Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 868
Filtrar
1.
Protein Sci ; 30(12): 2418-2432, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34651380

RESUMO

N-acetylated sugars are often found, for example, on the lipopolysaccharides of Gram-negative bacteria, on the S-layers of Gram-positive bacteria, and on the capsular polysaccharides. Key enzymes involved in their biosynthesis are the sugar N-acetyltransferases. Here, we describe a structural and functional analysis of one such enzyme from Helicobacter pullorum, an emerging pathogen that may be associated with gastroenteritis and gallbladder and liver diseases. For this analysis, the gene BA919-RS02330 putatively encoding an N-acetyltransferase was cloned, and the corresponding protein was expressed and purified. A kinetic analysis demonstrated that the enzyme utilizes dTDP-3-amino-3,6-dideoxy-d-glucose as a substrate as well as dTDP-3-amino-3,6-dideoxy-d-galactose, albeit at a reduced rate. In addition to this kinetic analysis, a similar enzyme from Helicobacter bilis was cloned and expressed, and its kinetic parameters were determined. Seven X-ray crystallographic structures of various complexes of the H. pullorum wild-type enzyme (or the C80T variant) were determined to resolutions of 1.7 Å or higher. The overall molecular architecture of the H. pullorum N-acetyltransferase places it into the Class II left-handed-ß-helix superfamily (LßH). Taken together, the data presented herein suggest that 3-acetamido-3,6-dideoxy-d-glucose (or the galactose derivative) is found on either the H. pullorum O-antigen or in another of its complex glycoconjugates. A BLAST search suggests that more than 50 non-pylori Helicobacter spp. have genes encoding N-acetyltransferases. Given that there is little information concerning the complex glycans in non-pylori Helicobacter spp. and considering their zoonotic potential, our results provide new biochemical insight into these pathogens.


Assuntos
Acetiltransferases/química , Proteínas de Bactérias/química , Desoxiaçúcares/química , Helicobacter/enzimologia , Lipopolissacarídeos/química , Nucleotídeos de Timina/química , Acetiltransferases/genética , Acetiltransferases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X , Desoxiaçúcares/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Glicoconjugados/química , Glicoconjugados/metabolismo , Helicobacter/química , Cinética , Lipopolissacarídeos/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Nucleotídeos de Timina/metabolismo
2.
Int J Mol Sci ; 22(11)2021 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-34073255

RESUMO

Whole genome sequences of two Acinetobacter baumannii clinical isolates, 48-1789 and MAR24, revealed that they carry the KL106 and KL112 capsular polysaccharide (CPS) biosynthesis gene clusters, respectively, at the chromosomal K locus. The KL106 and KL112 gene clusters are related to the previously described KL11 and KL83 gene clusters, sharing genes for the synthesis of l-rhamnose (l-Rhap) and 6-deoxy-l-talose (l-6dTalp). CPS material isolated from 48-1789 and MAR24 was studied by sugar analysis and Smith degradation along with one- and two-dimensional 1H and 13C NMR spectroscopy. The structures of K106 and K112 oligosaccharide repeats (K units) l-6dTalp-(1→3)-D-GlcpNAc tetrasaccharide fragment share the responsible genes in the respective gene clusters. The K106 and K83 CPSs also have the same linkage between K units. The KL112 cluster includes an additional glycosyltransferase gene, Gtr183, and the K112 unit includes α l-Rhap side chain that is not found in the K106 structure. K112 further differs in the linkage between K units formed by the Wzy polymerase, and a different wzy gene is found in KL112. However, though both KL106 and KL112 share the atr8 acetyltransferase gene with KL83, only K83 is acetylated.


Assuntos
Acinetobacter baumannii , Desoxiaçúcares , Hexoses , Polissacarídeos Bacterianos , Acinetobacter baumannii/química , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Desoxiaçúcares/química , Desoxiaçúcares/genética , Desoxiaçúcares/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Hexoses/química , Hexoses/genética , Hexoses/metabolismo , Polissacarídeos Bacterianos/química , Polissacarídeos Bacterianos/genética , Polissacarídeos Bacterianos/metabolismo , Especificidade da Espécie
3.
Biochem Biophys Res Commun ; 545: 112-118, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33548623

RESUMO

We previously reported the alginate lyase, SjAly, from a brown alga, Saccharina japonica, providing the first experimental evidence for a functional alginate-degradation enzyme in brown algae. 4-deoxy-L-erythro-5-hexoseulose uronate (DEHU), derived from an unsaturated monosaccharide, was identified as the minimum degradation product produced by SjAly-mediated lysis of alginate. DEHU was hitherto reported to be reduced to 2-keto-3-deoxy-gluconate (KDG) by a DEHU-specific reductase with NAD(P)H in alginate-assimilating organisms and its metabolism in alginate-producing organisms is unknown. Here, we report the functional identification of a DEHU reductase, SjRed, in S. japonica. Among the 14 tested compounds, only DEHU was used as a substrate and was converted to KDG in the presence of NADPH. Optimum temperature, pH, and KCl concentration required for SjRed activity were determined to be 25 °C, 7.2, and 100 mM, respectively. SjRed consists of 341 amino acid residues and is proposed to be a member of the aldo-keto reductase superfamily. Sequencing of SjRed revealed that it is composed of at least three exons. These results indicate the existence of an enzyme that reduces DEHU to KDG in S. japonica. This is the first report on the functional identification of a DEHU-reductase in alginate-producing organisms.


Assuntos
Aldo-Ceto Redutases/metabolismo , Proteínas de Algas/metabolismo , Alginatos/metabolismo , Phaeophyceae/enzimologia , Aldo-Ceto Redutases/química , Aldo-Ceto Redutases/genética , Proteínas de Algas/química , Proteínas de Algas/genética , Sequência de Aminoácidos , Desoxiaçúcares/metabolismo , Ácidos Hexurônicos/metabolismo , Phaeophyceae/genética , Polissacarídeo-Liases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
4.
mBio ; 11(2)2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32345640

RESUMO

All enterococci produce a complex polysaccharide called the enterococcal polysaccharide antigen (EPA). This polymer is required for normal cell growth and division and for resistance to cephalosporins and plays a critical role in host-pathogen interaction. The EPA contributes to host colonization and is essential for virulence, conferring resistance to phagocytosis during the infection. Recent studies revealed that the "decorations" of the EPA polymer, encoded by genetic loci that are variable between isolates, underpin the biological activity of this surface polysaccharide. In this work, we investigated the structure of the EPA polymer produced by the high-risk enterococcal clonal complex Enterococcus faecalis V583. We analyzed purified EPA from the wild-type strain and a mutant lacking decorations and elucidated the structure of the EPA backbone and decorations. We showed that the rhamnan backbone of EPA is composed of a hexasaccharide repeat unit of C2- and C3-linked rhamnan chains, partially substituted in the C3 position by α-glucose (α-Glc) and in the C2 position by ß-N-acetylglucosamine (ß-GlcNAc). The so-called "EPA decorations" consist of phosphopolysaccharide chains corresponding to teichoic acids covalently bound to the rhamnan backbone. The elucidation of the complete EPA structure allowed us to propose a biosynthetic pathway, a first essential step toward the design of antimicrobials targeting the synthesis of this virulence factor.IMPORTANCE Enterococci are opportunistic pathogens responsible for hospital- and community-acquired infections. All enterococci produce a surface polysaccharide called EPA (enterococcal polysaccharide antigen) required for biofilm formation, antibiotic resistance, and pathogenesis. Despite the critical role of EPA in cell growth and division and as a major virulence factor, no information is available on its structure. Here, we report the complete structure of the EPA polymer produced by the model strain E. faecalis V583. We describe the structure of the EPA backbone, made of a rhamnan hexasaccharide substituted by Glc and GlcNAc residues, and show that teichoic acids are covalently bound to this rhamnan chain, forming the so-called "EPA decorations" essential for host colonization and pathogenesis. This report represents a key step in efforts to identify the structural properties of EPA that are essential for its biological activity and to identify novel targets to develop preventive and therapeutic approaches against enterococci.


Assuntos
Antígenos de Bactérias/química , Enterococcus faecalis/metabolismo , Polissacarídeos/química , Antígenos de Bactérias/metabolismo , Desoxiaçúcares/química , Desoxiaçúcares/metabolismo , Humanos , Mananas/química , Mananas/metabolismo , Polissacarídeos/metabolismo , Ácidos Teicoicos/química , Ácidos Teicoicos/metabolismo , Enterococos Resistentes à Vancomicina/metabolismo
5.
J Biol Chem ; 295(16): 5519-5532, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32169901

RESUMO

Extracytoplasmic sugar decoration of glycopolymer components of the bacterial cell wall contributes to their structural diversity. Typically, the molecular mechanism that underpins such a decoration process involves a three-component glycosylation system (TGS) represented by an undecaprenyl-phosphate (Und-P) sugar-activating glycosyltransferase (Und-P GT), a flippase, and a polytopic glycosyltransferase (PolM GT) dedicated to attaching sugar residues to a specific glycopolymer. Here, using bioinformatic analyses, CRISPR-assisted recombineering, structural analysis of cell wall-associated polysaccharides (CWPS) through MALDI-TOF MS and methylation analysis, we report on three such systems in the bacterium Lactococcus lactis On the basis of sequence similarities, we first identified three gene pairs, csdAB, csdCD, and csdEF, each encoding an Und-P GT and a PolM GT, as potential TGS component candidates. Our experimental results show that csdAB and csdCD are involved in Glc side-chain addition on the CWPS components rhamnan and polysaccharide pellicle (PSP), respectively, whereas csdEF plays a role in galactosylation of lipoteichoic acid (LTA). We also identified a potential flippase encoded in the L. lactis genome (llnz_02975, cflA) and confirmed that it participates in the glycosylation of the three cell wall glycopolymers rhamnan, PSP, and LTA, thus indicating that its function is shared by the three TGSs. Finally, we observed that glucosylation of both rhamnan and PSP can increase resistance to bacteriophage predation and that LTA galactosylation alters L. lactis resistance to bacteriocin.


Assuntos
Parede Celular/metabolismo , Lactococcus lactis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Desoxiaçúcares/metabolismo , Galactose/metabolismo , Glicosilação , Lactococcus lactis/genética , Lipopolissacarídeos/metabolismo , Mananas/metabolismo , Ácidos Teicoicos/metabolismo
6.
J Biol Chem ; 295(5): 1338-1349, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31914410

RESUMO

The genetic context in bacterial genomes and screening for potential substrates can help identify the biochemical functions of bacterial enzymes. The Gram-negative, strictly anaerobic bacterium Veillonella ratti possesses a gene cluster that appears to be related to l-fucose metabolism and contains a putative dihydrodipicolinate synthase/N-acetylneuraminate lyase protein (FucH). Here, screening of a library of 2-keto-3-deoxysugar acids with this protein and biochemical characterization of neighboring genes revealed that this gene cluster encodes enzymes in a previously unknown "route I" nonphosphorylating l-fucose pathway. Previous studies of other aldolases in the dihydrodipicolinate synthase/N-acetylneuraminate lyase protein superfamily used only limited numbers of compounds, and the approach reported here enabled elucidation of the substrate specificities and stereochemical selectivities of these aldolases and comparison of them with those of FucH. According to the aldol cleavage reaction, the aldolases were specific for (R)- and (S)-stereospecific groups at the C4 position of 2-keto-3-deoxysugar acid but had no structural specificity or preference of methyl groups at the C5 and C6 positions, respectively. This categorization corresponded to the (Re)- or (Si)-facial selectivity of the pyruvate enamine on the (glycer)aldehyde carbonyl in the aldol-condensation reaction. These properties are commonly determined by whether a serine or threonine residue is positioned at the equivalent position close to the active site(s), and site-directed mutagenesis markedly modified C4-OH preference and selective formation of a diastereomer. I propose that substrate specificity of 2-keto-3-deoxysugar acid aldolases was convergently acquired during evolution and report the discovery of another l-2-keto-3-deoxyfuconate aldolase involved in the same nonphosphorylating l-fucose pathway in Campylobacter jejuni.


Assuntos
Aldeído Liases/metabolismo , Aldeídos/metabolismo , Fucose/metabolismo , Veillonella/enzimologia , Aldeído Liases/química , Aldeído Liases/genética , Aldeídos/química , Sequência de Aminoácidos/genética , Sítios de Ligação/genética , Campylobacter jejuni/enzimologia , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Domínio Catalítico/genética , Desoxiaçúcares/química , Desoxiaçúcares/metabolismo , Evolução Molecular , Hidroliases/química , Hidroliases/metabolismo , Cinética , Modelos Moleculares , Família Multigênica/genética , Mutagênese Sítio-Dirigida , Mutação , Oxo-Ácido-Liases/química , Oxo-Ácido-Liases/metabolismo , Filogenia , Especificidade por Substrato/genética , Veillonella/genética , Veillonella/metabolismo
7.
J Biol Chem ; 295(5): 1225-1239, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31819007

RESUMO

Glycan biosynthesis relies on nucleotide sugars (NSs), abundant metabolites that serve as monosaccharide donors for glycosyltransferases. In vivo, signal-dependent fluctuations in NS levels are required to maintain normal cell physiology and are dysregulated in disease. However, how mammalian cells regulate NS levels and pathway flux remains largely uncharacterized. To address this knowledge gap, here we examined UDP-galactose 4'-epimerase (GALE), which interconverts two pairs of essential NSs. Using immunoblotting, flow cytometry, and LC-MS-based glycolipid and glycan profiling, we found that CRISPR/Cas9-mediated GALE deletion in human cells triggers major imbalances in NSs and dramatic changes in glycolipids and glycoproteins, including a subset of integrins and the cell-surface death receptor FS-7-associated surface antigen. In particular, we observed substantial decreases in total sialic acid, galactose, and GalNAc levels in glycans. These changes also directly impacted cell signaling, as GALE-/- cells exhibited FS-7-associated surface antigen ligand-induced apoptosis. Our results reveal a role of GALE-mediated NS regulation in death receptor signaling and may have implications for the molecular etiology of illnesses characterized by NS imbalances, including galactosemia and metabolic syndrome.


Assuntos
Glicolipídeos/metabolismo , Glicoproteínas/metabolismo , Açúcares/metabolismo , UDPglucose 4-Epimerase/química , UDPglucose 4-Epimerase/metabolismo , Receptor fas/metabolismo , Apoptose/genética , Cromatografia Líquida , Desoxiaçúcares/metabolismo , Técnicas de Inativação de Genes , Glicolipídeos/biossíntese , Glicolipídeos/química , Glicoproteínas/biossíntese , Glicoproteínas/química , Glicosilação , Células HEK293 , Células HeLa , Humanos , Espectrometria de Massas , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/química , Polissacarídeos/metabolismo , Receptores de Superfície Celular/metabolismo , UDPglucose 4-Epimerase/genética , Receptor fas/química
8.
J Biol Chem ; 294(46): 17612-17625, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31582566

RESUMO

In Lactococcus lactis, cell-wall polysaccharides (CWPSs) act as receptors for many bacteriophages, and their structural diversity among strains explains, at least partially, the narrow host range of these viral predators. Previous studies have reported that lactococcal CWPS consists of two distinct components, a variable chain exposed at the bacterial surface, named polysaccharide pellicle (PSP), and a more conserved rhamnan chain anchored to, and embedded inside, peptidoglycan. These two chains appear to be covalently linked to form a large heteropolysaccharide. The molecular machinery for biosynthesis of both components is encoded by a large gene cluster, named cwps In this study, using a CRISPR/Cas-based method, we performed a mutational analysis of the cwps genes. MALDI-TOF MS-based structural analysis of the mutant CWPS combined with sequence homology, transmission EM, and phage sensitivity analyses enabled us to infer a role for each protein encoded by the cwps cluster. We propose a comprehensive CWPS biosynthesis scheme in which the rhamnan and PSP chains are independently synthesized from two distinct lipid-sugar precursors and are joined at the extracellular side of the cytoplasmic membrane by a mechanism involving a membrane-embedded glycosyltransferase with a GT-C fold. The proposed scheme encompasses a system that allows extracytoplasmic modification of rhamnan by complex substituting oligo-/polysaccharides. It accounts for the extensive diversity of CWPS structures observed among lactococci and may also have relevance to the biosynthesis of complex rhamnose-containing CWPSs in other Gram-positive bacteria.


Assuntos
Parede Celular/metabolismo , Lactococcus lactis/metabolismo , Polissacarídeos Bacterianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Vias Biossintéticas , Parede Celular/química , Parede Celular/genética , Desoxiaçúcares/análise , Desoxiaçúcares/genética , Desoxiaçúcares/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Lactococcus lactis/química , Lactococcus lactis/genética , Mananas/análise , Mananas/genética , Mananas/metabolismo , Família Multigênica , Polissacarídeos Bacterianos/análise , Polissacarídeos Bacterianos/genética
9.
Appl Environ Microbiol ; 84(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29247057

RESUMO

The earthworm gut is an anoxic, saccharide-rich microzone in aerated soils. The apparent degradation of diverse saccharides in the alimentary canal of the model earthworm Lumbricusterrestris is concomitant with the production of diverse organic acids, indicating that fermentation is an ongoing process in the earthworm gut. However, little is known about how different gut-associated saccharides are fermented. The hypothesis of this investigation was that different gut-associated saccharides differentially stimulate fermentative microorganisms in gut contents of L. terrestris This hypothesis was addressed by (i) assessing the fermentation profiles of anoxic gut content microcosms that were supplemented with gut-associated saccharides and (ii) the concomitant phylogenic analysis of 16S rRNA sequences. Galactose, glucose, maltose, mannose, arabinose, fucose, rhamnose, and xylose stimulated the production of fermentation products, including H2, CO2, acetate, lactate, propionate, formate, succinate, and ethanol. Fermentation profiles were dependent on the supplemental saccharide (e.g., glucose yielded large amounts of H2 and ethanol, whereas fucose did not, and maltose yielded large amounts of lactate, whereas mannose did not). Approximately 1,750,000 16S rRNA sequences were affiliated with 37 families, and phylogenic analyses indicated that a respective saccharide stimulated a subset of the diverse phylotypes. An Aeromonas-related phylotype displayed a high relative abundance in all treatments, whereas key Enterobacteriaceae-affiliated phylotypes were stimulated by some but not all saccharides. Collectively, these results reinforce the likelihood that (i) different saccharides stimulate different fermentations in gut contents of the earthworm and (ii) facultative aerobes related to Aeromonadaceae and Enterobacteriaceae can be important drivers of these fermentations.IMPORTANCE The feeding habits of earthworms influence the turnover of elements in the terrestrial biosphere. The alimentary tract of the earthworm constitutes an anoxic saccharide-rich microzone in aerated soils that offers ingested microbes a unique opportunity for anaerobic growth. The fermentative activity of microbes in the alimentary tract are responsible for the in situ production of (i) organic compounds that can be assimilated by the earthworm and (ii) H2 that is subject to in vivo emission by the earthworm and can be trophically linked to secondary microbial events in soils. To gain insight on how fermentative members of the gut microbiome might respond to the saccharide-rich alimentary canal, this study examines the impact of diverse gut-associated saccharides on the differential activation of fermentative microbes in gut contents of the model earthworm L. terrestris.


Assuntos
Bactérias/metabolismo , Desoxiaçúcares/metabolismo , Dissacarídeos/metabolismo , Microbioma Gastrointestinal , Monossacarídeos/metabolismo , Oligoquetos/microbiologia , Animais , Bactérias/classificação , Bactérias/isolamento & purificação , Fermentação , Oligoquetos/fisiologia , RNA Bacteriano/análise , RNA Ribossômico 16S/análise
10.
Biochemistry ; 56(29): 3818-3825, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28665588

RESUMO

The causative agent of tuberculosis, Mycobacterium tuberculosis, is a bacterium with a complex cell wall and a complicated life cycle. The genome of M. tuberculosis contains well over 4000 genes thought to encode proteins. One of these codes for a putative enzyme referred to as Rv3404c, which has attracted research attention as a potential virulence factor for over 12 years. Here we demonstrate that Rv3404c functions as a sugar N-formyltransferase that converts dTDP-4-amino-4,6-dideoxyglucose into dTDP-4-formamido-4,6-dideoxyglucose using N10-formyltetrahydrofolate as the carbon source. Kinetic analyses demonstrate that Rv3404c displays a significant catalytic efficiency of 1.1 × 104 M-1 s-1. In addition, we report the X-ray structure of a ternary complex of Rv3404c solved in the presence of N5-formyltetrahydrofolate and dTDP-4-amino-4,6-dideoxyglucose. The final model of Rv3404c was refined to an overall R-factor of 16.8% at 1.6 Å resolution. The results described herein are especially intriguing given that there have been no published reports of N-formylated sugars associated with M. tuberculosis. The data thus provide a new avenue of research into this fascinating, yet deadly, organism that apparently has been associated with human infection since ancient times.


Assuntos
Proteínas de Bactérias/química , Hidroximetil e Formil Transferases/química , Modelos Moleculares , Mycobacterium tuberculosis/enzimologia , Fatores de Virulência/química , Proteínas de Bactérias/metabolismo , Catálise , Cristalografia por Raios X , Desoxiaçúcares/química , Desoxiaçúcares/metabolismo , Formiltetra-Hidrofolatos/química , Formiltetra-Hidrofolatos/metabolismo , Hidroximetil e Formil Transferases/metabolismo , Cinética , Mycobacterium tuberculosis/patogenicidade , Nucleotídeos de Timina/química , Nucleotídeos de Timina/metabolismo , Fatores de Virulência/metabolismo
11.
Int J Biol Macromol ; 95: 1280-1288, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27838420

RESUMO

Thiothrix strains are filamentous sulfur-oxidizing bacteria common in activated sludge. Some of the members, including Thiothrix nivea and T. fructosivorans, are known to form a microtubular sheath that covers a line of cells. The sheaths are assemblages of [→4)-ß-d-GlcN-(1→4)-ß-d-Glc-(1→]n modified with unusual deoxy sugars. In an attempt to elucidate the sheath-forming mechanism, the patterns of sheath formation and cell proliferation were determined in this study. Prior to analysis, both sheaths were confirmed to be highly de-N-acetylated. Sheaths in viable filaments were N-biotinylated followed by cultivation and then fluorescently immunostained. Epifluorescence microscopy of the filaments revealed ubiquitous elongation of the sheaths. For visualization of the cell proliferation pattern, the cell membrane was fluorescently stained. The epifluorescence images demonstrated that cell proliferation also proceeds ubiquitously, suggesting that sheath elongation proceeds surrounding an elongating cell. In addition, the fine structure of the Thiothrix filaments was analyzed by transmission electron microscopy employing a freeze-substitution technique. The micrographs of freeze-substituted filaments showed that the sheaths were thin and single layered. In contrast, the sheaths in chemically fixed filaments appeared thick and multilayered. Treatment with glutaraldehyde probably caused deformation of the sheaths. Supporting this possibility, the sheaths were found to be deformed or solubilized by N-acetylation.


Assuntos
Biofilmes/crescimento & desenvolvimento , Esgotos/microbiologia , Thiothrix/química , Acetilação , Sequência de Carboidratos , Reagentes de Ligações Cruzadas/química , Desoxiaçúcares/química , Desoxiaçúcares/metabolismo , Corantes Fluorescentes/química , Glutaral/química , Humanos , Hidrólise , Coloração e Rotulagem/métodos , Propriedades de Superfície , Thiothrix/crescimento & desenvolvimento , Thiothrix/ultraestrutura , Eliminação de Resíduos Líquidos
12.
Methods Mol Biol ; 1512: 51-63, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27885598

RESUMO

Despite many years of research into bacterial chemotaxis, the only well characterized system to date is that of E. coli. Even for E. coli, the direct ligand binding had been fully characterized only for aspartate and serene receptors Tar and Tsr. In 30 years since, no other direct receptor-ligand interaction had been described for bacteria, until the characterization of the C. jejuni aspartate and multiligand receptors (Hartley-Tassell et al. Mol Microbiol 75:710-730, 2010). While signal transduction components of many sensory pathways have now been characterized, ligand-receptor interactions remain elusive due to paucity of high-throughput screening methods. Here, we describe the use of microarray screening we developed to identify ligands, surface plasmon resonance, and saturation transfer difference nuclear magnetic resonance (STD-NMR) we used to verify the hits and to determine the affinity constants of the interactions, allowing for more targeted verification of ligands with traditional chemotaxis and in vivo assays described in Chapter 13 .


