Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Food Funct ; 15(18): 9368-9389, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39189385

RESUMEN

Inflammation significantly influences the degeneration of dopaminergic neurons in Parkinson's disease (PD), which is potentially intensified by associated gut dysbiosis. The therapeutic potential of probiotics, due to their antioxidant, anti-inflammatory, and gut microbiota modulatory properties, is explored herein as a means to improve gut health and influence the gut-brain-microbiota axis in the context of PD. In this study, we investigated the role and possible mechanism of Bifidobacterium animalis subsp. lactis MH-022 (B. lactis MH-022) supplementation in a 6-hydroxydopamine (6-OHDA)-induced rat model of PD. Findings demonstrated that B. lactis MH-022 supplementation markedly ameliorated motor deficits, preserved dopaminergic neurons, enhanced the antioxidant capacity, and mitigated inflammation through restoring mitochondrial function. Furthermore, B. lactis MH-022 supplementation significantly altered the gut microbiota composition, augmenting the production of short-chain fatty acids and promoting the proliferation of beneficial bacterial taxa, thereby reinforcing their anti-inflammatory properties. Correlation analyses established strong associations between specific bacterial taxa and improvements in motor function, antioxidant levels, and reductions in inflammation markers. These insights emphasize the therapeutic potential of B. lactis MH-022 in modulating diverse aspects of PD, particularly highlighting its role in reducing inflammation, restoring mitochondrial function, enhancing antioxidant capacity, and reshaping the gut microbiota. This multifaceted approach underscores the probiotic's potential in reducing neuroinflammation and protecting dopaminergic neurons, thus offering a promising avenue for PD treatment.


Asunto(s)
Bifidobacterium animalis , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Inflamación , Oxidopamina , Enfermedad de Parkinson , Probióticos , Animales , Ratas , Probióticos/farmacología , Probióticos/uso terapéutico , Masculino , Enfermedad de Parkinson/terapia , Enfermedad de Parkinson/metabolismo , Bifidobacterium animalis/fisiología , Ratas Sprague-Dawley , Disbiosis/microbiología , Disbiosis/terapia , Suplementos Dietéticos
2.
Nutrients ; 15(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38004159

RESUMEN

It has been found that Streptococcus thermophilus (S. thermophilus) influenced the gut microbiota and host metabolism with strain specificity in C57BL/6J mice in the previous study, though it remains unclear whether lactose as a dietary factor associated with dairy consumption is involved as the mediator in the interaction. In the present study, integrated analysis of 16S rRNA gene sequencing and untargeted metabolomics by liquid chromatography-mass spectrometry of fecal samples in C57BL/6J mice was applied to evaluate the effect of lactose on the regulation of gut microbiota by two S. thermophilus strains (4M6 and DYNDL13-4). The results showed that the influence of lactose supplementation on gut microbiota induced by S. thermophilus ingestion was strain-specific. Although two S. thermophilus strains ingestion introduced similar perturbations in the fecal microbiota and gut microbial metabolism, the regulation of DYNDL13-4 on the gut microbiota and metabolism was more affected by lactose than 4M6. More specifically, lactose and 4M6 supplementation mainly enriched pathways of d-glutamine and d-glutamate metabolism, alanine, aspartate, and glutamate metabolism, and tryptophan and phenylalanine metabolism in the gut, whereas 4M6 only enriched tryptophan and phenylalanine metabolism. DYNDL13-4-L (DYNDL13-4 with lactose) had significant effects on sulfur, taurine, and hypotaurine metabolism in the gut and on phenylalanine, tyrosine, tryptophan biosynthesis, and linoleic acid metabolism in serum relative to the DYNDL13-4. Our study demonstrated the strain-specific effect of lactose and S. thermophilus supplementation on gut microbiota and host metabolism. However, considering the complexity of the gut microbiota, further research is necessary to provide insights to facilitate the design of personalized fermented milk products as a dietary therapeutic strategy for improving host health.


Asunto(s)
Microbioma Gastrointestinal , Streptococcus thermophilus , Ratones , Animales , Streptococcus thermophilus/metabolismo , Lactosa/metabolismo , Triptófano/metabolismo , ARN Ribosómico 16S/metabolismo , Ratones Endogámicos C57BL , Metaboloma , Fenilalanina/metabolismo , Suplementos Dietéticos
3.
Microorganisms ; 10(12)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36557585

RESUMEN

In natural settings, approximately 40-80% of bacteria exist as biofilms, most of which are mixed-species biofilms. Previous studies have typically focused on single- or dual-species biofilms. To expand the field of study on gut biofilms, we found a group of gut microbiota that can form biofilms well in vitro: Bifidobacterium longum subsp. infantis, Enterococcus faecalis, Bacteroides ovatus, and Lactobacillus gasseri. The increase in biomass and bio-volume of the mixed-species biofilm was confirmed via crystal violet staining, field emission scanning electron microscopy, and confocal laser scanning microscopy, revealing a strong synergistic relationship in these communities, with B. longum being the key biofilm-contributing species. This interaction may be related to changes in the cell number, biofilm-related genes, and metabolic activities. After quantifying the cell number using quantitative polymerase chain reaction, B. longum and L. gasseri were found to be the dominant flora in the mixed-species biofilm. In addition, this study analyzed biological properties of mixed-species biofilms, such as antibiotic resistance, cell metabolic activity, and concentration of water-insoluble polysaccharides. Compared with single-species biofilms, mixed-species biofilms had higher metabolic activity, more extracellular matrix, and greater antibiotic resistance. From these results, we can see that the formation of biofilms is a self-protection mechanism of gut microbiota, and the formation of mixed-species biofilms can greatly improve the survival rate of different strains. Finally, this study is a preliminary exploration of the biological characteristics of gut biofilms, and the molecular mechanisms underlying the formation of biofilms warrant further research.

4.
Food Funct ; 13(21): 11153-11168, 2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36205751

RESUMEN

Glucan is the most widely distributed glycan. Many probiotics such as lactic acid bacteria (LAB) encoded corresponding hydrolytic enzymes, which could use these glucans as energy substances. Brown alga is rich in glucan and has high edible and medicinal value, but research on its regulation to probiotics is not detailed enough. In this study, we determined a novel neutral α type gluco-oligosaccharide from the brown alga Laminaria japonica with a degree of polymerization (DP) of 2-8 and a structure that mainly consists of α-(1→4)-linked glycosidic bonds called Laminaria japonica gluco-oligosaccharide (LJGO). Fermentation in vitro and gene-phenotype correlation analyses revealed that LJGO selectively stimulated the growth of the LAB strain encoding a specific ATP-binding cassette (ABC) transport system in a GH13 gene cluster, with apparent differences among 14 tested species. Comparative genomics further revealed that this transport system is species-specific, implying a potential contribution to species evolution. Transcriptomic analysis based on LAB strains cultured on LJGO and 1H-NMR findings of LJGO residues after strain utilization showed that the GH13 gene cluster contains functional LAB genes involved in LJGO utilization. Further verification by gene knockout studies is needed to expand our findings.


Asunto(s)
Lactobacillales , Laminaria , Laminaria/química , Oligosacáridos , Glucanos , Polisacáridos
5.
Front Microbiol ; 13: 1001313, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36090099

RESUMEN

Respiratory syncytial virus (RSV) infection is a constant threat to the health of young children, and this is mainly attributed to the lack of effective prevention strategies. This study aimed to determine whether Lactobacillus (L.) mucosae, a potential probiotic, could protect against respiratory viral infection in a mouse model. Naive 3-4-week-old BALB/c mice were orally administered with three L. mucosae strains (2.5 × 108 CFU/mouse) 7 days before RSV infection (105 TCID50/mouse). Results showed that all three strains inhibited RSV replication and reduced the proportions of inflammatory cells, including granulocytes and monocytes in the blood. The L. mucosae M104R01L3 treatment maintained stable weight in mice and increased interferon (IFN)-ß and tumor necrosis factor (TNF)-α levels. The L. mucosae DCC1HL5 treatment increased interleukin (IL)-1ß and IL-10 levels. Moreover, the M104R01L3 and DCC1HL5 strains increased the proportions of Akkermansia, Alistipes, and Anaeroplasma which contributed to the advantageous modulation of the gut microbiota. Besides, L. mucosae affected the gut levels of short-chain fatty acids (SCFAs) that are important for the antiviral response. L. mucosae 1,025 increased acetate, propionate, and butyrate levels, whereas L. mucosae M104R01L3 increased the level of acetate in the gut. L. mucosae M104R01L3 may protect against viral infection by upregulating the IFN-ß levels in the lungs and its antiviral effect may be related to the increase of acetate levels in the gut. In conclusion, the three L. mucosae strains exerted antiviral effects against RSV infection by differentially regulating immune responses and intestinal micro-ecological balance. This study can provide a reference for studying the mechanisms underlying the antiviral effects of L. mucosae.

6.
Front Nutr ; 9: 862934, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35464005

RESUMEN

The coexistence of allergic rhinitis (AR) and asthma reinforces the concept of "one airway, one disease," which has prompted the exploration for a single intervention to treat both diseases. Lactobacillus reuteri CCFM1040 (CCFM1040) was found to be an inhibitor of the common pathogenesis of AR and asthma in our previous studies. This study presented a randomized, placebo-controlled trial to investigate the clinical effects of CCFM1040 on both diseases. The total symptom score (TSS), the quality of life (QoL), and the modulation in the gut microbiota of patients with AR, the Asthma Control and Test (ACT) of patients with asthma, and the safety of both AR and asthma were measured. In patients with AR, CCFM1040 numerically decreased TSS, Rhinoconjunctivitis Quality of Life Questionnaire (RQLQ), 3 nasal scores in TSS (nasal congestion, watery eyes, and rhinorrhea), and sleep and significantly improved (P = 0.014) non-nose/eye symptoms. The ACT score was numerically increased in patients with asthma (from partially controlled to well-controlled). Significant microbial (from class level to genus level) and metabolic differences (P < 0.05) were found in patients with AR. No adverse reactions were observed. No effect on the blood and urine routine indexes. CCFM1040 has a potential benefit on both diseases. Further studies based on these findings will help to optimize the management of AR and asthma.

7.
Gene ; 826: 146449, 2022 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-35337850

RESUMEN

Biofilm formation by bacteria represents an adaptation strategy to the environment, and some special genes may lead to a strong biofilm phenotype. In this study, we attempted to find functional genes associated with bifidobacterial biofilm formation. Firstly, we evaluated the biofilm formation ability of bifidobacterial strains from six species, which showed that Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium animalis, Bifidobacterium adolescentis, and Bifidobacterium pseudocatenulatum had biofilm-forming and non-biofilm-forming strains, while all Bifidobacterium bifidum strains could form strong biofilms. Then 48 strains were selected for genome sequencing and comparative analysis. The gene-trait matching analysis revealed that B. bifidum biofilm formation phenotype may associate with their unique genes, involving in stress response, quorum sensing, two components, and peptide synthesis. B. pseudocatenulatum biofilm formation was positively correlated with the eps cluster (rfbX). While no genotype related to the biofilm phenotype was found in B. longum using this analysis, but all contain autoinducer-2 (AI-2) receptor genes. Moreover, luxS, rbsB, rfbX were selected for real-time qPCR analysis, suggesting that their expression are important to biofilm formation. These results indicated that strains carrying certain genes tend to form stronger biofilms than those formed by strains without these genes.


Asunto(s)
Bifidobacterium , Percepción de Quorum , Bacterias , Bifidobacterium/genética , Bifidobacterium/metabolismo , Biopelículas , Fenotipo , Percepción de Quorum/genética
8.
Food Funct ; 13(6): 3704-3719, 2022 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-35266474

RESUMEN

Food allergy (FA) is a common immune disorder caused by food antigens. Probiotic strains showed alleviating effects on FA, such as the alleviation of FA pathological symptoms, serum OVA-sIgE levels, and the gut microbiota diversity and composition. The results showed that intragastric administration of Lactiplantibacillus plantarum CCFM1189, Limosilactobacillus reuteri CCFM1190, and Bifidobacterium longum CCFM1029 alleviated the weight loss and FA pathological symptoms of FA mice and decreased OVA-specific IgE and histamine (HIS) levels. CCFM1189 and CCFM1190 decreased IL-4, IL-5, and IL-13 levels, while CCFM1189 and CCFM 1029 decreased IL-17 levels. The gut microbiota analysis demonstrated that CCFM1189 increased the abundance of Akkermansia, while CCFM1190 improved immune regulation bacteria such as Faecalibaculum. CCFM1029 increased Bifidobacterium and the bacteria involved in short-chain fatty acid (SCFA) production, such as Dubosiella. L. plantarum CCFM1189 and L. reuteri CCFM1190 improved indoleacrylic acid levels in mouse fecal samples using untargeted metabolomics analysis. In conclusion, CCFM1189, CCFM1190, and CCFM1029 decreased Th2 immune responses and alleviated FA pathological symptoms by regulating the gut microbiota diversity and composition, and altering gut microbial metabolites, which could provide support in clinical tests and probiotic production in the future.


Asunto(s)
Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Probióticos , Animales , Indoles , Ratones , Ovalbúmina , Probióticos/farmacología
9.
Mol Nutr Food Res ; 66(10): e2101057, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35286767

RESUMEN

Major depressive disorder (MDD) is an enfeebling disease with a lifetime incidence of 20%. While accumulating studies implicate a correlation between the disease and gut microbiota, data show that not every patient responded to probiotic treatments. To comprehensively assess the potential role of probiotics in MDD, this study first summarizes the current pathological hypothesis of the disease from a life-stage perspective, focuses on the potential role of "depression gut microbiota." Currently available managements are then briefly summarized and novel bio-materials having potential therapeutic effects on MDD are also evaluated. To harness the positive effect of probiotics, prebiotics, and postbiotics, clinical evidence and their applications on MDD patients are listed. Factors that may counteract the pre/probiotic applications, such as diet, physiology, gender difference, and use of antibiotics and antidepressants are also discussed. The endocannabinoid (eCBs) system may be promising targets for probiotic therapy. More evidence is needed to demonstrate the hierarchical factors in the complex network driving the disease, and probiotic can be one promising adjunct for patients with MDD.


Asunto(s)
Trastorno Depresivo Mayor , Microbioma Gastrointestinal , Probióticos , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Humanos , Prebióticos , Probióticos/uso terapéutico
10.
Neurosci Biobehav Rev ; 135: 104556, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35122783

RESUMEN

Alzheimer's disease (AD) is one of the fastest growing cognitive decline-related neurological diseases. To date, effective curative strategies have remained elusive. A growing body of evidence indicates that dietary patterns have significant effects on cognitive function and the risk of developing AD. Previous studies on the association between diet and AD risk have mainly focused on individual food components and specific nutrients, and the mechanisms responsible for the beneficial effects of dietary patterns on AD are not well understood. This article provides a comprehensive overview of the effects of dietary patterns, including the Mediterranean diet (MedDiet), dietary approaches to stop hypertension (DASH) diet, Mediterranean-DASH diet intervention for neurological delay (MIND), ketogenic diet, caloric restriction, intermittent fasting, methionine restriction, and low-protein and high-carbohydrate diet, on cognitive impairment and summarizes the underlying mechanisms by which dietary patterns attenuate cognitive impairment, especially highlighting the modulation of dietary patterns on cognitive impairment through gut microbiota. Furthermore, considering the variability in individual metabolic responses to dietary intake, we put forward a framework to develop personalized dietary patterns for people with cognitive disorders or AD based on individual gut microbiome compositions.


Asunto(s)
Enfermedad de Alzheimer , Trastornos del Conocimiento , Disfunción Cognitiva , Dieta Mediterránea , Enfoques Dietéticos para Detener la Hipertensión , Enfermedad de Alzheimer/psicología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Humanos
11.
Food Funct ; 12(24): 12606-12620, 2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34821239

RESUMEN

Brown algae glycan from Laminaria japonica (LJNP) is a heterogeneous glycan with two apparent molecular weights of 1.1 and 37.3 kDa, and is mainly composed of α ß-glucan and a few fucosyl residues. To explore the regulation of gut microbiota and the host, LJNP and 2'-fucosyllactose (2'FL) were compared to investigate their effect on mice via oral administration. Using metagenomic and metabolomic analyses, we found that 2'FL mainly relied on Adlercreutzia equolifaciens and Akkermansia muciniphila to improve gut amino acid and bile acid metabolism, whereas LJNP mainly drove Bacteroides vulgatus and Bacteroides uniformis to regulate gut amino acid metabolism and glycometabolism. Moreover, LJNP showed a weight loss effect and better protection of the intestinal barrier than 2'FL. We further employed LJNP and 2'FL on a germ-free mice model. Interestingly, the body weight management was not microbiome mediated. This study showed that LJNP can ameliorate the intestinal barrier through modulation of the gut microbiota, maintain the blood glucose level, and regulate body weight and the antioxidant function. Although the benefits of LJNP on host health were partly revealed, mechanisms such as the weight loss effect require further study.


Asunto(s)
Peso Corporal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Laminaria/metabolismo , Polisacáridos/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Animales
12.
Genes (Basel) ; 12(10)2021 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-34680899

RESUMEN

The potential probiotic benefits of Bifidobacterium bifidum have received increasing attention recently. We used comparative genomic analysis to explore the differences in the genome and the physiological characteristics of B. bifidum isolated from the fecal samples of Chinese adults and infants. The relationships between genotypes and phenotypes were analyzed to assess the effects of isolation sources on the genetic variation of B. bifidum. The phylogenetic tree results indicated that the phylogeny of B. bifidum may be related to the geographical features of its isolation source. B. bifidum was found to have an open pan-genome and a conserved core genome. The genetic diversity of B. bifidum is mainly reflected in carbohydrate metabolism- and immune/competition-related factors, such as the glycoside hydrolase gene family, bacteriocin operons, antibiotic resistance genes, and clustered regularly interspaced short palindromic repeats (CRISPR)-Cas. Additionally, the type III A CRISPR-Cas system was discovered in B. bifidum for the first time. B. bifidum strains exhibited niche-specific characteristics, and the results of this study provide an improved understanding of the genetics of this species.


Asunto(s)
Bifidobacterium bifidum/genética , Adulto , Bifidobacterium bifidum/aislamiento & purificación , Heces/microbiología , Genes Bacterianos , Humanos , Lactante
13.
Microorganisms ; 9(10)2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34683449

RESUMEN

Lactic acid bacteria (LAB) are widely used as probiotics in the food industry owing to their beneficial effects on human health. However, numerous antibiotic resistance genes have been found in LAB strains, especially tetracycline resistance genes. Notably, the potential transferability of these genes poses safety risks. To comprehensively evaluate tetracycline resistance in LAB, we determined the tetracycline susceptibility patterns of 478 LAB strains belonging to four genera and eight species. By comparing phenotypes with genotypes based on genome-wide annotations, five tetracycline resistance genes, tet(M), tet(W/N/W), tet(L), tet(S), and tet(45), were detected in LAB. Multiple LAB strains without tetracycline resistance genes were found to be resistant to tetracycline at the currently recommended cutoff values. Thus, based on the minimum inhibitory concentrations of tetracycline for these LAB strains, the species-specific microbiological cutoff values for Lactobacillus (para)gasseri, Lactobacillus johnsonii, and Lactobacillus crispatus to tetracycline were first developed using the Turnidge, Kronvall, and eyeball methods. The cutoff values for Lactiplantibacillus plantarum were re-established and could be used to better distinguish susceptible strains from strains with acquired resistance. Finally, we verified that these five genes play a role in tetracycline resistance and found that tet(M) and tet(W/N/W) are the most widely distributed tetracycline resistance genes in LAB.

14.
Microorganisms ; 9(10)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34683469

RESUMEN

(1) Background: Constipation is a common condition that affects the health and the quality of life of patients. Recent studies have suggested that the gut microbiome is associated with constipation, but these studies were mainly focused on a single research cohort. Thus, we aimed to construct a classification model based on fecal bacterial and identify the potential gut microbes' biomarkers. (2) Methods: We collected 3056 fecal amplicon sequence data from five research cohorts. The data were subjected to a series of analyses, including alpha- and beta-diversity analyses, phylogenetic profiling analyses, and systematic machine learning to obtain a comprehensive understanding of the association between constipation and the gut microbiome. (3) Results: The alpha diversity of the bacterial community composition was higher in patients with constipation. Beta diversity analysis evidenced significant partitions between the two groups on the base of gut microbiota composition. Further, machine learning based on feature selection was performed to evaluate the utility of the gut microbiome as the potential biomarker for constipation. The Gradient Boosted Regression Trees after chi2 feature selection was the best model, exhibiting a validation performance of 70.7%. (4) Conclusions: We constructed an accurate constipation discriminant model and identified 15 key genera, including Serratia, Dorea, and Aeromonas, as possible biomarkers for constipation.

16.
Microorganisms ; 9(9)2021 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-34576890

RESUMEN

The infant gut microbiota plays a critical role in early life growth and derives mainly from maternal gut and breast milk. This study aimed to analyze the differences in the gut microbiota, namely Bifidobacterium and Lactobacillus communities at species level among breast milk as well as maternal and infant feces at different time points after delivery. Fifty-one mother-infant pairs from Indonesia were recruited, and the breast milk and maternal and infant feces were collected and analyzed by high throughput sequencing (16S rRNA, Bifidobacterium groEL and Lactobacillus groEL genes). PCoA results showed bacterial composition was different among breast milk and maternal and infant feces within the first two years. The abundance of Bifidobacterium and Bacteroides were significantly higher in infant feces compared to their maternal feces from birth to two years of age, and maternal breast milk within six months after birth (p < 0.05), whereas the abundance of Blautia, Prevotella, and Faecalibacterium was higher in maternal feces compared to that in breast milk within six months and infant feces within one year after birth, respectively (p < 0.05). The relative abundances of Bacteroides and Lactobacillus was higher and lower in infant feces compared to that in maternal feces only between one and two years of age, respectively (p < 0.05). For Bifidobacterium community at species level, B. adolescentis, B. ruminantium, B. longum subsp. infantis, B. bifidum, and B. pseudolongum were identified in all samples. However, the profile of Bifidobacterium was different between maternal and infant feces at different ages. The relative abundances of B. adolescentis and B. ruminantium were higher in maternal feces compared to those in infant feces from birth to one year of age (p < 0.05), while the relative abundances of B. longum subsp. infantis and B. bifidum were higher in infant feces compared to those in maternal feces beyond three months, and the relative abundance of B. pseudolongum was only higher in infant feces between three and six months (p < 0.05). For Lactobacillus community, L. paragasseri showed higher relative abundance in infant feces when the infant was younger than one year of age (p < 0.05). This study showed bacterial composition at the genus level and Bifidobacterium and Lactobacillus communities at the species level were stage specific in maternal breast milk as well as and maternal and infant feces.

17.
NPJ Biofilms Microbiomes ; 7(1): 71, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489454

RESUMEN

The gut microbiota could affect human health and disease. Although disease-associated microbiota alteration has been extensively investigated in the Chinese population, a nationwide Chinese gut microbiota baseline is still lacking. Here we performed 16 S rRNA gene sequencing on fecal samples from 2678 healthy Chinese individuals, who belonged to eight ethnic groups and resided in 63 counties/cities of 28 provinces. We identified four enterotypes, three of which were enriched for Prevotella, Bacteroides, and Escherichia, respectively, whereas the fourth one had no dominant genus. By assessing the association between the gut microbiota and 20 variables belonging to six categories, geography, demography, diet, urbanization, lifestyle, and sampling month, we revealed that geography explained the largest microbiota variation, and clarified the distinct patterns in the associations with staple food type, ethnicity, and urban/rural residence. Specifically, the gut microbiota of Han Chinese and ethnic minority groups from the same sites was more alike than that of the same ethnic minority groups from different sites. Individuals consuming wheat as staple food were predicted to have more microbial genes involving in glucan 1,3-beta-glucosidase and S-adenosyl-L-methionine biosynthesis than those who consumed rice, based on functional prediction. Besides, an appreciable effect of urbanization on decreased intra-individual diversity, increased inter-individual diversity, and increased proportion of the Bacteroides enterotype was observed. Collectively, our study provided a nationwide gut microbiota baseline of the Chinese population and knowledge on important covariates, which are fundamental to translational microbiota research.


Asunto(s)
Pueblo Asiatico , Dieta , Etnicidad , Tracto Gastrointestinal , Urbanización , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Bacterias/clasificación , Bacterias/genética , Biodiversidad , Niño , Preescolar , China , Minorías Étnicas y Raciales , Heces/microbiología , Femenino , Humanos , Estilo de Vida , Masculino , Persona de Mediana Edad , Grupos Minoritarios , ARN Ribosómico 16S/genética , Adulto Joven
18.
Int J Mol Sci ; 22(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34299216

RESUMEN

Bifidobacterium bifidum strains, an important component of probiotic foods, can form biofilms on abiotic surfaces, leading to increased self-resistance. However, little is known about the molecular mechanism of B. bifidum biofilm formation. A time series transcriptome sequencing and untargeted metabolomics analysis of both B. bifidum biofilm and planktonic cells was performed to identify key genes and metabolites involved in biofilm formation. Two hundred thirty-five nonredundant differentially expressed genes (DEGs) (including vanY, pstS, degP, groS, infC, groL, yajC, tadB and sigA) and 219 nonredundant differentially expressed metabolites (including L-threonine, L-cystine, L-tyrosine, ascorbic acid, niacinamide, butyric acid and sphinganine) were identified. Thirteen pathways were identified during the integration of both transcriptomics and metabolomics data, including ABC transporters; quorum sensing; two-component system; oxidative phosphorylation; cysteine and methionine metabolism; glutathione metabolism; glycine, serine and threonine metabolism; and valine, leucine and isoleucine biosynthesis. The DEGs that relate to the integration pathways included asd, atpB, degP, folC, ilvE, metC, pheA, pstS, pyrE, serB, ulaE, yajC and zwf. The differentially accumulated metabolites included L-cystine, L-serine, L-threonine, L-tyrosine, methylmalonate, monodehydroascorbate, nicotinamide, orthophosphate, spermine and tocopherol. These results indicate that quorum sensing, two-component system and amino acid metabolism are essential during B. bifidum biofilm formation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bifidobacterium bifidum/fisiología , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/genética , Bifidobacterium bifidum/genética , Bifidobacterium bifidum/metabolismo , Perfilación de la Expresión Génica , Metaboloma , Percepción de Quorum , Transcriptoma , Triticum/microbiología
19.
BMC Microbiol ; 21(1): 169, 2021 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-34090326

RESUMEN

BACKGROUND: Campylobacter jejuni is the major micro-bacillary pathogen responsible for human coloenteritis. Lactic acid bacteria (LAB) have been shown to protect against Campylobacter infection. However, LAB with a good ability to inhibit the growth of C. jejuni in vitro are less effective in animals and animal models, and have the disadvantages of high cost, a long cycle, cumbersome operation and insignificant immune response indicators. Caenorhabditis elegans is increasingly used to screen probiotics for their anti-pathogenic properties. However, no research on the use of C. elegans to screen for probiotic candidates antagonistic to C. jejuni has been conducted to date. RESULTS: This study established a lifespan model of C. elegans, enabling the preselection of LAB to counter C. jejuni infection. A potential protective mechanism of LAB was identified. Some distinct LAB species offered a high level of protection to C. elegans against C. jejuni. The LAB strains with a high protection rate reduced the load of C. jejuni in C. elegans. The transcription of antibacterial peptide genes, MAPK and Daf-16 signalling pathway-related genes was elevated using the LAB isolates with a high protection rate. The reliability of the lifespan model of C. elegans was verified using mice and chickens infected with C. jejuni. CONCLUSIONS: The results showed that different LAB had different abilities to protect C. elegans against C. jejuni. C. elegans provides a reliable model for researchers to screen for LAB that are antagonistic to C. jejuni on a large scale.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/inmunología , Infecciones por Campylobacter/tratamiento farmacológico , Campylobacter jejuni/efectos de los fármacos , Modelos Animales de Enfermedad , Lactobacillales/fisiología , Probióticos/administración & dosificación , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/inmunología , Infecciones por Campylobacter/genética , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/crecimiento & desarrollo , Pollos/genética , Pollos/inmunología , Pollos/microbiología , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Humanos , Ratones/genética , Ratones/inmunología , Ratones/microbiología , Ratones Endogámicos C57BL , Nematodos/genética , Nematodos/inmunología , Nematodos/microbiología
20.
J Nutr ; 151(7): 1703-1716, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33982127

RESUMEN

The rate of obesity is rapidly increasing and has become a health and economic burden worldwide. As recent studies have revealed that the gut microbiota is closely linked to obesity, researchers have used various approaches to modulate the gut microbiota to treat the condition. Dietary composition and energy intake strongly affect the composition and function of the gut microbiota. Intestinal microbial changes alter the composition of bile acids and fatty acids and regulate bacterial lipopolysaccharide production, all of which influence energy metabolism and immunity. Evidence also suggests that remodeling the gut microbiota through intake of probiotics, prebiotics, fermented foods, and dietary plants, as well as by fecal microbiota transplantation, are feasible methods to remediate obesity.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Metabólicas , Probióticos , Humanos , Obesidad , Prebióticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA