Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 299(6): 104781, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37146969

RESUMO

Intestinal mucous layers mediate symbiosis and dysbiosis of host-microbe interactions. These interactions are influenced by the mucin O-glycan degrading ability of several gut microbes. The identities and prevalence of many glycoside hydrolases (GHs) involved in microbial mucin O-glycan breakdown have been previously reported; however, the exact mechanisms and extent to which these GHs are dedicated to mucin O-glycan degradation pathways warrant further research. Here, using Bifidobacterium bifidum as a model mucinolytic bacterium, we revealed that two ß-N-acetylglucosaminidases belonging to the GH20 (BbhI) and GH84 (BbhIV) families play important roles in mucin O-glycan degradation. Using substrate specificity analysis of natural oligosaccharides and O-glycomic analysis of porcine gastric mucin (PGM) incubated with purified enzymes or B. bifidum carrying bbhI and/or bbhIV mutations, we showed that BbhI and BbhIV are highly specific for ß-(1→3)- and ß-(1→6)-GlcNAc linkages of mucin core structures, respectively. Interestingly, we found that efficient hydrolysis of the ß-(1→3)-linkage by BbhI of the mucin core 4 structure [GlcNAcß1-3(GlcNAcß1-6)GalNAcα-O-Thr] required prior removal of the ß-(1→6)-GlcNAc linkage by BbhIV. Consistent with this, inactivation of bbhIV markedly decreased the ability of B. bifidum to release GlcNAc from PGM. When combined with a bbhI mutation, we observed that the growth of the strain on PGM was reduced. Finally, phylogenetic analysis suggests that GH84 members may have gained diversified functions through microbe-microbe and host-microbe horizontal gene transfer events. Taken together, these data strongly suggest the involvement of GH84 family members in host glycan breakdown.


Assuntos
Acetilglucosaminidase , Proteínas de Bactérias , Bifidobacterium bifidum , Mucinas , Animais , Acetilglucosaminidase/química , Acetilglucosaminidase/metabolismo , Proteínas de Bactérias/metabolismo , Bifidobacterium bifidum/classificação , Bifidobacterium bifidum/enzimologia , Bifidobacterium bifidum/genética , Mucinas/metabolismo , Filogenia , Suínos
2.
Acta Crystallogr D Struct Biol ; 77(Pt 12): 1564-1578, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34866612

RESUMO

ß-Galactosidases catalyse the hydrolysis of lactose into galactose and glucose; as an alternative reaction, some ß-galactosidases also catalyse the formation of galactooligosaccharides by transglycosylation. Both reactions have industrial importance: lactose hydrolysis is used to produce lactose-free milk, while galactooligosaccharides have been shown to act as prebiotics. For some multi-domain ß-galactosidases, the hydrolysis/transglycosylation ratio can be modified by the truncation of carbohydrate-binding modules. Here, an analysis of BbgIII, a multidomain ß-galactosidase from Bifidobacterium bifidum, is presented. The X-ray structure has been determined of an intact protein corresponding to a gene construct of eight domains. The use of evolutionary covariance-based predictions made sequence docking in low-resolution areas of the model spectacularly easy, confirming the relevance of this rapidly developing deep-learning-based technique for model building. The structure revealed two alternative orientations of the CBM32 carbohydrate-binding module relative to the GH2 catalytic domain in the six crystallographically independent chains. In one orientation the CBM32 domain covers the entrance to the active site of the enzyme, while in the other orientation the active site is open, suggesting a possible mechanism for switching between the two activities of the enzyme, namely lactose hydrolysis and transgalactosylation. The location of the carbohydrate-binding site of the CBM32 domain on the opposite site of the module to where it comes into contact with the catalytic GH2 domain is consistent with its involvement in adherence to host cells. The role of the CBM32 domain in switching between hydrolysis and transglycosylation modes offers protein-engineering opportunities for selective ß-galactosidase modification for industrial purposes in the future.


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium bifidum/metabolismo , beta-Galactosidase/metabolismo , Proteínas de Bactérias/química , Bifidobacterium bifidum/enzimologia , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Galactose/metabolismo , Hidrólise , Lactose/metabolismo , Especificidade por Substrato , beta-Galactosidase/química
3.
J Agric Food Chem ; 68(17): 4930-4938, 2020 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-32279499

RESUMO

The transglycosylation activity of a novel commercial ß-galactosidase from Bifidobacterium bifidum (Saphera) was evaluated. The optimal conditions for the operation of this enzyme, measured with o-nitrophenyl-ß-d-galactopyranoside, were 40 °C and pH around 6.0. Although at low lactose concentrations the property of this enzyme was basically hydrolytic, an increase of lactose concentration to 400 g/L resulted in a significant formation (107.2 g/L, 27% yield) of prebiotic galactooligosaccharides (GOS). The maximum amount of GOS was obtained at a lactose conversion of approximately 90%, which contrasts with other ß-galactosidases, for which the highest GOS yield is achieved at 40-50% lactose conversion. Using high-performance anion-exchange chromatography with pulsed amperometric detection, semipreparative high-performance liquid chromatography-hydrophilic interaction liquid chromatography, mass spectrometry, and 1D and 2D NMR, we determined the structure of most of the GOS synthesized by this enzyme. The main identified products were Gal-ß(1→3)-Gal-ß(1→4)-Glc (3'-O-ß-galactosyl-lactose), Gal-ß(1→6)-Glc (allolactose), Gal-ß(1→3)-Glc (3-galactosyl-glucose), Gal-ß(1→3)-Gal (3-galactobiose), and the tetrasaccharide Gal-ß(1→3)-Gal-ß(1→3)-Gal-ß(1→4)-Glc. In general, B. bifidum ß-galactosidase showed a tendency to form ß(1→3) linkages followed by ß(1→6) and more scarcely ß(1→4).


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium bifidum/enzimologia , Oligossacarídeos/biossíntese , beta-Galactosidase/metabolismo , Proteínas de Bactérias/genética , Bifidobacterium bifidum/química , Bifidobacterium bifidum/genética , Configuração de Carboidratos , Cromatografia Líquida de Alta Pressão , Galactose/metabolismo , Lactose/metabolismo , Espectrometria de Massas , Oligossacarídeos/química , beta-Galactosidase/genética
4.
Biotechnol Prog ; 36(4): e2982, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32083812

RESUMO

Hydrolysis efficiency of ß-galactosidases is affected due to a strong inhibition by galactose, hampering the complete lactose hydrolysis. One alternative to reduce this inhibition is to perform mutations in the enzyme's active site. The aim of this study was to evaluate the effect of point mutations on the active site of different microbial ß-galactosidases, using computational techniques. The enzymes of Aspergillus niger (AnßGal), Aspergillus oryzae (AoßGal), Bacillus circulans (BcßGal), Bifidobacterium bifidum (BbßGal), and Kluyveromyces lactis (KlßGal) were used. The mutations were carried out in all residues that were up to 4.5 Å from the galactose/lactose molecules and binding energy was computed. The mutants Tyr96Ala (AnßGal), Asn140Ala and Asn199Ala (AoßGal), Arg111Ala and Glu355Ala (BcßGal), Arg122Ala and Phe358Ala (BbßGal), Tyr523Ala, Phe620Ala, and Trp582Ala (KlßGal) had the best results, with higher effect on galactose binding energy and lower effect on lactose affinity. To maximize enzyme reactions by reducing galactose affinity, double mutations were proposed for BcßGal, BbßGal, and KlßGal. The double mutations in BcßGal and BbßGal caused the highest reduction in galactose affinity, while no satisfactory results were observed to KlßGal. Using computational tools, mutants that reduced galactose affinity without significantly affecting lactose binding were proposed. The mutations proposed can be used to reduce the negative feedback process, improving the catalytic characteristics of ß-galactosidases and rendering them promising for industrial applications.


Assuntos
Galactose/química , Lactose/química , beta-Galactosidase/genética , Aspergillus niger/enzimologia , Aspergillus oryzae/enzimologia , Bacillus/enzimologia , Bifidobacterium bifidum/enzimologia , Catálise , Hidrólise , Cinética , Kluyveromyces/enzimologia , Mutação Puntual/genética , beta-Galactosidase/química , beta-Galactosidase/ultraestrutura
5.
Biotechnol Bioeng ; 117(5): 1597-1602, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32017022

RESUMO

The D746E variant of Bifidobacterium bifidum ß-N-acetyl-hexosaminidase is a promising glycosynthase (engineered glycosidase deficient in hydrolase activity) for the synthesis of lacto-N-triose II (LNT II), a core structural unit of human milk oligosaccharides. Here, we develop a flow process for the glycosynthase reaction, which is the regioselective ß-1,3-glycosylation of lactose from a d-glucosamine 1,2-oxazoline donor. Using the glycosynthase immobilized on agarose beads (∼30 mg/g) packed into a fixed bed (1 ml), we show stable continuous production of LNT II (145-200 mM) at quantitative yield from the donor substrate. The wild-type ß-N-acetyl-hexosaminidase used under exactly comparable conditions gives primarily (∼85%) the hydrolysis product d-glucosamine. By enabling short residence times (2 min) that are challenging for mixed-vessel types of reactor to establish, the glycosynthase flow reactor succeeds in an effective uncoupling of the LNT II formation (∼80-100 mM/min) from the slower side reactions (decomposition of donor substrate, enzymatic hydrolysis of LNT II) to obtain optimum synthetic efficiency. Our study thus provides a strong case for the application of flow chemistry principles to glycosynthase reactions and by that, it reveals the important synergy between enzyme and reaction engineering for biocatalytic synthesis of oligosaccharides.


Assuntos
Enzimas Imobilizadas , Engenharia Metabólica/métodos , Trissacarídeos , beta-N-Acetil-Hexosaminidases , Bifidobacterium bifidum/enzimologia , Bifidobacterium bifidum/genética , Enzimas Imobilizadas/genética , Enzimas Imobilizadas/metabolismo , Leite Humano/química , Trissacarídeos/análise , Trissacarídeos/metabolismo , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo
6.
Appl Microbiol Biotechnol ; 104(2): 661-673, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31822984

RESUMO

We have recently derived a ß-N-acetylhexosaminidase, BbhI, from Bifidobacterium bifidum JCM 1254, which could regioselectively synthesize GlcNAcß1-3Galß1-4Glc with a yield of 44.9%. Here, directed evolution of BbhI by domain-targeted mutagenesis was carried out. Firstly, the GH20 domain was selected for random mutagenesis using MEGAWHOP method and a small library of 1300 clones was created. A total of 734 colonies with reduced hydrolytic activity were isolated, and three mutants with elevated transglycosylation yields, GlcNAcß1-3Galß1-4Glc yields of 68.5%, 74.7%, and 81.1%, respectively, were obtained. Subsequently, nineteen independent mutants were constructed according to all the mutation sites in these three mutants. After transglycosylation analysis, Asp714 and Trp773 were identified as key residues for improvement in transglycosylation ability and were chosen for the second round of directed evolution by site-saturation mutagenesis. Two most efficient mutants D714T and W773R that acted as trans-ß-N-acetylhexosaminidase were finally achieved. D714T with the substitution at the putative nucleophile assistant residue Asp714 by threonine showed high yield of 84.7% with unobserved hydrolysis towards transglycosylation product. W773R with arginine substitution at Trp773 residue locating at the entrance of catalytic cavity led to the yield up to 81.8%. The kcat/Km values of D714T and W773R for hydrolysis of pNP-ß-GlcNAc displayed drastic decreases. NMR investigation of protein-substrate interaction revealed an invariable mode of the catalytic cavity of D714T, W773R, and WT BbhI. The collective motions of protein model showed the mutations Thr714 and Arg773 exerted little effect on the dynamics of the inside but a large effect on the dynamics of the outside of catalytic cavity.


Assuntos
Bifidobacterium bifidum/enzimologia , Mutagênese , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/metabolismo , Bifidobacterium bifidum/genética , Evolução Molecular Direcionada , Cinética , Espectroscopia de Ressonância Magnética , Proteínas Mutantes/química , beta-N-Acetil-Hexosaminidases/química
7.
Org Biomol Chem ; 17(23): 5661-5665, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31094393

RESUMO

Lacto-N-biose 1,2-oxazoline was prepared chemo-enzymatically and shown to be a donor substrate for ß-1,3-glycosylation of lactose by the wild-type and glycosynthase variants (D320E, D320A, Y419F) of Bifidobacterium bifidum ß-N-hexosaminidase. Lacto-N-tetraose, a core structure of human milk oligosaccharides, was formed in 20-60% yield of donor substrate (up to 8 mM product titre), depending on the degree of selectivity control by the enzyme used.


Assuntos
Bifidobacterium bifidum/enzimologia , Hexosaminidases/metabolismo , Oligossacarídeos/síntese química , Configuração de Carboidratos , Domínio Catalítico , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Variação Genética , Hexosaminidases/química , Hexosaminidases/genética , Isoenzimas , Modelos Moleculares , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Conformação Proteica
8.
Anaerobe ; 57: 45-54, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30880149

RESUMO

Bifidobacterium is beneficial for host health and exhibits different O2 sensitivity levels among species or strains via unknown mechanisms. Bifidobacterium bifidum JCM1255T, a type species of Bifidobacterium, is an O2-sensitive bacterium that can grow under low-O2 (5%) conditions, and the growth of this species is inhibited under high-O2 conditions (10% ∼) with accumulation of H2O2. We previously reported that NADH or NAD(P)H oxidase-active fractions were detected during purification using microaerobically grown B. bifidum cells, and the active enzyme was purified from the NADH oxidase-active fraction. The purified enzyme was identified as b-type dihydroorotate dehydrogenase (DHODb) and characterized as a dominant H2O2 producer in B. bifidum. In this study, we performed further purification of the enzyme from the NAD(P)H oxidase-active fraction and characterized the purified enzyme as a part of the H2O2 degradation system in B. bifidum. This purified enzyme was identified as thioredoxin reductase (TrxR); the NAD(P)H oxidase activity of this enzyme was not expressed in anaerobically grown B. bifidum, and mRNA expression was induced by O2 exposure. Furthermore, the purified B. bifidum TrxR interacted with recombinant alkyl hydroperoxide reductase (rAhpC) and exhibited NAD(P)H peroxidase activity. These results suggest that TrxR responds to O2 and protects B. bifidum from oxidative stress by degrading H2O2 via the TrxR-AhpC system.


Assuntos
Bifidobacterium bifidum/enzimologia , Peróxido de Hidrogênio/metabolismo , Oxidantes/metabolismo , Peroxirredoxinas/metabolismo , Tiorredoxina Dissulfeto Redutase/metabolismo , Anaerobiose , Bifidobacterium bifidum/metabolismo , Oxigênio/metabolismo , Oxigênio/toxicidade , Tiorredoxina Dissulfeto Redutase/isolamento & purificação
9.
Biosci Biotechnol Biochem ; 82(11): 2030-2039, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30027820

RESUMO

Sialidases catalyze the removal of terminal sialic acid from various complex carbohydrates. In the gastrointestinal tract, sialic acid is commonly found in the sugar chain of mucin, and many enteric commensals use mucin as a nutrient source. We previously identified two different sialidase genes in Bifidobacterium bifidum, and one was cloned and expressed as an extracellular protein designated as exo-α-sialidase SiaBb2. The other exo-α-sialidase gene (siabb1) from the same bifidobacterium encodes an extracellular protein (SiaBb1) consisting of 1795 amino acids with a molecular mass of 189 kDa. SiaBb1 possesses a catalytic domain that classifies this enzyme as a glycoside hydrolase family 33 member. SiaBb1 preferentially hydrolyzes α2,3-linked sialic acid over α2,6-linked sialic acid from sialoglycan, which is the same as SiaBb2. However, SiaBb1 has an SGNH hydrolase domain with sialate-O-acetylesterase activity and an N-terminal signal sequence and C-terminal transmembrane region. SiaBb1 is the first bifunctional sialidase identified with esterase activity. Abbreviations: GalNAc: N-acetyl-D-galactosamine; Fuc: L-fucose; Gal: D-galactose.


Assuntos
Acetilesterase/metabolismo , Bifidobacterium bifidum/enzimologia , Neuraminidase/metabolismo , Acetilesterase/química , Acetilesterase/genética , Sequência de Aminoácidos , Domínio Catalítico , Clonagem Molecular , Hidrólise , Mucinas/metabolismo , Neuraminidase/química , Neuraminidase/genética , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
10.
Anaerobe ; 52: 22-28, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29787815

RESUMO

We investigated the roles of extracellular sialidases (SiaBb1 and SiaBb2) in cross-feeding between sialidase-carrying Bifidobacterium bifidum and sialic acid-utilizing Bifidobacterium breve. Using 6' sialyllactose (6'SL) as a carbon source, the number of wild-type B. bifidum cells increased while that of a siabb2-inactivated strain (Δsiabb2) did not. Coculture of these two strains in the presence of 6'SL resulted in similar increase in cell numbers. Coculture of wild-type B. bifidum, but not the Δsiabb2 strain, with sialic acid-utilizing Bifidobacterium breve, which cannot release sialic acids from carbohydrates, in the presence of 6'SL increased the number of B. breve cells. Moreover, when mucin was used as a carbon source, B. breve growth was increased in cocultures with B. bifidum wild-type and Δsiabb2 strains, suggesting that SiaBb1 may be involved. Additionally, B. breve cell numbers increased during cultivation with recombinant SiaBb1-and SiaBb2-treated mucin as the sole carbon source. These results indicated that B. bifidum SiaBb2 liberated sialic acid from sialyl-human milk oligosaccharides and -mucin glycans, supporting the growth of B. breve through sialic acid cross-feeding. SiaBb1 may assist in the degradation of mucin glycan. Collectively, our results revealed that both the B. bifidum extracellular sialidases promote the utilization of sialylated carbohydrates and supply free sialic acid to other Bifidobacterium strains.


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium bifidum/enzimologia , Bifidobacterium breve/crescimento & desenvolvimento , Neuraminidase/metabolismo , Oligossacarídeos/metabolismo , Proteínas de Bactérias/genética , Bifidobacterium bifidum/genética , Bifidobacterium breve/metabolismo , Meios de Cultura/metabolismo , Feminino , Humanos , Lactose/análogos & derivados , Lactose/metabolismo , Leite Humano/microbiologia , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/genética , Polissacarídeos/metabolismo
11.
Appl Environ Microbiol ; 84(13)2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29678922

RESUMO

Gut bacteria provide a rich source of glycosidases that can recognize and/or hydrolyze glycans for nutrition. Interestingly, some glycosidases have also been found to catalyze transglycosylation reactions in vitro and thus can be used for oligosaccharide synthesis. In this work, six putative and one known exo-α-sialidase genes-three from Bacteroides fragilis NCTC9343, three from Clostridium perfringens ATCC 13124, and one known from Bifidobacterium bifidum JCM1254-were subjected to gene cloning and heterogeneous expression in Escherichia coli The recombinant enzymes were purified, characterized for substrate specificity, and screened for transglycosylation activity. A sialidase, named BfGH33C, from B. fragilis NCTC9343 was found to possess excellent transglycosylation activity for the synthesis of sialylated human milk oligosaccharide. The native BfGH33C was a homodimer with a molecular weight of 113.6 kDa. The Km and kcat values for 4-methylumbelliferyl N-acetyl-α-d-neuraminic acid and sialic acid dimer were determined to be 0.06 mM and 283.2 s-1, and 0.75 mM and 329.6 s-1, respectively. The enzyme was able to transfer sialyl from sialic acid dimer or oligomer to lactose with high efficiency and strict α2-6 regioselectivity. The influences of the initial substrate concentration, pH, temperature, and reaction time on transglycosylation were investigated in detail. Using 40 mM sialic acid dimer (or 40 mg/ml oligomer) and 1 M lactose (pH 6.5) at 50°C for 10 min, BfGH33C could specifically produce 6'-sialyllactose, a dominant sialylated human milk oligosaccharide, at a maximal conversion ratio above 20%. It provides a promising alternative to the current chemical and enzymatic methods for obtaining sialylated oligosaccharides.IMPORTANCE Sialylated human milk oligosaccharides are significantly beneficial to the neonate, as they play important roles in supporting resistance to pathogens, gut maturation, immune function, and brain and cognitive development. Therefore, access to the sialylated oligosaccharides has attracted increasing attention both for the study of saccharide functions and for the development of infant formulas that could mimic the nutritional value of human milk. Nevertheless, nine-carbon sialic acids are rather complicated for the traditional chemical modifications, which require multiple protection and deprotection steps to achieve a specific glycosidic bond. Here, the exo-α-sialidase BfGH33C synthesized 6'-sialyllactose in a simple step with high transglycosylation activity and strict regioselectivity. Additionally, it could utilize oligosialic acid, which was newly prepared in an easy, economical way to reduce the substrate cost, as a glycosyl donor. All the studies laid a foundation for the practical use of BfGH33C in large-scale synthesis of sialylated oligosaccharides in the future.


Assuntos
Bacteroides fragilis/enzimologia , Bacteroides fragilis/genética , Lactose/análogos & derivados , Leite Humano/química , Neuraminidase/genética , Neuraminidase/metabolismo , Oligossacarídeos/metabolismo , Bifidobacterium bifidum/enzimologia , Bifidobacterium bifidum/genética , Clonagem Molecular , Clostridium perfringens/enzimologia , Clostridium perfringens/genética , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Fórmulas Infantis , Lactose/biossíntese , Lactose/metabolismo , Modelos Moleculares , Peso Molecular , Neuraminidase/isolamento & purificação , Domínios Proteicos , Proteínas Recombinantes , Alinhamento de Sequência , Análise de Sequência , Ácidos Siálicos/metabolismo , Especificidade por Substrato , Temperatura , Fatores de Tempo
12.
Benef Microbes ; 9(4): 675-682, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29633643

RESUMO

Some food-derived opioid peptides have been reported to cause diseases, such as gastrointestinal inflammation, celiac disease, and mental disorders. Bifidobacterium is a major member of the dominant human gut microbiota, particularly in the gut of infants. In this study, we evaluated the potential of Bifidobacterium in the degradation of food-derived opioid peptides. All strains tested showed some level of dipeptidyl peptidase activity, which is thought to be involved in the degradation of food-derived opioid peptides. However, this activity was higher in bifidobacterial strains that are commonly found in the intestines of human infants, such as Bifidobacterium longum subsp. longum, B. longum subsp. infantis, Bifidobacterium breve and Bifidobacterium bifidum, than in those of other species, such as Bifidobacterium animalis and Bifidobacterium pseudolongum. In addition, some B. longum subsp. infantis and B. bifidum strains showed degradative activity in food-derived opioid peptides such as human and bovine milk-derived casomorphin-7 and wheat gluten-derived gliadorphin-7. A further screening of B. bifidum strains revealed some bifidobacterial strains that could degrade all three peptides. Our results revealed the potential of Bifidobacterium species in the degradation of food-derived opioid peptides, particularly for species commonly found in the intestine of infants. Selected strains of B. longum subsp. infantis and B. bifidum with high degradative capabilities can be used as probiotic microorganisms to eliminate food-derived opioid peptides and contribute to host health.


Assuntos
Bifidobacterium/enzimologia , Intestinos/microbiologia , Peptídeos Opioides/metabolismo , Probióticos , Bifidobacterium bifidum/enzimologia , Bifidobacterium breve/enzimologia , Bifidobacterium longum/enzimologia , Dipeptidil Peptidases e Tripeptidil Peptidases , Alimentos/efeitos adversos , Humanos , Lactente
13.
mBio ; 8(5)2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974612

RESUMO

Bifidobacterium is a natural inhabitant of the human gastrointestinal (GI) tract. We studied the role of the extracellular sialidase (SiaBb2, 835 amino acids [aa]) from Bifidobacterium bifidum ATCC 15696 in mucosal surface adhesion and carbohydrate catabolism. Human milk oligosaccharides (HMOs) or porcine mucin oligosaccharides as the sole carbon source enhanced B. bifidum growth. This was impaired in a B. bifidum ATCC 15696 strain harboring a mutation in the siabb2 gene. Mutant cells in early to late exponential growth phase also showed decreased adhesion to human epithelial cells and porcine mucin relative to the wild-type strain. These results indicate that SiaBb2 removes sialic acid from HMOs and mucin for metabolic purposes and may promote bifidobacterial adhesion to the mucosal surface. To further characterize SiaBb2-mediated bacterial adhesion, we examined the binding of His-tagged recombinant SiaBb2 peptide to colonic mucins and found that His-SiaBb2 as well as a conserved sialidase domain peptide (aa 187 to 553, His-Sia) bound to porcine mucin and murine colonic sections. A glycoarray assay revealed that His-Sia bound to the α2,6-linked but not to the α2,3-linked sialic acid on sialyloligosaccharide and blood type A antigen [GalNAcα1-3(Fucα1-2)Galß] at the nonreducing termini of sugar chains. These results suggest that the sialidase domain of SiaBb2 is responsible for this interaction and that the protein recognizes two distinct carbohydrate structures. Thus, SiaBb2 may be involved in Bifidobacterium-mucosal surface interactions as well as in the assimilation of a variety of sialylated carbohydrates.IMPORTANCE Adhesion to the host mucosal surface and carbohydrate assimilation are important for bifidobacterium colonization and survival in the host gastrointestinal tract. In this study, we investigated the mechanistic basis for B. bifidum extracellular sialidase (SiaBb2)-mediated adhesion. SiaBb2 cleaved sialyl-human milk oligosaccharides and mucin glycans to produce oligosaccharides that supported B. bifidum growth. Moreover, SiaBb2 enhanced B. bifidum adhesion to mucosal surfaces via specific interactions with the α2,6 linkage of sialyloligosaccharide and blood type A antigen on mucin carbohydrates. These findings provide insight into the bifunctional role of SiaBb2 and the adhesion properties of B. bifidum strains.


Assuntos
Aderência Bacteriana , Bifidobacterium bifidum/enzimologia , Bifidobacterium bifidum/fisiologia , Metabolismo dos Carboidratos , Neuraminidase/metabolismo , Animais , Bifidobacterium bifidum/efeitos dos fármacos , Bifidobacterium bifidum/genética , Colo/microbiologia , Células Epiteliais/microbiologia , Humanos , Camundongos , Mucinas/metabolismo , Oligossacarídeos/química , Oligossacarídeos/farmacologia , Polissacarídeos/metabolismo , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Suínos
14.
Biosci Biotechnol Biochem ; 81(10): 2018-2027, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28814130

RESUMO

Human gut symbiont bifidobacteria possess carbohydrate-degrading enzymes that act on the O-linked glycans of intestinal mucins to utilize those carbohydrates as carbon sources. However, our knowledge about mucin type O-glycan degradation by bifidobacteria remains fragmentary, especially regarding how they decompose sulfated glycans, which are abundantly found in mucin sugar-chains. Here, we examined the abilities of several Bifidobacterium strains to degrade a sulfated glycan substrate and identified a 6-sulfo-ß-d-N-acetylglucosaminidase, also termed sulfoglycosidase, encoded by bbhII from Bifidobacterium bifidum JCM 7004. A recombinant BbhII protein showed a substrate preference toward 6-sulfated and 3,4-disulfated N-acetylglucosamines over non-sulfated and 3-sulfated N-acetylglucosamines. The purified BbhII directly released 6-sulfated N-acetylglucosamine from porcine gastric mucin and the expression of bbhII was moderately induced in the presence of mucin. This de-capping activity may promote utilization of sulfated glycans of mucin by other bacteria including bifidobacteria, thereby establishing the symbiotic relationship between human and gut microbes.


Assuntos
Acetilglucosaminidase/metabolismo , Bifidobacterium bifidum/enzimologia , Mucinas/metabolismo , Polissacarídeos/metabolismo , Acetilglucosaminidase/química , Acetilglucosaminidase/genética , Sequência de Aminoácidos , Bifidobacterium bifidum/genética , Bifidobacterium bifidum/metabolismo , Regulação Bacteriana da Expressão Gênica
15.
J Biol Chem ; 292(29): 12126-12138, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28546425

RESUMO

The α-N-acetylgalactosaminidase from the probiotic bacterium Bifidobacterium bifidum (NagBb) belongs to the glycoside hydrolase family 129 and hydrolyzes the glycosidic bond of Tn-antigen (GalNAcα1-Ser/Thr). NagBb is involved in assimilation of O-glycans on mucin glycoproteins by B. bifidum in the human gastrointestinal tract, but its catalytic mechanism has remained elusive because of a lack of sequence homology around putative catalytic residues and of other structural information. Here we report the X-ray crystal structure of NagBb, representing the first GH129 family structure, solved by the single-wavelength anomalous dispersion method based on sulfur atoms of the native protein. We determined ligand-free, GalNAc, and inhibitor complex forms of NagBb and found that Asp-435 and Glu-478 are located in the catalytic domain at appropriate positions for direct nucleophilic attack at the anomeric carbon and proton donation for the glycosidic bond oxygen, respectively. A highly conserved Asp-330 forms a hydrogen bond with the O4 hydroxyl of GalNAc in the -1 subsite, and Trp-398 provides a stacking platform for the GalNAc pyranose ring. Interestingly, a metal ion, presumably Ca2+, is involved in the recognition of the GalNAc N-acetyl group. Mutations at Asp-435, Glu-478, Asp-330, and Trp-398 and residues involved in metal coordination (including an all-Ala quadruple mutant) significantly reduced the activity, indicating that these residues and the metal ion play important roles in substrate recognition and catalysis. Interestingly, NagBb exhibited some structural similarities to the GH101 endo-α-N-acetylgalactosaminidases, but several critical differences in substrate recognition and reaction mechanism account for the different activities of these two enzymes.


Assuntos
Acetilgalactosamina/metabolismo , Proteínas de Bactérias/metabolismo , Bifidobacterium bifidum/enzimologia , Coenzimas/metabolismo , Glicosídeo Hidrolases/metabolismo , Metais/metabolismo , alfa-N-Acetilgalactosaminidase/metabolismo , Acetilgalactosamina/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Domínio Catalítico , Coenzimas/química , Sequência Conservada , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Glicosídeo Hidrolases/antagonistas & inibidores , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/genética , Ligantes , Metais/química , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Probióticos , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína , alfa-N-Acetilgalactosaminidase/antagonistas & inibidores , alfa-N-Acetilgalactosaminidase/química , alfa-N-Acetilgalactosaminidase/genética
16.
FEBS J ; 283(22): 4097-4112, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27685756

RESUMO

In a search for better comprehension of ß-galactosidase function and specificity, we solved the crystal structures of the GH42 ß-galactosidase BbgII from Bifidobacterium bifidum S17, a well-adapted probiotic microorganism from the human digestive tract, and its complex with d-α-galactose. BbgII is a three-domain molecule that forms barrel-shaped trimers in solution. BbgII interactions with d-α-galactose, a competitive inhibitor, showed a number of residues that are involved in the coordination of ligands. A combination of site-directed mutagenesis of these amino acid residues with enzymatic activity measurements confirmed that Glu161 and Glu320 are fundamental for catalysis and their substitution by alanines led to catalytically inactive mutants. Mutation Asn160Ala resulted in a two orders of magnitude decrease of the enzyme kcat without significant modification in its Km , whereas mutations Tyr289Phe and His371Phe simultaneously decreased kcat and increased Km values. Enzymatic activity of Glu368Ala mutant was too low to be detected. Our docking and molecular dynamics simulations showed that the enzyme recognizes and tightly binds substrates with ß1→6 and ß1→3 bonds, while binding of the substrates with ß1→4 linkages is less favorable. DATABASE: Structural data are available in the PDB under the accession numbers 4UZS and 4UCF.


Assuntos
Proteínas de Bactérias/metabolismo , Bifidobacterium bifidum/enzimologia , Galactose/metabolismo , Galactosidases/metabolismo , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bifidobacterium bifidum/genética , Sítios de Ligação/genética , Biocatálise/efeitos dos fármacos , Domínio Catalítico , Cristalografia por Raios X , Galactose/química , Galactose/farmacologia , Galactosidases/química , Galactosidases/genética , Cinética , Conformação Molecular , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto , Domínios Proteicos , Multimerização Proteica , Especificidade por Substrato
17.
Glycobiology ; 26(11): 1235-1247, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27550195

RESUMO

Fucα1-2 Gal linkages, or H-antigens, constitute histo-blood group antigens and are involved in various physiological processes. In addition, recent studies have shown that the H-antigen-containing glycans play an important role, not only in establishing harmonious relationship between gut microbes and the host, but also in preventing gut dysbiosis-related diseases. Therefore, development of an efficient method for introducing Fuc residue at Gal residue at the nonreducing end of glycans via α-(1→2) linkage is desired for research as well as medicinal purposes. In this study, we succeeded in derivatizing inverting 1,2-α-l-fucosidase (AfcA) into a highly efficient 1,2-α-l-fucosynthase. The synthase specifically synthesized H type 1-, type 2-, type 3- and type 4-chain-containing oligosaccharides with yields of 57-75% based on acceptor depletion. The synthase was also able to specifically introduce Fuc residues into Lewis a/x antigens to produce Lewis b/y antigens, with yields of 43% and 62%, respectively. In addition, the enzyme efficiently introduced H-antigens into sugar chains of porcine gastric mucins, as revealed by lectin blotting and mass spectroscopy analysis of the sugars. Detailed acceptor specificity analysis using various monosaccharides and oligosaccharides unraveled unique substrate recognition feature of this synthase at the subsite (+1), which can be explained by our previous X-ray crystallographic study of AfcA. These results show that the synthase developed in this study could serve as an alternative to other H-antigen synthesis methods involving α-1,2-fucosyltransferases and retaining α-fucosidase.


Assuntos
Antígenos de Bactérias/metabolismo , Glicoproteínas/metabolismo , Oligossacarídeos/metabolismo , Açúcares/metabolismo , alfa-L-Fucosidase/metabolismo , Antígenos de Bactérias/química , Bifidobacterium bifidum/enzimologia , Biocatálise , Configuração de Carboidratos , Glicoproteínas/química , Modelos Moleculares , Oligossacarídeos/química , Açúcares/química
18.
Appl Environ Microbiol ; 82(18): 5642-52, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27422836

RESUMO

UNLABELLED: ß-N-Acetylhexosaminidases have attracted interest particularly for oligosaccharide synthesis, but their use remains limited by the rarity of enzyme sources , low efficiency, and relaxed regioselectivity of transglycosylation. In this work, genes of 13 ß-N-acetylhexosaminidases, including 5 from Bacteroides fragilis ATCC 25285, 5 from Clostridium perfringens ATCC 13124, and 3 from Bifidobacterium bifidum JCM 1254, were cloned and heterogeneously expressed in Escherichia coli The resulting recombinant enzymes were purified and screened for transglycosylation activity. A ß-N-acetylhexosaminidase named BbhI, which belongs to glycoside hydrolase family 20 and was obtained from B. bifidum JCM 1254, possesses the bifunctional property of efficiently transferring both GalNAc and GlcNAc residues through ß1-3 linkage to the Gal residue of lactose. The effects of initial substrate concentration, pH, temperature, and reaction time on transglycosylation activities of BbhI were studied in detail. With the use of 10 mM pNP-ß-GalNAc or 20 mM pNP-ß-GlcNAc as the donor and 400 mM lactose as the acceptor in phosphate buffer (pH 5.8), BbhI synthesized GalNAcß1-3Galß1-4Glc and GlcNAcß1-3Galß1-4Glc at maximal yields of 55.4% at 45°C and 4 h and 44.9% at 55°C and 1.5 h, respectively. The model docking of BbhI with lactose showed the possible molecular basis of strict regioselectivity of ß1-3 linkage in ß-N-acetylhexosaminyl lactose synthesis. IMPORTANCE: Oligosaccharides play a crucial role in many biological events and therefore are promising potential therapeutic agents. However, their use is limited because large-scale production of oligosaccharides is difficult. The chemical synthesis requires multiple protecting group manipulations to control the regio- and stereoselectivity of glycosidic bonds. In comparison, enzymatic synthesis can produce oligosaccharides in one step by using glycosyltransferases and glycosidases. Given the lower price of their glycosyl donor and their broader acceptor specificity, glycosidases are more advantageous than glycosyltransferases for large-scale synthesis. ß-N-Acetylhexosaminidases have attracted interest particularly for ß-N-acetylhexosaminyl oligosaccharide synthesis, but their application is affected by having few enzyme sources, low efficiency, and relaxed regioselectivity of transglycosylation. In this work, we describe a microbial ß-N-acetylhexosaminidase that exhibited strong transglycosylation activity and strict regioselectivity for ß-N-acetylhexosaminyl lactose synthesis and thus provides a powerful synthetic tool to obtain biologically important GalNAcß1-3Lac and GlcNAcß1-3Lac.


Assuntos
Acetilgalactosamina/metabolismo , Acetilglucosamina/metabolismo , Bifidobacterium bifidum/enzimologia , Lactose/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo , Bacteroides fragilis/enzimologia , Bacteroides fragilis/genética , Bifidobacterium bifidum/genética , Clonagem Molecular , Clostridium perfringens/enzimologia , Clostridium perfringens/genética , Estabilidade Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Concentração de Íons de Hidrogênio , Cinética , Simulação de Acoplamento Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Temperatura , beta-N-Acetil-Hexosaminidases/química , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/isolamento & purificação
19.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 4): 288-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27050262

RESUMO

Given the current interest in second-generation biofuels, carbohydrate-active enzymes have become the most important tool to overcome the structural recalcitrance of the plant cell wall. While some glycoside hydrolase families have been exhaustively described, others remain poorly characterized, especially with regard to structural information. The family 43 glycoside hydrolases are a diverse group of inverting enzymes; the available structure information on these enzymes is mainly from xylosidases and arabinofuranosidase. Currently, only one structure of an exo-ß-1,3-galactanase is available. Here, the production, crystallization and structure determination of a putative exo-ß-1,3-galactanase from Bifidobacterium bifidum S17 (BbGal43A) are described. BbGal43A was successfully produced and showed activity towards synthetic galactosides. BbGal43A was subsequently crystallized and data were collected to 1.4 Šresolution. The structure shows a single-domain molecule, differing from known homologues, and crystal contact analysis predicts the formation of a dimer in solution. Further biochemical studies are necessary to elucidate the differences between BbGal43A and its characterized homologues.


Assuntos
Bifidobacterium bifidum/enzimologia , Enzimas/metabolismo , Galactanos/metabolismo , Sequência de Aminoácidos , Clonagem Molecular , Cristalografia por Raios X , Enzimas/química , Conformação Proteica , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...