Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
PLoS Pathog ; 19(10): e1011721, 2023 10.
Article in English | MEDLINE | ID: mdl-37812645

ABSTRACT

V-ATPase, which comprises 13-14 subunits, is essential for pH homeostasis in all eukaryotes, but its proper function requires a regulator to assemble its subunits. While RAVE (regulator of H+-ATPase of vacuolar and endosomal membranes) and Raboconnectin-3 complexes assemble V-ATPase subunits in Saccharomyces cerevisiae and humans, respectively, the function of the RAVE complex in fungal pathogens remains largely unknown. In this study, we identified two RAVE complex components, Rav1 and Wdr1, in the fungal meningitis pathogen Cryptococcus neoformans, and analyzed their roles. Rav1 and Wdr1 are orthologous to yeast RAVE and human Rabconnectin-3 counterparts, respectively, forming the hybrid RAVE (hRAVE) complex. Deletion of RAV1 caused severe defects in growth, cell cycle control, morphogenesis, sexual development, stress responses, and virulence factor production, while the deletion of WDR1 resulted in similar but modest changes, suggesting that Rav1 and Wdr1 play central and accessary roles, respectively. Proteomics analysis confirmed that Wdr1 was one of the Rav1-interacting proteins. Although the hRAVE complex generally has V-ATPase-dependent functions, it also has some V-ATPase-independent roles, suggesting a unique role beyond conventional intracellular pH regulation in C. neoformans. The hRAVE complex played a critical role in the pathogenicity of C. neoformans, and RAV1 deletion attenuated virulence and impaired blood-brain barrier crossing ability. This study provides comprehensive insights into the pathobiological roles of the fungal RAVE complex and suggests a novel therapeutic strategy for controlling cryptococcosis.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Saccharomyces cerevisiae Proteins , Vacuolar Proton-Translocating ATPases , Humans , Saccharomyces cerevisiae Proteins/metabolism , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Saccharomyces cerevisiae/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism
2.
Mar Drugs ; 20(3)2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35323494

ABSTRACT

Six new ß-resorcylic acid derivatives (1-5 and 7) were isolated from a halophyte-associated fungus, Colletotrichum gloeosporioides JS0419, together with four previously reported ß-resorcylic acid lactones (RALs). The relative and absolute stereochemistry of 1 was completely established by a combination of spectroscopic data and chemical reactions. The structures of the isolated compounds were elucidated by analysis of HRMS and NMR data. Notably, compounds 1-3 had a ß-resorcylic acid harboring a long unesterified aliphatic side chain, whereas the long aliphatic chains were esterified to form macrolactones in 4-9. Among the isolated compounds, monocillin I and radicicol showed potent antifungal activities against Cryptococcus neoformans, comparable to clinically available antifungal agents and radicicol showed weak antifungal activity against Candida albicans. These findings provide insight into the chemical diversity of fungal RAL-type compounds and their pharmacological potential.


Subject(s)
Antifungal Agents/pharmacology , Candida albicans/drug effects , Chenopodiaceae/microbiology , Colletotrichum/chemistry , Cryptococcus neoformans/drug effects , Hydroxybenzoates/pharmacology , Salt-Tolerant Plants/microbiology , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Candida albicans/growth & development , Cryptococcus neoformans/growth & development , Hydroxybenzoates/chemistry , Hydroxybenzoates/isolation & purification , Molecular Structure , Stereoisomerism
3.
Microbiology (Reading) ; 166(9): 797-799, 2020 09.
Article in English | MEDLINE | ID: mdl-32956032

ABSTRACT

Cryptococcus neoformans is a lethal fungus disguised in a polysaccharide coat. It can remain dormant in the host for decades prior to reactivation, causing systemic cryptococcosis in humans and other mammals. Cryptococcus deploys a multitude of traits to adapt to and survive within the host, including immunosuppression, an ability to replicate intra- and extra-cellularly in phagocytes, changes in morphology and ploidy, a predilection to infect the CNS, and the capacity to utilize neurotransmitters and unique carbon sources available in the brain. These pathogenic strategies displayed by this fungus might have evolved through its interactions with microbial predators in the environment.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans , Adaptation, Physiological , Animals , Cryptococcus neoformans/classification , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/physiology , Genome, Fungal , Humans , Phylogeny , Virulence
4.
PLoS Genet ; 13(9): e1006982, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28898238

ABSTRACT

Morphotype switch is a cellular response to external and internal cues. The Cryptococcus neoformans species complex can undergo morphological transitions between the yeast and the hypha form, and such morphological changes profoundly affect cryptococcal interaction with various hosts. Filamentation in Cryptococcus was historically considered a mating response towards pheromone. Recent studies indicate the existence of pheromone-independent signaling pathways but their identity or the effectors remain unknown. Here, we demonstrated that glucosamine stimulated the C. neoformans species complex to undergo self-filamentation. Glucosamine-stimulated filamentation was independent of the key components of the pheromone pathway, which is distinct from pheromone-elicited filamentation. Glucosamine stimulated self-filamentation in H99, a highly virulent serotype A clinical isolate and a widely used reference strain. Through a genetic screen of the deletion sets made in the H99 background, we found that Crz1, a transcription factor downstream of calcineurin, was essential for glucosamine-stimulated filamentation despite its dispensability for pheromone-mediated filamentation. Glucosamine promoted Crz1 translocation from the cytoplasm to the nucleus. Interestingly, multiple components of the high osmolality glycerol response (HOG) pathway, consisting of the phosphorelay system and some of the Hog1 MAPK module, acted as repressors of glucosamine-elicited filamentation through their calcineurin-opposing effect on Crz1's nuclear translocation. Surprisingly, glucosamine-stimulated filamentation did not require Hog1 itself and was distinct from the conventional general stress response. The results demonstrate that Cryptococcus can resort to multiple genetic pathways for morphological transition in response to different stimuli. Given that the filamentous form attenuates cryptococcal virulence and is immune-stimulatory in mammalian models, the findings suggest that morphogenesis is a fertile ground for future investigation into novel means to compromise cryptococcal pathogenesis.


Subject(s)
Cryptococcus neoformans/genetics , Fungal Proteins/genetics , Glucosamine/metabolism , Morphogenesis/genetics , Calcineurin/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cryptococcus neoformans/growth & development , Cytoplasm/genetics , Cytoplasm/metabolism , Fungal Proteins/metabolism , Hyphae/genetics , Hyphae/growth & development , Mitogen-Activated Protein Kinases/genetics , Pheromones/genetics , Pheromones/metabolism , Protein Transport/genetics , Signal Transduction , Transcription Factors/genetics
6.
J Nat Prod ; 82(8): 2078-2086, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31321978

ABSTRACT

A reduction in the strong immunosuppressive activity of FK506 (1) is essential for developing this compound as an antifungal agent. Seven new FK506 analogues modified at both the FK506-binding protein 12- and the calcineurin-binding regions were biosynthesized. 9-DeoxoFK520 (7) exhibited a >900-fold reduction in the in vitro immunosuppressive activity but maintained significant antifungal activity, indicating that the C-9 and C-21 positions are critical for separation of immunosuppressive and antifungal activities. 7 exhibited robust synergistic antifungal activity with fluconazole. FK506 (1) is a 23-membered macrolide produced by several Streptomyces species and is used as an immunosuppressive drug to prevent the rejection of transplanted organs. FK506 has also exhibited antifungal, neuroprotective, and neuroregenerative activities. In humans, FK506 binds to FK506-binding protein (FKBP) 12, and the resulting FKBP12-FK506 complex interacts with a Ca2+-calmodulin-dependent phosphatase, calcineurin (CaN). Inactivation of CaN by forming the FKBP12-FK506-CaN ternary complex prevents the activation of nuclear factor of activated T cells (NF-AT), inhibiting the production of interleukin-2 and subsequent T-cell proliferation. This CaN signaling pathway also plays a critical role in the growth and pathogenesis of major fungal pathogens such as Cryptococcus neoformans, Candida albicans, and Aspergillus fumigatus. Therefore, the synthesis of FK506 analogues that can discriminate human FKBP12/CaN from its fungal counterparts may separate antifungal activity from the immunosuppressive activity, thereby allowing the development of a novel antifungal agent.


Subject(s)
Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Animals , Antifungal Agents/chemistry , Aspergillus fumigatus/drug effects , Candida albicans/drug effects , Cryptococcus neoformans/drug effects , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Microbial Sensitivity Tests , Molecular Structure , Tacrolimus/chemistry , Tacrolimus/metabolism
7.
PLoS Genet ; 12(10): e1006350, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27695031

ABSTRACT

Fungal pathogens have evolved diverse strategies to sense host-relevant cues and coordinate cellular responses, which enable virulence and drug resistance. Defining circuitry controlling these traits opens new opportunities for chemical diversity in therapeutics, as the cognate inhibitors are rarely explored by conventional screening approaches. This has great potential to address the pressing need for new therapeutic strategies for invasive fungal infections, which have a staggering impact on human health. To explore this approach, we focused on a leading human fungal pathogen, Candida albicans, and screened 1,280 pharmacologically active compounds to identify those that potentiate the activity of echinocandins, which are front-line therapeutics that target fungal cell wall synthesis. We identified 19 compounds that enhance activity of the echinocandin caspofungin against an echinocandin-resistant clinical isolate, with the broad-spectrum chelator DTPA demonstrating the greatest synergistic activity. We found that DTPA increases susceptibility to echinocandins via chelation of magnesium. Whole genome sequencing of mutants resistant to the combination of DTPA and caspofungin identified mutations in the histidine kinase gene NIK1 that confer resistance to the combination. Functional analyses demonstrated that DTPA activates the mitogen-activated protein kinase Hog1, and that NIK1 mutations block Hog1 activation in response to both caspofungin and DTPA. The combination has therapeutic relevance as DTPA enhanced the efficacy of caspofungin in a mouse model of echinocandin-resistant candidiasis. We found that DTPA not only reduces drug resistance but also modulates morphogenesis, a key virulence trait that is normally regulated by environmental cues. DTPA induced filamentation via depletion of zinc, in a manner that is contingent upon Ras1-PKA signaling, as well as the transcription factors Brg1 and Rob1. Thus, we establish a new mechanism by which metal chelation modulates morphogenetic circuitry and echinocandin resistance, and illuminate a novel facet to metal homeostasis at the host-pathogen interface, with broad therapeutic potential.


Subject(s)
Candida albicans/genetics , Candidiasis/drug therapy , Metals/metabolism , Mitogen-Activated Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Antifungal Agents/pharmacology , Candida albicans/drug effects , Candida albicans/pathogenicity , Candidiasis/microbiology , Caspofungin , Cell Wall/drug effects , Chelating Agents/chemistry , Chelating Agents/pharmacology , Drug Resistance, Fungal/genetics , Echinocandins/pharmacology , Genome, Fungal , Humans , Lipopeptides/pharmacology , Metals/chemistry , Mice , Morphogenesis/drug effects , Morphogenesis/genetics , Mutation , Pentetic Acid/pharmacology , Signal Transduction
8.
Article in English | MEDLINE | ID: mdl-30181374

ABSTRACT

FK506 (tacrolimus) is an FDA-approved immunosuppressant indicated for the prevention of allograft rejections in patients undergoing organ transplants. In mammals, FK506 inhibits the calcineurin-nuclear factor of activated T cells (NFAT) pathway to prevent T-cell proliferation by forming a ternary complex with its binding protein, FKBP12, and calcineurin. FK506 also exerts antifungal activity by inhibiting calcineurin, which is essential for the virulence of human-pathogenic fungi. Nevertheless, FK506 cannot be used directly as an antifungal drug due to its immunosuppressive action. In this study, we analyzed the cytotoxicity, immunosuppressive activity, and antifungal activity of four FK506 analogs, 31-O-demethyl-FK506, 9-deoxo-FK506, 9-deoxo-31-O-demethyl-FK506, and 9-deoxo-prolyl-FK506, in comparison with that of FK506. The four FK506 analogs generally possessed lower cytotoxicity and immunosuppressive activity than FK506. The FK506 analogs, except for 9-deoxo-prolyl-FK506, had strong antifungal activity against Cryptococcus neoformans and Candida albicans, which are two major invasive pathogenic yeasts, due to the inhibition of the calcineurin pathway. Furthermore, the FK506 analogs, except for 9-deoxo-prolyl-FK506, had strong antifungal activity against the invasive filamentous fungus Aspergillus fumigatus Notably, 9-deoxo-31-O-demethyl-FK506 and 31-O-demethyl-FK506 exhibited robust synergistic antifungal activity with fluconazole, similar to FK506. Considering the antifungal efficacy, cytotoxicity, immunosuppressive activity, and synergistic effect with commercial antifungal drugs, we selected 9-deoxo-31-O-demethyl-FK506 for further evaluation of its in vivo antifungal efficacy in a murine model of systemic cryptococcosis. Although 9-deoxo-31-O-demethyl-FK506 alone was not sufficient to treat the cryptococcal infection, when it was used in combination with fluconazole, it significantly extended the survival of C. neoformans-infected mice, confirming the synergistic in vivo antifungal efficacy between these two agents.


Subject(s)
Antifungal Agents/pharmacology , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Animals , Aspergillosis/drug therapy , Aspergillosis/microbiology , Aspergillus fumigatus/drug effects , Calcineurin/pharmacology , Calcineurin Inhibitors/pharmacology , Candida albicans/drug effects , Candidiasis/drug therapy , Candidiasis/microbiology , Cells, Cultured , Cryptococcosis/drug therapy , Cryptococcosis/microbiology , Cryptococcus neoformans/drug effects , Female , Fluconazole/pharmacology , Immunosuppressive Agents/pharmacology , Male , Mice , Microbial Sensitivity Tests/methods , Tacrolimus Binding Protein 1A/pharmacology
9.
Nat Chem Biol ; 12(10): 867-75, 2016 10.
Article in English | MEDLINE | ID: mdl-27571477

ABSTRACT

There is an urgent need for new strategies to treat invasive fungal infections, which are a leading cause of human mortality. Here, we establish two activities of the natural product beauvericin, which potentiates the activity of the most widely deployed class of antifungal against the leading human fungal pathogens, blocks the emergence of drug resistance, and renders antifungal-resistant pathogens responsive to treatment in mammalian infection models. Harnessing genome sequencing of beauvericin-resistant mutants, affinity purification of a biotinylated beauvericin analog, and biochemical and genetic assays reveals that beauvericin blocks multidrug efflux and inhibits the global regulator TORC1 kinase, thereby activating the protein kinase CK2 and inhibiting the molecular chaperone Hsp90. Substitutions in the multidrug transporter Pdr5 that enable beauvericin efflux impair antifungal efflux, thereby impeding resistance to the drug combination. Thus, dual targeting of multidrug efflux and TOR signaling provides a powerful, broadly effective therapeutic strategy for treating fungal infectious disease that evades resistance.


Subject(s)
Antifungal Agents/pharmacology , Depsipeptides/pharmacology , Fungi/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Antifungal Agents/chemistry , Depsipeptides/chemical synthesis , Depsipeptides/chemistry , Drug Resistance, Fungal/drug effects , Drug Resistance, Multiple/drug effects , Fungi/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Microbial Sensitivity Tests , Mycoses/drug therapy , Mycoses/microbiology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry , TOR Serine-Threonine Kinases/metabolism
10.
Bioorg Med Chem ; 26(1): 232-244, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29198609

ABSTRACT

Benzyloxyphenyl moiety is a common structure of highly potent, selective and reversible inhibitors of monoamine oxidase B (MAO-B), safinamide and sembragiline. We synthesized 4-(benzyloxy)phenyl and biphenyl-4-yl derivatives including halogen substituents on the terminal aryl unit. In addition, we modified the carbon linker between amine group and the biaryl linked unit. Among synthesized compounds, 12c exhibited the most potent and selective MAO-B inhibitory effect (hMAO-B IC50: 8.9 nM; >10,000-fold selectivity over MAO-A) as a competitive inhibitor. In addition, 12c showed greater MAO-B inhibitory activity and selectivity compared to well-known MAO-B inhibitors such as selegiline, safinamide and sembragiline. In the MPTP-induced mouse model of Parkinson's disease (PD), 12c significantly protected the tyrosine hydroxylase (TH)-immunopositive DAergic neurons and attenuated the PD-associated behavioral deficits. This study suggests characteristic structures as a MAO-B inhibitor that may provide a good insight for the development of therapeutic agents for PD.


Subject(s)
Benzene Derivatives/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase/metabolism , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Animals , Benzene Derivatives/chemical synthesis , Benzene Derivatives/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Parkinson Disease/metabolism , Parkinson Disease/pathology , Structure-Activity Relationship
11.
J Struct Biol ; 200(2): 118-123, 2017 11.
Article in English | MEDLINE | ID: mdl-28919350

ABSTRACT

The fungal pathogen Cryptococcus neoformans is a causative agent of meningoencephalitis in humans. For its pathogenicity, the inositol polyphosphate biosynthetic pathway plays critical roles. Recently, Ipk1 from C. neoformans (CnIpk1) was identified as an inositol 1,3,4,5,6-pentakisphosphate 2-kinase that catalyzes the phosphorylation of IP5 to form IP6, a substrate for subsequent reaction to produce inositol pyrophosphates, such as PP-IP5/IP7. Furthermore, it was shown that deletion of IPK1 significantly reduces the virulence of C. neoformans, indicating that Ipk1 is a major virulence contributor. In this study, we determined a crystal structure of the apo-form of CnIpk1 at 2.35Å resolution, the first structure for a fungal Ipk1, using a single-wavelength anomalous dispersion method. Even with a low sequence similarity of 26-28%, its overall structure resembles two other Ipk1 orthologs from Arabidopsis thaliana (AtIpk1) and Mus musculus (MmIpk1), and the most crucial residues in the active site are conserved. Unlike AtIpk1 and MmIpk1, however, metal-binding sites for structural stabilization and conformational variations are absent in CnIpk1. The binding environments for substrate IP5 could be inferred by the two different binding sites for sulfate ion in CnIpk1. Taken together, these observations suggest structural similarities and discrepancies for fungal Ipk1 among members of the Ipk1 family and provide structural information for the possible development of drug design for treatment of cryptococcosis.


Subject(s)
Catalytic Domain/genetics , Cryptococcus neoformans/enzymology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Arabidopsis/enzymology , Binding Sites/genetics , Cryptococcosis/drug therapy , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Crystallography, X-Ray , Drug Design , Mice , Protein Structure, Secondary
12.
PLoS Genet ; 10(4): e1004261, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743168

ABSTRACT

Cryptococcus neoformans is a pathogenic basidiomycetous yeast responsible for more than 600,000 deaths each year. It occurs as two serotypes (A and D) representing two varieties (i.e. grubii and neoformans, respectively). Here, we sequenced the genome and performed an RNA-Seq-based analysis of the C. neoformans var. grubii transcriptome structure. We determined the chromosomal locations, analyzed the sequence/structural features of the centromeres, and identified origins of replication. The genome was annotated based on automated and manual curation. More than 40,000 introns populating more than 99% of the expressed genes were identified. Although most of these introns are located in the coding DNA sequences (CDS), over 2,000 introns in the untranslated regions (UTRs) were also identified. Poly(A)-containing reads were employed to locate the polyadenylation sites of more than 80% of the genes. Examination of the sequences around these sites revealed a new poly(A)-site-associated motif (AUGHAH). In addition, 1,197 miscRNAs were identified. These miscRNAs can be spliced and/or polyadenylated, but do not appear to have obvious coding capacities. Finally, this genome sequence enabled a comparative analysis of strain H99 variants obtained after laboratory passage. The spectrum of mutations identified provides insights into the genetics underlying the micro-evolution of a laboratory strain, and identifies mutations involved in stress responses, mating efficiency, and virulence.


Subject(s)
Cryptococcus neoformans/genetics , Genome, Fungal/genetics , RNA, Fungal/genetics , Transcriptome/genetics , Virulence/genetics , Chromosomes, Fungal/genetics , DNA, Fungal/genetics , Introns/genetics
13.
J Biol Chem ; 290(3): 1861-73, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25477510

ABSTRACT

Cryptococcus neoformans is an encapsulated basidiomycete causing cryptococcosis in immunocompromised humans. The cell surface mannoproteins of C. neoformans were reported to stimulate the host T-cell response and to be involved in fungal pathogenicity; however, their O-glycan structure is uncharacterized. In this study, we performed a detailed structural analysis of the O-glycans attached to cryptococcal mannoproteins using HPLC combined with exoglycosidase treatment and showed that the major C. neoformans O-glycans were short manno-oligosaccharides that were connected mostly by α1,2-linkages but connected by an α1,6-linkage at the third mannose residue. Comparison of the O-glycan profiles from wild-type and uxs1Δ mutant strains strongly supports the presence of minor O-glycans carrying a xylose residue. Further analyses of C. neoformans mutant strains identified three mannosyltransferase genes involved in O-glycan extensions in the Golgi. C. neoformans KTR3, the only homolog of the Saccharomyces cerevisiae KRE2/MNT1 family genes, was shown to encode an α1,2-mannosyltransferase responsible for the addition of the second mannose residue via an α1,2-linkage to the major O-glycans. C. neoformans HOC1 and HOC3, homologs of the Saccharomyces cerevisiae OCH1 family genes, were shown to encode α1,6-mannosyltransferases that can transfer the third mannose residue, via an α1,6-linkage, to minor O-glycans containing xylose and to major O-glycans without xylose, respectively. Moreover, the C. neoformans ktr3Δ mutant strain, which displayed increased sensitivity to SDS, high salt, and high temperature, showed attenuated virulence in a mouse model of cryptococcosis, suggesting that the extended structure of O-glycans is required for cell integrity and full pathogenicity of C. neoformans.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans/metabolism , Mannosyltransferases/metabolism , Polysaccharides/biosynthesis , Animals , Cell Wall/metabolism , Chromatography, High Pressure Liquid , Cryptococcosis/metabolism , Female , Fungal Proteins/metabolism , Genetic Complementation Test , Glycosylation , Glycosyltransferases/metabolism , Mannose/chemistry , Mannosyltransferases/genetics , Melanins/chemistry , Mice , Mutation , Phenotype , Phylogeny , Plasmids/metabolism , Protein Conformation , Xylose/chemistry
15.
Eukaryot Cell ; 13(6): 796-812, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24728196

ABSTRACT

Protein tyrosine phosphatases (PTPs) serve as key negative-feedback regulators of mitogen-activated protein kinase (MAPK) signaling cascades. However, their roles and regulatory mechanisms in human fungal pathogens remain elusive. In this study, we characterized the functions of two PTPs, Ptp1 and Ptp2, in Cryptococcus neoformans, which causes fatal meningoencephalitis. PTP1 and PTP2 were found to be stress-inducible genes, which were controlled by the MAPK Hog1 and the transcription factor Atf1. Ptp2 suppressed the hyperphosphorylation of Hog1 and was involved in mediating vegetative growth, sexual differentiation, stress responses, antifungal drug resistance, and virulence factor regulation through the negative-feedback loop of the HOG pathway. In contrast, Ptp1 was not essential for Hog1 regulation, despite its Hog1-dependent induction. However, in the absence of Ptp2, Ptp1 served as a complementary PTP to control some stress responses. In differentiation, Ptp1 acted as a negative regulator, but in a Hog1- and Cpk1-independent manner. Additionally, Ptp1 and Ptp2 localized to the cytosol but were enriched in the nucleus during the stress response, affecting the transient nuclear localization of Hog1. Finally, Ptp1 and Ptp2 played minor and major roles, respectively, in the virulence of C. neoformans. Taken together, our data suggested that PTPs could be exploited as novel antifungal targets.


Subject(s)
Cryptococcus neoformans/enzymology , Fungal Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Virulence/genetics , Active Transport, Cell Nucleus , Animals , Base Sequence , Cell Nucleus/metabolism , Cryptococcus neoformans/genetics , Cryptococcus neoformans/growth & development , Cryptococcus neoformans/pathogenicity , Female , Fungal Proteins/chemistry , Fungal Proteins/genetics , Genes, Fungal , MAP Kinase Signaling System , Mice , Molecular Sequence Data , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Stress, Physiological , Transcription Factors/metabolism
16.
Mol Microbiol ; 90(3): 630-648, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23998805

ABSTRACT

Mechanisms of oxidative stress resistance are crucial virulence factors for survival and proliferation of fungal pathogens within the human host. In this study we have identified and functionally characterized the role of sulphiredoxin, Srx1, in oxidative stress resistance of Cryptococcus neoformans causing fungal meningoencephalitis and regulation of peroxiredoxins, Tsa1 and Tsa3, and thioredoxins, Trx1 and Trx2. The C. neoformans HOG (High Osmolarity Glycerol response) pathway was essential for the transcriptional regulation of SRX1 under peroxide stress conditions. A gene deletion study revealed that Srx1 was required for cells to counteract peroxide stress, but not other oxidative damaging agents. HOG1 was found to be essential for the induction of adaptive response to peroxide stress with concurrent repression of ergosterol biosynthesis in an SRX1-independent manner. Consistent with this, phosphorylation of C. neoformans Hog1 was modulated by both low and high doses of exogenous hydrogen peroxide treatment. Immunoblot analysis using the C. neoformans Tsa1 specific antibody revealed that both Srx1 and Trx1 were essential for recycling of oxidized Tsa1. In addition to its role in peroxide sensing and response C. neoformans Srx1 was also found to be required for a peroxiredoxin-independent function in promoting fungicide-dependent cell swelling and growth arrest. Finally we showed the importance of C. neoformans Srx1 in fungal pathogenesis by demonstrating its requirement for full virulence using a mouse infection model.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Cryptococcus neoformans/pathogenicity , Ergosterol/biosynthesis , Fungal Proteins/genetics , Fungal Proteins/metabolism , Hydrogen Peroxide/pharmacology , Peroxiredoxins/metabolism , Animals , Cryptococcosis , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/metabolism , Dioxoles/pharmacology , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation, Fungal/drug effects , Genes, Fungal , Humans , Mice , Mice, Inbred CBA , Pyrroles/pharmacology , Signal Transduction/drug effects
17.
Eukaryot Cell ; 12(12): 1564-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24078305

ABSTRACT

Sensing, responding, and adapting to the surrounding environment are crucial for all living organisms to survive, proliferate, and differentiate in their biological niches. This ability is also essential for Cryptococcus neoformans and its sibling species Cryptococcus gattii, as these pathogens have saprobic and parasitic life cycles in natural and animal host environments. The ability of Cryptococcus to cause fatal meningoencephalitis is highly related to its capability to remodel and optimize its metabolic and physiological status according to external cues. These cues act through multiple stress signaling pathways through a panoply of signaling components, including receptors/sensors, small GTPases, secondary messengers, kinases, transcription factors, and other miscellaneous adaptors or regulators. In this minireview, we summarize and highlight the importance of several stress signaling pathways that influence the pathogenicity of Cryptococcus and discuss future challenges in these areas.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus/metabolism , Cryptococcus/pathogenicity , Signal Transduction , Animals , Humans , Virulence
18.
J Sci Food Agric ; 94(3): 445-52, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23765595

ABSTRACT

BACKGROUND: Gromwell is known to have diverse pharmacological, cosmetic and nutritional benefits for humans. Nevertheless, the biological influence of gromwell extract (GE) on the general physiology of eukaryotic cells remains unknown. In this study a global transcriptome analysis was performed to identify genes affected by the addition of GE with Cryptococcus neoformans as the model system. RESULTS: In response to GE treatment, genes involved in signal transduction were immediately regulated, and the evolutionarily conserved sets of genes involved in the core cellular functions, including DNA replication, RNA transcription/processing and protein translation/processing, were generally up-regulated. In contrast, a number of genes involved in carbohydrate metabolism and transport, inorganic ion transport and metabolism, post-translational modification/protein turnover/chaperone functions and signal transduction were down-regulated. Among the GE-responsive genes that are also evolutionarily conserved in the human genome, the expression patterns of YSA1, TPO2, CFO1 and PZF1 were confirmed by northern blot analysis. Based on the functional characterization of some GE-responsive genes, it was found that GE treatment may promote cellular tolerance against a variety of environmental stresses in eukaryotes. CONCLUSIONS: GE treatment affects the expression levels of a significant portion of the Cryptococcus genome, implying that GE significantly affects the general physiology of eukaryotic cells.


Subject(s)
Adaptation, Physiological/genetics , Cryptococcus/drug effects , Eukaryotic Cells/drug effects , Lithospermum , Plant Extracts/pharmacology , Stress, Physiological/genetics , Transcriptome/drug effects , Biological Transport/drug effects , Biological Transport/genetics , Carbohydrate Metabolism/drug effects , Carbohydrate Metabolism/genetics , Cryptococcus/cytology , Cryptococcus/genetics , DNA Replication/drug effects , DNA Replication/genetics , Eukaryotic Cells/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genome , Oligonucleotide Array Sequence Analysis , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Protein Processing, Post-Translational/drug effects , Signal Transduction
19.
mSphere ; 9(1): e0055723, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38085018

ABSTRACT

The N6-threonylcarbamoyl adenosine (t6A) tRNA modification is critical for ensuring translation fidelity across three domains of life. Our prior work highlighted the KEOPS complex, organized in a Pcc1-Kae1-Bud32-Cgi121 linear arrangement, not only serves an evolutionarily conserved role in t6A tRNA modification but also exerts diverse functional impacts on pathobiological characteristics in Cryptococcus neoformans, a leading cause of fungal meningitis worldwide. However, the extent to which the pleiotropic functions of the KEOPS complex are specifically tied to tRNA modification remains uncertain. To address this, we undertook a functional characterization of Sua5, responsible for generating the precursor threonylcarbamoyl-adenylate (TC-AMP) for t6A tRNA modification, using a reverse genetics approach. Comparative phenotypic analyses with KEOPS mutants revealed that Sua5 plays a vital role in multiple cellular processes, such as t6A tRNA modification, growth, sexual development, stress response, and virulence factor production, thus reflecting the multifaceted functions of the KEOPS complex. In support of this, sua5Δ bud32Δ double mutants showed phenotypes comparable to those of the corresponding single mutants. Intriguingly, a SUA5 allele lacking a mitochondria targeting sequence (SUA5MTSΔ) was sufficient to restore the wild-type phenotypes in the sua5Δ mutant, suggesting that Sua5's primary functional locus may be cytosolic, akin to the KEOPS complex. Further supporting this, the deletion of Qri7, a mitochondrial paralog of Kae1, had no discernible phenotypic impact on C. neoformans. We concluded that cytosolic t6A tRNA modifications, orchestrated by Sua5 and the KEOPS complex, are central to the regulation of diverse pathobiological functions in C. neoformans.IMPORTANCEUnderstanding cellular functions at the molecular level is crucial for advancing disease treatments. Our research reveals a critical connection between the KEOPS complex and Sua5 in Cryptococcus neoformans, a significant cause of fungal meningitis. While the KEOPS complex is known for its versatile roles in cellular processes, Sua5 is specialized in t6A tRNA modification. Our key finding is that the diverse roles of the KEOPS complex, ranging from cell growth and stress response to virulence, are fundamentally linked to its function in t6A tRNA modification. This conclusion is supported by the remarkable similarities between the impacts of Sua5 and KEOPS on these processes, despite their roles in different steps of the t6A modification pathway. This newfound understanding deepens our insight into fungal biology and opens new avenues for developing potential therapies against dangerous fungal diseases.


Subject(s)
Cryptococcus neoformans , Meningitis, Fungal , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Adenosine/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism
20.
J Microbiol Biotechnol ; 34(7): 1-11, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38881183

ABSTRACT

The rise of Candida auris, a multidrug-resistant fungal pathogen, across more than 40 countries, has signaled an alarming threat to global health due to its significant resistance to existing antifungal therapies. Characterized by its rapid spread and robust drug resistance, C. auris presents a critical challenge in managing infections, particularly in healthcare settings. With research on its biological traits and genetic basis of virulence and resistance still in the early stages, there is a pressing need for a concerted effort to understand and counteract this pathogen. This review synthesizes current knowledge on the epidemiology, biology, genetic manipulation, pathogenicity, diagnostics, and resistance mechanisms of C. auris, and discusses future directions in research and therapeutic development. By exploring the complexities surrounding C. auris, we aim to underscore the importance of advancing research to devise effective control and treatment strategies.

SELECTION OF CITATIONS
SEARCH DETAIL