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

Country/Region as subject
Publication year range
1.
Mol Cell Proteomics ; 17(12): 2462-2479, 2018 12.
Article in English | MEDLINE | ID: mdl-30206180

ABSTRACT

Hmt1p is the predominant arginine methyltransferase in Saccharomyces cerevisiae Its substrate proteins are involved in transcription, transcriptional regulation, nucleocytoplasmic transport and RNA splicing. Hmt1p-catalyzed methylation can also modulate protein-protein interactions. Hmt1p is conserved from unicellular eukaryotes through to mammals where its ortholog, PRMT1, is lethal upon knockout. In yeast, however, the effect of knockout on the transcriptome and proteome has not been described. Transcriptome analysis revealed downregulation of phosphate-responsive genes in hmt1Δ, including acid phosphatases PHO5, PHO11, and PHO12, phosphate transporters PHO84 and PHO89 and the vacuolar transporter chaperone VTC3 Analysis of the hmt1Δ proteome revealed decreased abundance of phosphate-associated proteins including phosphate transporter Pho84p, vacuolar alkaline phosphatase Pho8p, acid phosphatase Pho3p and subunits of the vacuolar transporter chaperone complex Vtc1p, Vtc3p and Vtc4p. Consistent with this, phosphate homeostasis was dysregulated in hmt1Δ cells, showing decreased extracellular phosphatase levels and decreased total Pi in phosphate-depleted medium. In vitro, we showed that transcription factor Pho4p can be methylated at Arg-241, which could explain phosphate dysregulation in hmt1Δ if interplay exists with phosphorylation at Ser-242 or Ser-243, or if Arg-241 methylation affects the capacity of Pho4p to homodimerize or interact with Pho2p. However, the Arg-241 methylation site was not validated in vivo and the localization of a Pho4p-GFP fusion in hmt1Δ was not different from wild type. To our knowledge, this is the first study to reveal an association between Hmt1p and phosphate homeostasis and one which suggests a regulatory link between S-adenosyl methionine and intracellular phosphate.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Phosphates/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Acid Phosphatase/genetics , Arginine/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Homeostasis/genetics , Methylation , Microscopy, Fluorescence , Proteome/genetics , Tandem Mass Spectrometry , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
2.
Am J Pathol ; 188(7): 1653-1665, 2018 07.
Article in English | MEDLINE | ID: mdl-29929915

ABSTRACT

The innate immune system is the primary defense against cryptococcal infection, but paradoxically it promotes infection of the central nervous system. We performed a detailed longitudinal study of neurocryptococcosis in normal, chimeric, green fluorescent protein phagocyte-positive mice and phagocyte-depleted mice and interrogated the central nervous system innate immune response to Cryptococcus neoformans H99 using confocal microscopy, histology, flow cytometry, and quantification of brain cytokine/chemokines and fungal burdens. C. neoformans was present in the perivascular space (PVS) of post-capillary venules. This was associated with a massive influx of blood-derived monocytes, neutrophils, and T lymphocytes into the PVS and a predominantly proinflammatory cytokine/chemokine response. Phagocytes containing cryptococci were present only in the lumen and corresponding PVS of post-capillary venules. Free cryptococci were observed breaching the glia limitans, the protective barrier between the PVS and the cerebral parenchyma. Parenchymal cryptococcomas were typically in direct contact with post-capillary venules and lacked surrounding immune cell infiltrates. Phagocyte depletion abrogated cryptococcoma formation and PVS infiltrates. Together, these observations suggest that cryptococcomas can originate via phagocyte-dependent transport across post-capillary venular endothelium into the PVS and thence via passage of free cryptococci into the brain. In conclusion, we demonstrate for the first time that the PVS of cortical post-capillary venules is the major site of the early innate immune response to, and phagocyte-dependent entry of, C. neoformans.


Subject(s)
Brain/immunology , Cryptococcus neoformans/immunology , Immunity, Innate/immunology , Meningitis, Cryptococcal/immunology , Phagocytes/immunology , T-Lymphocytes/immunology , Venules/immunology , Animals , Brain/microbiology , Brain/pathology , Disease Models, Animal , Female , Meningitis, Cryptococcal/microbiology , Meningitis, Cryptococcal/pathology , Mice , Mice, Inbred C57BL , Monocytes , Phagocytes/microbiology , Phagocytes/pathology , T-Lymphocytes/microbiology , T-Lymphocytes/pathology , Venules/microbiology , Venules/pathology
3.
Cell Microbiol ; 20(2)2018 02.
Article in English | MEDLINE | ID: mdl-29113016

ABSTRACT

Cryptococcus neoformans is a basidiomycetous yeast and the cause of cryptococcosis in immunocompromised individuals. The most severe form of the disease is meningoencephalitis, which is one of the leading causes of death in HIV/AIDS patients. In order to access the central nervous system, C. neoformans relies on the activity of certain virulence factors such as urease, which allows transmigration through the blood-brain barrier. In this study, we demonstrate that the calcium transporter Pmc1 enables C. neoformans to penetrate the central nervous system, because the pmc1 null mutant failed to infect and to survive within the brain parenchyma in a murine systemic infection model. To investigate potential alterations in transmigration pathways in these mutants, global expression profiling of the pmc1 mutant strain was undertaken, and genes associated with urease, the Ca2+ -calcineurin pathway, and capsule assembly were identified as being differentially expressed. Also, a decrease in urease activity was observed in the calcium transporter null mutants. Finally, we demonstrate that the transcription factor Crz1 regulates urease activity and that the Ca2+ -calcineurin signalling pathway positively controls the transcription of calcium transporter genes and factors related to transmigration.


Subject(s)
Central Nervous System/microbiology , Cryptococcus neoformans/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Proteins/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Brain/metabolism , Brain/microbiology , Calcineurin/metabolism , Calcium/metabolism , Cell Line , Cryptococcosis/metabolism , Cryptococcosis/microbiology , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells , Humans , Meningoencephalitis/metabolism , Meningoencephalitis/microbiology , Mice , Mice, Inbred BALB C , Vacuoles/metabolism , Vacuoles/microbiology , Virulence/physiology , Virulence Factors/metabolism
4.
Microbiology (Reading) ; 163(10): 1457-1465, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28949902

ABSTRACT

With the growing demand for new antibiotics to combat increasing multi-drug resistance, a family of antimicrobial peptides known as cathelicidins has emerged as potential candidates. Expansions in cathelicidin-encoding genes in marsupials and monotremes are of specific interest as the peptides they encode have evolved to protect immunologically naive young in the harsh conditions of the pouch and burrow. Our previous work demonstrated that some marsupial and monotreme cathelicidins have broad-spectrum antibacterial activity and kill resistant bacteria, but the activity of many cathelicidins is unknown. To investigate associations between peptide antimicrobial activity and physiochemical properties, we tested 15 cathelicidin mature peptides from tammar wallaby, grey short-tailed opossum, platypus and echidna for antimicrobial activity against a range of bacterial and fungal clinical isolates. One opossum cathelicidin ModoCath4, tammar wallaby MaeuCath7 and echidna Taac-CATH1 had broad-spectrum antibacterial activity and killed methicillin-resistant Staphylococcus aureus. However, antimicrobial activity was reduced in the presence of serum or whole blood, and non-specific toxicity was observed at high concentrations. The active peptides were highly charged, potentially increasing binding to microbial surfaces, and contained amphipathic helical structures, which may facilitate membrane permeabilisation. Peptide sequence homology, net charge, amphipathicity and alpha helical content did not correlate with antimicrobial activity. However active peptides contained a significantly higher percentage of cationic residues than inactive ones, which may be used to predict active peptides in future work. Along with previous studies, our results indicate that marsupial and monotreme cathelicidins show potential for development as novel therapeutics to combat increasing antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cathelicidins/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/chemistry , Bacteria/growth & development , Cathelicidins/chemistry , Cell Membrane/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Marsupialia , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/physiology , Microbial Sensitivity Tests , Monotremata
5.
Infect Immun ; 83(4): 1296-304, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605772

ABSTRACT

Cryptococcus neoformans is an opportunistic fungal pathogen and a leading cause of fungal-infection-related fatalities, especially in immunocompromised hosts. Several virulence factors are known to play a major role in the pathogenesis of cryptococcal infections, including the enzyme phospholipase B1 (Plb1). Compared to other well-studied Cryptococcus neoformans virulence factors such as the polysaccharide capsule and melanin production, very little is known about the contribution of Plb1 to cryptococcal virulence. Phospholipase B1 is a phospholipid-modifying enzyme that has been implicated in multiple stages of cryptococcal pathogenesis, including initiation and persistence of pulmonary infection and dissemination to the central nervous system, but the underlying reason for these phenotypes remains unknown. Here we demonstrate that a Δplb1 knockout strain of C. neoformans has a profound defect in intracellular growth within host macrophages. This defect is due to a combination of a 50% decrease in proliferation and a 2-fold increase in cryptococcal killing within the phagosome. In addition, we show for the first time that the Δplb1 strain undergoes a morphological change during in vitro and in vivo intracellular infection, resulting in a subpopulation of very large titan cells, which may arise as a result of the attenuated mutant's inability to cope within the macrophage.


Subject(s)
Cryptococcosis/pathology , Cryptococcus neoformans/pathogenicity , Fungal Proteins/genetics , Lysophospholipase/genetics , Macrophages/immunology , Animals , Cell Line , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/immunology , Female , Gene Knockout Techniques , Macrophages/microbiology , Mice , Mice, Inbred A , Virulence Factors/genetics
6.
Fungal Genet Biol ; 67: 71-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24731805

ABSTRACT

Miltefosine (MI) is a novel, potential antifungal agent with activity against some yeast and filamentous fungal pathogens. We previously demonstrated in the model yeast, Saccharomyces cerevisiae, that MI causes disruption of mitochondrial membrane potential and apoptosis-like cell death via interaction with the Cox9p sub-unit of cytochrome c oxidase (COX). To identify additional mechanisms of antifungal action, MI resistance was induced in S. cerevisiae by exposure to the mutagen, ethyl methanesulfonate, and gene mutation(s) responsible for resistance were investigated. An MI-resistant haploid strain (H-C101) was created. Resistance was retained in the diploid strain (D-C101) following mating, confirming dominant inheritance. Phenotypic assessment of individual D-C101 tetrads revealed that only one mutant gene contributed to the MI-resistance phenotype. To identify this gene, the genome of H-C101 was sequenced and 17 mutated genes, including metacaspase-encoding MCA1, were identified. The MCA1 mutation resulted in substitution of asparagine (N) with aspartic acid (D) at position 164 (MCA1(N164D)). MI resistance was found to be primarily due to MCA1(N164D), as single-copy episomal expression of MCA1(N164D), but not two other mutated genes (FAS1(T1417I) and BCK2(T104A)), resulted in MI resistance in the wild-type strain. Furthermore, an MCA1 deletion mutant (mca1Δ) was MI-resistant. MI treatment led to accumulation of reactive oxygen species (ROS) in MI-resistant (MCA1(N164D)-expressing and mca1Δ) strains and MI-susceptible (MCA1-expressing) strains, but failed to activate Mca1 in the MI-resistant strains, demonstrating that ROS accumulation does not contribute to the fungicidal effect of MI. In conclusion, functional disruption of Mca1, leads to MI resistance and inability to mediate MI-induced apoptotic effects. Mca1-mediated apoptosis is therefore a major mechanism of MI-induced antifungal action.


Subject(s)
Antifungal Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Drug Resistance, Fungal , Phosphorylcholine/analogs & derivatives , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Caspases/genetics , Mutation , Phosphorylcholine/pharmacology , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
7.
PLoS Pathog ; 8(10): e1002957, 2012.
Article in English | MEDLINE | ID: mdl-23071437

ABSTRACT

We have investigated the potential of the GTP synthesis pathways as chemotherapeutic targets in the human pathogen Cryptococcus neoformans, a common cause of fatal fungal meningoencephalitis. We find that de novo GTP biosynthesis, but not the alternate salvage pathway, is critical to cryptococcal dissemination and survival in vivo. Loss of inosine monophosphate dehydrogenase (IMPDH) in the de novo pathway results in slow growth and virulence factor defects, while loss of the cognate phosphoribosyltransferase in the salvage pathway yielded no phenotypes. Further, the Cryptococcus species complex displays variable sensitivity to the IMPDH inhibitor mycophenolic acid, and we uncover a rare drug-resistant subtype of C. gattii that suggests an adaptive response to microbial IMPDH inhibitors in its environmental niche. We report the structural and functional characterization of IMPDH from Cryptococcus, revealing insights into the basis for drug resistance and suggesting strategies for the development of fungal-specific inhibitors. The crystal structure reveals the position of the IMPDH moveable flap and catalytic arginine in the open conformation for the first time, plus unique, exploitable differences in the highly conserved active site. Treatment with mycophenolic acid led to significantly increased survival times in a nematode model, validating de novo GTP biosynthesis as an antifungal target in Cryptococcus.


Subject(s)
Cryptococcus neoformans/enzymology , Cryptococcus neoformans/pathogenicity , Guanosine Triphosphate/biosynthesis , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Mycophenolic Acid/pharmacology , Animals , Antifungal Agents/pharmacology , Caenorhabditis elegans/microbiology , Cryptococcus gattii/drug effects , Cryptococcus gattii/genetics , Cryptococcus gattii/isolation & purification , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/metabolism , Crystallography, X-Ray , Drug Resistance, Fungal/genetics , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/genetics , Meningoencephalitis/microbiology
8.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798512

ABSTRACT

Many genetic studies have established the kinase activity of inositol phosphate multikinase (IPMK) is required for the synthesis of higher-order inositol phosphate signaling molecules, the regulation of gene expression and control of the cell cycle. These genetic studies await orthogonal validation by specific IPMK inhibitors, but no such inhibitors have been synthesized. Here, we report complete chemical synthesis, cellular characterization, structure-activity relationships and rodent pharmacokinetics of a novel series of highly potent IPMK inhibitors. The first-generation compound 1 (UNC7437) decreased cellular proliferation and tritiated inositol phosphate levels in metabolically labeled human U251-MG glioblastoma cells. Compound 1 also regulated the transcriptome of these cells, selectively regulating genes that are enriched in cancer, inflammatory and viral infection pathways. Further optimization of compound 1 eventually led to compound 15 (UNC9750), which showed improved potency and pharmacokinetics in rodents. Compound 15 specifically inhibited cellular accumulation of InsP 5 , a direct product of IPMK kinase activity, while having no effect on InsP 6 levels, revealing a novel metabolic signature detected for the first time by rapid chemical attenuation of cellular IPMK activity. These studies designed, optimized and synthesized a new series of IPMK inhibitors, which reduces glioblastoma cell growth, induces a novel InsP 5 metabolic signature, and reveals novel aspects inositol phosphate cellular metabolism and signaling.

9.
Infect Immun ; 81(4): 1245-55, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23381992

ABSTRACT

Phospholipase C (PLC) of Cryptococcus neoformans (CnPlc1) is crucial for virulence of this fungal pathogen. To investigate the mechanism of CnPlc1-mediated signaling, we established that phosphatidylinositol 4,5-bisphosphate (PIP(2)) is a major CnPlc1 substrate, which is hydrolyzed to produce inositol trisphosphate (IP(3)). In Saccharomyces cerevisiae, Plc1-derived IP(3) is a substrate for the inositol polyphosphate kinase Arg82, which converts IP(3) to more complex inositol polyphosphates. In this study, we show that in C. neoformans, the enzyme encoded by ARG1 is the major IP(3) kinase, and we further demonstrate that catalytic activity of Arg1 is essential for cellular homeostasis and virulence in the Galleria mellonella infection model. IP(3) content was reduced in the CnΔplc1 mutant and markedly increased in the CnΔarg1 mutant, while PIP(2) was increased in both mutants. The CnΔplc1 and CnΔarg1 mutants shared significant phenotypic similarity, including impaired thermotolerance, compromised cell walls, reduced capsule production and melanization, defective cell separation, and the inability to form mating filaments. In contrast to the S. cerevisiae ARG82 deletion mutant (ScΔarg82) strain, the CnΔarg1 mutant exhibited dramatically enlarged vacuoles indicative of excessive vacuolar fusion. In mammalian cells, PLC-derived IP(3) causes Ca(2+) release and calcineurin activation. Our data show that, unlike mammalian PLCs, CnPlc1 does not contribute significantly to calcineurin activation. Collectively, our findings provide the first evidence that the inositol polyphosphate anabolic pathway is essential for virulence of C. neoformans and further show that production of IP(3) as a precursor for synthesis of more complex inositol polyphosphates is the key biochemical function of CnPlc1.


Subject(s)
Arginase/metabolism , Cryptococcus neoformans/enzymology , Cryptococcus neoformans/pathogenicity , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Kinases/metabolism , Type C Phospholipases/metabolism , Virulence Factors/metabolism , Animals , Gene Deletion , Inositol 1,4,5-Trisphosphate/metabolism , Lepidoptera/microbiology , Metabolic Networks and Pathways/genetics , Models, Animal , Signal Transduction , Survival Analysis , Virulence
10.
Mol Biol Evol ; 29(8): 1987-2000, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22334577

ABSTRACT

The subtelomeric regions of organisms ranging from protists to fungi undergo a much higher rate of rearrangement than is observed in the rest of the genome. While characterizing these ~40-kb regions of the human fungal pathogen Cryptococcus neoformans, we have identified a recent gene amplification event near the right telomere of chromosome 3 that involves a gene encoding an arsenite efflux transporter (ARR3). The 3,177-bp amplicon exists in a tandem array of 2-15 copies and is present exclusively in strains with the C. neoformans var. grubii subclade VNI A5 MLST profile. Strains bearing the amplification display dramatically enhanced resistance to arsenite that correlates with the copy number of the repeat; the origin of increased resistance was verified as transport-related by functional complementation of an arsenite transporter mutant of Saccharomyces cerevisiae. Subsequent experimental evolution in the presence of increasing concentrations of arsenite yielded highly resistant strains with the ARR3 amplicon further amplified to over 50 copies, accounting for up to ~1% of the whole genome and making the copy number of this repeat as high as that seen for the ribosomal DNA. The example described here therefore represents a rare evolutionary intermediate-an array that is currently in a state of dynamic flux, in dramatic contrast to relatively common, static relics of past tandem duplications that are unable to further amplify due to nucleotide divergence. Beyond identifying and engineering fungal isolates that are highly resistant to arsenite and describing the first reported instance of microevolution via massive gene amplification in C. neoformans, these results suggest that adaptation through gene amplification may be an important mechanism that C. neoformans employs in response to environmental stresses, perhaps including those encountered during infection. More importantly, the ARR3 array will serve as an ideal model for further molecular genetic analyses of how tandem gene duplications arise and expand.


Subject(s)
Cryptococcus neoformans/genetics , Evolution, Molecular , Gene Amplification/genetics , Animals , Arsenites/metabolism , Arsenites/toxicity , Chromosomes, Fungal/genetics , Cryptococcosis/genetics , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/isolation & purification , Disease Models, Animal , Fungal Proteins/genetics , Gene Deletion , Gene Dosage/genetics , Genes, Fungal/genetics , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Phylogeny , Telomere/metabolism
11.
Fungal Genet Biol ; 61: 23-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24076076

ABSTRACT

Miltefosine (MI) has in vitro fungicidal activity against pathogenic fungi. However, mechanisms of resistance to MI have not been studied. By screening a genomic library of the model yeast, Saccharomyces cerevisiae, we identified HXT13 as a candidate genetic determinant of MI resistance. HXT13 belongs to the yeast hexose transporter family, which mediates hexose sugar uptake and is included in the major facilitator superfamily (MFS). We now report that overexpression of HXT13, but not of the closely-related genes, HXT15 and HXT17, and the more distantly related HXT14, resulted in a stable MI-resistant phenotype in S. cerevisiae. Resistance of the HXT13 overexpressing strain to MI correlated with higher cell viability following MI exposure as assessed by SYTOX® green staining compared with the control and overexpressing HXT14 strains. The mechanism of resistance in the HXT13 overexpressing strain was due to increased ATP-independent MI efflux. However, resistance to MI of the HXT13-overexpressing strain did not extend to other drugs including the echinocandins, amphotericin B, azoles, cycloheximide and sulfometuron methyl, ruling out the involvement of HXT13 in multidrug resistance. In summary, we have identified a new function of the hexose sugar transporter gene HXT13 when overexpressed in S. cerevisiae, namely, in efflux of MI and in mediating MI resistance.


Subject(s)
Antifungal Agents/metabolism , Drug Resistance, Fungal , Monosaccharide Transport Proteins/metabolism , Phosphorylcholine/analogs & derivatives , Saccharomyces cerevisiae/enzymology , Gene Expression , Microbial Viability/drug effects , Monosaccharide Transport Proteins/genetics , Phosphorylcholine/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics
12.
J Antimicrob Chemother ; 68(12): 2842-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23861311

ABSTRACT

OBJECTIVES: Antifungal treatment of uncommon filamentous fungal infections is problematic. This study determined the in vitro susceptibility of miltefosine, as a single agent and in combination with posaconazole or voriconazole, against these pathogens. METHODS: Susceptibility to miltefosine of 34 uncommon filamentous fungi was tested using CLSI broth microdilution M38-A2 methodology. Twenty isolates were studied for potential synergy using miltefosine/posaconazole and miltefosine/voriconazole combinations and the chequerboard microdilution assay. RESULTS: MICs of miltefosine were high (in general, >8 mg/L) for most isolates compared with amphotericin B, echinocandins and the azoles. Miltefosine had greatest activity against Scedosporium spp., Lichtheimia corymbifera and Rhizomucor sp. (MICs ≤ 4 mg/L). Miltefosine in combination either with posaconazole or voriconazole demonstrated synergy [fractional inhibitory concentration index (FICI) ≤ 0.5] in 12 instances (11 isolates): miltefosine/posaconazole combinations were synergistic against 3 of 4 Fusarium oxysporum strains (FICI range 0.37-0.5) and 5 of 10 mucormycete strains (FICI range 0.06-0.5). The combination of voriconazole with miltefosine showed synergy against one Scedosporium prolificans isolate and three mucormycetes-a single strain each of L. corymbifera, Rhizopus oryzae and Rhizomucor sp. No antagonism was observed. CONCLUSIONS: Miltefosine demonstrated synergy in 8/20 (40%) and 4/20 (20%) instances when combined with posaconazole and voriconazole, respectively. Synergy was most often observed against F. oxysporum and the mucormycetes. Study of miltefosine/azole combinations as a novel antifungal approach is indicated.


Subject(s)
Antifungal Agents/pharmacology , Fungi/drug effects , Phosphorylcholine/analogs & derivatives , Pyrimidines/pharmacology , Triazoles/pharmacology , Drug Synergism , Fungi/isolation & purification , Humans , Microbial Sensitivity Tests , Mycoses/microbiology , Phosphorylcholine/pharmacology , Voriconazole
13.
Eukaryot Cell ; 11(4): 532-44, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22286093

ABSTRACT

Recent studies indicate that mitochondrial functions impinge on cell wall integrity, drug tolerance, and virulence of human fungal pathogens. However, the mechanistic aspects of these processes are poorly understood. We focused on the mitochondrial outer membrane SAM (Sorting and Assembly Machinery) complex subunit Sam37 in Candida albicans. Inactivation of SAM37 in C. albicans leads to a large reduction in fitness, a phenotype not conserved with the model yeast Saccharomyces cerevisiae. Our data indicate that slow growth of the sam37ΔΔ mutant results from mitochondrial DNA loss, a new function for Sam37 in C. albicans, and from reduced activity of the essential SAM complex subunit Sam35. The sam37ΔΔ mutant was hypersensitive to drugs that target the cell wall and displayed altered cell wall structure, supporting a role for Sam37 in cell wall integrity in C. albicans. The sensitivity of the mutant to membrane-targeting antifungals was not significantly altered. The sam37ΔΔ mutant was avirulent in the mouse model, and bioinformatics showed that the fungal Sam37 proteins are distant from their animal counterparts and could thus represent potential drug targets. Our study provides the first direct evidence for a link between mitochondrial function and cell wall integrity in C. albicans and is further relevant for understanding mitochondrial function in fitness, antifungal drug tolerance, and virulence of this major pathogen. Beyond the relevance to fungal pathogenesis, this work also provides new insight into the mitochondrial and cellular roles of the SAM complex in fungi.


Subject(s)
Candida albicans/metabolism , Cell Wall/metabolism , Fungal Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Animals , Antifungal Agents/pharmacology , Candida albicans/drug effects , Candida albicans/growth & development , Candida albicans/pathogenicity , Candidemia/microbiology , Cell Wall/ultrastructure , Cells, Cultured , DNA, Mitochondrial/metabolism , Fluconazole/pharmacology , Fungal Proteins/genetics , Hyphae/metabolism , Kidney/microbiology , Kidney/pathology , Macrophages/microbiology , Membrane Potential, Mitochondrial , Mice , Microbial Sensitivity Tests , Mitochondrial Proteins/genetics , Nematoda/microbiology , Organelle Shape , Phenotype , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Homology, Amino Acid , Virulence
14.
Mol Microbiol ; 79(4): 968-89, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21299651

ABSTRACT

The cell wall is essential for viability of fungi and is an effective drug target in pathogens such as Candida albicans. The contribution of post-transcriptional gene regulators to cell wall integrity in C. albicans is unknown. We show that the C. albicans Ccr4-Pop2 mRNA deadenylase, a regulator of mRNA stability and translation, is required for cell wall integrity. The ccr4/pop2 mutants display reduced wall ß-glucans and sensitivity to the echinocandin caspofungin. Moreover, the deadenylase mutants are compromised for filamentation and virulence. We demonstrate that defective cell walls in the ccr4/pop2 mutants are linked to dysfunctional mitochondria and phospholipid imbalance. To further understand mitochondrial function in cell wall integrity, we screened a Saccharomyces cerevisiae collection of mitochondrial mutants. We identify several mitochondrial proteins required for caspofungin tolerance and find a connection between mitochondrial phospholipid homeostasis and caspofungin sensitivity. We focus on the mitochondrial outer membrane SAM complex subunit Sam37, demonstrating that it is required for both trafficking of phospholipids between the ER and mitochondria and cell wall integrity. Moreover, in C. albicans also Sam37 is essential for caspofungin tolerance. Our study provides the basis for an integrative view of mitochondrial function in fungal cell wall biogenesis and resistance to echinocandin antifungal drugs.


Subject(s)
Candida albicans/genetics , Cell Wall/ultrastructure , Fungal Proteins/metabolism , Mitochondria/metabolism , Ribonucleases/metabolism , Animals , Candida albicans/drug effects , Candida albicans/metabolism , Candida albicans/pathogenicity , Caspofungin , Cell Wall/chemistry , Cell Wall/drug effects , Echinocandins/pharmacology , Fungal Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Fungal , Homeostasis , Lipopeptides , Mice , Mice, Inbred BALB C , Mitochondria/ultrastructure , Mutation , Oligonucleotide Array Sequence Analysis , Phospholipids/analysis , Polyadenylation , RNA, Fungal/genetics , Ribonucleases/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Virulence , beta-Glucans/analysis
15.
Eukaryot Cell ; 10(7): 895-901, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602483

ABSTRACT

Adaptation to host temperature is a prerequisite for any pathogen capable of causing deep infection in humans. Our previous studies demonstrated that a Cryptococcus neoformans ccr4Δ mutant lacking the major deadenylase involved in regulated mRNA decay was defective in host temperature adaptation and therefore virulence. In this study, the ccr4Δ mutant was found to exhibit characteristics of chronic unfolded-protein response (UPR) engagement in both the gene expression profile and phenotype. We demonstrate that host temperature adaptation in C. neoformans is accompanied by transient induction of the endoplasmic reticulum (ER) stress response and that Ccr4-dependent posttranscriptional gene regulation contributes to resolution of ER stress during host temperature adaptation.


Subject(s)
Adaptation, Physiological/genetics , Cryptococcus neoformans/metabolism , Endoplasmic Reticulum/metabolism , Receptors, CCR4/genetics , Stress, Physiological/genetics , Body Temperature , Cryptococcus neoformans/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Fungal , Gene Knockout Techniques , Microscopy, Fluorescence , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , Polymerase Chain Reaction , RNA, Messenger/metabolism , Unfolded Protein Response
16.
Mycopathologia ; 173(5-6): 407-18, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21898146

ABSTRACT

Secretion pathways in fungi are essential for the maintenance of cell wall architecture and for the export of a number of virulence factors. In the fungal pathogen, Cryptococcus neoformans, much evidence supports the existence of more than one route taken by secreted molecules to reach the cell periphery and extracellular space, and a significant degree of crosstalk between conventional and non-conventional secretion routes. The need for such complexity may be due to differences in the nature of the exported cargo, the spatial and temporal requirements for constitutive and non-constitutive protein secretion, and/or as a means of compensating for the extra burden on the secretion machinery imposed by the elaboration of the polysaccharide capsule. This review focuses on the role of specific components of the C. neoformans secretion machinery in protein and/or polysaccharide export, including Sec4, Sec6, Sec14, Golgi reassembly and stacking protein and extracellular exosome-like vesicles. We also address what is known about traffic of the lipid, glucosylceramide, a target of therapeutic antibodies and an important regulator of C. neoformans pathogenicity, and the role of signalling pathways in the regulation of secretion.


Subject(s)
Cryptococcus neoformans/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Proteins/metabolism , Polysaccharides/metabolism , Virulence Factors/metabolism , Membrane Transport Proteins/metabolism
17.
Pathogens ; 11(11)2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36364991

ABSTRACT

Cryptococcus neoformans (Cn) is a pathogenic yeast that is the leading cause of fungal meningitis in immunocompromised patients. Various Cn virulence factors, such as the enzyme laccase and its product melanin, phospholipase, and capsular polysaccharide have been identified. During a screen of knockout mutants, the gene resistance to aminocholesterol 1 (RTA1) was identified, the function of which is currently unknown in Cn. Rta1 homologs in S. cerevisiae belong to a lipid-translocating exporter family of fungal proteins with transmembrane regions and confer resistance to the antimicrobial agent 7-aminocholesterol when overexpressed. To determine the role of RTA1 in Cn, the knock-out (rta1Δ) and reconstituted (rta1Δ+RTA1) strains were created and phenotypically tested. RTA1 was involved in resistance to 7-aminocholesterol, and also in exocyst complex component 3 (Sec6)-mediated secretion of urease, laccase, and the major capsule component, glucuronoxylomannan (GXM), which coincided with significantly smaller capsules in the rta1Δ and rta1Δ+RTA1 strains compared to the wild-type H99 strain. Furthermore, RTA1 expression was reduced in a secretory 14 mutant (sec14Δ) and increased in an RNAi Sec6 mutant. Transmission electron microscopy demonstrated vesicle accumulation inside the rta1Δ strain, predominantly near the cell membrane. Given that Rta1 is likely to be a transmembrane protein located at the plasma membrane, these data suggest that Rta1 may be involved in both secretion of various fungal virulence factors and resistance to 7-aminocholesterol in Cn.

18.
Mol Pharmacol ; 80(3): 476-85, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21610197

ABSTRACT

Miltefosine has antifungal properties and potential for development as a therapeutic for invasive fungal infections. However, its mode of action in fungi is poorly understood. We demonstrate that miltefosine is rapidly incorporated into yeast, where it penetrates the mitochondrial inner membrane, disrupting mitochondrial membrane potential and leading to an apoptosis-like cell death. COX9, which encodes subunit VIIa of the cytochrome c oxidase (COX) complex in the electron transport chain of the mitochondrial membrane, was identified as a potential target of miltefosine from a genomic library screen of the model yeast Saccharomyces cerevisiae. When overexpressed in S. cerevisiae, COX9, but not COX7 or COX8, led to a miltefosine-resistant phenotype. The effect of miltefosine on COX activity was assessed in cells expressing different levels of COX9. Miltefosine inhibited COX activity in a dose-dependent manner in Cox9p-positive cells. This inhibition most likely contributed to the miltefosine-induced apoptosis-like cell death.


Subject(s)
Antifungal Agents/pharmacology , Apoptosis/drug effects , Electron Transport Complex IV/metabolism , Phosphorylcholine/analogs & derivatives , Saccharomyces cerevisiae/drug effects , Base Sequence , DNA Primers , Phosphorylcholine/pharmacology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology
19.
Mol Microbiol ; 76(2): 517-34, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20384682

ABSTRACT

The polysaccharide beta-1,6-glucan is a major component of the cell wall of Cryptococcus neoformans, but its function has not been investigated in this fungal pathogen. We have identified and characterized seven genes, belonging to the KRE family, which are putatively involved in beta-1,6-glucan synthesis. The H99 deletion mutants kre5Delta and kre6Deltaskn1Delta contained less cell wall beta-1,6-glucan, grew slowly with an aberrant morphology, were highly sensitive to environmental and chemical stress and were avirulent in a mouse inhalation model of infection. These two mutants displayed alterations in cell wall chitosan and the exopolysaccharide capsule, a primary cryptococcal virulence determinant. The cell wall content of the GPI-anchored phospholipase B1 (Plb1) enzyme, which is required for cryptococcal cell wall integrity and virulence, was reduced in kre5Delta and kre6Deltaskn1Delta. Our results indicate that KRE5, KRE6 and SKN1 are involved in beta-1,6-glucan synthesis, maintenance of cell wall integrity and retention of mannoproteins and known cryptococcal virulence factors in the cell wall of C. neoformans. This study sets the stage for future investigations into the function of this abundant cell wall polymer.


Subject(s)
Cell Wall/metabolism , Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Polysaccharides/metabolism , beta-Glucans/metabolism , Animals , Architecture , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/cytology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Disease Models, Animal , Fungal Proteins/genetics , Gene Deletion , Maintenance , Mice , Protein Binding , Survival Analysis , Virulence
20.
PLoS One ; 16(4): e0249658, 2021.
Article in English | MEDLINE | ID: mdl-33852625

ABSTRACT

Devastating fires in Australia over 2019-20 decimated native fauna and flora, including koalas. The resulting population bottleneck, combined with significant loss of habitat, increases the vulnerability of remaining koala populations to threats which include disease. Chlamydia is one disease which causes significant morbidity and mortality in koalas. The predominant pathogenic species, Chlamydia pecorum, causes severe ocular, urogenital and reproductive tract disease. In marsupials, including the koala, gene expansions of an antimicrobial peptide family known as cathelicidins have enabled protection of immunologically naïve pouch young during early development. We propose that koala cathelicidins are active against Chlamydia and other bacteria and fungi. Here we describe ten koala cathelicidins, five of which contained full length coding sequences that were widely expressed in tissues throughout the body. Focusing on these five, we investigate their antimicrobial activity against two koala C. pecorum isolates from distinct serovars; MarsBar and IPTaLE, as well as other bacteria and fungi. One cathelicidin, PhciCath5, inactivated C. pecorum IPTaLE and MarsBar elementary bodies and significantly reduced the number of inclusions compared to the control (p<0.0001). Despite evidence of cathelicidin expression within tissues known to be infected by Chlamydia, natural PhciCath5 concentrations may be inadequate in vivo to prevent or control C. pecorum infections in koalas. PhciCath5 also displayed antimicrobial activity against fungi and Gram negative and positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA). Electrostatic interactions likely drive PhciCath5 adherence to the pathogen cell membrane, followed by membrane permeabilisation leading to cell death. Activity against E. coli was reduced in the presence of 10% serum and 20% whole blood. Future modification of the PhciCath5 peptide to enhance activity, including in the presence of serum/blood, may provide a novel solution to Chlamydia infection in koalas and other species.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Phascolarctidae/microbiology , Animals , Anti-Infective Agents , Antimicrobial Cationic Peptides/pharmacology , Australia , Chlamydia/genetics , Chlamydia/pathogenicity , Chlamydia Infections/epidemiology , Chlamydia Infections/prevention & control , Escherichia coli/genetics , Marsupialia/genetics , Marsupialia/microbiology , Methicillin-Resistant Staphylococcus aureus/genetics , Phascolarctidae/genetics , Phascolarctidae/metabolism , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL