimiento & desarrollo , Hifa/genética , Saccharomyces cerevisiae/genética , Septinas/genética , Animales , Candida albicans/patogenicidad , Candidiasis/genética , Candidiasis/patología , Proteínas de Ciclo Celular/genética , División Celular/genética , Pared Celular/genética , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Citoesqueleto/genética , Análisis Mutacional de ADN , Proteínas de Unión al ADN/biosíntesis , Proteínas Fúngicas/biosíntesis , Masculino , Ratones , Ratones Endogámicos ICR , Proteínas de Saccharomyces cerevisiae/biosíntesis , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal/genética , Factores de Transcripción/biosíntesis
10.
PLoS Pathog ; 8(2): e1002525, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22359502

RESUMEN

Cell-substrate adherence is a fundamental property of microorganisms that enables them to exist in biofilms. Our study focuses on adherence of the fungal pathogen Candida albicans to one substrate, silicone, that is relevant to device-associated infection. We conducted a mutant screen with a quantitative flow-cell assay to identify thirty transcription factors that are required for adherence. We then combined nanoString gene expression profiling with functional analysis to elucidate relationships among these transcription factors, with two major goals: to extend our understanding of transcription factors previously known to govern adherence or biofilm formation, and to gain insight into the many transcription factors we identified that were relatively uncharacterized, particularly in the context of adherence or cell surface biogenesis. With regard to the first goal, we have discovered a role for biofilm regulator Bcr1 in adherence, and found that biofilm regulator Ace2 is a major functional target of chromatin remodeling factor Snf5. In addition, Bcr1 and Ace2 share several target genes, pointing to a new connection between them. With regard to the second goal, our findings reveal existence of a large regulatory network that connects eleven adherence regulators, the zinc-response regulator Zap1, and approximately one quarter of the predicted cell surface protein genes in this organism. This limited yet sensitive glimpse of mutant gene expression changes had thus defined one of the broadest cell surface regulatory networks in C. albicans.


Asunto(s)
Candida albicans/fisiología , Adhesión Celular/genética , Proteínas Fúngicas/genética , Perfilación de la Expresión Génica , Biopelículas/crecimiento & desarrollo , Genes Fúngicos
11.
Eukaryot Cell ; 11(10): 1201-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865500

RESUMEN

The Rim101/PacC pathway governs adaptation to alkaline pH in many fungi. Output of the pathway is mediated by transcription factors of the Rim101/PacC family, which are activated by proteolytic cleavage. The proteolytic complex includes scaffold protein Rim20 and endosome-associated subunits of the endosomal sorting complex required for transport (ESCRT). We provide here evidence that Saccharomyces cerevisiae Rim13, the protease that is implicated in Rim101 cleavage, is associated with the Rim20-ESCRT complex, and we investigate its regulation. Rim13-GFP is dispersed in cells grown in acidic medium but forms punctate foci when cells encounter alkaline conditions. A vps4Δ mutant, which accumulates elevated levels of endosomal ESCRT, also accumulates elevated levels of Rim13-GFP foci, independently of external pH. In the vps4Δ background, mutation of ESCRT subunit Snf7 or of Rim20 blocks the formation of Rim13 foci, and we found that Rim13 and Rim20 are colocalized. The Rim13 ortholog PalB of Aspergillus nidulans has been shown to undergo ESCRT and membrane association through an N-terminal MIT domain, but Rim13 orthologs in the Saccharomyces clade lack homology to this N-terminal region. Instead, there is a clade-limited C-terminal region, and we show that point mutations in this region prevent punctate localization and impair Rim13 function. We suggest that RIM13 arose from its ancestral gene through two genome rearrangements. The ancestor lost the coding region for its MIT domain through a 5' rearrangement and acquired the coding region for the Saccharomyces-specific functional equivalent through a 3' rearrangement.


Asunto(s)
Proteasas de Cisteína/química , Péptido Hidrolasas/química , Señales de Clasificación de Proteína , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Proteasas de Cisteína/genética , Proteasas de Cisteína/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Datos de Secuencia Molecular , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Transporte de Proteínas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
Eukaryot Cell ; 11(7): 896-904, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22544909

RESUMEN

Candida albicans is a causative agent of oropharyngeal candidiasis (OPC), a biofilm-like infection of the oral mucosa. Biofilm formation depends upon the C. albicans transcription factor Bcr1, and previous studies indicate that Bcr1 is required for OPC in a mouse model of infection. Here we have used a nanoString gene expression measurement platform to elucidate the role of Bcr1 in OPC-related gene expression. We chose for assays a panel of 134 genes that represent a range of morphogenetic and cell cycle functions as well as environmental and stress response pathways. We assayed gene expression in whole infected tongue samples. The results sketch a portrait of C. albicans gene expression in which numerous stress response pathways are activated during OPC. This one set of experiments identifies 64 new genes with significantly altered RNA levels during OPC, thus increasing substantially the number of known genes in this expression class. The bcr1Δ/Δ mutant had a much more limited gene expression defect during OPC infection than previously reported for in vitro growth conditions. Among major functional Bcr1 targets, we observed that ALS3 was Bcr1 dependent in vivo while HWP1 was not. We used null mutants and complemented strains to verify that Bcr1 and Hwp1 are required for OPC infection in this model. The role of Als3 is transient and mild, though significant. Our findings suggest that the versatility of C. albicans as a pathogen may reflect its ability to persist in the face of multiple stresses and underscore that transcriptional circuitry during infection may be distinct from that detailed during in vitro growth.


Asunto(s)
Biopelículas , Candida albicans/fisiología , Candidiasis Bucal/microbiología , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Animales , Candida albicans/genética , Modelos Animales de Enfermedad , Proteínas Fúngicas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Mucosa Bucal/microbiología
13.
14.
Genetics ; 218(3)2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-33989396

RESUMEN

The fungal pathogen Candida albicans is surrounded by a cell wall that is the target of caspofungin and other echinocandin antifungals. Candida albicans can grow in several morphological forms, notably budding yeast and hyphae. Yeast and hyphal forms differ in cell wall composition, leading us to hypothesize that there may be distinct genes required for yeast and hyphal responses to caspofungin. Mutants in 27 genes reported previously to be caspofungin hypersensitive under yeast growth conditions were all caspofungin hypersensitive under hyphal growth conditions as well. However, a screen of mutants defective in transcription factor genes revealed that Cup9 is required for normal caspofungin tolerance under hyphal and not yeast growth conditions. In a hyphal-defective efg1Δ/Δ background, Cup9 is still required for normal caspofungin tolerance. This result argues that Cup9 function is related to growth conditions rather than cell morphology. RNA-seq conducted under hyphal growth conditions indicated that 361 genes were up-regulated and 145 genes were down-regulated in response to caspofungin treatment. Both classes of caspofungin-responsive genes were enriched for cell wall-related proteins, as expected for a response to disruption of cell wall integrity and biosynthesis. The cup9Δ/Δ mutant, treated with caspofungin, had reduced RNA levels of 40 caspofungin up-regulated genes, and had increased RNA levels of 8 caspofungin down-regulated genes, an indication that Cup9 has a narrow rather than global role in the cell wall integrity response. Five Cup9-activated surface-protein genes have roles in cell wall integrity, based on mutant analysis published previously (PGA31 and IFF11) or shown here (ORF19.3499, ORF19.851, or PGA28), and therefore may explain the hypersensitivity of the cup9Δ/Δmutant to caspofungin. Our findings define Cup9 as a new determinant of caspofungin susceptibility.


Asunto(s)
Candida albicans/metabolismo , Pared Celular/metabolismo , Farmacorresistencia Fúngica , Proteínas Fúngicas/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Antifúngicos/toxicidad , Candida albicans/efectos de los fármacos , Candida albicans/genética , Caspofungina/toxicidad , Pared Celular/efectos de los fármacos , Pared Celular/genética , Proteínas Fúngicas/genética , Regulación Fúngica de la Expresión Génica , Proteínas de Homeodominio/genética , Factores de Transcripción/genética
15.
J Vis Exp ; (157)2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32202525

RESUMEN

The microbial fungus Candida albicans can undergo a change from commensal colonization to virulence that is strongly correlated with its ability to switch from yeast-form growth to hyphal growth. Cells initiating this process become adherent to surfaces as well as to each other, with the resulting development of a biofilm colony. This commonly occurs not only on mucosal tissue surfaces in yeast infections, but also on medical implants such as catheters. It is well known that biofilm cells are resistant to antifungal drugs, and that cells that shed from the biofilm can lead to dangerous systemic infections. Biofilms range from heavily translucent to opaque due to refractive heterogeneity. Therefore, fungal biofilms are difficult to study by optical microscopy. To visualize internal structural, cellular, and subcellular features, we clarify fixed intact biofilms by stepwise solvent exchange to a point of optimal refractive index matching. For C. albicans biofilms, sufficient clarification is attained with methyl salicylate (n = 1.537) to enable confocal microscopy from apex to base in 600 µm biofilms with little attenuation. In this visualization protocol we outline phase contrast refractometry, the growth of laboratory biofilms, fixation, staining, solvent exchange, the setup for confocal fluorescence microscopy, and representative results.


Asunto(s)
Biopelículas , Candida albicans/fisiología , Imagenología Tridimensional , Biopelículas/crecimiento & desarrollo , Hifa/fisiología , Microscopía Confocal , Mutación/genética , Refractometría
16.
Bioconjug Chem ; 20(10): 1843-1847, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20976031

RESUMEN

We demonstrate the effectiveness of a genetically encoded Malachite Green (MG) binding fluorogen activating protein (FAP) for live cell stimulated emission depletion nanoscopy (STED). Both extracellular and intracellular FAPs were tested in living cells using fluorogens with either membrane expressed FAP or as an intracellular FAP-actin fusion. Structures with FWHM of 110-122nm were observed. Depletion data however suggests a resolution of 70nm with the given instrument.

17.
Cell Host Microbe ; 25(3): 432-443.e6, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30870623

RESUMEN

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.


Asunto(s)
Candida albicans/crecimiento & desarrollo , Candida albicans/patogenicidad , Tracto Gastrointestinal/microbiología , Regulación Fúngica de la Expresión Génica , Simbiosis , Animales , Candida albicans/citología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Hifa/citología , Hifa/crecimiento & desarrollo , Ratones , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Virulencia
18.
Methods Mol Biol ; 1968: 79-88, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30929207

RESUMEN

Bacterial cells modify their gene expression profiles throughout different stages of growth and in response to environmental cues. Analyses of gene expression across conditions reveal both conserved and condition-specific gene responses of bacteria to adapt to these dynamic conditions. In this chapter, we present a guide to pneumococcal RNA extraction for use in the NanoString nCounter platform. The nCounter is a highly effective method to measure gene expression of bacteria not only in a planktonic mode of growth but also in the presence of host cells where the RNA of interest represents only a small portion of the total material.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Streptococcus pneumoniae/genética , ARN Mensajero/genética
19.
mSphere ; 3(2)2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29695626

RESUMEN

The biological function of a gene is often probed through its interactions with other genes. This general approach has been especially useful to build knowledge about poorly understood genes upon the bedrock of well-characterized genes. Genetic interaction analysis requires the construction of strains with mutations in two or more genes. Single-gene mutants of microbial pathogens are generally validated through introduction of a wild-type copy of the affected gene to create a complemented or reconstituted strain, followed by testing for restoration of a wild-type phenotype. This practice, formalized as one of Falkow's "molecular Koch's postulates" ensures that the phenotype of the mutant depends upon the known mutation. However, multigene mutants are seldom validated because of the labor required to assemble multiple genomic segments into a vector that can be introduced into the mutant strain. We present here an approach, concatemer assembly for rescue of mutant abilities (CARMA), that circumvents this impediment through an in vivo recombinational assembly strategy that does not require cloning at all. Our results show that CARMA allows genetic rescue of two double-gene mutant strains of the fungal pathogen Candida albicansIMPORTANCE Our understanding of new genes is often built upon the knowledge of well-characterized genes. One avenue toward revealing such connections involves creation of strains with mutations in two or more defined genes to permit genetic interaction analysis. Strain manipulations can yield unexpected mutations at loci outside the defined targeted genes. In this report, we describe a method for rapid validation of multigene mutants, thus allowing an appraisal of the contribution of the defined targeted genes to the strain's phenotype.


Asunto(s)
Candida albicans/genética , ADN Concatenado , Proteínas Fúngicas/genética , Mutación , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Eliminación de Gen , Genes Fúngicos , Ingeniería Genética , Fenotipo
20.
Mol Biol Cell ; 15(6): 2593-605, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15047864

RESUMEN

The Sec1/Munc18 (SM) family of proteins is thought to impart compartmental specificity to vesicle fusion reactions. Here we report characterization of Vps33p, an SM family member previously thought to act exclusively at the vacuolar membrane with the vacuolar syntaxin Vam3p. Vacuolar morphology of vps33Delta cells resembles that of cells lacking both Vam3p and the endosomal syntaxin Pep12p, suggesting that Vps33p may function with these syntaxins at the vacuole and the endosome. Consistent with this, vps33 mutants secrete the Golgi precursor form of the vacuolar hydrolase CPY into the medium. We also demonstrate that Vps33p acts at other steps, for vps33 mutants show severe defects in endocytosis at the late endosome. At the endosome, Vps33p and other class C members exist as a complex with Vps8p, a protein previously known to act in transport between the late Golgi and the endosome. Vps33p also interacts with Pep12p, a known interactor of the SM protein Vps45p. High copy PEP7/VAC1 suppresses vacuolar morphology defects of vps33 mutants. These findings demonstrate that Vps33p functions at multiple trafficking steps and is not limited to action at the vacuolar membrane. This is the first report demonstrating the involvement of a single syntaxin with two SM proteins at the same organelle.


Asunto(s)
Endosomas/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Vacuolas/metabolismo , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Catepsina A/metabolismo , Proteínas del Citoesqueleto/metabolismo , Endocitosis , Eliminación de Gen , Glicosilación , Aparato de Golgi/metabolismo , Factor de Apareamiento , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos , Proteínas Munc18 , Péptidos/metabolismo , Unión Proteica , Precursores de Proteínas/metabolismo , Proteínas Qa-SNARE , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Vacuolas/ultraestructura , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA