Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Clin Invest ; 131(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33792561

RESUMEN

Airway eosinophilia is a hallmark of allergic asthma and is associated with mucus production, airway hyperresponsiveness, and shortness of breath. Although glucocorticoids are widely used to treat asthma, their prolonged use is associated with several side effects. Furthermore, many individuals with eosinophilic asthma are resistant to glucocorticoid treatment, and they have an unmet need for novel therapies. Here, we show that UDP-glucose (UDP-G), a nucleotide sugar, is selectively released into the airways of allergen-sensitized mice upon their subsequent challenge with that same allergen. Mice lacking P2Y14R, the receptor for UDP-G, had decreased airway eosinophilia and airway hyperresponsiveness compared with wild-type mice in a protease-mediated model of asthma. P2Y14R was dispensable for allergic sensitization and for the production of type 2 cytokines in the lung after challenge. However, UDP-G increased chemokinesis in eosinophils and enhanced their response to the eosinophil chemoattractant, CCL24. In turn, eosinophils triggered the release of UDP-G into the airway, thereby amplifying eosinophilic recruitment. This positive feedback loop was sensitive to therapeutic intervention, as a small molecule antagonist of P2Y14R inhibited airway eosinophilia. These findings thus reveal a pathway that can be therapeutically targeted to treat asthma exacerbations and glucocorticoid-resistant forms of this disease.


Asunto(s)
Asma/inmunología , Eosinófilos/inmunología , Eosinofilia Pulmonar/inmunología , Receptores Purinérgicos P2Y/inmunología , Uridina Difosfato Glucosa/inmunología , Alérgenos/inmunología , Animales , Asma/genética , Asma/patología , Quimiocina CCL24/genética , Quimiocina CCL24/inmunología , Eosinófilos/patología , Masculino , Ratones , Ratones Noqueados , Eosinofilia Pulmonar/genética , Eosinofilia Pulmonar/patología , Receptores Purinérgicos P2Y/deficiencia , Células Th2/inmunología , Células Th2/patología , Uridina Difosfato Glucosa/genética
2.
Metab Eng ; 61: 106-119, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32492511

RESUMEN

Glycolipids are target molecules in biotechnology and biomedicine as biosurfactants, biomaterials and bioactive molecules. An engineered E. coli strain for the production of glycoglycerolipids (GGL) used the MG517 glycolipid synthase from M. genitalium for glucosyl transfer from UDPGlc to diacylglycerol acceptor (Mora-Buyé et al., 2012). The intracellular diacylglycerol pool proved to be the limiting factor for GGL production. Here we designed different metabolic engineering strategies to enhance the availability of precursor substrates for the glycolipid synthase by modulating fatty acids, acyl donor and phosphatidic acid biosynthesis. Knockouts of tesA, fadE and fabR genes involved in fatty acids degradation, overexpression of the transcriptional regulator FadR, the acyltransferases PlsB and C, and the pyrophosphatase Cdh for phosphatidic acid biosynthesis, as well as the phosphatase PgpB for conversion to diacylglycerol were explored with the aim of improving GGL titers. Among the different engineered strains, the ΔtesA strain co-expressing MG517 and a fusion PlsCxPgpB protein was the best producer, with a 350% increase of GGL titer compared to the parental strain expressing MG517 alone. Attempts to boost UDPGlc availability by overexpressing the uridyltransferase GalU or knocking out the UDP-sugar diphosphatase encoding gene ushA did not further improve GGL titers. Most of the strains produced GGL containing a variable number of glucosyl units from mono-to tetra-saccharides. Interestingly, the strains co-expressing Cdh showed a shift in the GGL profile towards the diglucosylated lipid (up to 80% of total GGLs) whereas the strains with a fadR knockout presented a higher amount of unsaturated acyl chains. In all cases, GGL production altered the lipidic composition of the E. coli membrane, observing that GGL replace phosphatidylethanolamine to maintain the overall membrane charge balance.


