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










Publication year range
1.
Microbiol Spectr ; : e0341923, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842336

ABSTRACT

Cryptococcus neoformans is a fungal pathogen responsible for >200,000 yearly cases with a mortality as high as 81%. This burden results, in part, from an incomplete understanding of its pathogenesis and ineffective antifungal treatments; hence, there is a pressing need to understand the biology and host interactions of this yeast to develop improved treatments. Protein palmitoylation is important for cryptococcal virulence, and we previously identified the substrates of its main palmitoyl transferase. One of them was encoded by the uncharacterized gene CNAG_02129. In the filamentous fungus Neurospora crassa, a homolog of this gene named hyphal anastomosis protein 13 plays a role in proper cellular communication and filament fusion. In Cryptococcus, cellular communication is essential during mating; therefore, we hypothesized that CNAG_02129, which we named hyphal anastomosis protein 1 (HAM1), may play a role in mating. We found that ham1Δ mutants produce more fusion products during mating, filament more robustly, and exhibit competitive fitness defects under mating and non-mating conditions. Additionally, we found several differences with the major virulence factor, the polysaccharide capsule, that may affect virulence, consistent with prior studies linking virulence to mating. We observed that ham1Δ mutants have decreased capsule attachment and transfer but exhibit higher amounts of exopolysaccharide shedding and biofilm production. Finally, HAM1 expression is significantly lower in mating media relative to non-mating conditions, consistent with it acting as a negative regulator of mating. Understanding the connection between mating and virulence in C. neoformans may open new avenues of investigation into ways to improve the treatment of this disease. IMPORTANCE: Fungal mating is a vital part of the lifecycle of the pathogenic yeast Cryptococcus neoformans. More than just ensuring the propagation of the species, mating allows for sexual reproduction to occur and generates genetic diversity as well as infectious propagules that can invade mammalian hosts. Despite its importance in the biology of this pathogen, we still do not know all of the major players regulating the mating process and if they are involved or impact its pathogenesis. Here, we identified a novel negative regulator of mating that also affects certain cellular characteristics known to be important for virulence. This gene, which we call HAM1, is widely conserved across the cryptococcal family as well as in many pathogenic fungal species. This study will open new avenues of exploration regarding the function of uncharacterized but conserved genes in a variety of pathogenic fungal species and specifically in serotype A of C. neoformans.

2.
bioRxiv ; 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38585947

ABSTRACT

Cryptococcus neoformans is a fungal pathogen responsible for >200,000 yearly cases with a mortality as high as 81%. This burden results, in part, from an incomplete understanding of its pathogenesis and ineffective antifungal treatments; hence, there is a pressing need to understand the biology and host interactions of this yeast to develop improved treatments. Protein palmitoylation is important for cryptococcal virulence, and we previously identified the substrates of its main palmitoyl transferase. One of them was encoded by the uncharacterized gene CNAG_02129. In the filamentous fungus Neurospora crassa, a homolog of this gene named HAM-13 plays a role in proper cellular communication and filament fusion. In Cryptococcus, cellular communication is essential during mating, therefore we hypothesized that CNAG_02129, which we named HAM1, may play a role in mating. We found that ham1Δ mutants produce more fusion products during mating, filament more robustly, and exhibit competitive fitness defects under mating and non-mating conditions. Additionally, we found several differences with the major virulence factor, the polysaccharide capsule, that may affect virulence, consistent with prior studies linking virulence to mating. We observed that ham1Δ mutants have decreased capsule attachment and transfer but exhibit higher amounts of exopolysaccharide shedding and biofilm production. Lastly, HAM1 expression is significantly lower in mating media relative to non-mating conditions, consistent with it acting as a negative regulator of mating. Understanding the connection between mating and virulence in C. neoformans may open new avenues of investigation into ways to improve the treatment of this disease.

3.
mSphere ; 9(4): e0064323, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38470131

ABSTRACT

Although fungi have been important model organisms for solving genetic, molecular, and ecological problems, recently, they are also becoming an important source of infectious disease. Despite their high medical burden, fungal pathogens are understudied, and relative to other pathogenic microbes, less is known about how their gene functions contribute to disease. This is due, in part, to a lack of powerful genetic tools to study these organisms. In turn, this has resulted in inappropriate treatments and diagnostics and poor disease management. There are a variety of reasons genetic studies were challenging in pathogenic fungi, but in recent years, most of them have been overcome or advances have been made to circumvent these barriers. In this minireview, we highlight how recent advances in genetic studies in fungal pathogens have resulted in the discovery of important biology and potential new antifungals and have created the tools to comprehensively study these important pathogens.


Subject(s)
Fungi , Mycoses , Fungi/genetics , Fungi/classification , Fungi/pathogenicity , Mycoses/microbiology , Genetic Techniques , Humans , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use
4.
Infect Immun ; 91(9): e0043422, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37506189

ABSTRACT

Fungal infections represent a major, albeit neglected, public health threat with serious medical and economic burdens globally. With unacceptably high mortality rates, invasive fungal pathogens are responsible for millions of deaths each year, with a steadily increasing incidence primarily in immunocompromised individuals. The poor therapeutic options and rise of antifungal drug resistance pose further challenges in controlling these infections. These fungal pathogens have adapted to survive within mammalian hosts and can establish intracellular niches to promote survival within host immune cells. To do that, they have developed diverse methods to circumvent the innate immune system attack. This includes strategies such as altering their morphology, counteracting macrophage antimicrobial action, and metabolic adaptation. This is reminiscent of how bacterial pathogens have adapted to survive within host cells and cause disease. However, relative to the great deal of information available concerning intracellular bacterial pathogenesis, less is known about the mechanisms fungal pathogens employ. Therefore, here we review our current knowledge and recent advances in our understanding of how fungi can evade and persist within host immune cells. This review will focus on the major fungal pathogens, including Cryptococcus neoformans, Candida albicans, and Aspergillus fumigatus, among others. As we discover and understand the strategies used by these fungi, similarities with their bacterial counterparts are becoming apparent, hence we can use the abundant information from bacteria to guide our studies in fungi. By understanding these strategies, new lines of research will open that can improve the treatments of these devastating fungal diseases.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Mycoses , Animals , Humans , Mycoses/microbiology , Candida albicans/metabolism , Aspergillus fumigatus , Cryptococcosis/microbiology , Mammals
5.
mBio ; 14(1): e0338422, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36749043

ABSTRACT

The fungal pathogen Cryptococcus neoformans is distinguished by a cell-wall-anchored polysaccharide capsule that is critical for virulence. Biogenesis of both cell wall and capsule relies on the secretory pathway. Protein secretion begins with polypeptide translocation across the endoplasmic reticulum (ER) membrane through a highly conserved channel formed by three proteins: Sec61, Sbh1, and Sss1. Sbh1, the most divergent, contains multiple phosphorylation sites, which may allow it to regulate entry into the secretory pathway in a species- and protein-specific manner. Absence of SBH1 causes a cell-wall defect in both Saccharomyces cerevisiae and C. neoformans, although other phenotypes differ. Notably, proteomic analysis showed that when cryptococci are grown in conditions that mimic aspects of the mammalian host environment (tissue culture medium, 37°C, 5% CO2), a set of secretory and transmembrane proteins is upregulated in wild-type, but not in Δsbh1 mutant cells. The Sbh1-dependent proteins show specific features of their ER targeting sequences that likely cause them to transit less efficiently into the secretory pathway. Many also act in cell-wall biogenesis, while several are known virulence factors. Consistent with these observations, the C. neoformans Δsbh1 mutant is avirulent in a mouse infection model. We conclude that, in the context of conditions encountered during infection, Sbh1 controls the entry of virulence factors into the secretory pathway of C. neoformans, and thereby regulates fungal pathogenicity. IMPORTANCE Cryptococcus neoformans is a yeast that causes almost 200,000 deaths worldwide each year, mainly of immunocompromised individuals. The surface structures of this pathogen, a protective cell wall surrounded by a polysaccharide capsule, are made and maintained by proteins that are synthesized inside the cell and travel outwards through the secretory pathway. A protein called Sbh1 is part of the machinery that determines which polypeptides enter this export pathway. We found that when Sbh1 is absent, both C. neoformans and the model yeast S. cerevisiae show cell-wall defects. Lack of Sbh1 also changes the pattern of secretion of both transmembrane and soluble proteins, in a manner that depends on characteristics of their sequences. Notably, multiple proteins that are normally upregulated in conditions similar to those encountered during infection, including several needed for cryptococcal virulence, are no longer increased. Sbh1 thereby regulates the ability of this important pathogen to cause disease.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Saccharomyces cerevisiae Proteins , Animals , Mice , Cryptococcosis/microbiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mammals/metabolism , Polysaccharides/metabolism , Protein Transport , Proteomics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , SEC Translocation Channels/genetics , Translocation, Genetic , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Endoplasmic Reticulum/metabolism
6.
Front Cell Infect Microbiol ; 12: 967486, 2022.
Article in English | MEDLINE | ID: mdl-36211949

ABSTRACT

Understanding of how intracellular pathogens survive in their host cells is important to improve management of their diseases. This has been fruitful for intracellular bacteria, but it is an understudied area in fungal pathogens. Here we start elucidating and characterizing the strategies used by one of the commonest fungal pathogens, Cryptococcus neoformans, to survive intracellularly. The ability of the fungus to survive inside host cells is one of the main drivers of disease progression, yet it is unclear whether C. neoformans resides in a fully acidified, partially acidic, or neutral phagosome. Using a dye that only fluoresce under acidic conditions to stain C. neoformans, a hypha-defective Candida albicans mutant, and the nonpathogenic Saccharomyces cerevisiae, we characterized the fungal behaviors in infected macrophages by live microscopy. The main behavior in the C. albicans mutant strain and S. cerevisiae-phagosomes was rapid acidification after internalization, which remained for the duration of the imaging. In contrast, a significant number of C. neoformans-phagosomes exhibited alternative behaviors distinct from the normal phagosomal maturation: some phagosomes acidified with subsequent loss of acidification, and other phagosomes never acidified. Moreover, the frequency of these behaviors was affected by the immune status of the host cell. We applied the same technique to a flow cytometry analysis and found that a substantial percentage of C. neoformans-phagosomes showed impaired acidification, whereas almost 100% of the S. cerevisiae-phagosomes acidify. Lastly, using a membrane-damage reporter, we show phagosome permeabilization correlates with acidification alterations, but it is not the only strategy that C. neoformans uses to manipulate phagosomal acidification. The different behaviors described here provide an explanation to the confounding literature regarding cryptococcal-phagosome acidification and the methods can be applied to study other intracellular fungal pathogens.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Candida albicans , Cryptococcosis/microbiology , Hydrogen-Ion Concentration , Phagosomes/microbiology , Saccharomyces cerevisiae/genetics
7.
mBio ; 13(4): e0153922, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35726920

ABSTRACT

ATP-binding cassette (ABC) transporters represent one of the largest protein superfamilies. Functionally diverse, ABC transporters have been implicated in many aspects of microbial physiology. The genome of the human fungal pathogen Cryptococcus neoformans encodes 54 putative ABC transporters and most of them remain uncharacterized. In a previous genetic screen for fungal regulators of phagocytosis, we identified an uncharacterized gene, CNAG_06909, that modulates host interactions. This gene encoded a half-size ABC transporter of the PDR-type, and phenotypic studies of a strain with this gene deleted revealed an altered antifungal susceptibility profile, including hypersensitivity to fluconazole (FLC). This gene, which we named PDR6, localized to the endoplasmic reticulum (ER) and plasma membrane (PM), and when absent, less ergosterol was observed in the PM. Additionally, we observed that the pdr6Δ strain displayed a reduction in secreted polysaccharide capsular material. These changes to the cellular surface may explain the observed increased uptake by macrophages and the reduced intracellular survival. Finally, studies in mice demonstrated that Pdr6 function was required for the normal progression of cryptococcal infection. Taken together, this study demonstrates a novel dual role for PDR transporters in C. neoformans, which could represent a potential target for antifungal therapeutics. Furthermore, the atypical half-size transporter encoded by PDR6 is conserved in many fungal pathogens, but absent in model nonpathogenic fungi. Hence, this study provided a function for this unique group of fungal half-size PDR transporters that, although conserved, remain largely understudied. IMPORTANCE Conserved across all kingdoms of life, ABC transporters comprise one of the largest protein families. They are associated with multidrug resistance, affecting aspects such as resistance to antimicrobials or anti-cancer drugs. Despite their importance, they are understudied in fungal pathogens. In the environmental fungus Cryptococcus neoformans, a leading cause of fungal infections, only a few ABC transporters have been studied. Here, we characterized an atypical, half-size, ABC transporter of the PDR-type, that affected both antifungal resistance and host-pathogen interactions. PDR-type transporters are only present in fungi and plants, and this subgroup of half-size transporters was conserved in fungal pathogens, yet their function was completely unknown. Because the current treatments for cryptococcal infection are suboptimal, understanding the mechanisms of antifungal resistance and the host interactions that drive the infection is critical to improving the management of this disease. Here, we provide insights into these important aspects of cryptococcal pathogenesis.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Mycoses , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Cryptococcosis/microbiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Drug Resistance, Fungal/genetics , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mycoses/microbiology
8.
mSphere ; 4(5)2019 10 02.
Article in English | MEDLINE | ID: mdl-31578250

ABSTRACT

Felipe Santiago-Tirado studies the cell biology of cryptococcal infections. In this mSphere of Influence article, he reflects on how the papers "Systematic Genetic Analysis of Virulence in the Human Fungal Pathogen Cryptococcus neoformans" (https://doi.org/10.1016/j.cell.2008.07.046) and "Unraveling the Biology of a Fungal Meningitis Pathogen Using Chemical Genetics" (https://doi.org/10.1016/j.cell.2014.10.044) by the Noble and Madhani groups influenced his thinking by showcasing the various modern applications of yeast genetics in an organism where genetic manipulation was difficult.


Subject(s)
Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Virulence Factors/genetics , Fungal Proteins/genetics , Gene Deletion , Humans , Virulence
9.
Curr Protoc Microbiol ; 53(1): e78, 2019 06.
Article in English | MEDLINE | ID: mdl-30776307

ABSTRACT

Cryptococcus neoformans is an environmental yeast found worldwide that causes lethal brain infections, particularly in immunocompromised hosts. In 2016, there were 280,000 cases of cryptococcal meningitis in the HIV+ population, two-thirds of them fatal; other immunocompromised patients are also affected. The burden of cryptococcal disease and the limits of current chemotherapy create a pressing need for improved treatment. One hindrance to the development of new therapies is lack of understanding of how this pathogen breaches the barriers protecting the brain. Here we describe a tool for investigating this process. This simple in vitro blood-brain-barrier (BBB) model, based on a human brain endothelial cell line grown on a permeable membrane, may be used to assay the BBB transmigration of C. neoformans or other neurotropic pathogens. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Brain/microbiology , Cell Culture Techniques/methods , Cryptococcosis/microbiology , Cryptococcus neoformans/physiology , Endothelial Cells/microbiology , Blood-Brain Barrier/microbiology , Brain/cytology , Cell Line , Central Nervous System/microbiology , Humans , Models, Biological
11.
mBio ; 8(1)2017 01 31.
Article in English | MEDLINE | ID: mdl-28143979

ABSTRACT

The blood-brain barrier (BBB) protects the central nervous system (CNS) by restricting the passage of molecules and microorganisms. Despite this barrier, however, the fungal pathogen Cryptococcus neoformans invades the brain, causing a meningoencephalitis that is estimated to kill over 600,000 people annually. Cryptococcal infection begins in the lung, and experimental evidence suggests that host phagocytes play a role in subsequent dissemination, although this role remains ill defined. Additionally, the disparate experimental approaches that have been used to probe various potential routes of BBB transit make it impossible to assess their relative contributions, confounding any integrated understanding of cryptococcal brain entry. Here we used an in vitro model BBB to show that a "Trojan horse" mechanism contributes significantly to fungal barrier crossing and that host factors regulate this process independently of free fungal transit. We also, for the first time, directly imaged C. neoformans-containing phagocytes crossing the BBB, showing that they do so via transendothelial pores. Finally, we found that Trojan horse crossing enables CNS entry of fungal mutants that cannot otherwise traverse the BBB, and we demonstrate additional intercellular interactions that may contribute to brain entry. Our work elucidates the mechanism of cryptococcal brain invasion and offers approaches to study other neuropathogens. IMPORTANCE: The fungal pathogen Cryptococcus neoformans invades the brain, causing a meningoencephalitis that kills hundreds of thousands of people each year. One route that has been proposed for this brain entry is a Trojan horse mechanism, whereby the fungus crosses the blood-brain barrier (BBB) as a passenger inside host phagocytes. Although indirect experimental evidence supports this intriguing mechanism, it has never been directly visualized. Here we directly image Trojan horse transit and show that it is regulated independently of free fungal entry, contributes to cryptococcal BBB crossing, and allows mutant fungi that cannot enter alone to invade the brain.


Subject(s)
Blood-Brain Barrier/immunology , Cryptococcus neoformans/pathogenicity , Immune Evasion , Phagocytes/microbiology , Humans , Models, Biological , Models, Theoretical
12.
mBio ; 7(2): e00313-16, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27094327

ABSTRACT

UNLABELLED: Cryptococcus neoformans is a ubiquitous, opportunistic fungal pathogen that kills over 600,000 people annually. Here, we report integrated computational and experimental investigations of the role and mechanisms of transcriptional regulation in cryptococcal infection. Major cryptococcal virulence traits include melanin production and the development of a large polysaccharide capsule upon host entry; shed capsule polysaccharides also impair host defenses. We found that both transcription and translation are required for capsule growth and that Usv101 is a master regulator of pathogenesis, regulating melanin production, capsule growth, and capsule shedding. It does this by directly regulating genes encoding glycoactive enzymes and genes encoding three other transcription factors that are essential for capsule growth: GAT201, RIM101, and SP1. Murine infection with cryptococci lacking Usv101 significantly alters the kinetics and pathogenesis of disease, with extended survival and, unexpectedly, death by pneumonia rather than meningitis. Our approaches and findings will inform studies of other pathogenic microbes. IMPORTANCE: Cryptococcus neoformans causes fatal meningitis in immunocompromised individuals, mainly HIV positive, killing over 600,000 each year. A unique feature of this yeast, which makes it particularly virulent, is its polysaccharide capsule; this structure impedes host efforts to combat infection. Capsule size and structure respond to environmental conditions, such as those encountered in an infected host. We have combined computational and experimental tools to elucidate capsule regulation, which we show primarily occurs at the transcriptional level. We also demonstrate that loss of a novel transcription factor alters virulence factor expression and host cell interactions, changing the lethal condition from meningitis to pneumonia with an exacerbated host response. We further demonstrate the relevant targets of regulation and kinetically map key regulatory and host interactions. Our work elucidates mechanisms of capsule regulation, provides methods and resources to the research community, and demonstrates an altered pathogenic outcome that resembles some human conditions.


Subject(s)
Cryptococcosis/microbiology , Cryptococcus neoformans/pathogenicity , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Transcription Factors/metabolism , Animals , Computational Biology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Female , Fungal Proteins/genetics , Gene Regulatory Networks , Humans , Melanins/metabolism , Mice , Transcription Factors/genetics , Virulence
13.
J Microbiol ; 54(3): 212-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26920881

ABSTRACT

Lipid modification of proteins is a widespread, essential process whereby fatty acids, cholesterol, isoprenoids, phospholipids, or glycosylphospholipids are attached to polypeptides. These hydrophobic groups may affect protein structure, function, localization, and/or stability; as a consequence such modifications play critical regulatory roles in cellular systems. Recent advances in chemical biology and proteomics have allowed the profiling of modified proteins, enabling dissection of the functional consequences of lipid addition. The enzymes that mediate lipid modification are specific for both the lipid and protein substrates, and are conserved from fungi to humans. In this article we review these enzymes, their substrates, and the processes involved in eukaryotic lipid modification of proteins. We further focus on its occurrence in the fungal pathogen Cryptococcus neoformans, highlighting unique features that are both relevant for the biology of the organism and potentially important in the search for new therapies.


Subject(s)
Cryptococcus neoformans/metabolism , Fungal Proteins/metabolism , Lipid Metabolism , Acylation , Cryptococcosis/microbiology , Cryptococcosis/therapy , Cryptococcus neoformans/enzymology , Cryptococcus neoformans/pathogenicity , Fungal Proteins/chemistry , Humans , Lipoylation , Models, Molecular , Prenylation , Protein Processing, Post-Translational , Proteomics/methods
14.
PLoS Pathog ; 11(5): e1004908, 2015 May.
Article in English | MEDLINE | ID: mdl-25970403

ABSTRACT

Cryptococcus neoformans is an opportunistic yeast that kills over 625,000 people yearly through lethal meningitis. Host phagocytes serve as the first line of defense against this pathogen, but fungal engulfment and subsequent intracellular proliferation also correlate with poor patient outcome. Defining the interactions of this facultative intracellular pathogen with host phagocytes is key to understanding the latter's opposing roles in infection and how they contribute to fungal latency, dissemination, and virulence. We used high-content imaging and a human monocytic cell line to screen 1,201 fungal mutants for strains with altered host interactions and identified multiple genes that influence fungal adherence and phagocytosis. One of these genes was PFA4, which encodes a protein S-acyl transferase (PAT), one of a family of DHHC domain-containing proteins that catalyzes lipid modification of proteins. Deletion of PFA4 caused dramatic defects in cryptococcal morphology, stress tolerance, and virulence. Bioorthogonal palmitoylome-profiling identified Pfa4-specific protein substrates involved in cell wall synthesis, signal transduction, and membrane trafficking responsible for these phenotypic alterations. We demonstrate that a single PAT is responsible for the modification of a subset of proteins that are critical in cryptococcal pathogenesis. Since several of these palmitoylated substrates are conserved in other pathogenic fungi, protein palmitoylation represents a potential avenue for new antifungal therapeutics.


Subject(s)
Acyltransferases/metabolism , Cryptococcosis/metabolism , Cryptococcus neoformans/physiology , Fungal Proteins/metabolism , Host-Pathogen Interactions , Monocytes/microbiology , Protein Processing, Post-Translational , Acylation , Acyltransferases/genetics , Cell Adhesion , Cell Line , Cell Wall/immunology , Cell Wall/metabolism , Cell Wall/pathology , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/cytology , Cryptococcus neoformans/enzymology , Cryptococcus neoformans/pathogenicity , Fungal Proteins/genetics , Gene Deletion , Humans , Meningitis, Cryptococcal/immunology , Meningitis, Cryptococcal/metabolism , Meningitis, Cryptococcal/microbiology , Meningitis, Cryptococcal/pathology , Microbial Viability , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Mutation , Phagocytosis , Signal Transduction , Stress, Physiological , Substrate Specificity , Virulence , Virus Latency
15.
Eukaryot Cell ; 13(5): 560-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24585882

ABSTRACT

The cryptococcal capsule is a critical virulence factor of an important pathogen, but little is known about how it is associated with the cell or released into the environment. Two mutants lacking PBX1 and PBX2 were found to shed reduced amounts of the capsule polysaccharide glucuronoxylomannan (GXM). Nuclear magnetic resonance, composition, and physical analyses showed that the shed material was of normal mass but was slightly enriched in xylose. In contrast to previous reports, this material contained no glucose. Notably, the capsule fibers of pbxΔ mutant cells grown under capsule-inducing conditions were present at a lower than usual density and were loosely attached to the cell wall. Mutant cell walls were also defective, as indicated by phenotypes including abnormal cell morphology, reduced mating filamentation, and altered cell integrity. All observed phenotypes were shared between the two mutants and exacerbated in a double mutant. Consistent with a role in surface glycan synthesis, the Pbx proteins localized to detergent-resistant membrane domains. These results, together with the sequence motifs in the Pbx proteins, suggest that Pbx1 and Pbx2 are redundant proteins that act in remodeling the cell wall to maintain normal cell morphology and precursor availability for other glycan synthetic processes. Their absence results in aberrant cell wall growth and metabolic imbalance, which together impact cell wall and capsule synthesis, cell morphology, and capsule association. The surface changes also lead to increased engulfment by host phagocytes, consistent with the lack of virulence of pbx mutants in animal models.


Subject(s)
Cell Wall/metabolism , Cryptococcus neoformans/metabolism , Fungal Capsules/metabolism , Fungal Proteins/metabolism , Polysaccharides/biosynthesis , Carbohydrate Sequence , Cell Wall/chemistry , Cell Wall/genetics , Cryptococcosis/microbiology , Cryptococcus neoformans/chemistry , Cryptococcus neoformans/genetics , Fungal Capsules/chemistry , Fungal Capsules/genetics , Fungal Proteins/genetics , Humans , Molecular Sequence Data , Polysaccharides/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism
16.
Yeast ; 31(2): 47-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24375706

ABSTRACT

The importance of the Basidiomycete Cryptococcus neoformans to human health has stimulated its development as an experimental model for both basic physiology and pathogenesis. We briefly review the history of this fascinating and versatile fungus, some notable aspects of its biology that contribute to virulence, and current tools available for its study.


Subject(s)
Cryptococcosis/history , Cryptococcosis/microbiology , Cryptococcus neoformans/physiology , Microbiology/history , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cryptococcosis/epidemiology , Cryptococcus neoformans/genetics , Cryptococcus neoformans/isolation & purification , Cryptococcus neoformans/pathogenicity , History, 19th Century , History, 20th Century , History, 21st Century , Humans
17.
Mol Biol Cell ; 23(3): 412-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22160598

ABSTRACT

Formins are conserved proteins that assemble unbranched actin filaments in a regulated, localized manner. Budding yeast's two formins, Bni1p and Bnr1p, assemble actin cables necessary for polarized cell growth and organelle segregation. Here we define four regions in Bni1p that contribute to its localization to the bud and at the bud neck. The first (residues 1-333) requires dimerization for its localization and encompasses the Rho-binding domain. The second (residues 334-821) covers the Diaphanous inhibitory-dimerization-coiled coil domains, and the third is the Spa2p-binding domain. The fourth region encompasses the formin homology 1-formin homology 2-COOH region of the protein. These four regions can each localize to the bud cortex and bud neck at the right stage of the cell cycle independent of both F-actin and endogenous Bni1p. The first three regions contribute cumulatively to the proper localization of Bni1p, as revealed by the effects of progressive loss of these regions on the actin cytoskeleton and fidelity of spindle orientation. The fourth region contributes to the localization of Bni1p in tiny budded cells. Expression of mislocalized Bni1p constructs has a dominant-negative effect on both growth and nuclear segregation due to mislocalized actin assembly. These results define an unexpected complexity in the mechanism of formin localization and function.


Subject(s)
Actin Cytoskeleton/physiology , Microfilament Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/physiology , Actins/chemistry , Actins/physiology , Binding Sites , Dimerization , Microfilament Proteins/chemistry , Microfilament Proteins/physiology , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/physiology , Spindle Apparatus/physiology , Structure-Activity Relationship
18.
Trends Cell Biol ; 21(9): 515-25, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21764313

ABSTRACT

Cell polarity in eukaryotes requires constant sorting, packaging and transport of membrane-bound cargo within the cell. These processes occur in two sorting hubs: the recycling endosome for incoming material and the trans-Golgi network for outgoing material. Phosphatidylinositol 3-phosphate and phosphatidylinositol 4-phosphate are enriched at the endocytic and exocytic sorting hubs, respectively, where they act together with small GTPases to recruit factors to segregate cargo and regulate carrier formation and transport. In this review, we summarize the current understanding of how these lipids and GTPases regulate membrane trafficking directly, emphasizing the recent discoveries of phosphatidylinositol 4-phosphate functions at the trans-Golgi network.


Subject(s)
GTP Phosphohydrolases/metabolism , Phosphatidylinositol Phosphates/metabolism , trans-Golgi Network/metabolism , Animals , Cell Membrane/metabolism , Cell Polarity , Endocytosis , Endosomes/metabolism , Protein Binding , Protein Transport , Saccharomyces cerevisiae/metabolism , Signal Transduction
19.
Dev Cell ; 20(1): 47-59, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21238924

ABSTRACT

Cell polarity involves transport of specific membranes and macromolecules at the right time to the right place. In budding yeast, secretory vesicles are transported by the myosin-V Myo2p to sites of cell growth. We show that phosphatidylinositol 4-phosphate (PI4P) is present in late secretory compartments and is critical for their association with, and transport by, Myo2p. Further, the trans-Golgi network Rab Ypt31/32p and secretory vesicle Rab Sec4p each bind directly, but distinctly, to Myo2p, and these interactions are also required for secretory compartment transport. Enhancing the interaction of Myo2p with PI4P bypasses the requirement for interaction with Ypt31/32p and Sec4p. Together with additional genetic data, the results indicate that Rab proteins and PI4P collaborate in the association of secretory compartments with Myo2p. Thus, we show that a coincidence detection mechanism coordinates inputs from PI4P and the appropriate Rab for secretory compartment transport.


Subject(s)
Cell Compartmentation , Myosin Type V/metabolism , Phosphatidylinositol Phosphates/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Secretory Vesicles/metabolism , rab GTP-Binding Proteins/metabolism , Alleles , Biological Transport , Cell Polarity , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , Models, Biological , Mutation/genetics , Saccharomyces cerevisiae Proteins/metabolism
20.
Mol Biol Cell ; 17(4): 1812-21, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16467380

ABSTRACT

The tail of the yeast myosin V encoded by Myo2p is known to bind several receptors for cargo delivery along polarized actin cables. However, it is not known how Myo2p activity is regulated or how it selects between cargoes. Here we show that Myo2p is reversibly phosphorylated in vivo. A short peptide at the N-terminal end of the cargo-binding domain contains three residues contributing to single or doubly phosphorylated species. We confirm that the tail consists of two proteolytically resistant subdomains and identify a functionally important region N-terminal to subdomain 1 that includes the phosphorylation sites. Mutagenesis of the phosphorylation sites to alanine abolished a mobility shift diagnostic of phosphorylation, whereas mutagenesis to glutamic acid produced the shift and the formation of an additional phosphorylated species. These substitutions did not affect overall cell growth. However, one of the sites is predicted to be a substrate of cAMP-dependent protein kinase (PKA), and yeast expressing Myo2p with alanine substitutions is resistant to otherwise lethal overexpression of PKA, whereas the glutamic acid mutant is supersensitive to overexpression of PKA. These results suggest that in yeast, Myo2p is subject to phosphoregulation involving a PKA-related signaling pathway.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence/genetics , Amino Acid Substitution , Mutation , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Phenotype , Phosphorylation , Saccharomyces cerevisiae Proteins/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...