Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
Add more filters

Publication year range
1.
Nature ; 588(7839): 688-692, 2020 12.
Article in English | MEDLINE | ID: mdl-33268895

ABSTRACT

Inflammasomes are important sentinels of innate immune defence that are activated in response to diverse stimuli, including pathogen-associated molecular patterns (PAMPs)1. Activation of the inflammasome provides host defence against aspergillosis2,3, which is a major health concern for patients who are immunocompromised. However, the Aspergillus fumigatus PAMPs that are responsible for inflammasome activation are not known. Here we show that the polysaccharide galactosaminogalactan (GAG) of A. fumigatus is a PAMP that activates the NLRP3 inflammasome. The binding of GAG to ribosomal proteins inhibited cellular translation machinery, and thus activated the NLRP3 inflammasome. The galactosamine moiety bound to ribosomal proteins and blocked cellular translation, which triggered activation of the NLRP3 inflammasome. In mice, a GAG-deficient Aspergillus mutant (Δgt4c) did not elicit protective activation of the inflammasome, and this strain exhibited enhanced virulence. Moreover, administration of GAG protected mice from colitis induced by dextran sulfate sodium in an inflammasome-dependent manner. Thus, ribosomes connect the sensing of this fungal PAMP to the activation of an innate immune response.


Subject(s)
Aspergillosis/prevention & control , Aspergillus fumigatus/metabolism , Inflammasomes/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Polysaccharides/metabolism , Animals , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillus fumigatus/immunology , Biofilms , Colitis/chemically induced , Colitis/prevention & control , Dextran Sulfate , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Immunity, Innate , Inflammasomes/immunology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polysaccharides/immunology , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism
2.
Nature ; 555(7696): 382-386, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29489751

ABSTRACT

Resistance to infection is critically dependent on the ability of pattern recognition receptors to recognize microbial invasion and induce protective immune responses. One such family of receptors are the C-type lectins, which are central to antifungal immunity. These receptors activate key effector mechanisms upon recognition of conserved fungal cell-wall carbohydrates. However, several other immunologically active fungal ligands have been described; these include melanin, for which the mechanism of recognition is hitherto undefined. Here we identify a C-type lectin receptor, melanin-sensing C-type lectin receptor (MelLec), that has an essential role in antifungal immunity through recognition of the naphthalene-diol unit of 1,8-dihydroxynaphthalene (DHN)-melanin. MelLec recognizes melanin in conidial spores of Aspergillus fumigatus as well as in other DHN-melanized fungi. MelLec is ubiquitously expressed by CD31+ endothelial cells in mice, and is also expressed by a sub-population of these cells that co-express epithelial cell adhesion molecule and are detected only in the lung and the liver. In mouse models, MelLec was required for protection against disseminated infection with A. fumigatus. In humans, MelLec is also expressed by myeloid cells, and we identified a single nucleotide polymorphism of this receptor that negatively affected myeloid inflammatory responses and significantly increased the susceptibility of stem-cell transplant recipients to disseminated Aspergillus infections. MelLec therefore recognizes an immunologically active component commonly found on fungi and has an essential role in protective antifungal immunity in both mice and humans.


Subject(s)
Aspergillus fumigatus/immunology , Lectins, C-Type/immunology , Melanins/immunology , Naphthols/immunology , Animals , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillosis/prevention & control , Aspergillus fumigatus/chemistry , Aspergillus fumigatus/pathogenicity , Cell Wall/chemistry , Cell Wall/immunology , Female , Humans , Macrophages/immunology , Melanins/chemistry , Mice , Mice, Inbred C57BL , Naphthols/chemistry , Rats , Rats, Sprague-Dawley , Spores, Fungal/chemistry , Spores, Fungal/immunology , Substrate Specificity
3.
Cell ; 135(4): 726-37, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-19013280