Assuntos
Proteínas de Bactérias/metabolismo , Campylobacter jejuni/genética , Escherichia coli/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Receptores de Superfície Celular/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Proteínas de Bactérias/genética , Campylobacter jejuni/metabolismo , Quimiotaxia/genética , Desoxiaçúcares/metabolismo , Desoxiaçúcares/farmacologia , Escherichia coli/genética , Escherichia coli/metabolismo , Dispositivos Lab-On-A-Chip , Ligantes , Espectroscopia de Ressonância Magnética , Procedimentos Analíticos em Microchip , Ligação Proteica , Receptores de Superfície Celular/genética , Ribose/metabolismo , Ribose/farmacologia , Bibliotecas de Moléculas Pequenas/metabolismo , Ressonância de Plasmônio de Superfície
13.
Appl Microbiol Biotechnol ; 100(24): 10403-10415, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27368739

RESUMO

Pseudomonas cichorii D-tagatose 3-epimerase (PcDTE), which has a broad substrate specificity, efficiently catalyzes the epimerization of not only D-tagatose to D-sorbose but also D-fructose to D-psicose (D-allulose) and also recognizes the deoxy sugars as substrates. In an attempt to elucidate the substrate recognition and catalytic reaction mechanisms of PcDTE for deoxy sugars, the X-ray structures of the PcDTE mutant form with the replacement of Cys66 by Ser (PcDTE_C66S) in complexes with deoxy sugars were determined. These X-ray structures showed that substrate recognition by the enzyme at the 1-, 2-, and 3-positions is responsible for enzymatic activity and that substrate-enzyme interactions at the 4-, 5-, and 6-positions are not essential for the catalytic reaction of the enzyme leading to the broad substrate specificity of PcDTE. They also showed that the epimerization site of 1-deoxy 3-keto D-galactitol is shifted from C3 to C4 and that 1-deoxy sugars may bind to the catalytic site in the inhibitor-binding mode. The hydrophobic groove that acts as an accessible surface for substrate binding is formed through the dimerization of PcDTE. In PcDTE_C66S/deoxy sugar complex structures, bound ligand molecules in both the linear and ring forms were detected in the hydrophobic groove, while bound ligand molecules in the catalytic site were in the linear form. This result suggests that the sugar-ring opening of a substrate may occur in the hydrophobic groove and also that the narrow channel of the passageway to the catalytic site allows a substrate in the linear form to pass through.


Assuntos
Carboidratos Epimerases/química , Carboidratos Epimerases/metabolismo , Desoxiaçúcares/química , Desoxiaçúcares/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
14.
Microb Cell Fact ; 15: 56, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-27001601

RESUMO

BACKGROUND: Streptomyces albus J1074 produces glycosylated antibiotics paulomycin A, B and E that derive from chorismate and contain an isothiocyanate residue in form of paulic acid. Paulomycins biosynthesis pathway involves two glycosyltransferases, three acyltransferases, enzymes required for paulic acid biosynthesis (in particular an aminotransferase and a sulfotransferase), and enzymes involved in the biosynthesis of two deoxysugar moieties: D-allose and L-paulomycose. RESULTS: Inactivation of genes encoding enzymes involved in deoxysugar biosynthesis, paulic acid biosynthesis, deoxysugar transfer, and acyl moieties transfer has allowed the identification of several biosynthetic intermediates and shunt products, derived from paulomycin intermediates, and to propose a refined version of the paulomycin biosynthesis pathway. Furthermore, several novel bioactive derivatives of paulomycins carrying modifications in the L-paulomycose moiety have been generated by combinatorial biosynthesis using different plasmids that direct the biosynthesis of alternative deoxyhexoses. CONCLUSIONS: The paulomycins biosynthesis pathway has been defined by inactivation of genes encoding glycosyltransferases, acyltransferases and enzymes involved in paulic acid and L-paulomycose biosynthesis. These experiments have allowed the assignment of each of these genes to specific paulomycin biosynthesis steps based on characterization of products accumulated by the corresponding mutant strains. In addition, novel derivatives of paulomycin A and B containing L-paulomycose modified moieties were generated by combinatorial biosynthesis. The production of such derivatives shows that L-paulomycosyl glycosyltransferase Plm12 possesses a certain degree of flexibility for the transfer of different deoxysugars. In addition, the pyruvate dehydrogenase system form by Plm8 and Plm9 is also flexible to catalyze the attachment of a two-carbon side chain, derived from pyruvate, into both 2,6-dideoxyhexoses and 2,3,6-trideoxyhexoses. The activity of the novel paulomycin derivatives carrying modifications in the L-paulomycose moiety is lower than the original compounds pointing to some interesting structure-activity relationships.


Assuntos
Antibacterianos/biossíntese , Metabolismo dos Carboidratos/genética , Engenharia Metabólica/métodos , Streptomyces/genética , Streptomyces/metabolismo , Vias Biossintéticas/genética , Cicloexenos , Desoxiaçúcares/metabolismo , Dissacarídeos/biossíntese , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Família Multigênica , Organismos Geneticamente Modificados , Streptomyces/enzimologia
15.
Glycobiology ; 26(5): 493-500, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26703456

RESUMO

Colitose, also known as 3,6-dideoxy-L-galactose or 3-deoxy-L-fucose, is one of only five naturally occurring 3,6-dideoxyhexoses. Colitose was found in lipopolysaccharide of a number of infectious bacteria, including Escherichia coli O55 & O111 and Vibrio cholera O22 & O139. To date, no colitosyltransferase (ColT) has been characterized, probably due to the inaccessibility of the sugar donor, GDP-colitose. In this study, starting with chemically prepared colitose, 94.6 mg of GDP-colitose was prepared via a facile and efficient one-pot two-enzyme system involving an L-fucokinase/GDP-L-Fuc pyrophosphorylase and an inorganic pyrophosphatase (EcPpA). WbgN, a putative ColT from E. coliO55:H5 was then cloned, overexpressed, purified and biochemically characterized by using GDP-colitose as a sugar donor. Activity assay and structural identification of the synthetic product clearly demonstrated that wbgN encodes an α1,2-ColT. Biophysical study showed that WbgN does not require metal ion, and is highly active at pH 7.5-9.0. In addition, acceptor specificity study indicated that WbgN exclusively recognizes lacto-N-biose (Galß1,3-GlcNAc). Most interestingly, it was found that WbgN exhibits similar activity toward GDP-l-Fuc (kcat/Km= 9.2 min(-1)mM(-1)) as that toward GDP-colitose (kcat/Km= 12 min(-1)mM(-1)). Finally, taking advantage of this, type 1 H-antigen was successfully synthesized in preparative scale.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Glucosiltransferases/química , Glucosiltransferases/metabolismo , Desoxiaçúcares/química , Desoxiaçúcares/genética , Desoxiaçúcares/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Glucosiltransferases/genética , Açúcares de Guanosina Difosfato/química , Açúcares de Guanosina Difosfato/genética , Açúcares de Guanosina Difosfato/metabolismo
16.
Biosci Biotechnol Biochem ; 78(2): 317-25, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25036688

RESUMO

6-Deoxy-L-glucose, 6-deoxy-L-altrose, and 6-deoxy-L-allose were produced from L-rhamnose with an immobilized enzyme that was partially purified (IE) and an immobilized Escherichia coli recombinant treated with toluene (TT). 6-Deoxy-L-psicose was produced from L-rhamnose by a combination of L-rhamnose isomerase (TT-PsLRhI) and D-tagatose 3-epimerase (TT-PcDTE). The purified 6-deoxy-L-psicose was isomerized to 6-deoxy-L-altrose and 6-deoxy-L-allose with L-arabinose isomerase (TT-EaLAI) and L-ribose isomerase (TT-AcLRI), respectively, and then was epimerized to L-rhamnulose with immobilized D-tagatose 3-epimerase (IE-PcDTE). Following purification, L-rhamnulose was converted to 6-deoxy-L-glucose with D-arabinose isomerase (TT-BpDAI). The equilibrium ratios of 6-deoxy-L-psicose:6-deoxy-L-altrose, 6-deoxy-L-psicose:6-deoxy-L-allose, and L-rhamnulose:6-deoxy-L-glucose were 60:40, 40:60, and 27:73, respectively. The production yields of 6-deoxy-L-glucose, 6-deoxy-L-altrose, and 6-deoxy-L-allose from L-rhamnose were 5.4, 14.6, and 25.1%, respectively. These results indicate that the aldose isomerases used in this study acted on 6-deoxy aldohexoses.


Assuntos
Desoxiaçúcares/metabolismo , Hexoses/metabolismo , Oxirredutases Intramoleculares/metabolismo , Ramnose/metabolismo , Bactérias/enzimologia , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Oxirredutases Intramoleculares/química
17.
Biotechnol Lett ; 36(9): 1809-18, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24793498

RESUMO

Daunorubicin (DNR) is a representative anthracycline with anti-tumor bioactivity. Its convergent biosynthetic pathway has promoted the research on pursuing novel anthracyclines by combinatorial biosynthesis. SnoaL is a special polyketide cyclase that catalyzes the closure of nogalonic acid methyl ester with the C9-S stereochemistry. In this study, the gene cluster of DNR was cloned, and snoaL was integrated into the DNR biosynthetic pathway for the substitution of dnrD in Streptomyces coeruleobidus DM, which resulted in the production of epi-aklaviketone. The biosynthetic pathway of NDP-4-deacetyl-L-chromose B was then expressed in the engineered strain, which led to the production of corresponding glycosylated anthracycline compounds. Finally, the bioactivities of these engineering strains were evaluated.


Assuntos
Vias Biossintéticas/genética , Daunorrubicina/metabolismo , Desoxiaçúcares/metabolismo , Engenharia Metabólica , Streptomyces/genética , Streptomyces/metabolismo , Biotransformação , Carboidratos/análise , Citosol/química , Família Multigênica
18.
Protein Sci ; 23(6): 683-92, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24616215

RESUMO

Unusual di- and trideoxysugars are often found on the O-antigens of Gram-negative bacteria, on the S-layers of Gram-positive bacteria, and on various natural products. One such sugar is 3-acetamido-3,6-dideoxy-D-glucose. A key step in its biosynthesis, catalyzed by a 3,4-ketoisomerase, is the conversion of thymidine diphosphate (dTDP)-4-keto-6-deoxyglucose to dTDP-3-keto-6-deoxyglucose. Here we report an X-ray analysis of a 3,4-ketoisomerase from Thermoanaerobacterium thermosaccharolyticum. For this investigation, the wild-type enzyme, referred to as QdtA, was crystallized in the presence of dTDP and its structure solved to 2.0-Å resolution. The dimeric enzyme adopts a three-dimensional architecture that is characteristic for proteins belonging to the cupin superfamily. In order to trap the dTDP-4-keto-6-deoxyglucose substrate into the active site, a mutant protein, H51N, was subsequently constructed, and the structure of this protein in complex with the dTDP-sugar ligand was solved to 1.9-Å resolution. Taken together, the structures suggest that His 51 serves as a catalytic base, that Tyr 37 likely functions as a catalytic acid, and that His 53 provides a proton shuttle between the C-3' hydroxyl and the C-4' keto group of the hexose. This study reports the first three-dimensional structure of a 3,4-ketoisomerase in complex with its dTDP-sugar substrate and thus sheds new molecular insight into this fascinating class of enzymes.


Assuntos
Proteínas de Bactérias/química , Carboidratos Epimerases/química , Carboidratos Epimerases/metabolismo , Thermoanaerobacterium/enzimologia , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Desoxiaçúcares/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
19.
Protein Sci ; 23(3): 273-83, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24347283

RESUMO

N-formylated sugars have been observed on the O-antigens of such pathogenic Gram-negative bacteria as Campylobacter jejuni and Francisella tularensis. Until recently, however, little was known regarding the overall molecular architectures of the N-formyltransferases that are required for the biosynthesis of these unusual sugars. Here we demonstrate that the protein encoded by the wbtj gene from F. tularensis is an N-formyltransferase that functions on dTDP-4-amino-4,6-dideoxy-d-glucose as its substrate. The enzyme, hereafter referred to as WbtJ, demonstrates a strict requirement for N(10) -formyltetrahydrofolate as its carbon source. In addition to the kinetic analysis, the three-dimensional structure of the enzyme was solved in the presence of dTDP-sugar ligands to a nominal resolution of 2.1 Å. Each subunit of the dimeric enzyme is dominated by a "core" domain defined by Met 1 to Ser 185. This core motif harbors the active site residues. Following the core domain, the last 56 residues fold into two α-helices and a ß-hairpin motif. The hairpin motif is responsible primarily for the subunit:subunit interface, which is characterized by a rather hydrophobic pocket. From the study presented here, it is now known that WbtJ functions on C-4' amino sugars. Another enzyme recently investigated in the laboratory, WlaRD, formylates only C-3' amino sugars. Strikingly, the quaternary structures of WbtJ and WlaRD are remarkably different. In addition, there are several significant variations in the side chains that line their active site pockets, which may be important for substrate specificity. Details concerning the kinetic and structural properties of WbtJ are presented.


Assuntos
Desoxiaçúcares/metabolismo , Formiltetra-Hidrofolatos/metabolismo , Francisella tularensis/enzimologia , Hidroximetil e Formil Transferases/química , Hidroximetil e Formil Transferases/metabolismo , Nucleotídeos de Timina/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Francisella tularensis/química , Hidroximetil e Formil Transferases/genética , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Especificidade por Substrato
20.
Metab Eng ; 20: 92-100, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24060454

RESUMO

The overall erythromycin biosynthetic pathway can be sub-divided into macrocyclic polyketide formation and polyketide tailoring to produce the final bioactive molecule. In this study, the native deoxysugar tailoring reactions were exchanged for the purpose of demonstrating the production of alternative final erythromycin compounds. Both the d-desosamine and l-mycarose deoxysugar pathways were replaced with the alternative d-mycaminose and d-olivose pathways to produce new erythromycin analogues through the Escherichia coli heterologous system. Both analogues exhibited bioactivity against multiple antibiotic-resistant Bacillus subtilis strains. Besides demonstrating an intrinsic flexibility for the biosynthetic system to accommodate alternative tailoring pathways, the results offer an initial attempt to leverage the E. coli platform for erythromycin analogue production.


Assuntos
Amino Açúcares , Desoxiaçúcares , Eritromicina , Escherichia coli , Glucosamina/análogos & derivados , Amino Açúcares/genética , Amino Açúcares/metabolismo , Bacillus subtilis/enzimologia , Bacillus subtilis/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Desoxiaçúcares/genética , Desoxiaçúcares/metabolismo , Eritromicina/análogos & derivados , Eritromicina/biossíntese , Escherichia coli/enzimologia , Escherichia coli/genética , Glucosamina/genética , Glucosamina/metabolismo , Hexoses , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Streptomyces/enzimologia , Streptomyces/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...