Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Curr Res Food Sci ; 7: 100603, 2023.
Article in English | MEDLINE | ID: mdl-37840697

ABSTRACT

Non-conventional yeast species, or non-Saccharomyces yeasts, are increasingly recognized for their involvement in fermented foods. Many of them exhibit probiotic characteristics that are mainly due to direct contacts with other cell types through various molecular components of their cell wall. The biochemical composition and/or the molecular structure of the cell wall components are currently considered the primary determinant of their probiotic properties. Here we first present the techniques that are used to extract and analyze the cell wall components of food industry-related non-Saccharomyces yeasts. We then review the current understanding of the cell wall composition and structure of each polysaccharide from these yeasts. Finally, the data exploring the potential beneficial role of their cell wall components, which could be a source of innovative functional ingredients, are discussed. Such research would allow the development of high value-added products and provide the food industry with novel inputs beyond the well-established S. cerevisiae.

2.
J Mol Biol ; 435(10): 168048, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36933821

ABSTRACT

Knr4/Smi1 proteins are specific to the fungal kingdom and their deletion in the model yeast Saccharomyces cerevisiae and the human pathogen Candida albicans results in hypersensitivity to specific antifungal agents and a wide range of parietal stresses. In S. cerevisiae, Knr4 is located at the crossroads of several signalling pathways, including the conserved cell wall integrity and calcineurin pathways. Knr4 interacts genetically and physically with several protein members of those pathways. Its sequence suggests that it contains large intrinsically disordered regions. Here, a combination of small-angle X-ray scattering (SAXS) and crystallographic analysis led to a comprehensive structural view of Knr4. This experimental work unambiguously showed that Knr4 comprises two large intrinsically disordered regions flanking a central globular domain whose structure has been established. The structured domain is itself interrupted by a disordered loop. Using the CRISPR/Cas9 genome editing technique, strains expressing KNR4 genes deleted from different domains were constructed. The N-terminal domain and the loop are essential for optimal resistance to cell wall-binding stressors. The C-terminal disordered domain, on the other hand, acts as a negative regulator of this function of Knr4. The identification of molecular recognition features, the possible presence of secondary structure in these disordered domains and the functional importance of the disordered domains revealed here designate these domains as putative interacting spots with partners in either pathway. Targeting these interacting regions is a promising route to the discovery of inhibitory molecules that could increase the susceptibility of pathogens to the antifungals currently in clinical use.


Subject(s)
Intrinsically Disordered Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Cell Wall/metabolism , Intrinsically Disordered Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Scattering, Small Angle , Transcription Factors/metabolism , X-Ray Diffraction
3.
Cell Microbiol ; 18(9): 1217-27, 2016 09.
Article in English | MEDLINE | ID: mdl-27199081

ABSTRACT

The most highly connected proteins in protein-protein interactions networks are called hubs; they generally connect signalling pathways. In Saccharomyces cerevisiae, Knr4 constitutes a connecting node between the two main signal transmission pathways involved in cell wall maintenance upon stress: the cell wall integrity and the calcium-calcineurin pathway. Knr4 is required to enable the cells to resist many cell wall-affecting stresses, and KNR4 gene deletion is synthetic lethal with the simultaneous deletion of numerous other genes involved in morphogenesis and cell wall biogenesis. Knr4 has been shown to engage in multiple physical interactions, an ability conferred by the intrinsic structural adaptability of major disordered regions present in the N-terminal and C-terminal parts of the protein. Taking all together, Knr4 is an intrinsically disordered hub protein. Available data from other fungi indicate the conservation of Knr4 homologs cellular function and localization at sites of polarized growth among fungal species, including pathogenic species. Because of their particular role in morphogenesis control and of their fungal specificity, these proteins could constitute interesting new pharmaceutical drug targets for antifungal combination therapy.


Subject(s)
Cell Wall/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Transcription Factors/physiology , Cell Cycle Checkpoints , Humans , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Saccharomyces cerevisiae Proteins/chemistry , Signal Transduction , Transcription Factors/chemistry , Transcription, Genetic
4.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 9): 1120-4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26323295

ABSTRACT

The potentially structured core domain of the intrinsically disordered protein Knr4 from Saccharomyces cerevisiae, comprising residues 80-340, was expressed in Escherichia coli and crystallized using the hanging-drop vapour-diffusion method. Selenomethionine-containing (SeMet) protein was also purified and crystallized. Crystals of both proteins belonged to space group P6522, with unit-cell parameters a = b = 112.44, c = 265.21 Šfor the native protein and a = b = 112.49, c = 262.21 Šfor the SeMet protein, and diffracted to 3.50 and 3.60 Šresolution, respectively. There are two molecules in the asymmetric unit related by a twofold axis. The anomalous signal of selenium was recorded and yielded an electron-density map of sufficient quality to allow the identification of secondary-structure elements.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Transcription Factors/chemistry , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Static Electricity , Ultraviolet Rays
5.
PLoS One ; 9(5): e97148, 2014.
Article in English | MEDLINE | ID: mdl-24817274

ABSTRACT

Understanding the mechanism that controls space-time coordination of elongation and division of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is critical for fighting the tubercle bacillus. Most of the numerous enzymes involved in the synthesis of Mycolic acid - Arabinogalactan-Peptidoglycan complex (MAPc) in the cell wall are essential in vivo. Using a dynamic approach, we localized Mtb enzymes belonging to the fatty acid synthase-II (FAS-II) complexes and involved in mycolic acid (MA) biosynthesis in a mycobacterial model of Mtb: M. smegmatis. Results also showed that the MA transporter MmpL3 was present in the mycobacterial envelope and was specifically and dynamically accumulated at the poles and septa during bacterial growth. This localization was due to its C-terminal domain. Moreover, the FAS-II enzymes were co-localized at the poles and septum with Wag31, the protein responsible for the polar localization of mycobacterial peptidoglycan biosynthesis. The dynamic localization of FAS-II and of the MA transporter with Wag31, at the old-growing poles and at the septum suggests that the main components of the mycomembrane may potentially be synthesized at these precise foci. This finding highlights a major difference between mycobacteria and other rod-shaped bacteria studied to date. Based on the already known polar activities of envelope biosynthesis in mycobacteria, we propose the existence of complex polar machinery devoted to the biogenesis of the entire envelope. As a result, the mycobacterial pole would represent the Achilles' heel of the bacillus at all its growing stages.


Subject(s)
Bacterial Proteins/metabolism , Biosynthetic Pathways/physiology , Cell Growth Processes/physiology , Multiprotein Complexes/biosynthesis , Mycobacterium tuberculosis/physiology , Mycolic Acids/metabolism , Fatty Acid Synthase, Type II/metabolism , Galactans/metabolism , Gene Knockout Techniques , Green Fluorescent Proteins , Microscopy, Video , Molecular Structure , Multiprotein Complexes/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Peptidoglycan/metabolism , Spindle Poles/metabolism
6.
PLoS One ; 6(12): e29564, 2011.
Article in English | MEDLINE | ID: mdl-22216317

ABSTRACT

BACKGROUND: The human pathogen Mycobacterium tuberculosis (Mtb) has the originality of possessing a multifunctional mega-enzyme FAS-I (Fatty Acid Synthase-I), together with a multi-protein FAS-II system, to carry out the biosynthesis of common and of specific long chain fatty acids: the mycolic acids (MA). MA are the main constituents of the external mycomembrane that represents a tight permeability barrier involved in the pathogenicity of Mtb. The MA biosynthesis pathway is essential and contains targets for efficient antibiotics. We have demonstrated previously that proteins of FAS-II interact specifically to form specialized and interconnected complexes. This finding suggested that the organization of FAS-II resemble to the architecture of multifunctional mega-enzyme like the mammalian mFAS-I, which is devoted to the fatty acid biosynthesis. PRINCIPAL FINDINGS: Based on conventional and reliable studies using yeast-two hybrid, yeast-three-hybrid and in vitro Co-immunoprecipitation, we completed here the analysis of the composition and architecture of the interactome between the known components of the Mtb FAS-II complexes. We showed that the recently identified dehydratases HadAB and HadBC are part of the FAS-II elongation complexes and may represent a specific link between the core of FAS-II and the condensing enzymes of the system. By testing four additional methyltransferases involved in the biosynthesis of mycolic acids, we demonstrated that they display specific interactions with each type of complexes suggesting their coordinated action during MA elongation. SIGNIFICANCE: These results provide a global update of the architecture and organization of a FAS-II system. The FAS-II system of Mtb is organized in specialized interconnected complexes and the specificity of each elongation complex is given by preferential interactions between condensing enzymes and dehydratase heterodimers. This study will probably allow defining essential and specific interactions that correspond to promising targets for Mtb FAS-II inhibitors.


Subject(s)
Fatty Acid Synthase, Type II/metabolism , Hydro-Lyases/metabolism , Mycobacterium tuberculosis/enzymology , Mycolic Acids/metabolism , Dimerization , Fatty Acid Synthase, Type II/chemistry , Hydro-Lyases/chemistry , Immunoprecipitation
7.
Biochim Biophys Acta ; 1774(2): 303-11, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17240207

ABSTRACT

The (R)-specific 3-hydroxyacyl dehydratases/trans-enoyl hydratases are key proteins in the biosynthesis of fatty acids. In mycobacteria, such enzymes remain unknown, although they are involved in the biosynthesis of major and essential lipids like mycolic acids. First bioinformatic analyses allowed to identify a single candidate protein, namely Rv3389c, that belongs to the hydratases 2 family and is most likely made of a distinctive asymmetric double hot dog fold. The purified recombinant Rv3389c protein was shown to efficiently catalyze the hydration of (C(8)-C(16)) enoyl-CoA substrates. Furthermore, it catalyzed the dehydration of a 3-hydroxyacyl-CoA in coupled reactions with both reductases (MabA and InhA) of the acyl carrier protein (ACP)-dependent M. tuberculosis fatty acid synthase type II involved in mycolic acid biosynthesis. Yet, the facts that Rv3389c activity decreased in the presence of ACP, versus CoA, derivative and that Rv3389c knockout mutant had no visible variation of its fatty acid content suggested the occurrence of additional hydratase/dehydratase candidates. Accordingly, further and detailed bioinformatic analyses led to the identification of other members of the hydratases 2 family in M. tuberculosis.


Subject(s)
Bacterial Proteins/chemistry , Mycobacterium tuberculosis/chemistry , Amino Acid Sequence , Catalysis , Models, Molecular , Molecular Sequence Data , Mycobacterium tuberculosis/enzymology , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
J Mol Biol ; 353(4): 847-58, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16213523

ABSTRACT

Tuberculosis kills about two million people every year and remains one of the leading causes of mortality worldwide. As a result of the increasing antibiotic resistance of Mycobacterium tuberculosis (Mtb) strains, there is an urgent need for new antitubercular drugs. Several efficient antibiotics, including isoniazid, specifically target the fatty acid synthase-II (FAS-II) complex of mycolic acid biosynthesis. We have previously shown that there are protein-protein interactions between the components of FAS-II that are essential for mycobacterial survival. We have now looked at the potential partners of FAS-II, mtFabD, the methyltransferases MmaAs, and Pks13. A combination of yeast two-hybrid and co-immunoprecipitation experiments showed that mtFabD interacts with each beta-ketoacyl-synthase (KasA, KasB and mtFabH) and with the core of FAS-II (InhA and MabA). The methyltransferases have a greater affinity for KasA and KasB than for mtFabH, suggesting that modifications on the meromycolic chains may occur during their elongation. Finally, Pks13, which catalyzes the final Claisen condensation of mycolic acids, interacts specifically with KasB. These data allowed us to determine the architecture of the multiple specialized FAS-II complexes, giving us insights into the organization of the complete mycolic acids biosynthesis. Our studies suggest a new and crucial interaction (KasB-Pks13) as a putative target for peptidomimetic antibiotics.


Subject(s)
Acetyltransferases/metabolism , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/enzymology , Mycolic Acids/metabolism , Protein Interaction Mapping , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Bacterial Proteins/metabolism , Fatty Acid Synthase, Type II , Immunoprecipitation , Methyltransferases/metabolism , Mitochondrial Proteins/metabolism , Polyketide Synthases/metabolism , Protein Binding , Two-Hybrid System Techniques
9.
Microbiology (Reading) ; 151(Pt 6): 2075-2086, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15942014

ABSTRACT

Mycobacterium smegmatis is often used as a surrogate host for pathogenic mycobacteria, especially since the isolation of the transformable smooth morphotype strain mc(2)155 from the isogenic rough wild-type strain ATCC 607. Biochemical analysis of the cell envelope components revealed a lack of polar glycolipids, namely the lipooligosaccharides and the polar subfamilies of glycopeptidolipids, in the mc(2)155 strain. In addition, the latter strain differs from its parent by the distribution of various species of glycolipids and phospholipids between the outermost and deeper layers of the cell envelope. The presence of filamentous and rope-like structures at the cell surface of mc(2)155 cells grown in complex media further supported an ultrastructural change in the cell envelope of the mutant. Importantly, a significantly more rapid uptake of the hydrophobic chenodeoxycholate was observed for the mutant compared to wild-type cells. Taken together, these data indicate that the nature of the surface-exposed and envelope constituents is crucial for the surface properties, cell wall permeability and bacterial phenotype, and suggest that the transformable character of the mc(2)155 strain may be in part explained by these profound modifications of its cell envelope.


Subject(s)
Cell Membrane/chemistry , Cell Wall/chemistry , Mycobacterium smegmatis/chemistry , Mycobacterium smegmatis/genetics , Biological Transport , Cell Wall/ultrastructure , Chenodeoxycholic Acid/metabolism , Chromatography, Thin Layer , Glycolipids/analysis , Glycopeptides/analysis , Lipopolysaccharides/analysis , Microscopy, Electron, Transmission , Mycobacterium smegmatis/ultrastructure , Permeability , Phospholipids/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Mol Microbiol ; 54(5): 1161-72, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15554959

ABSTRACT

Despite the existence of efficient chemotherapy, tuberculosis remains a leading cause of mortality worldwide. New drugs are urgently needed to reduce the potential impact of the emergence of multidrug-resistant strains of the causative agent Mycobacterium tuberculosis (Mtb). The front-line antibiotic isoniazid (INH), and several other drugs, target the biosynthesis of mycolic acids and especially the Fatty Acid Synthase-II (FAS-II) elongation system. This biosynthetic pathway is essential and specific for mycobacteria and still represents a valuable system for the search of new anti-tuberculous agents. Several data, in the literature, suggest the existence of protein-protein interactions within the FAS-II system. These interactions themselves might serve as targets for a new generation of drugs directed against Mtb. By using an extensive in vivo yeast two-hybrid approach and in vitro co-immunoprecipitation, we have demonstrated the existence of both homotypic and heterotypic interactions between the known components of FAS-II. The condensing enzymes KasA, KasB and mtFabH interact with each other and with the reductases MabA and InhA. Furthermore, we have designed and constructed point mutations of the FAS-II reductase MabA, able to disrupt its homotypic interactions and perturb the interaction pattern of this protein within FAS-II. Finally, we showed by a transdominant genetic approach that these mutants are dominant negative in both non-pathogenic and pathogenic mycobacteria. These data allowed us to draw a dynamic model of the organization of FAS-II. They also represent an important step towards the design of a new generation of anti-tuberculous agents, as being inhibitors of essential protein-protein interactions.


Subject(s)
Acetyltransferases/metabolism , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/enzymology , Protein Interaction Mapping , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acetyltransferases/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fatty Acid Synthase, Type II , Genetic Complementation Test , Immunoprecipitation , Models, Molecular , Multienzyme Complexes/genetics , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/physiology , Oxidoreductases/genetics , Oxidoreductases/metabolism , Point Mutation , Protein Binding , Two-Hybrid System Techniques
11.
Antimicrob Agents Chemother ; 48(1): 242-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14693546

ABSTRACT

The first-line specific antituberculous drug isoniazid inhibits the fatty acid elongation system (FAS) FAS-II involved in the biosynthesis of mycolic acids, which are major lipids of the mycobacterial envelope. The MabA protein that catalyzes the second step of the FAS-II elongation cycle is structurally and functionally related to the in vivo target of isoniazid, InhA, an NADH-dependent enoyl-acyl carrier protein reductase. The present work shows that the NADPH-dependent beta-ketoacyl reduction activity of MabA is efficiently inhibited by isoniazid in vitro by a mechanism similar to that by which isoniazid inhibits InhA activity. It involves the formation of a covalent adduct between Mn(III)-activated isoniazid and the MabA cofactor. Liquid chromatography-mass spectrometry analyses revealed that the isonicotinoyl-NADP adduct has multiple chemical forms in dynamic equilibrium. Both kinetic experiments with isolated forms and purification of the enzyme-ligand complex strongly suggested that the molecules active against MabA activity are the oxidized derivative and a major cyclic form. Spectrofluorimetry showed that the adduct binds to the MabA active site. Modeling of the MabA-adduct complex predicted an interaction between the isonicotinoyl moiety of the inhibitor and Tyr185. This hypothesis was supported by the fact that a higher 50% inhibitory concentration of the adduct was measured for MabA Y185L than for the wild-type enzyme, while both proteins presented similar affinities for NADP(+). The crystal structure of MabA Y185L that was solved showed that the substitution of Tyr185 induced no significant conformational change. The description of the first inhibitor of the beta-ketoacyl reduction step of fatty acid biosynthesis should help in the design of new antituberculous drugs efficient against multidrug-resistant tubercle bacilli.


Subject(s)
Antitubercular Agents/pharmacology , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Acyl Carrier Protein/genetics , Acyl Carrier Protein/metabolism , Chromatography, High Pressure Liquid , Chromatography, Liquid , Crystallography, X-Ray , Fatty Acids/metabolism , Models, Molecular , Mycobacterium tuberculosis/genetics , NADP/metabolism , Protein Conformation , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization
12.
J Mol Biol ; 320(2): 249-61, 2002 Jul 05.
Article in English | MEDLINE | ID: mdl-12079383

ABSTRACT

The fatty acid elongation system FAS-II is involved in the biosynthesis of mycolic acids, which are major and specific long-chain fatty acids of the cell envelope of Mycobacterium tuberculosis and other mycobacteria, including Mycobacterium smegmatis. The protein MabA, also named FabG1, has been shown recently to be part of FAS-II and to catalyse the NADPH-specific reduction of long chain beta-ketoacyl derivatives. This activity corresponds to the second step of an FAS-II elongation round. FAS-II is inhibited by the antituberculous drug isoniazid through the inhibition of the 2-trans-enoyl-acyl carrier protein reductase InhA. Thus, the other enzymes making up this enzymatic complex represent potential targets for designing new antituberculous drugs. The crystal structure of the apo-form MabA was solved to 2.03 A resolution by molecular replacement. MabA is tetrameric and shares the conserved fold of the short-chain dehydrogenases/reductases (SDRs). However, it exhibits some significant local rearrangements of the active-site loops in the absence of a cofactor, particularly the beta5-alpha5 region carrying the unique tryptophan residue, in agreement with previous fluorescence spectroscopy data. A similar conformation has been observed in the beta-ketoacyl reductase from Escherichia coli and the distantly related dehydratase. The distinctive enzymatic and structural properties of MabA are discussed in view of its crystal structure and that of related enzymes.


Subject(s)
Alcohol Oxidoreductases/chemistry , Fatty Acids/biosynthesis , Mycobacterium tuberculosis/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase , Amino Acid Sequence , Binding Sites , Cesium/chemistry , Cross-Linking Reagents/pharmacology , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Escherichia coli/enzymology , Kinetics , Light , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Conformation , Protein Folding , Scattering, Radiation , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...