ABSTRACT

The budding yeast, Saccharomyces cerevisiae, has emerged as an archetype of eukaryotic cell biology. Here we show that S. cerevisiae is also a model for the evolution of cooperative behavior by revisiting flocculation, a self-adherence phenotype lacking in most laboratory strains. Expression of the gene FLO1 in the laboratory strain S288C restores flocculation, an altered physiological state, reminiscent of bacterial biofilms. Flocculation protects the FLO1 expressing cells from multiple stresses, including antimicrobials and ethanol. Furthermore, FLO1(+) cells avoid exploitation by nonexpressing flo1 cells by self/non-self recognition: FLO1(+) cells preferentially stick to one another, regardless of genetic relatedness across the rest of the genome. Flocculation, therefore, is driven by one of a few known "green beard genes," which direct cooperation toward other carriers of the same gene. Moreover, FLO1 is highly variable among strains both in expression and in sequence, suggesting that flocculation in S. cerevisiae is a dynamic, rapidly evolving social trait.


Subject(s)
Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/physiology , Biofilms , Drug Resistance, Fungal , Flow Cytometry , Fungal Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Fungal , Genes, Fungal , Mannose-Binding Lectins , Membrane Proteins/metabolism , Microscopy , Models, Biological , Oligonucleotide Array Sequence Analysis , Phenotype , Saccharomyces cerevisiae Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 117(26): 14948-14957, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32541034

ABSTRACT

Diverting aminoacyl-transfer RNAs (tRNAs) from protein synthesis is a well-known process used by a wide range of bacteria to aminoacylate membrane constituents. By tRNA-dependently adding amino acids to glycerolipids, bacteria change their cell surface properties, which intensifies antimicrobial drug resistance, pathogenicity, and virulence. No equivalent aminoacylated lipids have been uncovered in any eukaryotic species thus far, suggesting that tRNA-dependent lipid remodeling is a process restricted to prokaryotes. We report here the discovery of ergosteryl-3ß-O-l-aspartate (Erg-Asp), a conjugated sterol that is produced by the tRNA-dependent addition of aspartate to the 3ß-OH group of ergosterol, the major sterol found in fungal membranes. In fact, Erg-Asp exists in the majority of "higher" fungi, including species of biotechnological interest, and, more importantly, in human pathogens like Aspergillus fumigatus We show that a bifunctional enzyme, ergosteryl-3ß-O-l-aspartate synthase (ErdS), is responsible for Erg-Asp synthesis. ErdS corresponds to a unique fusion of an aspartyl-tRNA synthetase-that produces aspartyl-tRNAAsp (Asp-tRNAAsp)-and of a Domain of Unknown Function 2156, which actually transfers aspartate from Asp-tRNAAsp onto ergosterol. We also uncovered that removal of the Asp modifier from Erg-Asp is catalyzed by a second enzyme, ErdH, that is a genuine Erg-Asp hydrolase participating in the turnover of the conjugated sterol in vivo. Phylogenomics highlights that the entire Erg-Asp synthesis/degradation pathway is conserved across "higher" fungi. Given the central roles of sterols and conjugated sterols in fungi, we propose that this tRNA-dependent ergosterol modification and homeostasis system might have broader implications in membrane remodeling, trafficking, antimicrobial resistance, or pathogenicity.


Subject(s)
Aspartic Acid/metabolism , Aspergillus fumigatus/metabolism , RNA, Fungal/metabolism , RNA, Transfer, Amino Acyl/metabolism , Sterols/metabolism , Aminoacylation , Aspartic Acid/chemistry , Aspergillus fumigatus/chemistry , Aspergillus fumigatus/genetics , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/genetics , Sterols/chemistry
5.
Annu Rev Microbiol ; 71: 99-116, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28701066

ABSTRACT

More than 90% of the cell wall of the filamentous fungus Aspergillus fumigatus comprises polysaccharides. Biosynthesis of the cell wall polysaccharides is under the control of three types of enzymes: transmembrane synthases, which are anchored to the plasma membrane and use nucleotide sugars as substrates, and cell wall-associated transglycosidases and glycosyl hydrolases, which are responsible for remodeling the de novo synthesized polysaccharides and establishing the three-dimensional structure of the cell wall. For years, the cell wall was considered an inert exoskeleton of the fungal cell. The cell wall is now recognized as a living organelle, since the composition and cellular localization of the different constitutive cell wall components (especially of the outer layers) vary when the fungus senses changes in the external environment. The cell wall plays a major role during infection. The recognition of the fungal cell wall by the host is essential in the initiation of the immune response. The interactions between the different pattern-recognition receptors (PRRs) and cell wall pathogen-associated molecular patterns (PAMPs) orientate the host response toward either fungal death or growth, which would then lead to disease development. Understanding the molecular determinants of the interplay between the cell wall and host immunity is fundamental to combatting Aspergillus diseases.


Subject(s)
Aspergillus fumigatus/metabolism , Aspergillus fumigatus/pathogenicity , Cell Wall/immunology , Cell Wall/metabolism , Polysaccharides/metabolism , Aspergillosis/immunology , Aspergillosis/pathology , Aspergillus fumigatus/enzymology , Host-Pathogen Interactions , Humans , Virulence
7.
Curr Top Microbiol Immunol ; 425: 331-369, 2020.
Article in English | MEDLINE | ID: mdl-32418033

ABSTRACT

The beginning of our understanding of the cell wall construction came from the work of talented biochemists in the 70-80's. Then came the era of sequencing. Paradoxically, the accumulation of fungal genomes complicated rather than solved the mystery of cell wall construction, by revealing the involvement of a much higher number of proteins than originally thought. The situation has become even more complicated since it is now recognized that the cell wall is an organelle whose composition continuously evolves with the changes in the environment or with the age of the fungal cell. The use of new and sophisticated technologies to observe cell wall construction at an almost atomic scale should improve our knowledge of the cell wall construction. This essay will present some of the major and still unresolved questions to understand the fungal cell wall biosynthesis and some of these exciting futurist approaches.


Subject(s)
Cell Wall/metabolism , Fungi/cytology , Fungi/metabolism , Cell Wall/chemistry
8.
Curr Top Microbiol Immunol ; 425: 167-186, 2020.
Article in English | MEDLINE | ID: mdl-32418035

ABSTRACT

Glycosylphosphatidylinositol (GPI) anchored proteins are a class of proteins attached to the extracellular leaflet of the plasma membrane via a post-translational modification, the glycolipid anchor. GPI anchored proteins are expressed in all eukaryotes, from fungi to plants and animals. They display very diverse functions ranging from enzymatic activity, signaling, cell adhesion, cell wall metabolism, and immune response. In this review, we investigated for the first time an exhaustive list of all the GPI anchored proteins present in the Aspergillus fumigatus genome. An A. fumigatus mutant library of all the genes that encode in silico identified GPI anchored proteins has been constructed and the phenotypic analysis of all these mutants has been characterized including their growth, conidial viability or morphology, adhesion and the ability to form biofilms. We showed the presence of different fungal categories of GPI anchored proteins in the A. fumigatus genome associated to their role in cell wall remodeling, adhesion, and biofilm formation.


Subject(s)
Aspergillus fumigatus/cytology , Aspergillus fumigatus/metabolism , Cell Wall/metabolism , Fungal Proteins/metabolism , Glycosylphosphatidylinositols/metabolism , Morphogenesis , Animals , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Fungal Proteins/genetics
9.
Clin Microbiol Rev ; 33(1)2019 12 18.
Article in English | MEDLINE | ID: mdl-31722890

ABSTRACT

Aspergillus fumigatus is a saprotrophic fungus; its primary habitat is the soil. In its ecological niche, the fungus has learned how to adapt and proliferate in hostile environments. This capacity has helped the fungus to resist and survive against human host defenses and, further, to be responsible for one of the most devastating lung infections in terms of morbidity and mortality. In this review, we will provide (i) a description of the biological cycle of A. fumigatus; (ii) a historical perspective of the spectrum of aspergillus disease and the current epidemiological status of these infections; (iii) an analysis of the modes of immune response against Aspergillus in immunocompetent and immunocompromised patients; (iv) an understanding of the pathways responsible for fungal virulence and their host molecular targets, with a specific focus on the cell wall; (v) the current status of the diagnosis of different clinical syndromes; and (vi) an overview of the available antifungal armamentarium and the therapeutic strategies in the clinical context. In addition, the emergence of new concepts, such as nutritional immunity and the integration and rewiring of multiple fungal metabolic activities occurring during lung invasion, has helped us to redefine the opportunistic pathogenesis of A. fumigatus.


Subject(s)
Aspergillosis/epidemiology , Aspergillosis/microbiology , Aspergillus fumigatus/physiology , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Aspergillosis/diagnosis , Aspergillosis/history , Aspergillus fumigatus/drug effects , Disease Susceptibility , History, 21st Century , Host-Pathogen Interactions/immunology , Humans , Immunity , Immunocompromised Host , Public Health Surveillance , Treatment Outcome , Virulence
10.
J Am Chem Soc ; 142(3): 1175-1179, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31913631

ABSTRACT

Using 3-O-benzoyl-4,6-O-di-tert-butylsilylidene-2-azido-2-deoxy-selenogalactoside, biotinylated oligo-α-(1 → 4)-d-galactosamines comprising from two to six GalN units were prepared for the first time together with their N-acetylated derivatives. The combination of blocking groups used herein provided stereocontrol for the α-stereospecific glycosylation, to show also high efficiency of phenyl 2-azido-2-deoxy-selenogalactosides as glycosyl donors. The obtained glycoconjugates are related to fragments of exopolysaccharide galactosaminogalactan (GG) found in Aspergillus fumigatus, which is the most important airborne human fungal pathogen in industrialized countries. The synthesized glycoconjugates were arrayed on streptavidin-coated plates and used to investigate the GG epitopes recognized by mouse monoclonal antibodies against GG and by human antibodies in the sera of patients with aspergillosis. The obtained data showed that the oligo-α-(1 → 4)-d-galactosamines and their N-acetylated derivatives allowed the first precise analysis of the specificity of the antibody responses to this extremely complex fungal polysaccharide.


Subject(s)
Biotinylation , Galactosamine/chemistry , Acetylation , Galactosamine/immunology , Humans , Stereoisomerism , Structure-Activity Relationship
11.
Eur J Immunol ; 49(6): 918-927, 2019 06.
Article in English | MEDLINE | ID: mdl-30903663

ABSTRACT

Aspergillus fumigatus is an important cause of pulmonary and systemic infections in immune compromised individuals, and of corneal ulcers and blindness in immune competent patients. To examine the role of chitin synthases in Aspergillus corneal infection, we analyzed Aspergillus mutants of chitin synthase family 1 and family 2, and found that compared with the parent strain, the quadruple mutants from both families were more readily killed by neutrophils in vitro, and that both also exhibited impaired hyphal growth in the cornea. Further, inhibition of chitin synthases using Nikkomycin Z enhanced neutrophil killing in vitro and in vivo in a murine model of A. fumigatus corneal infection. Acidic mammalian chitinase (AMCase) is mostly produced by macrophages in asthmatic lungs; however, we now demonstrate that neutrophils are a major source of AMCase, which inhibits hyphal growth. In A. fumigatus corneal infection, neutrophils are the major source of AMCase, and addition of AMCase inhibitors or adoptive transfer of neutrophils from AMCase-/- mice resulted in impaired hyphal killing. Together, these findings identify chitin synthases as important fungal virulence factors and neutrophil-derived AMCase as an essential mediator of host defense.


Subject(s)
Aspergillosis/immunology , Chitin Synthase/immunology , Chitinases/metabolism , Keratitis/immunology , Neutrophils/immunology , Animals , Aspergillus fumigatus/immunology , Aspergillus fumigatus/pathogenicity , Chitin Synthase/biosynthesis , Humans , Keratitis/metabolism , Keratitis/microbiology , Mice, Inbred C57BL , Neutrophils/enzymology , Virulence
12.
Cell Microbiol ; 21(12): e13102, 2019 12.
Article in English | MEDLINE | ID: mdl-31424155

ABSTRACT

The cell wall of Aspergillus fumigatus is predominantly composed of polysaccharides. The central fibrillar core of the cell wall is composed of a branched ß(1-3)glucan, to which the chitin and the galactomannan are covalently bound. Softening of the cell wall is an essential event during fungal morphogenesis, wherein rigid cell wall structures are cleaved by glycosyl hydrolases. In this study, we characterised the role of the glycosyl hydrolase GH55 members in A. fumigatus fungal morphogenesis. We showed that deletion of the six genes of the GH55 family stopped conidial cell wall maturation at the beginning of the development process, leading to abrogation of conidial separation: the shape of conidia became ovoid, and germination was delayed. In conclusion, the reorganisation and structuring of the conidial cell wall mediated by members of the GH55 family is essential for their maturation, normal dissemination, and germination.


Subject(s)
Aspergillus fumigatus/genetics , Cell Wall/genetics , Fungal Proteins/genetics , Morphogenesis/genetics , Spores, Fungal/genetics , Chitin/genetics
13.
Cell Microbiol ; 21(5): e12994, 2019 05.
Article in English | MEDLINE | ID: mdl-30552790

ABSTRACT

If the mycelium of Aspergillus fumigatus is very short-lived in the laboratory, conidia can survive for years. This survival capacity and extreme resistance to environmental insults is a major biological characteristic of this fungal species. Moreover, conidia, which easily reach the host alveola, are the infective propagules. Earlier studies have shown the role of some molecules of the outer conidial layer in protecting the fungus against the host defense. The outer layer of the conidial cell wall, directly in contact with the host cells, consists of α-(1,3)-glucan, melanin, and proteinaceous rodlets. This study is focused on the global importance of this outer layer. Single and multiple mutants without one to three major components of the outer layer were constructed and studied. The results showed that the absence of the target molecules resulting from multiple gene deletions led to unexpected phenotypes without any logical additivity. Unexpected compensatory cell wall surface modifications were indeed observed, such as the synthesis of the mycelial virulence factor galactosaminogalactan, the increase in chitin and glycoprotein concentration or particular changes in permeability. However, sensitivity of the multiple mutants to killing by phagocytic host cells confirmed the major importance of melanin in protecting conidia.


Subject(s)
Aspergillus fumigatus/metabolism , Cell Wall/metabolism , Melanins/metabolism , Spores, Fungal/metabolism , Aspergillosis/immunology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Aspergillus fumigatus/pathogenicity , Azoles/pharmacology , Benzenesulfonates/pharmacology , Caspofungin/pharmacology , Cell Wall/drug effects , Cell Wall/genetics , Chitin/metabolism , Congo Red/pharmacology , Fungal Proteins/metabolism , Glucans/genetics , Glucans/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Melanins/genetics , Melanins/physiology , Monocytes/immunology , Mycelium/metabolism , Phagocytes/metabolism , Polysaccharides/metabolism , Pyocyanine/pharmacology , Spores, Fungal/cytology , Spores, Fungal/genetics , Virulence Factors/metabolism
14.
J Biol Chem ; 293(40): 15538-15555, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30139746

ABSTRACT

Innate immunity in animals including humans encompasses the complement system, which is considered an important host defense mechanism against Aspergillus fumigatus, one of the most ubiquitous opportunistic human fungal pathogens. Previously, it has been shown that the alkaline protease Alp1p secreted from A. fumigatus mycelia degrades the complement components C3, C4, and C5. However, it remains unclear how the fungal spores (i.e. conidia) defend themselves against the activities of the complement system immediately after inhalation into the lung. Here, we show that A. fumigatus conidia contain a metalloprotease Mep1p, which is released upon conidial contact with collagen and inactivates all three complement pathways. In particular, Mep1p efficiently inactivated the major complement components C3, C4, and C5 and their activation products (C3a, C4a, and C5a) as well as the pattern-recognition molecules MBL and ficolin-1, either by directly cleaving them or by cleaving them to a form that is further broken down by other proteases of the complement system. Moreover, incubation of Mep1p with human serum significantly inhibited the complement hemolytic activity and conidial opsonization by C3b and their subsequent phagocytosis by macrophages. Together, these results indicate that Mep1p associated with and released from A. fumigatus conidia likely facilitates early immune evasion by disarming the complement defense in the human host.


Subject(s)
Aspergillus fumigatus/immunology , Complement C3/genetics , Complement C4/genetics , Complement C5/genetics , Invasive Pulmonary Aspergillosis/immunology , Metalloendopeptidases/immunology , Animals , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/pathogenicity , Collagen/genetics , Collagen/immunology , Complement C3/metabolism , Complement C4/metabolism , Complement C5/metabolism , Disease Models, Animal , Fungal Proteins/genetics , Fungal Proteins/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Invasive Pulmonary Aspergillosis/genetics , Invasive Pulmonary Aspergillosis/microbiology , Invasive Pulmonary Aspergillosis/pathology , Lectins/genetics , Lectins/immunology , Lung/immunology , Lung/pathology , Macrophages/immunology , Macrophages/microbiology , Male , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Phagocytosis , Spores, Fungal/growth & development , Spores, Fungal/immunology , Spores, Fungal/pathogenicity , Ficolins
15.
J Biol Chem ; 293(13): 4901-4912, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29414772

ABSTRACT

Surfactant protein D (SP-D), a C-type lectin and pattern-recognition soluble factor, plays an important role in immune surveillance to detect and eliminate human pulmonary pathogens. SP-D has been shown to protect against infections with the most ubiquitous airborne fungal pathogen, Aspergillus fumigatus, but the fungal surface component(s) interacting with SP-D is unknown. Here, we show that SP-D binds to melanin pigment on the surface of A. fumigatus dormant spores (conidia). SP-D also exhibited an affinity to two cell-wall polysaccharides of A. fumigatus, galactomannan (GM) and galactosaminogalactan (GAG). The immunolabeling pattern of SP-D was punctate on the conidial surface and was uniform on germinating conidia, in accordance with the localization of melanin, GM, and GAG. We also found that the collagen-like domain of SP-D is involved in its interaction with melanin, whereas its carbohydrate-recognition domain recognized GM and GAG. Unlike un-opsonized conidia, SP-D-opsonized conidia were phagocytosed more efficiently and stimulated the secretion of proinflammatory cytokines by human monocyte-derived macrophages. Furthermore, SP-D-/- mice challenged intranasally with wildtype conidia or melanin ghosts (i.e. hollow melanin spheres) displayed significantly reduced proinflammatory cytokines in the lung compared with wildtype mice. In summary, SP-D binds to melanin present on the dormant A. fumigatus conidial surface, facilitates conidial phagocytosis, and stimulates the host immune response.


Subject(s)
Aspergillus fumigatus/immunology , Fungal Polysaccharides/immunology , Melanins/immunology , Phagocytosis , Pulmonary Aspergillosis/immunology , Pulmonary Surfactant-Associated Protein D/immunology , Spores, Fungal/immunology , Animals , Aspergillus fumigatus/genetics , Fungal Polysaccharides/genetics , Melanins/genetics , Mice , Mice, Knockout , Pulmonary Aspergillosis/genetics , Pulmonary Aspergillosis/pathology , Pulmonary Surfactant-Associated Protein D/genetics , Spores, Fungal/genetics
16.
Appl Environ Microbiol ; 85(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31126941

ABSTRACT

The elongation growth of the mushroom stipe is a characteristic but not well-understood morphogenetic event of basidiomycetes. We found that extending native stipe cell walls of Coprinopsis cinerea were associated with the release of N-acetylglucosamine and chitinbiose and with chitinase activity. Two chitinases among all detected chitinases from C. cinerea, ChiE1 and ChiIII, reconstituted heat-inactivated stipe wall extension and released N-acetylglucosamine and chitinbiose. Interestingly, both ChiE1 and ChiIII hydrolyze insoluble crystalline chitin powder, while other C. cinerea chitinases do not, suggesting that crystalline chitin components of the stipe cell wall are the target of action for ChiE1 and ChiIII. ChiE1- or ChiIII-reconstituted heat-inactivated stipe walls showed maximal extension activity at pH 4.5, consistent with the optimal pH for native stipe wall extension in vitro; ChiE1- or ChiIII-reconstituted heat-inactivated stipe wall extension activities were associated with stipe elongation growth regions; and the combination of ChiE1 and ChiIII showed a synergism to reconstitute heat-inactivated stipe wall extension at a low action concentration. Field emission scanning electron microscopy (FESEM) images showed that the inner surface of acid-induced extended native stipe cell walls and ChiE1- or ChiIII-reconstituted extended heat-inactivated stipe cell walls exhibited a partially broken parallel microfibril architecture; however, these broken transversely arranged microfibrils were not observed in the unextended stipe cell walls that were induced by neutral pH buffer or heat inactivation. Double knockdown of ChiE1 and ChiIII resulted in the reduction of stipe elongation, mycelium growth, and heat-sensitive cell wall extension of native stipes. These results indicate a chitinase-hydrolyzing mechanism for stipe cell wall extension.IMPORTANCE A remarkable feature in the development of basidiomycete fruiting bodies is stipe elongation growth that results primarily from manifold cell elongation. Some scientists have suggested that stipe elongation is the result of enzymatic hydrolysis of cell wall polysaccharides, while other scientists have proposed the possibility that stipe elongation results from nonhydrolytic disruption of the hydrogen bonds between cell wall polysaccharides. Here, we show direct evidence for a chitinase-hydrolyzing mechanism of stipe cell wall elongation in the model mushroom Coprinopsis cinerea that is different from the expansin nonhydrolysis mechanism of plant cell wall extension. We presumed that in the growing stipe cell walls, parallel chitin microfibrils are tethered by ß-1,6-branched ß-1,3-glucans, and that the breaking of the tether by chitinases leads to separation of these microfibrils to increase their spacing for insertion of new synthesized chitin and ß-1,3-glucans under turgor pressure in vivo.


Subject(s)
Acetylglucosamine/metabolism , Agaricales/genetics , Cell Wall/metabolism , Chitin/metabolism , Chitinases/genetics , Fungal Proteins/genetics , Agaricales/metabolism , Chitinases/metabolism , Fungal Proteins/metabolism , Hydrolysis
17.
Bioconjug Chem ; 30(6): 1788-1797, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31125199

ABSTRACT

ß-(1,3)-Glucan is one of the antigenic components of the bacterial as well as fungal cell wall. We designed microcapsules (MCs) ligated with ß-(1,3)-glucan, to study its immunomodulatory effect. The MCs were obtained by interfacial polycondensation between diacyl chloride (sebacoyl chloride and terephtaloyl chloride) and diethylenetriamine in organic and aqueous phases, respectively. Planar films were first designed to optimize monomer compositions and to examine the kinetics of film formation. MCs with aqueous fluorescent core were then obtained upon controlled emulsification-polycondensation reactions using optimized monomer compositions and adding fluorescein into the aqueous phase. The selected MC-formulation was grafted with Curdlan, a linear ß-(1,3)-glucan from  Agrobacterium species or branched ß-(1,3)-glucan isolated from the cell wall of Aspergillus fumigatus. These ß-(1,3)-glucan grafted MCs were phagocytosed by human monocyte-derived macrophages, and stimulated cytokine secretion. Moreover, the blocking of dectin-1, a ß-(1,3)-glucan recognizing receptor, did not completely inhibit the phagocytosis of these ß-(1,3)-glucan grafted MCs, suggesting the involvement of other receptors in the recognition and uptake of ß-(1,3)-glucan. Overall, grafted MCs are a useful tool for the study of the mechanism of phagocytosis and immunomodulatory effect of the microbial polysaccharides.


Subject(s)
Adjuvants, Immunologic/pharmacology , Agrobacterium/chemistry , Aspergillus fumigatus/chemistry , Capsules , Cell Wall/chemistry , Polysaccharides/pharmacology , beta-Glucans/chemistry , Microscopy, Electron, Scanning , Rheology
18.
J Infect Dis ; 218(8): 1306-1313, 2018 09 08.
Article in English | MEDLINE | ID: mdl-29846638

ABSTRACT

Background: The airway epithelium is the first barrier interacting with Aspergillus fumigatus conidia after their inhalation, suggesting that this structure functions as point of entry of this fungus to initiate pulmonary aspergillosis. Methods: To study epithelial entry by A fumigatus, primary human reconstituted pseudostratified epithelium cultured in air-liquid interface as well as bronchial epithelial cell monolayers were infected with conidia. Results: Under these experimental conditions, we found that A fumigatus hyphae traversed the bronchial epithelium through a mechanism involving the recruitment of actin, which formed a tunnel that allows hyphae to enter the cells without disturbing their integrity. Conclusions: These findings describe a new mechanism by which A fumigatus hyphae penetrate the airway epithelial barrier and can infect its human host.


Subject(s)
Aspergillus fumigatus/physiology , Epithelial Cells/microbiology , Epithelium/microbiology , Hyphae/physiology , Lung/microbiology , Aspergillus fumigatus/ultrastructure , Cell Culture Techniques , Cells, Cultured , Epithelial Cells/ultrastructure , Humans , Hyphae/ultrastructure , Microscopy, Confocal , Microscopy, Electron, Transmission
19.
Mol Microbiol ; 105(6): 880-900, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28677124

ABSTRACT

Aspergillus fumigatus, a ubiquitous human fungal pathogen, produces asexual spores (conidia), which are the main mode of propagation, survival and infection of this human pathogen. In this study, we present the molecular characterization of a novel regulator of conidiogenesis and conidial survival called MybA because the predicted protein contains a Myb DNA binding motif. Cellular localization of the MybA::Gfp fusion and immunoprecipitation of the MybA::Gfp or MybA::3xHa protein showed that MybA is localized to the nucleus. RNA sequencing data and a uidA reporter assay indicated that the MybA protein functions upstream of wetA, vosA and velB, the key regulators involved in conidial maturation. The deletion of mybA resulted in a very significant reduction in the number and viability of conidia. As a consequence, the ΔmybA strain has a reduced virulence in an experimental murine model of aspergillosis. RNA-sequencing and biochemical studies of the ΔmybA strain suggested that MybA protein controls the expression of enzymes involved in trehalose biosynthesis as well as other cell wall and membrane-associated proteins and ROS scavenging enzymes. In summary, MybA protein is a new key regulator of conidiogenesis and conidial maturation and survival, and plays a crucial role in propagation and virulence of A. fumigatus.


Subject(s)
Aspergillus fumigatus/genetics , Spores, Fungal/genetics , Aspergillosis/microbiology , Aspergillus fumigatus/metabolism , Cell Wall/metabolism , Fungal Proteins/metabolism , Gene Deletion , Gene Expression Regulation, Fungal/genetics , Humans , Membrane Proteins/metabolism , Sequence Deletion , Transcription Factors/metabolism , Virulence/genetics
20.
Article in English | MEDLINE | ID: mdl-29632009

ABSTRACT

Aspergillus fumigatus can cause pulmonary aspergillosis in immunocompromised patients and is associated with a high mortality rate due to a lack of reliable treatment options. This opportunistic pathogen requires zinc in order to grow and cause disease. Novel compounds that interfere with fungal zinc metabolism may therefore be of therapeutic interest. We screened chemical libraries containing 59,223 small molecules using a resazurin assay that compared their effects on an A. fumigatus wild-type strain grown under zinc-limiting conditions and on a zinc transporter knockout strain grown under zinc-replete conditions to identify compounds affecting zinc metabolism. After a first screen, 116 molecules were selected whose inhibitory effects on fungal growth were further tested by using luminescence assays and hyphal length measurements to confirm their activity, as well as by toxicity assays on HeLa cells and mice. Six compounds were selected following a rescreening, of which two were pyrazolones, two were porphyrins, and two were polyaminocarboxylates. All three groups showed good in vitro activity, but only one of the polyaminocarboxylates was able to significantly improve the survival of immunosuppressed mice suffering from pulmonary aspergillosis. This two-tier screening approach led us to the identification of a novel small molecule with in vivo fungicidal effects and low murine toxicity that may lead to the development of new treatment options for fungal infections by administration of this compound either as a monotherapy or as part of a combination therapy.


Subject(s)
Antifungal Agents/therapeutic use , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/pathogenicity , Pulmonary Aspergillosis/drug therapy , Pulmonary Aspergillosis/metabolism , Zinc/metabolism , Animals , Disease Models, Animal , Luminescent Measurements , Mice , Microbial Sensitivity Tests , Pyrazolones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL