Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.084
Filtrar
1.
PeerJ ; 12: e17502, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38952971

RESUMEN

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.


Asunto(s)
Cocos , Gelatina , Lacticaseibacillus rhamnosus , Probióticos , Cocos/química , Cocos/microbiología , Gelatina/química , Antioxidantes/farmacología , Antioxidantes/química , Fermentación
2.
BMC Microbiol ; 24(1): 233, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951788

RESUMEN

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.


Asunto(s)
Bifidobacterium , Microbioma Gastrointestinal , Probióticos , Humanos , Bifidobacterium/aislamiento & purificación , Bifidobacterium/clasificación , Bifidobacterium/genética , Adulto , Femenino , Masculino , Persona de Mediana Edad , Enfermedades Inflamatorias del Intestino/microbiología , Adulto Joven , Anciano , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/microbiología , Filogenia , Heces/microbiología , ARN Ribosómico 16S/genética , Fenotipo , Adolescente , Antibacterianos/farmacología
3.
Front Immunol ; 15: 1389920, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957464

RESUMEN

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.


Asunto(s)
Probióticos , Infecciones del Sistema Respiratorio , Humanos , Infecciones del Sistema Respiratorio/inmunología , Método Doble Ciego , Masculino , Adulto , Probióticos/administración & dosificación , Femenino , Adulto Joven , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A Secretora/inmunología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 7/inmunología , Inmunomodulación
4.
FASEB J ; 38(13): e23791, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38963340

RESUMEN

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.


Asunto(s)
Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , FN-kappa B , Transducción de Señal , Sirtuina 1 , Weissella , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Sirtuina 1/metabolismo , Ratones , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Sulfato de Dextran/toxicidad , Transducción de Señal/efectos de los fármacos , FN-kappa B/metabolismo , Weissella/metabolismo , Masculino , Probióticos/farmacología
5.
Front Public Health ; 12: 1342313, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38962766

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Diabetes Mellitus Tipo 2/microbiología , Humanos , Disbiosis , Glucemia/análisis , Hemoglobina Glucada/análisis , Probióticos
6.
Parasit Vectors ; 17(1): 284, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956725

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas , Microbioma Gastrointestinal , Ratones Noqueados , Toxoplasma , Animales , Ratones , Toxoplasma/inmunología , Lesiones Encefálicas/inmunología , Probióticos/administración & dosificación , Encéfalo/inmunología , Lactobacillus , Modelos Animales de Enfermedad , Huésped Inmunocomprometido , Toxoplasmosis/inmunología , ARN Ribosómico 16S/genética , Masculino , Intestinos/inmunología
7.
PLoS One ; 19(7): e0297351, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38954692

RESUMEN

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.


Asunto(s)
Halitosis , Ligilactobacillus salivarius , Fotoquimioterapia , Probióticos , Humanos , Halitosis/microbiología , Halitosis/tratamiento farmacológico , Halitosis/terapia , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Adulto , Fotoquimioterapia/métodos , Masculino , Femenino , Adolescente , Adulto Joven , Lengua/microbiología , Antiinfecciosos/uso terapéutico
8.
Hepatol Commun ; 8(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38967596

RESUMEN

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.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Mitocondrias , Humanos , Microbioma Gastrointestinal/fisiología , Mitocondrias/metabolismo , Probióticos/uso terapéutico , Hígado Graso/metabolismo , Hígado Graso/microbiología , Hígado Graso/terapia , Prebióticos , Antibacterianos/uso terapéutico , Animales , Estrés Oxidativo
9.
JAMA Netw Open ; 7(7): e2418129, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38967929

RESUMEN

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.


Asunto(s)
Antibacterianos , Diarrea , Heces , Microbioma Gastrointestinal , Probióticos , Humanos , Probióticos/uso terapéutico , Femenino , Masculino , Antibacterianos/uso terapéutico , Antibacterianos/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Niño , Preescolar , Heces/microbiología , Diarrea/inducido químicamente , Diarrea/prevención & control , Diarrea/tratamiento farmacológico , Diarrea/microbiología , Adolescente , Lactante
10.
Sci Rep ; 14(1): 15471, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969697

RESUMEN

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.


Asunto(s)
Alginatos , Helados , Lacticaseibacillus casei , Ficoeritrina , Simbióticos , Lacticaseibacillus casei/metabolismo , Helados/microbiología , Alginatos/química , Ficoeritrina/química , Simbióticos/administración & dosificación , Antioxidantes/química , Nostoc/metabolismo , Probióticos
11.
Trials ; 25(1): 458, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970042

RESUMEN

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.


Asunto(s)
Sepsis Neonatal , Humanos , Recién Nacido , Sepsis Neonatal/diagnóstico , Sepsis Neonatal/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Determinación de Punto Final/normas , India , Probióticos/uso terapéutico , Probióticos/efectos adversos , Resultado del Tratamiento , Mortalidad Infantil , Proyectos de Investigación , Tamaño de la Muestra
12.
Allergol Immunopathol (Madr) ; 52(4): 84-90, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38970270

RESUMEN

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.


Asunto(s)
Glucemia , Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Hemoglobina Glucada , Hipoglucemiantes , Metformina , Probióticos , Humanos , Metformina/farmacología , Metformina/administración & dosificación , Probióticos/administración & dosificación , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/terapia , Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Masculino , Femenino , Persona de Mediana Edad , Hipoglucemiantes/farmacología , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/uso terapéutico , Hemoglobina Glucada/metabolismo , Glucemia/efectos de los fármacos , Adulto , Suplementos Dietéticos , Insulina/sangre , Anciano
13.
Curr Microbiol ; 81(8): 258, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38960917

RESUMEN

Imidazole-chalcone compounds are recognised for their broad-spectrum antimicrobial properties. Probiotic-friendly, selective new-generation antimicrobials prove to be more efficient in combating gastrointestinal system pathogens. The aim of this study is to identify imidazole-chalcone derivatives that probiotics tolerate and evaluate their in vitro synergistic antimicrobial effects on pathogens. In this study, fifteen previously identified imidazole-chalcone derivatives were analyzed for their in vitro antimicrobial properties against gastrointestinal microorganisms. Initially, the antimicrobial activity of pathogens was measured using the agar well diffusion method, while the susceptibility of probiotics was determined by microdilution. The chosen imidazole-chalcone derivatives were assessed for synergistic effects using the checkerboard method. Four imidazole-chalcone derivatives to which probiotic bacteria were tolerant exhibited antibacterial and antifungal activity against the human pathogens tested. To our knowledge, this study is the first to reveal the fractional inhibitory concentration (FIC) of combinations of imidazole-chalcone derivatives. Indeed, the minimum inhibitory concentrations (MIC) for morpholinyl- (ZDO-3f) and 4-ethylpiperazinyl- (ZDO-3 m) imidazole-chalcones were notably low when tested against E. coli and B. subtilis, with values of 31.25 µg/mL and 125 µg/mL, respectively. The combination of morpholinyl- and 4-ethylpiperazinyl derivatives demonstrated an indifferent effect against E. coli, but an additive effect was observed for B. subtilis. Additionally, it was observed that imidazole-chalcone derivatives did not exhibit any inhibitory effects on probiotic organisms like Lactobacillus fermentum (CECT-5716), Lactobacillus rhamnosus (GG), and Lactobacillus casei (RSSK-591). This study demonstrates that imidazole-chalcone derivatives that are well tolerated by probiotics can potentially exert a synergistic effect against gastrointestinal system pathogens.


Asunto(s)
Sinergismo Farmacológico , Imidazoles , Pruebas de Sensibilidad Microbiana , Probióticos , Probióticos/farmacología , Imidazoles/farmacología , Imidazoles/química , Chalcona/farmacología , Chalcona/química , Chalcona/análogos & derivados , Antibacterianos/farmacología , Antibacterianos/química , Chalconas/farmacología , Chalconas/química , Tracto Gastrointestinal/microbiología , Humanos , Bacterias/efectos de los fármacos , Antifúngicos/farmacología , Antifúngicos/química
14.
Sci Rep ; 14(1): 15460, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965287

RESUMEN

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.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Ratones Endogámicos C57BL , Probióticos , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Lacticaseibacillus rhamnosus/fisiología , Masculino , Probióticos/farmacología , Probióticos/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/microbiología , Cuerpo Estriado/metabolismo , Intoxicación por MPTP/microbiología , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/tratamiento farmacológico , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/efectos de los fármacos , Dopamina/metabolismo
15.
Sci Rep ; 14(1): 15466, 2024 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-38965336

RESUMEN

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.


Asunto(s)
Heces , Microbioma Gastrointestinal , Probióticos , Destete , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Porcinos , Proyectos Piloto , Probióticos/administración & dosificación , Heces/microbiología , Salmonelosis Animal/microbiología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/prevención & control , Lactobacillaceae , Salmonella typhimurium/efectos de los fármacos
16.
Sci Rep ; 14(1): 15387, 2024 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965339

RESUMEN

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.


Asunto(s)
Biopelículas , Lactobacillus , Probióticos , Triptaminas , Escherichia coli Uropatógena , Vagina , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Humanos , Triptaminas/farmacología , Femenino , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/fisiología , Probióticos/farmacología , Vagina/microbiología , Lactobacillus/efectos de los fármacos , Lactobacillus/metabolismo , Lactobacillus/fisiología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/prevención & control , Adulto , Antibacterianos/farmacología
17.
Sci Rep ; 14(1): 15565, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38971851

RESUMEN

Lactic acid bacteria (LAB) isolated from medicinal herb Murraya koenigii, commonly known as curry leaf, which promotes the growth and maintenance of gut microbiota, were studied for their probiotic potential. The key objective of this research was to isolate and evaluate probiotic characteristics, test adherence capabilities, and confirm their safety. Lactococcus lactis (MKL8), isolated from Murraya koenigii, was subjected to in vitro analysis to assess its resistance to the gastric environment, ability to adhere Caco-2 cells, anti-microbial activity, hydrophobicity, auto-aggregation, and safety profiling through MTT assay and hemolytic. MKL8 exhibited growth at 0.5% phenol concentrations (> 80%) and was able to survive in conditions with high bile concentrations (> 79%) and a relatively low pH (72%-91%). It shows high tolerance to high osmotic conditions (> 73%) and simulated gastric juice (> 72%). Additionally, MKL8 demonstrated strong hydrophobicity (85%), auto-aggregation (87.3%-91.7%), and adherence to Caco-2 cells. Moreover, it had an inhibitory effect against pathogens too. By performing the hemolytic and MTT assays, the non-toxicity of MKL8 isolate was examined, and it exhibited no harmful characteristics. Considering MKL8's resistance to gastrointestinal tract conditions, high surface hydrophobicity, non-toxicity, and ability to inhibit the tested pathogens, it can be concluded that MKL8 demonstrated promising probiotic properties and has potential for use in the food industry.


Asunto(s)
Adhesión Bacteriana , Lactococcus lactis , Murraya , Probióticos , Humanos , Células CACO-2 , Lactococcus lactis/aislamiento & purificación , Adhesión Bacteriana/efectos de los fármacos , Murraya/química , Interacciones Hidrofóbicas e Hidrofílicas , Antibacterianos/farmacología
18.
Front Immunol ; 15: 1418594, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975337

RESUMEN

Introduction: Maternal synbiotic supplementation during pregnancy and lactation can significantly influence the immune system. Prebiotics and probiotics have a positive impact on the immune system by preventing or ameliorating among others intestinal disorders. This study focused on the immunomodulatory effects of B. breve M-16V and short chain galacto-oligosaccharides (scGOS)/long chain fructo-oligosachairdes (lcFOS), including systemic and mucosal compartments and milk composition. Methods: Lewis rats were orally administered with the synbiotic or vehicle during pregnancy (21 days) and lactation (21 days). At the weaning day, small intestine (SI), mammary gland (MG), adipose tissue, milk, mesenteric lymph nodes (MLN), salivary gland (SG), feces and cecal content were collected from the mothers. Results: The immunoglobulinome profile showed increased IgG2c in plasma and milk, as well as elevated sIgA in feces at weaning. The supplementation improved lipid metabolism through enhanced brown adipose tissue activity and reinforced the intestinal barrier by increasing the expression of Muc3, Cldn4, and Ocln. The higher production of short chain fatty acids in the cecum and increased Bifidobacterium counts suggest a potential positive impact on the gastrointestinal tract. Discussion: These findings indicate that maternal synbiotic supplementation during gestation and lactation improves their immunological status and improved milk composition.


Asunto(s)
Bifidobacterium breve , Lactancia , Leche , Oligosacáridos , Animales , Femenino , Embarazo , Bifidobacterium breve/inmunología , Leche/inmunología , Leche/química , Ratas , Ratas Endogámicas Lew , Suplementos Dietéticos , Simbióticos/administración & dosificación , Probióticos/administración & dosificación , Probióticos/farmacología
19.
ISME J ; 18(1)2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38896583

RESUMEN

Probiotics have gained significant attention as a potential strategy to improve health by modulating host-microbe interactions, particularly in situations where the normal microbiota has been disrupted. However, evidence regarding their efficacy has been inconsistent, with considerable interindividual variability in response. We aimed to explore whether a common genetic variant that affects the production of mucosal α(1,2)-fucosylated glycans, present in around 20% of the population, could explain the observed interpersonal differences in the persistence of commonly used probiotics. Using a mouse model with varying α(1,2)-fucosylated glycans secretion (Fut2WT or Fut2KO), we examined the abundance and persistence of Bifidobacterium strains (infantis, breve, and bifidum). We observed significant differences in baseline gut microbiota characteristics between Fut2WT and Fut2KO littermates, with Fut2WT mice exhibiting enrichment of species able to utilize α(1,2)-fucosylated glycans. Following antibiotic exposure, only Fut2WT animals showed persistent engraftment of Bifidobacterium infantis, a strain able to internalize α(1,2)-fucosylated glycans, whereas B. breve and B. bifidum, which cannot internalize α(1,2)-fucosylated glycans, did not exhibit this difference. In mice with an intact commensal microbiota, the relationship between secretor status and B. infantis persistence was reversed, with Fut2KO animals showing greater persistence compared to Fut2WT. Our findings suggest that the interplay between a common genetic variation and antibiotic exposure plays a crucial role in determining the dynamics of B. infantis in the recipient gut, which could potentially contribute to the observed variation in response to this commonly used probiotic species.


Asunto(s)
Antibacterianos , Fucosiltransferasas , Galactósido 2-alfa-L-Fucosiltransferasa , Microbioma Gastrointestinal , Probióticos , Animales , Ratones , Fucosiltransferasas/genética , Fucosiltransferasas/metabolismo , Probióticos/administración & dosificación , Antibacterianos/farmacología , Bifidobacterium longum subspecies infantis/genética , Bifidobacterium longum subspecies infantis/metabolismo , Polisacáridos/metabolismo , Interacciones Microbiota-Huesped , Ratones Endogámicos C57BL , Ratones Noqueados , Bifidobacterium/genética , Bifidobacterium/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...