Asunto(s)
Proteínas Bacterianas , Escherichia coli , Glucolípidos/biosíntesis , Glicosiltransferasas , Ingeniería Metabólica , Mycoplasma genitalium/genética , Ácidos Fosfatidicos/metabolismo , Uridina Difosfato Glucosa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Glucolípidos/genética , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Mycoplasma genitalium/enzimología , Ácidos Fosfatidicos/genética , Uridina Difosfato Glucosa/genética
3.
Metab Eng ; 47: 243-253, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29596994

RESUMEN

Synthetic microbial coculture to express heterologous biosynthetic pathway for de novo production of medicinal ingredients is an emerging strategy for metabolic engineering and synthetic biology. Here, taking efficient production of salidroside as an example of glycosides, we design and construct a syntrophic Escherichia coli-E. coli coculture composed of the aglycone (AG) strain and the glycoside (GD) strain, which convergently accommodate biosynthetic pathways of tyrosol and salidroside, respectively. To accomplish this the phenylalanine-deficient AG strain was engineered to utilize xylose preferentially and to overproduce precursor tyrosol, while the tyrosine-deficient GD strain was constructed to consume glucose exclusively and to enhance another precursor UDP-glucose availability for synthesis of salidroside. The AG and GD strains in the synthetic consortium are obligatory cooperators through crossfeeding of tyrosine and phenylalanine and compatible in glucose and xylose mixture. Through balancing the metabolic pathway strength, we show that the syntrophic coculture was robust and stable, and produced 6.03 g/L of salidroside. It was the de novo production of salidroside for the first time in E. coli coculture system, which would be applicable for production of other important glycosides and natural products.


Asunto(s)
Glucósidos , Ingeniería Metabólica , Fenoles , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Glucósidos/biosíntesis , Glucósidos/genética , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/metabolismo , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo , Xilosa/genética , Xilosa/metabolismo
4.
Glycobiology ; 26(5): 520-31, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26692049

RESUMEN

Formation of the heterocyst envelope polysaccharide (HEP) is a key process for cyanobacterial heterocyst differentiation. The maturation of HEP in Anabaena sp. strain PCC 7120 is controlled by a gene cluster termed HEP island in addition to an operon alr3698-alr3699, which encodes two putative proteins termed Alr3698/HepD and Alr3699/HepE. Here we report the crystal structures of HepE in the apo-form and three complex forms that bind to UDP-glucose (UDPG), UDP&glucose, and UDP, respectively. The overall structure of HepE displays a typical GT-B fold of glycosyltransferases, comprising two separate ß/α/ß Rossmann-fold domains that form an inter-domain substrate-binding crevice. Structural analyses combined with enzymatic assays indicate that HepE is a glucosyltransferase using UDPG as a sugar donor. Further site-directed mutageneses enable us to assign the key residues that stabilize the sugar donor and putative acceptor. Based on the comparative structural analyses, we propose a putative catalytic cycle of HepE, which undergoes "open-closed-open" conformational changes upon binding to the substrates and release of products. These findings provide structural and catalytic insights into the first enzyme involved in the HEP biosynthesis pathway.


Asunto(s)
Anabaena/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Glucosiltransferasas/química , Glucosiltransferasas/metabolismo , Polisacáridos Bacterianos/biosíntesis , Polisacáridos Bacterianos/química , Anabaena/genética , Proteínas Bacterianas/genética , Glucosiltransferasas/genética , Familia de Multigenes , Polisacáridos Bacterianos/genética , Dominios Proteicos , Uridina Difosfato Glucosa/química , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo
5.
Appl Biochem Biotechnol ; 175(8): 3729-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25724977

RESUMEN

We constructed and applied a recombinant, permeabilized Escherichia coli strain for the multistep synthesis of UDP-glucose. Sucrose phosphorylase (E.C. 2.4.1.7) of Leuconostoc mesenteroides was over expressed and the pgm gene encoding for phosphoglucomutase (E.C. 5.4.2.2) was deleted in E. coli to yield the E. coli JW 0675-1 SP strain. The cells were permeabilized with the detergent Triton X-100 at 0.05 % v/v. The synthesis of UDP-glucose with permeabilized cells was then optimized with regard to pH, cell density during the synthesis and growth phase during cell harvest, metal cofactor, other media components, and temperature. In one configuration sucrose, phosphate, UMP, and ATP were used as substrates. At pH 7.8, 27 mg/ml cell dry weight, cell harvest during the early stationary phase of growth and Mn(2+) as cofactor a yield of 37 % with respect to UMP was achieved at 33 °C. In a second step, ATP was regenerated by feeding glucose and using only catalytic amounts of ATP and NAD(+). A UDP-glucose yield of 60 % with respect to UMP was obtained using this setup. With the same setup but without addition of external ATP, the yield was 54%.


Asunto(s)
Escherichia coli/enzimología , Fosfoglucomutasa/metabolismo , Uridina Difosfato Glucosa/biosíntesis , Adenosina Trifosfato/metabolismo , Escherichia coli/genética , Glucosa/metabolismo , Glucosiltransferasas/biosíntesis , Glucosiltransferasas/genética , Ingeniería Metabólica , Fosfoglucomutasa/genética , Uridina Difosfato Glucosa/genética
6.
Arch Biochem Biophys ; 564: 120-7, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25317963

RESUMEN

Glucosyl-3-phosphoglycerate synthase (GpgS) catalyzes the first step in the biosynthesis of glucosyl glycerate, the putative precursor used in building methylated polysaccharides in mycobacteria. Enzymes from Mycobacterium tuberculosis (MtGpgS) and related species have been structurally characterized and subjected to basic kinetic analyses, but more in-depth kinetic analysis is currently lacking. Dead-end inhibition studies with MtGpgS suggest an ordered kinetic mechanism with 3-phosphoglycerate (3-PGA) binding first, followed by UDP-glucose, in contrast to previous reports. At higher concentrations, 3-PGA exhibits competitive substrate inhibition vs. UDP-glucose, suggesting 3-PGA can bind to either binding site on the enzyme. Parabolic noncompetitive inhibition plots by a 3-PGA analog also support this conclusion. The effect of varying pH on the catalytic parameters indicates single ionizable residue involved catalysis (pKa=6.3) that must be deprotonated for full activity. A solvent kinetic isotope effect of 2.0±0.3 on kcat is consistent with a proton in flight during the rate-determining step. Site-directed mutagenesis studies identify several residues critical for interactions with substrates. Although the residues are conserved among other glycosyltransferase families catalyzing similar reactions, the effect of substitutions varies between families suggesting that conserved areas play different catalytic roles in each family.


Asunto(s)
Proteínas Bacterianas/química , Glucosiltransferasas/química , Ácidos Glicéricos/química , Mycobacterium tuberculosis/enzimología , Uridina Difosfato Glucosa/química , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Catálisis , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Ácidos Glicéricos/metabolismo , Mutagénesis Sitio-Dirigida , Mutación Missense , Mycobacterium tuberculosis/genética , Unión Proteica , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo
7.
J Biol Chem ; 289(14): 10104-14, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24558041

RESUMEN

Glycolipids are mainly found in phototrophic organisms (like plants and cyanobacteria), in Gram-positive bacteria, and a few other bacterial phyla. Besides the function as bulk membrane lipids, they often play a role under phosphate deprivation as surrogates for phospholipids. The Gram-negative Agrobacterium tumefaciens accumulates four different glycolipids under phosphate deficiency, including digalactosyl diacylglycerol and glucosylgalactosyl diacylglycerol synthesized by a processive glycosyltransferase. The other two glycolipids have now been identified by mass spectrometry and nuclear magnetic resonance spectroscopy as monoglucosyl diacylglycerol and glucuronosyl diacylglycerol. These two lipids are synthesized by a single promiscuous glycosyltransferase encoded by the ORF atu2297, with UDP-glucose or UDP-glucuronic acid as sugar donors. The transfer of sugars differing in their chemistry is a novel feature not observed before for lipid glycosyltransferases. Furthermore, this enzyme is the first glucuronosyl diacylglycerol synthase isolated. Deletion mutants of Agrobacterium lacking monoglucosyl diacylglycerol and glucuronosyl diacylglycerol or all glycolipids are not impaired in growth or virulence during infection of tobacco leaf discs. Our data suggest that the four glycolipids and the nonphospholipid diacylglyceryl trimethylhomoserine can mutually replace each other during phosphate deprivation. This redundancy of different nonphospholipids may represent an adaptation mechanism to enhance the competitiveness in nature.


Asunto(s)
Agrobacterium tumefaciens/enzimología , Proteínas Bacterianas/metabolismo , Glucosiltransferasas/metabolismo , Glucolípidos/biosíntesis , Agrobacterium tumefaciens/genética , Proteínas Bacterianas/genética , Glucosiltransferasas/genética , Glucolípidos/genética , Fosfatos/metabolismo , Hojas de la Planta/microbiología , Nicotiana/microbiología , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo , Uridina Difosfato Ácido Glucurónico/genética , Uridina Difosfato Ácido Glucurónico/metabolismo
8.
PLoS Genet ; 9(7): e1003663, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935518

RESUMEN

Growth rate and nutrient availability are the primary determinants of size in single-celled organisms: rapidly growing Escherichia coli cells are more than twice as large as their slow growing counterparts. Here we report the identification of the glucosyltransferase OpgH as a nutrient-dependent regulator of E. coli cell size. During growth under nutrient-rich conditions, OpgH localizes to the nascent septal site, where it antagonizes assembly of the tubulin-like cell division protein FtsZ, delaying division and increasing cell size. Biochemical analysis is consistent with OpgH sequestering FtsZ from growing polymers. OpgH is functionally analogous to UgtP, a Bacillus subtilis glucosyltransferase that inhibits cell division in a growth rate-dependent fashion. In a striking example of convergent evolution, OpgH and UgtP share no homology, have distinct enzymatic activities, and appear to inhibit FtsZ assembly through different mechanisms. Comparative analysis of E. coli and B. subtilis reveals conserved aspects of growth rate regulation and cell size control that are likely to be broadly applicable. These include the conservation of uridine diphosphate glucose as a proxy for nutrient status and the use of moonlighting enzymes to couple growth rate-dependent phenomena to central metabolism.


Asunto(s)
Tamaño de la Célula , Escherichia coli/crecimiento & desarrollo , Glucosiltransferasas/metabolismo , Uridina Difosfato Glucosa/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , División Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Glucosiltransferasas/genética , Uridina Difosfato Glucosa/genética
9.
Planta ; 238(4): 683-93, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23801300

RESUMEN

Flavonoids are predominantly found as glycosides in plants. The glycosylation of flavonoids is mediated by uridine diphosphate-dependent glycosyltransferases (UGT). UGTs attach various sugars, including arabinose, glucose, galactose, xylose, and glucuronic acid, to flavonoid aglycones. Two UGTs isolated from Arabidopsis thaliana, AtUGT78D2 and AtUGT78D3, showed 89 % amino acid sequence similarity (75 % amino acid sequence identity) and both attached a sugar to the 3-hydroxyl group of flavonols using a UDP-sugar. The two enzymes used UDP-glucose and UDP-arabinose, respectively, and AtUGT78D2 was approximately 90-fold more efficient than AtUGT78D3 when judged by the k(cat)/K(m) value. Domain exchanges between AtUGT78D2 and AtUGT78D3 were carried out to find UGTs with better catalytic efficiency for UDP-arabinose and exhibiting dual sugar selectivity. Among 19 fusion proteins examined, three showed dual sugar selectivity, and one fusion protein had better catalytic efficiency for UDP-arabinose compared with AtUGT78D3. Using molecular modeling, the changes in enzymatic properties in the chimeric proteins were elucidated. To the best of our knowledge, this is the first report on the construction of fusion proteins with expanded sugar-donor range and enhanced catalytic efficiencies for sugar donors.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Glicosiltransferasas/genética , Uridina Difosfato Glucosa/metabolismo , Azúcares de Uridina Difosfato/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes de Fusión/genética , Especificidad por Sustrato/genética , Uridina Difosfato Glucosa/genética , Azúcares de Uridina Difosfato/genética
10.
PLoS One ; 7(7): e41515, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848518

RESUMEN

The cyst nematode Heterodera schachtii infects roots of Arabidopsis plants and establishes feeding sites called syncytia, which are the only nutrient source for nematodes. Development of syncytia is accompanied by changes in cell wall structures including the development of cell wall ingrowths. UDP-glucuronic acid is a precursor of several cell wall polysaccharides and can be produced by UDP-glucose dehydrogenase through oxidation of UDP-glucose. Four genes in Arabidopsis encode this enzyme. Promoter::GUS analysis revealed that UGD2 and UGD3 were expressed in syncytia as early as 1 dpi while expression of UGD1 and UGD4 could only be detected starting at 2 dpi. Infection assays showed no differences between Δugd1 and Δugd4 single mutants and wild type plants concerning numbers of males and females and the size of syncytia and cysts. On single mutants of Δugd2 and Δugd3, however, less and smaller females, and smaller syncytia formed compared to wild type plants. The double mutant ΔΔugd23 had a stronger effect than the single mutants. These data indicate that UGD2 and UGD3 but not UGD1 and UGD4 are important for syncytium development. We therefore studied the ultrastructure of syncytia in the ΔΔugd23 double mutant. Syncytia contained an electron translucent cytoplasm with degenerated cellular organelles and numerous small vacuoles instead of the dense cytoplasm as in syncytia developing in wild type roots. Typical cell wall ingrowths were missing in the ΔΔugd23 double mutant. Therefore we conclude that UGD2 and UGD3 are needed for the production of cell wall ingrowths in syncytia and that their lack leads to a reduced host suitability for H. schachtii resulting in smaller syncytia, lower number of developing nematodes, and smaller females.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis , Pared Celular/enzimología , Células Gigantes/enzimología , Interacciones Huésped-Parásitos/fisiología , Nematodos/fisiología , Raíces de Plantas/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo , Animales , Arabidopsis/enzimología , Arabidopsis/genética , Arabidopsis/parasitología , Proteínas de Arabidopsis/genética , Pared Celular/genética , Citoplasma/enzimología , Citoplasma/genética , Femenino , Masculino , Mutación , Enfermedades de las Plantas/parasitología , Raíces de Plantas/genética , Raíces de Plantas/parasitología , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/genética
11.
J Biol Chem ; 287(3): 2119-29, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22123821

RESUMEN

Biosynthesis of UDP-glucuronic acid by UDP-glucose 6-dehydrogenase (UGDH) occurs through the four-electron oxidation of the UDP-glucose C6 primary alcohol in two NAD(+)-dependent steps. The catalytic reaction of UGDH is thought to involve a Cys nucleophile that promotes formation of a thiohemiacetal enzyme intermediate in the course of the first oxidation step. The thiohemiacetal undergoes further oxidation into a thioester, and hydrolysis of the thioester completes the catalytic cycle. Herein we present crystallographic and kinetic evidence for the human form of UGDH that clarifies participation of covalent catalysis in the enzymatic mechanism. Substitution of the putative catalytic base for water attack on the thioester (Glu(161)) by an incompetent analog (Gln(161)) gave a UGDH variant (E161Q) in which the hydrolysis step had become completely rate-limiting so that a thioester enzyme intermediate accumulated at steady state. By crystallizing E161Q in the presence of 5 mm UDP-glucose and 2 mm NAD(+), we succeeded in trapping a thiohemiacetal enzyme intermediate and determined its structure at 2.3 Å resolution. Cys(276) was covalently modified in the structure, establishing its role as catalytic nucleophile of the reaction. The thiohemiacetal reactive C6 was in a position suitable to become further oxidized by hydride transfer to NAD(+). The proposed catalytic mechanism of human UGDH involves Lys(220) as general base for UDP-glucose alcohol oxidation and for oxyanion stabilization during formation and breakdown of the thiohemiacetal and thioester enzyme intermediates. Water coordinated to Asp(280) deprotonates Cys(276) to function as an aldehyde trap and also provides oxyanion stabilization. Glu(161) is the Brønsted base catalytically promoting the thioester hydrolysis.


Asunto(s)
NAD/química , Uridina Difosfato Glucosa Deshidrogenasa/química , Uridina Difosfato Glucosa/química , Sustitución de Aminoácidos , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Cinética , Mutación Missense , NAD/genética , NAD/metabolismo , Oxidación-Reducción , Relación Estructura-Actividad , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo , Uridina Difosfato Glucosa Deshidrogenasa/genética , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo
12.
J Biol Chem ; 286(30): 27048-57, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21653318

RESUMEN

Serine-rich repeat glycoproteins (SRRPs) are a growing family of bacterial adhesins found in many streptococci and staphylococci; they play important roles in bacterial biofilm formation and pathogenesis. Glycosylation of this family of adhesins is essential for their biogenesis. A glucosyltransferase (Gtf3) catalyzes the second step of glycosylation of a SRRP (Fap1) from an oral streptococcus, Streptococcus parasanguinis. Although Gtf3 homologs are highly conserved in SRRP-containing streptococci, they share minimal homology with functionally known glycosyltransferases. We report here the 2.3 Å crystal structure of Gtf3. The structural analysis indicates that Gtf3 forms a tetramer and shares significant structural homology with glycosyltransferases from GT4, GT5, and GT20 subfamilies. Combining crystal structural analysis with site-directed mutagenesis and in vitro glycosyltransferase assays, we identified residues that are required for UDP- or UDP-glucose binding and for oligomerization of Gtf3 and determined their contribution to the enzymatic activity of Gtf3. Further in vivo studies revealed that the critical amino acid residues identified by the structural analysis are crucial for Fap1 glycosylation in S. parasanguinis in vivo. Moreover, Gtf3 homologs from other streptococci were able to rescue the gtf3 knock-out mutant of S. parasanguinis in vivo and catalyze the sugar transfer to the modified SRRP substrate in vitro, demonstrating the importance and conservation of the Gtf3 homologs in glycosylation of SRRPs. As the Gtf3 homologs only exist in SRRP-containing streptococci, we conclude that the Gtf3 homologs represent a unique subfamily of glycosyltransferases.


Asunto(s)
Proteínas Fimbrias/química , Glucosiltransferasas/química , Multimerización de Proteína/fisiología , Streptococcus/enzimología , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Técnicas de Silenciamiento del Gen , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Glicosilación , Mutación , Estructura Cuaternaria de Proteína , Streptococcus/genética , Uridina Difosfato Glucosa/química , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo
13.
J Biol Chem ; 286(27): 23708-16, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21558268

RESUMEN

The biosynthetic enzymes involved in wall teichoic acid biogenesis in gram-positive bacteria have been the subject of renewed investigation in recent years with the benefit of modern tools of biochemistry and genetics. Nevertheless, there have been only limited investigations into the enzymes that glycosylate wall teichoic acid. Decades-old experiments in the model gram-positive bacterium, Bacillus subtilis 168, using phage-resistant mutants implicated tagE (also called gtaA and rodD) as the gene coding for the wall teichoic acid glycosyltransferase. This study and others have provided only indirect evidence to support a role for TagE in wall teichoic acid glycosylation. In this work, we showed that deletion of tagE resulted in the loss of α-glucose at the C-2 position of glycerol in the poly(glycerol phosphate) polymer backbone. We also reported the first kinetic characterization of pure, recombinant wall teichoic acid glycosyltransferase using clean synthetic substrates. We investigated the substrate specificity of TagE using a wide variety of acceptor substrates and found that the enzyme had a strong kinetic preference for the transfer of glucose from UDP-glucose to glycerol phosphate in polymeric form. Further, we showed that the enzyme recognized its polymeric (and repetitive) substrate with a sequential kinetic mechanism. This work provides direct evidence that TagE is the wall teichoic acid glycosyltransferase in B. subtilis 168 and provides a strong basis for further studies of the mechanism of wall teichoic acid glycosylation, a largely uncharted aspect of wall teichoic acid biogenesis.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/metabolismo , Pared Celular/enzimología , Glicosiltransferasas/metabolismo , Ácidos Teicoicos/metabolismo , Bacillus subtilis/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Pared Celular/genética , Glicosilación , Glicosiltransferasas/química , Glicosiltransferasas/genética , Cinética , Mutación , Ácidos Teicoicos/genética , Uridina Difosfato Glucosa/química , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo
14.
Sheng Wu Gong Cheng Xue Bao ; 24(9): 1531-7, 2008 Sep.
Artículo en Chino | MEDLINE | ID: mdl-19160833

RESUMEN

In plants, UDP-L-rhamnose is one of the major components of cell wall skeleton. Rhamnose synthase plays a key role in rhamnose synthesis which converts UDP-D-glucose into UDP-L-rhamnose in plants. In this study, we isolated the 1058 bp promoter region of the rhamnose synthase gene AtRHM1 from Arabidopsis genome by PCR, and created a series of deletions of AtRHM1 promoter ranging from -931 bp to +127 bp. The full length of the promoter and its deletion derivatives fused with GUS reporter gene were introduced into wild-type Arabidopsis by Agrobacterium-mediated transformation respectively. The GUS staining and GUS enzymatic activity assay showed that the expression of AtRHM1 is induced at transcriptional level by glucose and the regulatory elements involved in the glucose response are located in the region of -931 bp - -752 bp which contains three G-box motifs.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Glucosiltransferasas/genética , Regiones Promotoras Genéticas , Azúcares de Uridina Difosfato/genética , Plantas Modificadas Genéticamente/genética , Uridina Difosfato Glucosa/genética , Uridina Difosfato Glucosa/metabolismo , Azúcares de Uridina Difosfato/metabolismo
15.
J Biol Chem ; 282(8): 5389-403, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17190829

RESUMEN

UDP-L-rhamnose is required for the biosynthesis of cell wall rhamnogalacturonan-I, rhamnogalacturonan-II, and natural compounds in plants. It has been suggested that the RHM2/MUM4 gene is involved in conversion of UDP-D-glucose to UDP-L-rhamnose on the basis of its effect on rhamnogalacturonan-I-directed development in Arabidopsis thaliana. RHM2/MUM4-related genes, RHM1 and RHM3, can be found in the A. thaliana genome. Here we present direct evidence that all three RHM proteins have UDP-D-glucose 4,6-dehydratase, UDP-4-keto-6-deoxy-D-glucose 3,5-epimerase, and UDP-4-keto-L-rhamnose 4-keto-reductase activities in the cytoplasm when expressed in the yeast Saccharomyces cerevisiae. Functional domain analysis revealed that the N-terminal region of RHM2 (RHM2-N; amino acids 1-370) has the first activity and the C-terminal region of RHM2 (RHM2-C; amino acids 371-667) has the two following activities. This suggests that RHM2 converts UDP-d-glucose to UDP-L-rhamnose via an UDP-4-keto-6-deoxy-D-glucose intermediate. Site-directed mutagenesis of RHM2 revealed that mucilage defects in MUM4-1 and MUM4-2 mutant seeds of A. thaliana are caused by abolishment of RHM2 enzymatic activity in the mutant strains and furthermore, that the GXXGXX(G/A) and YXXXK motifs are important for enzymatic activity. Moreover, a kinetic analysis of purified His(6)-tagged RHM2-N protein revealed 5.9-fold higher affinity of RHM2 for UDP-D-glucose than for dTDP-D-glucose, the preferred substrate for dTDP-D-glucose 4,6-dehydratase from bacteria. RHM2-N activity is strongly inhibited by UDP-L-rhamnose, UDP-D-xylose, and UDP but not by other sugar nucleotides, suggesting that RHM2 maintains cytoplasmic levels of UDP-D-glucose and UDP-L-rhamnose via feedback inhibition by UDP-L-rhamnose and UDP-D-xylose.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Pared Celular/metabolismo , Genoma de Planta/fisiología , Complejos Multienzimáticos/metabolismo , Ramnosa/metabolismo , Uridina Difosfato Glucosa/metabolismo , Secuencias de Aminoácidos/genética , Arabidopsis/química , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Mutagénesis Sitio-Dirigida , Pectinas/biosíntesis , Pectinas/química , Pectinas/genética , Ramnosa/química , Ramnosa/genética , Saccharomyces cerevisiae/genética , Semillas/química , Semillas/enzimología , Semillas/genética , Uridina Difosfato Glucosa/química , Uridina Difosfato Glucosa/genética , Uridina Difosfato Xilosa/química , Uridina Difosfato Xilosa/genética , Uridina Difosfato Xilosa/metabolismo
16.
Biochem J ; 341 ( Pt 2): 395-400, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10393098

RESUMEN

Glucosylceramide synthase (GCS) catalyses the transfer of glucose from UDP-glucose (UDP-Glc) to ceramide to form glucosylceramide, the common precursor of most higher-order glycosphingolipids. Inhibition of GCS activity has been proposed as a possible target of chemotherapeutic agents for a number of diseases, including cancer. Design of new GCS inhibitors with desirable pharmaceutical properties is hampered by lack of knowledge of the secondary structure or catalytic mechanism of the GCS protein. Thus we cloned the rat homologue of GCS to begin studies to identify its catalytic regions. The histidine-modifying agent diethyl pyrocarbonate (DEPC) inhibited recombinant rat GCS expressed in bacteria; this inhibition was rapidly reversible by hydroxylamine and could be diminished by preincubation of GCS with UDP-Glc. These data suggest that DEPC acts on histidine residues within or near the UDP-Glc-binding site of GCS. Mutant proteins were expressed in which the eight histidine residues in GCS were individually replaced by other amino acids. H193A (His193-->Ala) and H193N (His193-->Asn) mutants were unaffected by 0.1 mM DEPC, a concentration that inhibited other histidine mutants and the wild-type enzyme by at least 60%. These results indicate that His193 is the primary target of DEPC and is at, or near, the UDP-Glc-binding site of GCS. His193 mutants were also insensitive to the GCS inhibitor d-threo-1-phenyl-2- decanoylamino-3-morpholinopropan-1-ol, at concentrations which inhibited the wild-type enzyme by >80%. These results have significance for both an understanding of the GCS active site and also for the possible design of new and specific inhibitors of GCS.


Asunto(s)
Glucosiltransferasas/química , Glucosiltransferasas/genética , Morfolinas/química , Uridina Difosfato Glucosa/química , Uridina Difosfato Glucosa/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Glucosiltransferasas/metabolismo , Histidina , Humanos , Datos de Secuencia Molecular , Morfolinas/metabolismo , Mutagénesis Sitio-Dirigida , Mutación Puntual , Ratas , Alineación de Secuencia , Uridina Difosfato Glucosa/metabolismo
17.
J Biol Chem ; 273(45): 29497-505, 1998 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-9792656

RESUMEN

The major core oligosaccharide biosynthesis operons from prototype Escherichia coli strains displaying R1 and R4 lipopolysaccharide core types were polymerase chain reaction-amplified and analyzed. Comparison of deduced products of the open reading frames between the two regions indicate that all but two share total similarities of 94% or greater. Core oligosaccharide structures resulting from nonpolar insertion mutations in each gene of the core OS biosynthesis operon in the R1 strain allowed assignment of all of the glycosyltransferase enzymes required for outer core assembly. The difference between the R1 and R4 core oligosaccharides results from the specificity of the WaaV protein (a beta1, 3-glucosyltransferase) in R1 and WaaX (a beta1, 4-galactosyltransferase) in R4. Complementation of the waaV mutant of the R1 prototype strain with the waaX gene of the R4 strain converted the core oligosaccharide from an R1- to an R4-type lipopolysaccharide core molecule. Aside from generating core oligosaccharide specificity, the unique beta-linked glucopyranosyl residue of the R1 core plays a crucial role in organization of the lipopolysaccharide. This residue provides a novel attachment site for lipid A-core-linked polysaccharides and distinguishes the R1-type LPS from existing models for enterobacterial lipopolysaccharides.


Asunto(s)
Escherichia coli/metabolismo , Glucosiltransferasas/metabolismo , Antígenos O/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Secuencia de Carbohidratos , Cartilla de ADN , Escherichia coli/genética , Datos de Secuencia Molecular , Antígenos O/química , Operón , Uridina Difosfato Glucosa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA