Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.344
Filtrar
1.
Sci Adv ; 10(31): eado5555, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39093965

RESUMEN

Because of the decreasing supply of new antibiotics, recent outbreaks of infectious diseases, and the emergence of antibiotic-resistant microorganisms, it is imperative to develop new effective strategies for deactivating a broad spectrum of microorganisms and viruses. We have implemented electrically polarized nanoscale metallic (ENM) coatings that deactivate a wide range of microorganisms including Gram-negative and Gram-positive bacteria with greater than 6-log reduction in less than 10 minutes of treatment. The electrically polarized devices were also effective in deactivating lentivirus and Candida albicans. The key to the high deactivation effectiveness of ENM devices is electrochemical production of micromolar cuprous ions, which mediated reduction of oxygen to hydrogen peroxide. Formation of highly damaging species, hydroxyl radicals and hypochlorous acid, from hydrogen peroxide contributed to antimicrobial properties of the ENM devices. The electric polarization of nanoscale coatings represents an unconventional tool for deactivating a broad spectrum of microorganisms through in situ production of reactive oxygenated and chlorinated species.


Asunto(s)
Peróxido de Hidrógeno , Peróxido de Hidrógeno/metabolismo , Oxígeno/metabolismo , Oxígeno/química , Candida albicans/efectos de los fármacos , Candida albicans/metabolismo , Propiedades de Superficie , Especies Reactivas de Oxígeno/metabolismo , Antiinfecciosos/farmacología , Antiinfecciosos/química , Oxidación-Reducción
2.
PLoS One ; 19(8): e0308665, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39121069

RESUMEN

Development of resistance and tolerance to antifungal drugs in Candida albicans can compromise treatment of infections caused by this pathogenic yeast species. The uniquely expanded C. albicans TLO gene family is comprised of 14 paralogous genes which encode Med2, a subunit of the multiprotein Mediator complex which is involved in the global control of transcription. This study investigates the acquisition of fluconazole tolerance in a mutant in which the entire TLO gene family has been deleted. This phenotype was reversed to varying degrees upon reintroduction of representative members of the alpha- and beta-TLO clades (i.e. TLO1 and TLO2), but not by TLO11, a gamma-clade representative. Comparative RNA sequencing analysis revealed changes in the expression of genes involved in a range of cellular functions, including ergosterol biosynthesis, mitochondrial function, and redox homeostasis. This was supported by the results of mass spectrometry analysis, which revealed alterations in sterol composition of the mutant cell membrane. Our data suggest that members of the C. albicans TLO gene family are involved in the control of ergosterol biosynthesis and mitochondrial function and may play a role in the responses of C. albicans to azole antifungal agents.


Asunto(s)
Antifúngicos , Candida albicans , Farmacorresistencia Fúngica , Fluconazol , Proteínas Fúngicas , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/metabolismo , Fluconazol/farmacología , Antifúngicos/farmacología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Farmacorresistencia Fúngica/genética , Esteroles/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Ergosterol/biosíntesis , Ergosterol/metabolismo , Eliminación de Gen , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Familia de Multigenes , Pruebas de Sensibilidad Microbiana , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética
3.
Sci Signal ; 17(849): eads1861, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39137247
4.
Front Cell Infect Microbiol ; 14: 1392564, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983116

RESUMEN

Antifungal resistance and antifungal tolerance are two distinct terms that describe different cellular responses to drugs. Antifungal resistance describes the ability of a fungus to grow above the minimal inhibitory concentration (MIC) of a drug. Antifungal tolerance describes the ability of drug susceptible strains to grow slowly at inhibitory drug concentrations. Recent studies indicate antifungal resistance and tolerance have distinct evolutionary trajectories. Superficial candidiasis bothers millions of people yearly. Miconazole has been used for topical treatment of yeast infections for over 40 years. Yet, fungal resistance to miconazole remains relatively low. Here we found different clinical isolates of Candida albicans had different profile of tolerance to miconazole, and the tolerance was modulated by physiological factors including temperature and medium composition. Exposure of non-tolerant strains with different genetic backgrounds to miconazole mainly induced development of tolerance, not resistance, and the tolerance was mainly due to whole chromosomal or segmental amplification of chromosome R. The efflux gene CDR1 was required for maintenance of tolerance in wild type strains but not required for gain of aneuploidy-mediated tolerance. Heat shock protein Hsp90 and calcineurin were essential for maintenance as well as gain of tolerance. Our study indicates development of aneuploidy-mediated tolerance, not resistance, is the predominant mechanism of rapid adaptation to miconazole in C. albicans, and the clinical relevance of tolerance deserves further investigations.


Asunto(s)
Aneuploidia , Antifúngicos , Calcineurina , Candida albicans , Farmacorresistencia Fúngica , Proteínas Fúngicas , Proteínas HSP90 de Choque Térmico , Miconazol , Pruebas de Sensibilidad Microbiana , Miconazol/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Antifúngicos/farmacología , Farmacorresistencia Fúngica/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Calcineurina/metabolismo , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Candidiasis/microbiología , Candidiasis/tratamiento farmacológico , Tolerancia a Medicamentos
5.
Commun Biol ; 7(1): 814, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965424

RESUMEN

In human pathogenic fungi, receiver domains from hybrid histidine kinases (hHK) have to recognize one HPt. To understand the recognition mechanism, we have assessed phosphorelay from receiver domains of five hHKs of group III, IV, V, VI, and XI to HPt from Chaetomium thermophilum and obtained the structures of Ct_HPt alone and in complex with the receiver domain of hHK group VI. Our data indicate that receiver domains phosphotransfer to Ct_HPt, show a low affinity for complex formation, and prevent a Leu-Thr switch to stabilize phosphoryl groups, also derived from the structures of the receiver domains of hHK group III and Candida albicans Sln1. Moreover, we have elucidated the envelope structure of C. albicans Ypd1 using small-angle X-ray scattering which reveals an extended flexible conformation of the long loop αD-αE which is not involved in phosphotransfer. Finally, we have analyzed the role of salt bridges in the structure of Ct_HPt alone.


Asunto(s)
Chaetomium , Proteínas Fúngicas , Histidina Quinasa , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Chaetomium/metabolismo , Chaetomium/genética , Chaetomium/enzimología , Histidina Quinasa/metabolismo , Histidina Quinasa/química , Histidina Quinasa/genética , Candida albicans/metabolismo , Candida albicans/enzimología , Fosforilación , Modelos Moleculares , Dispersión del Ángulo Pequeño , Conformación Proteica
6.
NPJ Syst Biol Appl ; 10(1): 77, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39025861

RESUMEN

Energy metabolism is crucial for all living cells, especially during fast growth or stress scenarios. Many cancer and activated immune cells (Warburg effect) or yeasts (Crabtree effect) mostly rely on aerobic glucose fermentation leading to lactate or ethanol, respectively, to generate ATP. In recent years, several mathematical models have been proposed to explain the Warburg effect on theoretical grounds. Besides glucose, glutamine is a very important substrate for eukaryotic cells-not only for biosynthesis, but also for energy metabolism. Here, we present a minimal constraint-based stoichiometric model for explaining both the classical Warburg effect and the experimentally observed respirofermentation of glutamine (WarburQ effect). We consider glucose and glutamine respiration as well as the respective fermentation pathways. Our resource allocation model calculates the ATP production rate, taking into account enzyme masses and, therefore, pathway costs. While our calculation predicts glucose fermentation to be a superior energy-generating pathway in human cells, different enzyme characteristics in yeasts reduce this advantage, in some cases to such an extent that glucose respiration is preferred. The latter is observed for the fungal pathogen Candida albicans, which is a known Crabtree-negative yeast. Further, optimization results show that glutamine is a valuable energy source and important substrate under glucose limitation, in addition to its role as a carbon and nitrogen source of biomass in eukaryotic cells. In conclusion, our model provides insights that glutamine is an underestimated fuel for eukaryotic cells during fast growth and infection scenarios and explains well the observed parallel respirofermentation of glucose and glutamine in several cell types.


Asunto(s)
Metabolismo Energético , Fermentación , Glucosa , Glutamina , Modelos Biológicos , Neoplasias , Glutamina/metabolismo , Metabolismo Energético/fisiología , Humanos , Neoplasias/metabolismo , Glucosa/metabolismo , Fermentación/fisiología , Candida albicans/metabolismo , Adenosina Trifosfato/metabolismo , Saccharomyces cerevisiae/metabolismo
7.
Biomolecules ; 14(7)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-39062502

