Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.668
Filter
1.
Virulence ; 15(1): 2412750, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39370643

ABSTRACT

Candida albicans is an opportunistic fungal pathogen known for surviving in various nutrient-limited conditions within the host and causing infections. Our prior research revealed that Hfl1p, an archaeal histone-like or Hap5-like protein, is linked to mitochondrial ATP generation and yeast-hyphae morphogenesis. However, the specific roles of Hfl1p in these virulence behaviours, through its function in the CBF/NF-Y complex or as a DNA polymerase II subunit, remain unclear. This study explores Hfl1p's diverse functions in energy metabolism and morphogenesis. By combining proteomic analysis and phenotypic evaluations of the hfl1Δ/hfl1Δ mutant with ChIP data, we found that Hfl1p significantly impacts mitochondrial DNA-encoded CI subunits, the tricarboxylic acid (TCA) cycle, and morphogenetic pathways. This influence occurs either independently or alongside other transcription factors recognizing a conserved DNA motif (TAXXTAATTA). These findings emphasize Hfl1p's critical role in linking carbon metabolism and mitochondrial respiration to the yeast-to-filamentous form transition, enhancing our understanding of C. albicans' metabolic adaptability during morphological transition, an important pathogenic trait of this fungus. This could help identify therapeutic targets by disrupting the relationship between energy metabolism and cell morphology in C. albicans.


Subject(s)
Candida albicans , Energy Metabolism , Fungal Proteins , Gene Expression Regulation, Fungal , Transcription Factors , Candida albicans/genetics , Candida albicans/growth & development , Candida albicans/metabolism , Candida albicans/pathogenicity , Fungal Proteins/genetics , Fungal Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Hyphae/growth & development , Hyphae/genetics , Mitochondria/metabolism , Mitochondria/genetics , Proteomics , Genome, Mitochondrial , Cell Nucleus/metabolism
2.
Med Mycol J ; 65(3): 67-74, 2024.
Article in English | MEDLINE | ID: mdl-39218649

ABSTRACT

Candida species are common human pathogens that cause a wide range of diseases ranging from superficial to invasive candidiasis. However, basic studies focusing on the mechanisms underlying these diseases are limited. This article reviews our previous research on the mechanisms of superficial and invasive candidiasis, the virulence of Candida species, and Candida species fitness to hosts. Regarding invasive candidiasis, we focused on two types of infections: ocular candidiasis and endogenous candidiasis from the gastrointestinal tract. Using an established ocular candidiasis mouse model, along with retrospective epidemiological research, we found a strong association between Candida albicans and ocular candidiasis. Regarding endogenous candidiasis, research using Candida auris indicated that invasive strains had a higher capability for gastrointestinal tract colonization and showed greater dissemination compared with non-invasive strains. In terms of superficial candidiasis, we focused on the defense mechanism in vulvovaginal candidiasis. The results suggested that stimulated invariant natural killer T cells played a protective role against C. albicans vaginal infection and might be a therapeutic target for vulvovaginal candidiasis. Concerning Candida species fitness, we focused on environmental factors, particularly oxygen concentration, and evaluated biofilm formation under various oxygen concentrations, revealing that each Candida species favored different oxygen concentrations. In particular, Candida tropicalis showed greater biofilm formation under hypoxic conditions. Our research revealed several insights for understanding the exact mechanisms of candidiasis, which might lead to better control of Candida species infections and appropriate treatment.


Subject(s)
Biofilms , Candida , Candidiasis , Disease Models, Animal , Animals , Mice , Candida/pathogenicity , Candidiasis/microbiology , Humans , Biofilms/growth & development , Virulence , Female , Candidiasis, Vulvovaginal/microbiology , Eye Infections, Fungal/microbiology , Candida albicans/pathogenicity , Candidiasis, Invasive/microbiology
3.
Virulence ; 15(1): 2405616, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39316797

ABSTRACT

Candida albicans, a part of normal flora, is an opportunistic fungal pathogen and causes severe health issues in immunocompromised patients. Its pathogenicity is intricately linked to the transcriptional regulation of its metabolic pathways. Paf1 complex (Paf1C) is a crucial transcriptional regulator that is highly conserved in eukaryotes. The objective of this study was to explore the role of Paf1C in the metabolic pathways and how it influences the pathogenicity of C. albicans. Paf1C knockout mutant strains of C. albicans (ctr9Δ/Δ, leo1Δ/Δ, and cdc73Δ/Δ) were generated using the CRISPR-Cas9 system. To investigate the effect of Paf1C on pathogenicity, macrophage interaction assays and mouse survival tests were conducted. The growth patterns of the Paf1C knockout mutants were analyzed through spotting assays and growth curve measurements. Transcriptome analysis was conducted under yeast conditions (30°C without serum) and hyphal conditions (37°C with 10% FBS), to further elucidate the role of Paf1C in the pathogenicity of C. albicans. CTR9 deletion resulted in the attenuation of C. albicans virulence, in macrophage and mouse models. Furthermore, we confirmed that the reduced virulence of the ctr9Δ/Δ mutant can be attributed to a decrease in C. albicans cell abundance. Moreover, transcriptome analysis revealed that metabolic processes required for cell proliferation are impaired in ctr9Δ/Δ mutant. Notably, CTR9 deletion led to the downregulation of methionine biosynthetic genes and the cAMP-PKA signaling pathway-related hypha essential genes, which are pivotal for virulence. Our results suggest that Ctr9-regulated methionine metabolism is a crucial factor for determining C. albicans pathogenicity.


Subject(s)
Candida albicans , Candidiasis , Fungal Proteins , Gene Expression Regulation, Fungal , Macrophages , Methionine , Candida albicans/pathogenicity , Candida albicans/genetics , Candida albicans/metabolism , Animals , Mice , Virulence , Fungal Proteins/genetics , Fungal Proteins/metabolism , Methionine/metabolism , Candidiasis/microbiology , Macrophages/microbiology , Mice, Inbred BALB C , Female , RAW 264.7 Cells , Hyphae/growth & development , Hyphae/genetics , Hyphae/metabolism , Gene Expression Profiling
4.
Cell Rep ; 43(8): 114601, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39126650

ABSTRACT

Fungal pathogens such as Candida albicans pose a significant threat to human health with limited treatment options available. One strategy to expand the therapeutic target space is to identify genes important for pathogen growth in host-relevant environments. Here, we leverage a pooled functional genomic screening strategy to identify genes important for fitness of C. albicans in diverse conditions. We identify an essential gene with no known Saccharomyces cerevisiae homolog, C1_09670C, and demonstrate that it encodes subunit 3 of replication factor A (Rfa3). Furthermore, we apply computational analyses to identify functionally coherent gene clusters and predict gene function. Through this approach, we predict the cell-cycle-associated function of C3_06880W, a previously uncharacterized gene required for fitness specifically at elevated temperatures, and follow-up assays confirm that C3_06880W encodes Iml3, a component of the C. albicans kinetochore with roles in virulence in vivo. Overall, this work reveals insights into the vulnerabilities of C. albicans.


Subject(s)
Candida albicans , Fungal Proteins , Candida albicans/genetics , Candida albicans/pathogenicity , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genetic Fitness , Genomics/methods , Virulence/genetics , Genome, Fungal , Humans
5.
mSphere ; 9(9): e0028724, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39115319

ABSTRACT

The uridine derivatives UDP-glucose and UDP-N-acetylglucosamine are important for cell wall construction as they are the precursors for the synthesis of ß-1,3-glucan and chitin, respectively. Previous studies have demonstrated attenuated virulence of uridine auxotrophs in mice, which has been attributed to insufficient uridine levels for growth in the host. We have discovered that uridine deprivation in the uridine auxotroph ura3ΔΔ disrupts cell wall architecture by increasing surface mannans, exposing ß-1,3-glucan and chitin, and decreasing UDP-sugar levels. Cell wall architecture and UDP-sugars can be rescued with uridine supplementation. The cell wall architectural disruptions in the ura3ΔΔ mutant also impact immune activation since the mutant elicited greater TNFα secretion from RAW264.7 macrophages than wild type. To determine if cell wall defects contributed to decreased virulence in the ura3ΔΔ mutant, we used a murine model of systemic infection. Mice infected with the ura3ΔΔ mutant exhibited increased survival and reduced kidney fungal burden compared with mice infected with wild type. However, suppression of the immune response with cyclophosphamide did not rescue virulence in mice infected with the ura3ΔΔ mutant, indicating the attenuation in virulence of uridine auxotrophs can be attributed to decreased growth in the host but not increased exposure of ß-1,3-glucan. Moreover, the ura3ΔΔ mutant is unable to grow on ex vivo kidney agar, which demonstrates its inability to colonize the kidneys due to poor growth. Thus, although uridine auxotrophy elicits changes to cell wall architecture that increase the exposure of immunogenic polymers, metabolic fitness costs more strongly drive the observed virulence attenuation.IMPORTANCECandida albicans is a common cause of bloodstream infections (candidemia). Treatment of these bloodstream infections is made difficult because of increasing antifungal resistance and drug toxicity. Thus, new tactics are needed for antifungal drug development, with immunotherapy being of particular interest. The cell wall of C. albicans is composed of highly immunogenic polymers, particularly ß-1,3-glucan. However, ß-1,3-glucan is naturally masked by an outer layer of mannoproteins, which hampers the detection of the fungus by the host immune system. Alteration in cell wall components has been shown to increase ß-1,3-glucan exposure; however, it is unknown how the inability to synthesize precursors to cell wall components affects unmasking. Here, we demonstrate how cell wall architecture is altered in response to a deficit in precursors for cell wall synthesis and how uridine is a crucial component of these precursors.


Subject(s)
Candida albicans , Candidiasis , Cell Wall , Uridine , beta-Glucans , Animals , Uridine/metabolism , Mice , Cell Wall/metabolism , beta-Glucans/metabolism , Virulence , Candida albicans/pathogenicity , Candida albicans/genetics , Candida albicans/metabolism , Candidiasis/microbiology , RAW 264.7 Cells , Female , Disease Models, Animal , Macrophages/microbiology , Macrophages/immunology , Chitin/metabolism
6.
mSphere ; 9(9): e0028224, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39171917

ABSTRACT

Microbial gene expression measurements derived from infected organs are invaluable to understand pathogenesis. However, current methods are limited to "bulk" analyses that neglect microbial cell heterogeneity and the lesion's spatial architecture. Here, we report the use of hybridization chain reaction RNA fluorescence in situ hybridization (HCR RNA-FISH) to visualize and quantify Candida albicans transcripts at single-cell resolution in tongues of infected mice. The method is compatible with fixed-frozen and formalin-fixed paraffin-embedded tissues. We document cell-to-cell variation and intriguing spatiotemporal expression patterns for C. albicans mRNAs that encode products implicated in oral candidiasis. The approach provides a spatial dimension to gene expression analyses of host-Candida interactions. IMPORTANCE: Candida albicans is a fungal pathobiont inhabiting multiple mucosal surfaces of the human body. Immunosuppression, antibiotic-induced microbial dysbiosis, or implanted medical devices can impair mucosal integrity enabling C. albicans to overgrow and disseminate, causing either mucosal diseases such as oropharyngeal candidiasis or life-threatening systemic infections. Profiling fungal genes that are expressed in the infected mucosa or in any other infected organ is paramount to understand pathogenesis. Ideally, these transcript profiling measurements should reveal the expression of any gene at the single-cell level. The resolution typically achieved with current approaches, however, limits most gene expression measurements to cell population averages. The approach described in this report provides a means to dissect fungal gene expression in infected tissues at single-cell resolution.


Subject(s)
Candida albicans , Candidiasis, Oral , In Situ Hybridization, Fluorescence , Single-Cell Analysis , Tongue , Animals , Candida albicans/genetics , Candida albicans/pathogenicity , Mice , Single-Cell Analysis/methods , Candidiasis, Oral/microbiology , Tongue/microbiology , In Situ Hybridization, Fluorescence/methods , Gene Expression Profiling , Spatio-Temporal Analysis , Female , Gene Expression Regulation, Fungal , Disease Models, Animal , Host-Pathogen Interactions/genetics
7.
Microb Biotechnol ; 17(8): e14547, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39160430

ABSTRACT

Root caries is a subtype of dental caries that predominantly impacts older adults. The occurrence and progression of root caries are associated with the homeostasis of dental plaque biofilm, and microbial synergistic and antagonistic interactions in the biofilm play a significant role in maintaining the oral microecological balance. The objective of the current study was to investigate the role of Veillonella parvula in the microbial interactions and the pathogenesis of root caries. The analysis of clinical samples from patients with/without root caries revealed that Veillonella and V. parvula were abundant in the saliva of patients with root caries. More importantly, a significantly increased colonization of V. parvula was observed in root carious lesions. Further in vitro biofilm and animal study showed that V. parvula colonization increased the abundance and virulence of Streptococcus mutans and Candida albicans, leading to the formation of a polymicrobial biofilm with enhanced anti-stress capacity and cariogenicity, consequently exacerbating the severity of carious lesions. Our results indicate the critical role of V. parvula infection in the occurrence of root caries, providing a new insight for the etiological investigation and prevention of root caries.


Subject(s)
Biofilms , Candida albicans , Microbial Interactions , Root Caries , Streptococcus mutans , Veillonella , Streptococcus mutans/physiology , Streptococcus mutans/pathogenicity , Streptococcus mutans/genetics , Candida albicans/pathogenicity , Candida albicans/physiology , Humans , Biofilms/growth & development , Root Caries/microbiology , Animals , Veillonella/genetics , Veillonella/physiology , Saliva/microbiology , Disease Models, Animal , Male , Female
8.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125628

ABSTRACT

Staphylococcus aureus, particularly drug-resistant strains, poses significant challenges in healthcare due to its ability to form biofilms, which confer increased resistance to antibiotics and immune responses. Building on previous knowledge that several flavonoids exhibit antibiofilm activity, this study sought to identify a novel flavonoid capable of effectively inhibiting biofilm formation and virulence factor production in S. aureus strains including MRSA. Among the 19 flavonoid-like compounds tested, 3,2'-dihydroxyflavone (3,2'-DHF) was identified for the first time as inhibiting biofilm formation and virulence factors in S. aureus with an MIC 75 µg/mL. The antibiofilm activity was further confirmed by microscopic methods. Notably, 3,2'-DHF at 5 µg/mL was effective in inhibiting both mono- and polymicrobial biofilms involving S. aureus and Candida albicans, a common co-pathogen. 3,2'-DHF reduces hemolytic activity, slime production, and the expression of key virulence factors such as hemolysin gene hla and nuclease gene nuc1 in S. aureus. These findings highlight the potential of 3,2'-DHF as a novel antibiofilm and antivirulence agent against both bacterial and fungal biofilms, offering a promising alternative to traditional antibiotics in the treatment of biofilm-associated infections.


Subject(s)
Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Staphylococcus aureus , Virulence Factors , Biofilms/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Virulence Factors/genetics , Anti-Bacterial Agents/pharmacology , Candida albicans/drug effects , Candida albicans/pathogenicity , Flavones/pharmacology , Flavonoids/pharmacology , Virulence/drug effects , Humans
9.
Cell Host Microbe ; 32(9): 1536-1551.e6, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39084229

ABSTRACT

Candida albicans stably colonizes humans but is the leading cause of hospital-acquired fungemia. Traditionally, masking immunogenic moieties has been viewed as a tactic for immune evasion. Here, we demonstrate that C. albicans blocks type I interferon (IFN-I) signaling via translocating an effector protein Cmi1 into host cells. Mechanistically, Cmi1 binds and inhibits TANK-binding kinase 1 (TBK1) to abrogate IFN-regulatory factor 3 (IRF3) phosphorylation, thereby suppressing the IFN-I cascade. Murine infection with a cmi1 mutant displays an exaggerated IFN-I response in both kidneys and bone-marrow-derived macrophages, leading to rapid fungal clearance and host survival. Remarkably, the lack of CMI1 compromises gut commensalism and increases IFN-I response in mouse colonic cells. These phenotypes of cmi1 are rescued by the depletion of IFN-I receptor. This work establishes the importance of TBK1 inhibition in fungal pathogenesis and reveals that a human commensal-pathogenic fungus significantly impacts host immunity during gut colonization and infection via delivering effector proteins into host cells.


Subject(s)
Candida albicans , Interferon Regulatory Factor-3 , Macrophages , Protein Serine-Threonine Kinases , Animals , Humans , Mice , Candida albicans/immunology , Candida albicans/pathogenicity , Candidiasis/immunology , Candidiasis/microbiology , Fungal Proteins/metabolism , Fungal Proteins/genetics , Host-Pathogen Interactions , Immune Evasion , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Interferon Type I/immunology , Kidney/microbiology , Kidney/immunology , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Symbiosis
10.
J Glob Antimicrob Resist ; 38: 341-348, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39059553

ABSTRACT

OBJECTIVES: This study evaluated the role of Upc2 in the development of azole resistance in Candida albicans isolates from Lebanese hospitalized patients and determined a correlation between resistance and virulence. METHODS: The UPC2 gene which codes for an ergosterol biosynthesis regulator was sequenced and analysed in two azole-resistant and one azole-susceptible C. albicans isolates. An amino acid substitution screening was carried out on Upc2 with a focus on its ligand binding domain (LBD) known to interact with ergosterol. Then, Upc2 protein secondary structure prediction and homology modelling were conducted, followed by total plasma membrane ergosterol and cell wall chitin quantifications. For virulence, mouse models of systemic infection were generated and an agar adhesion and invasion test was performed. RESULTS: Azole-resistant isolates harboured novel amino acid substitutions in the LBD of Upc2 and changes in protein secondary structures were observed. In addition, these isolates exhibited a significant increase in plasma membrane ergosterol content. Resistance and virulence were inversely correlated while increased cell wall chitin concentration does not seem to be linked to resistance since even though we observed an increase in chitin concentration, it was not statistically significant. CONCLUSIONS: The azole-resistant C. albicans isolates harboured novel amino acid substitutions in the LBD of Upc2 which are speculated to induce an increase in plasma membrane ergosterol content, preventing the binding of azoles to their target, resulting in resistance.


Subject(s)
Antifungal Agents , Azoles , Candida albicans , Candidiasis , Drug Resistance, Fungal , Ergosterol , Fungal Proteins , Microbial Sensitivity Tests , Mutation , Candida albicans/genetics , Candida albicans/drug effects , Candida albicans/isolation & purification , Candida albicans/pathogenicity , Lebanon , Humans , Azoles/pharmacology , Antifungal Agents/pharmacology , Animals , Fungal Proteins/genetics , Fungal Proteins/metabolism , Virulence , Mice , Candidiasis/microbiology , Amino Acid Substitution , Chitin/metabolism , Female , Cell Wall , Disease Models, Animal
11.
Mol Cell Proteomics ; 23(9): 100818, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39047911

ABSTRACT

Candida albicans is a diploid pathogen known for its ability to live as a commensal fungus in healthy individuals but causing both superficial infections and disseminated candidiasis in immunocompromised patients where it is associated with high morbidity and mortality. Its success in colonizing the human host is attributed to a wide range of virulence traits that modulate interactions between the host and the pathogen, such as optimal growth rate at 37 °C, the ability to switch between yeast and hyphal forms, and a remarkable genomic and phenotypic plasticity. A fascinating aspect of its biology is a prominent heterogeneous proteome that arises from frequent genomic rearrangements, high allelic variation, and high levels of amino acid misincorporations in proteins. This leads to increased morphological and physiological phenotypic diversity of high adaptive potential, but the scope of such protein mistranslation is poorly understood due to technical difficulties in detecting and quantifying amino acid misincorporation events in complex protein samples. We have developed and optimized mass spectrometry and bioinformatics pipelines capable of identifying rare amino acid misincorporation events at the proteome level. We have also analyzed the proteomic profile of an engineered C. albicans strain that exhibits high level of leucine misincorporation at protein CUG sites and employed an in vivo quantitative gain-of-function fluorescence reporter system to validate our LC-MS/MS data. C. albicans misincorporates amino acids above the background level at protein sites of diverse codons, particularly at CUG, confirming our previous data on the quantification of leucine incorporation at single CUG sites of recombinant reporter proteins, but increasing misincorporation of Leucine at these sites does not alter the translational fidelity of the other codons. These findings indicate that the C. albicans statistical proteome exceeds prior estimates, suggesting that its highly plastic phenome may also be modulated by environmental factors due to translational ambiguity.


Subject(s)
Candida albicans , Fungal Proteins , Protein Biosynthesis , Proteogenomics , Candida albicans/metabolism , Candida albicans/genetics , Candida albicans/pathogenicity , Fungal Proteins/metabolism , Fungal Proteins/genetics , Proteogenomics/methods , Humans , Proteome/metabolism , Codon
12.
mSphere ; 9(8): e0064123, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39012122

ABSTRACT

Fungi were some of the earliest organismal systems used to explore mutational processes and its phenotypic consequences on members of a species. Yeasts that cause significant human disease were quickly incorporated into these investigations to define the genetic and phenotypic drivers of virulence. Among Candida species, Candida albicans has emerged as a model for studying genomic processes of evolution because of its clinical relevance, relatively small genome, and ability to tolerate complex chromosomal changes. Here, we describe major recent findings that used evolution of strains from defined genetic backgrounds to delineate mutational and adaptative processes and include how nascent exploration into naturally occurring variation is contributing to these conceptual frameworks. Ultimately, efforts to discern adaptive mechanisms used by C. albicans will continue to divulge new biology and can better inform treatment regimens for the increasing prevalence of fungal disease.


Subject(s)
Candida albicans , Genetic Variation , Candida albicans/genetics , Candida albicans/pathogenicity , Candida albicans/classification , Humans , Evolution, Molecular , Genome, Fungal , Virulence/genetics , Candidiasis/microbiology , Biological Evolution
13.
mBio ; 15(8): e0124924, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38949302

ABSTRACT

Protein kinases are critical regulatory proteins in both prokaryotes and eukaryotes. Accordingly, protein kinases represent a common drug target for a wide range of human diseases. Therefore, understanding protein kinase function in human pathogens such as the fungus Candida albicans is likely to extend our knowledge of its pathobiology and identify new potential therapies. To facilitate the study of C. albicans protein kinases, we constructed a library of 99 non-essential protein kinase homozygous deletion mutants marked with barcodes in the widely used SN genetic background. Here, we describe the construction of this library and the characterization of the competitive fitness of the protein kinase mutants under 11 different growth and stress conditions. We also screened the library for protein kinase mutants with altered filamentation and biofilm formation, two critical virulence traits of C. albicans. An extensive network of protein kinases governs these virulence traits in a manner highly dependent on the specific environmental conditions. Studies on specific protein kinases revealed that (i) the cell wall integrity MAPK pathway plays a condition-dependent role in filament initiation and elongation; (ii) the hyper-osmolar glycerol MAPK pathway is required for both filamentation and biofilm formation, particularly in the setting of in vivo catheter infection; and (iii) Sok1 is dispensable for filamentation in hypoxic environments at the basal level of a biofilm but is required for filamentation in normoxia. In addition to providing a new genetic resource for the community, these observations emphasize the environmentally contingent function of C. albicans protein kinases.IMPORTANCECandida albicans is one of the most common causes of fungal disease in humans for which new therapies are needed. Protein kinases are key regulatory proteins and are increasingly targeted by drugs for the treatment of a wide range of diseases. Understanding protein kinase function in C. albicans pathogenesis may facilitate the development of new antifungal drugs. Here, we describe a new library of 99 protein kinase deletion mutants to facilitate the study of protein kinases. Furthermore, we show that the function of protein kinases in two virulence-related processes, filamentation and biofilm formation, is dependent on the specific environmental conditions.


Subject(s)
Biofilms , Candida albicans , Protein Kinases , Candida albicans/genetics , Candida albicans/enzymology , Candida albicans/pathogenicity , Candida albicans/physiology , Biofilms/growth & development , Protein Kinases/genetics , Protein Kinases/metabolism , Virulence , Animals , Fungal Proteins/genetics , Fungal Proteins/metabolism , Candidiasis/microbiology , Gene Expression Regulation, Fungal , Mice , Hyphae/growth & development , Hyphae/genetics
14.
mBio ; 15(8): e0153524, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38980041

ABSTRACT

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.


Subject(s)
Candida albicans , Fungal Proteins , Gene Expression Regulation, Fungal , Spliceosomes , Candida albicans/genetics , Candida albicans/pathogenicity , Candida albicans/growth & development , Candida albicans/physiology , Candida albicans/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Humans , Fungal Proteins/genetics , Fungal Proteins/metabolism , Morphogenesis/genetics , RNA Splicing , Virulence , Hyphae/growth & development , Hyphae/genetics , Introns/genetics
15.
mSphere ; 9(7): e0037224, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38980069

ABSTRACT

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.


Subject(s)
Candida albicans , Fungal Proteins , Gene Expression Regulation, Fungal , Iron , Candida albicans/genetics , Candida albicans/pathogenicity , Candida albicans/metabolism , Iron/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mutation , Virulence , Gene Expression Profiling , Transcription Factors/genetics , Transcription Factors/metabolism , Phenotype , Cell Wall/metabolism , Cell Wall/genetics , Genetic Variation , Genotype
16.
Curr Opin Microbiol ; 80: 102507, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38955050

ABSTRACT

The human microbiota is a complex microbial ecosystem populated by bacteria, fungi, viruses, protists, and archaea. The coexistence of fungi alongside with many billions of bacteria, especially in the gut, involves complex interactions, ranging from antagonistic to beneficial, between the members of these two kingdoms. Bacteria can impact fungi through various means, such as physical interactions, secretion of metabolites, or alteration of the host immune response, thereby affecting fungal growth and virulence. This review summarizes recent progress in this field, delving into the latest understandings of bacterial-fungal-immune interactions and innovative therapeutic approaches addressing the challenges of treating fungal infections associated with microbiota imbalances.


Subject(s)
Candida albicans , Candidiasis , Gastrointestinal Microbiome , Humans , Candida albicans/pathogenicity , Candidiasis/microbiology , Candidiasis/immunology , Animals , Bacteria/pathogenicity , Bacteria/genetics , Host-Pathogen Interactions , Virulence , Microbiota , Microbial Interactions
17.
Free Radic Biol Med ; 222: 344-360, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38945457

ABSTRACT

Innovative solutions for rapid protection against broad-spectrum infections are very important in dealing with complex infection environments. We utilized a functionally inactive mutated endolysin protein of Streptococcus pneumoniae (ΔA146Ply) to immunize mice against pneumonic infections by multidrug-resistant bacteria, Candida albicans and influenza virus type A. ΔA146Ply protection relied on both immunized tissue-resident and monocyte-derived alveolar macrophages and inhibited infection induced ferroptosis that upregulated expression of GPX4 (glutathione peroxidase) in alveolar macrophages. Ferroptosis resistance endowed macrophages with enhanced phagocytosis by inhibiting lipid peroxidation during infection. Moreover, we demonstrated ΔA146Ply upregulated GPX4 through the TLR4/IRG1/NRF2 pathway. ΔA146Ply also induced ferroptosis inhibition and phagocytosis improvement in human monocytes. This mode of action is a novel and potentially prophylactic and rapid broad-spectrum anti-infection mechanism. Our study provides new insights into protective interventions that act by regulating ferroptosis to improve multiple pathogen resistance via GPX4 targeting.


Subject(s)
Ferroptosis , Macrophages, Alveolar , NF-E2-Related Factor 2 , Phospholipid Hydroperoxide Glutathione Peroxidase , Streptococcus pneumoniae , Toll-Like Receptor 4 , Animals , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mice , Streptococcus pneumoniae/pathogenicity , Ferroptosis/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Humans , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Phagocytosis , Candida albicans/pathogenicity , Candidiasis/immunology , Candidiasis/genetics , Candidiasis/microbiology , Candidiasis/metabolism , Up-Regulation , Female
18.
Curr Opin Microbiol ; 80: 102493, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833793

ABSTRACT

Candida albicans is a ubiquitous fungus of humans. It is not only a component of the oral and intestinal microbiota of most healthy adults but also a major cause of mucosal disorders and life-threatening disseminated infections. Until recently, research on the biology and pathogenesis of the fungus was largely based on a single clinical isolate. We review investigations that have started to dissect a diverse set of C. albicans strains. Using different approaches to leverage the species' phenotypic and/or genetic diversity, these studies illuminate the wide range of interactions between fungus and host. While connecting genetic variants to phenotypes of interest remains challenging, research on C. albicans' natural diversity is central to understand fungal commensalism and pathogenesis.


Subject(s)
Candida albicans , Genetic Variation , Symbiosis , Candida albicans/genetics , Candida albicans/pathogenicity , Humans , Candidiasis/microbiology , Animals , Host-Pathogen Interactions , Gastrointestinal Microbiome
19.
G3 (Bethesda) ; 14(8)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38874344

ABSTRACT

Candida albicans is a major fungal pathogen of humans that can cause serious systemic infections in vulnerable immunocompromised populations. One of its virulence attributes is its capacity to transition between yeast and filamentous morphologies, but our understanding of this process remains incomplete. Here, we analyzed data from a functional genomic screen performed with the C. albicans Gene Replacement And Conditional Expression collection to identify genes crucial for morphogenesis in host-relevant conditions. Through manual scoring of microscopy images coupled with analysis of each image using a deep learning-based method termed Candescence, we identified 307 genes important for filamentation in tissue culture medium at 37°C with 5% CO2. One such factor was orf19.5963, which is predicted to encode the prenyltransferase Nus1 based on sequence homology to Saccharomyces cerevisiae. We further showed that Nus1 and its predicted interacting partner Rer2 are important for filamentation in multiple liquid filament-inducing conditions as well as for wrinkly colony formation on solid agar. Finally, we highlight that Nus1 and Rer2 likely govern C. albicans morphogenesis due to their importance in intracellular trafficking, as well as maintaining lipid homeostasis. Overall, this work identifies Nus1 and Rer2 as important regulators of C. albicans filamentation and highlights the power of functional genomic screens in advancing our understanding of gene function in human fungal pathogens.


Subject(s)
Candida albicans , Fungal Proteins , Candida albicans/genetics , Candida albicans/pathogenicity , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Dimethylallyltranstransferase/genetics , Dimethylallyltranstransferase/metabolism , Hyphae/growth & development
20.
Microbiol Mol Biol Rev ; 88(2): e0002123, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38832801

ABSTRACT

SUMMARYA significant increase in the incidence of Candida-mediated infections has been observed in the last decade, mainly due to rising numbers of susceptible individuals. Recently, the World Health Organization published its first fungal pathogen priority list, with Candida species listed in medium, high, and critical priority categories. This review is a synthesis of information and recent advances in our understanding of two of these species-Candida albicans and Candida glabrata. Of these, C. albicans is the most common cause of candidemia around the world and is categorized as a critical priority pathogen. C. glabrata is considered a high-priority pathogen and has become an increasingly important cause of candidemia in recent years. It is now the second most common causative agent of candidemia in many geographical regions. Despite their differences and phylogenetic divergence, they are successful as pathogens and commensals of humans. Both species can cause a broad variety of infections, ranging from superficial to potentially lethal systemic infections. While they share similarities in certain infection strategies, including tissue adhesion and invasion, they differ significantly in key aspects of their biology, interaction with immune cells, host damage strategies, and metabolic adaptations. Here we provide insights on key aspects of their biology, epidemiology, commensal and pathogenic lifestyles, interactions with the immune system, and antifungal resistance.


Subject(s)
Antifungal Agents , Candida albicans , Candida glabrata , Candidiasis , Drug Resistance, Fungal , Candida glabrata/pathogenicity , Humans , Candida albicans/pathogenicity , Candidiasis/microbiology , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Animals , Candidemia/microbiology , Candidemia/epidemiology , Phylogeny , Host-Pathogen Interactions
SELECTION OF CITATIONS
SEARCH DETAIL