Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 868
Filter
1.
J Biol Chem ; 295(16): 5519-5532, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32169901

ABSTRACT

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.


Subject(s)
Cell Wall/metabolism , Lactococcus lactis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Deoxy Sugars/metabolism , Galactose/metabolism , Glycosylation , Lactococcus lactis/genetics , Lipopolysaccharides/metabolism , Mannans/metabolism , Teichoic Acids/metabolism
2.
J Biol Chem ; 295(5): 1225-1239, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31819007

ABSTRACT

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.


Subject(s)
Glycolipids/metabolism , Glycoproteins/metabolism , Sugars/metabolism , UDPglucose 4-Epimerase/chemistry , UDPglucose 4-Epimerase/metabolism , fas Receptor/metabolism , Apoptosis/genetics , Chromatography, Liquid , Deoxy Sugars/metabolism , Gene Knockout Techniques , Glycolipids/biosynthesis , Glycolipids/chemistry , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Glycosylation , HEK293 Cells , HeLa Cells , Humans , Mass Spectrometry , N-Acetylneuraminic Acid/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Receptors, Cell Surface/metabolism , UDPglucose 4-Epimerase/genetics , fas Receptor/chemistry
3.
J Biol Chem ; 295(5): 1338-1349, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31914410

ABSTRACT

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.


Subject(s)
Aldehyde-Lyases/metabolism , Aldehydes/metabolism , Fucose/metabolism , Veillonella/enzymology , Aldehyde-Lyases/chemistry , Aldehyde-Lyases/genetics , Aldehydes/chemistry , Amino Acid Sequence/genetics , Binding Sites/genetics , Campylobacter jejuni/enzymology , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Catalytic Domain/genetics , Deoxy Sugars/chemistry , Deoxy Sugars/metabolism , Evolution, Molecular , Hydro-Lyases/chemistry , Hydro-Lyases/metabolism , Kinetics , Models, Molecular , Multigene Family/genetics , Mutagenesis, Site-Directed , Mutation , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Phylogeny , Substrate Specificity/genetics , Veillonella/genetics , Veillonella/metabolism
4.
Biochem Biophys Res Commun ; 545: 112-118, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33548623

ABSTRACT

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.


Subject(s)
Aldo-Keto Reductases/metabolism , Algal Proteins/metabolism , Alginates/metabolism , Phaeophyceae/enzymology , Aldo-Keto Reductases/chemistry , Aldo-Keto Reductases/genetics , Algal Proteins/chemistry , Algal Proteins/genetics , Amino Acid Sequence , Deoxy Sugars/metabolism , Hexuronic Acids/metabolism , Phaeophyceae/genetics , Polysaccharide-Lyases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
5.
Int J Mol Sci ; 22(11)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073255

ABSTRACT

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.


Subject(s)
Acinetobacter baumannii , Deoxy Sugars , Hexoses , Polysaccharides, Bacterial , Acinetobacter baumannii/chemistry , Acinetobacter baumannii/genetics , Acinetobacter baumannii/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Deoxy Sugars/chemistry , Deoxy Sugars/genetics , Deoxy Sugars/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Hexoses/chemistry , Hexoses/genetics , Hexoses/metabolism , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/metabolism , Species Specificity
6.
J Biol Chem ; 294(46): 17612-17625, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31582566

ABSTRACT

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.


Subject(s)
Cell Wall/metabolism , Lactococcus lactis/metabolism , Polysaccharides, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways , Cell Wall/chemistry , Cell Wall/genetics , Deoxy Sugars/analysis , Deoxy Sugars/genetics , Deoxy Sugars/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Lactococcus lactis/chemistry , Lactococcus lactis/genetics , Mannans/analysis , Mannans/genetics , Mannans/metabolism , Multigene Family , Polysaccharides, Bacterial/analysis , Polysaccharides, Bacterial/genetics
7.
Appl Environ Microbiol ; 84(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29247057

ABSTRACT

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.


Subject(s)
Bacteria/metabolism , Deoxy Sugars/metabolism , Disaccharides/metabolism , Gastrointestinal Microbiome , Monosaccharides/metabolism , Oligochaeta/microbiology , Animals , Bacteria/classification , Bacteria/isolation & purification , Fermentation , Oligochaeta/physiology , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis
8.
Biochemistry ; 56(29): 3818-3825, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28665588

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Hydroxymethyl and Formyl Transferases/chemistry , Models, Molecular , Mycobacterium tuberculosis/enzymology , Virulence Factors/chemistry , Bacterial Proteins/metabolism , Catalysis , Crystallography, X-Ray , Deoxy Sugars/chemistry , Deoxy Sugars/metabolism , Formyltetrahydrofolates/chemistry , Formyltetrahydrofolates/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Kinetics , Mycobacterium tuberculosis/pathogenicity , Thymine Nucleotides/chemistry , Thymine Nucleotides/metabolism , Virulence Factors/metabolism
9.
Glycobiology ; 26(5): 493-500, 2016 May.
Article in English | MEDLINE | ID: mdl-26703456

ABSTRACT

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.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism , Deoxy Sugars/chemistry , Deoxy Sugars/genetics , Deoxy Sugars/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Glucosyltransferases/genetics , Guanosine Diphosphate Sugars/chemistry , Guanosine Diphosphate Sugars/genetics , Guanosine Diphosphate Sugars/metabolism
10.
Microb Cell Fact ; 15: 56, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-27001601

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Carbohydrate Metabolism/genetics , Metabolic Engineering/methods , Streptomyces/genetics , Streptomyces/metabolism , Biosynthetic Pathways/genetics , Cyclohexenes , Deoxy Sugars/metabolism , Disaccharides/biosynthesis , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Multigene Family , Organisms, Genetically Modified , Streptomyces/enzymology
11.
Appl Microbiol Biotechnol ; 100(24): 10403-10415, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27368739

ABSTRACT

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.


Subject(s)
Carbohydrate Epimerases/chemistry , Carbohydrate Epimerases/metabolism , Deoxy Sugars/chemistry , Deoxy Sugars/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Protein Binding , Protein Conformation , Substrate Specificity
12.
Biosci Biotechnol Biochem ; 78(2): 317-25, 2014.
Article in English | MEDLINE | ID: mdl-25036688

ABSTRACT

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.


Subject(s)
Deoxy Sugars/metabolism , Hexoses/metabolism , Intramolecular Oxidoreductases/metabolism , Rhamnose/metabolism , Bacteria/enzymology , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Intramolecular Oxidoreductases/chemistry
13.
Biotechnol Lett ; 36(9): 1809-18, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24793498

ABSTRACT

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.


Subject(s)
Biosynthetic Pathways/genetics , Daunorubicin/metabolism , Deoxy Sugars/metabolism , Metabolic Engineering , Streptomyces/genetics , Streptomyces/metabolism , Biotransformation , Carbohydrates/analysis , Cytosol/chemistry , Multigene Family
14.
Metab Eng ; 20: 92-100, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24060454

ABSTRACT

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.


Subject(s)
Amino Sugars , Deoxy Sugars , Erythromycin , Escherichia coli , Glucosamine/analogs & derivatives , Amino Sugars/genetics , Amino Sugars/metabolism , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Deoxy Sugars/genetics , Deoxy Sugars/metabolism , Erythromycin/analogs & derivatives , Erythromycin/biosynthesis , Escherichia coli/enzymology , Escherichia coli/genetics , Glucosamine/genetics , Glucosamine/metabolism , Hexoses , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Streptomyces/enzymology , Streptomyces/genetics
15.
Biochemistry ; 51(4): 867-78, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22220494

ABSTRACT

The unusual dideoxy sugar d-anthrose has been identified as an important component in the endospores of infectious agents such as Bacillus anthracis and Bacillus cereus. Specifically, it is the terminal sugar on the bacterium's exosporium, and it provides a point of interaction between the spore and the host. The biosynthesis of d-anthrose involves numerous steps starting from α-d-glucose 1-phosphate. Here we present a combined structural and functional investigation of AntD from B. cereus. This enzyme plays a key role in d-anthrose biosynthesis by catalyzing the acylation of the C-4″ amino group of dTDP-4-amino-4,6-dideoxyglucose using 3-hydroxy-3-methylbutyryl-CoA as the acyl donor. For this investigation, two ternary complexes of AntD were determined to 1.8 Å resolution: one in which the protein contained bound ß-hydroxybutyryl-CoA and dTDP and the second with CoA and dTDP-4-amino-4,6-dideoxyglucose. On the basis of these high-resolution structures, it was shown that the side chain of Asp 94 lies within hydrogen bonding distance of the sugar C-4″ amino group, and the side chain of Ser 84 resides near the carbonyl oxygen of ß-hydroxybutyryl-CoA. To test the roles of these residues in the catalytic mechanism of AntD, various site-directed mutant proteins were prepared and subjected to kinetic and structural analyses. The D94A and D94N mutant proteins demonstrated enzymatic activity, albeit with significantly reduced catalytic efficiencies. The S84A mutant protein showed an approximate 10-fold decrease in activity. Interestingly, the S84C and S84T mutant proteins were both active but demonstrated substrate inhibition. The three-dimensional structures of all of the mutant proteins were nearly identical to that of the wild-type enzyme, indicating that the changes in their kinetic parameters were not due to major conformational changes. Taken together, these data suggest that Asp 94 is important for substrate binding, but probably does not function as an enzymatic base, and that Ser 84 most likely plays a role in the formation of an oxyanion hole.


Subject(s)
Acyltransferases/chemistry , Acyltransferases/metabolism , Amino Sugars/metabolism , Bacillus cereus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Deoxyglucose/analogs & derivatives , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Acyltransferases/genetics , Amino Acid Substitution , Aspartic Acid/chemistry , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Coenzyme A/chemistry , Coenzyme A/metabolism , Crystallography, X-Ray , Deoxy Sugars/chemistry , Deoxy Sugars/metabolism , Deoxyglucose/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine/chemistry , Thymine Nucleotides/chemistry , Thymine Nucleotides/metabolism
16.
Glycobiology ; 22(9): 1236-44, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22661447

ABSTRACT

O-Antigen is a component of the outer membrane of Gram-negative bacteria and one of the most variable cell surface constituents, giving rise to major antigenic variability. The diversity of O-antigen is almost entirely attributed to genetic variations in O-antigen gene clusters. Bacteria of the genus Providencia are facultative pathogens, which can cause urinary tract infections, wound infections and enteric diseases. Recently, the O-antigen gene cluster of Providencia was localized between the cpxA and yibK genes in the genome. However, few genes involved in the synthesis of Providencia O-antigens have been functionally identified. In this study, the putative O-antigen gene cluster of Providencia alcalifaciens O30 was sequenced and analyzed. Almost all putative genes for the O-antigen synthesis were found, including a novel formyltransferase gene vioF that was proposed to be responsible for the conversion of dTDP-4-amino-4,6- dideoxy-D-glucose (dTDP-D-Qui4N) to dTDP-4,6-dideoxy-4-formamido-D-glucose (dTDP-D-Qui4NFo). vioF was cloned, and the enzyme product was expressed as a His-tagged fusion protein, purified and assayed for its activity. High-performance liquid chromatography was used to monitor the enzyme-substrate reaction, and the structure of the product dTDP-D-Qui4NFo was established by electrospray ionization tandem mass spectrometry and nuclear magnetic resonance spectroscopy. Kinetic parameters of VioF were determined, and effects of temperature and cations on its activity were also examined. Together, the functional analyses support the identification of the O-antigen gene cluster of P. alcalifaciens O30.


Subject(s)
Bacterial Proteins/genetics , Glucosamine/analogs & derivatives , Hydroxymethyl and Formyl Transferases/genetics , O Antigens/metabolism , Providencia/metabolism , Antigenic Variation , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Base Sequence , Carbohydrate Sequence , Chromatography, High Pressure Liquid , Cloning, Molecular , Deoxy Sugars/metabolism , Escherichia coli/genetics , Glucosamine/biosynthesis , Hydroxymethyl and Formyl Transferases/chemistry , Hydroxymethyl and Formyl Transferases/metabolism , Kinetics , Molecular Sequence Data , Multigene Family , O Antigens/chemistry , O Antigens/genetics , Providencia/chemistry , Providencia/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Spectrometry, Mass, Electrospray Ionization , Temperature , Thymine Nucleotides/metabolism
17.
Angew Chem Int Ed Engl ; 51(42): 10638-42, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22997042

ABSTRACT

Two bifunctional enzymes cooperate in the assembly and the positioning of two sugars, D-olivose and D-mycarose, of the anticancer antibiotic mithramycin. MtmC finishes the biosynthesis of both sugar building blocks depending on which MtmGIV activity is supported. MtmGIV transfers these two sugars onto two structurally distinct acceptor substrates. The dual function of these enzymes explains two essential but previously unidentified activities.


Subject(s)
Antibiotics, Antineoplastic/biosynthesis , Deoxy Sugars/metabolism , Glycosyltransferases/metabolism , Plicamycin/biosynthesis , Carbohydrate Sequence , Deoxy Sugars/biosynthesis , Glycosylation , Glycosyltransferases/biosynthesis , Molecular Sequence Data
18.
Appl Environ Microbiol ; 77(2): 435-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21075894

ABSTRACT

A combinatorial biosynthetic approach was used to interrogate the donor substrate flexibility of GilGT, the glycosyltransferase involved in C-glycosylation during gilvocarcin biosynthesis. Complementation of gilvocarcin mutant Streptomyces lividans TK24 (cosG9B3-U(-)), in which the biosynthesis of the natural sugar donor substrate was compromised, with various deoxysugar plasmids led to the generation of six gilvocarcin analogues with altered saccharide moieties. Characterization of the isolated gilvocarcin derivatives revealed five new compounds, including 4-ß-C-D-olivosyl-gilvocarcin V (D-olivosyl GV), 4-ß-C-D-olivosyl-gilvocarcin M (D-olivosyl GM), 4-ß-C-D-olivosyl-gilvocarcin E (D-olivosyl GE), 4-α-C-L-rhamnosyl-gilvocarcin M (polycarcin M), 4-α-C-L-rhamnosyl-gilvocarcin E (polycarcin E), and the recently characterized 4-α-C-L-rhamnosyl-gilvocarcin V (polycarcin V). Preliminary anticancer assays showed that D-olivosyl-gilvocarcin and polycarcin V exhibit antitumor activities comparable to that of their parent drug congener, gilvocarcin V, against human lung cancer (H460), murine lung cancer (LL/2), and breast cancer (MCF-7) cell lines. Our findings demonstrate GilGT to be a moderately flexible C-glycosyltransferase able to transfer both D- and L-hexopyranose moieties to the unique angucyclinone-derived benzo[D]naphtho[1,2b]pyran-6-one backbone of the gilvocarcins.


Subject(s)
Aminoglycosides/metabolism , Antineoplastic Agents/metabolism , Deoxy Sugars/metabolism , Streptomyces lividans/genetics , Streptomyces lividans/metabolism , Animals , Biosynthetic Pathways/genetics , Cell Line, Tumor , Cell Survival/drug effects , Coumarins/chemistry , Coumarins/metabolism , Humans , Mice , Streptomyces lividans/enzymology
19.
Appl Microbiol Biotechnol ; 90(4): 1347-59, 2011 May.
Article in English | MEDLINE | ID: mdl-21336933

ABSTRACT

To investigate the distribution of dTDP-glucose-4,6-dehydratase (dTGD) gene and diversity of the potential 6-deoxyhexose (6DOH) glycosylated compounds in marine microorganisms, a total of 91 marine sediment-derived bacteria, representing 48 operational taxonomic units and belonging to 25 genera, were screened by polymerase chain reaction. In total, 84% of the strains were dTGD gene positive, suggesting 6DOH biosynthetic pathway is widespread in these marine sediment-derived bacteria. BLASTp results of dTGD gene fragments indicate a high chemical diversity of the potential 6DOH glycosylated compounds. Close phylogenetic relationship occurred between dTGDs involved in the production of same or similar 6DOH glycosylated compounds, suggesting dTGD can be used to predict the structure of potential 6DOH glycosylated compounds produced by new strains. In two cases, where dTGD shared ≥85% amino acid identity and close phylogenetic relationship with their counterparts, 6DOH glycosylated compounds were accurately predicted. Our results demonstrate that phylogenetic analysis of dTGD gene is useful for structure prediction of glycosylated compounds from newly isolated strains and can therefore guide the chemical purification and structure identification process. The rapid identification of strains that possess dTGD gene provides a bioinformatics assessment of the greatest potential to produce glycosylated compounds despite the absence of fully biosynthetic pathways or genome sequences.


Subject(s)
Bacteria/enzymology , Bacteria/isolation & purification , Bacterial Proteins/genetics , Geologic Sediments/microbiology , Hydro-Lyases/genetics , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Deoxy Sugars/metabolism , Glycosylation , Hydro-Lyases/chemistry , Hydro-Lyases/metabolism , Molecular Sequence Data , Phylogeny , Substrate Specificity
20.
Protein Sci ; 30(12): 2418-2432, 2021 12.
Article in English | MEDLINE | ID: mdl-34651380

ABSTRACT

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.


Subject(s)
Acetyltransferases/chemistry , Bacterial Proteins/chemistry , Deoxy Sugars/chemistry , Helicobacter/enzymology , Lipopolysaccharides/chemistry , Thymine Nucleotides/chemistry , Acetyltransferases/genetics , Acetyltransferases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Deoxy Sugars/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycoconjugates/chemistry , Glycoconjugates/metabolism , Helicobacter/chemistry , Kinetics , Lipopolysaccharides/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Thymine Nucleotides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL