Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.719
Filter
1.
Proc Natl Acad Sci U S A ; 121(32): e2304382121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39088389

ABSTRACT

Microbes rarely exist in isolation and instead form complex polymicrobial communities. As a result, microbes have developed intricate offensive and defensive strategies that enhance their fitness in these complex communities. Thus, identifying and understanding the molecular mechanisms controlling polymicrobial interactions is critical for understanding the function of microbial communities. In this study, we show that the gram-negative opportunistic human pathogen Pseudomonas aeruginosa, which frequently causes infection alongside a plethora of other microbes including fungi, encodes a genetic network which can detect and defend against gliotoxin, a potent, disulfide-containing antimicrobial produced by the ubiquitous filamentous fungus Aspergillus fumigatus. We show that gliotoxin exposure disrupts P. aeruginosa zinc homeostasis, leading to transcriptional activation of a gene encoding a previously uncharacterized dithiol oxidase (herein named as DnoP), which detoxifies gliotoxin and structurally related toxins. Despite sharing little homology to the A. fumigatus gliotoxin resistance protein (GliT), the enzymatic mechanism of DnoP from P. aeruginosa appears to be identical that used by A. fumigatus. Thus, DnoP and its transcriptional induction by low zinc represent a rare example of both convergent evolution of toxin defense and environmental cue sensing across kingdoms. Collectively, these data provide compelling evidence that P. aeruginosa has evolved to survive exposure to an A. fumigatus disulfide-containing toxin in the natural environment.


Subject(s)
Aspergillus fumigatus , Gliotoxin , Pseudomonas aeruginosa , Gliotoxin/metabolism , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/genetics , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/genetics , Zinc/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Microbial Interactions , Humans , Fungal Proteins/metabolism , Fungal Proteins/genetics
2.
Nat Commun ; 15(1): 6312, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060235

ABSTRACT

Azole antifungals inhibit the sterol C14-demethylase (CYP51/Erg11) of the ergosterol biosynthesis pathway. Here we show that the azole-induced synthesis of fungicidal cell wall carbohydrate patches in the pathogenic mold Aspergillus fumigatus strictly correlates with the accumulation of the CYP51 substrate eburicol. A lack of other essential ergosterol biosynthesis enzymes, such as sterol C24-methyltransferase (Erg6A), squalene synthase (Erg9) or squalene epoxidase (Erg1) does not trigger comparable cell wall alterations. Partial repression of Erg6A, which converts lanosterol into eburicol, increases azole resistance. The sterol C5-desaturase (ERG3)-dependent conversion of eburicol into 14-methylergosta-8,24(28)-dien-3ß,6α-diol, the "toxic diol" responsible for the fungistatic activity against yeasts, is not required for the fungicidal effects in A. fumigatus. While ERG3-lacking yeasts are azole resistant, ERG3-lacking A. fumigatus becomes more susceptible. Mutants lacking mitochondrial complex III functionality, which are much less effectively killed, but strongly inhibited in growth by azoles, convert eburicol more efficiently into the supposedly "toxic diol". We propose that the mode of action of azoles against A. fumigatus relies on accumulation of eburicol which exerts fungicidal effects by triggering cell wall carbohydrate patch formation.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Azoles , Fungal Proteins , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/genetics , Antifungal Agents/pharmacology , Fungal Proteins/metabolism , Fungal Proteins/genetics , Azoles/pharmacology , Ergosterol/metabolism , Ergosterol/biosynthesis , Cell Wall/metabolism , Cell Wall/drug effects , Drug Resistance, Fungal/genetics , Bicyclic Monoterpenes/pharmacology , Bicyclic Monoterpenes/metabolism , Microbial Sensitivity Tests , Sterol 14-Demethylase/metabolism , Sterol 14-Demethylase/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Oxidoreductases/metabolism , Oxidoreductases/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Squalene Monooxygenase/metabolism , Squalene Monooxygenase/genetics , Lanosterol/analogs & derivatives
3.
mSphere ; 9(7): e0042524, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38975761