RESUMEN

Current microbiological methods for pneumonia diagnosis require invasive specimen collection and time-consuming analytical procedures. There is a need for less invasive and faster methods to detect lower respiratory tract infections. The analysis of volatile metabolites excreted by pathogenic microorganisms provides the basis for developing such a method. Given the synergistic role of Candida albicans in increasing the virulence of pathogenic bacteria causing pneumonia and the cross-kingdom metabolic interactions between microorganisms, we compare the emission of volatiles from Candida albicans yeasts and the bacteria Staphylococcus aureus using single and mixed co-cultures and apply that knowledge to human in vivo investigations. Gas chromatography-mass spectrometry (GC-MS) analysis resulted in the identification of sixty-eight volatiles that were found to have significantly different levels in cultures compared to reference medium samples. Certain volatiles were found in co-cultures that mainly originated from C. albicans metabolism (e.g., isobutyl acetate), whereas other volatiles primarily came from S. aureus (e.g., ethyl 2-methylbutyrate). Isopentyl valerate reflects synergic interactions of both microbes, as its level in co-cultures was found to be approximately three times higher than the sum of its amounts in monocultures. Hydrophilic-lipophilic-balanced (HLB) coated meshes for thin-film microextraction (TFME) were used to preconcentrate volatiles directly from bronchoalveolar lavage (BAL) specimens collected from patients suffering from ventilation-associated pneumonia (VAP), which was caused explicitly by C. albicans and S. aureus. GC-MS analyses confirmed the existence of in vitro-elucidated microbial VOCs in human specimens. Significant differences in BAL-extracted amounts respective to the pathogen-causing pneumonia were found. The model in vitro experiments provided evidence that cross-kingdom interactions between pathogenic microorganisms affect the synthesis of volatile compounds. The TFME meshes coated with HLB particles proved to be suitable for extracting VOCs from human material, enabling the translation of in vitro experiments on the microbial volatilome to the in vivo situation involving infected patients. This indicates the direction that should be taken for further clinical studies on VAP diagnosis based on volatile analysis.


Asunto(s)
Líquido del Lavado Bronquioalveolar , Candida albicans , Cromatografía de Gases y Espectrometría de Masas , Staphylococcus aureus , Compuestos Orgánicos Volátiles , Candida albicans/metabolismo , Staphylococcus aureus/metabolismo , Humanos , Compuestos Orgánicos Volátiles/metabolismo , Compuestos Orgánicos Volátiles/análisis , Líquido del Lavado Bronquioalveolar/microbiología , Líquido del Lavado Bronquioalveolar/química , Técnicas de Cocultivo , Neumonía/microbiología , Neumonía/metabolismo
8.
Nat Commun ; 15(1): 5746, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982056

RESUMEN

Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.


Asunto(s)
Candida albicans , Candidiasis , Coinfección , Proteínas Fúngicas , Infecciones Estafilocócicas , Staphylococcus aureus , Candida albicans/metabolismo , Candida albicans/patogenicidad , Candida albicans/genética , Animales , Coinfección/microbiología , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/metabolismo , Staphylococcus aureus/genética , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/metabolismo , Candidiasis/microbiología , Ratones , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Infecciones Intraabdominales/microbiología , Femenino , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Percepción de Quorum/genética , Virulencia , Regulación Fúngica de la Expresión Génica , Modelos Animales de Enfermedad , Transactivadores/metabolismo , Transactivadores/genética
9.
mSphere ; 9(7): e0037224, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38980069

RESUMEN

Iron acquisition is critical for pathogens to proliferate during invasive infection, and the human fungal pathogen Candida albicans is no exception. The iron regulatory network, established in reference strain SC5314 and derivatives, includes the central player Sef1, a transcription factor that activates iron acquisition genes in response to iron limitation. Here, we explored potential variation in this network among five diverse C. albicans strains through mutant analysis, Nanostring gene expression profiling, and, for two strains, RNA-Seq. Our findings highlight four features that may inform future studies of natural variation and iron acquisition in this species. (i) Conformity: In all strains, major iron acquisition genes are upregulated during iron limitation, and a sef1Δ/Δ mutation impairs that response and growth during iron limitation. (ii) Response variation: Some aspects of the iron limitation response vary among strains, notably the activation of hypha-associated genes. As this gene set is tied to tissue damage and virulence, variation may impact the progression of infection. (iii) Genotype-phenotype variation: The impact of a sef1Δ/Δ mutation on cell wall integrity varies, and for the two strains examined the phenotype correlated with sef1Δ/Δ impact on several cell wall integrity genes. (iv) Phenotype discovery: DNA repair genes were induced modestly by iron limitation in sef1Δ/Δ mutants, with fold changes we would usually ignore. However, the response occurred in both strains tested and was reminiscent of a much stronger response described in Cryptococcus neoformans, a suggestion that it may have biological meaning. In fact, we observed that the iron limitation of a sef1Δ/Δ mutant caused recessive phenotypes to emerge at two heterozygous loci. Overall, our results show that a network that is critical for pathogen proliferation presents variation outside of its core functions.IMPORTANCEA key virulence factor of Candida albicans is the ability to maintain iron homeostasis in the host where iron is scarce. We focused on a central iron regulator, SEF1. We found that iron regulator Sef1 is required for growth, cell wall integrity, and genome integrity during iron limitation. The novel aspect of this work is the characterization of strain variation in a circuit that is required for survival in the host and the connection of iron acquisition to genome integrity in C. albicans.


Asunto(s)
Candida albicans , Proteínas Fúngicas , Regulación Fúngica de la Expresión Génica , Hierro , Candida albicans/genética , Candida albicans/patogenicidad , Candida albicans/metabolismo , Hierro/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Mutación , Virulencia , Perfilación de la Expresión Génica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Fenotipo , Pared Celular/metabolismo , Pared Celular/genética , Variación Genética , Genotipo
10.
mBio ; 15(8): e0153524, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38980041

RESUMEN

At human body temperature, the fungal pathogen Candida albicans can transition from yeast to filamentous morphologies in response to host-relevant cues. Additionally, elevated temperatures encountered during febrile episodes can independently induce C. albicans filamentation. However, the underlying genetic pathways governing this developmental transition in response to elevated temperatures remain largely unexplored. Here, we conducted a functional genomic screen to unravel the genetic mechanisms orchestrating C. albicans filamentation specifically in response to elevated temperature, implicating 45% of genes associated with the spliceosome or pre-mRNA splicing in this process. Employing RNA-Seq to elucidate the relationship between mRNA splicing and filamentation, we identified greater levels of intron retention in filaments compared to yeast, which correlated with reduced expression of the affected genes. Intriguingly, homozygous deletion of a gene encoding a spliceosome component important for filamentation (PRP19) caused even greater levels of intron retention compared with wild type and displayed globally dysregulated gene expression. This suggests that intron retention is a mechanism for fine-tuning gene expression during filamentation, with perturbations of the spliceosome exacerbating this process and blocking filamentation. Overall, this study unveils a novel biological process governing C. albicans filamentation, providing new insights into the complex regulation of this key virulence trait.IMPORTANCEFungal pathogens such as Candida albicans can cause serious infections with high mortality rates in immunocompromised individuals. When C. albicans is grown at temperatures encountered during human febrile episodes, yeast cells undergo a transition to filamentous cells, and this process is key to its virulence. Here, we expanded our understanding of how C. albicans undergoes filamentation in response to elevated temperature and identified many genes involved in mRNA splicing that positively regulate filamentation. Through transcriptome analyses, we found that intron retention is a mechanism for fine-tuning gene expression in filaments, and perturbation of the spliceosome exacerbates intron retention and alters gene expression substantially, causing a block in filamentation. This work adds to the growing body of knowledge on the role of introns in fungi and provides new insights into the cellular processes that regulate a key virulence trait in C. albicans.


Asunto(s)
Candida albicans , Proteínas Fúngicas , Regulación Fúngica de la Expresión Génica , Empalmosomas , Candida albicans/genética , Candida albicans/patogenicidad , Candida albicans/crecimiento & desarrollo , Candida albicans/fisiología , Candida albicans/metabolismo , Empalmosomas/genética , Empalmosomas/metabolismo , Humanos , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Morfogénesis/genética , Empalme del ARN , Virulencia , Hifa/crecimiento & desarrollo , Hifa/genética , Intrones/genética
11.
mBio ; 15(8): e0073224, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38953353

RESUMEN

Candida albicans, an opportunistic fungal pathogen, produces the quorum-sensing molecule farnesol, which we have shown alters the transcriptional response and phenotype of human monocyte-derived dendritic cells (DCs), including their cytokine secretion and ability to prime T cells. This is partially dependent on the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-γ), which has numerous ligands, including the sphingolipid metabolite sphingosine 1-phosphate. Sphingolipids are a vital component of membranes that affect membrane protein arrangement and phagocytosis of C. albicans by DCs. Thus, we quantified sphingolipid metabolites in monocytes differentiating into DCs by High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Farnesol increased the activity of serine palmitoyltransferase, leading to increased levels of 3-keto-dihydrosphingosine, dihydrosphingosine, and dihydrosphingosine 1-phosphate and inhibited dihydroceramide desaturase by inducing oxidative stress, leading to increased levels of dihydroceramide and dihydrosphingomyelin species and reduced ceramide levels. Accumulation of dihydroceramides can inhibit mitochondrial function; accordingly, farnesol reduced mitochondrial respiration. Dihydroceramide desaturase inhibition increases lipid droplet formation, which we observed in farnesol-treated cells, coupled with an increase in intracellular triacylglycerol species. Furthermore, inhibition of dihydroceramide desaturase with either farnesol or specific inhibitors impaired the ability of DCs to prime interferon-γ-producing T cells. The effect of farnesol on sphingolipid metabolism, triacylglycerol synthesis, and mitochondrial respiration was not dependent on PPAR-γ. In summary, our data reveal novel effects of farnesol on sphingolipid metabolism, neutral lipid synthesis, and mitochondrial function in DCs that affect their instruction of T cell cytokine secretion, indicating that C. albicans can manipulate host cell metabolism via farnesol secretion.IMPORTANCECandida albicans is a common commensal yeast, but it is also an opportunistic pathogen which is one of the leading causes of potentially lethal hospital-acquired infections. There is growing evidence that its overgrowth in the gut can influence diseases as diverse as alcohol-associated liver disease and COVID-19. Previously, we found that its quorum-sensing molecule, farnesol, alters the phenotype of dendritic cells differentiating from monocytes, impairing their ability to drive protective T cell responses. Here, we demonstrate that farnesol alters the metabolism of sphingolipids, important structural components of the membrane that also act as signaling molecules. In monocytes differentiating to dendritic cells, farnesol inhibited dihydroceramide desaturase, resulting in the accumulation of dihydroceramides and a reduction in ceramide levels. Farnesol impaired mitochondrial respiration, known to occur with an accumulation of dihydroceramides, and induced the accumulation of triacylglycerol and oil bodies. Inhibition of dihydroceramide desaturase resulted in the impaired ability of DCs to induce interferon-γ production by T cells. Thus, farnesol production by C. albicans could manipulate the function of dendritic cells by altering the sphingolipidome.


Asunto(s)
Candida albicans , Células Dendríticas , Farnesol , Monocitos , Percepción de Quorum , Esfingolípidos , Farnesol/farmacología , Farnesol/metabolismo , Humanos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/inmunología , Candida albicans/efectos de los fármacos , Candida albicans/metabolismo , Esfingolípidos/metabolismo , Percepción de Quorum/efectos de los fármacos , Monocitos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/microbiología , Monocitos/inmunología , PPAR gamma/metabolismo , PPAR gamma/genética , Espectrometría de Masas en Tándem , Citocinas/metabolismo
12.
PLoS Pathog ; 20(7): e1012389, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39078851

RESUMEN

Ergosterol is essential for fungal cell membrane integrity and growth, and numerous antifungal drugs target ergosterol. Inactivation or modification of ergosterol biosynthetic genes can lead to changes in antifungal drug susceptibility, filamentation and stress response. Here, we found that the ergosterol biosynthesis gene ERG251 is a hotspot for point mutations during adaptation to antifungal drug stress within two distinct genetic backgrounds of Candida albicans. Heterozygous point mutations led to single allele dysfunction of ERG251 and resulted in azole tolerance in both genetic backgrounds. This is the first known example of point mutations causing azole tolerance in C. albicans. Importantly, single allele dysfunction of ERG251 in combination with recurrent chromosome aneuploidies resulted in bona fide azole resistance. Homozygous deletions of ERG251 caused increased fitness in low concentrations of fluconazole and decreased fitness in rich medium, especially at low initial cell density. Homozygous deletions of ERG251 resulted in accumulation of ergosterol intermediates consistent with the fitness defect in rich medium. Dysfunction of ERG251, together with FLC exposure, resulted in decreased accumulation of the toxic sterol (14-ɑ-methylergosta-8,24(28)-dien-3ß,6α-diol) and increased accumulation of non-toxic alternative sterols. The altered sterol composition of the ERG251 mutants had pleiotropic effects on transcription, filamentation, and stress responses including cell membrane, osmotic and oxidative stress. Interestingly, while dysfunction of ERG251 resulted in azole tolerance, it also led to transcriptional upregulation of ZRT2, a membrane-bound Zinc transporter, in the presence of FLC, and overexpression of ZRT2 is sufficient to increase azole tolerance in wild-type C. albicans. Finally, in a murine model of systemic infection, homozygous deletion of ERG251 resulted in decreased virulence while the heterozygous deletion mutants maintain their pathogenicity. Overall, this study demonstrates that single allele dysfunction of ERG251 is a recurrent and effective mechanism of acquired azole tolerance. We propose that altered sterol composition resulting from ERG251 dysfunction mediates azole tolerance as well as pleiotropic effects on stress response, filamentation and virulence.


Asunto(s)
Antifúngicos , Candida albicans , Candidiasis , Farmacorresistencia Fúngica , Ergosterol , Proteínas Fúngicas , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/metabolismo , Antifúngicos/farmacología , Ratones , Farmacorresistencia Fúngica/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Animales , Candidiasis/microbiología , Candidiasis/metabolismo , Candidiasis/tratamiento farmacológico , Ergosterol/metabolismo , Azoles/farmacología , Esteroles/metabolismo , Fenotipo , Estrés Fisiológico , Pruebas de Sensibilidad Microbiana , Fluconazol/farmacología
13.
J Proteomics ; 303: 105228, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38878881

RESUMEN

Candida albicans, a significant human pathogenic fungus, employs hydrolytic proteases for host invasion. Conventional antifungal agents are reported with resistance issues from around the world. This study investigates the role of Bacillus licheniformis extracellular proteins (ECP) as effective antifungal peptides (AFPs). The aim was to identify and characterize the ECP of B. licheniformis through LC-MS/MS and bioinformatics analysis. LC-MS/MS analysis identified 326 proteins with 69 putative ECP, further analyzed in silico. Of these, 21 peptides exhibited antifungal properties revealed by classAMP tool and are predominantly anionic. Peptide-protein docking revealed interactions between AFPs like Peptide chain release factor 1 (Q65DV1_Seq1: SASEQLSDAK) and Putative carboxy peptidase (Q65IF0_Seq7: SDSSLEDQDFILESK) with C. albicans virulent SAP5 proteins (PDB ID 2QZX), forming hydrogen bonds and significant Pi-Pi interactions. The identification of B. licheniformis ECP is the novelty of the study that sheds light on their antifungal potential. The identified AFPs, particularly those interacting with bonafide pharmaceutical targets SAP5 of C. albicans represent promising avenues for the development of antifungal treatments with AFPs that could be the pursuit of a novel therapeutic strategy against C. albicans. SIGNIFICANCE OF STUDY: The purpose of this work was to carry out proteomic profiling of the secretome of B. licheniformis. Previously, the efficacy of Bacillus licheniformis extracellular proteins against Candida albicans was investigated and documented in a recently communicated manuscript, showcasing the antifungal activity of these proteins. In order to achieve high-throughput identification of ES (Excretory-secretory) proteins, the utilization of liquid chromatography tandem mass spectrometry (LC-MS) was utilized. There was a lack of comprehensive research on AFPs in B. licheniformis, nevertheless. The proteins secreted by B. licheniformis in liquid medium were initially discovered using liquid chromatography-tandem mass spectrometry (LC-MS) analysis and identification in order to immediately characterize the unidentified active metabolites in fermentation broth.


Asunto(s)
Antifúngicos , Bacillus licheniformis , Proteínas Bacterianas , Candida albicans , Espectrometría de Masas en Tándem , Candida albicans/efectos de los fármacos , Candida albicans/metabolismo , Antifúngicos/farmacología , Bacillus licheniformis/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Cromatografía Liquida , Humanos , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/química , Cromatografía Líquida con Espectrometría de Masas
14.
PLoS Biol ; 22(6): e3002694, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38900845

RESUMEN

Fungi and bacteria coexist in many polymicrobial communities, yet the molecular basis of their interactions remains poorly understood. Here, we show that the fungus Candida albicans sequesters essential magnesium ions from the bacterium Pseudomonas aeruginosa. To counteract fungal Mg2+ sequestration, P. aeruginosa expresses the Mg2+ transporter MgtA when Mg2+ levels are low. Thus, loss of MgtA specifically impairs P. aeruginosa in co-culture with C. albicans, but fitness can be restored by supplementing Mg2+. Using a panel of fungi and bacteria, we show that Mg2+ sequestration is a general mechanism of fungal antagonism against gram-negative bacteria. Mg2+ limitation enhances bacterial resistance to polymyxin antibiotics like colistin, which target gram-negative bacterial membranes. Indeed, experimental evolution reveals that P. aeruginosa evolves C. albicans-dependent colistin resistance via non-canonical means; antifungal treatment renders resistant bacteria colistin-sensitive. Our work suggests that fungal-bacterial competition could profoundly impact polymicrobial infection treatment with antibiotics of last resort.


Asunto(s)
Antibacterianos , Candida albicans , Colistina , Magnesio , Pseudomonas aeruginosa , Magnesio/farmacología , Magnesio/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Antibacterianos/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/metabolismo , Colistina/farmacología , Pruebas de Sensibilidad Microbiana , Polimixinas/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Interacciones Microbianas/efectos de los fármacos
15.
Appl Environ Microbiol ; 90(7): e0087424, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-38940563

RESUMEN

Farnesol salvage, a two-step pathway converting farnesol to farnesyl pyrophosphate (FPP), occurs in bacteria, plants, and animals. This paper investigates the presence of this pathway in fungi. Through bioinformatics, biochemistry, and physiological analyses, we demonstrate its absence in the yeasts Saccharomyces cerevisiae and Candida albicans, suggesting a likely absence across fungi. We screened 1,053 fungal genomes, including 34 from C. albicans, for potential homologs to four genes (Arabidopsis thaliana AtFOLK, AtVTE5, AtVTE6, and Plasmodium falciparum PfPOLK) known to accomplish farnesol/prenol salvage in other organisms. Additionally, we showed that 3H-farnesol was not converted to FPP or any other phosphorylated prenol, and exogenous farnesol was not metabolized within 90 minutes at any phase of growth and did not rescue cells from the toxic effects of atorvastatin, but it did elevate the levels of intracellular farnesol (Fi). All these experiments were conducted with C. albicans. In sum, we found no evidence for farnesol salvage in fungi. IMPORTANCE: The absence of farnesol salvage constitutes a major difference in the metabolic capabilities of fungi. In terms of fungal physiology, the lack of farnesol salvage pathways relates to how farnesol acts as a quorum-sensing molecule in Candida albicans and why farnesol should be investigated for use in combination with other known antifungal antibiotics. Its absence is essential for a model (K. W. Nickerson et al., Microbiol Mol Biol Rev 88:e00081-22, 2024), wherein protein farnesylation, protein chaperones, and the unfolded protein response are combined under the unifying umbrella of a cell's intracellular farnesol (Fi). In terms of human health, farnesol should have at least two different modes of action depending on whether those cells have farnesol salvage. Because animals have farnesol salvage, we can now see the importance of dietary prenols as well as the potential importance of farnesol in treating neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and multiple sclerosis.


Asunto(s)
Candida albicans , Farnesol , Farnesol/metabolismo , Candida albicans/efectos de los fármacos , Candida albicans/genética , Candida albicans/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/crecimiento & desarrollo , Genoma Fúngico , Sesquiterpenos
16.
PLoS Biol ; 22(6): e3002693, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38905306

RESUMEN

Candida albicans is a commensal of the human microbiota that can form biofilms on implanted medical devices. These biofilms are tolerant to antifungals and to the host immune system. To identify novel genes modulating C. albicans biofilm formation, we performed a large-scale screen with 2,454 C. albicans doxycycline-dependent overexpression strains and identified 16 genes whose overexpression significantly hampered biofilm formation. Among those, overexpression of the ZCF15 and ZCF26 paralogs that encode transcription factors and have orthologs only in biofilm-forming species of the Candida clade, caused impaired biofilm formation both in vitro and in vivo. Interestingly, overexpression of ZCF15 impeded biofilm formation without any defect in hyphal growth. Transcript profiling, transcription factor binding, and phenotypic microarray analyses conducted upon overexpression of ZCF15 and ZCF26 demonstrated their role in reprogramming cellular metabolism by regulating central metabolism including glyoxylate and tricarboxylic acid cycle genes. Taken together, this study has identified a new set of biofilm regulators, including ZCF15 and ZCF26, that appear to control biofilm development through their specific role in metabolic remodeling.


Asunto(s)
Biopelículas , Candida albicans , Proteínas Fúngicas , Regulación Fúngica de la Expresión Génica , Factores de Transcripción , Biopelículas/crecimiento & desarrollo , Candida albicans/genética , Candida albicans/metabolismo , Candida albicans/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Animales , Plancton/metabolismo , Glioxilatos/metabolismo , Perfilación de la Expresión Génica/métodos , Ratones , Ciclo del Ácido Cítrico , Hifa/metabolismo , Hifa/crecimiento & desarrollo , Hifa/genética , Candidiasis/microbiología , Reprogramación Metabólica
17.
J Biol Chem ; 300(7): 107444, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38838772

RESUMEN

Candida albicans is an opportunistic fungal pathogen that can switch between yeast and hyphal morphologies depending on the environmental cues it receives. The switch to hyphal form is crucial for the establishment of invasive infections. The hyphal form is also characterized by the cell surface expression of hyphae-specific proteins, many of which are GPI-anchored and important determinants of its virulence. The coordination between hyphal morphogenesis and the expression of GPI-anchored proteins is made possible by an interesting cross-talk between GPI biosynthesis and the cAMP-PKA signaling cascade in the fungus; a parallel interaction is not found in its human host. On the other hand, in the nonpathogenic yeast, Saccharomyces cerevisiae, GPI biosynthesis is shut down when filamentation is activated and vice versa. This too is achieved by a cross-talk between GPI biosynthesis and cAMP-PKA signaling. How are diametrically opposite effects obtained from the cross-talk between two reasonably well-conserved pathways present ubiquitously across eukarya? This Review attempts to provide a model to explain these differences. In order to do so, it first provides an overview of the two pathways for the interested reader, highlighting the similarities and differences that are observed in C. albicans versus the well-studied S. cerevisiae model, before going on to explain how the different mechanisms of regulation are effected. While commonalities enable the development of generalized theories, it is hoped that a more nuanced approach, that takes into consideration species-specific differences, will enable organism-specific understanding of these processes and contribute to the development of targeted therapies.


Asunto(s)
Candida albicans , Proteínas Quinasas Dependientes de AMP Cíclico , AMP Cíclico , Hifa , Saccharomyces cerevisiae , Transducción de Señal , Candida albicans/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Hifa/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Glicosilfosfatidilinositoles/metabolismo , Glicosilfosfatidilinositoles/biosíntesis , Humanos , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
18.
Mol Biol Cell ; 35(7): ar99, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38776129

RESUMEN

The human fungal pathogen Candida albicans can cause lethal systemic infections due to its ability to resist stress from the host and to undergo invasive hyphal growth. Previous studies showed that plasma membrane MCC/eisosome domains were important for virulence by promoting the ability of Sur7 to mediate normal cell wall morphogenesis and stress resistance. The sur7Δ mutant displayed abnormal clusters of PI4,5P2, suggesting that misregulation of this lipid underlies the sur7Δ phenotype. To test this, we increased PI4,5P2 levels by deleting combinations of the three PI4,5P2 5' phosphatase genes (INP51, INP52, and INP54) and found that some combinations, such as inp51Δ inp52Δ, gave phenotypes similar the sur7Δ mutant. In contrast, deleting one copy of MSS4, the gene that encodes the 5' kinase needed to create PI4,5P2, reduced the abnormal PI4,5P2 clusters and also decreased the abnormal cell wall and stress sensitive phenotypes of the sur7Δ mutant. Additional studies support a model that the abnormal PI4,5P2 patches recruit septin proteins, which in turn promote aberrant cell wall growth. These results identify Sur7 as a novel regulator of PI4,5P2 and highlight the critical role of PI4,5P2 in the regulation of C. albicans virulence properties.


Asunto(s)
Candida albicans , Pared Celular , Proteínas Fúngicas , Morfogénesis , Candida albicans/metabolismo , Candida albicans/patogenicidad , Candida albicans/genética , Candida albicans/fisiología , Pared Celular/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Virulencia , Estrés Fisiológico , Fosfatidilinositol 4,5-Difosfato/metabolismo , Hifa/metabolismo , Membrana Celular/metabolismo , Regulación Fúngica de la Expresión Génica , Monoéster Fosfórico Hidrolasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética
19.
Nanomedicine ; 59: 102750, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38734040

RESUMEN

The human pathogenic fungus Candida albicans damages epithelial cells during superficial infections. Here we use three-dimensional-sequential-confocal Raman spectroscopic imaging and atomic force microscopy to investigate the interaction of C. albicans wild type cells, the secreted C. albicans peptide toxin candidalysin and mutant cells lacking candidalysin with epithelial cells. The candidalysin is responsible for epithelial cell damage and exhibits in its deuterated form an identifiable Raman signal in a frequency region distinct from the cellular frequency region. Vibration modes at 2100-2200 cm-1 attributed to carbon­deuterium bending and at 477 cm-1, attributed to the nitrogen­deuterium out-of-plane bending, found around the nucleus, can be assigned to deuterated candidalysin. Atomic force microscopy visualized 100 nm deep lesions on the cell and force-distance curves indicate the higher adhesion on pore surrounding after incubation with candidalysin. Candidalysin targets the plasma membrane, but is also found inside of the cytosol of epithelial cells during C. albicans infection.


Asunto(s)
Candida albicans , Células Epiteliales , Microscopía de Fuerza Atómica , Espectrometría Raman , Candida albicans/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Microscopía de Fuerza Atómica/métodos , Espectrometría Raman/métodos , Humanos , Candidiasis/microbiología , Microscopía Confocal/métodos , Marcaje Isotópico , Imagenología Tridimensional , Deuterio/química
20.
Cell Host Microbe ; 32(6): 964-979.e7, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38754418

RESUMEN

The gut microbiota is closely linked to atherosclerosis. However, the role of intestinal fungi, essential members of the complex microbial community, in atherosclerosis is poorly understood. Herein, we show that gut fungi dysbiosis is implicated in patients with dyslipidemia, characterized by higher levels of Candida albicans (C. albicans), which are positively correlated with plasma total cholesterol and low-density lipoprotein-cholesterol (LDL-C) levels. Furthermore, C. albicans colonization aggravates atherosclerosis progression in a mouse model of the disease. Through gain- and loss-of-function studies, we show that an intestinal hypoxia-inducible factor 2α (HIF-2α)-ceramide pathway mediates the effect of C. albicans. Mechanistically, formyl-methionine, a metabolite of C. albicans, activates intestinal HIF-2α signaling, which drives increased ceramide synthesis to accelerate atherosclerosis. Administration of the HIF-2α selective antagonist PT2385 alleviates atherosclerosis in mice by reducing ceramide levels. Our findings identify a role for intestinal fungi in atherosclerosis progression and highlight the intestinal HIF-2α-ceramide pathway as a target for atherosclerosis treatment.


Asunto(s)
Aterosclerosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Candida albicans , Ceramidas , Transducción de Señal , Animales , Candida albicans/metabolismo , Aterosclerosis/microbiología , Aterosclerosis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ratones , Humanos , Ceramidas/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino , Microbioma Gastrointestinal/fisiología , Intestinos/microbiología , Intestinos/patología , Disbiosis/microbiología , Femenino , Candidiasis/microbiología , Candidiasis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...