Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 16(1): 2305477, 2024.
Article in English | MEDLINE | ID: mdl-38298145

ABSTRACT

Non-LEE-encoded Effector A (NleA) is a type III secreted effector protein of enterohaemorrhagic and enteropathogenic Escherichia coli as well as the related mouse pathogen Citrobacter rodentium. NleA translocation into host cells is essential for virulence. We previously published several lines of evidence indicating that NleA is modified by host-mediated mucin-type O-linked glycosylation, the first example of a bacterial effector protein modified in this way. In this study, we use lectins to provide direct evidence for the modification of NleA by O-linked glycosylation and determine that the interaction of NleA with the COPII complex is necessary for this modification to occur.


Subject(s)
Enteropathogenic Escherichia coli , Escherichia coli Proteins , Gastrointestinal Microbiome , Animals , Mice , Escherichia coli Proteins/metabolism , Virulence Factors/metabolism , Glycosylation , Bacterial Proteins/metabolism
2.
Curr Protoc ; 3(8): e853, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37555775

ABSTRACT

The fungal cell wall and secreted exopolysaccharides play an important role in the interactions between fungi and their environment. Despite their central role in fungal biology, ecology, and host-pathogen interactions, the composition of these polymers and their synthetic pathways are not well understood. The protocols presented in this article describe an approach to isolate fungal cell wall polysaccharides and to identify and quantify the monosaccharide composition of these polymers by gas chromatography-mass spectrometry (GC-MS). © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: O-methyl trimethylsilyl monosaccharide derivatives composition analysis by GC-MS Support Protocol: Fungal cell wall extraction.


Subject(s)
Cell Wall , Fungal Polysaccharides , Gas Chromatography-Mass Spectrometry , Monosaccharides , Biopolymers/analysis , Biopolymers/isolation & purification , Cell Wall/chemistry , Fungal Polysaccharides/analysis , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/isolation & purification , Gas Chromatography-Mass Spectrometry/methods , Monosaccharides/analysis , Monosaccharides/chemistry , Monosaccharides/isolation & purification , Reference Standards , Calibration
3.
NPJ Biofilms Microbiomes ; 9(1): 7, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36732330

ABSTRACT

Pel exopolysaccharide biosynthetic loci are phylogenetically widespread biofilm matrix determinants in bacteria. In Pseudomonas aeruginosa, Pel is crucial for cell-to-cell interactions and reducing susceptibility to antibiotic and mucolytic treatments. While genes encoding glycoside hydrolases have long been linked to biofilm exopolysaccharide biosynthesis, their physiological role in biofilm development is unclear. Here we demonstrate that the glycoside hydrolase activity of P. aeruginosa PelA decreases adherent biofilm biomass and is responsible for generating the low molecular weight secreted form of the Pel exopolysaccharide. We show that the generation of secreted Pel contributes to the biomechanical properties of the biofilm and decreases the virulence of P. aeruginosa in Caenorhabditis elegans and Drosophila melanogaster. Our results reveal that glycoside hydrolases found in exopolysaccharide biosynthetic systems can help shape the soft matter attributes of a biofilm and propose that secreted matrix components be referred to as matrix associated to better reflect their influence.


Subject(s)
Biofilms , Glycoside Hydrolases , Polysaccharides, Bacterial , Pseudomonas aeruginosa , Animals , Biomechanical Phenomena , Drosophila melanogaster/microbiology , Glycoside Hydrolases/genetics , Pseudomonas aeruginosa/physiology , Virulence , Caenorhabditis elegans/microbiology
4.
Cell Surf ; 9: 100095, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36691652

ABSTRACT

Half a century after their discovery, polymers of N-acetylgalactosamine produced by the Aspergilli have garnered new interest as mediators of fungal virulence. Recent work has focused on the Aspergillus fumigatus secreted and cell wall-associated heteropolymer, galactosaminogalactan (GAG). This polymer, composed of galactose (Gal) and partially deacetylated N-acetylgalactosamine (GalNAc), plays a role in a variety of pathogenic processes including biofilm formation, immune modulation and evasion, and resistance to antifungals. Given its many potential contributions to fungal pathogenesis, GAG is a promising therapeutic target for novel antifungal strategies. As such, several studies have sought to elucidate the biosynthetic pathways required for GAG production and secretion. Herein we review the progress made in the understanding of the molecular mechanisms underlying GAG synthesis and identify several gaps in our understanding of this process.

5.
NPJ Biofilms Microbiomes ; 8(1): 83, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261442

ABSTRACT

The filamentous fungus Aspergillus fumigatus is an ubiquitous mold that can cause invasive pulmonary infections in immunocompromised patients. Within the lung, A. fumigatus forms biofilms that can enhance resistance to antifungals and immune defenses, highlighting the importance of defining the mechanisms underlying biofilm development and associated emergent properties. A. fumigatus biofilms display a morphology and architecture that is distinct from bacterial and yeast biofilms. Moreover, A. fumigatus biofilms display unique characteristics in the composition of their extracellular matrix (ECM) and the regulatory networks governing biofilm formation. This review will discuss our current understanding of the form and function of A. fumigatus biofilms, including the unique components of ECM matrix, potential drug resistance mechanisms, the regulatory networks governing A. fumigatus biofilm formation, and potential therapeutics targeting these structures.


Subject(s)
Antifungal Agents , Aspergillus fumigatus , Humans , Aspergillus fumigatus/genetics , Antifungal Agents/pharmacology , Biofilms , Fungi , Extracellular Matrix , Drug Resistance
7.
Commun Biol ; 5(1): 502, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35618750

ABSTRACT

The genetic capacity to synthesize the biofilm matrix exopolysaccharide Pel is widespread among Gram-negative and Gram-positive bacteria. However, its exact chemical structure has been challenging to determine. Using a Pseudomonas aeruginosa strain engineered to overproduce Pel, improvements to the isolation procedure, and selective hydrolysis with the glycoside hydrolase PelAh, we demonstrate that Pel is a partially de-N-acetylated linear polymer of α-1,4-N-acetylgalactosamine comprised predominantly of dimeric repeats of galactosamine and N-acetylgalactosamine.


Subject(s)
Acetylgalactosamine , Polysaccharides, Bacterial , Biofilms , Galactosamine , Polymers
8.
J Fungi (Basel) ; 8(4)2022 Mar 24.
Article in English | MEDLINE | ID: mdl-35448567

ABSTRACT

The mold Aspergillus fumigatus and bacterium Pseudomonas aeruginosa form biofilms in the airways of individuals with cystic fibrosis. Biofilm formation by A. fumigatus depends on the self-produced cationic exopolysaccharide galactosaminogalactan (GAG), while P. aeruginosa biofilms can contain the cationic exopolysaccharide Pel. GAG and Pel are rendered cationic by deacetylation mediated by either the secreted deacetylase Agd3 (A. fumigatus) or the periplasmic deacetylase PelA (P. aeruginosa). Given the similarities between these polymers, the potential for biofilm interactions between these organisms were investigated. P. aeruginosa were observed to adhere to A. fumigatus hyphae in a GAG-dependent manner and to GAG-coated coverslips of A. fumigatus biofilms. In biofilm adherence assays, incubation of P. aeruginosa with A. fumigatus culture supernatants containing de-N-acetylated GAG augmented the formation of adherent P. aeruginosa biofilms, increasing protection against killing by the antibiotic colistin. Fluorescence microscopy demonstrated incorporation of GAG within P. aeruginosa biofilms, suggesting that GAG can serve as an alternate biofilm exopolysaccharide for this bacterium. In contrast, Pel-containing bacterial culture supernatants only augmented the formation of adherent A. fumigatus biofilms when antifungal inhibitory molecules were removed. This study demonstrates biofilm interaction via exopolysaccharides as a potential mechanism of co-operation between these organisms in chronic lung disease.

9.
mBio ; 13(2): e0293321, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35254131

ABSTRACT

Alanine metabolism has been suggested as an adaptation strategy to oxygen limitation in organisms ranging from plants to mammals. Within the pulmonary infection microenvironment, Aspergillus fumigatus forms biofilms with steep oxygen gradients defined by regions of oxygen limitation. An alanine aminotransferase, AlaA, was observed to function in alanine catabolism and is required for several aspects of A. fumigatus biofilm physiology. Loss of alaA, or its catalytic activity, results in decreased adherence of biofilms through a defect in the maturation of the extracellular matrix polysaccharide galactosaminogalactan (GAG). Additionally, exposure of cell wall polysaccharides is also impacted by loss of alaA, and loss of AlaA catalytic activity confers increased biofilm susceptibility to echinocandin treatment, which is correlated with enhanced fungicidal activity. The increase in echinocandin susceptibility is specific to biofilms, and chemical inhibition of alaA by the alanine aminotransferase inhibitor ß-chloro-l-alanine is sufficient to sensitize A. fumigatus biofilms to echinocandin treatment. Finally, loss of alaA increases susceptibility of A. fumigatus to in vivo echinocandin treatment in a murine model of invasive pulmonary aspergillosis. Our results provide insight into the interplay of metabolism, biofilm formation, and antifungal drug resistance in A. fumigatus and describe a mechanism of increasing susceptibility of A. fumigatus biofilms to the echinocandin class of antifungal drugs. IMPORTANCE Aspergillus fumigatus is a ubiquitous filamentous fungus that causes an array of diseases depending on the immune status of an individual, collectively termed aspergillosis. Antifungal therapy for invasive pulmonary aspergillosis (IPA) or chronic pulmonary aspergillosis (CPA) is limited and too often ineffective. This is in part due to A. fumigatus biofilm formation within the infection environment and the resulting emergent properties, particularly increased antifungal resistance. Thus, insights into biofilm formation and mechanisms driving increased antifungal drug resistance are critical for improving existing therapeutic strategies and development of novel antifungals. In this work, we describe an unexpected observation where alanine metabolism, via the alanine aminotransferase AlaA, is required for several aspects of A. fumigatus biofilm physiology, including resistance of A. fumigatus biofilms to the echinocandin class of antifungal drugs. Importantly, we observed that chemical inhibition of alanine aminotransferases is sufficient to increase echinocandin susceptibility and that loss of alaA increases susceptibility to echinocandin treatment in a murine model of IPA. AlaA is the first gene discovered in A. fumigatus that confers resistance to an antifungal drug specifically in a biofilm context.


Subject(s)
Aspergillus fumigatus , Invasive Pulmonary Aspergillosis , Alanine/metabolism , Alanine/pharmacology , Alanine/therapeutic use , Alanine Transaminase/metabolism , Alanine Transaminase/pharmacology , Animals , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Biofilms , Disease Models, Animal , Echinocandins/metabolism , Echinocandins/pharmacology , Echinocandins/therapeutic use , Mammals , Mice , Oxygen/metabolism
10.
mBio ; 11(6)2020 11 10.
Article in English | MEDLINE | ID: mdl-33173002

ABSTRACT

Polysaccharides are key components of both the fungal cell wall and biofilm matrix. Despite having distinct assembly and regulation pathways, matrix exopolysaccharide and cell wall polysaccharides share common substrates and intermediates in their biosynthetic pathways. It is not clear, however, if the biosynthetic pathways governing the production of these polysaccharides are cooperatively regulated. Here, we demonstrate that cell wall stress promotes production of the exopolysaccharide galactosaminogalactan (GAG)-depend biofilm formation in the major fungal pathogen of humans Aspergillus fumigatus and that the transcription factor SomA plays a crucial role in mediating this process. A core set of SomA target genes were identified by transcriptome sequencing and chromatin immunoprecipitation coupled to sequencing (ChIP-Seq). We identified a novel SomA-binding site in the promoter regions of GAG biosynthetic genes agd3 and ega3, as well as its regulators medA and stuA Strikingly, this SomA-binding site was also found in the upstream regions of genes encoding the cell wall stress sensors, chitin synthases, and ß-1,3-glucan synthase. Thus, SomA plays a direct regulation of both GAG and cell wall polysaccharide biosynthesis. Consistent with these findings, SomA is required for the maintenance of normal cell wall architecture and compositions in addition to its function in biofilm development. Moreover, SomA was found to globally regulate glucose uptake and utilization, as well as amino sugar and nucleotide sugar metabolism, which provides precursors for polysaccharide synthesis. Collectively, our work provides insight into fungal adaptive mechanisms in response to cell wall stress where biofilm formation and cell wall homeostasis were synchronously regulated.IMPORTANCE The cell wall is essential for fungal viability and is absent from human hosts; thus, drugs disrupting cell wall biosynthesis have gained more attention. Caspofungin is a member of a new class of clinically approved echinocandin drugs to treat invasive aspergillosis by blocking ß-1,3-glucan synthase, thus damaging the fungal cell wall. Here, we demonstrate that caspofungin and other cell wall stressors can induce galactosaminogalactan (GAG)-dependent biofilm formation in the human pathogen Aspergillus fumigatus We further identified SomA as a master transcription factor playing a dual role in both biofilm formation and cell wall homeostasis. SomA plays this dual role by direct binding to a conserved motif upstream of GAG biosynthetic genes and genes involved in cell wall stress sensors, chitin synthases, and ß-1,3-glucan synthase. Collectively, these findings reveal a transcriptional control pathway that integrates biofilm formation and cell wall homeostasis and suggest SomA as an attractive target for antifungal drug development.


Subject(s)
Aspergillus fumigatus/metabolism , Biofilms , Cell Wall/metabolism , Fungal Proteins/metabolism , Transcription Factors/metabolism , Aspergillosis/microbiology , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Cell Wall/genetics , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Glucose/metabolism , Homeostasis , Humans , Polysaccharides/biosynthesis , Promoter Regions, Genetic , Transcription Factors/genetics
11.
Nat Commun ; 11(1): 2450, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415073

ABSTRACT

The exopolysaccharide galactosaminogalactan (GAG) is an important virulence factor of the fungal pathogen Aspergillus fumigatus. Deletion of a gene encoding a putative deacetylase, Agd3, leads to defects in GAG deacetylation, biofilm formation, and virulence. Here, we show that Agd3 deacetylates GAG in a metal-dependent manner, and is the founding member of carbohydrate esterase family CE18. The active site is formed by four catalytic motifs that are essential for activity. The structure of Agd3 includes an elongated substrate-binding cleft formed by a carbohydrate binding module (CBM) that is the founding member of CBM family 87. Agd3 homologues are encoded in previously unidentified putative bacterial exopolysaccharide biosynthetic operons and in other fungal genomes.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/metabolism , Aspergillus fumigatus/enzymology , Aspergillus fumigatus/physiology , Biofilms/growth & development , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Polysaccharides/metabolism , Acetylation , Amino Acid Sequence , Aspergillus fumigatus/genetics , Catalytic Domain , Conserved Sequence , Gene Expression Regulation, Fungal , Glycosaminoglycans/biosynthesis , Metals/metabolism , Protein Domains , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity , Time Factors
12.
Curr Top Microbiol Immunol ; 425: 225-254, 2020.
Article in English | MEDLINE | ID: mdl-32072265

ABSTRACT

During infection, many fungal pathogens form biofilms within tissues or on biomedical devices. The growth of fungi within biofilms increases dramatically their resistance to both immune defences and antifungal therapies. In the last twenty years, studies have begun to shed light on many of the steps involved in biofilm synthesis and composition, revealing new antifungal strategies. This chapter will focus on the biofilm exopolysaccharides produced by A. fumigatus and C. albicans, the two main causes of human fungal infections. We will review the current state of our understanding of the structure, biosynthesis, and role of exopolysaccharides in biofilm development and function with a view to identifying future strategies for prophylaxis and treatment of these devastating infections.


Subject(s)
Biofilms , Fungal Polysaccharides/metabolism , Mycoses/drug therapy , Mycoses/microbiology , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/metabolism , Biofilms/drug effects , Candida albicans/drug effects , Candida albicans/metabolism , Fungal Polysaccharides/biosynthesis , Fungal Polysaccharides/chemistry , Humans
13.
J Biol Chem ; 294(37): 13833-13849, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31416836

ABSTRACT

Aspergillus fumigatus is an opportunistic fungal pathogen that causes both chronic and acute invasive infections. Galactosaminogalactan (GAG) is an integral component of the A. fumigatus biofilm matrix and a key virulence factor. GAG is a heterogeneous linear α-1,4-linked exopolysaccharide of galactose and GalNAc that is partially deacetylated after secretion. A cluster of five co-expressed genes has been linked to GAG biosynthesis and modification. One gene in this cluster, ega3, is annotated as encoding a putative α-1,4-galactosaminidase belonging to glycoside hydrolase family 114 (GH114). Herein, we show that recombinant Ega3 is an active glycoside hydrolase that disrupts GAG-dependent A. fumigatus and Pel polysaccharide-dependent Pseudomonas aeruginosa biofilms at nanomolar concentrations. Using MS and functional assays, we demonstrate that Ega3 is an endo-acting α-1,4-galactosaminidase whose activity depends on the conserved acidic residues, Asp-189 and Glu-247. X-ray crystallographic structural analysis of the apo Ega3 and an Ega3-galactosamine complex, at 1.76 and 2.09 Å resolutions, revealed a modified (ß/α)8-fold with a deep electronegative cleft, which upon ligand binding is capped to form a tunnel. Our structural analysis coupled with in silico docking studies also uncovered the molecular determinants for galactosamine specificity and substrate binding at the -2 to +1 binding subsites. The findings in this study increase the structural and mechanistic understanding of the GH114 family, which has >600 members encoded by plant and opportunistic human pathogens, as well as in industrially used bacteria and fungi.


Subject(s)
Aspergillus fumigatus/metabolism , Glycoside Hydrolases/genetics , Hexosaminidases/metabolism , Aspergillus fumigatus/genetics , Aspergillus fumigatus/ultrastructure , Biofilms/drug effects , Crystallography, X-Ray/methods , Fungal Proteins/genetics , Fungi/metabolism , Glycoside Hydrolases/metabolism , Hexosaminidases/pharmacology , Hexosaminidases/ultrastructure , Polysaccharides/metabolism , Virulence
14.
J Biol Chem ; 294(28): 10760-10772, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31167793

ABSTRACT

During infection, the fungal pathogen Aspergillus fumigatus forms biofilms that enhance its resistance to antimicrobials and host defenses. An integral component of the biofilm matrix is galactosaminogalactan (GAG), a cationic polymer of α-1,4-linked galactose and partially deacetylated N-acetylgalactosamine (GalNAc). Recent studies have shown that recombinant hydrolase domains from Sph3, an A. fumigatus glycoside hydrolase involved in GAG synthesis, and PelA, a multifunctional protein from Pseudomonas aeruginosa involved in Pel polysaccharide biosynthesis, can degrade GAG, disrupt A. fumigatus biofilms, and attenuate fungal virulence in a mouse model of invasive aspergillosis. The molecular mechanisms by which these enzymes disrupt biofilms have not been defined. We hypothesized that the hydrolase domains of Sph3 and PelA (Sph3h and PelAh, respectively) share structural and functional similarities given their ability to degrade GAG and disrupt A. fumigatus biofilms. MALDI-TOF enzymatic fingerprinting and NMR experiments revealed that both proteins are retaining endo-α-1,4-N-acetylgalactosaminidases with a minimal substrate size of seven residues. The crystal structure of PelAh was solved to 1.54 Å and structure alignment to Sph3h revealed that the enzymes share similar catalytic site residues. However, differences in the substrate-binding clefts result in distinct enzyme-substrate interactions. PelAh hydrolyzed partially deacetylated substrates better than Sph3h, a finding that agrees well with PelAh's highly electronegative binding cleft versus the neutral surface present in Sph3h Our insight into PelAh's structure and function necessitate the creation of a new glycoside hydrolase family, GH166, whose structural and mechanistic features, along with those of GH135 (Sph3), are reported here.


Subject(s)
Biofilms/drug effects , Glycoside Hydrolases/metabolism , Polysaccharide-Lyases/ultrastructure , Anti-Infective Agents/metabolism , Aspergillus fumigatus/metabolism , Biofilms/growth & development , Catalytic Domain , Fungal Proteins/metabolism , Fungi/metabolism , Glycoside Hydrolases/physiology , Hydrolysis , Polysaccharide-Lyases/metabolism , Polysaccharides/metabolism , Pseudomonas aeruginosa/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Substrate Specificity/physiology , Virulence
15.
PLoS Pathog ; 14(4): e1006998, 2018 04.
Article in English | MEDLINE | ID: mdl-29684093

ABSTRACT

Poly-ß(1,6)-N-acetyl-D-glucosamine (PNAG) is a major biofilm component of many pathogenic bacteria. The production, modification, and export of PNAG in Escherichia coli and Bordetella species require the protein products encoded by the pgaABCD operon. PgaB is a two-domain periplasmic protein that contains an N-terminal deacetylase domain and a C-terminal PNAG binding domain that is critical for export. However, the exact function of the PgaB C-terminal domain remains unclear. Herein, we show that the C-terminal domains of Bordetella bronchiseptica PgaB (PgaBBb) and E. coli PgaB (PgaBEc) function as glycoside hydrolases. These enzymes hydrolyze purified deacetylated PNAG (dPNAG) from Staphylococcus aureus, disrupt PNAG-dependent biofilms formed by Bordetella pertussis, Staphylococcus carnosus, Staphylococcus epidermidis, and E. coli, and potentiate bacterial killing by gentamicin. Furthermore, we found that PgaBBb was only able to hydrolyze PNAG produced in situ by the E. coli PgaCD synthase complex when an active deacetylase domain was present. Mass spectrometry analysis of the PgaB-hydrolyzed dPNAG substrate showed a GlcN-GlcNAc-GlcNAc motif at the new reducing end of detected fragments. Our 1.76 Å structure of the C-terminal domain of PgaBBb reveals a central cavity within an elongated surface groove that appears ideally suited to recognize the GlcN-GlcNAc-GlcNAc motif. The structure, in conjunction with molecular modeling and site directed mutagenesis led to the identification of the dPNAG binding subsites and D474 as the probable catalytic acid. This work expands the role of PgaB within the PNAG biosynthesis machinery, defines a new glycoside hydrolase family GH153, and identifies PgaB as a possible therapeutic agent for treating PNAG-dependent biofilm infections.


Subject(s)
Amidohydrolases/metabolism , Biofilms/growth & development , Bordetella/enzymology , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Glycoside Hydrolases/metabolism , beta-Glucans/chemistry , Acetylation , Amidohydrolases/chemistry , Bordetella/growth & development , Crystallography, X-Ray , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Glycoside Hydrolases/chemistry , Operon , Protein Conformation , beta-Glucans/metabolism
16.
Plant Biotechnol J ; 15(3): 285-296, 2017 03.
Article in English | MEDLINE | ID: mdl-27483398

ABSTRACT

Influenza virus-like particles (VLPs) have been shown to induce a safe and potent immune response through both humoral and cellular responses. They represent promising novel influenza vaccines. Plant-based biotechnology allows for the large-scale production of VLPs of biopharmaceutical interest using different model organisms, including Nicotiana benthamiana plants. Through this platform, influenza VLPs bud from the plasma membrane and accumulate between the membrane and the plant cell wall. To design and optimize efficient production processes, a better understanding of the plant cell wall composition of infiltrated tobacco leaves is a major interest for the plant biotechnology industry. In this study, we have investigated the alteration of the biochemical composition of the cell walls of N. benthamiana leaves subjected to abiotic and biotic stresses induced by the Agrobacterium-mediated transient transformation and the resulting high expression levels of influenza VLPs. Results show that abiotic stress due to vacuum infiltration without Agrobacterium did not induce any detectable modification of the leaf cell wall when compared to non infiltrated leaves. In contrast, various chemical changes of the leaf cell wall were observed post-Agrobacterium infiltration. Indeed, Agrobacterium infection induced deposition of callose and lignin, modified the pectin methylesterification and increased both arabinosylation of RG-I side chains and the expression of arabinogalactan proteins. Moreover, these modifications were slightly greater in plants expressing haemagglutinin-based VLP than in plants infiltrated with the Agrobacterium strain containing only the p19 suppressor of silencing.


Subject(s)
Agrobacterium/metabolism , Biotechnology/methods , Cell Wall/metabolism , Hemagglutinins/metabolism , Nicotiana/metabolism , Agrobacterium/genetics , Hemagglutinins/genetics , Influenza Vaccines/genetics , Influenza Vaccines/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Nicotiana/genetics
17.
mBio ; 7(2): e00252-16, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27048799

ABSTRACT

UNLABELLED: The mold Aspergillus fumigatus causes invasive infection in immunocompromised patients. Recently, galactosaminogalactan (GAG), an exopolysaccharide composed of galactose and N-acetylgalactosamine (GalNAc), was identified as a virulence factor required for biofilm formation. The molecular mechanisms underlying GAG biosynthesis and GAG-mediated biofilm formation were unknown. We identified a cluster of five coregulated genes that were dysregulated in GAG-deficient mutants and whose gene products share functional similarity with proteins that mediate the synthesis of the bacterial biofilm exopolysaccharide poly-(ß1-6)-N-acetyl-D-glucosamine (PNAG). Bioinformatic analyses suggested that the GAG cluster gene agd3 encodes a protein containing a deacetylase domain. Because deacetylation of N-acetylglucosamine residues is critical for the function of PNAG, we investigated the role of GAG deacetylation in fungal biofilm formation. Agd3 was found to mediate deacetylation of GalNAc residues within GAG and render the polysaccharide polycationic. As with PNAG, deacetylation is required for the adherence of GAG to hyphae and for biofilm formation. Growth of the Δagd3 mutant in the presence of culture supernatants of the GAG-deficient Δuge3 mutant rescued the biofilm defect of the Δagd3 mutant and restored the adhesive properties of GAG, suggesting that deacetylation is an extracellular process. The GAG biosynthetic gene cluster is present in the genomes of members of the Pezizomycotina subphylum of the Ascomycota including a number of plant-pathogenic fungi and a single basidiomycete species,Trichosporon asahii, likely a result of recent horizontal gene transfer. The current study demonstrates that the production of cationic, deacetylated exopolysaccharides is a strategy used by both fungi and bacteria for biofilm formation. IMPORTANCE: This study sheds light on the biosynthetic pathways governing the synthesis of galactosaminogalactan (GAG), which plays a key role in A. fumigatus virulence and biofilm formation. We find that bacteria and fungi use similar strategies to synthesize adhesive biofilm exopolysaccharides. The presence of orthologs of the GAG biosynthetic gene clusters in multiple fungi suggests that this exopolysaccharide may also be important in the virulence of other fungal pathogens. Further, these studies establish a molecular mechanism of adhesion in which GAG interacts via charge-charge interactions to bind to both fungal hyphae and other substrates. Finally, the importance of deacetylation in the synthesis of functional GAG and the extracellular localization of this process suggest that inhibition of deacetylation may be an attractive target for the development of novel antifungal therapies.


Subject(s)
Aspergillosis/microbiology , Aspergillus fumigatus/physiology , Biofilms , Polysaccharides/metabolism , Acetylation , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans
18.
Plant Signal Behav ; 10(6): e1026023, 2015.
Article in English | MEDLINE | ID: mdl-26176901

ABSTRACT

Two independent studies have shown that the cell wall of pollen tubes from tobacco and tomato species contained fucosylated xyloglucan (XyG). These findings are intriguing as many reports have shown that XyG of somatic cells of these species is not fucosylated but instead is arabinosylated. In order to produce fucosylated XyG, plants must express a functional galactoside α-2-fucosyltransferase. Here, using a bioinformatics approach, we show that several candidate genes coding for XyG fucosyltransferases are present in the genome of coffee and several Solanaceae species including tomato, tobacco, potato, eggplant and pepper. BLAST and protein alignments with the 2 well-characterized XyG fucosyltransferases from Arabidopsis thaliana and Pisum sativum revealed that at least 6 proteins from different Solanaceae species and from coffee displayed the 3 conserved motifs required for XyG fucosyltransferase activity.


Subject(s)
Fucosyltransferases/metabolism , Genome, Plant , Plant Proteins/metabolism , Solanaceae/enzymology , Solanaceae/genetics , Algorithms , Amino Acid Motifs , Amino Acid Sequence , Coffea/enzymology , Computer Simulation , Fucosyltransferases/chemistry , Molecular Sequence Data , Phylogeny , Plant Proteins/chemistry , Protein Structure, Tertiary , Sequence Alignment
19.
BMC Microbiol ; 15: 72, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25886496

ABSTRACT

BACKGROUND: Pseudomonas fluorescens strain MFE01 secretes in abundance two Hcp proteins (haemolysin co-regulated proteins) Hcp1 and Hcp2, characteristic of a functional type 6 secretion system. Phenotypic studies have shown that MFE01 has antibacterial activity against a wide range of competitor bacteria, including rhizobacteria and clinically relevant bacteria. Mutagenesis of the hcp2 gene abolishes or reduces, depending on the target strain, MFE01 antibacterial activity. Hcp1, encoded by hcp1, may also be involved in bacterial competition. We therefore assessed the contribution of Hcp1 to competition of P. fluorescens MFE01 with other bacteria, by studying MFE01 mutants in various competitive conditions. RESULTS: Mutation of hcp1 had pleiotropic effects on the MFE01 phenotype. It affected mucoidy of the strain and its motility and was associated with the loss of flagella, which were restored by introduction of plasmid expressing hcp1. The hcp1 mutation had no effect on bacterial competition during incubation in solid medium. MFE01 was able to sequester another P. fluorescens strain, MFN1032, under swimming conditions. The hcp2 mutant but not the hcp1 mutant conserved this ability. In competition assays on swarming medium, MFE01 impaired MFN1032 swarming and displayed killing activity. The hcp2 mutant, but not the hcp1 mutant, was able to reduce MFN1032 swarming. The hcp1 and hcp2 mutations each abolished killing activity in these conditions. CONCLUSION: Our findings implicate type 6 secretion of Hcp1 in mucoidy and motility of MFE01. Our study is the first to establish a link between a type 6 secretion system and flagellin and mucoidy. Hcp1 also appears to contribute to limiting the motility of prey cells to facilitate killing mediated by Hcp2. Inhibition of motility associated with an Hcp protein has never been described. With this work, we illustrate the importance and versatility of type 6 secretion systems in bacterial adaptation and fitness.


Subject(s)
Antibiosis , Bacterial Proteins/metabolism , Locomotion , Polysaccharides, Bacterial/metabolism , Pseudomonas fluorescens/physiology , Type VI Secretion Systems/metabolism , Bacterial Proteins/genetics , Gene Deletion , Genetic Complementation Test , Pseudomonas fluorescens/genetics , Pseudomonas fluorescens/metabolism
20.
Ann Bot ; 115(1): 55-66, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25434027

ABSTRACT

BACKGROUND AND AIMS: In flowering plants, fertilization relies on the delivery of the sperm cells carried by the pollen tube to the ovule. During the tip growth of the pollen tube, proper assembly of the cell wall polymers is required to maintain the mechanical properties of the cell wall. Xyloglucan (XyG) is a cell wall polymer known for maintaining the wall integrity and thus allowing cell expansion. In most angiosperms, the XyG of somatic cells is fucosylated, except in the Asterid clade (including the Solanaceae), where the fucosyl residues are replaced by arabinose, presumably due to an adaptive and/or selective diversification. However, it has been shown recently that XyG of Nicotiana alata pollen tubes is mostly fucosylated. The objective of the present work was to determine whether such structural differences between somatic and gametophytic cells are a common feature of Nicotiana and Solanum (more precisely tomato) genera. METHODS: XyGs of pollen tubes of domesticated (Solanum lycopersicum var. cerasiforme and var. Saint-Pierre) and wild (S. pimpinellifolium and S. peruvianum) tomatoes and tobacco (Nicotiana tabacum) were analysed by immunolabelling, oligosaccharide mass profiling and GC-MS analyses. KEY RESULTS: Pollen tubes from all the species were labelled with the mAb CCRC-M1, a monoclonal antibody that recognizes epitopes associated with fucosylated XyG motifs. Analyses of the cell wall did not highlight major structural differences between previously studied N. alata and N. tabacum XyG. In contrast, XyG of tomato pollen tubes contained fucosylated and arabinosylated motifs. The highest levels of fucosylated XyG were found in pollen tubes from the wild species. CONCLUSIONS: The results clearly indicate that the male gametophyte (pollen tube) and the sporophyte have structurally different XyG. This suggests that fucosylated XyG may have an important role in the tip growth of pollen tubes, and that they must have a specific set of functional XyG fucosyltransferases, which are yet to be characterized.


Subject(s)
Glucans/metabolism , Nicotiana/metabolism , Solanum lycopersicum/metabolism , Solanum/metabolism , Xylans/metabolism , Arabinose/metabolism , Fucosyltransferases/metabolism , Gas Chromatography-Mass Spectrometry , Immunohistochemistry , Solanum lycopersicum/enzymology , Oligosaccharides/chemistry , Plant Proteins/metabolism , Pollen Tube/metabolism , Solanum/enzymology , Nicotiana/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...