Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Gastroenterol Hepatol ; 39(3): 480-488, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38149305

RESUMO

BACKGROUND AND AIM: Potassium-competitive acid blockers more strongly suppress the gastric acid barrier than proton pump inhibitors and cause dysbiosis. However, preventive measures in this regard have not been established. We aimed to evaluate whether 1-kestose, a known prebiotic, was effective at alleviating dysbiosis caused by potassium-competitive acid blockers. METHODS: Patients scheduled to undergo endoscopic resection for superficial gastroduodenal tumors were enrolled and randomized 1:1 to receive either 1-kestose or placebo. All patients were started on potassium-competitive acid blocker (vonoprazan 20 mg/day) and took 1-kestose 10 g/day or placebo (maltose) 5 g/day for 8 weeks. The primary outcome was the effect of 1-kestose on potassium-competitive acid blocker-induced alterations in the microbiome. The fecal microbiome was analyzed before and after potassium-competitive acid blocker treatment via MiSeq (16S rRNA gene V3-V4 region). RESULTS: Forty patients were enrolled, and 16 in each group were analyzed. In the placebo group, the Simpson index, an alpha diversity, was significantly decreased and relative abundance of Streptococcus was significantly increased by 1.9-fold. In the kestose group, the Simpson index did not change significantly and relative abundance of Streptococcus increased 1.3-fold, but this was not a significant change. In both groups, no adverse events occurred, ulcers were well healed, and pretreatment and posttreatment short-chain fatty acid levels did not differ. CONCLUSIONS: The potassium-competitive acid blocker caused dysbiosis in the placebo group; this effect was prevented by 1-kestose. Thus, 1-kestose may be useful in dysbiosis treatment.


Assuntos
Disbiose , Microbiota , Pirróis , Sulfonamidas , Trissacarídeos , Humanos , Disbiose/etiologia , RNA Ribossômico 16S , Projetos Piloto , Inibidores da Bomba de Prótons/efeitos adversos , Potássio
2.
BMC Microbiol ; 23(1): 266, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37737162

RESUMO

BACKGROUND: It has become clear that the intestinal microbiota plays a role in food allergies. The objective of this study was to assess the food allergy-preventive effects of combined intake of a short fructan (1-kestose [Kes]) and a long fructan (inulin ([Inu]) in an ovalbumin (OVA)-induced food allergy mouse model. RESULTS: Oral administration of fructans lowered the allergenic symptom score and alleviated the decreases in rectal temperature and total IgA levels and increases in OVA-specific IgE and IgA levels induced by high-dose OVA challenge, and in particular, combined intake of Kes and Inu significantly suppressed the changes in all these parameters. The expression of the pro-inflammatory cytokine IL-4, which was increased in the allergy model group, was significantly suppressed by fructan administration, and the expression of the anti-inflammatory cytokine IL-10 was significantly increased upon Kes administration. 16 S rRNA amplicon sequencing of the gut microbiota and beta diversity analysis revealed that fructan administration may induce gut microbiota resistance to food allergy sensitization, rather than returning the gut microbiota to a non-sensitized state. The relative abundances of the genera Parabacteroides B 862,066 and Alloprevotella, which were significantly reduced by food allergy sensitization, were restored by fructan administration. In Parabacteroides, the relative abundances of Parabacteroides distasonis, Parabacteroides goldsteinii, and their fructan-degrading glycoside hydrolase family 32 gene copy numbers were increased upon Kes or Inu administration. The concentrations of short-chain fatty acids (acetate and propionate) and lactate were increased by fructan administration, especially significantly in the Kes + Inu, Kes, and Inu-fed (Inu, Kes + Inu) groups. CONCLUSION: Combined intake of Kes and Inu suppressed allergy scores more effectively than single intake, suggesting that Kes and Inu have different allergy-preventive mechanisms. This indicates that the combined intake of these short and long fructans may have an allergy-preventive benefit.


Assuntos
Hipersensibilidade Alimentar , Microbioma Gastrointestinal , Animais , Camundongos , Frutanos/farmacologia , Hipersensibilidade Alimentar/prevenção & controle , Citocinas , Imunoglobulina A
3.
Pediatr Res ; 94(3): 1067-1074, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36918660

RESUMO

BACKGROUND: Interventions targeting the gut microbiota for treating food allergy (FA) have been gaining much attention. Although several studies have examined the effects of probiotics, few have verified the effects of prebiotic intervention on FA in humans. METHODS: We conducted a preliminary open-label, parallel-group comparison trial in children diagnosed with severe cow's milk allergy (CMA) who were instructed to ingest baked milk (BM; bread or cookies) daily. The subjects either received or did not receive the prebiotic 1-kestose (kestose) daily for 6 months. CMA symptoms and the threshold dose for milk protein were evaluated by oral food challenge with heated milk or BM. Blood and fecal samples were also collected for investigations of the antigen-specific immunoglobulin (Ig) E levels and microbiota composition. RESULTS: Kestose treatment significantly increased the threshold dose for milk protein, and decreased the milk- and casein-specific IgE levels in serum. In those treated with kestose, the abundance of Fusicatenibacter spp. significantly increased in the feces, and a significant inverse correlation was seen between the abundance of Fusicatenibacter spp. and the milk- and casein-specific IgE levels. CONCLUSION: Kestose treatment induced some tolerance to milk protein via changes in the gut microbiota composition in children with FA. IMPACT: A 6-month treatment with the prebiotic kestose increased the threshold dose for milk protein, and decreased the serum levels of milk- and casein-specific IgE in children diagnosed with cow's milk allergy. The kestose treatment increased the abundance of Fusicatenibacter spp. in the gut, which was inversely correlated with the antigen-specific IgE levels. This is the first study to demonstrate that a prebiotic intervention induced some tolerance to an allergen in children with food allergy.


Assuntos
Hipersensibilidade Alimentar , Microbioma Gastrointestinal , Hipersensibilidade a Leite , Animais , Bovinos , Feminino , Humanos , Criança , Lactente , Hipersensibilidade a Leite/diagnóstico , Caseínas , Proteínas do Leite , Imunoglobulina E , Alérgenos
4.
Scand J Gastroenterol ; 58(2): 193-198, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36036243

RESUMO

BACKGROUND: The relationship between pancreatic ductal adenocarcinoma (PDAC) and the intestinal environment is not fully understood. The purpose of this study was to elucidate the characteristics of the intestinal environment in PDAC. METHODS: We performed a case-control study of 5 Japanese patients with unresectable PDAC located in the body or tail (PDAC-bt). The number of patients analyzed was limited for this preliminary study. We included 68 healthy subjects, herein control, of pre-printed study in the preliminary study. 16S rRNA amplicon sequencing and metabolomic analysis were performed using fecal samples from the subjects. RESULTS: There was no difference in the Shannon index and Principal Coordinate Analysis between PDAC-bt and the control. However, a significant increase in oral-associated bacteria (Actinomyces, Streptococcus, Veillonella, Lactobacillus) was observed. A significant decrease of Anaerostipes was demonstrated in the feces of PDAC-bt compared with the control. The intestinal propionic acid and deoxycholic acid were significantly lower in PDAC-bt compared with the control. CONCLUSIONS: We showed that the intestinal environment of PDAC-bt is characterized by an increase in oral-associated bacteria and an imbalance of metabolites but without changes in alpha and beta diversity of the gut microbiota profiles.Clinical Trial Registration: www.umin.ac.jp, UMIN 000041974, 000023675, 000023970.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Estudos de Casos e Controles , RNA Ribossômico 16S/genética , População do Leste Asiático , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Intestinos/patologia , Bactérias/genética , Neoplasias Pancreáticas
5.
BMC Vet Res ; 19(1): 146, 2023 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-37679756

RESUMO

BACKGROUND: Erythritol was found to inhibit the growth of microorganisms. The present study aimed to demonstrate the growth inhibition of Staphylococcus pseudintermedius by erythritol and to define the changes in gene transcription signatures induced by erythritol. Changes in the gene transcription profiles were analysed by RNA sequencing and quantitative reverse transcription PCR. Gene ontology analysis was performed to assign functional descriptions to the genes. RESULTS: Erythritol inhibited S. pseudintermedius growth in a dose-dependent manner. We then performed a transcriptome analysis of S. pseudintermedius with and without 5% (w/w) erythritol exposure to validate the mechanism of growth inhibition. We revealed that erythritol induced up-regulation of three genes (ptsG, ppdK, and ppdkR) that are related to the phosphoenolpyruvate-dependent sugar phosphotransferase system (PTS). Glucose supplementation restored the up-regulation of the PTS-related genes in response to erythritol. In addition, erythritol down-regulated eleven genes that are located in a single pur-operon and inhibited biofilm formation of S. pseudintermedius. CONCLUSIONS: These findings indicated that erythritol antagonistically inhibits PTS-mediated glucose uptake, thereby exerting a growth inhibitory effect on S. pseudintermedius. Moreover, erythritol inhibits the 'de novo' IMP biosynthetic pathway that may contribute to biofilm synthesis in S. pseudintermedius.


Assuntos
Eritritol , Transcriptoma , Animais , Proliferação de Células , Eritritol/farmacologia , Biofilmes
6.
Biosci Biotechnol Biochem ; 87(9): 981-990, 2023 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-37280168

RESUMO

The trisaccharide 1-kestose, a major constituent of fructooligosaccharide, has strong prebiotic effects. We used high-performance liquid chromatography and 1H nuclear magnetic resonance spectroscopy to show that BiBftA, a ß-fructosyltransferase belonging to glycoside hydrolase family 68, from Beijerinckia indica subsp. indica catalyzes transfructosylation of sucrose to produce mostly 1-kestose and levan polysaccharides. We substituted His395 and Phe473 in BiBftA with Arg and Tyr, respectively, and analyzed the reactions of the mutant enzymes with 180 g/L sucrose. The ratio of the molar concentrations of glucose and 1-kestose in the reaction mixture with wild-type BiBftA was 100:8.1, whereas that in the reaction mixture with the variant H395R/F473Y was 100:45.5, indicating that H395R/F473Y predominantly accumulated 1-kestose from sucrose. The X-ray crystal structure of H395R/F473Y suggests that its catalytic pocket is unfavorable for binding of sucrose while favorable for transfructosylation.


Assuntos
Proteínas de Bactérias , Hexosiltransferases , Hexosiltransferases/genética , Hexosiltransferases/metabolismo , Sacarose/metabolismo
7.
Langenbecks Arch Surg ; 408(1): 364, 2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37725176

RESUMO

PURPOSE: Postoperative diarrhea (PD) remains one of the significant complications. Only a few studies focused on PD after minimally invasive surgery. We aimed to investigate PD after minimally invasive gastrectomy for gastric cancer. METHODS: A total of 1476 consecutive patients with gastric cancer undergoing laparoscopic or robotic gastrectomy between 2009 and 2019 at our institution were retrospectively reviewed. PD was defined as continuous diarrhea for ≥ 2 days, positive stool culture, or positive clostridial antigen test. The incidence, causes, and related clinical factors were analyzed. RESULTS: Of the 1476 patients, the median age was 69 years. Laparoscopic and robotic approaches were performed in 1072 (72.6%) and 404 (27.4%), respectively. Postoperative complications with Clavien-Dindo classification grade of ≥ IIIa occurred in 108 (7.4%) patients. PD occurred in 89 (6.0%) patients. Of the 89 patients with PD, Clostridium difficile, enteropathogenic Escherichia coli, and methicillin-resistant Staphylococcus aureus were detected in 24 (27.0%), 16 (33.3%), and 7 (14.6%) patients, respectively. Multivariate analysis revealed that age ≥ 75 years (OR 1.62, 95% CI [1.02-2.60], p = 0.042) and postoperative complications (OR 6.04, 95% CI [3.54-10.32], p < 0.001) were independent risk factors for PD. In patients without complications, TG (OR 1.88) and age of ≥ 75 years(OR 1.71) were determined as independent risk factors. CONCLUSION: The incidence of PD following minimally invasive gastrectomy for gastric cancer was 6.0%. Older age and TG were obvious risk factors in such a surgery, with the latter being a significant risk even in the absence of complications.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Neoplasias Gástricas , Humanos , Idoso , Neoplasias Gástricas/cirurgia , Relevância Clínica , Incidência , Estudos Retrospectivos , Diarreia , Gastrectomia/efeitos adversos , Complicações Pós-Operatórias/epidemiologia
8.
Biochem Biophys Res Commun ; 613: 81-86, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35537289

RESUMO

Butyrate producing bacteria are one of the major components of the human gut microbiota. Their major metabolite, butyrate, has several beneficial properties for host health. Fructooligosaccharides (FOSs) are well documented prebiotics and are hydrolyzed by intracellular glycoside hydrolase family 32 (GH32) enzyme in several butyrate producers, whereas butyrate producers Anaerostipes hadrus and Anaerostipes butyraticus possess extracellular GH32 enzymes. The present study characterized the extracellular GH32 enzymes in the organisms to consider possible cross-feeding of FOSs with other microbes. Culture supernatant of A. hadrus actively hydrolyzed kestose and nystose, i.e., degrees of polymerization 3 and 4 FOSs, respectively, whereas that of A. butyraticus did not hydrolyzed. When co-cultured with Lacticaseibacillus rhamnosus GG in the presence of nystose, which was negative for growth on the FOSs but positive for growth on FOS degradants, A. hadrus promoted the growth of L. rhamnosus GG, but A. butyraticus did not. The observed negative results in A. butyraticus would be due to the presence of a stop codon in the gene encoding extracellular GH32. Genomic analysis revealed that A. hadrus conserved a single extracellular GH32 enzyme at the species level. The enzyme was phylogenetically distinguished into two groups, but the two groups shared similar FOS degradation properties. The results obtained here suggested that A. hadrus is active for extracellular degradation of FOSs and provides its degradants to other microbes. This study provides a basis of knowledge to understand how ingested FOSs are co-metabolized in gut microbiota.


Assuntos
Microbioma Gastrointestinal , Oligossacarídeos , Butiratos/metabolismo , Clostridiales , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Humanos , Oligossacarídeos/metabolismo , Prebióticos
9.
Br J Nutr ; 127(4): 513-525, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-33849681

RESUMO

SCFA increase serotonin (5-hydroxytryptamine, 5-HT) synthesis and content in the colon in vitro and ex vivo, but little is known in vivo. We tested whether dietary indigestible saccharides, utilised as a substrate to produce SCFA by gut microbiota, would increase colonic 5-HT content in mice. Male C57BL/6J mice were fed a purified diet and water supplemented with 4 % (w/v) 1-kestose (KES) for 2 weeks. Colonic 5-HT content and enterochromaffin (EC) cell numbers were lower in mice supplemented with KES than those without supplementation, while monoamine oxidase A activity and mRNA levels of tryptophan hydroxylase 1 (Tph1), chromogranin A (Chga), Slc6a4 and monoamine oxidase A (Maoa) genes in the colonic mucosa, serum 5-HT concentration and total 5-HT content in the colonic contents did not differ between groups. Caecal acetate concentration and Bifidobacterium pseudolongum population were higher in KES-supplemented mice. Similar trends were observed in mice supplemented with other indigestible saccharides, that is, fructo-oligosaccharides, inulin and raffinose. Intragastric administration of live B. pseudolongum (108 colony-forming units/d) for 2 weeks reduced colonic 5-HT content and EC cell numbers. These results suggest that changes in synthesis, reuptake, catabolism and overflow of 5-HT in the colonic mucosa are not involved in the reduction of colonic 5-HT content by dietary indigestible saccharides in mice. We propose that gut microbes including B. pseudolongum could contribute to the reduction of 5-HT content in the colonic mucosa via diminishing EC cells.


Assuntos
Colo , Serotonina , Animais , Bifidobacterium , Colo/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monoaminoxidase/metabolismo , Serotonina/metabolismo
10.
Appl Microbiol Biotechnol ; 106(7): 2455-2470, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35267055

RESUMO

Fructooligosaccharide is a mixture of mostly the trisaccharide 1-kestose (GF2), tetrasaccharide nystose (GF3), and fructosyl nystose (GF4). Enzymes that hydrolyze GF3 may be useful for preparing GF2 from the fructooligosaccharide mixture. A ß-fructofuranosidase belonging to glycoside hydrolase family 32 (GH32) from the honeybee gut bacterium Frischella perrara (FperFFase) was expressed in Escherichia coli and purified. The time course of the hydrolysis of 60 mM sucrose, GF2, and GF3 by FperFFase was analyzed, showing that the hydrolytic activity of FperFFase for trisaccharide GF2 was lower than those for disaccharide sucrose and tetrasaccharide GF3. The crystal structure of FperFFase and its structure in complex with fructose were determined. FperFFase was found to be structurally homologous to bifidobacterial ß-fructofuranosidases even though bifidobacterial enzymes preferably hydrolyze GF2 and the amino acid residues interacting with fructose at subsite - 1 are mostly conserved between them. A proline residue was inserted between Asp298 and Ser299 using site-directed mutagenesis, and the activity of the variant 298P299 was measured. The ratio of activities for 60 mM GF2/GF3 by wild-type FperFFase was 35.5%, while that of 298P299 was 23.6%, indicating that the structure of the loop comprising Trp297-Asp298-Ser299 correlated with the substrate preference of FperFFase. The crystal structure also shows that a loop consisting of residues 117-127 is likely to contribute to the substrate binding of FperFFase. The results obtained herein suggest that FperFFase is potentially useful for the manufacture of GF2. KEY POINTS: • Frischella ß-fructofuranosidase hydrolyzed nystose more efficiently than 1-kestose. • Trp297-Asp298-Ser299 was shown to be correlated with the substrate preference. • Loop consisting of residues 117-127 appears to contribute to the substrate binding.


Assuntos
Oligossacarídeos , beta-Frutofuranosidase , Animais , Abelhas , Frutose , Gammaproteobacteria , Oligossacarídeos/metabolismo , Sacarose , Trissacarídeos/metabolismo , beta-Frutofuranosidase/metabolismo
11.
Biochem Biophys Res Commun ; 534: 808-814, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33162030

RESUMO

The role of microRNAs (miRNAs) in how microbiota influence the host intestinal immune system is not fully understood. We compared the expression profiles of miRNAs and mRNAs in lamina propria leukocytes (LPL) in the large intestines of germ-free (GF) and specific pathogen-free (SPF) mice. Microarray analysis revealed different expression profiles of miRNAs and mRNAs between GF and SPF mice. Quantitative real time-PCR (qRT-PCR) showed that the level of miR-200 family members was significantly higher in SPF mice than in GF mice. In silico prediction followed by qRT-PCR suggested that Bcl11b, Ets1, Gbp7, Stat5b, and Zeb1 genes were downregulated by the miR-200 family. Western blotting revealed that the expression of BCL11B and ETS-1, but not ZEB1, in large intestinal LPL was significantly lower in SPF mice than in GF mice. Interleukin (IL)-2 production in cultured LPL upon stimulation with phorbol 12-myristate 13-acetate and ionomycin for 24 h was significantly lower in SPF mice than in GF mice. Conventionalization of GF mice substantially recapitulated SPF mice in terms of the expression of miR-200 family members and their target genes and IL-2 production in large intestinal LPL. Considering that BCL11B and ETS-1 reportedly function as transcription factors to activate the Il2 gene, we propose that the presence of gut commensals suppresses IL-2 production in large intestinal LPL, at least in part through post-transcriptional downregulation of Bcl11b and Ets1 genes by miR-200 family members.


Assuntos
Interleucina-2/metabolismo , Intestino Grosso/citologia , MicroRNAs/genética , Proteína Proto-Oncogênica c-ets-1/genética , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor/genética , Animais , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Vida Livre de Germes , Interleucina-2/genética , Leucócitos/fisiologia , Masculino , Camundongos Endogâmicos BALB C , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Supressoras de Tumor/metabolismo
12.
Biochem Biophys Res Commun ; 529(1): 64-69, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32560820

RESUMO

RegIIIß and RegIIIγ are antimicrobial peptides expressed in intestinal epithelial cells. Expression of these peptides is reportedly decreased by high-fat diet (HFD) and increased by indigestible oligosaccharides in mice. Clearly, these dietary regimens change the structure of intestinal microbiota. We employed an intestinal microbiota transplantation (IMT) to test whether diet-induced changes in the expression of these peptides are mediated by gut microbiota. C57BL/6J mice were fed either a normal-fat diet (NFD), a HFD, or a NFD supplemented with or without 1-kestose (KES), an indigestible oligosaccharide. Ileal RegIIIß and RegIIIγ mRNA levels were lower in mice receiving IMT from HFD-fed mice than in those receiving NFD-fed mice and higher in mice receiving IMT from KES-supplemented mice than in those receiving the mice without KES supplementation. Western blot analysis showed that serum RegIIIß levels changed in parallel with the ileal mRNA levels. We propose that HFD- and KES-induced changes in the ileal RegIIIß and RegIIIγ expression and in the circulating RegIIIß levels are mediated, at least in part, by intestinal microbiota.


Assuntos
Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Proteínas Associadas a Pancreatite/sangue , Proteínas Associadas a Pancreatite/genética , Animais , Dieta , Dieta Hiperlipídica , Íleo/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trissacarídeos/administração & dosagem , Interleucina 22
13.
Biosci Biotechnol Biochem ; 84(12): 2508-2520, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32752982

RESUMO

An enzyme belonging to glycoside hydrolase family 68 (GH68) from Beijerinckia indica subsp. indica NBRC 3744 was expressed in Escherichia coli. Biochemical characterization showed that the enzyme was identified to be a ß-fructosyltransferase (BiBftA). Crystallization of a full-length BiBftA was initially attempted, but no crystals were obtained. We constructed a variant in which 5 residues (Pro199-Gly203) and 13 residues (Leu522-Gln534) in potentially flexible regions were deleted, and we successfully crystallized this variant BiBftA. BiBftA is composed of a five-bladed ß-propeller fold as in other GH68 enzymes. The structure of BiBftA in complex with fructose unexpectedly indicated that one ß-fructofuranose (ß-Fruf) molecule and one ß-fructopyranose molecule bind to the catalytic pocket. The orientation of ß-Fruf at subsite -1 is tilted from the orientation observed in most GH68 enzymes, presenting a second structure of a GH68 enzyme in complex with the tilted binding mode of ß-Fruf.


Assuntos
Beijerinckiaceae/enzimologia , Frutose/metabolismo , Glicosídeo Hidrolases/química , Glicosídeo Hidrolases/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Glicosídeo Hidrolases/genética , Modelos Moleculares , Mutagênese , Conformação Proteica , Relação Estrutura-Atividade
14.
Anaerobe ; 61: 102076, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31326442

RESUMO

Prebiotics are widely used to shape a balanced microbiota in humans and animals. 1-Kestose (kestose) is one of the major components in commercialized short-chain fructooligosaccharide and is a promising prebiotic for infants. We herein studied the impact of kestose on the healthy adult microbiota in an in vitro fecal batch culture model. Stool samples obtained from seven healthy adults were diluted, inoculated into broth supplemented with or without 0.5% (w/v) kestose (kestose group and control group, respectively), and cultured under anaerobic conditions. Microbiota in the groups and stool samples were analyzed using 16S rRNA gene sequencing. At the phylum level, the kestose group showed increases in Bacteroidetes, whereas the control group showed increases in Proteobacteria. At the species level, Bifidobacterium longum was the only species showing significantly higher levels in the kestose group than in the control group and stool samples. On the other hand, levels of Escherichia coli were significantly higher in the control group than in stool samples, while the levels were not significantly different between the kestose group and stool samples. Quantitative PCR assays also revealed significantly higher levels of B. longum and lower tendency of E. coli in the kestose group than in the control group. These results suggest that supplementation with kestose increased the levels of beneficial microorganism and prevented the growth of risk-associated microorganisms related to disease development. Further interventional studies are needed to understand the health benefits of kestose in adult humans.


Assuntos
Suplementos Nutricionais , Fezes/microbiologia , Microbioma Gastrointestinal , Trissacarídeos/administração & dosagem , Adulto , Fatores Etários , Feminino , Fermentação , Voluntários Saudáveis , Humanos , Masculino , Metabolômica/métodos , Metagenoma , Metagenômica/métodos , Adulto Jovem
15.
Int J Mol Sci ; 21(3)2020 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-32019174

RESUMO

The gut microbiota remains relatively stable during adulthood; however, certain intrinsic and environmental factors can lead to microbiota dysbiosis. Its restoration towards a healthy condition using best-suited prebiotics requires previous development of in vitro models for evaluating their functionality. Herein, we carried out fecal cultures with microbiota from healthy normal-weight and morbid obese adults. Cultures were supplemented with different inulin-type fructans (1-kestose, Actilight, P95, Synergy1 and Inulin) and a galactooligosaccharide. Their impact on the gut microbiota was assessed by monitoring gas production and evaluating changes in the microbiota composition (qPCR and 16S rRNA gene profiling) and metabolic activity (gas chromatography). Additionally, the effect on the bifidobacterial species was assessed (ITS-sequencing). Moreover, the functionality of the microbiota before and after prebiotic-modulation was determined in an in vitro model of interaction with an intestinal cell line. In general, 1-kestose was the compound showing the largest effects. The modulation with prebiotics led to significant increases in the Bacteroides group and Faecalibacterium in obese subjects, whereas in normal-weight individuals, substantial rises in Bifidobacterium and Faecalibacterium were appreciated. Notably, the results obtained showed differences in the responses among the tested compounds but also among the studied human populations, indicating the need for developing population-specific products.


Assuntos
Bactérias/crescimento & desenvolvimento , Fezes/microbiologia , Microbioma Gastrointestinal/fisiologia , Obesidade Mórbida/tratamento farmacológico , Prebióticos/administração & dosagem , Magreza/tratamento farmacológico , Adulto , Bactérias/efeitos dos fármacos , Estudos de Casos e Controles , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Técnicas In Vitro , Masculino , Obesidade Mórbida/metabolismo , Obesidade Mórbida/patologia , Magreza/metabolismo , Magreza/patologia
16.
Biochem Biophys Res Commun ; 518(2): 294-298, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31420164

RESUMO

Kestose and nystose are short chain fructooligosaccharides (scFOSs) with degrees of polymerization of 3 and 4, respectively. A previous study revealed that these scFOSs have different growth stimulation properties against two human commensals, i.e. Bifidobacterium longum subsp. longum and butyrogenic Anaerostipes caccae. The present study characterized genes involved in FOS metabolism in these organisms. A. caccae possesses a single gene cluster consisting of four genes, including a gene encoding the putative FOS degradation enzyme sucrose-6-phosphate hydrolase (S6PH). B. longum possesses two gene clusters consisting of three genes each, including genes encoding ß-fructofuranosidase (CscA) and sucrose phosphorylase (ScrP). In A. caccae, the genes were highly transcribed in cells cultured with sucrose or kestose but poorly in cells cultured with glucose or nystose. Heterologously expressed S6PH degraded sucrose and kestose but not nystose. In B. longum, transcription of the genes was high in cells cultured with sucrose or kestose but was poor or not detected in cells cultured with glucose or nystose. Heterologously expressed CscA degraded sucrose, kestose and nystose, but ScrP degraded only sucrose. These data suggested that the different growth stimulation activities of kestose and nystose are due to different substrate specificities of FOS degradation enzymes in the organisms and/or induction activity of the genes in the two scFOSs. This is the first study characterizing the FOS metabolism at the transcriptional level and substrate-specificity of the degradation enzyme in butyrogenic human gut anaerobes.


Assuntos
Bifidobacterium longum/enzimologia , Clostridiales/enzimologia , Oligossacarídeos/metabolismo , Bifidobacterium longum/genética , Bifidobacterium longum/metabolismo , Clostridiales/genética , Clostridiales/metabolismo , Genes Bacterianos , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Humanos , Família Multigênica , Especificidade por Substrato , beta-Frutofuranosidase/genética , beta-Frutofuranosidase/metabolismo
17.
Biosci Biotechnol Biochem ; 82(9): 1599-1605, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29873621

RESUMO

1-Kestose is a key prebiotic fructooligosaccharide (FOS) sugar. Some ß-fructofuranosidases (FFases) have high transfructosylation activity, which is useful for manufacturing FOS. Therefore, obtaining FFases that produce 1-kestose efficiently is important. Here, we established a rapid FFase evaluation method using Escherichia coli that display different FFases fused to a PgsA anchor protein from Bacillus subtilis. E. coli cell suspensions expressing the PgsA-FFase fusion efficiently produce FOS from sucrose. Using this screening technique, we found that the E. coli transformant expressing Aspergillus kawachii FFase (AkFFase) produced a larger amount of 1-kestose than those expressing FFases from A. oryzae and A. terreus. Saturation mutagenesis of AkFFase was performed, and the mutant G85W was obtained. The E. coli transformant expressing AkFFase G85W markedly increased production of 1-kestose. Our results indicate that the surface display technique using PgsA is useful for screening of FFases, and AkFFase G85W is likely to be suitable for 1-kestose production. ABBREVIATIONS: AkFFase: Aspergillus kawachii FFase; AoFFase: Aspergillus oryzae FFase; AtFFase: Aspergillus terreus FFase; FFase: ß-fructofuranosidase; FOS: fructooligosaccharide; fructosylnystose: 1F-ß-fructofuranosylnystose.


Assuntos
Aspergillus/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Trissacarídeos/metabolismo , beta-Frutofuranosidase/biossíntese , Aspergillus/enzimologia , Escherichia coli/genética , Mutagênese , beta-Frutofuranosidase/genética , beta-Frutofuranosidase/metabolismo
18.
Anaerobe ; 51: 110-119, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29734011

RESUMO

Prebiotic oligosaccharides are known to have significant impacts on gut microbiota and are thus widely used to program healthy microbiota composition and activity from infants to the elderly. Bifidobacteria and lactobacilli are among the major target microorganisms of oligosaccharides, but the metabolic properties of oligosaccharides in other predominant gut microbes have not been well characterized. In the present study, we demonstrated the metabolic properties of six oligosaccharides in 31 key gut anaerobes. Bifidobacteria readily metabolized fructooligosaccharide (FOSs) with degree of polymerization (DP) 3, i.e. 1-kestose, but several strains used did not actively metabolize FOSs with DP4 and DP5, i.e. nystose and fructosylnystose. Akkermansia muciniphila, a potential new probiotic against obesity, did not show significant growth with any of the oligosaccharides tested. The butyrate producer Anaerostipes caccae grew well on 1-kestose but poorly on FOS mixtures, whereas it contained 1-kestose at 30%. Bacteroides-Parabacteroides group species were separated into two groups based on oligosaccharide metabolic properties. One group metabolized well most of the oligosaccharides tested, but the others metabolized only 1 or 2 selected oligosaccharides. Oligosaccharide profiles after culturing revealed that Bifidobacterium spp. preferentially metabolized shorter oligosaccharides (DP3) in the mixtures, whereas Bacteroides-Parabacteroides spp. did not show oligosaccharide selectivity for metabolism or rather preferred longer oligosaccharides (>DP4). The fermentation profiles indicated specific links between the microbial end-products and specific gut microbes. Available carbohydrates had a significant impact on the accumulation of amino acid-derived bacterial metabolites (i.e. phenol, p-cresol, indole and skatole) and short chain fatty acids. The results assist in predicting the impact of oligosaccharides in human intervention and gut microbiota modulation.


Assuntos
Bactérias Anaeróbias/crescimento & desenvolvimento , Bactérias Anaeróbias/metabolismo , Oligossacarídeos/metabolismo , Prebióticos , Bactérias Anaeróbias/isolamento & purificação , Fermentação , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Microbiota/efeitos dos fármacos
19.
Biosci Biotechnol Biochem ; 81(9): 1786-1795, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28715279

RESUMO

ß-Fructofuranosidases belonging to glycoside hydrolase family (GH) 32 are enzymes that hydrolyze sucrose. Some GH32 enzymes also catalyze transfructosylation to produce fructooligosaccharides. We found that Aspergillus kawachii IFO 4308 ß-fructofuranosidase (AkFFase) produces fructooligosaccharides, mainly 1-kestose, from sucrose. We determined the crystal structure of AkFFase. AkFFase is composed of an N-terminal small component, a ß-propeller catalytic domain, an α-helical linker, and a C-terminal ß-sandwich, similar to other GH32 enzymes. AkFFase forms a dimer, and the dimerization pattern is different from those of other oligomeric GH32 enzymes. The complex structure of AkFFase with fructose unexpectedly showed that fructose binds both subsites -1 and +1, despite the fact that the catalytic residues were not mutated. Fructose at subsite +1 interacts with Ile146 and Glu296 of AkFFase via direct hydrogen bonds.


Assuntos
Aspergillus/enzimologia , Frutose/metabolismo , beta-Frutofuranosidase/química , beta-Frutofuranosidase/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Glicosilação , Modelos Moleculares
20.
Pediatr Res ; 80(6): 844-851, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27537603

RESUMO

BACKGROUND: Although Faecalibacterium prausnitzii is a major bacterium in the intestine of adults, which is known to have anti-inflammatory effects, the development in infants or the response to prebiotics remains unclear. METHODS: The counts of F. prausnitzii in the feces were examined by real-time polymerase chain reaction (PCR). Fecal samples were obtained from 65 atopic dermatitis (AD) infants who participated in a randomized controlled clinical trial to investigate the therapeutic effect of kestose, the smallest fructooligosaccharide. RESULTS: Although the F. prausnitzii count was undetectable level in most 0- to 1-y-old infants, the count reached a level comparable to that in adults in 2- to 5-y-old infants. The bacterial number increased about 10-fold by oral administration of kestose every day for 12 wk in the younger infants, but not so much in the older infants. This bacterial increase was significantly correlated with an improvement in the AD symptoms in the older infants. CONCLUSION: The F. prausnitzii population in the intestine reaches a level comparable to that in adult at approximately 2 y of age. Kestose efficiently stimulates the growth of this bacterium in the intestine, which might lead to an improvement in AD symptoms in infants.


Assuntos
Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/microbiologia , Faecalibacterium prausnitzii/efeitos dos fármacos , Oligossacarídeos/uso terapêutico , Prebióticos/administração & dosagem , Fatores Etários , Carga Bacteriana , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Pré-Escolar , Faecalibacterium prausnitzii/genética , Faecalibacterium prausnitzii/isolamento & purificação , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Lactente , Recém-Nascido , Masculino , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA