Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Methods Mol Biol ; 2775: 225-237, 2024.
Article in English | MEDLINE | ID: mdl-38758321

ABSTRACT

The polysaccharide capsule of Cryptococcus neoformans is the primary virulence factor and one of the most commonly studied aspects of this pathogenic yeast. Capsule size varies widely between strains, has the ability to grow rapidly when introduced to stressful or low-nutrient conditions, and has been positively correlated with strain virulence. For these reasons, the size of the capsule is of great interest to C. neoformans researchers. Inducing the growth of the C. neoformans capsule is used during phenotypic testing to help understand the effects of different treatments on the yeast or size differences between strains. Here, we describe one of the standard methods of capsule induction and detail two accepted methods of staining: (i) India ink, a negative stain, used in conjunction with conventional light microscopy and (ii) co-staining with fluorescent dyes of both the cell wall and capsule followed by confocal microscopy. Finally, we outline how to measure capsule diameter manually and offer a protocol for automated diameter measurement of India ink-stained samples using computational image analysis.


Subject(s)
Cryptococcus neoformans , Staining and Labeling , Cryptococcus neoformans/cytology , Staining and Labeling/methods , Microscopy, Confocal/methods , Cell Wall/metabolism , Cell Wall/ultrastructure , Fungal Capsules/metabolism , Image Processing, Computer-Assisted/methods , Fluorescent Dyes/chemistry , Carbon
2.
Methods Mol Biol ; 2775: 367-373, 2024.
Article in English | MEDLINE | ID: mdl-38758330

ABSTRACT

Glucuronoxylomannan (GXM) is the principal capsular component in the Cryptococcus genus. This complex polysaccharide participates in numerous events related to the physiology and pathogenesis of Cryptococcus, which highlights the importance of establishing methods for its isolation and analysis. Conventional methods for GXM isolation have been extensively discussed in the literature. In this chapter, we describe two fast methods for obtaining extracellular fractions enriched with cryptococcal GXM.


Subject(s)
Cryptococcus , Polysaccharides , Polysaccharides/chemistry , Antigens, Fungal/immunology , Cryptococcus neoformans , Fungal Capsules/metabolism , Fungal Capsules/chemistry , Humans
3.
Front Cell Infect Microbiol ; 14: 1369301, 2024.
Article in English | MEDLINE | ID: mdl-38774630

ABSTRACT

Dual-specificity LAMMER kinases are highly evolutionarily conserved in eukaryotes and play pivotal roles in diverse physiological processes, such as growth, differentiation, and stress responses. Although the functions of LAMMER kinase in fungal pathogens in pathogenicity and stress responses have been characterized, its role in Cryptococcus neoformans, a human fungal pathogen and a model yeast of basidiomycetes, remains elusive. In this study, we identified a LKH1 homologous gene and constructed a strain with a deleted LKH1 and a complemented strain. Similar to other fungi, the lkh1Δ mutant showed intrinsic growth defects. We observed that C. neoformans Lkh1 was involved in diverse stress responses, including oxidative stress and cell wall stress. Particularly, Lkh1 regulates DNA damage responses in Rad53-dependent and -independent manners. Furthermore, the absence of LKH1 reduced basidiospore formation. Our observations indicate that Lkh1 becomes hyperphosphorylated upon treatment with rapamycin, a TOR protein inhibitor. Notably, LKH1 deletion led to defects in melanin synthesis and capsule formation. Furthermore, we found that the deletion of LKH1 led to the avirulence of C. neoformans in a systemic cryptococcosis murine model. Taken together, Lkh1 is required for the stress response, sexual differentiation, and virulence of C. neoformans.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Fungal Proteins , Virulence , Animals , Female , Humans , Mice , Cell Wall/metabolism , Cryptococcosis/microbiology , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/genetics , Cryptococcus neoformans/enzymology , Disease Models, Animal , DNA Damage , Fungal Capsules/metabolism , Fungal Capsules/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Melanins/metabolism , Mice, Inbred BALB C , Oxidative Stress , Phosphorylation , Sirolimus/pharmacology , Spores, Fungal/growth & development , Stress, Physiological
4.
J Biol Chem ; 298(4): 101769, 2022 04.
Article in English | MEDLINE | ID: mdl-35218774

ABSTRACT

The polysaccharide capsule of fungal pathogen Cryptococcus neoformans is a critical virulence factor that has historically evaded complete characterization. Cryptococcal polysaccharides are known to either remain attached to the cell as capsular polysaccharides (CPSs) or to be shed into the extracellular space as exopolysaccharides (EPSs). While many studies have examined the properties of EPS, far less is known about CPS. In this work, we detail the development of new physical and enzymatic methods for the isolation of CPS which can be used to explore the architecture of the capsule and isolated capsular material. We show that sonication or Glucanex enzyme cocktail digestion yields soluble CPS preparations, while use of a French pressure cell press or Glucanex digestion followed by cell disruption removed the capsule and produced cell wall-associated polysaccharide aggregates that we call "capsule ghosts", implying an inherent organization that allows the CPS to exist independent of the cell wall surface. Since sonication and Glucanex digestion were noncytotoxic, it was also possible to observe the cryptococcal cells rebuilding their capsule, revealing the presence of reducing end glycans throughout the capsule. Finally, analysis of dimethyl sulfoxide-extracted and sonicated CPS preparations revealed the conservation of previously identified glucuronoxylomannan motifs only in the sonicated CPS. Together, these observations provide new insights into capsule architecture and synthesis, consistent with a model in which the capsule is assembled from the cell wall outward using smaller polymers, which are then compiled into larger ones.


Subject(s)
Cryptococcus neoformans , Fungal Capsules , Polysaccharides , Cell Wall/chemistry , Cell Wall/metabolism , Cryptococcosis/microbiology , Cryptococcus neoformans/metabolism , Fungal Capsules/chemistry , Fungal Capsules/metabolism , Polysaccharides/metabolism , Virulence Factors/metabolism
5.
BMC Microbiol ; 21(1): 341, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903172

ABSTRACT

BACKGROUND: Fungal infections impact over 25% of the global population. For the opportunistic fungal pathogen, Cryptococcus neoformans, infection leads to cryptococcosis. In the presence of the host, disease is enabled by elaboration of sophisticated virulence determinants, including polysaccharide capsule, melanin, thermotolerance, and extracellular enzymes. Conversely, the host protects itself from fungal invasion by regulating and sequestering transition metals (e.g., iron, zinc, copper) important for microbial growth and survival. RESULTS: Here, we explore the intricate relationship between zinc availability and fungal virulence via mass spectrometry-based quantitative proteomics. We observe a core proteome along with a distinct zinc-regulated protein-level signature demonstrating a shift away from transport and ion binding under zinc-replete conditions towards transcription and metal acquisition under zinc-limited conditions. In addition, we revealed a novel connection among zinc availability, thermotolerance, as well as capsule and melanin production through the detection of a Wos2 ortholog in the secretome under replete conditions. CONCLUSIONS: Overall, we provide new biological insight into cellular remodeling at the protein level of C. neoformans under regulated zinc conditions and uncover a novel connection between zinc homeostasis and fungal virulence determinants.


Subject(s)
Cryptococcus neoformans/pathogenicity , Molecular Chaperones/metabolism , Proteome/metabolism , Secretome/metabolism , Zinc/metabolism , Cryptococcus neoformans/metabolism , Fungal Capsules/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Melanins/metabolism , Molecular Chaperones/genetics , Mutation , Proteomics , Thermotolerance , Virulence/genetics
6.
mBio ; 12(6): e0279021, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34724824

ABSTRACT

The environmental yeast Cryptococcus neoformans is the most common cause of deadly fungal meningitis in primarily immunocompromised populations. A number of factors contribute to cryptococcal pathogenesis. Among them, inositol utilization has been shown to promote C. neoformans development in nature and invasion of central nervous system during dissemination. The mechanisms of the inositol regulation of fungal virulence remain incompletely understood. In this study, we analyzed inositol-induced capsule growth and the contribution of a unique inositol catabolic pathway in fungal development and virulence. We found that genes involved in the inositol catabolic pathway are highly induced by inositol, and they are also highly expressed in the cerebrospinal fluid of patients with meningoencephalitis. This pathway in C. neoformans contains three genes encoding myo-inositol oxygenases that convert myo-inositol into d-glucuronic acid, a substrate of the pentose phosphate cycle and a component of the polysaccharide capsule. Our mutagenesis analysis demonstrates that inositol catabolism is required for C. neoformans virulence and deletion mutants of myo-inositol oxygenases result in altered capsule growth as well as the polysaccharide structure, including O-acetylation. Our study indicates that the ability to utilize the abundant inositol in the brain may contribute to fungal pathogenesis in this neurotropic fungal pathogen. IMPORTANCE The human pathogen Cryptococcus neoformans is the leading cause of fungal meningitis in primarily immunocompromised populations. Understanding how this environmental organism adapts to the human host to cause deadly infection will guide our development of novel disease control strategies. Our recent studies revealed that inositol utilization by the fungus promotes C. neoformans development in nature and invasion of the central nervous system during infection. The mechanisms of the inositol regulation in fungal virulence remain incompletely understood. In this study, we found that C. neoformans has three genes encoding myo-inositol oxygenase, a key enzyme in the inositol catabolic pathway. Expression of these genes is highly induced by inositol, and they are highly expressed in the cerebrospinal fluid of patients with meningoencephalitis. Our mutagenesis analysis indeed demonstrates that inositol catabolism is required for C. neoformans virulence by altering the growth and structure of polysaccharide capsule, a major virulence factor. Considering the abundance of free inositol and inositol-related metabolites in the brain, our study reveals an important mechanism of host inositol-mediated fungal pathogenesis for this neurotropic fungal pathogen.


Subject(s)
Cryptococcus neoformans/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Capsules/chemistry , Inositol/metabolism , Meningitis, Cryptococcal/microbiology , Animals , Brain/metabolism , Brain/microbiology , Cryptococcus neoformans/chemistry , Cryptococcus neoformans/genetics , Female , Fungal Capsules/genetics , Fungal Capsules/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Humans , Male , Meningitis, Cryptococcal/metabolism , Mice , Oxygenases/genetics , Oxygenases/metabolism , Rabbits , Virulence
7.
N Biotechnol ; 58: 55-60, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32562862

ABSTRACT

Co-culture conditions are beneficial for study due to the advances which arise from symbiotic interactions and which cannot be replicated under pure culture conditions. Here, the focus is on the connection between two fungi - a yeast, Saccharomyces cerevisiae, and a filamentous fungus, Penicillium chrysogenum - in a yeast immobilization system termed' yeast biocapsules', where the yeast and filamentous fungus are strongly attached to one another, forming spherical structures. This co-culture condition hinders filamentous fungal biomass growth, while immobilization of yeast cells continues to increase. The effect of the co-culture condition on endometabolites or intracellular metabolites were tracked during the beginning and end of the yeast biocapsule formation period, and metabolites analyzed by Gas Chromatography-Mass Spectrometry Detector (GC-MSD). Distinct metabolite profiles were found between single culture conditions, involving each organism separately, and with the co-culture condition, where there were differences in 54 endometabolites. Specifically, co-culture condition compounds such as fructose, glycolic acid and glyceric acid were present in higher concentrations at the end of biocapsule formation. These results shed light on the mechanisms and biochemical impact of the interaction between the yeast and filamentous fungus and serve as a basis to apply and further develop yeast biocapsules as a new biotechnological tool with benefits for industry.


Subject(s)
Fungal Capsules/metabolism , Penicillium chrysogenum/metabolism , Saccharomyces cerevisiae/metabolism , Biomass , Biotechnology , Coculture Techniques , Fructose/chemistry , Fructose/metabolism , Fungal Capsules/chemistry , Gas Chromatography-Mass Spectrometry , Glyceric Acids/chemistry , Glyceric Acids/metabolism , Glycolates/chemistry , Glycolates/metabolism , Penicillium chrysogenum/chemistry , Penicillium chrysogenum/cytology , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/cytology
8.
J Mycol Med ; 30(1): 100905, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31706700

ABSTRACT

INTRODUCTION: Iron chelator has previously demonstrated fungicidal effects. This study aimed to investigate the antifungal activity of the iron chelators deferoxamine (DFO) and deferasirox (DSX) against Cryptococcus. MATERIALS AND METHODS: Cryptococcus neoformans and Cryptococcus gattii were used to determine the minimal inhibitory concentrations (MICs) of DFO and DSX, and the fractional inhibitory concentration index (FICI) of DFO and DSX when combined with amphotericin B (AMB). Expression of cryptococcal CFT1, CFT2, and CIR1 genes was determined using real-time polymerase chain reaction (PCR). RESULTS: Neither DFO nor DSX alone showed antifungal activity against Cryptococcus strains. When combined with AMB, the MICs of DFO and DSX decreased from>200µg/mL to 6.25 or 12.5µg/mL. The MIC of AMB decreased one-fold dilution in most strains when combined with iron chelators. The FICI of DFO+AMB and DSX+AMB was 0.5 and 1, respectively. C. neoformans showed significant growth retardation when incubated with a combination of sub-MIC concentrations of AMB and DFO; whereas, C. gattii demonstrated lesser growth retardation in DFO+AMB. No cryptococcal growth retardation was observed when DSX was combined with AMB. When C. neoformans was grown in DFO, the CFT1, CFT2, and CIR1 proteins were expressed 1.7, 2.0, and 0.9 times, respectively. When C. neoformans was grown in DSX, the CFT1, CFT2, and CIR1 genes were expressed 0.5, 0.6, and 0.3 times, respectively. CONCLUSION: Synergistic antifungal activity of combination DFO and AMB was observed in Cryptococcus. Relatively increased CFT1 and CFT2 expression may be associated with the effect of DFO that inhibits the growth of fungi.


Subject(s)
Cryptococcus/drug effects , Cryptococcus/growth & development , Cryptococcus/genetics , Iron Chelating Agents/pharmacology , Iron/metabolism , Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus/metabolism , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/genetics , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/metabolism , Deferasirox/pharmacology , Deferoxamine/pharmacology , Drug Synergism , Fungal Capsules/drug effects , Fungal Capsules/genetics , Fungal Capsules/metabolism , Gene Expression Regulation, Fungal/drug effects , Humans , Invasive Fungal Infections/complications , Invasive Fungal Infections/drug therapy , Invasive Fungal Infections/microbiology , Iron Overload/complications , Iron Overload/drug therapy , Iron Overload/microbiology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Microbial Sensitivity Tests , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Fungal Genet Biol ; 124: 59-72, 2019 03.
Article in English | MEDLINE | ID: mdl-30630094

ABSTRACT

Cryptococcus neoformans is a ubiquitous yeast pathogen that often infects the human central nervous system (CNS) to cause meningitis in immunocompromised individuals. Although numerous signaling pathways and factors important for fungal sexual reproduction and virulence have been investigated, their precise mechanism of action remains to be further elucidated. In this study, we identified and characterized a novel zinc finger protein Zfp1 that regulates fungal sexual reproduction and virulence in C. neoformans. qRT-PCR and ZFP1 promoter regulatory activity assays revealed a ubiquitous expression pattern of ZFP1 in all stages during mating. Subcellular localization analysis indicates that Zfp1 is targeted to the cytoplasm of C. neoformans. In vitro assays of stress responses showed that zfp1Δ mutants and the ZFP1 overexpressed strains ZFP1OE are hypersensitive to SDS, but not Congo red, indicating that Zfp1 may regulate cell membrane integrity. Zfp1 is also essential for fungal sexual reproduction because basidiospore production was blocked in bilateral mating between zfp1Δ mutants or ZFP1 overexpressed strains. Fungal nuclei development assay showed that nuclei in the bilateral mating of zfp1Δ mutants or ZFP1 overexpressed strains failed to undergo meiosis after fusion, indicating Zfp1 is important for regulating meiosis during mating. Although zfp1Δ mutants showed normal growth and produced normal major virulence factors, virulence was attenuated in a murine model. Interestingly, we found that the ZFP1 overexpressed strains were avirulent in a murine systemic-infection model. Overall, our study showed that the zinc finger protein Zfp1 is essential for fungal sporulation and virulence in C. neoformans.


Subject(s)
Cryptococcus neoformans/physiology , Cryptococcus neoformans/pathogenicity , Fungal Proteins/physiology , Zinc Fingers/physiology , Amino Acid Motifs , Animals , Blotting, Western , Cell Membrane/metabolism , Cell Nucleus Division/physiology , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/genetics , Female , Fungal Capsules/metabolism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Gene Expression Profiling , Meiosis/physiology , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Virulence , Zinc/metabolism , Zinc Fingers/genetics
10.
Sci Rep ; 8(1): 16378, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30401972

ABSTRACT

Cryptococcus neoformans is an opportunistic fungus that can cause lethal brain infections in immunosuppressed individuals. Infection usually occurs via the inhalation of a spore or desiccated yeast which can then disseminate from the lung to the brain and other tissues. Dissemination and disease is largely influence by the production of copious amounts of cryptococcal polysaccharides, both which are secreted to the extracellular environment or assembled into a thick capsule surrounding the cell body. There are two important polysaccharides: glucuronoxylomannan (GXM) and galactoxylomannan, also called as glucuronoxylomanogalactan (GXMGal or GalXM). Although GXM is more abundant, GalXM has a more potent modulatory effect. In the present study, we show that GalXM is a potent activator of murine dendritic cells, and when co-cultured with T cells, induces a Th17 cytokine response. We also demonstrated that treating mice with GalXM prior to infection with C. neoformans protects from infection, and this phenomenon is dependent on IL-6 and IL-17. These findings help us understand the immune biology of capsular polysaccharides in fungal pathogenesis.


Subject(s)
Cryptococcosis/metabolism , Cryptococcus neoformans/physiology , Fungal Capsules/metabolism , Interleukin-17/metabolism , Polysaccharides/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cryptococcosis/immunology , Cryptococcus neoformans/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Mice , Th17 Cells/cytology , Th17 Cells/drug effects
11.
Article in English | MEDLINE | ID: mdl-29844051

ABSTRACT

Cryptococcus spp. are common opportunistic fungal pathogens, particularly in HIV patients. The approved drug miltefosine (MFS) has potential as an alternative antifungal against cryptococcosis; however, the mechanism of action of MFS in Cryptococcus is poorly understood. Here, we examined the effects of MFS on C. neoformans and C. gattii yeasts (planktonic and biofilm lifestyles) to clarify its mechanism of action. MFS presented inhibitory and fungicidal effects against planktonic Cryptococcus cells, with similar activities against dispersion biofilm cells, while sessile biofilm cells were less sensitive to MFS. Interestingly, MFS had postantifungal effect on Cryptococcus, with a proliferation delay of up to 8.15 h after a short exposure to fungicidal doses. MFS at fungicidal concentrations increased the plasma membrane permeability, likely due to a direct interaction with ergosterol, as suggested by competition assays with exogenous ergosterol. Moreover, MFS reduced the mitochondrial membrane potential, increased reactive oxygen species (ROS) production, and induced DNA fragmentation and condensation, all of which are hallmarks of apoptosis. Transmission electron microscopy analysis showed that MFS-treated yeasts had a reduced mucopolysaccharide capsule (confirmed by morphometry with light microscopy), plasma membrane irregularities, mitochondrial swelling, and a less conspicuous cell wall. Our results suggest that MFS increases the plasma membrane permeability in Cryptococcus via an interaction with ergosterol and also affects the mitochondrial membrane, eventually leading to apoptosis, in line with its fungicidal activity. These findings confirm the potential of MFS as an antifungal against C. neoformans and C. gattii and warrant further studies to establish clinical protocols for MFS use against cryptococcosis.


Subject(s)
Antifungal Agents/pharmacology , Apoptosis/drug effects , Cryptococcus gattii/drug effects , Cryptococcus neoformans/drug effects , Phosphorylcholine/analogs & derivatives , Amphotericin B/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus gattii/metabolism , Cryptococcus gattii/ultrastructure , Cryptococcus neoformans/metabolism , Cryptococcus neoformans/ultrastructure , DNA Fragmentation/drug effects , Ergosterol/metabolism , Fungal Capsules/drug effects , Fungal Capsules/metabolism , Fungal Capsules/ultrastructure , Humans , Membrane Potential, Mitochondrial/drug effects , Microbial Sensitivity Tests , Opportunistic Infections/drug therapy , Opportunistic Infections/microbiology , Phosphorylcholine/pharmacology , Plankton/drug effects , Plankton/growth & development , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism
12.
Mem Inst Oswaldo Cruz ; 113(7): e180040, 2018.
Article in English | MEDLINE | ID: mdl-29742198

ABSTRACT

Cryptococcus neoformans is an opportunistic fungal pathogen that is ubiquitous in the environment. It causes a deadly meningitis that is responsible for over 180,000 deaths worldwide each year, including 15% of all AIDS-related deaths. The high mortality rates for this infection, even with treatment, suggest a need for improved therapy. Unique characteristics of C. neoformans may suggest directions for drug discovery. These include features of three structures that surround the cell: the plasma membrane, the cell wall around it, and the outermost polysaccharide capsule. We review current knowledge of the fundamental biology of these fascinating structures and highlight open questions in the field, with the goal of stimulating further investigation that will advance basic knowledge and human health.


Subject(s)
Cryptococcus neoformans , Fungal Capsules/metabolism , Fungal Proteins/biosynthesis , Polysaccharides/biosynthesis , Cell Wall , Cryptococcus neoformans/chemistry , Cryptococcus neoformans/cytology , Cryptococcus neoformans/pathogenicity , Virulence
13.
Med Mycol J ; 59(1): E1-E6, 2018.
Article in Japanese | MEDLINE | ID: mdl-29491337

ABSTRACT

This article presents the ultrastructural patterns of interactions between the murine lung macrophages and cells of low- (RKPGY-881, -1165, -1178) and high-virulence (RKPGY-1090, -1095, -1106) strains of Cryptococcus neoformans at the seventh post-experimental day. It was found that if macrophages ingest living yeast cells, the latter can: 1) become completely free from polysaccharide capsules, after that their contents undergo lysis, and cell wall debris are extruded from the macrophage (first scenario); 2) become partly free from their capsules, destroy the phagosomal plasma membrane and induce destructive processes inside the macrophage causing their death (second scenario); or 3) not lose their capsules and localize inside macrophage in latent state (third scenario). Macrophages can also ingest senescent and dead C. neoformans cells surrounded by capsules that are lost at the ingesting and phagosome stages (fourth scenario). The study revealed the dependence of cell-mediated immunity on the stage of development of ingested C. neoformans yeast cells. Here we describe a new mechanism of capsular polysaccharide elimination of C. neoformans yeast cells by murine macrophages.


Subject(s)
Cryptococcus neoformans/immunology , Cryptococcus neoformans/ultrastructure , Macrophages, Alveolar/immunology , Macrophages, Alveolar/ultrastructure , Phagocytosis , Animals , Cryptococcus neoformans/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Capsules/metabolism , Fungal Capsules/ultrastructure , Fungal Polysaccharides/metabolism , Immunity, Cellular/immunology , Male , Mice , Phagosomes , Virulence
14.
PLoS Pathog ; 14(1): e1006765, 2018 01.
Article in English | MEDLINE | ID: mdl-29346417

ABSTRACT

Cryptococcus neoformans, an AIDS-defining opportunistic pathogen, is the leading cause of fungal meningitis worldwide and is responsible for hundreds of thousands of deaths annually. Cryptococcal glycans are required for fungal survival in the host and for pathogenesis. Most glycans are made in the secretory pathway, although the activated precursors for their synthesis, nucleotide sugars, are made primarily in the cytosol. Nucleotide sugar transporters are membrane proteins that solve this topological problem, by exchanging nucleotide sugars for the corresponding nucleoside phosphates. The major virulence factor of C. neoformans is an anti-phagocytic polysaccharide capsule that is displayed on the cell surface; capsule polysaccharides are also shed from the cell and impede the host immune response. Xylose, a neutral monosaccharide that is absent from model yeast, is a significant capsule component. Here we show that Uxt1 and Uxt2 are both transporters specific for the xylose donor, UDP-xylose, although they exhibit distinct subcellular localization, expression patterns, and kinetic parameters. Both proteins also transport the galactofuranose donor, UDP-galactofuranose. We further show that Uxt1 and Uxt2 are required for xylose incorporation into capsule and protein; they are also necessary for C. neoformans to cause disease in mice, although surprisingly not for fungal viability in the context of infection. These findings provide a starting point for deciphering the substrate specificity of an important class of transporters, elucidate a synthetic pathway that may be productively targeted for therapy, and contribute to our understanding of fundamental glycobiology.


Subject(s)
Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Glycoproteins/metabolism , Nucleotide Transport Proteins/metabolism , Uridine Diphosphate Xylose/metabolism , Animals , Biological Transport , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/ultrastructure , Female , Fungal Capsules/metabolism , Fungal Capsules/ultrastructure , Fungal Proteins/genetics , Galactose/analogs & derivatives , Galactose/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Glycoproteins/genetics , Kinetics , Mice , Microscopy, Electron, Transmission , Mutation , Nucleotide Transport Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Uridine Diphosphate/analogs & derivatives , Uridine Diphosphate/metabolism , Virulence
15.
Diagn Microbiol Infect Dis ; 89(2): 143-145, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28784461

ABSTRACT

Diagnosis of central nervous system cryptococcosis relies on a spectrum of methods but has improved with lateral flow diagnostic assays that detect capsular polysaccharide antigens of Cryptococcus. Here, we present the case of an HIV-infected African-American man with cryptococcal meningoencephalitis caused by a strain producing little or no capsule.


Subject(s)
Antigens, Fungal/metabolism , Cryptococcus neoformans/metabolism , Fungal Capsules/metabolism , Fungal Polysaccharides/metabolism , Meningitis, Cryptococcal/diagnosis , Meningoencephalitis/diagnosis , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/microbiology , Adult , Amphotericin B/therapeutic use , Antifungal Agents/therapeutic use , Antigens, Fungal/immunology , Flucytosine/therapeutic use , Humans , Male , Meningitis, Cryptococcal/microbiology , Meningoencephalitis/microbiology
16.
Environ Microbiol Rep ; 9(3): 268-278, 2017 06.
Article in English | MEDLINE | ID: mdl-28251810

ABSTRACT

In this study, an aquaporin protein, Aqp1, in Cryptococcus neoformans, which can lead either saprobic or parasitic lifestyles and causes life-threatening fungal meningitis was identified and characterized. AQP1 expression was rapidly induced (via the HOG pathway) by osmotic or oxidative stress. In spite of such transcriptional regulation, Aqp1 was found to be largely unnecessary for adaptation to diverse environmental stressors, regardless of the presence of the polysaccharide capsule. The latter is shown here to be a key environmental-stress protectant for C. neoformans. Furthermore, Aqp1 was not required for the development and virulence of C. neoformans. Deletion of AQP1 increased hydrophobicity of the cell surface. The comparative metabolic profiling analysis of the aqp1Δ mutant and AQP1-overexpressing strains revealed that deletion of AQP1 significantly increased cellular accumulation of primary and secondary metabolites, whereas overexpression of AQP1 depleted such metabolites, suggesting that this water channel protein performs a critical function in metabolic homeostasis. In line with this result, it was found that the aqp1Δ mutant (which is enriched with diverse metabolites) survived better than the wild type and a complemented strain, indicating that Aqp1 is likely to be involved in competitive fitness of this fungal pathogen.


Subject(s)
Aquaporin 1/genetics , Aquaporin 1/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Proteins/genetics , Osmotic Pressure/physiology , Oxidative Stress/physiology , Animals , Cryptococcus neoformans/metabolism , Diamide/pharmacology , Fungal Capsules/genetics , Fungal Capsules/metabolism , Fungal Polysaccharides/genetics , Fungal Polysaccharides/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Homeostasis/physiology , Hydrophobic and Hydrophilic Interactions , Mice , Mitogen-Activated Protein Kinases/genetics , Virulence/genetics , tert-Butylhydroperoxide/pharmacology
17.
mBio ; 7(1): e01862-15, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26758180

ABSTRACT

UNLABELLED: The opportunistic fungal pathogen Cryptococcus neoformans causes life-threatening meningitis in immunocompromised individuals. The expression of virulence factors, including capsule and melanin, is in part regulated by the cyclic-AMP/protein kinase A (cAMP/PKA) signal transduction pathway. In this study, we investigated the influence of PKA on the composition of the intracellular proteome to obtain a comprehensive understanding of the regulation that underpins virulence. Through quantitative proteomics, enrichment and bioinformatic analyses, and an interactome study, we uncovered a pattern of PKA regulation for proteins associated with translation, the proteasome, metabolism, amino acid biosynthesis, and virulence-related functions. PKA regulation of the ubiquitin-proteasome pathway in C. neoformans showed a striking parallel with connections between PKA and protein degradation in chronic neurodegenerative disorders and other human diseases. Further investigation of proteasome function with the inhibitor bortezomib revealed an impact on capsule production as well as hypersusceptibility for strains with altered expression or activity of PKA. Parallel studies with tunicamycin also linked endoplasmic reticulum stress with capsule production and PKA. Taken together, the data suggest a model whereby expression of PKA regulatory and catalytic subunits and the activation of PKA influence proteostasis and the function of the endoplasmic reticulum to control the elaboration of the polysaccharide capsule. Overall, this study revealed both broad and conserved influences of the cAMP/PKA pathway on the proteome and identified proteostasis as a potential therapeutic target for the treatment of cryptococcosis. IMPORTANCE: Fungi cause life-threatening diseases, but very few drugs are available to effectively treat fungal infections. The pathogenic fungus Cryptococcus neoformans causes a substantial global burden of life-threatening meningitis in patients suffering from HIV/AIDS. An understanding of the mechanisms by which fungi deploy virulence factors to cause disease is critical for developing new therapeutic approaches. We employed a quantitative proteomic approach to define the changes in the protein complement that occur upon modulating the cAMP signaling pathway that regulates virulence in C. neoformans. This approach identified a conserved role for cAMP signaling in the regulation of the ubiquitin-proteasome pathway and revealed a link between this pathway and elaboration of a major virulence determinant, the polysaccharide capsule. Targeting the ubiquitin-proteasome pathway opens new therapeutic options for the treatment of cryptococcosis.


Subject(s)
Cryptococcus neoformans/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fungal Capsules/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Cyclic AMP/metabolism , Proteome/analysis , Proteomics
18.
J Microbiol Biotechnol ; 26(5): 918-27, 2016 May 28.
Article in English | MEDLINE | ID: mdl-26437944

ABSTRACT

Cryptococcus neoformans is a life-threatening pathogenic yeast that causes devastating meningoencephalitis. The mechanism of cryptococcal brain invasion is largely unknown, and recent studies suggest that its extracellular microvesicles may be involved in the invasion process. The 14-3-3 protein is abundant in the extracellular microvesicles of C. neoformans, and the 14-3-3-GFP fusion has been used as the microvesicle's marker. However, the physiological role of 14-3-3 has not been explored. In this report, we have found that C. neoformans contains a single 14-3-3 gene that apparently is an essential gene. To explore the functions of 14-3-3, we substituted the promoter region of the 14-3-3 with the copper-controllable promoter CTR4. The CTR4 regulatory strain showed an enlarged cell size, drastic changes in morphology, and a decrease in the thickness of the capsule under copper-enriched conditions. Furthermore, the mutant cells produced a lower amount of total proteins in their extracellular microvesicles and reduced adhesion to human brain microvascular endothelial cells in vitro. Proteomic analyses of the protein components under 14-3-3-overexpressed and -suppressed conditions revealed that the 14-3-3 function(s) might be associated with the microvesicle biogenesis. Our results support that 14-3-3 has diverse pertinent roles in both physiology and pathogenesis in C. neoformans. Its gene functions are closely relevant to the pathogenesis of this fungus.


Subject(s)
14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Cryptococcus neoformans/genetics , Acid Phosphatase/metabolism , Biomarkers/metabolism , Brain/blood supply , Brain/microbiology , Cell Adhesion/physiology , Copper/metabolism , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/pathogenicity , Endothelial Cells/microbiology , Fungal Capsules/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Laccase/metabolism , Mutation , Phenotype , Promoter Regions, Genetic , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
19.
Eur J Clin Microbiol Infect Dis ; 34(12): 2421-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26463450

ABSTRACT

The purpose of this investigation was to characterise the interactions of Cryptococcus neoformans with mammalian host alveolar epithelial cells and alveolar macrophages, with emphasis on the roles of the cryptococcal capsule and the host cell cytoskeletons. The adherence and internalisation of C. neoformans into mammalian lung cells and the roles of host cell cytoskeletons in host-pathogen interactions were studied using in vitro models coupled with a differential fluorescence assay, fluorescence staining, immunofluorescence and drug inhibition of actin and microtubule polymerisation. Under conditions devoid of opsonin and macrophage activation, C. neoformans has a high affinity towards MH-S alveolar macrophages, yet associated poorly to A549 alveolar epithelial cells. Acapsular C. neoformans adhered to and internalised into the mammalian cells more effectively compared to encapsulated cryptococci. Acapsular C. neoformans induced prominent actin reorganisation at the host-pathogen interface in MH-S alveolar macrophages, but minimally affected actin reorganisation in A549 alveolar epithelial cells. Acapsular C. neoformans also induced localisation of microtubules to internalised cryptococci in MH-S cells. Drug inhibition of actin and microtubule polymerisation both reduced the association of acapsular C. neoformans to alveolar macrophages. The current study visualises and confirms the interactions of C. neoformans with mammalian alveolar cells during the establishment of infection in the lungs. The acapsular form of C. neoformans effectively adhered to and internalised into alveolar macrophages by inducing localised actin reorganisation, relying on the host's actin and microtubule activities.


Subject(s)
Actin Cytoskeleton/metabolism , Cryptococcus neoformans/physiology , Epithelial Cells/physiology , Host-Pathogen Interactions , Macrophages/physiology , Microtubules/metabolism , Animals , Cell Adhesion , Cell Line , Endocytosis , Epithelial Cells/microbiology , Fungal Capsules/genetics , Fungal Capsules/metabolism , Humans , Macrophages/microbiology , Mice
20.
Med Mycol ; 53(8): 885-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26333356

ABSTRACT

This study evaluated the synergistic interactions between amphotericin B (AMB) and azithromycin (AZM), daptomycin (DAP), linezolid (LNZ), minocycline (MINO), fluconazole (FLZ), flucytosine (5FC), linezolid (LZD), or tigecycline (TIG) against clinical isolates of Cryptococcus neoformans var. grubii before and after capsule induction. High synergism (>75%) was observed for the combinations, AMB+5FC, AMB+TIG, AMB+AZM, AMB+LZD and AMB+MINO but only in the strains after capsule induction. The results show that the presence of the capsule may lower the minimum inhibitory concentrations (MICs) of antifungal agents, but antimicrobial activity can be improved by combining antifungal and antibacterial agents.


Subject(s)
Amphotericin B/pharmacology , Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Cryptococcus neoformans/drug effects , Drug Interactions , Fungal Capsules/metabolism , Cryptococcosis/microbiology , Cryptococcus neoformans/isolation & purification , Cryptococcus neoformans/metabolism , Humans , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL