Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 650
Filtrar
1.
Front Immunol ; 12: 772532, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970262

RESUMO

Emerging evidence supports that the efficacy of immune checkpoint blockade (ICB) therapy is associated with the host's gut microbiota, as prior antibiotic intake often leads to poor outcome and low responsiveness toward ICB treatment. Therefore, we hypothesized that the efficacy of ICB therapy like anti-programmed cell death protein-1 (PD-1) treatment required an intact host gut microbiota, and it was established that probiotics could enhance the recovery of gut microbiota disruption by external stimuli. Thus, the present study aimed to evaluate the effect of the probiotics, Lactobacillus rhamnosus Probio-M9, on recovering antibiotic-disrupted gut microbiota and its impact on the outcome of ICB therapy in tumor-bearing mice. We first disrupted the mouse microbiota by antibiotics and then remediated the gut microbiota by probiotics or naturally. Tumor transplantation was then performed, followed by anti-PD-1-based antitumor therapy. Changes in the fecal metagenomes and the tumor suppression effect were monitored during different stages of the experiment. Our results showed that Probio-M9 synergized with ICB therapy, significantly improving tumor inhibition compared with groups not receiving the probiotic treatment (P < 0.05 at most time points). The synergistic effect was accompanied by effective restoration of antibiotic-disrupted fecal microbiome that was characterized by a drastically reduced Shannon diversity value and shifted composition of dominating taxa. Moreover, probiotic administration significantly increased the relative abundance of beneficial bacteria (e.g., Bifidobacterium pseudolongum, Parabacteroides distasonis, and some Bacteroides species; 0.0001 < P < 0.05). The gut microbiome changes were accompanied by mild reshaping of the functional metagenomes characterized by enrichment in sugar degradation and vitamin and amino acid synthesis pathways. Collectively, this study supported that probiotic administration could enhance the efficacy and responsiveness of anti-PD-1-based immunotherapy, and Probio-M9 could be a potential candidate of microbe-based synergistic tumor therapeutics. The preclinical data obtained here would support the design of future human clinical trials for further consolidating the current findings and for safety assessment of probiotic adjunctive treatment in ICB therapy.


Assuntos
Antibacterianos/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/administração & dosagem , Lacticaseibacillus rhamnosus , Neoplasias/terapia , Probióticos/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Bacteroides/efeitos dos fármacos , Bacteroides/crescimento & desenvolvimento , Bifidobacterium/efeitos dos fármacos , Bifidobacterium/crescimento & desenvolvimento , Linhagem Celular Tumoral , Fezes/microbiologia , Camundongos Endogâmicos BALB C , Neoplasias/microbiologia
2.
Biomolecules ; 11(12)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34944439

RESUMO

Diverse members of the Bacteroidetes phylum have general protein O-glycosylation systems that are essential for processes such as host colonization and pathogenesis. Here, we analyzed the function of a putative fucosyltransferase (FucT) family that is widely encoded in Bacteroidetes protein O-glycosylation genetic loci. We studied the FucT orthologs of three Bacteroidetes species-Tannerella forsythia, Bacteroides fragilis, and Pedobacter heparinus. To identify the linkage created by the FucT of B. fragilis, we elucidated the full structure of its nine-sugar O-glycan and found that l-fucose is linked ß1,4 to glucose. Of the two fucose residues in the T. forsythia O-glycan, the fucose linked to the reducing-end galactose was shown by mutational analysis to be l-fucose. Despite the transfer of l-fucose to distinct hexose sugars in the B. fragilis and T. forsythia O-glycans, the FucT orthologs from B. fragilis, T. forsythia, and P. heparinus each cross-complement the B. fragilis ΔBF4306 and T. forsythia ΔTanf_01305 FucT mutants. In vitro enzymatic analyses showed relaxed acceptor specificity of the three enzymes, transferring l-fucose to various pNP-α-hexoses. Further, glycan structural analysis together with fucosidase assays indicated that the T. forsythia FucT links l-fucose α1,6 to galactose. Given the biological importance of fucosylated carbohydrates, these FucTs are promising candidates for synthetic glycobiology.


Assuntos
Bacteroides/crescimento & desenvolvimento , Fucosiltransferases/química , Fucosiltransferases/genética , Polissacarídeos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteroides/enzimologia , Bacteroides fragilis/enzimologia , Bacteroides fragilis/crescimento & desenvolvimento , Configuração de Carboidratos , Evolução Molecular , Fucosiltransferases/metabolismo , Regulação Bacteriana da Expressão Gênica , Glicosilação , Modelos Moleculares , Pedobacter/enzimologia , Pedobacter/crescimento & desenvolvimento , Polissacarídeos/metabolismo , Tannerella forsythia/enzimologia , Tannerella forsythia/crescimento & desenvolvimento
3.
Int J Biol Macromol ; 187: 664-674, 2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34339781

RESUMO

Symbiotic bacteria, including members of the Bacteroides genus, are known to digest dietary fibers in the gastrointestinal tract. The metabolism of complex carbohydrates is restricted to a specified subset of species and is likely orchestrated by polysaccharide utilization loci (PULs) in these microorganisms. ß-Mannans are plant cell wall polysaccharides that are commonly found in human nutrients. Here, we report the structural basis of a PUL cluster, BdPUL12, which controls ß-mannan-like glycan catabolism in Bacteroides dorei. Detailed biochemical characterization and targeted gene disruption studies demonstrated that a key glycoside hydrolase, BdP12GH26, performs the initial attack on galactomannan or glucomannan likely via an endo-acting mode, generating mannooligosaccharides and mannose. Importantly, coculture assays showed that the B. dorei promoted the proliferation of Lactobacillus helveticus and Bifidobacterium adolescentis, likely by sharing mannooligosaccharides and mannose with these gut probiotics. Our findings provide new insights into carbohydrate metabolism in gut-inhabiting bacteria and lay a foundation for novel probiotic development.


Assuntos
Proteínas de Bactérias/metabolismo , Bacteroides/enzimologia , Galactose/análogos & derivados , Mananas/metabolismo , Manose/metabolismo , Manosidases/metabolismo , Oligossacarídeos/metabolismo , Probióticos , Proteínas de Bactérias/genética , Bacteroides/genética , Bacteroides/crescimento & desenvolvimento , Bifidobacterium adolescentis/crescimento & desenvolvimento , Bifidobacterium adolescentis/metabolismo , Galactose/metabolismo , Microbioma Gastrointestinal , Hidrólise , Lactobacillus helveticus/crescimento & desenvolvimento , Lactobacillus helveticus/metabolismo , Manosidases/genética , Simbiose
4.
mBio ; 12(4): e0136821, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34340552

RESUMO

The human gut microbiota (HGM) contributes to the physiology and health of its host. The health benefits provided by dietary manipulation of the HGM require knowledge of how glycans, the major nutrients available to this ecosystem, are metabolized. Arabinogalactan proteins (AGPs) are a ubiquitous feature of plant polysaccharides available to the HGM. Although the galactan backbone and galactooligosaccharide side chains of AGPs are conserved, the decorations of these structures are highly variable. Here, we tested the hypothesis that these variations in arabinogalactan decoration provide a selection mechanism for specific Bacteroides species within the HGM. The data showed that only a single bacterium, B. plebeius, grew on red wine AGP (Wi-AGP) and seaweed AGP (SW-AGP) in mono- or mixed culture. Wi-AGP thus acts as a privileged nutrient for a Bacteroides species within the HGM that utilizes marine and terrestrial plant glycans. The B. plebeius polysaccharide utilization loci (PULs) upregulated by AGPs encoded a polysaccharide lyase, located in the enzyme family GH145, which hydrolyzed Rha-Glc linkages in Wi-AGP. Further analysis of GH145 identified an enzyme with two active sites that displayed glycoside hydrolase and lyase activities, respectively, which conferred substrate flexibility for different AGPs. The AGP-degrading apparatus of B. plebeius also contained a sulfatase, BpS1_8, active on SW-AGP and Wi-AGP, which played a pivotal role in the utilization of these glycans by the bacterium. BpS1_8 enabled other Bacteroides species to access the sulfated AGPs, providing a route to introducing privileged nutrient utilization into probiotic and commensal organisms that could improve human health. IMPORTANCE Dietary manipulation of the HGM requires knowledge of how glycans available to this ecosystem are metabolized. The variable structures that decorate the core component of plant AGPs may influence their utilization by specific organisms within the HGM. Here, we evaluated the ability of Bacteroides species to utilize a marine and terrestrial AGP. The data showed that a single bacterium, B. plebeius, grew on Wi-AGP and SW-AGP in mono- or mixed culture. Wi-AGP is thus a privileged nutrient for a Bacteroides species that utilizes marine and terrestrial plant glycans. A key component of the AGP-degrading apparatus of B. plebeius is a sulfatase that conferred the ability of the bacterium to utilize these glycans. The enzyme enabled other Bacteroides species to access the sulfated AGPs, providing a route to introducing privileged nutrient utilization into probiotic and commensal organisms that could improve human health.


Assuntos
Bacteroides/metabolismo , Mucoproteínas/metabolismo , Nutrientes/metabolismo , Sulfatases/metabolismo , Bacteroides/enzimologia , Bacteroides/genética , Bacteroides/crescimento & desenvolvimento , Carboidratos da Dieta/metabolismo , Microbioma Gastrointestinal/fisiologia , Proteínas de Plantas/metabolismo , Polissacarídeo-Liases/genética , Polissacarídeo-Liases/metabolismo , Polissacarídeos/metabolismo
5.
Nat Rev Microbiol ; 19(12): 774-785, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34183820

RESUMO

The defining trait of obligate anaerobes is that oxygen blocks their growth, yet the underlying mechanisms are unclear. A popular hypothesis was that these microorganisms failed to evolve defences to protect themselves from reactive oxygen species (ROS) such as superoxide and hydrogen peroxide, and that this failure is what prevents their expansion to oxic habitats. However, studies reveal that anaerobes actually wield most of the same defences that aerobes possess, and many of them have the capacity to tolerate substantial levels of oxygen. Therefore, to understand the structures and real-world dynamics of microbial communities, investigators have examined how anaerobes such as Bacteroides, Desulfovibrio, Pyrococcus and Clostridium spp. struggle and cope with oxygen. The hypoxic environments in which these organisms dwell - including the mammalian gut, sulfur vents and deep sediments - experience episodic oxygenation. In this Review, we explore the molecular mechanisms by which oxygen impairs anaerobes and the degree to which bacteria protect their metabolic pathways from it. The emergent view of anaerobiosis is that optimal strategies of anaerobic metabolism depend upon radical chemistry and low-potential metal centres. Such catalytic sites are intrinsically vulnerable to direct poisoning by molecular oxygen and ROS. Observations suggest that anaerobes have evolved tactics that either minimize the extent to which oxygen disrupts their metabolism or restore function shortly after the stress has dissipated.


Assuntos
Bactérias Anaeróbias/metabolismo , Oxigênio/toxicidade , Espécies Reativas de Oxigênio/toxicidade , Anaerobiose , Bactérias Anaeróbias/crescimento & desenvolvimento , Bacteroides/crescimento & desenvolvimento , Bacteroides/metabolismo , Clostridium/crescimento & desenvolvimento , Clostridium/metabolismo , Desulfovibrio/crescimento & desenvolvimento , Desulfovibrio/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/toxicidade , Oxigênio/metabolismo , Pyrococcus/crescimento & desenvolvimento , Pyrococcus/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Superóxidos/toxicidade
6.
Gut Microbes ; 13(1): 1-17, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34132157

RESUMO

Dysbiosis of gut microbiota has been retrospectively linked to autism spectrum disorders but the temporal association between gut microbiota and early neurodevelopment in healthy infants is largely unknown. We undertook this study to determine associations between gut microbiota at two critical periods during infancy and neurodevelopment in a general population birth cohort.Here, we analyzed data from 405 infants (199 females) from the CHILD (Canadian Healthy Infant Longitudinal Development) Cohort Study. Neurodevelopmental outcomes were objectively assessed using the Bayley Scale of Infant Development (BSID-III) at 1 and 2 years of age. Microbiota profiling with 16S rRNA gene sequencing was conducted on fecal samples obtained at a mean age of 4 and 12 months.Using clustering methods, we identified three groups of infants based on relative abundance of gut microbiota at 12 months: Proteobacteria-dominant cluster (22.4% higher abundance at 12 months), Firmicutes-dominant cluster (46.0% higher abundance at 12 months) and Bacteroidetes-dominant cluster (31.6% higher abundance at 12 months). Relative to the Proteobacteria-dominant cluster, the Bacteroidetes-dominant cluster was associated with higher scores for cognitive (4.8 points; FDRp = .02), language (4.2 points; FDRp≤0.001), and motor (3.1 points; FDRp = .03) development at age 2 in models adjusted for covariates. When stratified by sex, only male infants with a Bacteroidetes-dominant microbiota had more favorable cognitive (5.9 points, FDRp = .06) and language (7.9 points; FDRp≤0.001) development. Genus Bacteroides abundance in gut microbiota was positively correlated with cognitive and language scores at age 2. Fully adjusted linear mixed model analysis revealed a positive association between Bacteroidetes-dominant cluster and change in cognitive and language performance from 1 to 2 years, predominantly among males. No associations were evident between 4-month microbiota clusters and BSID-II scores. Noteworthy is that enhanced sphingolipid synthesis and metabolism, and antagonism or competition between Bacteroides and Streptococcus were characteristic of a Bacteroidetes-dominant gut microbiota.This study found strong evidence of positive associations between Bacteroidetes gut microbiota in late infancy and subsequent neurodevelopment, most prominently among males but not females.


Assuntos
Bacteroides/crescimento & desenvolvimento , Desenvolvimento Infantil , Microbioma Gastrointestinal , Sistema Nervoso/crescimento & desenvolvimento , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Bacteroides/classificação , Bacteroides/genética , Bacteroides/isolamento & purificação , Canadá , Feminino , Seguimentos , Humanos , Lactente , Masculino , RNA Ribossômico 16S/genética , Estudos Retrospectivos
7.
Gastroenterology ; 161(3): 940-952.e15, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34111469

RESUMO

BACKGROUND & AIMS: Perturbations in the early-life gut microbiome are associated with increased risk for complex immune disorders like inflammatory bowel diseases. We previously showed that maternal antibiotic-induced gut dysbiosis vertically transmitted to offspring increases experimental colitis risk in interleukin (IL) 10 gene deficient (IL10-/-) mice, a finding that may result from the loss/lack of essential microbes needed for appropriate immunologic education early in life. Here, we aimed to identify key microbes required for proper development of the early-life gut microbiome that decrease colitis risk in genetically susceptible animals. METHODS: Metagenomic sequencing followed by reconstruction of metagenome-assembled genomes was performed on fecal samples of IL10-/- mice with and without antibiotic-induced dysbiosis to identify potential missing microbial members needed for immunologic education. One high-value target strain was then engrafted early and/or late into the gut microbiomes of IL10-/- mice with antibiotic-induced dysbiosis. RESULTS: Early-, but not late-, life engraftment of a single dominant Bacteroides strain of non-antibiotic-treated IL10-/- mice was sufficient to restore the development of the gut microbiome, promote immune tolerance, and prevent colitis in IL10-/- mice that had antibiotic-induced dysbiosis. CONCLUSIONS: Restitution of a keystone microbial strain missing in the early-life antibiotic-induced gut dysbiosis results in recovery of the microbiome, proper development of immune tolerance, and reduced risk for colitis in genetically prone hosts.


Assuntos
Bacteroides/crescimento & desenvolvimento , Colite/prevenção & controle , Colo/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Interleucina-10/deficiência , Animais , Antibacterianos , Bacteroides/imunologia , Colite/imunologia , Colite/metabolismo , Colite/microbiologia , Colo/imunologia , Colo/metabolismo , Colo/patologia , Modelos Animais de Doenças , Disbiose , Fezes/microbiologia , Interações Hospedeiro-Patógeno , Tolerância Imunológica , Interleucina-10/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estudo de Prova de Conceito , Fatores de Tempo
8.
Int J Biol Macromol ; 181: 357-368, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-33774071

RESUMO

Inflammatory bowel disease (IBD) afflicted individual and most medications have side-effects. Crataegus pinnatifida (Hawthorn), which is a safe medicine and food homolog plant, has been reported to prevent colitis in murine. Yet the bioactivity component and the underlying molecular mechanism remain unclear. Here, we established a direct link between colitis induced by dextran sulphate sodium (DSS) in mice and polysaccharide HAW1-2 isolated from hawthorn. Our results showed HAW1-2 restored the pathological lesions in colon and inhibited the expression of inflammatory cytokines including IL-1ß, IL-6 and TNF-α. Meanwhile, IKKα/ß, IκBα, NF-κB and the phosphorylation levels were inhibited significantly. These findings suggested HAW1-2 could alleviate the inflammation of colon. Further, we found the composition of gut microbiota was modified and Bacteroides including Alistipes and Odoribacter were significantly enriched. Besides, we showed Alistipes and Odoribacter were positively co-related with acetic acid and propionic acid while were negatively co-related with inflammatory cytokines. Finally, we demonstrated the anti-inflammation activity of HAW1-2 might be induced by acetic acid. Together, the present data revealed HAW1-2 could directly modify the gut microbiota, especially for Bacteroides, and generate SCFAs to inhibit colitis. It also implies microbiota-directed intervention in IBD patients should be particularly given more attention.


Assuntos
Colite/tratamento farmacológico , Colite/microbiologia , Crataegus/química , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal , Polissacarídeos/uso terapêutico , Ácido Acético/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Bacteroides/efeitos dos fármacos , Bacteroides/crescimento & desenvolvimento , Linhagem Celular , Colite/induzido quimicamente , Colite/patologia , Colo/efeitos dos fármacos , Colo/microbiologia , Colo/patologia , Sulfato de Dextrana , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação/patologia , Masculino , Metaboloma , Camundongos Endogâmicos C57BL , Modelos Biológicos , NF-kappa B/metabolismo , Polissacarídeos/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos
9.
J Biol Chem ; 296: 100552, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33744293

RESUMO

The Cellulosome is an intricate macromolecular protein complex that centralizes the cellulolytic efforts of many anaerobic microorganisms through the promotion of enzyme synergy and protein stability. The assembly of numerous carbohydrate processing enzymes into a macromolecular multiprotein structure results from the interaction of enzyme-borne dockerin modules with repeated cohesin modules present in noncatalytic scaffold proteins, termed scaffoldins. Cohesin-dockerin (Coh-Doc) modules are typically classified into different types, depending on structural conformation and cellulosome role. Thus, type I Coh-Doc complexes are usually responsible for enzyme integration into the cellulosome, while type II Coh-Doc complexes tether the cellulosome to the bacterial wall. In contrast to other known cellulosomes, cohesin types from Bacteroides cellulosolvens, a cellulosome-producing bacterium capable of utilizing cellulose and cellobiose as carbon sources, are reversed for all scaffoldins, i.e., the type II cohesins are located on the enzyme-integrating primary scaffoldin, whereas the type I cohesins are located on the anchoring scaffoldins. It has been previously shown that type I B. cellulosolvens interactions possess a dual-binding mode that adds flexibility to scaffoldin assembly. Herein, we report the structural mechanism of enzyme recruitment into B. cellulosolvens cellulosome and the identification of the molecular determinants of its type II cohesin-dockerin interactions. The results indicate that, unlike other type II complexes, these possess a dual-binding mode of interaction, akin to type I complexes. Therefore, the plasticity of dual-binding mode interactions seems to play a pivotal role in the assembly of B. cellulosolvens cellulosome, which is consistent with its unmatched complexity and size.


Assuntos
Proteínas de Bactérias/metabolismo , Bacteroides/metabolismo , Proteínas de Ciclo Celular/metabolismo , Celulossomas/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Clostridiales/metabolismo , Proteínas de Bactérias/genética , Bacteroides/genética , Bacteroides/crescimento & desenvolvimento , Proteínas de Ciclo Celular/genética , Celobiose/metabolismo , Celulose/metabolismo , Proteínas Cromossômicas não Histona/genética , Clostridiales/genética , Clostridiales/crescimento & desenvolvimento
10.
Biomed Pharmacother ; 133: 111014, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33246225

RESUMO

BACKGROUND: Intestinal microbiota is a novel drug target of metabolic diseases, especially for those with poor oral bioavailability. Nuciferine, with poor bioavailability, has an anti-hyperlipidemic effect at low dosages. PURPOSE: In the present study, we aimed to explore the role of intestinal microbiota in the anti-hyperlipidemic function of nuciferine and identify the key bacterial targets that might confer the therapeutic actions. METHODS: The contribution of gut microbes in the anti-hyperlipidemic effect of nuciferine was evaluated by conventional and antibiotic-established pseudo-sterile mice. Whole-metagenome shotgun sequencing was used to characterize the changes in microbial communities by various agents. RESULTS: Nuciferine exhibited potent anti-hyperlipidemic and liver steatosis-alleviating effects at the doses of 7.5-30 mg/kg. The beneficial effects of nuciferine were substantially abolished when combined with antibiotics. Metagenomic analysis showed that nuciferine significantly shifted the microbial structure, and the enrichment of Akkermansia muciniphila was closely related to the therapeutic effect of nuciferine. CONCLUSIONS: Our results revealed that gut microbiota played an essential role in the anti-hyperlipidemic effect of nuciferine, and enrichment of Akkermansia muciniphila represented a key mechanism through which nuciferine exerted its therapeutic effects.


Assuntos
Aporfinas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hiperlipidemias/tratamento farmacológico , Hipolipemiantes/farmacologia , Intestinos/microbiologia , Lipídeos/sangue , Akkermansia/efeitos dos fármacos , Akkermansia/genética , Akkermansia/crescimento & desenvolvimento , Animais , Antibacterianos/farmacologia , Bacteroides/efeitos dos fármacos , Bacteroides/genética , Bacteroides/crescimento & desenvolvimento , Biomarcadores/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Hiperlipidemias/sangue , Hiperlipidemias/microbiologia , Masculino , Metagenoma , Metagenômica , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/microbiologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/sangue , Obesidade/microbiologia , Obesidade/prevenção & controle , RNA-Seq
11.
Gut Microbes ; 12(1): 1847627, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33319645

RESUMO

Recent studies indicate that microbial enterotypes may influence the beneficial effects of wholegrain enriched diets including bodyweight regulation. In a 4-week intervention trial, overweight subjects were randomized to consume either arabinoxylan-oligosaccharides (AXOS) (10.4 g/d) from wheat bran or polyunsaturated fatty acids (PUFA) (3.6 g/d). In the present study, we have stratified the subjects participating in the intervention (n = 29) according to the baseline Prevotella-to-Bacteroides (P/B) ratios through a post-hoc analysis and applied a linear mixed model analysis to identify the influence of this P/B ratio on the differences in weight changes in the intervention arms. Following AXOS consumption (n = 15), the high P/B group showed no bodyweight changes [-0.14 kg (95% CI: -0.67; 0.38, p = .59)], while the low P/B group gained 0.65 kg (95% CI: 0.16; 1.14, p = .009). Consequently, a difference of -0.79 kg was found between P/B groups (95% CI: -1.51; -0.08, p = .030). No differences were found between P/B groups following PUFA consumption (0.61 kg, 95% CI: -0.13; 1.35, p = .10). Among the Bacteroides species, B. cellulosilyticus relative abundance exhibited the highest positive rank correlation (Kendall's tau = 0.51, FDR p = .070) with 4-week weight change on AXOS, and such association was further supported by using supervised classification methods (Random Forest). We outlined several carbohydrate-active enzyme (CAZy) genes involved in xylan-binding and degradation to be enriched in B. cellulosilyticus genomes, as well as multiple accessory genes, suggesting a supreme AXOS-derived glycan scavenging role of such species. This post-hoc analysis, ensuring species and strain demarcation at the human gut microbiota, permitted to uncover the predictive role of Bacteroides species over P/B enterotype in weight gain during a fiber-based intervention. The results of this pilot trial pave the way for future assessments on fiber fermentation outputs from Bacteroides species affecting lipid metabolism in the host and with direct impact on adiposity, thus helping to design personalized interventions.


Assuntos
Bacteroides/crescimento & desenvolvimento , Microbioma Gastrointestinal , Oligossacarídeos/metabolismo , Sobrepeso/dietoterapia , Sobrepeso/microbiologia , Xilanos/metabolismo , Adolescente , Adulto , Bacteroides/classificação , Bacteroides/genética , Bacteroides/isolamento & purificação , Biomarcadores/metabolismo , Peso Corporal , Fezes/microbiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Sobrepeso/fisiopatologia , Adulto Jovem
12.
Gut Microbes ; 12(1): 1-18, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33043794

RESUMO

ß-glucans are the dietary nutrients present in oats, barley, algae, and mushrooms. The macromolecules are well known for their immune-modulatory activity; however, how the human gut bacteria digest them is vaguely understood. In this study, Bacteroides uniformis JCM 13288 T was found to grow on laminarin, pustulan, and porphyran. We sequenced the genome of the strain, which was about 5.05 megabase pairs and contained 4868 protein-coding genes. On the basis of growth patterns of the bacterium, two putative polysaccharide utilization loci for ß-glucans were identified from the genome, and associated four putative genes were cloned, expressed, purified, and characterized. Three glycoside hydrolases (GHs) that were endo-acting enzymes (BuGH16, BuGH30, and BuGH158), and one which was an exo-acting (BuGH3) enzyme. The BuGH3, BuGH16, and BuGH158 can cleave linear exo/endo- ß- 1-3 linkages while BuGH30 can digest endo- ß- 1-6 linkages. BuGH30 and BuGH158 were further explored for their roles in digesting ß- glucans and generation of oligosaccharides, respectively. The BuGH30 predominately found to cleave long chain ß- 1-6 linked glucans, and obtained final product was gentiobiose. The BuGH158 used for producing oligosaccharides varying from degree of polymerization 2 to 7 from soluble curdlan. We demonstrated that these oligosaccharides can be utilized by gut bacteria, which either did not grow or poorly grew on laminarin. Thus, B. uniformis JCM 13288 T is not only capable of utilizing ß-glucans but also shares these glycans with human gut bacteria for potentially maintaining the gut microbial homeostasis.


Assuntos
Bacteroides/metabolismo , Microbioma Gastrointestinal , Glicosídeo Hidrolases/metabolismo , Bactérias Gram-Positivas/metabolismo , beta-Glucanas/metabolismo , Bacteroides/enzimologia , Bacteroides/crescimento & desenvolvimento , Configuração de Carboidratos , Loci Gênicos , Genoma Bacteriano , Glicosídeo Hidrolases/genética , Bactérias Gram-Positivas/genética , Bactérias Gram-Positivas/crescimento & desenvolvimento , Interações Microbianas , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , beta-Glucanas/química
13.
Sci Rep ; 10(1): 17351, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060635

RESUMO

Yeast mannan (YM) is an indigestible water-soluble polysaccharide of the yeast cell wall, with a notable prebiotic effect on the intestinal microbiota. We previously reported that YM increased Bacteroides thetaiotaomicron abundance in in vitro rat faeces fermentation, concluding that its effects on human colonic microbiota should be investigated. In this study, we show the effects of YM on human colonic microbiota and its metabolites using an in vitro human faeces fermentation system. Bacterial 16S rRNA gene sequence analysis showed that YM administration did not change the microbial diversity or composition. Quantitative real-time PCR analysis revealed that YM administration significantly increased the relative abundance of Bacteroides ovatus and B. thetaiotaomicron. Moreover, a positive correlation was observed between the relative ratio (with or without YM administration) of B. thetaiotaomicron and B. ovatus (r = 0.92), suggesting that these bacteria utilise YM in a coordinated manner. In addition, YM administration increased the production of acetate, propionate, and total short-chain fatty acids. These results demonstrate the potential of YM as a novel prebiotic that selectively increases B. thetaiotaomicron and B. ovatus and improves the intestinal environment. The findings also provide insights that might be useful for the development of novel functional foods.


Assuntos
Bacteroides/crescimento & desenvolvimento , Colo/microbiologia , Microbioma Gastrointestinal , Mananas/farmacologia , Prebióticos , Leveduras/metabolismo , Bacteroides/classificação , Alimento Funcional , Humanos , Especificidade da Espécie
14.
Proc Natl Acad Sci U S A ; 117(39): 24484-24493, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32938803

RESUMO

Mechanistic studies of anaerobic gut bacteria have been hindered by the lack of a fluorescent protein system to track and visualize proteins and dynamic cellular processes in actively growing bacteria. Although underappreciated, many gut "anaerobes" are able to respire using oxygen as the terminal electron acceptor. The oxygen continually released from gut epithelial cells creates an oxygen gradient from the mucus layer to the anaerobic lumen [L. Albenberg et al., Gastroenterology 147, 1055-1063.e8 (2014)], with oxygen available to bacteria growing at the mucus layer. Here, we show that Bacteroides species are metabolically and energetically robust and do not mount stress responses in the presence of 0.10 to 0.14% oxygen, defined as nanaerobic conditions [A. D. Baughn, M. H. Malamy, Nature 427, 441-444 (2004)]. Taking advantage of this metabolic capability, we show that nanaerobic growth provides sufficient oxygen for the maturation of oxygen-requiring fluorescent proteins in Bacteroides species. Type strains of four different Bacteroides species show bright GFP fluorescence when grown nanaerobically versus anaerobically. We compared four different red fluorescent proteins and found that mKate2 yields the highest red fluorescence intensity in our assay. We show that GFP-tagged proteins can be localized in nanaerobically growing bacteria. In addition, we used time-lapse fluorescence microscopy to image dynamic type VI secretion system processes in metabolically active Bacteroides fragilis The ability to visualize fluorescently labeled Bacteroides and fluorescently linked proteins in actively growing nanaerobic gut symbionts ushers in an age of imaging analyses not previously possible in these bacteria.


Assuntos
Bacteroides/metabolismo , Microbioma Gastrointestinal , Aerobiose , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteroides/classificação , Bacteroides/genética , Bacteroides/crescimento & desenvolvimento , Humanos , Oxigênio/metabolismo , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/metabolismo
15.
Medicine (Baltimore) ; 99(37): e21788, 2020 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-32925716

RESUMO

Hepatocellular carcinoma (HCC) is one of the most common neoplasms encountered, and its incidence is increasing worldwide. In this study, we explored the characteristics of gut microbiota in patients with primary hepatocellular carcinoma in advanced stage who received immune checkpoint inhibitors (ICIs) based on a large population with hepatitis B virus infection. An initial cohort of 65 patients with metastatic melanoma were included in this study. All patients were treated with ICIs at Fujian provincial geriatric hospital between August 2016 and June 2018. The 16S rDNA V4 region was amplified by Polymerase chain reaction and sequenced on the MiSeq platform. We found that the diversities of the gut microbiota in HCC who received ICIs were obviously increased. Negative feedback, which is controlled by interplay between microbial metabolic activities and host pathways, is thought to promote high bacterial diversity. We focused on the Faecalibacterium genus in response group, and Bacteroidales order in non-response group, and stratified patients into high versus low categories based on the median relative abundance of these taxa in the gut microbiome. Patients with high Faecalibacterium abundance had a significantly prolonged PFS versus those with a low abundance. Conversely, patients with a high abundance of Bacteroidales had a shortened progressive free survival compared to those with a low abundance. In summary, the present study examined the oral and gut microbiome of HCC patients undergoing immune checkpoint inhibitors immunotherapy. Significant differences were observed in the diversity and composition of the patient gut microbiome of responders versus non-responders.


Assuntos
Carcinoma Hepatocelular/microbiologia , Microbioma Gastrointestinal , Fatores Imunológicos/uso terapêutico , Imunoterapia/mortalidade , Neoplasias Hepáticas/microbiologia , Idoso , Bacteroides/crescimento & desenvolvimento , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/mortalidade , China , Faecalibacterium/crescimento & desenvolvimento , Feminino , Humanos , Imunoterapia/métodos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade , Intervalo Livre de Progressão , Estudos Prospectivos , RNA Ribossômico 16S/análise , Resultado do Tratamento
16.
Anaerobe ; 66: 102276, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32927049

RESUMO

B. ovatus is a member of the human gut microbiota with a broad capability to degrade complex glycans. Here we show that B. ovatus degrades plant polysaccharides in a preferential order, and that glycan structural complexity plays a role in determining the prioritisation of polysaccharide usage.


Assuntos
Bacteroides/crescimento & desenvolvimento , Bacteroides/metabolismo , Trato Gastrointestinal/microbiologia , Polissacarídeos/metabolismo , Microbioma Gastrointestinal , Humanos , Plantas/química , Polissacarídeos/química
17.
FASEB J ; 34(8): 10699-10719, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32584506

RESUMO

Heart failure (HF) is an increasing global health crisis, affecting 40 million people and causing 50% mortality within 5 years of diagnosis. A fuller understanding of the genetic and environmental factors underlying HF, and novel therapeutic approaches to address it, are urgently warranted. Here, we discovered that cardiac-specific germline deletion in mice of potassium channel ß subunit-encoding Kcne2 (Kcne2CS-/- ) causes dilated cardiomyopathy and terminal HF (median longevity, 28 weeks). Mice with global Kcne2 deletion (Kcne2Glo-/- ) exhibit multiple HF risk factors, yet, paradoxically survived over twice as long as Kcne2CS-/- mice. Global Kcne2 deletion, which inhibits gastric acid secretion, reduced the relative abundance of species within Bacteroidales, a bacterial order that positively correlates with increased lifetime risk of human cardiovascular disease. Strikingly, the proton-pump inhibitor omeprazole similarly altered the microbiome and delayed terminal HF in Kcne2CS-/- mice, increasing survival 10-fold at 44 weeks. Thus, genetic or pharmacologic induction of hypochlorhydria and decreased gut Bacteroidales species are associated with lifespan extension in a novel HF model.


Assuntos
Acloridria/genética , Acloridria/mortalidade , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/mortalidade , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Bacteroides/crescimento & desenvolvimento , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/mortalidade , Feminino , Ácido Gástrico/metabolismo , Microbioma Gastrointestinal/genética , Deleção de Genes , Coração/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Risco
18.
Gut Microbes ; 11(5): 1324-1347, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32404017

RESUMO

Multiple studies have identified changes within the gut microbiome in response to diarrheal-inducing bacterial pathogens. However, examination of the microbiome in response to viral pathogens remains understudied. Compounding this, many studies use fecal samples to assess microbiome composition; which may not accurately mirror changes within the small intestine, the primary site for most enteric virus infections. As a result, the functional significance of small intestinal microbiome shifts during infection is not well defined. To address these gaps, rotavirus-infected neonatal mice were examined for changes in bacterial community dynamics, host gene expression, and tissue recovery during infection. Profiling bacterial communities using 16S rRNA sequencing suggested significant and distinct changes in ileal communities in response to rotavirus infection, with no significant changes for other gastrointestinal (GI) compartments. At 1-d post-infection, we observed a loss in Lactobacillus species from the ileum, but an increase in Bacteroides and Akkermansia, both of which exhibit mucin-digesting capabilities. Concomitant with the bacterial community shifts, we observed a loss of mucin-filled goblet cells in the small intestine at d 1, with recovery occurring by d 3. Rotavirus infection of mucin-producing cell lines and human intestinal enteroids (HIEs) stimulated release of stored mucin granules, similar to in vivo findings. In vitro, incubation of mucins with Bacteroides or Akkermansia members resulted in significant glycan degradation, which altered the binding capacity of rotavirus in silico and in vitro. Taken together, these data suggest that the response to and recovery from rotavirus-diarrhea is unique between sub-compartments of the GI tract and may be influenced by mucin-degrading microbes.


Assuntos
Microbioma Gastrointestinal , Íleo/microbiologia , Polissacarídeos/metabolismo , Infecções por Rotavirus/patologia , Infecções por Rotavirus/virologia , Rotavirus/patogenicidade , Akkermansia/crescimento & desenvolvimento , Akkermansia/metabolismo , Animais , Animais Recém-Nascidos , Bactérias/classificação , Bactérias/crescimento & desenvolvimento , Bacteroides/crescimento & desenvolvimento , Bacteroides/metabolismo , Células Caliciformes/fisiologia , Íleo/patologia , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Lactobacillus/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos BALB C , Mucinas/metabolismo , RNA Ribossômico 16S/genética , Infecções por Rotavirus/microbiologia , Virulência
19.
Gut Microbes ; 11(5): 1374-1384, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32401138

RESUMO

BACKGROUND: Little data are available on the subject of gut microbiota composition in endurance athletes as well as connections between diet and specific bacteria abundance. However, most studies suggest that athletes' microbiota undergoes major alterations, which may contribute to increased physical performance. Therefore, we decided to investigate differences in gut microbiota between healthy controls and endurance athletes. MATERIALS AND METHODS: Stools samples were collected from 14 marathon runners, 11 cross-country skiers and 46 sedentary healthy controls. The athletes' diet evaluation was performed with 24-h diet recall, using the Aliant programme. The 16S gene sequencing was performed using the Ion 16S Metagenomics Kit on Ion Torrent PGM sequencer. Taxonomic classification and diversity indices computation was performed with Mothur. RESULTS: 20 and 5 taxa differentiated healthy controls from marathon runners and cross-country skiers, respectively. Both groups presented a lowered abundance of major gut microbiota genus, Bacteroidetes and higher abundance of Prevotella. The athletes' microbiome was also more diverse in cross-country skiers than the one of sedentary controls (Simpson index p-value at 0.025). Thirty-one strong correlations (Spearman's coefficient > 0.6) were uncovered between bacteria abundance and diet, including inverse correlation of Prevotella with sucrose intake, Phascolarctobacterium with polyunsaturated fatty acids as well as positive correlation of Christensenellaceae with folic acid intake and Agathobacter with fiber amount in diet. CONCLUSIONS: The excessive training associates with both differences in composition and promotion of higher bacterial diversity. Taxons enriched in athletes are known to participate in fiber fermentation.


Assuntos
Atletas , Bactérias/classificação , Treino Aeróbico , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Corrida de Maratona , Esqui , Adolescente , Adulto , Idoso , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bacteroides/crescimento & desenvolvimento , Dieta , Fibras na Dieta/administração & dosagem , Sacarose na Dieta/administração & dosagem , Fezes/microbiologia , Feminino , Genes de RNAr , Humanos , Masculino , Pessoa de Meia-Idade , Resistência Física , Polônia , Prevotella/crescimento & desenvolvimento , RNA Ribossômico 16S , Comportamento Sedentário , Adulto Jovem
20.
Gut Microbes ; 11(5): 1362-1373, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32453615

RESUMO

The human gut microbiota develops soon after birth and can acquire inter-individual variation upon exposure to intrinsic and environmental cues. However, inter-individual variation has not been comprehensively assessed in a multi-ethnic study. We studied a longitudinal birth cohort of 106 infants of three Asian ethnicities (Chinese, Malay, and Indian) that resided in the same geographical location (Singapore). Specific and temporal influences of ethnicity, mode of delivery, breastfeeding status, gestational age, birthweight, gender, and maternal education on the development of the gut microbiota in the first 2 years of life were studied. Mode of delivery, breastfeeding status, and ethnicity were identified as the main factors influencing the compositional development of the gut microbiota. Effects of delivery mode and breastfeeding status lasted until 6M and 3M, respectively, with the primary impact on the diversity and temporal colonization of the genera Bacteroides and Bifidobacterium. The effect of ethnicity was apparent at 3M post-birth, even before the introduction of weaning (complementary) foods, and remained significant after adjusting for delivery mode and breastfeeding status. Ethnic influences remained significant until 12M in the Indian and Chinese infants. The microbiota of Indian infants was characterized by higher abundances of Bifidobacterium and Lactobacillus, while Chinese infants had higher abundances of Bacteroides and Akkermansia. These findings provide a detailed insight into the specific and temporal influences of early life factors and ethnicity in the development of the human gut microbiota. Trial Registration: Clinicaltrials.gov registration no. NCT01174875.


Assuntos
Bactérias/crescimento & desenvolvimento , Aleitamento Materno , Parto Obstétrico , Etnicidade , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Akkermansia/crescimento & desenvolvimento , Bactérias/classificação , Bacteroides/crescimento & desenvolvimento , Bifidobacterium/crescimento & desenvolvimento , Pré-Escolar , China , Fezes/microbiologia , Feminino , Humanos , Índia , Lactente , Recém-Nascido , Lactobacillus/crescimento & desenvolvimento , Malásia , Masculino , Desmame
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...