首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 125 毫秒
1.
The lipopolysaccharide of Pseudomonas aeruginosa PAO1 contains an unusual sugar, 2,3-diacetamido-2,3-dideoxy-d-mannuronic acid (d-ManNAc3NAcA). wbpB, wbpE, and wbpD are thought to encode oxidase, transaminase, and N-acetyltransferase enzymes. To characterize their functions, recombinant proteins were overexpressed and purified from heterologous hosts. Activities of His6-WbpB and His6-WbpE were detected only when both proteins were combined in the same reaction. Using a direct MALDI-TOF mass spectrometry approach, we identified ions that corresponded to the predicted products of WbpB (UDP-3-keto-d-GlcNAcA) and WbpE (UDP-d-GlcNAc3NA) in the coupled enzyme-substrate reaction. Additionally, in reactions involving WbpB, WbpE, and WbpD, an ion consistent with the expected product of WbpD (UDP-d-GlcNAc3NAcA) was identified. Preparative quantities of UDP-d-GlcNAc3NA and UDP-d-GlcNAc3NAcA were enzymatically synthesized. These compounds were purified by high-performance liquid chromatography, and their structures were elucidated by NMR spectroscopy. This is the first report of the functional characterization of these proteins, and the enzymatic synthesis of UDP-d-GlcNAc3NA and UDP-d-GlcNAc3NAcA.Gram-negative organisms such as Pseudomonas aeruginosa produce lipopolysaccharide (LPS)4 as an essential component of the outer leaflet of the outer membrane. LPS can be conceptually divided into three parts: lipid A, which anchors LPS into the membrane; core oligosaccharide, which contributes to membrane stability; and the O-antigen, which is a polysaccharide that extends away from the cell surface. In P. aeruginosa, two types of O-antigen are observed: A-band O-antigen, which is common to most strains, and B-band O-antigen, which is variable and therefore used as the basis of the International Antigenic Typing Scheme (1). P. aeruginosa serotypes O2, O5, O16, O18, and O20 collectively belong to serogroup O2, because they all share common backbone sugar structures in their O-antigen repeat units consisting of two di-N-acetylated uronic acids and one 2-acetamido-2,6-dideoxy-d-galactose (N-acetyl-d-fucosamine). The minor structural variations in the O-antigen repeat units that differentiate this serogroup into five serotypes are: the type of glycosidic linkage between O-units (alpha versus beta) that is formed by the O-antigen polymerase (Wzy), isomers present (d-mannuronic or l-guluronic acid), and acetyl group substituents (24). The B-band O-antigen of P. aeruginosa PAO1 (serotype O5) contains a repeating trisaccharide of 2-acetamido-3-acetamidino-2,3-dideoxy-d-mannuronic acid (d-ManNAc3NAmA), 2,3-diacetamido-2,3-dideoxy-d-mannuronic acid (d-ManNAc3NAcA), and 2-acetamido-2,6-dideoxy-d-galactose (3).The biosynthesis of the two mannuronic acid derivatives has yet to be fully understood and has been the subject of investigation by our group. To produce UDP-d-ManNAc3NAcA, a five-step pathway has been proposed (Fig. 1) that requires the products of five genes localized to the B-band O-antigen biosynthesis cluster (5). The O-antigen biosynthesis cluster was shown to be identical for all serotypes within serogroup O2, which further underscores the high similarity between these serotypes (5). The five genes, including wbpA, wbpB, wbpE, wbpD, and wbpI, have been shown to be essential for B-band LPS biosynthesis, because knockout mutants of each of these genes are deficient in B-band O-antigen (68). Homologs of all five of the proteins required for the UDP-d-ManNAc3NAcA biosynthesis pathway are conserved in other bacterial pathogens, including Bordetella pertussis, Bordetella parapertussis, and Bordetella bronchiseptica. Cross-complementation of P. aeruginosa knockout mutants lacking wbpA, wbpB, wbpE, wbpD, or wbpI with the homologues from B. pertussis could fully restore LPS production in the P. aeruginosa LPS mutants, suggesting that the genes from B. pertussis are functional homologs of the wbp genes (7). Homologs of these genes could be identified in diverse bacterial species, demonstrating the importance of UDP-d-ManNAc3NAcA biosynthesis beyond its role in P. aeruginosa (7).Open in a separate windowFIGURE 1.Proposed pathway for the biosynthesis of UDP-d-ManNAc3NAcA in P. aeruginosa PAO1. The full names of the sugars are as follows: GlcNAc, 2-acetamido-2-deoxy-d-glucose; GlcNAcA, 2-acetamido-2-deoxy-d-glucuronic acid; 3-keto-d-GlcNAcA, 2-acetamido-2-deoxy-d-ribo-hex-3-uluronic acid; GlcNAc3NA, 2-acetamido-3-amino-2,3-dideoxy-d-glucuronic acid; GlcNAc3NAcA, 2,3-diacetamido-2,3-dideoxy-d-glucuronic acid; ManNAc3NAcA, 2,3-diacetamido-2,3-dideoxy-d-mannuronic acid. Adapted from Ref. 8.The first enzyme of the UDP-d-ManNAc3NAcA biosynthesis pathway, WbpA, is a 6-dehydrogenase that converts UDP-2-acetamido-2-deoxy-d-glucose (N-acetyl-d-glucosamine; UDP-d-GlcNAc) to UDP-2-acetamido-2-deoxy-d-glucuronic acid (N-acetyl-d-glucosaminuronic acid, UDP-d-GlcNAcA) using NAD+ as a coenzyme (9) (Fig. 1). Following this, the second step in UDP-d-ManNAc3NAcA biosynthesis is proposed to be an oxidation reaction catalyzed by WbpB, forming UDP-2-acetamido-2-deoxy-d-ribo-hex-3-uluronic acid (3-keto-d-GlcNAcA), which in turn is used as the substrate for transamination by WbpE, creating UDP-2-acetamido-3-amino-2,3-dideoxy-d-glucuronic acid (d-GlcNAc3NA).This residue is thought to be the substrate for WbpD, a putative N-acetyltransferase of the hexapeptide acyltransferase superfamily (10) that requires acetyl-CoA as a co-substrate (8). WbpD has been proposed to synthesize UDP-2,3-diacetamido-2,3-dideoxy-d-glucuronic acid (UDP-d-GlcNAc-3NAcA), which is utilized in the B-band O-antigen of P. aeruginosa serotype O1. In P. aeruginosa serogroup O2, the UDP-d-GlcNAc3NAcA is then epimerized by WbpI to create the UDP-d-ManNAc3NAcA required for incorporation into B-band LPS (11). A derivative of UDP-d-ManNAc3NAcA is also used in the synthesis of B-band O-antigen of P. aeruginosa serogroup O2. UDP-d-ManNAc3NAmA is thought to be produced through additional modification of UDP-d-ManNAc3NAcA via the action of WbpG, an amidotransferase, which has also been demonstrated to be essential for the production of B-band O-antigen (12, 13).In the current study, our aim was to define the function of WbpB, WbpE, and WbpD, because only genetic evidence has previously been given for the involvement of wbpB and wbpE (7), and the reaction catalyzed by WbpD could not be demonstrated due to the unavailability of its presumed substrate, UDP-d-GlcNAc3NA (8). The functional characterization of these proteins is also important for understanding LPS biosynthesis in B. pertussis, because the genes in the LPS locus of this species, wlbA, wlbC, and wlbB, could cross-complement knockouts of wbpB, wbpE, and wbpD, respectively, when expressed in P. aeruginosa PAO1 (7). Furthermore, these three proteins form a cassette for the generation of C-3 N-acetylated hexoses and may be important for the biosynthesis of a variety of other sugars. Capillary electrophoresis and MALDI-TOF mass spectrometry were used to analyze reaction mixtures of WbpB and WbpE and showed that the expected products were produced only when both enzymes were present together. Achieving the enzymatic synthesis of the product of both enzymes, which was demonstrated to be UDP-d-GlcNAc3NA by 1H NMR spectroscopy, was a key breakthrough, because this rare sugar has never before been produced by any means. UDP-d-GlcNAc3NA was also essential for use as the substrate of WbpD, which not only allowed us to determine the enzymatic activity of this protein but also allowed the enzymatic synthesis of UDP-d-GlcNAc3NAcA to be achieved as well. Although this sugar had previously been produced through a 17-step chemical synthesis (11, 14), the 4-step concurrent enzymatic reaction demonstrates the advantage of linking chemistry with biology and represents a significant saving of both time and reagents as compared with chemical synthesis. Finally, our data also showed the success in reconstituting in vitro the 5-step pathway for the biosynthesis of UDP-d-ManNAc3NAcA in P. aeruginosa.  相似文献   

2.
Helicobacter pylori causes gastrointestinal diseases, including gastric cancer. Its high motility in the viscous gastric mucosa facilitates colonization of the human stomach and depends on the helical cell shape and the flagella. In H. pylori, Csd6 is one of the cell shape-determining proteins that play key roles in alteration of cross-linking or by trimming of peptidoglycan muropeptides. Csd6 is also involved in deglycosylation of the flagellar protein FlaA. To better understand its function, biochemical, biophysical, and structural characterizations were carried out. We show that Csd6 has a three-domain architecture and exists as a dimer in solution. The N-terminal domain plays a key role in dimerization. The middle catalytic domain resembles those of l,d-transpeptidases, but its pocket-shaped active site is uniquely defined by the four loops I to IV, among which loops I and III show the most distinct variations from the known l,d-transpeptidases. Mass analyses confirm that Csd6 functions only as an l,d-carboxypeptidase and not as an l,d-transpeptidase. The d-Ala-complexed structure suggests possible binding modes of both the substrate and product to the catalytic domain. The C-terminal nuclear transport factor 2-like domain possesses a deep pocket for possible binding of pseudaminic acid, and in silico docking supports its role in deglycosylation of flagellin. On the basis of these findings, it is proposed that H. pylori Csd6 and its homologs constitute a new family of l,d-carboxypeptidase. This work provides insights into the function of Csd6 in regulating the helical cell shape and motility of H. pylori.  相似文献   

3.
Flagella of the bacteria Helicobacter pylori and Campylobacter jejuni are important virulence determinants, whose proper assembly and function are dependent upon glycosylation at multiple positions by sialic acid-like sugars, such as 5,7-diacetamido-3,5,7,9-tetradeoxy-l-glycero-l-manno-nonulosonic acid (pseudaminic acid (Pse)). The fourth enzymatic step in the pseudaminic acid pathway, the hydrolysis of UDP-2,4-diacetamido-2,4,6-trideoxy-β-l-altropyranose to generate 2,4-diacetamido-2,4,6-trideoxy-l-altropyranose, is performed by the nucleotide sugar hydrolase PseG. To better understand the molecular basis of the PseG catalytic reaction, we have determined the crystal structures of C. jejuni PseG in apo-form and as a complex with its UDP product at 1.8 and 1.85 Å resolution, respectively. In addition, molecular modeling was utilized to provide insight into the structure of the PseG-substrate complex. This modeling identifies a His17-coordinated water molecule as the putative nucleophile and suggests the UDP-sugar substrate adopts a twist-boat conformation upon binding to PseG, enhancing the exposure of the anomeric bond cleaved and favoring inversion at C-1. Furthermore, based on these structures a series of amino acid substitution derivatives were constructed, altering residues within the active site, and each was kinetically characterized to examine its contribution to PseG catalysis. In conjunction with structural comparisons, the almost complete inactivation of the PseG H17F and H17L derivatives suggests that His17 functions as an active site base, thereby activating the nucleophilic water molecule for attack of the anomeric C–O bond of the UDP-sugar. As the PseG structure reveals similarity to those of glycosyltransferase family-28 members, in particular that of Escherichia coli MurG, these findings may also be of relevance for the mechanistic understanding of this important enzyme family.The gastrointestinal pathogens Campylobacter jejuni and Helicobacter pylori have been shown to modify their flagellins with the sialic acid-like sugar 5,7-diacetamido-3,5,7,9-tetradeoxy-l-glycero-l-manno-nonulosonic acid or pseudaminic acid (Pse),3 via O-linkage at up to 19 sites per flagellin monomer (1, 2). Not only is this sialic acid-like modification necessary for flagellar assembly and motility (1, 2), it has also been shown to be important for C. jejuni virulence (3). In addition to its role in autoagglutination of bacterial cells, Pse and related derivatives may also influence pathogenesis through bacterial adhesion, invasion, and immune evasion (4, 5), since sialic acids in humans have been shown to mediate a myriad of cell-cell and cell-molecule interactions (6). As flagellin glycosylation in these organisms is required for host colonization and ultimately virulence (3, 7, 8), these novel sugar biosynthetic pathways provide an excellent platform for therapeutic development.The reliance of H. pylori pathogenicity on Pse biosynthesis, in combination with the prevalence of H. pylori resistance to existing antibiotic treatments (9), prompted and led to the complete elucidation of the CMP-pseudaminic acid (CMP-Pse) biosynthetic pathway in both C. jejuni and H. pylori (1015). The CMP-Pse biosynthetic pathway (Fig. 1) is similar to that of CMP-sialic acid, involving condensation of an N-acetylhexosamine intermediate with the three-carbon pyruvate molecule forming a nine-carbon sialic acid-like nonulosonate, although in contrast the CMP-Pse pathway consists of several more steps between the initial building block UDP-GlcNAc and the condensation reaction. PseG, a UDP-sugar hydrolase, produces the final 6-deoxy-N-acetylhexosamine intermediate in the CMP-Pse pathway by removing the nucleotide moiety from UDP-2,4-diacetamido-2,4,6-trideoxy-β-l-altropyranose or UDP-6-deoxy-AltdiNAc (Fig. 1). This sort of single enzymatic function is rare in nature, with the only other similar example being a GDP-mannose/GDP-glucose hydrolase (16), which belongs to the metal-dependent Nudix family of enzymes. In an elegant study, Liu and Tanner (11) demonstrated that PseG catalyzes nucleotide removal by a metal-independent C–O bond cleavage mechanism resulting in inversion of stereochemistry at C-1 of the product 2,4-diacetamido-2,4,6-trideoxy-l-altropyranose or 6-deoxy-AltdiNAc, similar to the catalytic properties of some GT-B glycosyltransferases.Open in a separate windowFIGURE 1.Role of PseG within the CMP-pseudaminic acid biosynthetic pathway of C. jejuni and H. pylori. The biosynthetic step involving PseG is highlighted in blue. The enzymes and biosynthetic intermediates of the CMP-pseudaminic acid pathway are, in the following order, PseB (Cj1293/HP0840), NADP-dependent dehydratase/epimerase; PseC (Cj1294/HP0366), pyridoxal phosphate-dependent aminotransferase; PseH (Cj1313/HP0327), N-acetyltransferase; PseG (Cj1312/HP0326B), NDP-sugar hydrolase; PseI (Cj1317/HP0178), pseudaminic acid synthase; PseF (Cj1311/HP0326A), CMP-pseudaminic acid synthetase; and I, UDP-GlcNAc; II, UDP-2-acetamido-2,6-dideoxy-β-l-arabino-hexos-4-ulose; III, UDP-4-amino-4,6-dideoxy-β-l-AltNAc; IV, UDP-2,4-diacetamido-2,4,6-trideoxy-β-l-altropyranose; V, 2,4-diacetamido-2,4,6-trideoxy-l-altropyranose; VI, pseudaminic acid; and VII, CMP-pseudaminic acid. Here, PEP refers to phosphoenolpyruvate. Pyranose rings are shown as their predominant chair conformation in solution as determined from nuclear Overhauser effects and JH,H coupling constants (13).Together, glycosyltransferases and glycoside hydrolases compose the majority of enzymes in both eukaryotes and prokaryotes that manipulate glycosidic bonds. Glycosyltransferases of the Leloir classification use sugar-nucleotide derivatives as glycosyl donors resulting in transfer to acceptors such as a monosaccharide, oligosaccharide, or polysaccharide. It is therefore plausible that a “glycosyltransferase fold” in PseG has evolved to efficiently utilize water as an acceptor, instead of another carbohydrate, consequently behaving as a hydrolase (11). Based on structure, most glycosyltransferases fall into two groups, GT-A and GT-B, that exhibit different folds, respectively (17). For both families, depending on the particular enzyme, the outcome may result in either inversion or retention of stereochemistry for the donor anomeric carbon (see Fig. 2). In addition, GT-B family enzymes are metal-independent, lacking an important DXD motif present in most GT-A members. Based on the novelty of PseG and its role in H. pylori pathogenicity, we sought a greater structural and mechanistic understanding of this important enzyme.Open in a separate windowFIGURE 2.Functional comparison of enzymes belonging to the GT-B superfamily. A, UDP-sugar hydrolase PseG catalyzes the removal of UDP from UDP-2,4-diacetamido-2,4,6-trideoxy-β-l-Alt or UDP-6-deoxy-AltdiNAc. B, UDP-GlcNAc hydrolyzing 2-epimerase NeuC catalyzes the removal of UDP and the formation of ManNAc from UDP-GlcNAc. C, GlcNAc transferase MurG catalyzes the formation of undecaprenyl-phosphoryl-muramyl-pentapeptide-GlcNAc via formation of a glycosidic linkage between UDP-GlcNAc and undecaprenyl-phosphoryl-muramyl-pentapeptide. R represents the phosphoryl-undecaprenyl moiety, with the pentapeptide having the specific sequence l-Ala-d-γGlu-l-Lys-d-Ala-d-Ala. Both A and C activities result in an initial inversion of stereochemistry at C-1 for the donor substrate. In contrast, the activity for B results in an initial retention of C-1 stereochemistry. Enzymatically altered anomeric bonds are indicated in red.Here we report the crystal structure of PseG alone at 1.8 Å resolution and in complex with UDP, a product of the reaction, at 1.85 Å resolution. Although very few homologs have been identified based on sequence similarity alone, PseG bears the closest structural similarity to MurG, a GT-B family member (18). In addition, computational docking and molecular dynamics simulations were performed to gain insight into the binding mode of the PseG substrate UDP-6-deoxy-AltdiNAc. Based on the crystallographic and modeled structures, several potential active site residues were selected for mutagenesis and kinetic analyses to further characterize the PseG active site. The relevance of these findings to the structurally related MurG family of enzymes is discussed.  相似文献   

4.
Studies on a glycopeptide from ovalbumin   总被引:1,自引:1,他引:0  
1. The structure of the carbohydrate component of the glycopeptide isolated from the proteolytic digest of ovalbumin has been investigated by chemical and enzymic methods. 2. The results are consistent with the presence of a single carbohydrate prosthetic group, linked through its reducing end group to the peptide chain. 3. Further, all the 2-amino-2-deoxy-d-glucose units appear to be in the N-acylated form, the phenolic hydroxyl group of tyrosine is free and the ω-carboxyl group of aspartic acid is substituted. 4. The carbohydrate component has a branched-chain structure, the two non-reducing ends being terminated by a d-mannopyranosyl and a 2-acetamido-2-deoxy-d-glucopyranosyl residue respectively. 5. The terminal d-mannopyranosyl unit is probably linked through at least one other d-mannopyranosyl residue to the remainder of the carbohydrate.  相似文献   

5.
N-acetyl-d-quinovosamine (2-acetamido-2,6-dideoxy-d-glucose, QuiNAc) occurs in the polysaccharide structures of many Gram-negative bacteria. In the biosynthesis of QuiNAc-containing polysaccharides, UDP-QuiNAc is the hypothetical donor of the QuiNAc residue. Biosynthesis of UDP-QuiNAc has been proposed to occur by 4,6-dehydration of UDP-N-acetyl-d-glucosamine (UDP-GlcNAc) to UDP-2-acetamido-2,6-dideoxy-d-xylo-4-hexulose followed by reduction of this 4-keto intermediate to UDP-QuiNAc. Several specific dehydratases are known to catalyze the first proposed step. A specific reductase for the last step has not been demonstrated in vitro, but previous mutant analysis suggested that Rhizobium etli gene wreQ might encode this reductase. Therefore, this gene was cloned and expressed in Escherichia coli, and the resulting His6-tagged WreQ protein was purified. It was tested for 4-reductase activity by adding it and NAD(P)H to reaction mixtures in which 4,6-dehydratase WbpM had acted on the precursor substrate UDP-GlcNAc. Thin layer chromatography of the nucleotide sugars in the mixture at various stages of the reaction showed that WbpM converted UDP-GlcNAc completely to what was shown to be its 4-keto-6-deoxy derivative by NMR and that addition of WreQ and NADH led to formation of a third compound. Combined gas chromatography-mass spectrometry analysis of acid hydrolysates of the final reaction mixture showed that a quinovosamine moiety had been synthesized after WreQ addition. The two-step reaction progress also was monitored in real time by NMR. The final UDP-sugar product after WreQ addition was purified and determined to be UDP-d-QuiNAc by one-dimensional and two-dimensional NMR experiments. These results confirmed that WreQ has UDP-2-acetamido-2,6-dideoxy-d-xylo-4-hexulose 4-reductase activity, completing a pathway for UDP-d-QuiNAc synthesis in vitro.  相似文献   

6.
All of the 2,6-dideoxy sugars contained within the structure of chromomycin A3 are derived from d-glucose. Enzyme assays were used to confirm the presence of hexokinase, phosphoglucomutase, UDPG pyrophosphorylase (UDPGP), and UDPG oxidoreductase (UDPGO), all of which are involved in the pathway of glucose activation and conversion into 2,6-dideoxyhexoses during chromomycin biosynthesis. Levels of the four enzymes in Streptomyces spp. cell extracts were correlated with the production of chromomycins. The pathway of sugar activation in Streptomyces spp. involves glucose 6-phosphorylation by hexokinase, isomerization to G-1-P catalyzed by phosphoglucomutase, synthesis of UDPG catalyzed by UDPGP, and formation of UDP-4-keto-6-deoxyglucose by UDPGO.Dideoxy sugars occur commonly in the structures of cardiac glycosides from plants, in antibiotics like chromomycin A3 (Fig. (Fig.1),1), and in macrolides produced by microorganisms. On the basis of stable isotope-labeling experiments, biosynthetic studies conducted in Rosazza’s laboratory have indicated that all the deoxy sugars of chromomycin A3 are derived from d-glucose (21). While the assembly of the polyketide aglycone is reasonably well understood, relatively little is known of the details of 2,6-dideoxy sugar biogenesis in streptomycetes. Earlier studies with Streptomyces rimosus indicated that TDP-mycarose is synthesized from TDP-d-glucose (TDPG) and S-adenosyl-l-methionine (10, 23). The reaction requires NADPH as a cofactor, and TDP-4-keto-6-deoxy-d-glucose is an intermediate. Formation of TDP-4-keto-6-deoxy-d-glucose was catalyzed by the enzyme TDPG oxidoreductase (TDPG-4,6-dehydratase; EC 4.2.1.46). Similar 4-keto sugar nucleotides are intermediates for the biosynthesis of polyene macrolide antibiotic amino sugars (18). Similar pathways have been elaborated for the formation of 2,6-dideoxy-d-threo-4-hexulose of granaticin in Escherichia coli (6, 25) and 2,6-dideoxy-d-arabino-hexose of chlorothricin (12). The initial 6-deoxygenation of glucose during 3,6-dideoxy sugar formation involves a similar mechanism (32). In all of these processes, glucose is first activated by conversion into a sugar nucleotide such as UDPG followed by NAD+ oxidation of the 4 position to the corresponding 4-oxo derivative. Position 6 deoxygenation involves a general tautomerization, dehydration, and NADH,H+-catalyzed reduction process (6, 12, 25). A similar tautomerization and dehydration followed by reduction may produce C-3-deoxygenated products, such as CDP-3,6-dideoxyglucose (27). The pathway for formation of 3,6-dideoxyhexoses from CDPG in Yersinia pseudotuberculosis was clearly elucidated by Liu and Thorson (14). However, none of this elegant work was focused on the earlier steps of hexose nucleotide formation. Open in a separate windowFIG. 1Structures of chromomycins A2 and A3.On the basis of previous work (7), it is reasonable to postulate that the biosynthesis of 2,6-dideoxyglucose in Streptomyces griseus involves phosphorylation to glucose-6-phosphate by hexokinase (HK; E.C.2.7.7.1), as in glycolysis; conversion to glucose-1-phosphate by phosphoglucomutase (PGM; EC 2.7.5.1); reaction with UTP to form UDPG in a reaction catalyzed by UDPG pyrophosphorylase (UDPGP) (glucose-1-phosphate uridylyltransferase; EC 2.7.7.9), and C-6 deoxygenation catalyzed by UDP-d-glucose-4,6-dehydratase with NAD+ as a cofactor (Fig. (Fig.2).2). UDPG and GDPG have been detected in cell extracts of S. griseus and Streptomyces sp. strain MRS202, suggesting that these compounds are active sugar nucleotides involved in the formation of dideoxyhexoses (15). UDPGP genes from several bacteria have been cloned and sequenced (1, 3, 4, 11, 29, 30). Although nucleotidyl diphosphohexose-4,6-dehydratases (NDP-hexose-4,6-dehydratases) have been purified and characterized from several sources (5, 8, 9, 13, 19, 25, 26, 31, 33), the occurrence of the glucose-activating enzymes HK, PGM, UDPGP, and UDPG oxidoreductase (UDPGO) involved in 2,6-dideoxyhexose formation has not been established in streptomycetes. This work provides evidence for the presence of these enzymes involved in the biosynthetic activation of glucose to the 2,6-dideoxyhexoses in chromomycin A3.Open in a separate windowFIG. 2Proposed pathway for the formation of 2,6-dideoxy sugars in streptomycetes involving HK, PGM, UDPGP, and UDPGO.  相似文献   

7.
Uniquely modified heptoses found in surface carbohydrates of bacterial pathogens are potential therapeutic targets against such pathogens. Our recent biochemical characterization of the GDP-6-deoxy-d-manno- and GDP-6-deoxy-d-altro-heptose biosynthesis pathways has provided the foundation for elucidation of the more complex l-gluco-heptose synthesis pathway of Campylobacter jejuni strain NCTC 11168. In this work we use GDP-4-keto,6-deoxy-d-lyxo-heptose as a surrogate substrate to characterize three enzymes predicted to be involved in this pathway: WcaGNCTC (also known as Cj1427), MlghB (Cj1430), and MlghC (Cj1428). We compare them with homologues involved in d-altro-heptose production: WcaG81176 (formerly WcaG), DdahB (Cjj1430), and DdahC (Cjj1427). We show that despite high levels of similarity, the enzymes have pathway-specific catalytic activities and substrate specificities. MlghB forms three products via C3 and C5 epimerization activities, whereas its DdahB homologue only had C3 epimerase activity along its cognate pathway. MlghC is specific for the double C3/C5 epimer generated by MlghB and produces l-gluco-heptose via stereospecific C4 reductase activity. In contrast, its homologue DdahC only uses the C3 epimer to yield d-altro-heptose via C4 reduction. Finally, we show that WcaGNCTC is not necessary for l-gluco-heptose synthesis and does not affect its production by MlghB and MlghC, in contrast to its homologue WcaG81176, that has regulatory activity on d-altro-heptose synthesis. These studies expand our fundamental understanding of heptose modification, provide new glycobiology tools to synthesize novel heptose derivatives with biomedical applications, and provide a foundation for the structure function analysis of these enzymes.  相似文献   

8.
Several weakly transported sugars were tested for transport by the Na+-dependent sugar carrier with slices of everted hamster intestinal tissue. Sugars were assumed to be transported by this carrier if the accumulation was diminished in the absence of Na+ and in the presence of the competitive inhibitor 1,5-anhydro-d-glucitol. The extent of accumulation was correlated with the number of hydroxyl groups in the d-gluco configuration if the ring oxygen was placed in the normal d-glucose position. 5-Thio-d-glucose, with a sulphur atom in the ring, was transported at about the same rate as d-glucose and had a similar Ki for d-galactose transport, but myoinositol was poorly accumulated. It is suggested that there is no hydrogen bonding at the ring oxygen atom, but that the oxygen atom is found at this position as a result of steric constraints. No sugar without a hydroxyl group in the d-gluco position at C-2 of the sugar, including d-mannose, 2-deoxy-d-glucose, 2-chloro-2-deoxy-d-glucose and 2-deoxy-2-fluoro-d-glucose, was transported by the Na+-dependent carrier, but these sugars and l-fucose weakly and competitively inhibit the Na+-dependent accumulation of l-glucose into slices of everted hamster intestinal tissue. It is concluded that the bond between the carrier and C-2 of the sugar may be covalent, and a possible mechanism for active intestinal transport is proposed.  相似文献   

9.
The metabolism of myo-inositol-2-14C, d-glucuronate-1-14C, d-glucuronate-6-14C, and l-methionine-methyl-14C to cell wall polysaccharides was investigated in excised root-tips of 3 day old Zea mays seedlings. From myo-inositol, about one-half of incorporated label was recovered in ethanol insoluble residues. Of this label, about 90% was solubilized by treatment, first with a preparation of pectinase-EDTA, then with dilute hydrochloric acid. The only labeled constituents in these hydrolyzates were d-galacturonic acid, d-glucuronic acid, 4-O-methyl-d-glucuronic acid, d-xylose, and l-arabinose, or larger oligosaccharide fragments containing these units. Medium external to excised root-tips grown under sterile conditions in myo-inositol-2-14C contained labeled polysaccharide.  相似文献   

10.
Uptake of monosaccharides by guinea-pig cerebral-cortex slices   总被引:1,自引:1,他引:0       下载免费PDF全文
By the use of 1mm-iodoacetate to inhibit glycolysis in guinea-pig cerebral tissue slices, the kinetics of the uptake of monosaccharides on transfer of tissue from 0° to 37° were studied. d-Ribose, d-galactose, d-mannose, l-sorbose, and d-fructose showed diffusion kinetics, whereas 2-deoxy-d-glucose, d-glucose, d-arabinose and d-xylose showed saturation kinetics.  相似文献   

11.
SalM is a short-chain dehydrogenase/reductase enzyme from the marine actinomycete Salinispora tropica that is involved in the biosynthesis of chloroethylmalonyl-CoA, a novel halogenated polyketide synthase extender unit of the proteasome inhibitor salinosporamide A. SalM was heterologously overexpressed in Escherichia coli and characterized in vitro for its substrate specificity, kinetics, and reaction profile. A sensitive real-time 13C NMR assay was developed to visualize the oxidation of 5-chloro-5-deoxy-d-ribose to 5-chloro-5-deoxy-d-ribono-γ-lactone in an NAD+-dependent reaction, followed by spontaneous lactone hydrolysis to 5-chloro-5-deoxy-d-ribonate. Although short-chain dehydrogenase/reductase enzymes are widely regarded as metal-independent, a strong divalent metal cation dependence for Mg2+, Ca2+, or Mn2+ was observed with SalM. Oxidative activity was also measured with the alternative substrates d-erythrose and d-ribose, making SalM the first reported stereospecific non-phosphorylative ribose 1-dehydrogenase.  相似文献   

12.
l-Glutamine d-fructose 6-phosphate amidotransferase (EC 2.6.1.16) was extracted and purified 600-fold by acetone fractionation and diethylaminoethyl cellulose column chromatography from mung bean seeds (Phaseolus aureus). The partially purified enzyme was highly specific for l-glutamine as an amide nitrogen donor, and l-asparagine could not replace it. The enzyme showed a pH optimum in the range of 6.2 to 6.7 in phosphate buffer. Km values of 3.8 mm and 0.5 mm were obtained for d-fructose 6-phosphate and l-glutamine, respectively. The enzyme was competitively inhibited with respect to d-fructose 6-phosphate by uridine diphosphate-N-acetyl-d-glucosamine which had a Ki value of 13 μm. Upon removal of l-glutamine and its replacement by d-fructose 6-phosphate and storage over liquid nitrogen, the enzyme was completely desensitized to inhibition by uridine diphosphate-N-acetyl-d-glucosamine. This indicates that the inhibitor site is distinct from the catalytic site and that uridine diphosphate-N-acetyl-d-glucosamine acts as a feedback inhibitor of the enzyme.  相似文献   

13.
d-Galacturonic acid, the main monomer of pectin, is an attractive substrate for bioconversions, since pectin-rich biomass is abundantly available and pectin is easily hydrolyzed. l-Galactonic acid is an intermediate in the eukaryotic pathway for d-galacturonic acid catabolism, but extracellular accumulation of l-galactonic acid has not been reported. By deleting the gene encoding l-galactonic acid dehydratase (lgd1 or gaaB) in two filamentous fungi, strains were obtained that converted d-galacturonic acid to l-galactonic acid. Both Trichoderma reesei Δlgd1 and Aspergillus niger ΔgaaB strains produced l-galactonate at yields of 0.6 to 0.9 g per g of substrate consumed. Although T. reesei Δlgd1 could produce l-galactonate at pH 5.5, a lower pH was necessary for A. niger ΔgaaB. Provision of a cosubstrate improved the production rate and titer in both strains. Intracellular accumulation of l-galactonate (40 to 70 mg g biomass−1) suggested that export may be limiting. Deletion of the l-galactonate dehydratase from A. niger was found to delay induction of d-galacturonate reductase and overexpression of the reductase improved initial production rates. Deletion of the l-galactonate dehydratase from A. niger also delayed or prevented induction of the putative d-galacturonate transporter An14g04280. In addition, A. niger ΔgaaB produced l-galactonate from polygalacturonate as efficiently as from the monomer.  相似文献   

14.
d-Galacturonic acid can be obtained by hydrolyzing pectin, which is an abundant and low value raw material. By means of metabolic engineering, we constructed fungal strains for the conversion of d-galacturonate to meso-galactarate (mucate). Galactarate has applications in food, cosmetics, and pharmaceuticals and as a platform chemical. In fungi d-galacturonate is catabolized through a reductive pathway with a d-galacturonate reductase as the first enzyme. Deleting the corresponding gene in the fungi Hypocrea jecorina and Aspergillus niger resulted in strains unable to grow on d-galacturonate. The genes of the pathway for d-galacturonate catabolism were upregulated in the presence of d-galacturonate in A. niger, even when the gene for d-galacturonate reductase was deleted, indicating that d-galacturonate itself is an inducer for the pathway. A bacterial gene coding for a d-galacturonate dehydrogenase catalyzing the NAD-dependent oxidation of d-galacturonate to galactarate was introduced to both strains with disrupted d-galacturonate catabolism. Both strains converted d-galacturonate to galactarate. The resulting H. jecorina strain produced galactarate at high yield. The A. niger strain regained the ability to grow on d-galacturonate when the d-galacturonate dehydrogenase was introduced, suggesting that it has a pathway for galactarate catabolism.d-Galacturonate is the main component of pectin, an abundant and cheap raw material. Sugar beet pulp and citrus peel are both rich in pectin residues. At present, these residues are mainly used as cattle feed. However, since energy-consuming drying and pelletizing of the residues is required to prevent them from rotting, it is not always economical to process the residues, and it is desirable to find alternative uses.Various microbes which live on decaying plant material have the ability to catabolize d-galacturonate using various, completely different pathways (19). Eukaryotic microorganisms use a reductive pathway in which d-galacturonate is first reduced to l-galactonate by an NAD(P)H-dependent reductase (12, 17). In the following steps a dehydratase, aldolase, and reductase convert the l-galactonate to pyruvate and glycerol (9, 11, 14).In Hypocrea jecorina (anamorph Trichoderma reesei) the gar1 gene codes for a strictly NADPH-dependent d-galacturonate reductase. In Aspergillus niger a homologue gene sequence, gar2, exists; however, a different gene, gaaA, is upregulated during growth on d-galacturonate containing medium (16). The gaaA codes for a d-galacturonate reductase with different kinetic properties than the H. jecorina enzyme, having a higher affinity toward d-galacturonate and using either NADH or NADPH as cofactor. It is not known whether gar2 codes for an active protein.Some bacteria, such as Agrobacterium tumefaciens or Pseudomonas syringae, have an oxidative pathway for d-galacturonate catabolism. In this pathway d-galacturonate is first oxidized to meso-galactarate (mucate) by an NAD-utilizing d-galacturonate dehydrogenase. Galactarate is then converted in the following steps to α-ketoglutarate. This route is sometimes called the α-ketoglutarate pathway (20). Galactarate can also be catabolized through the glycerate pathway (20). The products of this pathway are pyruvate and d-glycerate. These pathways have been described in prokaryotes, and it is not certain whether similar pathways also exist in fungi, some of which are able to metabolize galactarate.d-Galacturonate dehydrogenase (EC 1.1.1.203) has been described in Agrobacterium tumefaciens and in Pseudomonas syringae, and the enzymes from these organisms have been purified and characterized (3, 6, 22). Recently, the corresponding genes were also identified (4, 24). Both enzymes are specific for NAD as a cofactor but are not specific for the substrate. They oxidize d-galacturonate and d-glucuronate to meso-galactarate (mucate) and d-glucarate (saccharate), respectively. The reaction product is probably the hexaro-lactone which spontaneously hydrolyzes. The reverse reaction can only be observed at acidic pH where some of the galactarate is in the lactone form (22).We describe here strains of filamentous fungi that have been genetically engineered to produce galactarate by disruption of d-galacturonate reductase and expression of d-galacturonate dehydrogenase (Fig. (Fig.1).1). Galactarate is currently commercially produced from d-galactose by oxidation with nitric acid (1) or from d-galacturonate by electrolytic oxidation (8). Oxidation with nitric acid is expensive and produces toxic wastes. Galactarate is used as a chelator and in skin care products. It was formerly used as a leavening agent in self-rising flour (2) and has potential applications in polymer synthesis (10) and as a platform chemical (for a review, see reference 13).Open in a separate windowFIG. 1.Engineering the d-galacturonic acid pathway in fungi. Deletion of the gene encoding d-galacturonate reductase resulted in strains unable to utilize d-galacturonic acid as a carbon source. The expression of a bacterial udh gene, encoding an NAD-dependent d-galacturonate dehydrogenase, resulted in fungal strains, which were able to oxidize d-galacturonic acid to meso-galactaric acid (mucic acid). d-Galacturonate dehydrogenase forms a galactaro-lactone which spontaneously hydrolyzes.  相似文献   

15.
To establish an advantageous method for the production of l-amino acids, microbial isomerization of d- and dl-amino acids to l-amino acids was studied. Screening experiments on a number of microorganisms showed that cell suspensions of Pseudomonas fluorescens and P. miyamizu were capable of isomerizing d- and dl-phenylalanines to l-phenylalanine. Various conditions suitable for isomerization by these organisms were investigated. Cells grown in a medium containing d-phenylalanine showed highest isomerization activity, and almost completely converted d- or dl-phenylalanine into l-phenylalanine within 24 to 48 hr of incubation. Enzymatic studies on this isomerizing system suggested that the isomerization of d- or dl-phenylalanine is not catalyzed by a single enzyme, “amino acid isomerase,” but the conversion proceeds by a two step system as follows: d-pheylalanine is oxidized to phenylpyruvic acid by d-amino acid oxidase, and the acid is converted to l-phenylalanine by transamination or reductive amination.  相似文献   

16.
The asexual spores (conidia) of Aspergillus niger germinate to produce hyphae under appropriate conditions. Germination is initiated by conidial swelling and mobilization of internal carbon and energy stores, followed by polarization and emergence of a hyphal germ tube. The effects of different pyranose sugars, all analogues of d-glucose, on the germination of A. niger conidia were explored, and we define germination as the transition from a dormant conidium into a germling. Within germination, we distinguish two distinct stages, the initial swelling of the conidium and subsequent polarized growth. The stage of conidial swelling requires a germination trigger, which we define as a compound that is sensed by the conidium and which leads to catabolism of d-trehalose and isotropic growth. Sugars that triggered germination and outgrowth included d-glucose, d-mannose, and d-xylose. Sugars that triggered germination but did not support subsequent outgrowth included d-tagatose, d-lyxose, and 2-deoxy-d-glucose. Nontriggering sugars included d-galactose, l-glucose, and d-arabinose. Certain nontriggering sugars, including d-galactose, supported outgrowth if added in the presence of a complementary triggering sugar. This division of functions indicates that sugars are involved in two separate events in germination, triggering and subsequent outgrowth, and the structural features of sugars that support each, both, or none of these events are discussed. We also present data on the uptake of sugars during the germination process and discuss possible mechanisms of triggering in the absence of apparent sugar uptake during the initial swelling of conidia.  相似文献   

17.
The mechanism of hexose transport into plasma membrane vesicles isolated from mature sugarbeet leaves (Beta vulgaris L.) was investigated. The initial rate of glucose uptake into the vesicles was stimulated approximately fivefold by imposing a transmembrane pH gradient (ΔpH), alkaline inside, and approximately fourfold by a negative membrane potential (ΔΨ), generated as a K+-diffusion potential, negative inside. The -fold stimulation was directly related to the relative ΔpH or ΔΨ gradient imposed, which were determined by the uptake of acetate or tetraphenylphosphonium, respectively. ΔΨ- and ΔpH-dependent glucose uptake showed saturation kinetics with a Km of 286 micromolar for glucose. Other hexose molecules (e.g. 2-deoxy-d-glucose, 3-O-methyl-d-glucose, and d-mannose) were also accumulated into plasma membrane vesicles in a ΔpH-dependent manner. Inhibition constants of a number of compounds for glucose uptake were determined. Effective inhibitors of glucose uptake included: 3-O-methyl-d-glucose, 5-thio-d-glucose, d-fructose, d-galactose, and d-mannose, but not 1-O-methyl-d-glucose, d- and l-xylose, l-glucose, d-ribose, and l-sorbose. Under all conditions of proton motive force magnitude and glucose and sucrose concentration tested, there was no effect of sucrose on glucose uptake. Thus, hexose transport on the sugarbeet leaf plasma membrane was by a H+-hexose symporter, and the carrier and possibly the energy source were not shared by the plasma membrane H+-sucrose symporter.  相似文献   

18.
The transport of some sugars at the antiluminal face of renal cells was studied using teased tubules of flounder (Pseudopleuronectes americanus). The analytical procedure allowed the determination of both free and total (free plus phosphorylated) tissue sugars. The inulin space of the preparation was 0.333 ± 0.017 kg/kg wet wt (7 animals, 33 analyses). The nonmetabolizable α-methyl-D-glucoside entered the cells by a carrier-mediated (phloridzin-sensitive), ouabain-insensitive process. The steady-state tissue/medium ratio was systematically below that for diffusion equilibrium. D-Glucose was a poor inhibitor of α-methyl-glucoside transport, D-galactose was ineffective. The phloridzin-sensitive transport processes of 2-deoxy-D-glucose,D-galactose,and 2-deoxy-D-galactose were associated with considerable phosphorylation. Kinetic evidence suggested that these sugars were transported in free form and subsequently were phosphorylated. 2-Deoxy-D-glucose accumulated in the cells against a slight concentration gradient. This transport was greatly inhibited by D-glucose, whereas α-methyl-glucoside and also D-galactose and its 2-deoxy-derivative were ineffective. D-Galactose and 2-deoxy-D-galactose mutually competed for transport; D-glucose, 2-deoxy-D-glucose, and α-methyl-D-glucoside were ineffective. Studies using various sugars as inhibitors suggest the presence of three carrier-mediated pathways of sugar transport at the antiluminal cell face of the flounder renal tubule: the pathway of α-methyl-D-glucoside (not shared by D-glucose); the pathway commonly shared by 2-deoxy-D-glucose and D-glucose; the pathway shared by D-galactose and 2-deoxy-D-galactose.  相似文献   

19.
Saito K  Kasai Z 《Plant physiology》1984,76(1):170-174
5-Keto-l-idionic acid (5-keto-d-gluconic acid, d-xylo-5-hexulosonic acid) was found as a metabolic product of l-ascorbic acid in slices of immature grapes, Vitis labrusca L. cv `Delaware'. Specifically labeled compounds, recognized as metabolic products of l-ascorbic acid in grapes, were fed to young grape tissues to investigate the metabolic pathway from l-ascorbic acid to l-(+)-tartaric acid.  相似文献   

20.
The first enzyme in the pathway for l-arabinose catabolism in eukaryotic microorganisms is a reductase, reducing l-arabinose to l-arabitol. The enzymes catalyzing this reduction are in general nonspecific and would also reduce d-xylose to xylitol, the first step in eukaryotic d-xylose catabolism. It is not clear whether microorganisms use different enzymes depending on the carbon source. Here we show that Aspergillus niger makes use of two different enzymes. We identified, cloned, and characterized an l-arabinose reductase, larA, that is different from the d-xylose reductase, xyrA. The larA is up-regulated on l-arabinose, while the xyrA is up-regulated on d-xylose. There is however an initial up-regulation of larA also on d-xylose but that fades away after about 4 h. The deletion of the larA gene in A. niger results in a slow growth phenotype on l-arabinose, whereas the growth on d-xylose is unaffected. The l-arabinose reductase can convert l-arabinose and d-xylose to their corresponding sugar alcohols but has a higher affinity for l-arabinose. The Km for l-arabinose is 54 ± 6 mm and for d-xylose 155 ± 15 mm.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号