Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27.155
Filtrer
1.
PeerJ ; 12: e17502, 2024.
Article de Anglais | MEDLINE | ID: mdl-38952971

RÉSUMÉ

Background: Desserts with vegetable ingredients are a constantly expanding global market due to the search for alternatives to cow's milk. Fermentation of these matrices by lactic acid bacteria can add greater functionality to the product, improving its nutritional, sensory, and food safety characteristics, as well as creating bioactive components with beneficial effects on health. Concern for health and well-being has aroused interest in byproducts of the industry that have functional properties for the body, such as mature coconut water, a normally discarded residue that is rich in nutrients. This study aimed to develop a probiotic gelatin based on pulp and water from mature coconuts and evaluate the physicochemical characteristics, viability of the Lacticaseibacillus rhamnosus LR32 strain in the medium, as well as the texture properties of the product. Methods: After collection and cleaning, the physicochemical characterization, mineral analysis, analysis of the total phenolic content and antioxidant activity of mature coconut water were carried out, as well as the centesimal composition of its pulp. Afterwards, the gelling was developed with the addition of modified corn starch, gelatin, sucrose, and probiotic culture, being subjected to acidity analysis, texture profile and cell count, on the first day and every 7 days during 21 days of storage, under refrigeration at 5 °C. An analysis of the centesimal composition was also carried out. Results: The main minerals in coconut water were potassium (1,932.57 mg L-1), sodium (19.57 mg L-1), magnesium (85.13 mg L-1) calcium (279.93 mg L-1) and phosphorus (11.17 mg L- 1), while the pulp had potassium (35.96 g kg-1), sodium (0.97 g kg-1), magnesium (2.18 g kg-1), 37 calcium (1.64 g kg-1), and phosphorus (3.32 g kg-1). The phenolic content of the water and pulp was 5.72 and 9.77 mg gallic acid equivalent (GAE) 100 g-1, respectively, and the antioxidant capacity was 1.67 and 0.98 39 g of 2, 2-diphenyl-1-picrylhydrazyl (DPPH) mg-1, respectively. The coconut pulp had 2.81 g 100 g-1of protein, 1.11 g 100 g-1 of 40 ash, 53% moisture, and 5.81 g 100 g-1 of carbohydrates. The gelatin produced during the storage period presented firmness parameters ranging from 145.82 to 206.81 grams-force (gf), adhesiveness from 692.85 to 1,028.63 gf sec, cohesiveness from 0.604 to 0.473, elasticity from 0.901 to 0.881, gumminess from 86.27 to 97.87 gf, and chewiness from 77.72 to 91.98 gf. Regarding the viability of the probiotic microorganism, the dessert had 7.49 log CFU g-1 that remained viable during the 21-day storage, reaching 8.51 CFU g-1. Acidity ranged from 0.15 to 0.64 g of lactic acid 100 g-1. The centesimal composition of the product showed 4.88 g 100 g-1 of protein, 0.54 g 100 g-1 of ash, 85.21% moisture, and 5.37g 100 g-1 of carbohydrates. The development of the gelatin made it possible to obtain a differentiated product, contributing to diversification in the food sector, providing a viable alternative for maintaining consumer health and reducing costs compared to desserts already available on the market.


Sujet(s)
Cocos , Gélatine , Lacticaseibacillus rhamnosus , Probiotiques , Cocos/composition chimique , Cocos/microbiologie , Gélatine/composition chimique , Antioxydants/pharmacologie , Antioxydants/composition chimique , Fermentation
2.
Parasit Vectors ; 17(1): 284, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956725

RÉSUMÉ

BACKGROUND: Toxoplasma gondii infection affects a significant portion of the global population, leading to severe toxoplasmosis and, in immunocompromised patients, even death. During T. gondii infection, disruption of gut microbiota further exacerbates the damage to intestinal and brain barriers. Therefore, identifying imbalanced probiotics during infection and restoring their equilibrium can regulate the balance of gut microbiota metabolites, thereby alleviating tissue damage. METHODS: Vimentin gene knockout (vim-/-) mice were employed as an immunocompromised model to evaluate the influence of host immune responses on gut microbiota balance during T. gondii infection. Behavioral experiments were performed to assess changes in cognitive levels and depressive tendencies between chronically infected vim-/- and wild-type (WT) mice. Fecal samples were subjected to 16S ribosomal RNA (rRNA) sequencing, and serum metabolites were analyzed to identify potential gut probiotics and their metabolites for the treatment of T. gondii infection. RESULTS: Compared to the immunocompetent WT sv129 mice, the immunocompromised mice exhibited lower levels of neuronal apoptosis and fewer neurobehavioral abnormalities during chronic infection. 16S rRNA sequencing revealed a significant decrease in the abundance of probiotics, including several species of Lactobacillus, in WT mice. Restoring this balance through the administration of Lactobacillus murinus and Lactobacillus gasseri significantly suppressed the T. gondii burden in the intestine, liver, and brain. Moreover, transplantation of these two Lactobacillus spp. significantly improved intestinal barrier damage and alleviated inflammation and neuronal apoptosis in the central nervous system. Metabolite detection studies revealed that the levels of various Lactobacillus-related metabolites, including indole-3-lactic acid (ILA) in serum, decreased significantly after T. gondii infection. We confirmed that L. gasseri secreted much more ILA than L. murinus. Notably, ILA can activate the aromatic hydrocarbon receptor signaling pathway in intestinal epithelial cells, promoting the activation of CD8+ T cells and the secretion of interferon-gamma. CONCLUSION: Our study revealed that host immune responses against T. gondii infection severely disrupted the balance of gut microbiota, resulting in intestinal and brain damage. Lactobacillus spp. play a crucial role in immune regulation, and the metabolite ILA is a promising therapeutic compound for efficient and safe treatment of T. gondii infection.


Sujet(s)
Lésions encéphaliques , Microbiome gastro-intestinal , Souris knockout , Toxoplasma , Animaux , Souris , Toxoplasma/immunologie , Lésions encéphaliques/immunologie , Probiotiques/administration et posologie , Encéphale/immunologie , Lactobacillus , Modèles animaux de maladie humaine , Sujet immunodéprimé , Toxoplasmose/immunologie , ARN ribosomique 16S/génétique , Mâle , Intestins/immunologie
3.
Hepatol Commun ; 8(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967596

RÉSUMÉ

With an increasing prevalence, metabolic dysfunction-associated steatotic liver disease (MASLD) has become a major global health problem. MASLD is well-known as a multifactorial disease. Mitochondrial dysfunction and alterations in the gut bacteria are 2 vital events in MASLD. Recent studies have highlighted the cross-talk between microbiota and mitochondria, and mitochondria are recognized as pivotal targets of the gut microbiota to modulate the host's physiological state. Mitochondrial dysfunction plays a vital role in MASLD and is associated with multiple pathological changes, including hepatocyte steatosis, oxidative stress, inflammation, and fibrosis. Metabolites are crucial mediators of the gut microbiota that influence extraintestinal organs. Additionally, regulation of the composition of gut bacteria may serve as a promising therapeutic strategy for MASLD. This study reviewed the potential roles of several common metabolites in MASLD, emphasizing their impact on mitochondrial function. Finally, we discuss the current treatments for MASLD, including probiotics, prebiotics, antibiotics, and fecal microbiota transplantation. These methods concentrate on restoring the gut microbiota to promote host health.


Sujet(s)
Transplantation de microbiote fécal , Microbiome gastro-intestinal , Mitochondries , Humains , Microbiome gastro-intestinal/physiologie , Mitochondries/métabolisme , Probiotiques/usage thérapeutique , Stéatose hépatique/métabolisme , Stéatose hépatique/microbiologie , Stéatose hépatique/thérapie , Prébiotiques , Antibactériens/usage thérapeutique , Animaux , Stress oxydatif
4.
JAMA Netw Open ; 7(7): e2418129, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967929

RÉSUMÉ

Importance: Probiotics are often considered in children to prevent antibiotic-associated diarrhea. However, the underlying mechanistic effects and impact of probiotics on antibiotic-induced microbiota changes are not well understood. Objective: To investigate the effects of a multispecies probiotic on the gut microbiota composition in children receiving antibiotics. Design, Setting, and Participants: This is a secondary analysis of a randomized, quadruple-blind, placebo-controlled clinical trial from February 1, 2018, to May 31, 2021, including 350 children receiving broad-spectrum antibiotics in the inpatient and outpatient settings. Patients were followed up until 1 month after the intervention period. Fecal samples and data were analyzed between September 1, 2022, and February 28, 2023. Eligibility criteria included 3 months to 18 years of age and recruitment within 24 hours following initiation of broad-spectrum systemic antibiotics. In total, 646 eligible patients were approached and 350 participated in the trial. Intervention: Participants were randomly assigned to receive daily placebo or a multispecies probiotic formulation consisting of 8 strains from 5 different genera during antibiotic treatment and for 7 days afterward. Main Outcomes and Measures: Fecal stool samples were collected at 4 predefined times: (1) inclusion, (2) last day of antibiotic use, (3) last day of the study intervention, and (4) 1 month after intervention. Microbiota analysis was performed by 16S ribosomal RNA gene sequencing. Results: A total of 350 children were randomized and collected stool samples from 88 were eligible for the microbiota analysis (54 boys and 34 girls; mean [SD] age, 47.09 [55.64] months). Alpha diversity did not significantly differ between groups at the first 3 times. Shannon diversity (mean [SD], 3.56 [0.75] vs 3.09 [1.00]; P = .02) and inverse Simpson diversity (mean [SD], 3.75 [95% CI, 1.66-5.82] vs -1.31 [95% CI, -3.17 to 0.53]; P = 1 × 10-4) indices were higher in the placebo group compared with the probiotic group 1 month after intervention. Beta diversity was not significantly different at any of the times. Three of 5 supplemented genera had higher relative abundance during probiotic supplementation, but this difference had disappeared after 1 month. Conclusions and Relevance: The studied probiotic mixture had minor and transient effects on the microbiota composition during and after antibiotic treatment. Further research is needed to understand their working mechanisms in manipulating the microbiome and preventing antibiotic-associated dysbiosis and adverse effects such as antibiotic-associated diarrhea. Trial Registration: ClinicalTrials.gov Identifier: NCT03334604.


Sujet(s)
Antibactériens , Diarrhée , Fèces , Microbiome gastro-intestinal , Probiotiques , Humains , Probiotiques/usage thérapeutique , Femelle , Mâle , Antibactériens/usage thérapeutique , Antibactériens/effets indésirables , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Enfant , Enfant d'âge préscolaire , Fèces/microbiologie , Diarrhée/induit chimiquement , Diarrhée/prévention et contrôle , Diarrhée/traitement médicamenteux , Diarrhée/microbiologie , Adolescent , Nourrisson
5.
Sci Rep ; 14(1): 15471, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969697

RÉSUMÉ

This study examines the effect of phycoerythrin (PE) from a cyanobacterial Nostoc strain encapsulated with alginate as a potential prebiotic to produce synbiotic ice cream products with Lactobacillus casei. It was found that the addition of the encapsulated PE affected, mostly favourably, the physicochemical properties, antioxidant activity, probiotic survival, volatile compound contents, and sensory acceptability of the synbiotic ice cream samples before and after aging at the freezing periods of one day to eight weeks. Thus, it confirms the prebiotic potential of PE for synbiotic ice creams with L. casei.


Sujet(s)
Alginates , Crème glacée , Lacticaseibacillus casei , Phycoérythrine , Synbiotiques , Lacticaseibacillus casei/métabolisme , Crème glacée/microbiologie , Alginates/composition chimique , Phycoérythrine/composition chimique , Synbiotiques/administration et posologie , Antioxydants/composition chimique , Nostoc/métabolisme , Probiotiques
6.
Trials ; 25(1): 458, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970042

RÉSUMÉ

Despite progress in reducing the infant mortality in India, the neonatal mortality decline has been slower, necessitating concerted efforts to achieve Sustainable Development Goal-3. A promising strategy aiming to prevent neonatal sepsis in high-risk, vulnerable, low birth weight neonates through an innovative intervention includes probiotic supplementation. This article communicates the decision by the ProSPoNS trial investigators to establish a Central Endpoint Adjudication Committee (CEAC) as an addendum to the protocol published in Trials in 2021 for the purpose of clarifying the primary outcome. In the published protocol, the study hypothesis and primary objective are based on "sepsis," the primary outcome has been specified as sepsis/PSBI, whereas the sample size estimation was performed based on the "physician diagnosed sepsis." To align all the three above, the investigators meeting, held on 17th-18th August 2023, at MGIMS Sevagram, Wardha, deliberated and unanimously agreed that "physician diagnosed sepsis" is the primary study outcome which includes sepsis/PSBI. The CEAC, chaired by an external subject expert and members including trial statistician, a microbiologist, and all site principal investigators will employ four criteria to determine "physician diagnosed sepsis": (1) blood culture status, (2) sepsis screen status, (3) PSBI/non-PSBI signs and symptoms, and (4) the clinical course for each sickness event. Importantly, this clarification maintains consistency with the approved study protocol (Protocol No. 5/7/915/2012 version 3.1 dated 14 Feb 2020), emphasizing the commitment to methodological transparency and adherence to predefined standards. The decision to utilize the guidance of a CEAC is recommended as the gold standard in multicentric complex clinical trials to achieve consistency and accuracy in assessment of outcomes.Trial registrationClinical Trial Registry of India (CTRI) CTRI/2019/05/019197. Registered on 16 May 2019.


Sujet(s)
Sepsis néonatal , Humains , Nouveau-né , Sepsis néonatal/diagnostic , Sepsis néonatal/traitement médicamenteux , Essais contrôlés randomisés comme sujet , Détermination du point final/normes , Inde , Probiotiques/usage thérapeutique , Probiotiques/effets indésirables , Résultat thérapeutique , Mortalité infantile , Plan de recherche , Taille de l'échantillon
7.
Allergol Immunopathol (Madr) ; 52(4): 84-90, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970270

RÉSUMÉ

PURPOSE: To investigate the effect of metformin on gut microbiota imbalance in patients with type 2 diabetes mellitus (T2DM), and the value of probiotic supplementation. METHODS: A total of 84 newly diagnosed T2DM patients were randomly divided into probiotics group, metformin group, and control group, with 28 patients in each group. The blood glucose control, islet function, gut microbiota, and inflammatory factors were compared between three groups. RESULTS: After 3 months of treatment, fasting plasma glucose (FPG), 2-h postprandial plasma glucose (2-h PG), and glycosylated hemoglobin A1c (HbA1c) were evidently decreased in both probiotics and metformin groups (P < 0.05) and were lower than that in the control group prior to treatment. Besides, FPG, 2-h PG, and HbA1c were lower in the metformin group than that in the control group. FPG, 2-h PG, and HbA1c were further lower in the probiotic group than in the metformin group (P < 0.05). Fasting insulin (FINS) and islet ß cell (HOMA-ß) -function were dramatically increased in the same group (P < 0.05), while insulin-resistant islet ß cells (HOMA-IR) were significantly lower in the same group (P < 0.05); FINS and HOMA-ß were significantly higher, while HOMA-IR was significantly lower (P < 0.05) in both groups than in the control group prior to treatment. HOMA-IR was also lower in the probiotic group than in the metformin group after treatment (P < 0.05); the number of lactobacilli and bifidobacteria increased (P < 0.05) in both probiotic and metformin groups than in the control group prior to treatment, and the number of Enterobacteriaceae and Enterococcus was lower in the control group prior to treatment (P < 0.05). In addition, the number of lactobacilli and bifidobacteria was higher and the number of enterobacteria and enterococci was lower in the probiotic group than that in the metformin group after treatment, and the differences were statistically significant (P < 0.05). Lipopolysaccharide (LPS), interleukin 6 (IL-6), and C-reactive protein (CRP) levels were lower in both probiotic and metformin groups (P < 0.05). The serum LPS, IL-6, and CRP levels were lower in both probiotic and metformin groups, compared to the control group prior to the treatment (P < 0.05). CONCLUSION: Metformin while treating T2DM assists in improving the imbalance of gut microbiota.


Sujet(s)
Glycémie , Diabète de type 2 , Microbiome gastro-intestinal , Hémoglobine glyquée , Hypoglycémiants , Metformine , Probiotiques , Humains , Metformine/pharmacologie , Metformine/administration et posologie , Probiotiques/administration et posologie , Diabète de type 2/traitement médicamenteux , Diabète de type 2/immunologie , Diabète de type 2/thérapie , Diabète de type 2/microbiologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/immunologie , Mâle , Femelle , Adulte d'âge moyen , Hypoglycémiants/pharmacologie , Hypoglycémiants/administration et posologie , Hypoglycémiants/usage thérapeutique , Hémoglobine glyquée/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Adulte , Compléments alimentaires , Insuline/sang , Sujet âgé
8.
Front Public Health ; 12: 1342313, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962766

RÉSUMÉ

Background: Studies have shown that gut dysbiosis contributes to the pathophysiology of type 2 diabetes mellitus (T2DM). Identifying specific gut microbiota dysbiosis may provide insight into the pathogenesis of T2DM. Purpose: This study investigated the causal relationship between gut microbiota and T2DM using meta-analysis and Mendelian randomization (MR). Methods: In the first part, we searched for literature on gut microbiota and T2DM, and conducted a meta-analysis. We observed differences in glycosylated hemoglobin and fasting blood glucose levels in both groups. Second, we obtained GWAS data from genome-wide association study database 19 (GWAS). We used two-sample MR analysis to verify the forward and reverse causal associations between gut microbiota and T2DM. Additionally, we selected the European GWAS data from the European Bioinformatics Institute (EBI) as a validation set for external validation of the MR analysis. In the third part, we aimed to clarify which gut microbiota contribute to the degree of causal association between group disorders and T2DM through multivariate MR analysis and Bayesian model averaging (MR-BMA). Results: 1. According to the meta-analysis results, the glycated hemoglobin concentration in the gut probiotic intervention group was significantly lower than in the control group. Following treatment, fasting blood glucose levels in the intervention group were significantly lower than those in the control group. 2. The results of two samples MR analysis revealed that there were causal relationships between six gut microbiota and T2DM. Genus Haemophilus and order Pasteurellaceae were negatively correlated with T2DM. Genus Actinomycetes, class Melanobacteria and genus Lactobacillus were positively correlated. Reverse MR analysis demonstrated that T2DM and gut microbiota did not have any reverse causal relationship. The external validation data set showed a causal relationship between gut microbiota and T2DM. 3. Multivariate MR analysis and MR-BMA results showed that the independent genus Haemophilus collection had the largest PP. Conclusion: Our research results suggest that gut microbiota is closely related to T2DM pathogenesis. The results of further MR research and an analysis of the prediction model indicate that a variety of gut microbiota disorders, including genus Haemophilus, are causally related to the development of T2DM. The findings of this study may provide some insight into the diagnosis and treatment of T2DM. Systematic review registration: https://www.crd.york.ac.uk/PROSPERO.


Sujet(s)
Diabète de type 2 , Microbiome gastro-intestinal , Étude d'association pangénomique , Analyse de randomisation mendélienne , Diabète de type 2/microbiologie , Humains , Dysbiose , Glycémie/analyse , Hémoglobine glyquée/analyse , Probiotiques
9.
Sci Rep ; 14(1): 15460, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965287

RÉSUMÉ

The gut microbiota plays a crucial role in neural development and progression of neural disorders like Parkinson's disease (PD). Probiotics have been suggested to impact neurodegenerative diseases via gut-brain axis. This study aims to investigate the therapeutic potential of Lacticaseibacillus rhamnosus E9, a high exopolysaccharide producer, on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP)-induced mouse model of PD. C57BL/6 mice subjected to MPTP were fed L. rhamnosus E9 for fifteen days and sacrificed after the last administration. Motor functions were determined by open-field, catalepsy, and wire-hanging tests. The ileum and the brain tissues were collected for ELISA, qPCR, and immunohistochemistry analyses. The cecum content was obtained for microbiota analysis. E9 supplementation alleviated MPTP-induced motor dysfunctions accompanied by decreased levels of striatal TH and dopamine. E9 also reduced the level of ROS in the striatum and decreased the DAT expression while increasing the DR1. Furthermore, E9 improved intestinal integrity by enhancing ZO-1 and Occludin levels and reversed the dysbiosis of the gut microbiota induced by MPTP. In conclusion, E9 supplementation improved the MPTP-induced motor deficits and neural damage as well as intestinal barrier by modulating the gut microbiota in PD mice. These findings suggest that E9 supplementation holds therapeutic potential in managing PD through the gut-brain axis.


Sujet(s)
1-Méthyl-4-phényl-1,2,3,6-tétrahydropyridine , Modèles animaux de maladie humaine , Microbiome gastro-intestinal , Lacticaseibacillus rhamnosus , Souris de lignée C57BL , Probiotiques , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Lacticaseibacillus rhamnosus/physiologie , Mâle , Probiotiques/pharmacologie , Probiotiques/administration et posologie , Maladie de Parkinson/traitement médicamenteux , Maladie de Parkinson/métabolisme , Maladie de Parkinson/microbiologie , Corps strié/métabolisme , Intoxication au MPTP/microbiologie , Intoxication au MPTP/métabolisme , Intoxication au MPTP/traitement médicamenteux , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Dopamine/métabolisme
10.
Sci Rep ; 14(1): 15466, 2024 07 05.
Article de Anglais | MEDLINE | ID: mdl-38965336

RÉSUMÉ

This study aimed to evaluate the efficacy of Lactiplantibacillus argentoratensis AGMB00912 (LA) in reducing Salmonella Typhimurium infection in weaned piglets. The investigation focused on the influence of LA on the gut microbiota composition, growth performance, and Salmonella fecal shedding. The results indicated that LA supplementation significantly improved average daily gain and reduced the prevalence and severity of diarrhea. Fecal analysis revealed reduced Salmonella shedding in the LA-supplemented group. Furthermore, LA notably altered the composition of the gut microbiota, increasing the levels of beneficial Bacillus and decreasing those of harmful Proteobacteria and Spirochaetes. Histopathological examination showed less intestinal damage in LA-treated piglets than in the controls. The study also observed that LA affected metabolic functions related to carbohydrate, amino acid, and fatty acid metabolism, thereby enhancing gut health and resilience against infection. Short-chain fatty acid concentrations in the feces were higher in the LA group, suggesting improved gut microbial activity. LA supplementation enriched the population of beneficial bacteria, including Streptococcus, Clostridium, and Bifidobacterium, while reducing the number of harmful bacteria, such as Escherichia and Campylobacter. These findings indicate the potential of LA as a probiotic alternative for swine nutrition, offering protective effects to the gut microbiota against Salmonella infection.


Sujet(s)
Fèces , Microbiome gastro-intestinal , Probiotiques , Sevrage , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Suidae , Projets pilotes , Probiotiques/administration et posologie , Fèces/microbiologie , Salmonelloses animales/microbiologie , Maladies des porcs/microbiologie , Maladies des porcs/prévention et contrôle , Lactobacillaceae , Salmonella typhimurium/effets des médicaments et des substances chimiques
11.
Sci Rep ; 14(1): 15387, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965339

RÉSUMÉ

Probiotics offer a promising prophylactic approach against various pathogens and represent an alternative strategy to combat biofilm-related infections. In this study, we isolated vaginal commensal microbiota from 54 healthy Indian women to investigate their probiotic traits. We primarily explored the ability of cell-free supernatant (CFS) from Lactobacilli to prevent Uropathogenic Escherichia coli (UPEC) colonization and biofilm formation. Our findings revealed that CFS effectively reduced UPEC's swimming and swarming motility, decreased cell surface hydrophobicity, and hindered matrix production by downregulating specific genes (fimA, fimH, papG, and csgA). Subsequent GC-MS analysis identified Tryptamine, a monoamine compound, as the potent bioactive substance from Lactobacilli CFS, inhibiting UPEC biofilms with an MBIC of 4 µg/ml and an MBEC of 8 µg/ml. Tryptamine induced significant changes in E. coli colony biofilm morphology, transitioning from the Red, Dry, and Rough (RDAR) to the Smooth and White phenotype, indicating reduced extracellular matrix production. Biofilm time-kill assays demonstrated a four-log reduction in UPEC viability when treated with Tryptamine, highlighting its potent antibacterial properties, comparable to CFS treatment. Biofilm ROS assays indicated a significant elevation in ROS generation within UPEC biofilms, suggesting a potential antibacterial mechanism. Gene expression studies with Tryptamine-treated samples showed a reduction in expression of curli gene (csgA), consistent with CFS treatment. This study underscores the potential of Tryptamine from probiotic Lactobacilli CFS as a promising antibiofilm agent against UPEC biofilms.


Sujet(s)
Biofilms , Lactobacillus , Probiotiques , Tryptamines , Escherichia coli uropathogène , Vagin , Biofilms/effets des médicaments et des substances chimiques , Biofilms/croissance et développement , Humains , Tryptamines/pharmacologie , Femelle , Escherichia coli uropathogène/effets des médicaments et des substances chimiques , Escherichia coli uropathogène/physiologie , Probiotiques/pharmacologie , Vagin/microbiologie , Lactobacillus/effets des médicaments et des substances chimiques , Lactobacillus/métabolisme , Lactobacillus/physiologie , Infections à Escherichia coli/microbiologie , Infections à Escherichia coli/traitement médicamenteux , Infections à Escherichia coli/prévention et contrôle , Adulte , Antibactériens/pharmacologie
12.
BMC Microbiol ; 24(1): 233, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951788

RÉSUMÉ

BACKGROUND: Inflammatory Bowel Diseases (IBD) are a major public health issue with unclear aetiology. Changes in the composition and functionality of the intestinal microbiota are associated with these pathologies, including the depletion of strict anaerobes such as Feacalibacterium prausnitzii. Less evidence is observed for depletion in other anaerobes, among which bifidobacteria. This study characterized the taxonomic and functional diversity of bifidobacteria isolated from the human intestinal microbiota in active and non-active IBD patients by a culturomics approach and evaluated if these bifidobacteria might be used as probiotics for gut health. RESULTS: A total of 341 bifidobacteria were isolated from the intestinal microbiota of IBD patients (52 Crohn's disease and 26 ulcerative colitis patients), with a high proportion of Bifidobacterium dentium strains (28% of isolated bifidobacteria). In ulcerative colitis, the major species identified was B. dentium (39% of isolated bifidobacteria), in active and non-active ulcerative colitis. In Crohn's disease, B. adolescentis was the major species isolated from non-active patients (40%), while similar amounts of B. dentium and B. adolescentis were found in active Crohn's disease patients. The relative abundance of B. dentium was increased with age, both in Crohn's disease and ulcerative colitis and active and non-active IBD patients. Antibacterial capacities of bifidobacteria isolated from non-active ulcerative colitis against Escherichia coli LF82 and Salmonella enterica ATCC 14028 were observed more often compared to strains isolated from active ulcerative colitis. Finally, B. longum were retained as strains with the highest probiotic potential as they were the major strains presenting exopolysaccharide synthesis, antibacterial activity, and anti-inflammatory capacities. Antimicrobial activity and EPS synthesis were further correlated to the presence of antimicrobial and EPS gene clusters by in silico analysis. CONCLUSIONS: Different bifidobacterial taxonomic profiles were identified in the microbiota of IBD patients. The most abundant species were B. dentium, mainly associated to the microbiota of ulcerative colitis patients and B. adolescentis, in the intestinal microbiota of Crohn's disease patients. Additionally, the relative abundance of B. dentium significantly increased with age. Furthermore, this study evidenced that bifidobacteria with probiotic potential (antipathogenic activity, exopolysaccharide production and anti-inflammatory activity), especially B. longum strains, can be isolated from the intestinal microbiota of both active and non-active Crohn's disease and ulcerative colitis patients.


Sujet(s)
Bifidobacterium , Microbiome gastro-intestinal , Probiotiques , Humains , Bifidobacterium/isolement et purification , Bifidobacterium/classification , Bifidobacterium/génétique , Adulte , Femelle , Mâle , Adulte d'âge moyen , Maladies inflammatoires intestinales/microbiologie , Jeune adulte , Sujet âgé , Rectocolite hémorragique/microbiologie , Maladie de Crohn/microbiologie , Phylogenèse , Fèces/microbiologie , ARN ribosomique 16S/génétique , Phénotype , Adolescent , Antibactériens/pharmacologie
13.
Front Immunol ; 15: 1389920, 2024.
Article de Anglais | MEDLINE | ID: mdl-38957464

RÉSUMÉ

Probiotic consumption strongly influences local intestinal immunity and systemic immune status. Heyndrickxia coagulans strain SANK70258 (HC) is a spore-forming lactic acid bacterium that has immunostimulatory properties on peripheral tissues. However, few reports have examined the detailed effectiveness of HC on human immune function and its mechanism of action. Therefore, we conducted a randomized, double-blind, placebo-controlled, parallel-group study to comprehensively evaluate the effects of HC on immunostimulatory capacity, upper respiratory tract infection (URTI) symptoms, and changes in intestinal organic-acid composition. Results of a questionnaire survey of URTI symptoms showed that runny nose, nasal congestion, sneezing, and sore throat scores as well as the cumulative number of days of these symptoms were significantly lower in the HC group than in the placebo group during the study period. Furthermore, the salivary secretory immunoglobulin A (sIgA) concentration was significantly higher, and the natural killer (NK) cell activity tended to be higher in the HC group than in the placebo group. In addition, we performed an exposure culture assay of inactivated influenza virus on peripheral blood mononuclear cells (PBMCs) isolated from the blood of participants in the HC and placebo groups. Gene-expression analysis in PBMCs after culture completion showed that IFNα and TLR7 expression levels were significantly higher in the HC group than in the placebo group. In addition, the expression levels of CD304 tended to be higher in the HC group than in the placebo group. On the other hand, the HC group showed a significantly higher increase in the intestinal butyrate concentration than the placebo group. HC intake also significantly suppressed levels of IL-6 and TNFα produced by PBMCs after exposure to inactivated influenza virus. Collectively, these results suggest that HC activated plasmacytoid dendritic cells expressing TLR7 and CD304 and strongly induced IFNα production, subsequently activating NK cells and increasing sIgA levels, and induced anti-inflammatory effects via increased intestinal butyrate levels. These changes may contribute to the acquisition of host resistance to viral infection and URTI prevention.


Sujet(s)
Probiotiques , Infections de l'appareil respiratoire , Humains , Infections de l'appareil respiratoire/immunologie , Méthode en double aveugle , Mâle , Adulte , Probiotiques/administration et posologie , Femelle , Jeune adulte , Agranulocytes/immunologie , Agranulocytes/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Microbiome gastro-intestinal/immunologie , Immunoglobuline A sécrétoire/immunologie , Récepteur de type Toll-7/métabolisme , Récepteur de type Toll-7/immunologie , Immunomodulation
14.
FASEB J ; 38(13): e23791, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38963340

RÉSUMÉ

Inflammatory bowel disease (IBD) is a kind of recurrent inflammatory disorder of the intestinal tract. The purpose of this study was to investigate the effects of Weissella paramesenteroides NRIC1542 on colitis in mice. A colitis model was induced by adding 1.5% DSS to sterile distilled water for seven consecutive days. During this process, mice were administered different concentrations of W. paramesenteroides NRIC1542. Colitis was assessed by DAI, colon length and hematoxylin-eosin staining of colon sections. The expressions of NF-κB signaling proteins and the tight junction proteins ZO-1 and occludin were detected by western blotting, and the gut microbiota was analyzed by 16S rDNA. The results showed that W. paramesenteroides NRIC1542 significantly reduced the degree of pathological tissue damage and the levels of TNF-α and IL-1ß in colonic tissue, inhibiting the NF-κB signaling pathway and increasing the expression of SIRT1, ZO-1 and occludin. In addition, W. paramesenteroides NRIC1542 can modulate the structure of the gut microbiota, characterized by increased relative abundance of Muribaculaceae_unclassified, Paraprevotella, Prevotellaceae_UCG_001 and Roseburia, and decrease the relative abundance of Akkermansia and Alloprevotella induced by DSS. The above results suggested that W. paramesenteroides NRIC1542 can protect against DSS-induced colitis in mice through anti-inflammatory, intestinal barrier maintenance and flora modulation.


Sujet(s)
Colite , Sulfate dextran , Microbiome gastro-intestinal , Facteur de transcription NF-kappa B , Transduction du signal , Sirtuine-1 , Weissella , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Sirtuine-1/métabolisme , Souris , Colite/induit chimiquement , Colite/métabolisme , Colite/microbiologie , Sulfate dextran/toxicité , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Weissella/métabolisme , Mâle , Probiotiques/pharmacologie
15.
PLoS One ; 19(7): e0297351, 2024.
Article de Anglais | MEDLINE | ID: mdl-38954692

RÉSUMÉ

OBJECTIVE: This study aimed to evaluate the effect of antimicrobial photodynamic therapy (aPDT) and the use of probiotics on the treatment of halitosis. METHODS: Fifty-two participants, aged from 18 to 25 years, exhaling sulfhydride (H2S) ≥ 112 ppb were selected. They were allocated into 4 groups (n = 13): Group 1: tongue scraper; Group 2: treated once with aPDT; Group 3: probiotic capsule containing Lactobacillus salivarius WB21 (6.7 x 108 CFU) and xylitol (280mg), 3 times a day after meals, for 14 days; Group 4: treated once with aPDT and with the probiotic capsule for 14 days. Halimetry with gas chromatography (clinical evaluation) and microbiological samples were collected from the dorsum of the tongue before and after aPDT, as well as after 7, 14, and 30 days. The clinical data failed to follow a normal distribution; therefore, comparisons were made using the Kruskal-Wallis test (independent measures) and Friedman ANOVA (dependent measures) followed by appropriate posthoc tests, when necessary. For the microbiological data, seeing as the data failed to follow a normal distribution, the Kruskal-Wallis rank sum test was performed with Dunn's post-test. The significance level was α = 0.05. RESULTS: Clinical results (halimetry) showed an immediate significant reduction in halitosis with aPDT (p = 0.0008) and/or tongue scraper (p = 0.0006). Probiotics showed no difference in relation to the initial levels (p = 0.7530). No significant differences were found in the control appointments. The amount of Porphyromonas gingivalis, Tannerella forsythia, and Treponema denticola were not altered throughout the analysis (p = 0.1616, p = 0.2829 and p = 0.2882, respectively). CONCLUSION: There was an immediate clinical reduction of halitosis with aPDT and tongue scraping, but there was no reduction in the number of bacteria throughout the study, or differences in the control times, both in the clinical and microbiological results. New clinical trials are necessary to better assess the tested therapies. TRIAL REGISTRATION: Clinical Trials NCT03996044.


Sujet(s)
Halitose , Ligilactobacillus salivarius , Photothérapie dynamique , Probiotiques , Humains , Halitose/microbiologie , Halitose/traitement médicamenteux , Halitose/thérapie , Probiotiques/usage thérapeutique , Probiotiques/administration et posologie , Adulte , Photothérapie dynamique/méthodes , Mâle , Femelle , Adolescent , Jeune adulte , Langue/microbiologie , Anti-infectieux/usage thérapeutique
16.
Benef Microbes ; 15(4): 411-415, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38960392

RÉSUMÉ

Clinical guidelines are recommendations for healthcare providers regarding patients with specific conditions. These guidelines should be based on practice experience and the best available research evidence. However, guidelines developed by various health organisations worldwide often do not agree with each other. This is also true for the current guidelines for the clinical use of probiotics. This article aims to provide examples of conflicting clinical guidelines for probiotics, define reasons for this phenomenon, describe standard tools for improving their quality, and suggest ways to enhance the development and assessment of suitable clinical guidelines for the appropriate clinical use of probiotics in specific conditions.


Sujet(s)
Guides de bonnes pratiques cliniques comme sujet , Probiotiques , Probiotiques/administration et posologie , Probiotiques/usage thérapeutique , Probiotiques/normes , Humains
17.
Benef Microbes ; 15(4): 387-396, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38955352

RÉSUMÉ

Group B Streptococcus (GBS) is the leading cause of bacterial neonatal sepsis. This study aimed to confirm the effect of Ligilactobacillus salivarius V4II-90 on GBS colonisation during pregnancy. A randomised, multicentre, double-blind, placebo-controlled, parallel-group study was conducted in seven hospitals in Madrid, Spain. The sample was broken down into two groups with 20 participants each (n = 40) in order to show reduced GBS colonisation frequency in the probiotic versus the placebo group. Pregnant participants positive for vaginal-rectal colonisation before or during the 13th week of gestation were randomly assigned to either the placebo or the probiotic group. The probiotic, L. salivarius V4II-90 at 1 × 109 cfu/day was administered for 12 weeks, starting at week 21-23 of gestation. The primary outcome was the percentage of participants with vaginal and/or rectal GBS colonisation at the end of the intervention period (35 weeks of gestation). Secondary outcomes were changes in the microbial composition of vaginal and rectal exudates; premature delivery; premature rupture of membranes; intrapartum antibiotics; new-borns with early or late-onset GBS sepsis; adverse events (AEs); and GBS test results performed at the hospital at week 35 of gestation. Of the 481 participants included, 44 were vaginal-rectal colonised with GBS and randomised. 43 completed the study (20 in the probiotic group and 23 in the placebo group). After intervention, GBS was eradicated in six participants (27%) from the placebo group and in twelve participants (63%) from the probiotic group ( P = 0.030). None of the 185 AEs reported were identified as possibly, probably, or definitely related to the investigational product. In conclusion, oral administration of L. salivarius V4II-90 is a safe and successful strategy to significantly decrease the rates of GBS colonisation at the end of pregnancy and, therefore, to reduce the exposure of subjects and their infants to intrapartum antibiotic prophylaxis. Trial registered at ClinicalTrials.gov: number NCT03669094.


Sujet(s)
Ligilactobacillus salivarius , Complications infectieuses de la grossesse , Probiotiques , Rectum , Infections à streptocoques , Streptococcus agalactiae , Vagin , Humains , Femelle , Grossesse , Probiotiques/administration et posologie , Méthode en double aveugle , Streptococcus agalactiae/croissance et développement , Streptococcus agalactiae/effets des médicaments et des substances chimiques , Infections à streptocoques/prévention et contrôle , Infections à streptocoques/microbiologie , Infections à streptocoques/traitement médicamenteux , Adulte , Vagin/microbiologie , Rectum/microbiologie , Ligilactobacillus salivarius/physiologie , Complications infectieuses de la grossesse/microbiologie , Complications infectieuses de la grossesse/prévention et contrôle , Complications infectieuses de la grossesse/traitement médicamenteux , Nouveau-né , Espagne , Jeune adulte
18.
Med Microbiol Immunol ; 213(1): 14, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38977511

RÉSUMÉ

Mycobacterium tuberculosis, a lethal pathogen in human history, causes millions of deaths annually, which demands the development of new concepts of drugs. Considering this fact, earlier research has explored the anti-tuberculosis potential of a probiotic strain, Lactocaseibacillus rhamnosus PMC203, leading to a subsequent focus on the molecular mechanism involved in its effect, particularly on autophagy. In this current study, immunoblotting-based assay exhibited a remarkable expression of autophagy marker LC3-II in the PMC203 treated group compared to an untreated group. A remarkable degradation of p62 was also noticed within treated cells compared to control. Furthermore, the immunofluorescence-based assay showed significant fold change in fluorescence intensity for alexa-647-LC3 and alexa-488-LC3, whereas p62 was degraded noticeably. Moreover, lysosomal biogenesis generation was elevated significantly in terms of LAMP1 and acidic vesicular organelles. As a result, PMC203-induced autophagy played a vital role in reducing M. tuberculosis burden within the macrophages in treated groups compared to untreated group. A colony -forming unit assay also revealed a significant reduction in M. tuberculosis in the treated cells over time. Additionally, the candidate strain significantly upregulated the expression of autophagy induction and lysosomal biogenesis genes. Together, these results could enrich our current knowledge of probiotics-mediated autophagy in tuberculosis and suggest its implications for innovatively managing tuberculosis.


Sujet(s)
Autophagie , Lacticaseibacillus rhamnosus , Macrophages , Mycobacterium tuberculosis , Probiotiques , Mycobacterium tuberculosis/génétique , Lacticaseibacillus rhamnosus/physiologie , Lacticaseibacillus rhamnosus/métabolisme , Macrophages/microbiologie , Humains , Lysosomes/métabolisme , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique , Charge bactérienne , Tuberculose/microbiologie
19.
BMC Microbiol ; 24(1): 253, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982403

RÉSUMÉ

BACKGROUND: Gut microbes play an important role in the growth and health of neonatal piglets. Probiotics can promote the healthy growth of neonatal piglets by regulating their gut microbes. The study investigated the effects of spraying Lactiplantibacillus plantarum P-8 (L. plantarum P-8) fermentation broth on the growth performance and gut microbes of neonatal piglets. RESULTS: The animals were randomly divided into probiotics groups (109 neonatal piglets) and control groups (113 neonatal piglets). The probiotics group was sprayed with L. plantarum P-8 fermented liquid from 3 day before the expected date of the sow to the 7-day-old of piglets, while the control group was sprayed with equal dose of PBS. Average daily gain (ADG), immune and antioxidant status and metagenome sequencing were used to assess the changes in growth performance and gut microbiota of neonatal piglets. The results showed that L. plantarum P-8 treatment significantly improved the average daily gain (P < 0.05) of neonatal piglets. L. plantarum P-8 increased the activities of CAT and SOD but reduced the levels of IL-2 and IL-6, effectively regulating the antioxidant capacity and immunity in neonatal piglets. L. plantarum P-8 adjusted the overall structure of gut microflora improving gut homeostasis to a certain extent, and significantly increased the relative abundance of gut beneficial bacteria such as L. mucosae and L. plantarum. CONCLUSION: Spraying L. plantarum P-8 can be a feasible and effective probiotic intervention not only improving the growth of neonatal piglets, regulating the antioxidant capacity and immunity of neonatal piglets, but also improving the gut homeostasis to a certain extent.


Sujet(s)
Animaux nouveau-nés , Microbiome gastro-intestinal , Probiotiques , Animaux , Probiotiques/administration et posologie , Probiotiques/pharmacologie , Suidae , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Lactobacillus plantarum , Fermentation , Antioxydants/métabolisme , Antioxydants/administration et posologie , Antioxydants/pharmacologie , Fèces/microbiologie
20.
Benef Microbes ; 15(4): 357-371, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38955353

RÉSUMÉ

Constipation during pregnancy can induce serious complications, including miscarriage and preterm labour, while the evidence of probiotics in improving constipation during pregnancy was little. In this study, 29 healthy pregnant women and 65 constipated pregnant women were enrolled to assess the effectiveness of probiotics on constipation during pregnancy. Our results showed that the probiotics were effective in improving the Constipation Severity Scale (CSS) and Bristol Stool Scale (BSS) scores, including increasing defecation frequency, decreasing defecation time, and improving fecal characteristics. 16S rRNA sequencing revealed that the probiotics effectively restored the diversity of intestinal microbiota. At the phylum level, Firmicutes (13.27% vs 57.20%) and Actinobacteria (3.77% vs 12.80%) were increased, while Bacteroidetes (77.82% vs 20.24%) was decreased. At the level of the genus, Faecalibacterium (2.03% vs 10.33%), Bifidobacterium (1.21% vs 8.56%), and Phascolarctobacterium (0.05% vs 2.88%), the beneficial bacteria were increased, while the Bacteroides (29.23% vs 12.28%) and Prevotella (24.32% vs 4.92%) were decreased. In conclusion, these results indicated that probiotics can effectively relieve the constipation symptoms by improving the diversity of intestinal microbiota, regulating the disturbance of microflorae, and restoring the balance of microflorae to exert a stronger moderating effect than diet and lifestyle modification. Our results provided clinical data and a theoretical basis for the exploitation of probiotics in treating constipation during pregnancy. Chinese Clinical Trial Registry: ChiCTR2100052069.


Sujet(s)
Constipation , Fèces , Microbiome gastro-intestinal , Probiotiques , ARN ribosomique 16S , Constipation/thérapie , Constipation/microbiologie , Constipation/traitement médicamenteux , Humains , Femelle , Probiotiques/administration et posologie , Probiotiques/usage thérapeutique , Grossesse , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Adulte , Fèces/microbiologie , ARN ribosomique 16S/génétique , Bactéries/classification , Bactéries/génétique , Bactéries/effets des médicaments et des substances chimiques , Bactéries/isolement et purification , Complications de la grossesse/microbiologie , Complications de la grossesse/thérapie , Complications de la grossesse/traitement médicamenteux , Jeune adulte , Défécation/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE