Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Microorganisms ; 11(11)2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38004768

ABSTRACT

The mortality rates of invasive fungal infections remain high because of the limited number of antifungal drugs available and antifungal drug resistance, which can rapidly evolve during treatment. Mutations in key resistance genes such as ERG11 were postulated to be the predominant cause of antifungal drug resistance in the clinic. However, recent advances in whole genome sequencing have revealed that there are multiple mechanisms leading to the microevolution of resistance. In many fungal species, resistance can emerge through ERG11-independent mechanisms and through the accumulation of mutations in many genes to generate a polygenic resistance phenotype. In addition, genome sequencing has revealed that full or partial aneuploidy commonly occurs in clinical or microevolved in vitro isolates to confer antifungal resistance. This review will provide an overview of the mutations known to be selected during the adaptive microevolution of antifungal drug resistance and focus on how recent advances in genome sequencing technology have enhanced our understanding of this process.

2.
JAC Antimicrob Resist ; 4(2): dlac033, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35402912

ABSTRACT

Background: Fungal infections are common life-threatening diseases amongst immunodeficient individuals. Invasive fungal disease is commonly treated with an azole antifungal agent, resulting in selection pressure and the emergence of drug resistance. Antifungal resistance is associated with higher mortality rates and treatment failure, making the current clinical management of fungal disease very challenging. Clinical isolates from a variety of fungi have been shown to contain mutations in the MSH2 gene, encoding a component of the DNA mismatch repair pathway. Mutation of MSH2 results in an elevated mutation rate that can increase the opportunity for selectively advantageous mutations to occur, accelerating the development of antifungal resistance. Objectives: To characterize the molecular mechanisms causing the microevolutionary emergence of antifungal resistance in msh2 mismatch repair mutants of Cryptococcus neoformans. Methods: The mechanisms resulting in the emergence of antifungal resistance were investigated using WGS, characterization of deletion mutants and measuring ploidy changes. Results: The genomes of resistant strains did not possess mutations in ERG11 or other genes of the ergosterol biosynthesis pathway. Antifungal resistance was due to small contributions from mutations in many genes. MSH2 does not directly affect ploidy changes. Conclusions: This study provides evidence that resistance to fluconazole can evolve independently of ERG11 mutations. A common microevolutionary route to the emergence of antifungal resistance involves the accumulation of mutations that alter stress signalling, cellular efflux, membrane trafficking, epigenetic modification and aneuploidy. This complex pattern of microevolution highlights the significant challenges posed both to diagnosis and treatment of drug-resistant fungal pathogens.

3.
Microorganisms ; 10(2)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35208897

ABSTRACT

Adaptation to the changing environmental conditions experienced within a host requires genetic diversity within a microbial population. Genetic diversity arises from mutations which occur due to DNA damage from exposure to exogenous environmental stresses or generated endogenously through respiration or DNA replication errors. As mutations can be deleterious, a delicate balance must be obtained between generating enough mutations for micro-evolution to occur while maintaining fitness and genomic integrity. Pathogenic microorganisms can actively modify their mutation rate to enhance adaptive micro-evolution by increasing expression of error-prone DNA polymerases or by mutating or decreasing expression of genes required for DNA repair. Strains which exhibit an elevated mutation rate are termed mutators. Mutators are found in varying prevalence in clinical populations where large-effect beneficial mutations enhance survival and are predominately caused by defects in the DNA mismatch repair (MMR) pathway. Mutators can facilitate the emergence of antibiotic resistance, allow phenotypic modifications to prevent recognition and destruction by the host immune system and enable switching to metabolic and cellular morphologies better able to survive in the given environment. This review will focus on recent advances in understanding the phenotypic and genotypic changes occurring in MMR mutators in both prokaryotic and eukaryotic pathogens.

4.
Molecules ; 26(13)2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34202224

ABSTRACT

Biofilms are assemblages of microbial cells, extracellular polymeric substances (EPS), and other components extracted from the environment in which they develop. Within biofilms, the spatial distribution of these components can vary. Here we present a fundamental characterization study to show differences between biofilms formed by Gram-positive methicillin-resistant Staphylococcus aureus (MRSA), Gram-negative Pseudomonas aeruginosa, and the yeast-type Candida albicans using synchrotron macro attenuated total reflectance-Fourier transform infrared (ATR-FTIR) microspectroscopy. We were able to characterise the pathogenic biofilms' heterogeneous distribution, which is challenging to do using traditional techniques. Multivariate analyses revealed that the polysaccharides area (1200-950 cm-1) accounted for the most significant variance between biofilm samples, and other spectral regions corresponding to amides, lipids, and polysaccharides all contributed to sample variation. In general, this study will advance our understanding of microbial biofilms and serve as a model for future research on how to use synchrotron source ATR-FTIR microspectroscopy to analyse their variations and spatial arrangements.


Subject(s)
Biofilms/growth & development , Candida albicans/physiology , Methicillin-Resistant Staphylococcus aureus/physiology , Pseudomonas aeruginosa/physiology , Synchrotrons , Fourier Analysis , Spectroscopy, Fourier Transform Infrared
5.
ACS Appl Mater Interfaces ; 13(15): 17340-17352, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33844492

ABSTRACT

Antimicrobial resistance has rendered many conventional therapeutic measures, such as antibiotics, ineffective. This makes the treatment of infections from pathogenic micro-organisms a major growing health, social, and economic challenge. Recently, nanomaterials, including two-dimensional (2D) materials, have attracted scientific interest as potential antimicrobial agents. Many of these studies, however, rely on the input of activation energy and lack real-world utility. In this work, we present the broad-spectrum antimicrobial activity of few-layered black phosphorus (BP) at nanogram concentrations. This property arises from the unique ability of layered BP to produce reactive oxygen species, which we harness to create this unique functionality. BP is shown to be highly antimicrobial toward susceptible and resistant bacteria and fungal species. To establish cytotoxicity with mammalian cells, we showed that both L929 mouse and BJ-5TA human fibroblasts were metabolically unaffected by the presence of BP. Finally, we demonstrate the practical utility of this approach, whereby medically relevant surfaces are imparted with antimicrobial properties via functionalization with few-layer BP. Given the self-degrading properties of BP, this study demonstrates a viable and practical pathway for the deployment of novel low-dimensional materials as antimicrobial agents without compromising the composition or nature of the coated substrate.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Phosphorus/chemistry , Animals , Dose-Response Relationship, Drug , Drug Resistance, Bacterial/drug effects , Drug Resistance, Fungal/drug effects , Humans , Mice
6.
ACS Appl Bio Mater ; 3(5): 2997-3004, 2020 May 18.
Article in English | MEDLINE | ID: mdl-35025346

ABSTRACT

The fabrication of antimicrobial surfaces that exhibit enhanced activity toward a large variety of microbial species is one of the major challenges of our time. In fact, the negative effects associated with both bacterial and fungal infections are enormous, especially considering that many microbial species are developing resistance to known antibiotics. In this work, we show how a combination of a specific surface morphology and surface chemistry can create a surface that exhibits nearly 100% antimicrobial activity toward both Gram-negative and Gram-positive bacteria and fungal cells. Arrays of vertically aligned, oxygen-deficient zinc oxide (ZnO) nanowires grown on a substrate exhibit enhanced antimicrobial activity compared with surfaces containing either less defective nanowires or highly oxygen-deficient flat films. This synergistic effect between physical activity (morphology) and chemical activity (surface composition) has been shown to be responsible for the outstanding antimicrobial activity of our surfaces, especially toward notoriously resilient bacterial or fungal species. These findings provide a series of design rules for tuning the activities of antibacterial and antifungal nanomaterials. These rules constitute an excellent platform for the development of next-generation antimicrobial surfaces.

7.
DNA Repair (Amst) ; 86: 102751, 2020 02.
Article in English | MEDLINE | ID: mdl-31838381

ABSTRACT

Passaging of microbes in vitro can lead to the selection of microevolved derivatives with differing properties to their original parent strains. One well characterised instance is the phenotypic differences observed between the series of strains derived from the type strain of the human pathogenic fungus Cryptococcus neoformans. A second case was reported in the close relative Cryptococcus deneoformans, in which a well-studied isolate ATCC 24067 (52D) altered its phenotypic characteristics after in vitro passaging in different laboratories. One of these derivatives, ATCC 24067A, has decreased virulence and also exhibits a hypermutator phenotype, in which the mutation rate is increased compared to wild type. In this study, the molecular basis behind the changes in the lineage of ATCC 24067 was determined by next-generation sequencing of the parent and passaged strain genomes. This analysis resulted in the identification of a point mutation that causes a D270G amino acid substitution within the exonuclease proofreading domain of the DNA polymerase delta subunit encoded by POL3. Complementation with POL3 confirmed that this mutation is responsible for the hypermutator phenotype of this strain. Regeneration of the mutation in C. neoformans, to eliminate the additional mutations present in the ATCC 24067A genetic background, demonstrated that the hypermutator phenotype of the pol3D270G mutant causes rapid microevolution in vitro but does not result in decreased virulence. These findings indicate that mutator strains can emerge in these pathogenic fungi without conferring a fitness cost, but the subsequent rapid accumulation of mutations can be deleterious.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans/growth & development , DNA Polymerase III/genetics , High-Throughput Nucleotide Sequencing/methods , Point Mutation , Amino Acid Substitution , Animals , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Fungal Proteins/genetics , Mice , Phenotype , Sequence Analysis, DNA , Serial Passage
8.
mBio ; 10(2)2019 04 23.
Article in English | MEDLINE | ID: mdl-31015333

ABSTRACT

Although mutation drives evolution over long and short terms, measuring and comparing mutation rates accurately have been particularly difficult. This is especially true when mutations lead to an alteration in fitness. E. Shor, J. Schuyler, and D. S. Perlin (https://doi.org/10.1128/mBio.00120-19) present a new method to compare mutation rates across fungal strains and under different growth conditions: they employ the green fluorescent protein (GFP) as the reporter and count mutations using fluorescence-activated cell sorting (FACS). The estimates of mutation rates using the GFP-FACS approach are similar to those calculated with other reporters, and the method was used to assess if different alleles of the mismatch repair pathway gene MSH2 impact the mutation rates in the human pathogen Candida glabrata The approach could be extended to other microbes and applications, opening the way for a better understanding of how mutation rates have impacted speciation and the emergence of antimicrobial resistance.


Subject(s)
Candida glabrata , Mutation Rate , Drug Resistance, Fungal , Fluorescence , Humans , Mutation
9.
mSphere ; 3(2)2018.
Article in English | MEDLINE | ID: mdl-29577082

ABSTRACT

Fungi are adept at occupying specific environmental niches and often exploit numerous secondary metabolites generated by the cytochrome P450 (CYP) monoxygenases. This report describes the characterization of a yeast-specific CYP encoded by simA ("survival in macrophages"). Deletion of simA does not affect yeast growth at 37°C in vitro but is essential for yeast cell production during macrophage infection. The ΔsimA strain exhibits reduced conidial germination and intracellular growth of yeast in macrophages, suggesting that the enzymatic product of SimA is required for normal fungal growth in vivo. Intracellular ΔsimA yeast cells exhibit cell wall defects, and metabolomic and chemical sensitivity data suggest that SimA may promote chitin synthesis or deposition in vitro. In vivo, ΔsimA yeast cells subsequently lyse and are degraded, suggesting that SimA may increase resistance to and/or suppress host cell biocidal effectors. The results suggest that simA synthesizes a secondary metabolite that allows T. marneffei to occupy the specific intracellular environmental niche within the macrophage. IMPORTANCE This study in a dimorphic fungal pathogen uncovered a role for a yeast-specific cytochrome P450 (CYP)-encoding gene in the ability of T. marneffei to grow as yeast cells within the host macrophages. This report will inspire further research into the role of CYPs and secondary metabolite synthesis during fungal pathogenic growth.

10.
Article in English | MEDLINE | ID: mdl-28861398

ABSTRACT

Fungal infections are an increasing public health problem, particularly in immunocompromised individuals. While these pathogenic fungi show polyphyletic origins with closely related non-pathogenic species, many undergo morphological transitions to produce pathogenic cell types that are associated with increased virulence. However, the characteristics of these pathogenic cells that contribute to virulence are poorly defined. Talaromyces marneffei grows as a non-pathogenic hyphal form at 25°C but undergoes a dimorphic transition to a pathogenic yeast form at 37°C in vitro and following inhalation of asexual conidia by a host. Here we show that this transition is associated with major changes in central carbon metabolism, and that these changes are correlated with increased virulence of the yeast form. Comprehensive metabolite profiling and 13C-labeling studies showed that hyphal cells exhibited very active glycolytic metabolism and contain low levels of internal carbohydrate reserves. In contrast, yeast cells fully catabolized glucose in the mitochondrial TCA cycle, and store excess glucose in large intracellular pools of trehalose and mannitol. Inhibition of the yeast TCA cycle inhibited replication in culture and in host cells. Yeast, but not hyphae, were also able to use myo-inositol and amino acids as secondary carbon sources, which may support their survival in host macrophages. These analyses suggest that T. marneffei yeast cells exhibit a more efficient oxidative metabolism and are capable of utilizing a diverse range of carbon sources, which contributes to their virulence in animal tissues, highlighting the importance of dimorphic switching in pathogenic yeast.


Subject(s)
Metabolomics , Talaromyces/growth & development , Talaromyces/metabolism , Talaromyces/pathogenicity , Amino Acids/metabolism , Animals , Carbohydrate Metabolism , Carbon/metabolism , Citric Acid Cycle , Gene Expression Regulation, Fungal , Host-Pathogen Interactions/physiology , Humans , Hyphae/growth & development , Hyphae/metabolism , Inositol/metabolism , Macrophages/microbiology , Mitochondria/metabolism , Mycoses , Spores, Fungal/growth & development , Spores, Fungal/metabolism , Staining and Labeling , THP-1 Cells , Talaromyces/cytology , Temperature , Virulence , Yeasts/cytology , Yeasts/growth & development , Yeasts/metabolism
11.
mBio ; 8(3)2017 05 30.
Article in English | MEDLINE | ID: mdl-28559486

ABSTRACT

The ability to adapt to a changing environment provides a selective advantage to microorganisms. In the case of many pathogens, a large change in their environment occurs when they move from a natural setting to a setting within a human host and then during the course of disease development to various locations within that host. Two clinical isolates of the human fungal pathogen Cryptococcus neoformans were identified from a collection of environmental and clinical strains that exhibited a mutator phenotype, which is a phenotype which provides the ability to change rapidly due to the accumulation of DNA mutations at high frequency. Whole-genome analysis of these strains revealed mutations in MSH2 of the mismatch repair pathway, and complementation confirmed that these mutations are responsible for the mutator phenotype. Comparison of mutation frequencies in deletion strains of eight mismatch repair pathway genes in C. neoformans showed that the loss of three of them, MSH2, MLH1, and PMS1, results in an increase in mutation rates. Increased mutation rates enable rapid microevolution to occur in these strains, generating phenotypic variations in traits associated with the ability to grow in vivo, in addition to allowing rapid generation of resistance to antifungal agents. Mutation of PMS1 reduced virulence, whereas mutation of MSH2 or MLH1 had no effect on the level of virulence. These findings thus support the hypothesis that this pathogenic fungus can take advantage of a mutator phenotype in order to cause disease but that it can do so only in specific pathways that lead to a mutator trait without a significant tradeoff in fitness.IMPORTANCE Fungi account for a large number of infections that are extremely difficult to treat; superficial fungal infections affect approximately 1.7 billion (25%) of the general population worldwide, and systemic fungal diseases result in an unacceptably high mortality rate. How fungi adapt to their hosts is not fully understood. This research investigated the role of changes to DNA sequences in adaption to the host environment and the ability to cause disease in Cryptococcus neoformans, one of the world's most common and most deadly fungal pathogens. The study results showed that microevolutionary rates are enhanced in either clinical isolates or in gene deletion strains with msh2 mutations. This gene has similar functions in regulating the rapid emergence of antifungal drug resistance in a distant fungal relative of C. neoformans, the pathogen Candida glabrata Thus, microevolution resulting from enhanced mutation rates may be a common contributor to fungal pathogenesis.


Subject(s)
Cryptococcus neoformans/genetics , DNA Mismatch Repair , DNA Replication , Evolution, Molecular , Mutation , Antifungal Agents/pharmacology , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/pathogenicity , DNA Damage/drug effects , DNA Repair , Drug Resistance, Fungal , Genes, Fungal , Genome, Fungal , Humans , Phenotype , Virulence/drug effects
12.
Curr Genet ; 63(3): 519-530, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27783209

ABSTRACT

Discovering the genes underlying fundamental processes that enable cells to live and reproduce is a technical challenge, because loss of gene function in mutants results in organisms that cannot survive. This study describes a forward genetics method to identify essential genes in fungi, based on the propensity for Agrobacterium tumefaciens to insert T-DNA molecules into the promoters or 5' untranslated regions of genes and by placing a conditional promoter within the T-DNA. Insertions of the promoter of the GAL7 gene were made in the human pathogen Cryptococcus neoformans. Nine strains of 960 T-DNA insertional mutants screened grew on media containing galactose, but had impaired growth on media containing glucose, which suppresses expression from GAL7. T-DNA insertions were found in the homologs of IDI1, MRPL37, NOC3, NOP56, PRE3 and RPL17, all of which are essential in ascomycete yeasts Saccharomyces cerevisiae or Schizosaccharomyces pombe. Altering the carbon source in the medium provided a system to identify phenotypes in response to stress agents. The pre3 proteasome subunit mutant was further characterized. The T-DNA insertion and phenotype co-segregate in progeny from a cross, and the growth defect is complemented by the reintroduction of the wild type gene into the insertional mutant. A deletion allele was generated in a diploid strain, this heterozygous strain was sporulated, and analysis of the progeny provided additional genetic evidence that PRE3 is essential. The experimental design is applicable to other fungi and has other forward genetic applications such as to isolate over-expression suppressors or enhance the production of traits of interest.


Subject(s)
Cell Survival/genetics , Cryptococcus neoformans/genetics , DNA, Bacterial/genetics , Genes, Essential/genetics , Agrobacterium tumefaciens/genetics , Gene Expression Regulation, Fungal/genetics , Mutagenesis, Insertional , Mutation , Phenotype , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/genetics , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/genetics
13.
Mol Microbiol ; 102(4): 715-737, 2016 11.
Article in English | MEDLINE | ID: mdl-27558514

ABSTRACT

Iron is a key trace element important for many biochemical processes and its availability varies with the environment. For human pathogenic fungi iron acquisition can be particularly problematical because host cells sequester free iron as part of the acute-phase response to infection. Fungi rely on high-affinity iron uptake systems, such as reductive iron assimilation (RIA) and siderophore-mediated iron uptake (non-RIA). These have been extensively studied in pathogenic fungi that exist outside of host cells, but much less is known for intracellular fungal pathogens. Talaromyces marneffei is a dimorphic fungal pathogen endemic to Southeast Asia. In the host T. marneffei resides within macrophages where it grows as a fission yeast. T. marneffei has genes of both iron assimilation systems as well as a paralogue of the siderophore biosynthetic gene sidA, designated sidX. Unlike other fungi, deletion of sidA or sidX resulted in cell type-specific effects. Mutant analysis showed that T. marneffei yeast cells also employ RIA for iron acquisition, providing an additional system in this cell type that differs substantially from hyphal cells. These data illustrate the specialized iron acquisition systems used by the different cell types of a dimorphic fungal pathogen and highlight the complexity in siderophore-biosynthetic pathways amongst fungi.


Subject(s)
Iron/metabolism , Siderophores/metabolism , Talaromyces/cytology , Talaromyces/metabolism , Biosynthetic Pathways , Food , Homeostasis , Hyphae/metabolism , Macrophages/microbiology , Talaromyces/genetics , Transcription, Genetic
14.
mSphere ; 1(1)2016.
Article in English | MEDLINE | ID: mdl-27303703

ABSTRACT

For successful infection to occur, a pathogen must be able to evade or tolerate the host's defense systems. This requires the pathogen to first recognize the host environment and then signal this response to elicit a complex adaptive program in order to activate its own defense strategies. In both prokaryotes and eukaryotes, two-component signaling systems are utilized to sense and respond to changes in the external environment. The hybrid histidine kinases (HHKs) at the start of the two-component signaling pathway have been well characterized in human pathogens. However, how these HHKs regulate processes downstream currently remains unclear. This study describes the role of a response regulator downstream of these HHKs, sskA, in Talaromyces marneffei, a dimorphic human pathogen. sskA is required for asexual reproduction, hyphal morphogenesis, cell wall integrity, osmotic adaptation, and the morphogenesis of yeast cells both in vitro at 37°C and during macrophage infection, but not during dimorphic switching. Comparison of the ΔsskA mutant with a strain in which the mitogen-activated protein kinase (MAPK) of the high-osmolarity glycerol pathway (SakA) has been deleted suggests that SskA acts upstream of this pathway in T. marneffei to regulate these morphogenetic processes. This was confirmed by assessing the amount of phosphorylated SakA in the ΔsskA mutant, antifungal resistance due to a lack of SakA activation, and the ability of a constitutively active sakA allele (sakA(F316L) ) to suppress the ΔsskA mutant phenotypes. We conclude that SskA regulates morphogenesis and osmotic stress adaptation in T. marneffei via phosphorylation of the SakA MAPK of the high-osmolarity glycerol pathway. IMPORTANCE This is the first study in a dimorphic fungal pathogen to investigate the role of a response regulator downstream of two-component signaling systems and its connection to the high-osmolarity glycerol pathway. This study will inspire further research into the downstream components of two-component signaling systems and their role during pathogenic growth.

15.
FEMS Microbiol Rev ; 39(6): 797-811, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26253139

ABSTRACT

The ability of pathogenic fungi to switch between a multicellular hyphal and unicellular yeast growth form is a tightly regulated process known as dimorphic switching. Dimorphic switching requires the fungus to sense and respond to the host environment and is essential for pathogenicity. This review will focus on the role of dimorphism in fungi commonly called thermally dimorphic fungi, which switch to a yeast growth form during infection. This group of phylogenetically diverse ascomycetes includes Talaromyces marneffei (recently renamed from Penicillium marneffei), Blastomyces dermatitidis (teleomorph Ajellomyces dermatitidis), Coccidioides species (C. immitis and C. posadasii), Histoplasma capsulatum (teleomorph Ajellomyces capsulatum), Paracoccidioides species (P. brasiliensis and P. lutzii) and Sporothrix schenckii (teleomorph Ophiostoma schenckii). This review will explore both the signalling pathways regulating the morphological transition and the transcriptional responses necessary for intracellular growth. The physiological requirements of yeast cells during infection will also be discussed, highlighting recent advances in the understanding of the role of iron and calcium acquisition during infection.


Subject(s)
Adaptation, Physiological , Fungi/physiology , Host-Pathogen Interactions , Gene Expression Regulation, Fungal , Hyphae/physiology , Signal Transduction , Yeasts/physiology
16.
PLoS One ; 10(4): e0122728, 2015.
Article in English | MEDLINE | ID: mdl-25866870

ABSTRACT

Talaromyces marneffei (Basionym: Penicillium marneffei) is a significant opportunistic fungal pathogen in patients infected with human immunodeficiency virus in Southeast Asia. T. marneffei cells have been shown to become melanized in vivo. Melanins are pigment biopolymers which act as a non-specific protectant against various stressors and which play an important role during virulence in fungi. The synthesis of the two most commonly found melanins in fungi, the eumelanin DOPA-melanin and the allomelanin DHN-melanin, requires the action of laccase enzymes. The T. marneffei genome encodes a number of laccases and this study describes the characterization of one of these, pbrB, during growth and development. A strain carrying a PbrB-GFP fusion shows that pbrB is expressed at high levels during asexual development (conidiation) but not in cells growing vegetatively. The pbrB gene is required for the synthesis of DHN-melanin in conidia and when deleted results in brown pigmented conidia, in contrast to the green conidia of the wild type.


Subject(s)
Genes, Fungal , Laccase/genetics , Melanins/biosynthesis , Penicillium/genetics , Spores, Fungal/metabolism , Microscopy, Fluorescence , Naphthols , Penicillium/classification , Penicillium/enzymology , Phylogeny , Pigments, Biological/biosynthesis
17.
PLoS Pathog ; 11(3): e1004790, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25812137

ABSTRACT

During infection, pathogens must utilise the available nutrient sources in order to grow while simultaneously evading or tolerating the host's defence systems. Amino acids are an important nutritional source for pathogenic fungi and can be assimilated from host proteins to provide both carbon and nitrogen. The hpdA gene of the dimorphic fungus Penicillium marneffei, which encodes an enzyme which catalyses the second step of tyrosine catabolism, was identified as up-regulated in pathogenic yeast cells. As well as enabling the fungus to acquire carbon and nitrogen, tyrosine is also a precursor in the formation of two types of protective melanin; DOPA melanin and pyomelanin. Chemical inhibition of HpdA in P. marneffei inhibits ex vivo yeast cell production suggesting that tyrosine is a key nutrient source during infectious growth. The genes required for tyrosine catabolism, including hpdA, are located in a gene cluster and the expression of these genes is induced in the presence of tyrosine. A gene (hmgR) encoding a Zn(II)2-Cys6 binuclear cluster transcription factor is present within the cluster and is required for tyrosine induced expression and repression in the presence of a preferred nitrogen source. AreA, the GATA-type transcription factor which regulates the global response to limiting nitrogen conditions negatively regulates expression of cluster genes in the absence of tyrosine and is required for nitrogen metabolite repression. Deletion of the tyrosine catabolic genes in the cluster affects growth on tyrosine as either a nitrogen or carbon source and affects pyomelanin, but not DOPA melanin, production. In contrast to other genes of the tyrosine catabolic cluster, deletion of hpdA results in no growth within macrophages. This suggests that the ability to catabolise tyrosine is not required for macrophage infection and that HpdA has an additional novel role to that of tyrosine catabolism and pyomelanin production during growth in host cells.


Subject(s)
Fungal Proteins/metabolism , Hydrolases/metabolism , Macrophages/metabolism , Mycoses/metabolism , Penicillium/growth & development , Tyrosine/metabolism , Animals , Cell Line , Fungal Proteins/genetics , Hydrolases/genetics , Macrophages/microbiology , Macrophages/pathology , Mice , Mycoses/pathology , Penicillium/genetics , Tyrosine/genetics
18.
G3 (Bethesda) ; 3(11): 1997-2014, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24062530

ABSTRACT

Penicillium marneffei is an opportunistic human pathogen endemic to Southeast Asia. At 25° P. marneffei grows in a filamentous hyphal form and can undergo asexual development (conidiation) to produce spores (conidia), the infectious agent. At 37° P. marneffei grows in the pathogenic yeast cell form that replicates by fission. Switching between these growth forms, known as dimorphic switching, is dependent on temperature. To understand the process of dimorphic switching and the physiological capacity of the different cell types, two microarray-based profiling experiments covering approximately 42% of the genome were performed. The first experiment compared cells from the hyphal, yeast, and conidiation phases to identify "phase or cell-state-specific" gene expression. The second experiment examined gene expression during the dimorphic switch from one morphological state to another. The data identified a variety of differentially expressed genes that have been organized into metabolic clusters based on predicted function and expression patterns. In particular, C-14 sterol reductase-encoding gene ergM of the ergosterol biosynthesis pathway showed high-level expression throughout yeast morphogenesis compared to hyphal. Deletion of ergM resulted in severe growth defects with increased sensitivity to azole-type antifungal agents but not amphotericin B. The data defined gene classes based on spatio-temporal expression such as those expressed early in the dimorphic switch but not in the terminal cell types and those expressed late. Such classifications have been helpful in linking a given gene of interest to its expression pattern throughout the P. marneffei dimorphic life cycle and its likely role in pathogenicity.


Subject(s)
Fungal Proteins/genetics , Genome, Fungal , Penicillium/genetics , Antifungal Agents/pharmacology , Cluster Analysis , Ergosterol/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal/drug effects , Humans , Membrane Fluidity , Oligonucleotide Array Sequence Analysis , Penicillium/drug effects , Penicillium/growth & development , Sequence Analysis, DNA , Temperature , Transcriptome/drug effects
19.
Eukaryot Cell ; 12(2): 154-60, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23204189

ABSTRACT

Penicillium marneffei is an emerging human-pathogenic fungus endemic to Southeast Asia. Like a number of other fungal pathogens, P. marneffei exhibits temperature-dependent dimorphic growth and grows in two distinct cellular morphologies, hyphae at 25°C and yeast cells at 37°C. Hyphae can differentiate to produce the infectious agents, asexual spores (conidia), which are inhaled into the host lung, where they are phagocytosed by pulmonary alveolar macrophages. Within macrophages, conidia germinate into unicellular yeast cells, which divide by fission. This minireview focuses on the current understanding of the genes required for the morphogenetic control of conidial germination, hyphal growth, asexual development, and yeast morphogenesis in P. marneffei.


Subject(s)
Hyphae/growth & development , Morphogenesis , Mycoses/microbiology , Penicillium/growth & development , Cell Polarity , Genes, Fungal , Host-Pathogen Interactions , Humans , Hyphae/genetics , Hyphae/physiology , Penicillium/genetics , Penicillium/physiology , Phylogeny , Reproduction, Asexual , Spores, Fungal/genetics , Spores, Fungal/growth & development , Spores, Fungal/physiology , Yeasts/growth & development
20.
PLoS Pathog ; 8(10): e1002851, 2012.
Article in English | MEDLINE | ID: mdl-23055919

ABSTRACT

Molecular genetic approaches typically detect recombination in microbes regardless of assumed asexuality. However, genetic data have shown the AIDS-associated pathogen Penicillium marneffei to have extensive spatial genetic structure at local and regional scales, and although there has been some genetic evidence that a sexual cycle is possible, this haploid fungus is thought to be genetically, as well as morphologically, asexual in nature because of its highly clonal population structure. Here we use comparative genomics, experimental mixed-genotype infections, and population genetic data to elucidate the role of recombination in natural populations of P. marneffei. Genome wide comparisons reveal that all the genes required for meiosis are present in P. marneffei, mating type genes are arranged in a similar manner to that found in other heterothallic fungi, and there is evidence of a putatively meiosis-specific mutational process. Experiments suggest that recombination between isolates of compatible mating types may occur during mammal infection. Population genetic data from 34 isolates from bamboo rats in India, Thailand and Vietnam, and 273 isolates from humans in China, India, Thailand, and Vietnam show that recombination is most likely to occur across spatially and genetically limited distances in natural populations resulting in highly clonal population structure yet sexually reproducing populations. Predicted distributions of three different spatial genetic clusters within P. marneffei overlap with three different bamboo rat host distributions suggesting that recombination within hosts may act to maintain population barriers within P. marneffei.


Subject(s)
Genes, Mating Type, Fungal , Mycoses/microbiology , Penicillium/genetics , Penicillium/physiology , Reproduction, Asexual/genetics , AIDS-Related Opportunistic Infections/microbiology , Animals , Asia, Southeastern , Comparative Genomic Hybridization , Genetic Variation , Genotype , Host-Pathogen Interactions , Linkage Disequilibrium , Male , Meiosis/genetics , Mice , Muridae/microbiology , Mycoses/veterinary , Penicillium/isolation & purification , Recombination, Genetic , Rodent Diseases/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...