ABSTRACT

Treatment of fungal infections associated with the filamentous fungus Aspergillus fumigatus is becoming more problematic as this organism is developing resistance to the main chemotherapeutic drug at an increasing rate. Azole drugs represent the current standard-of-care in the treatment of aspergillosis with this drug class acting by inhibiting a key step in the biosynthesis of the fungal sterol ergosterol. Azole compounds block the activity of the lanosterol α-14 demethylase, encoded by the cyp51A gene. A common route of azole resistance involves an increase in transcription of cyp51A. This transcriptional increase requires the function of a Zn2Cys6 DNA-binding domain-containing transcription activator protein called AtrR. AtrR was identified through its action as a positive regulator of expression of an ATP-binding cassette transporter (abcC/cdr1B here called abcG1). Using both deletion and alanine scanning mutagenesis, we demonstrate that a conserved C-terminal domain in A. fumigatus is required for the expression of abcG1 but dispensable for cyp51A transcription. This domain is also found in several other fungal pathogen AtrR homologs consistent with a conserved gene-selective function of this protein segment being conserved. Using RNA sequencing (RNA-seq), we find that this gene-specific transcriptional defect extends to several other membrane transporter-encoding genes including a second ABC transporter locus. Our data reveal that AtrR uses at least two distinct mechanisms to induce gene expression and that normal susceptibility to azole drugs cannot be provided by maintenance of wild-type expression of the ergosterol biosynthetic pathway when ABC transporter expression is reduced. IMPORTANCE: Aspergillus fumigatus is the primary human filamentous fungal pathogen. The principal chemotherapeutic drug used to control infections associated with A. fumigatus is the azole compound. These drugs are well-tolerated and effective, but resistance is emerging at an alarming rate. Most resistance is associated with mutations that lead to overexpression of the azole target enzyme, lanosterol α-14 demethylase, encoded by the cyp51A gene. A key regulator of cyp51A gene expression is the transcription factor AtrR. Very little is known of the molecular mechanisms underlying the effect of AtrR on gene expression. Here, we use deletion and clustered amino acid substitution mutagenesis to map a region of AtrR that confers gene-specific activation on target genes of this transcription factor. This region is highly conserved across AtrR homologs from other pathogenic species arguing that its importance in transcriptional regulation is maintained across evolution.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Fungal Proteins , Gene Expression Regulation, Fungal , Transcriptional Activation , Aspergillus fumigatus/genetics , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Antifungal Agents/pharmacology , Azoles/pharmacology , Cytochrome P-450 Enzyme System/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Drug Resistance, Fungal/genetics , Protein Domains
4.
Mycopathologia ; 189(4): 49, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864956

ABSTRACT

Aspergillosis encompasses a wide range of clinical conditions based on the interaction between Aspergillus and the host. It ranges from colonization to invasive aspergillosis. The human lung provides an entry door for Aspergillus. Aspergillus has virulence characteristics such as conidia, rapid growth at body temperature, and the production of specific proteins, carbohydrates, and secondary metabolites that allow A. fumigatus to infiltrate the lung's alveoli and cause invasive aspergillosis. Alveolar epithelial cells play an important role in both fungus clearance and immune cell recruitment via cytokine release. Although the innate immune system quickly clears conidia in immunocompetent hosts, A. fumigatus has evolved multiple virulence factors in order to escape immune response such as ROS detoxifying enzymes, the rodlet layer, DHN-melanin and toxins. Bacterial co-infections or interactions can alter the immune response, impact Aspergillus growth and virulence, enhance biofilm formation, confound diagnosis, and reduce treatment efficacy. The gut microbiome's makeup influences pulmonary immune responses generated by A. fumigatus infection and vice versa. The real-time PCR for Aspergillus DNA detection might be a particularly useful tool to diagnose pulmonary aspergillosis. Metagenomics analyses allow quick and easy detection and identification of a great variety of fungi in different clinical samples, although optimization is still required particularly for the use of NGS techniques. This review will analyze the current state of aspergillosis in light of recent discoveries in the microbiota and mycobiota.


Subject(s)
Aspergillosis , Mycobiome , Humans , Aspergillosis/microbiology , Aspergillosis/diagnosis , Aspergillosis/immunology , Aspergillus fumigatus/pathogenicity , Aspergillus fumigatus/genetics , Aspergillus fumigatus/immunology , Aspergillus/genetics , Aspergillus/pathogenicity , Virulence Factors/genetics , Microbiota , Virulence , Metagenomics , Host-Pathogen Interactions/immunology
6.
Arch Microbiol ; 206(7): 305, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878211

ABSTRACT

Aspergillus fumigatus is a ubiquitous filamentous fungus commonly found in the environment. It is also an opportunistic human pathogen known to cause a range of respiratory infections, such as invasive aspergillosis, particularly in immunocompromised individuals. Azole antifungal agents are widely used for the treatment and prophylaxis of Aspergillus infections due to their efficacy and tolerability. However, the emergence of azole resistance in A. fumigatus has become a major concern in recent years due to their association with increased treatment failures and mortality rates. The development of azole resistance in A. fumigatus can occur through both acquired and intrinsic mechanisms. Acquired resistance typically arises from mutations in the target enzyme, lanosterol 14-α-demethylase (Cyp51A), reduces the affinity of azole antifungal agents for the enzyme, rendering them less effective, while intrinsic resistance refers to a natural resistance of certain A. fumigatus isolates to azole antifungals due to inherent genetic characteristics. The current review aims to provide a comprehensive overview of azole antifungal resistance in A. fumigatus, discusses underlying resistance mechanisms, including alterations in the target enzyme, Cyp51A, and the involvement of efflux pumps in drug efflux. Impact of azole fungicide uses in the environment and the spread of resistant strains is also explored.


Subject(s)
Antifungal Agents , Aspergillosis , Aspergillus fumigatus , Azoles , Drug Resistance, Fungal , Fungal Proteins , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Azoles/pharmacology , Drug Resistance, Fungal/genetics , Antifungal Agents/pharmacology , Humans , Fungal Proteins/genetics , Fungal Proteins/metabolism , Aspergillosis/microbiology , Aspergillosis/drug therapy , Microbial Sensitivity Tests , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Mutation
7.
Commun Biol ; 7(1): 704, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851817

ABSTRACT

Aspergillus fumigatus represents a public health problem due to the high mortality rate in immunosuppressed patients and the emergence of antifungal-resistant isolates. Protein acetylation is a crucial post-translational modification that controls gene expression and biological processes. The strategic manipulation of enzymes involved in protein acetylation has emerged as a promising therapeutic approach for addressing fungal infections. Sirtuins, NAD+-dependent lysine deacetylases, regulate protein acetylation and gene expression in eukaryotes. However, their role in the human pathogenic fungus A. fumigatus remains unclear. This study constructs six single knockout strains of A. fumigatus and a strain lacking all predicted sirtuins (SIRTKO). The mutant strains are viable under laboratory conditions, indicating that sirtuins are not essential genes. Phenotypic assays suggest sirtuins' involvement in cell wall integrity, secondary metabolite production, thermotolerance, and virulence. Deletion of sirE attenuates virulence in murine and Galleria mellonella infection models. The absence of SirE alters the acetylation status of proteins, including histones and non-histones, and triggers significant changes in the expression of genes associated with secondary metabolism, cell wall biosynthesis, and virulence factors. These findings encourage testing sirtuin inhibitors as potential therapeutic strategies to combat A. fumigatus infections or in combination therapy with available antifungals.


Subject(s)
Aspergillosis , Aspergillus fumigatus , Sirtuins , Aspergillus fumigatus/pathogenicity , Aspergillus fumigatus/genetics , Aspergillus fumigatus/enzymology , Sirtuins/genetics , Sirtuins/metabolism , Virulence , Animals , Mice , Aspergillosis/microbiology , Aspergillosis/drug therapy , Acetylation , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Virulence Factors/genetics , Virulence Factors/metabolism , Moths/microbiology
8.
Nat Commun ; 15(1): 4984, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862481

ABSTRACT

More than 10 million people suffer from lung diseases caused by the pathogenic fungus Aspergillus fumigatus. Azole antifungals represent first-line therapeutics for most of these infections but resistance is rising, therefore the identification of antifungal targets whose inhibition synergises with the azoles could improve therapeutic outcomes. Here, we generate a library of 111 genetically barcoded null mutants of Aspergillus fumigatus in genes encoding protein kinases, and show that loss of function of kinase YakA results in hypersensitivity to the azoles and reduced pathogenicity. YakA is an orthologue of Candida albicans Yak1, a TOR signalling pathway kinase involved in modulation of stress responsive transcriptional regulators. We show that YakA has been repurposed in A. fumigatus to regulate blocking of the septal pore upon exposure to stress. Loss of YakA function reduces the ability of A. fumigatus to penetrate solid media and to grow in mouse lung tissue. We also show that 1-ethoxycarbonyl-beta-carboline (1-ECBC), a compound previously shown to inhibit C. albicans Yak1, prevents stress-mediated septal spore blocking and synergises with the azoles to inhibit A. fumigatus growth.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Dyrk Kinases , Fungal Proteins , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Aspergillus fumigatus/genetics , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/enzymology , Animals , Antifungal Agents/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/antagonists & inhibitors , Mice , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Azoles/pharmacology , Aspergillosis/microbiology , Aspergillosis/drug therapy , Lung/microbiology , Spores, Fungal/drug effects , Spores, Fungal/genetics , Female
9.
Mycopathologia ; 189(4): 50, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864903

ABSTRACT

Aspergillus fumigatus is a saprophytic fungal pathogen that causes opportunistic infections in animals and humans. Azole resistance has been reported globally in human A. fumigatus isolates, but the prevalence of resistance in isolates from animals is largely unknown. A retrospective resistance surveillance study was performed using a collection of clinical A. fumigatus isolates from various animal species collected between 2015 and 2020. Agar-based azole resistance screening of all isolates was followed by in vitro antifungal susceptibility testing and cyp51A gene sequencing of the azole-resistant isolates. Over the 5 year period 16 (11.3%) of 142 A. fumigatus culture-positive animals harbored an azole-resistant isolate. Resistant isolates were found in birds (15%; 2/13), cats (21%; 6/28), dogs (8%; 6/75) and free-ranging harbor porpoise (33%; 2/6). Azole-resistance was cyp51A mediated in all isolates: 81.3% (T-67G/)TR34/L98H, 12.5% TR46/Y121F/T289A. In one azole-resistant A. fumigatus isolate a combination of C(-70)T/F46Y/C(intron7)T/C(intron66)T/M172V/E427K single-nucleotide polymorphisms in the cyp51A gene was found. Of the animals with an azole-resistant isolate and known azole exposure status 71.4% (10/14) were azole naive. Azole resistance in A. fumigatus isolates from animals in the Netherlands is present and predominantly cyp51A TR-mediated, supporting an environmental route of resistance selection. Our data supports the need to include veterinary isolates in resistance surveillance programs. Veterinarians should consider azole resistance as a reason for therapy failure when treating aspergillosis and consider resistance testing of relevant isolates.


Subject(s)
Antifungal Agents , Aspergillosis , Aspergillus fumigatus , Azoles , Drug Resistance, Fungal , Microbial Sensitivity Tests , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Aspergillus fumigatus/isolation & purification , Animals , Azoles/pharmacology , Drug Resistance, Fungal/genetics , Aspergillosis/microbiology , Aspergillosis/veterinary , Antifungal Agents/pharmacology , Netherlands/epidemiology , Retrospective Studies , Fungal Proteins/genetics , Birds/microbiology , Cats , Dogs , Cytochrome P-450 Enzyme System
10.
mBio ; 15(7): e0118424, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38832777

ABSTRACT

Group III hybrid histidine kinases are fungal-specific proteins and part of the multistep phosphorelay, representing the initial part of the high osmolarity glycerol (HOG) pathway. TcsC, the corresponding kinase in Aspergillus fumigatus, was expected to be a cytosolic protein but is targeted to the nucleus. Activation of TcsC by the antifungal fludioxonil has lethal consequences for the fungus. The agent triggers a fast and TcsC-dependent activation of SakA and later on a redistribution of TcsC to the cytoplasm. High osmolarity also activates TcsC, which then exits the nucleus or concentrates in spot-like, intra-nuclear structures. The sequence corresponding to the N-terminal 208 amino acids of TcsC lacks detectable domains. Its loss renders TcsC cytosolic and non-responsive to hyperosmotic stress, but it has no impact on the antifungal activity of fludioxonil. A point mutation in one of the three putative nuclear localization sequences, which are present in the N-terminus, prevents the nuclear localization of TcsC, but not its ability to respond to hyperosmotic stress. Hence, this striking intracellular localization is no prerequisite for the role of TcsC in the adaptive response to hyperosmotic stress, instead, TcsC proteins that are present in the nuclei seem to modulate the cell wall composition of hyphae, which takes place in the absence of stress. The results of the present study underline that the spatiotemporal dynamics of the individual components of the multistep phosphorelay is a crucial feature of this unique signaling hub. IMPORTANCE: Signaling pathways enable pathogens, such as Aspergillus fumigatus, to respond to a changing environment. The TcsC protein is the major sensor of the high osmolarity glycerol (HOG) pathway of A. fumigatus and it is also the target of certain antifungals. Insights in its function are therefore relevant for the pathogenicity and new therapeutic treatment options. TcsC was expected to be cytoplasmic, but we detected it in the nucleus and showed that it translocates to the cytoplasm upon activation. We have identified the motif that guides TcsC to the nucleus. An exchange of a single amino acid in this motif prevents a nuclear localization, but this nuclear targeting is no prerequisite for the TcsC-mediated stress response. Loss of the N-terminal 208 amino acids prevents the nuclear localization and renders TcsC unable to respond to hyperosmotic stress demonstrating that this part of the protein is of crucial importance.


Subject(s)
Aspergillus fumigatus , Cell Nucleus , Dioxoles , Fungal Proteins , Histidine Kinase , Pyrroles , Aspergillus fumigatus/genetics , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/drug effects , Histidine Kinase/metabolism , Histidine Kinase/genetics , Histidine Kinase/chemistry , Cell Nucleus/metabolism , Pyrroles/pharmacology , Pyrroles/metabolism , Dioxoles/pharmacology , Dioxoles/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/metabolism , Osmotic Pressure , Cytoplasm/metabolism , Protein Transport , Gene Expression Regulation, Fungal , Osmolar Concentration , Signal Transduction
11.
mBio ; 15(7): e0116624, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38934618

ABSTRACT

Contemporary antifungal therapies utilized to treat filamentous fungal infections are inhibited by intrinsic and emerging drug resistance. Consequently, there is an urgent need to develop novel antifungal compounds that are effective against drug-resistant filamentous fungi. Here, we utilized an Aspergillus fumigatus cell-based high-throughput screen to identify small molecules with antifungal activity that also potentiated triazole activity. The screen identified 16 hits with promising activity against A. fumigatus. A nonspirocyclic piperidine, herein named MBX-7591, exhibited synergy with triazole antifungal drugs and activity against pan-azole-resistant A. fumigatus isolates. MBX-7591 has additional potent activity against Rhizopus species and CO2-dependent activity against Cryptococcus neoformans. Chemical, genetic, and biochemical mode of action analyses revealed that MBX-7591 increases cell membrane saturation by decreasing oleic acid content. MBX-7591 has low toxicity in vivo and shows good efficacy in decreasing fungal burden in a murine model of invasive pulmonary aspergillosis. Taken together, our results suggest MBX-7591 is a promising hit with a novel mode of action for further antifungal drug development to combat the rising incidence of triazole-resistant filamentous fungal infections.IMPORTANCEThe incidence of infections caused by fungi continues to increase with advances in medical therapies. Unfortunately, antifungal drug development has not kept pace with the incidence and importance of fungal infections, with only three major classes of antifungal drugs currently available for use in the clinic. Filamentous fungi, also called molds, are particularly recalcitrant to contemporary antifungal therapies. Here, a recently developed Aspergillus fumigatus cell reporter strain was utilized to conduct a high-throughput screen to identify small molecules with antifungal activity. An emphasis was placed on small molecules that potentiated the activity of contemporary triazole antifungals and led to the discovery of MBX-7591. MBX-7591 potentiates triazole activity against drug-resistant molds such as A. fumigatus and has activity against Mucorales fungi. MBX-7591's mode of action involves inhibiting the conversion of saturated to unsaturated fatty acids, thereby impacting fungal membrane integrity. MBX-7591 is a novel small molecule with antifungal activity poised for lead development.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Drug Resistance, Fungal , Fatty Acids, Unsaturated , Microbial Sensitivity Tests , Triazoles , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Antifungal Agents/pharmacology , Triazoles/pharmacology , Mice , Animals , Fatty Acids, Unsaturated/pharmacology , Humans , High-Throughput Screening Assays , Drug Synergism , Rhizopus/drug effects , Rhizopus/genetics , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/genetics , Piperidines/pharmacology , Disease Models, Animal , Invasive Pulmonary Aspergillosis/drug therapy , Invasive Pulmonary Aspergillosis/microbiology
12.
Appl Environ Microbiol ; 90(7): e0027124, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38842339

ABSTRACT

Airborne triazole-resistant spores of the human fungal pathogen Aspergillus fumigatus are a significant human health problem as the agricultural use of triazoles has been selecting for cross-resistance to life-saving clinical triazoles. However, how to quantify exposure to airborne triazole-resistant spores remains unclear. Here, we describe a method for cost-effective wide-scale outdoor air sampling to measure both spore abundance as well as antifungal resistance fractions. We show that prolonged outdoor exposure of sticky seals placed in delta traps, when combined with a two-layered cultivation approach, can regionally yield sufficient colony-forming units (CFUs) for the quantitative assessment of aerial resistance levels at a spatial scale that was up to now unfeasible. When testing our method in a European pilot sampling 12 regions, we demonstrate that there are significant regional differences in airborne CFU numbers, and the triazole-resistant fraction of airborne spores is widespread and varies between 0 and 0.1 for itraconazole (∼4 mg/L) and voriconazole (∼2 mg/L). Our efficient and accessible air sampling protocol opens up extensive options for fine-scale spatial sampling and surveillance studies of airborne A. fumigatus.IMPORTANCEAspergillus fumigatus is an opportunistic fungal pathogen that humans and other animals are primarily exposed to through inhalation. Due to the limited availability of antifungals, resistance to the first choice class of antifungals, the triazoles, in A. fumigatus can make infections by this fungus untreatable and uncurable. Here, we describe and validate a method that allows for the quantification of airborne resistance fractions and quick genotyping of A. fumigatus TR-types. Our pilot study provides proof of concept of the suitability of the method for use by citizen-scientists for large-scale spatial air sampling. Spatial air sampling can open up extensive options for surveillance, health-risk assessment, and the study of landscape-level ecology of A. fumigatus, as well as investigating the environmental drivers of triazole resistance.


Subject(s)
Air Microbiology , Antifungal Agents , Aspergillus fumigatus , Drug Resistance, Fungal , Triazoles , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Triazoles/pharmacology , Antifungal Agents/pharmacology , Spores, Fungal/drug effects , Spores, Fungal/genetics , Environmental Monitoring/methods
13.
Protein Sci ; 33(7): e5071, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38895984

ABSTRACT

Tuberculosis necrotizing toxin (TNT) is a protein domain discovered on the outer membrane of Mycobacterium tuberculosis (Mtb), and the fungal pathogen Aspergillus fumigatus. TNT domains have pure NAD(P) hydrolytic activity, setting them apart from other NAD-cleaving domains such as ADP-ribosyl cyclase and Toll/interleukin-1 receptor homology (TIR) domains which form a wider set of products. Importantly, the Mtb TNT domain has been shown to be involved in immune evasion via depletion of the intracellular NAD pool of macrophages. Therefore, an intriguing hypothesis is that TNT domains act as "NAD killers" in host cells facilitating pathogenesis. Here, we explore the phylogenetic distribution of TNT domains and detect their presence solely in bacteria and fungi. Within fungi, we discerned six TNT clades. In addition, X-ray crystallography and AlphaFold2 modeling unveiled clade-specific strategies to promote homodimer stabilization of the fungal enzymes, namely, Ca2+ binding, disulfide bonds, or hydrogen bonds. We show that dimer stabilization is a requirement for NADase activity and that the group-specific strategies affect the active site conformation, thereby modulating enzyme activity. Together, these findings reveal the evolutionary lineage of fungal TNT enzymes, corroborating the hypothesis of them being pure extracellular NAD (eNAD) cleavers, with possible involvement in microbial warfare and host immune evasion.


Subject(s)
Mycobacterium tuberculosis , NAD , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/chemistry , NAD/metabolism , Protein Domains , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungal Proteins/genetics , Crystallography, X-Ray , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/genetics , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/chemistry , Evolution, Molecular , Models, Molecular , Phylogeny , NAD+ Nucleosidase/metabolism , NAD+ Nucleosidase/chemistry , NAD+ Nucleosidase/genetics
14.
Front Cell Infect Microbiol ; 14: 1393242, 2024.
Article in English | MEDLINE | ID: mdl-38912204

ABSTRACT

Background: Invasive mold diseases of the central nervous (CNS IMD) system are exceedingly rare disorders, characterized by nonspecific clinical symptoms. This results in significant diagnostic challenges, often leading to delayed diagnosis and the risk of misdiagnosis for patients. Metagenomic Next-Generation Sequencing (mNGS) holds significant importance for the diagnosis of infectious diseases, especially in the rapid and accurate identification of rare and difficult-to-culture pathogens. Therefore, this study aims to explore the clinical characteristics of invasive mold disease of CNS IMD in children and assess the effectiveness of mNGS technology in diagnosing CNS IMD. Methods: Three pediatric patients diagnosed with Invasive mold disease brain abscess and treated in the Pediatric Intensive Care Unit (PICU) of the First Affiliated Hospital of Zhengzhou University from January 2020 to December 2023 were selected for this study. Results: Case 1, a 6-year-old girl, was admitted to the hospital with "acute liver failure." During her hospital stay, she developed fever, irritability, and seizures. CSF mNGS testing resulted in a negative outcome. Multiple brain abscesses were drained, and Aspergillus fumigatus was detected in pus culture and mNGS. The condition gradually improved after treatment with voriconazole combined with caspofungin. Case 2, a 3-year-old girl, was admitted with "acute B-lymphoblastic leukemia." During induction chemotherapy, she developed fever and seizures. Aspergillus fumigatus was detected in the intracranial abscess fluid by mNGS, and the condition gradually improved after treatment with voriconazole combined with caspofungin, followed by "right-sided brain abscess drainage surgery." Case 3, a 7-year-old girl, showed lethargy, fever, and right-sided limb weakness during the pending chemotherapy period for acute B-lymphoblastic leukemia. Rhizomucor miehei and Rhizomucor pusillus was detected in the cerebrospinal fluid by mNGS. The condition gradually improved after treatment with amphotericin B combined with posaconazole. After a six-month follow-up post-discharge, the three patients improved without residual neurological sequelae, and the primary diseases were in complete remission. Conclusion: The clinical manifestations of CNS IMD lack specificity. Early mNGS can assist in identifying the pathogen, providing a basis for definitive diagnosis. Combined surgical treatment when necessary can help improve prognosis.


Subject(s)
Antifungal Agents , Brain Abscess , High-Throughput Nucleotide Sequencing , Metagenomics , Humans , Female , Child , Metagenomics/methods , Brain Abscess/microbiology , Brain Abscess/diagnosis , Brain Abscess/drug therapy , Antifungal Agents/therapeutic use , Antifungal Agents/pharmacology , Invasive Fungal Infections/diagnosis , Invasive Fungal Infections/microbiology , Invasive Fungal Infections/drug therapy , Male , Central Nervous System Fungal Infections/diagnosis , Central Nervous System Fungal Infections/microbiology , Central Nervous System Fungal Infections/drug therapy , Child, Preschool , Aspergillus fumigatus/genetics , Aspergillus fumigatus/isolation & purification , Caspofungin/therapeutic use
15.
Emerg Infect Dis ; 30(8): 1531-1541, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38935978

ABSTRACT

Azole-resistant Aspergillus fumigatus (ARAf) fungi have been found inconsistently in the environment in Denmark since 2010. During 2018-2020, nationwide surveillance of clinical A. fumigatus fungi reported environmental TR34/L98H or TR46/Y121F/T289A resistance mutations in 3.6% of isolates, prompting environmental sampling for ARAf and azole fungicides and investigation for selection of ARAf in field and microcosmos experiments. ARAf was ubiquitous (20% of 366 samples; 16% TR34/L98H- and 4% TR46/Y121F/T289A-related mechanisms), constituting 4.2% of 4,538 A. fumigatus isolates. The highest proportions were in flower- and compost-related samples but were not correlated with azole-fungicide application concentrations. Genotyping showed clustering of tandem repeat-related ARAf and overlaps with clinical isolates in Denmark. A. fumigatus fungi grew poorly in the field experiment with no postapplication change in ARAf proportions. However, in microcosmos experiments, a sustained complete (tebuconazole) or partial (prothioconazole) inhibition against wild-type A. fumigatus but not ARAf indicated that, under some conditions, azole fungicides may favor growth of ARAf in soil.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Azoles , Drug Resistance, Fungal , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Aspergillus fumigatus/isolation & purification , Azoles/pharmacology , Denmark/epidemiology , Antifungal Agents/pharmacology , Humans , Aspergillosis/epidemiology , Aspergillosis/microbiology , Aspergillosis/drug therapy , Microbial Sensitivity Tests , Mutation , Fungicides, Industrial/pharmacology , Genotype
16.
Open Biol ; 14(6): 240033, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38919062

ABSTRACT

Aspergillus fumigatus is the predominant mould pathogen for humans. Adaption to host-imposed iron limitation has previously been demonstrated to be essential for its virulence. [2Fe-2S] clusters are crucial as cofactors of several metabolic pathways and mediate cytosolic/nuclear iron sensing in fungi including A. fumigatus. [2Fe-2S] cluster trafficking has been shown to involve BolA family proteins in both mitochondria and the cytosol/nucleus. Interestingly, both A. fumigatus homologues, termed Bol1 and Bol3, possess mitochondrial targeting sequences, suggesting the lack of cytosolic/nuclear versions. Here, we show by the combination of mutational, proteomic and fluorescence microscopic analyses that expression of the Bol3 encoding gene leads to dual localization of gene products to mitochondria and the cytosol/nucleus via alternative translation initiation downstream of the mitochondrial targeting sequence, which appears to be highly conserved in various Aspergillus species. Lack of either mitochondrial Bol1 or Bol3 was phenotypically inconspicuous while lack of cytosolic/nuclear Bol3 impaired growth during iron limitation but not iron sensing which indicates a particular importance of [2Fe-2S] cluster trafficking during iron limitation. Remarkably, cytosolic/nuclear Bol3 differs from the mitochondrial version only by N-terminal acetylation, a finding that was only possible by mutational hypothesis testing.


Subject(s)
Aspergillus fumigatus , Cytosol , Fungal Proteins , Iron , Mitochondria , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , Cytosol/metabolism , Mitochondria/metabolism , Iron/metabolism , Adaptation, Physiological , Cell Nucleus/metabolism , Protein Transport , Proteomics/methods , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Gene Expression Regulation, Fungal , Acetylation
17.
mSphere ; 9(6): e0025324, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38814077

ABSTRACT

Aspergillus fumigatus is the leading cause of severe mold infections in immunocompromised patients. This common fungus possesses innate attributes that allow it to evade the immune system, including its ability to survive the high copper (Cu) levels in phagosomes. Our previous work has revealed that under high Cu levels, the A. fumigatus transcription factor AceA is activated, inducing the expression of the copper exporter CrpA to expel excess Cu. To identify additional elements in Cu resistance, we evolved A. fumigatus wild-type and mutant ΔaceA or ΔcrpA strains under increasing Cu concentrations. Sequencing of the resultant resistant strains identified both shared and unique evolutionary pathways to resistance. Reintroduction of three of the most common mutations in genes encoding Pma1 (plasma membrane H+-ATPase), Gcs1 (glutamate cysteine-ligase), and Cpa1 (carbamoyl-phosphate synthetase), alone and in combination, into wild-type A. fumigatus confirmed their additive role in conferring Cu resistance. Detailed analysis indicated that the pma1 mutation L424I preserves Pma1 H+-ATPase activity under high Cu concentrations and that the cpa1 mutation A37V confers a survival advantage to conidia in the presence of Cu. Interestingly, simultaneous mutations of all three genes did not alter virulence in infected mice. Our work has identified novel Cu-resistance pathways and provides an evolutionary approach for dissecting the molecular basis of A. fumigatus adaptation to diverse environmental challenges.IMPORTANCEAspergillus fumigatus is the most common mold infecting patients with weakened immunity. Infection is caused by the inhalation of mold spores into the lungs and is often fatal. In healthy individuals, spores are engulfed by lung immune cells and destroyed by a combination of enzymes, oxidants, and high levels of copper. However, the mold can protect itself by pumping out excess copper with specific transporters. Here, we evolved A. fumigatus under high copper levels and identified new genetic mutations that help it resist the toxic effects of copper. We studied how these mutations affect the mold's ability to resist copper and how they impact its ability to cause disease. This is the first such study in a pathogenic mold, and it gives us a better understanding of how it manages to bypass our body's defenses during an infection.


Subject(s)
Aspergillus fumigatus , Copper , Fungal Proteins , Aspergillus fumigatus/genetics , Aspergillus fumigatus/pathogenicity , Copper/metabolism , Animals , Mice , Fungal Proteins/genetics , Fungal Proteins/metabolism , Aspergillosis/microbiology , Aspergillosis/immunology , Mutation , Drug Resistance, Fungal/genetics , Virulence , Evolution, Molecular , Glutamate-Cysteine Ligase/genetics , Female , Proton-Translocating ATPases/genetics
18.
Mycoses ; 67(5): e13732, 2024 May.
Article in English | MEDLINE | ID: mdl-38712846

ABSTRACT

BACKGROUND: Triazole-resistant Aspergillus fumigatus (TRAF) isolates are a growing public health problem with worldwide distribution. Epidemiological data on TRAF is limited in Africa, particularly in West Africa. OBJECTIVES: This study aimed to screen for the environmental presence of TRAF isolates in the indoor air of two hospitals in Burkina Faso. MATERIALS AND METHODS: Air samples were collected in wards housing patients at risk for invasive aspergillosis, namely infectious diseases ward, internal medicine ward, nephrology ward, pulmonology ward, medical emergency ward and paediatric ward. Sabouraud Dextrose Agar supplemented with triazoles was used to screen the suspected TRAF isolates and EUCAST method to confirm the resistance of suspected isolates. Sequencing of cyp51A gene was used to identify the resistance mechanism of confirmed TRAF isolates. RESULTS: Of the 198 samples collected and analysed, 67 showed growth of A. fumigatus isolates. The prevalence of TRAF isolates was 3.23% (4/124). One TRAF isolate exhibited a pan-triazole resistance. Sequencing of cyp51A gene identified the TR34/L98H mutation for this pan-triazole resistant isolate. This study showed for the first time the circulation of the pan-azole resistant isolate harbouring the TR34/L98H mutation in Burkina Faso. CONCLUSIONS: These findings emphasise the need to map these TRAF isolates in all parts of Burkina Faso and to establish local and national continuous surveillance of environmental and clinical TRAF isolates in this country.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Cytochrome P-450 Enzyme System , Drug Resistance, Fungal , Fungal Proteins , Mutation , Triazoles , Aspergillus fumigatus/genetics , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/isolation & purification , Drug Resistance, Fungal/genetics , Triazoles/pharmacology , Humans , Burkina Faso/epidemiology , Fungal Proteins/genetics , Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme System/genetics , Microbial Sensitivity Tests , Aspergillosis/microbiology , Aspergillosis/epidemiology , Air Microbiology
19.
J Clin Microbiol ; 62(7): e0036924, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38819167

ABSTRACT

Azole resistance screening in Aspergillus fumigatus sensu stricto can be routinely carried out by using azole-containing agar plates (E.Def 10.2 procedure); however, conidial suspension filtering and inoculum adjustment before inoculum preparation are time-consuming. We evaluated whether skipping the filtration and inoculum adjustment steps negatively influenced the performance of the E.Def 10.2 procedure. A. fumigatus sensu stricto isolates (n = 98), previously classified as azole susceptible or azole resistant (E.Def 9.4 method), were studied. Azole-resistant isolates had either the wild-type cyp51A gene sequence (n = 1) or the following cyp51A gene substitutions: TR34-L98H (n = 41), G54R (n = 5), TR46-Y121F-T289A (n = 1), or G448S (n = 1). In-house azole-containing agar plates were prepared according to the EUCAST E.Def 10.2 procedure. Conidial suspensions obtained by adding distilled water (Tween 20 0.1%) were either filtered and the inocula adjusted to 0.5 McFarland or left unfiltered and unadjusted. Agreements between the agar screening methods using inocula prepared by each procedure were high for itraconazole (99%), voriconazole (100%), and posaconazole (94.9%). Sensitivity and specificity (considering the susceptibility category as per the microdilution E.Def 9.4 method as the gold standard) of E.Def 10.2 were 100% to rule in or rule out resistance when unfiltered and unadjusted suspensions were used; the resistance phenotype of isolates harboring the TR34-L98H, G54R, or TR46-Y121F-T289A substitutions was correctly detected. Unfiltered and unadjusted conidial suspensions do not negatively influence the performance of the E.Def 10.2 method when screening for azole resistance in A. fumigatus sensu stricto. IMPORTANCE: Azole resistance screening in Aspergillus fumigatus sensu stricto can be routinely carried out by using azole-containing plates (E.Def 10.2 procedure); however, conidial suspension filtering and inoculum adjustment before inoculation of plates are time-consuming. We, here, showed that unfiltered and unadjusted conidial suspensions do not negatively influence the performance of the E.Def 10.2 method when screening for azole resistance in A. fumigatus sensu stricto.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Azoles , Drug Resistance, Fungal , Microbial Sensitivity Tests , Spores, Fungal , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Aspergillus fumigatus/isolation & purification , Azoles/pharmacology , Antifungal Agents/pharmacology , Microbial Sensitivity Tests/methods , Humans , Spores, Fungal/drug effects , Spores, Fungal/genetics , Culture Media/chemistry , Fungal Proteins/genetics , Agar , Cytochrome P-450 Enzyme System/genetics
20.
G3 (Bethesda) ; 14(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38696662

ABSTRACT

Aspergillus fumigatus is a deadly fungal pathogen, responsible for >400,000 infections/year and high mortality rates. A. fumigatus strains exhibit variation in infection-relevant traits, including in their virulence. However, most A. fumigatus protein-coding genes, including those that modulate its virulence, are shared between A. fumigatus strains and closely related nonpathogenic relatives. We hypothesized that A. fumigatus genes exhibit substantial genetic variation in the noncoding regions immediately upstream to the start codons of genes, which could reflect differences in gene regulation between strains. To begin testing this hypothesis, we identified 5,812 single-copy orthologs across the genomes of 263 A. fumigatus strains. In general, A. fumigatus noncoding regions showed higher levels of sequence variation compared with their corresponding protein-coding regions. Focusing on 2,482 genes whose protein-coding sequence identity scores ranged between 75 and 99%, we identified 478 total genes with signatures of positive selection only in their noncoding regions and 65 total genes with signatures only in their protein-coding regions. Twenty-eight of the 478 noncoding regions and 5 of the 65 protein-coding regions under selection are associated with genes known to modulate A. fumigatus virulence. Noncoding region variation between A. fumigatus strains included single-nucleotide polymorphisms and insertions or deletions of at least a few nucleotides. These results show that noncoding regions of A. fumigatus genes harbor greater sequence variation than protein-coding regions, raising the hypothesis that this variation may contribute to A. fumigatus phenotypic heterogeneity.


Subject(s)
Aspergillus fumigatus , Fungal Proteins , Genetic Variation , Genome, Fungal , Open Reading Frames , Aspergillus fumigatus/genetics , Aspergillus fumigatus/pathogenicity , Fungal Proteins/genetics , Polymorphism, Single Nucleotide , Untranslated Regions , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL