Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.350
Filtrer
1.
Sci Rep ; 14(1): 21372, 2024 09 12.
Article de Anglais | MEDLINE | ID: mdl-39266591

RÉSUMÉ

This study was designed to investigate the effect of vitamin D and/or synbiotics on the response to treatment, cytokines profile and hormonal biomarkers in breast cancer patients undergoing neoadjuvant therapy. A total of 76 patients were recruited and completed the course of the intervention between 2019 and 2021 in Kerman, Iran. breast cancer patients were randomly enrolled in this study. Patients divided into four groups to receive one of the following regimens: placebo, vitamin D, synbiotics and a combination of vitamin D and synbiotics. clinicopathologic parameters, inflammatory and anti-inflammatory biomarkers and hormonal levels were measured at the baseline and four months after intervention. The study results found no clear link between the interventions and achieving pathological complete response (pCR), and a similar trend was observed in Ki-67 index examination. After neoadjuvant therapy, TNF-α concentrations decreased, with vitamin D supplementation moderating this decline. Vitamin D supplemented groups showed a significant increase in serum IL-6 levels. While IL-10 levels decreased in the placebo group, all intervention groups were protected from this decline. Moreover, there was a notable increase in the anti-inflammatory index, particularly in the group receiving both vitamin D and synbiotic supplementation, suggesting potential synergistic anti-inflammatory effects from their combined administration. The outcomes suggest a potential anti-inflammatory function of this combination. Consequently, more extensive studies with prolonged follow-up periods and substantial sample sizes are warranted to thoroughly evaluate their potential benefits for breast cancer patients.


Sujet(s)
Tumeurs du sein , Cytokines , Synbiotiques , Vitamine D , Humains , Tumeurs du sein/traitement médicamenteux , Femelle , Vitamine D/sang , Vitamine D/administration et posologie , Synbiotiques/administration et posologie , Adulte d'âge moyen , Projets pilotes , Cytokines/sang , Cytokines/métabolisme , Adulte , Traitement néoadjuvant/méthodes , Iran , Résultat thérapeutique , Synergie des médicaments , Compléments alimentaires
2.
BMC Microbiol ; 24(1): 331, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39245724

RÉSUMÉ

BACKGROUND: The quest for candidate probiotics and prebiotics to develop novel synbiotics for sustainable and profitable fish farming remains a major focus for various stakeholders. In this study, we examined the effects of combining two fungal probiotics, Saccharomyces cerevisiae and Aspergillus niger with extracts of Jerusalem artichoke and white button mushroom to develop a synbiotic formulation to improve the growth and health status of zebrafish (Danio rerio). An initial in vitro study determined the most effective synbiotic combination, which was then tested in a 60-day in vivo nutritional trial using zebrafish (80 ± 1.0 mg) as a model animal. Four experimental diets were prepared: a control diet (basal diet), a prebiotic diet with 100% selected mushroom extract, a probiotic diet with 107 CFU of S. cerevisiae/g of diet, and a synbiotic diet with 107 CFU of S. cerevisiae/g of diet and 100% mushroom extract. As readouts, growth performance, survival, digestive enzyme activity and innate immune responses were evaluated. RESULTS: In vitro results showed that the S. cerevisiae cultured in a medium containing 100% mushroom extract exhibited the maximum specific growth rate and shortest doubling time. In the in vivo test with zebrafish, feeding them with a synbiotic diet, developed with S. cerevisiae and mushroom extract, led to a significant improvement in the growth performance of zebrafish (P < 0.05). The group of zebrafish fed with the synbiotic diet showed significantly higher levels of digestive enzyme activity and immune responses compared to the control group (P < 0.05). CONCLUSION: Taken together, these results indicated that the combination of S. cerevisiae and mushroom extract forms an effective synbiotic, capable of enhancing growth performance and immune response in zebrafish.


Sujet(s)
Agaricales , Probiotiques , Saccharomyces cerevisiae , Synbiotiques , Danio zébré , Animaux , Danio zébré/croissance et développement , Danio zébré/immunologie , Synbiotiques/administration et posologie , Probiotiques/pharmacologie , Aliment pour animaux/analyse , Aspergillus niger/croissance et développement , Immunité innée/effets des médicaments et des substances chimiques , Prébiotiques
3.
Sci Rep ; 14(1): 20333, 2024 09 02.
Article de Anglais | MEDLINE | ID: mdl-39223205

RÉSUMÉ

Association between metabolic syndrome (MetS) and oxidative stress has been shown in numerous studies. It has been shown that probiotics could be the effective treatment strategy in improving oxidative stress. This study aimed to determine the effects of a new developed synbiotic yogurt on oxidative stress status in adults with MetS. Forty-four individuals were assigned into two groups and given 300 g of synbiotic yogurt containing Lactobacillus plantarum, Lactobacillus pentosus, and Chloromyces marcosianos yeast or regular yogurt for 12 weeks in this randomized, placebo-controlled clinical trial. Before and after the intervention, biochemical parameters were assessed. Daily consumption of synbiotic yogurt in adults with MetS showed a statistically significant improvement in the level of glutathione peroxidase (p = 0.01) and total oxidant status (p = 0.006) compared to the regular yogurt. Total Antioxidant Capacity and superoxide dismutase levels increased significantly (p = 0.002 and p = 0.02, respectively) in the intervention group compared to the baseline levels. In adults with MetS, daily consumption of the synbiotic yogurt containing native strains of Lactobacillus plantarum, Lactobacillus pentosus, and Chloromyces marcosianos yeast for 12 weeks was associated with improvements in oxidative stress status.Trial registration number: Iranian Registry of Clinical Trials (IRCT20220426054667N1) (18/05/2022).


Sujet(s)
Syndrome métabolique X , Stress oxydatif , Synbiotiques , Yaourt , Humains , Yaourt/microbiologie , Mâle , Femelle , Syndrome métabolique X/diétothérapie , Syndrome métabolique X/thérapie , Syndrome métabolique X/microbiologie , Adulte d'âge moyen , Adulte , Antioxydants/métabolisme , Lactobacillus plantarum , Probiotiques/usage thérapeutique , Probiotiques/administration et posologie , Superoxide dismutase/métabolisme , Glutathione peroxidase/métabolisme
4.
Food Res Int ; 195: 114976, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39277213

RÉSUMÉ

Changes in dietary patterns and living habits have led to an increasing number of individuals with elevated cholesterol levels. Excessive consumption of high-cholesterol foods can disrupt the body's lipid metabolism. Numerous studies have firmly established the cholesterol-lowering effects of probiotics and prebiotics, with evidence showing that the synergistic use of synbiotics is functionally more potent than using probiotics or prebiotics alone. Currently, the screening strategy involves screening prebiotics for synbiotic development with probiotics as the core. However, in comparison to probiotics, there are fewer types of prebiotics available, leading to limited resources. Consequently, the combinations of synbiotics obtained are restricted, and probiotics and prebiotics are only relatively suitable. Therefore, in this study, a novel synbiotic screening strategy with prebiotics as the core was developed. The synbiotic combination of Lactobacillus rhamnosus S_82 and xylo-oligosaccharides was screened from the intestinal tract of young people through five generations of xylo-oligosaccharides. Subsequently, the cholesterol-lowering ability of the medium was simulated, and the two carbon sources of glucose and xylo-oligosaccharides were screened out. The results showed that synbiotics may participate in cholesterol-lowering regulation by down-regulating the expression of NPC1L1 gene, down-regulating ACAT2 and increasing the expression of ABCG8 gene in vitro through cell adsorption and cell absorption in vitro, and regulating the intestinal microbiota. Synbiotics hold promise as potential candidates for the prevention of hypercholesterolemia in humans and animals, and this study providing a theoretical foundation for the development of new synbiotic products.


Sujet(s)
Lacticaseibacillus rhamnosus , Oligosaccharides , Prébiotiques , Synbiotiques , Lacticaseibacillus rhamnosus/métabolisme , Oligosaccharides/pharmacologie , Humains , Hypolipémiants/pharmacologie , Cholestérol/métabolisme , Cholestérol/sang , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Probiotiques , Glucuronates
5.
Food Funct ; 15(19): 9954-9971, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39264166

RÉSUMÉ

Background: Probiotics, prebiotics, and synbiotics have been suggested as a possible therapy for non-alcoholic fatty liver disease (NAFLD). However, their efficacy in improving blood glucose levels in NAFLD patients remains uncertain. Objective: The aim of this study was to assess the effects of supplementation with probiotics, prebiotics, or synbiotics on fasting blood glucose (FBG) levels in NAFLD patients. Methods: We searched PubMed, Web of Science, and Google Scholar for relevant trials published up to March 2024. Out of 3369 identified studies, 24 randomized controlled trials (RCTs) were included. Results: Probiotic, prebiotic, or synbiotic supplementation substantially reduced FBG (n = 23; standard mean difference (SMD) = -0.17; 95% confidence interval (CI): -0.30, -0.03; P = 0.02), fasting insulin levels (n = 12; SMD = -0.28; 95% CI: -0.49, -0.07; P = 0.01), and homeostatic model assessment for insulin resistance (HOMA-IR; n = 14; SMD = -0.28; 95% CI: -0.47, -0.09; P = 0.004). However, glycosylated hemoglobin (HbA1c; n = 3; SMD = -0.17; 95% CI: -0.48, 0.13; P = 0.27) was not significantly affected. The FBG-decreasing effect diminished as the body mass index (BMI) of volunteers increased in the baseline. Additionally, the number of probiotic strains and geographic region were shown to significantly affect FBG levels. Conclusion: This meta-analysis indicates that supplementation with probiotics, prebiotics, or synbiotics may aid in controlling glucose homeostasis in patients with NAFLD.


Sujet(s)
Glycémie , Stéatose hépatique non alcoolique , Prébiotiques , Probiotiques , Essais contrôlés randomisés comme sujet , Synbiotiques , Humains , Stéatose hépatique non alcoolique/diétothérapie , Stéatose hépatique non alcoolique/thérapie , Stéatose hépatique non alcoolique/métabolisme , Synbiotiques/administration et posologie , Probiotiques/usage thérapeutique , Probiotiques/administration et posologie , Glycémie/métabolisme , Homéostasie , Insulinorésistance , Femelle , Mâle , Adulte , Adulte d'âge moyen
6.
Clin Med Res ; 22(2): 97-106, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39231625

RÉSUMÉ

Background: Although various treatments are developed against COVID-19 variants, probiotic, and synbiotic adjunct therapy with several benefits such as safety, low cost, and availability could be needed for preventing or treating COVID-19 infection.Objective: The present systematic review aimed to assess the prophylactic efficacy of the probiotic, prebiotic, and synbiotic administration against COVID-19.Methods: The protocol of this systematic review was registered at the PROSPERO (Code number: CRD42023418900). The Scopus, Cochrane Library, Web of Sciences, and PubMed databases were systematically searched to define the clinical trials published up to November 2022 in the English language. The comparison of the incidence of COVID-19 disease and levels of specific antibodies against SARS-cov2 between the intervention and placebo groups were evaluated in this systematic review.Results: According to the five included trials, four indicated the incidence of COVID-19, and no significant differences were observed between the probiotic and placebo groups during 1, 2, or 3 months of follow-up in the mentioned studies. Regarding the antibody assays against SARS-Cov2 including IgM, IgG, or IgA reported by three eligible trials, there were no significant differences between the intervention and control groups.Conclusion: It seems that the administration of single or multi-strain probiotics or synbiotics had no prophylactic effects in different populations such as high-risk staff exposed to COVID-19, elderly nursing home residents, healthy adults, and household contact with COVID-19 patients during 1-to-3-months of intervention.


Sujet(s)
COVID-19 , Prébiotiques , Probiotiques , Synbiotiques , Humains , COVID-19/prévention et contrôle , Prébiotiques/administration et posologie , Probiotiques/usage thérapeutique , Essais contrôlés randomisés comme sujet , Synbiotiques/administration et posologie
7.
Medicine (Baltimore) ; 103(37): e39647, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39287261

RÉSUMÉ

BACKGROUND: Necrotizing enterocolitis (NEC) is a multifactorial disease primarily affecting infants with very low birth weight (VLBW). Research has shown that the pathogenesis of NEC is associated with abnormal bacterial colonization. Synbiotics, dietary supplements containing probiotics (beneficial bacteria) and prebiotics (non-digestible food), can alter the gut microbiome. METHODS: A double-blind, randomized controlled trial was conducted to assess the efficacy of PediLact®, an oral drop multi-strain synbiotic containing Lactobacillus rhamnosus, Lactobacillus reuteri, and Bifidobacterium infantis, on nutritional parameters and the occurrence of NEC in VLBW neonates. In this study, 118 VLBW neonates from neonatal intensive care units were randomly allocated in a 1:1 ratio to receive either a synbiotic or a placebo. The synbiotic administration continued until the infant was fully fed. The primary outcomes were the occurrence of NEC and the number of days required to achieve full enteral feeding. Log-binomial regression and ANOVA/ANCOVA models were used for analysis. RESULTS: In the group that received the synbiotic, the incidence of NEC decreased significantly (adjusted RR = 0.22, 95% CI: 0.07-0.72, P value = .01; adjusted RD = -0.22, 95% CI: -0.33 to -0.12, P value < .001; NNT = 5). Additionally, feeding intolerance was less frequent in this group (adjusted RR = 0.27, 95% CI: 0.14-0.51, P value < .001; NNT = 3). Furthermore, consumption of the synbiotic was associated with significant weight gain (approximately 40 g) in infants (adjusted SMD = 0.63; 95% CI: 0.26-1.00, P value < .001). The duration of hospitalization and the time required to reach full enteral feeding were also significantly shorter in the synbiotic group (by approximately 3 days). No serious side effects were reported. CONCLUSION: Prescribing multi-strain synbiotics reduces the incidence of NEC in VLBW infants and has beneficial effects on breastfeeding tolerance and weight gain velocity. Therefore, physicians may consider prescribing synbiotics to VLBW neonates.


Sujet(s)
Nutrition entérale , Entérocolite nécrosante , Nourrisson très faible poids naissance , Synbiotiques , Humains , Synbiotiques/administration et posologie , Méthode en double aveugle , Entérocolite nécrosante/prévention et contrôle , Entérocolite nécrosante/épidémiologie , Nutrition entérale/méthodes , Nouveau-né , Femelle , Mâle , Unités de soins intensifs néonatals , Lacticaseibacillus rhamnosus , Microbiome gastro-intestinal
8.
Pharmacol Res ; 208: 107398, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39241935

RÉSUMÉ

BACKGROUND: Patients with non-alcoholic fatty liver disease (NAFLD) benefit from using synbiotics. However, findings from existing trials remain contentious. Therefore, this meta-analysis evaluated the effects of synbiotics on liver enzymes, blood pressure, inflammation, and lipid profiles in patients with NAFLD. METHODS: We searched PubMed, Embase, Cochrane, Scopus, and Web of Science for randomized controlled trials (RCTs) regarding synbiotics supplementation in patients with NAFLD. RESULTS: The meta-analysis revealed that synbiotics supplementation significantly improved liver enzymes (AST, WMD: -9.12 IU/L; 95 % CI: -13.19 to -5.05; ALT, WMD: -8.53 IU/L; 95 % CI: -15.07 to -1.99; GGT, WMD: -10.42 IU/L; 95 % CI: -15.19 to -5.65), lipid profile (TC, WMD: -7.74 mg/dL; 95 % CI: -12.56 to -2.92), obesity indices (body weight, WMD: -1.95 kg; 95 % CI: -3.69 to -0.22; WC, WMD: -1.40 cm; 95 % CI: -2.71 to -0.10), systolic blood pressure (SBP, WMD: -6.00 mmHg; 95 % CI: -11.52 to -0.49), and inflammatory markers (CRP, WMD: -0.69 mg/L; 95 % CI: -1.17 to -0.21; TNF-α, WMD: -14.01 pg/mL; 95 % CI: -21.81 to -6.20). CONCLUSION: Overall, supplementation with synbiotics positively improved liver enzymes, obesity indices, and inflammatory cytokines in patients with NAFLD.


Sujet(s)
Pression sanguine , Inflammation , Lipides , Foie , Stéatose hépatique non alcoolique , Obésité , Essais contrôlés randomisés comme sujet , Synbiotiques , Humains , Synbiotiques/administration et posologie , Pression sanguine/effets des médicaments et des substances chimiques , Foie/métabolisme , Lipides/sang
9.
Trials ; 25(1): 615, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39289685

RÉSUMÉ

BACKGROUND: Prematurity remains one of the main causes of neonatal morbidity and mortality. Approximately two thirds of preterm births are spontaneous, i.e. secondary to preterm labour, preterm prelabour rupture of membranes (PPROM) or cervical insufficiency. Etiologically, the vaginal microbiome plays an important role in spontaneous preterm birth (sPTB). Vaginal dysbiosis and bacterial vaginosis are well-known risk factors for ascending lower genital tract infections and sPTB, while a Lactobacillus crispatus-dominated vaginal microbiome is associated with term deliveries. Synbiotics may help to achieve and/or maintain a normal, Lactobacillus-dominated vaginal microbiome. METHODS: We will perform a multi-centre, double-blind, randomised, placebo-controlled trial. Women aged 18 years or older with a singleton pregnancy are eligible for inclusion at 80/7-106/7 weeks gestational age if they have one or more of the following risk factors for sPTB: previous sPTB at 240/7-356/7 weeks, prior PPROM before 360/7 weeks, or spontaneous pregnancy loss at 140/7-236/7 weeks of gestation. Exclusion criteria are multiple gestation, cervix conisation, inflammatory bowel disease, uterine anomaly, and the use of pro-/pre-/synbiotics. Patients will be randomised to oral synbiotics or placebo, starting before 11 weeks of gestation until delivery. The oral synbiotic consists of eight Lactobacillus species (including L. crispatus) and prebiotics. The primary outcome is the gestational age at delivery. Vaginal microbiome analysis once per trimester (at approximately 9, 20, and 30 weeks) and delivery will be performed using metataxonomic sequencing (16S rRNA gene) and microbial culture. Secondary outcomes include PPROM, the use of antibiotics, antenatal admission information, and neonatal outcomes. DISCUSSION: This study will evaluate the effect of oral synbiotics on the vaginal microbiome during pregnancy in a high-risk population and correlate the microbial changes with the gestational age at delivery and relevant pregnancy outcomes. TRIAL REGISTRATION: ClinicalTrials.gov, NCT05966649. Registered on April 5, 2024.


Sujet(s)
Études multicentriques comme sujet , Naissance prématurée , Essais contrôlés randomisés comme sujet , Synbiotiques , Vagin , Humains , Femelle , Méthode en double aveugle , Grossesse , Naissance prématurée/prévention et contrôle , Synbiotiques/administration et posologie , Vagin/microbiologie , Facteurs de risque , Microbiote , Âge gestationnel , Nouveau-né , Résultat thérapeutique , Vaginose bactérienne/microbiologie , Vaginose bactérienne/diagnostic
10.
J Agric Food Chem ; 72(33): 18497-18506, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39099138

RÉSUMÉ

Synbiotics, the combination of probiotics and prebiotics, are thought to be a pragmatic approach for the treatment of various diseases, including inflammatory bowel disease (IBD). The synergistic therapeutic effects of probiotics and prebiotics remain underexplored. Clostridium tyrobutyricum, a short-chain fatty acid (SCFA) producer, has been recognized as a promising probiotic candidate that can offer health benefits. In this study, the treatment effects of synbiotics containing C. tyrobutyricum and chitooligosaccharides (COSs) on IBD were evaluated. The results indicated that the synbiotic supplement effectively relieved inflammation and restored intestinal barrier function. Additionally, the synbiotic supplement could contribute to the elimination of reactive oxygen species (ROS) and improve the production of SCFAs through the SCFAs-producer of C. tyrobutyricum. Furthermore, such the synbiotic could also regulate the composition of gut microbiota. These findings underscore the potential of C. tyrobutyricum and COSs as valuable living biotherapeutics for the treatment of intestinal-related diseases.


Sujet(s)
Clostridium tyrobutyricum , Microbiome gastro-intestinal , Maladies inflammatoires intestinales , Oligosaccharides , Synbiotiques , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Oligosaccharides/administration et posologie , Maladies inflammatoires intestinales/microbiologie , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/thérapie , Maladies inflammatoires intestinales/métabolisme , Clostridium tyrobutyricum/métabolisme , Animaux , Humains , Synbiotiques/administration et posologie , Souris , Mâle , Acides gras volatils/métabolisme , Souris de lignée C57BL , Probiotiques/administration et posologie , Probiotiques/pharmacologie , Prébiotiques/administration et posologie , Chitosane
11.
Sci Rep ; 14(1): 18518, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39122808

RÉSUMÉ

In this study, a variety of probiotic strains, including Lactiplantibacillus plantarum, Lacticaseibacillus casei, Lactobacillus acidophilus, Streptococcus thermophilus, Bifidobacterium longum, Limosilactobacillus reuteri, Lactobacillus delbrueckii subsp. bulgaricus, Lacticaseibacillus rhamnosus, and Bifidobacterium bifidum, were utilized for soymilk fermentation both as free cells and as synbiotics on agro-industrial residuals such as okara, whey protein, banana peels, apple pomace, sugarcane bagasse, orange peels, and lemon peels. Among these, Lacticaseibacillus rhamnosus emerged as the most significant strain for soymilk fermentation, exhibiting a viability of 10.47 log cfu/mL, a pH of 4.41, total acidity of 1.12%, and organic acid contents (lactic and acetic acid) of 11.20 and 7.50 g/L, respectively. As a synbiotic Lacticaseibacillus rhamnosus immobilised on okara, showed even more impressive results, with a viability of 12.98 log cfu/mL, a pH of 4.31, total acidity of 1.27%, and organic acid contents of 13.90 and 9.30 g/L, respectively. Over a 12-h fermentation period, cell viability values increased by 10.47-fold in free cells and 11.19-fold in synbiotics. Synbiotic supplementation of fermented soymilk proved more beneficial than free cells in terms of viability, acidity, and organic acid content. Furthermore, when synbiotic fermented soymilk was freeze-dried to simulate the digestive system in vitro, synbiotics and freeze-dried cells demonstrated superior gastrointestinal tract survival compared to free cells. Both the probiotic bacteria and the synbiotics exhibited cytotoxicity against colon and liver cancer cell lines, with half-maximal inhibitory concentrations ranging from 41.96 to 61.52 µL/well.


Sujet(s)
Fermentation , Probiotiques , Jus de soja , Synbiotiques , Humains , Probiotiques/métabolisme , Jus de soja/composition chimique , Jus de soja/métabolisme , Antinéoplasiques/pharmacologie
12.
Sci Rep ; 14(1): 19120, 2024 08 18.
Article de Anglais | MEDLINE | ID: mdl-39155305

RÉSUMÉ

Cardiovascular disease is one of the leading causes of death worldwide. Evidence suggests that alterations in the gut microbiome could play a role in cardiovascular diseases, including heart failure. The purpose of this study was to evaluate the effect of synbiotics on serum paraoxonase 1(PON1), soluble CD163/soluble TNF-like weak inducer of apoptosis (sCD163/sTWEAK), and lipid profile, which are involved in heart failure in patients with chronic heart failure. In this triple-blind randomized clinical trial, 90 eligible patients were included in the study. They were randomly assigned to receive one capsule (500 mg) of synbiotics or a placebo daily for ten weeks. Serum PON1, sCD163/sTWEAK, and lipid profiles were measured at the beginning and end of the study. The data were analyzed by SPSS 24, and the p-value < 0.05 was considered statistically significant. Among 90 patients who met the inclusion criteria, 80 completed the study. The primary outcomes showed a small effect on sTWEAK, with an adjusted standard mean difference (SMD) of 0.2. However, no significant changes were observed in sCD163/sTWEAK (SMD: 0.16). Secondary outcomes indicated no changes in PON1, total cholesterol (TC), or LDL-C levels. However, there was an increase in HDL-C levels (adjusted SMD: 0.46, 95% CI: 0.02-0.91) and a decrease in TG and TC/HDL levels (adjusted SMD: - 0.5 and - 0.3, respectively) in the synbiotic group. A favorable effect of synbiotics on sTWEAK, HDL, TG, and TC/HDL of patients with heart failure was observed, but no statistically significant effect was found on sCD163/sTWEAK, PON1, LDL, and TC factors.


Sujet(s)
Aryldialkylphosphatase , Défaillance cardiaque , Synbiotiques , Humains , Aryldialkylphosphatase/sang , Mâle , Femelle , Synbiotiques/administration et posologie , Défaillance cardiaque/sang , Adulte d'âge moyen , Sujet âgé , Récepteurs de surface cellulaire/sang , Antigènes CD/sang , Cytokine TWEAK/sang , Lipoprotéines/sang , Maladie chronique , Marqueurs biologiques/sang , Antigènes de différenciation des myélomonocytes
13.
PLoS One ; 19(8): e0304692, 2024.
Article de Anglais | MEDLINE | ID: mdl-39110654

RÉSUMÉ

During the last years, growing interest in the use of mare's milk in food production is observed. The subject of the study was to evaluate the feasibility of mare's milk for the production of yogurt ice cream and synbiotic ice cream. Four variants of mare's milk ice cream were developed: ice cream with yogurt bacteria without inulin (YO) and with 2% of inulin (YO+I), synbiotic ice cream with 2% inulin and Lacticaseibacillus rhamnosus (LCR+I) and with Lactiplantibacillus plantarum (LP+I). Ice creams were enriched with inulin in order to evaluate its influence on the viability of LAB and on the product quality. Physicochemical, textural and sensory analyses were performed. Count of viable bacteria cells was also evaluated. Obtained ice creams did not differ in terms of protein, fat and total solids content (1.85-1.91%, 7.33-7.58% and 24.66-26.96% respectively), but differed in acidity. Ice cream YO, the only one without inulin, had the highest acidity, what suggests that inulin decrease this parameter. Regardless the type of LAB starter culture and inulin addition, samples had the same range of overrun (35.20-44.03%) and melting rate (73.49-79.87%). However the variant of ice cream influenced textural properties and colour parameters. All obtained mare's milk ice creams had high overall sensory quality. It was noticed, that ice cream with inulin had higher count of LAB (>7logCFU/g), than sample without inulin (>6logCFU/g). In conclusion, mare's milk may be considered as feasible raw material for yogurt ice cream and synbiotic ice cream production.


Sujet(s)
Crème glacée , Lait , Synbiotiques , Yaourt , Crème glacée/analyse , Crème glacée/microbiologie , Yaourt/analyse , Yaourt/microbiologie , Animaux , Synbiotiques/analyse , Lait/composition chimique , Equus caballus , Femelle , Inuline , Lacticaseibacillus rhamnosus/métabolisme , Humains , Microbiologie alimentaire
14.
Food Funct ; 15(18): 9471-9487, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39193624

RÉSUMÉ

Equol is a highly active product of soy isoflavones produced by specific bacteria in the human or animal colon. However, equol production is influenced by differences in the gut flora carried by the body. Our previous research has shown that a synbiotic preparation comprising the probiotic Lactobacillus rhamnosus ATCC 7469 and the prebiotic lactulose can enhance equol production by modulating the intestinal flora. Nevertheless, the harsh environment of the gastrointestinal tract limits this capability by diminishing the number of probiotics reaching the colon. Microencapsulation of probiotics is an effective strategy to enhance their viability. In this study, probiotic gel microspheres (SA-S-CS) were prepared using an extrusion method, with sodium alginate (SA) and chitosan (CS) serving as the encapsulating materials. Scanning electron microscopy (SEM) was employed to observe the surface morphology and the internal distribution of bacteria within the microcapsules. The structural characteristics of the microcapsules were investigated using Fourier-transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD). Furthermore, the thermal stability, storage stability, probiotic viability post-simulated gastrointestinal fluid treatment, and colon release rate were examined. Finally, the impact of probiotic microencapsulation on promoting equol production by the synbiotic preparation was assessed. The results indicated that the microcapsules exhibited a spherical structure with bacteria evenly distributed on the inner surface. Studies on thermal and storage stability showed that the number of viable cells in the probiotic microcapsule group significantly increased compared to the free probiotic group. Gastrointestinal tolerance studies revealed that after in vitro simulated gastrointestinal digestion, the amount of viable cells in the microcapsules was 7 log10 CFU g-1, demonstrating good gastrointestinal tolerance. Moreover, after incubation in simulated colonic fluid for 150 min, the release rate of probiotics reached 93.13%. This suggests that chitosan-coated sodium alginate microcapsules can shield Lactobacillus rhamnosus ATCC 7469 from the gastrointestinal environment, offering a novel model for synbiotic preparation to enhance equol production.


Sujet(s)
Équol , Lacticaseibacillus rhamnosus , Lactulose , Probiotiques , Synbiotiques , Lacticaseibacillus rhamnosus/métabolisme , Lactulose/métabolisme , Humains , Alginates/composition chimique , Préparation de médicament/méthodes , Chitosane/composition chimique , Côlon/microbiologie , Côlon/métabolisme
15.
Clin Nutr ESPEN ; 63: 813-820, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39178987

RÉSUMÉ

The intricate ecosystem of microorganisms residing within and on the human body, collectively known as the microbiome, significantly influences human health. Imbalances in this microbiome, referred to as dysbiosis, have been associated with various diseases, prompting the exploration of novel therapeutic approaches. Personalized medicine, Tailors treatments to individual patient characteristics, offers a promising avenue for addressing microbiome-related health issues. This review highlights recent developments in utilizing personalized medicine to target the microbiome, aiming to enhance health outcomes. Noteworthy strategies include fecal microbiota transplantation (FMT), where healthy donor microbes are transferred to patients, showing promise in treating conditions such as recurrent Clostridium difficile infection. Additionally, probiotics, which are live microorganisms similar to beneficial gut inhabitants, and prebiotics, non-digestible compounds promoting microbial growth, are emerging as tools to restore microbiome balance. The integration of these approaches, known as synbiotics, enhances microbial colonization and therapeutic effects. Advances in metagenomics and sequencing technologies provide the means to understand individual microbiome profiles, enabling tailored interventions. This paper aims to present the latest insights in leveraging personalized medicine to address microbiome-related health concerns, envisioning a future where microbiome-based therapies reshape disease management and promote human health.


Sujet(s)
Dysbiose , Transplantation de microbiote fécal , Microbiome gastro-intestinal , Prébiotiques , Médecine de précision , Probiotiques , Humains , Probiotiques/usage thérapeutique , Dysbiose/thérapie , Microbiote , Synbiotiques
16.
J Microbiol Biotechnol ; 34(9): 1739-1747, 2024 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-39099195

RÉSUMÉ

Alzheimer's disease (AD) is a progressive neurological disorder that represents a major cause of dementia worldwide. Its pathogenesis involves multiple pathways, including the amyloid cascade, tau protein, oxidative stress, and metal ion dysregulation. Recent studies have suggested a critical link between changes in gut microbial diversity and the disruption of the gut-brain axis in AD. Previous studies primarily explored the potential benefits of probiotics and prebiotics in managing AD. However, studies have yet to fully describe a novel promising approach involving the use of synbiotics, which include a combination of active probiotics and new-generation prebiotics. Synbiotics show potential for mitigating the onset and progression of AD, thereby offering a holistic approach to address the multifaceted nature of AD. This review article primarily aims to gain further insights into the mechanisms of AD, specifically the intricate interaction between gut bacteria and the brain via the gut-brain axis. By understanding this relationship, we can identify potential targets for intervention and therapeutic strategies to combat AD effectively. This review also discusses substantial evidence supporting the role of synbiotics as a promising AD treatment that surpasses traditional probiotic or prebiotic interventions. We find that synbiotics may be used not only to address cognitive decline but also to reduce AD-related psychological burden, thus enhancing the overall quality of life of patients with AD.


Sujet(s)
Maladie d'Alzheimer , Microbiome gastro-intestinal , Probiotiques , Synbiotiques , Maladie d'Alzheimer/thérapie , Maladie d'Alzheimer/prévention et contrôle , Synbiotiques/administration et posologie , Humains , Probiotiques/usage thérapeutique , Probiotiques/administration et posologie , Santé mentale , Axe cerveau-intestin/physiologie , Prébiotiques , Animaux , Encéphale , Qualité de vie
17.
Int J Biol Macromol ; 278(Pt 3): 134860, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39163956

RÉSUMÉ

Exploring nutritional therapies that manipulate tryptophan metabolism to activate AhR signaling represents a promising approach for mitigating chronic colitis. Arabinoxylan is a bioactive constituent abundant in wheat bran. Here, we comprehensively investigated anti-colitis potentials of wheat bran arabinoxylan (WBAX), its synbiotic and postbiotic derived from WBAX and Limosilactobacillus reuteri WX-94 (i.e., a probiotic strain exhibiting tryptophan metabolic activity). WBAX fueled L. reuteri and promoted microbial conversion of tryptophan to AhR ligands during in vitro fermentation in the culture medium and in the fecal microbiota from type 2 diabetes. The WBAX postbiotic outperformed WBAX and its synbiotic in augmenting efficacy of tryptophan in restoring DSS-disturbed serum immune markers, colonic tight junction proteins and gene profiles involved in amino acid metabolism and FoxO signaling. The WBAX postbiotic remodeled gut microbiota and superiorly enhanced AhR ligands (i.e., indole metabolites and bile acids), alongside with elevation in colonic AhR and IL-22. Associations between genera and metabolites modified by the postbiotic and colitis in human were verified and strong binding capacities between metabolites and colitis-related targets were demonstrated by molecular docking. Our study advances the novel perspective of WBAX in manipulating tryptophan metabolism and anti-colitis potentials of WBAX postbiotic via promoting gut microbiota-dependent AhR signaling.


Sujet(s)
Colite , Fibre alimentaire , Microbiome gastro-intestinal , Limosilactobacillus reuteri , Synbiotiques , Xylanes , Xylanes/pharmacologie , Xylanes/composition chimique , Xylanes/métabolisme , Limosilactobacillus reuteri/métabolisme , Colite/métabolisme , Colite/microbiologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Fibre alimentaire/métabolisme , Humains , Animaux , Souris , Récepteurs à hydrocarbure aromatique/métabolisme , Tryptophane/métabolisme , Simulation de docking moléculaire , Fermentation , Mâle , Diabète de type 2/métabolisme , Diabète de type 2/microbiologie , Multi-omique
18.
Commun Biol ; 7(1): 943, 2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39098939

RÉSUMÉ

To enhance health benefits, a probiotic can be co-administered with a metabolizable prebiotic forming a synergistic synbiotic. We assessed the synergies resulting from combining Bifidobacterium longum subsp. infantis LMG 11588 and an age-adapted blend of six human milk oligosaccharides (HMOs) in ex vivo colonic incubation bioreactors seeded with fecal background microbiota from infant and toddler donors. When HMOs were combined with B. infantis LMG 11588, they were rapidly and completely consumed. This resulted in increased short chain fatty acid (SCFA) production compared to the summed SCFA production from individual ingredients (synergy). Remarkably, HMOs were partially consumed for specific infant donors in the absence of B. infantis LMG 11588, yet all donors showed increased SCFA production upon B. infantis LMG 11588 supplementation. We found specific bacterial taxa associated with the differential response pattern to HMOs. Our study shows the importance of carefully selecting pre- and probiotic into a synergistic synbiotic that could benefit infants.


Sujet(s)
Bifidobacterium longum sous-espèce infantis , Acides gras volatils , Lait humain , Oligosaccharides , Humains , Lait humain/métabolisme , Lait humain/composition chimique , Oligosaccharides/métabolisme , Acides gras volatils/métabolisme , Nourrisson , Bifidobacterium longum sous-espèce infantis/métabolisme , Bifidobacterium/métabolisme , Probiotiques/administration et posologie , Microbiome gastro-intestinal , Fèces/microbiologie , Femelle , Synbiotiques/administration et posologie , Enfant d'âge préscolaire
19.
Nutrients ; 16(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125280

RÉSUMÉ

Postbiotics are defined as a preparation of inanimate microorganisms and/or their components that confers a health benefit to the host. They range from cell wall fragments to metabolites, bacterial lysates, extracellular vesicles, and short-chain fatty acids (SCFAs). Postbiotics may influence carcinogenesis via a variety of mechanisms. They can promote homeostatic immune responses, reduce inflammation, induce selective cytotoxicity against tumor cells, as well as the enabling the control of tumor cell proliferation and enhancing intestinal epithelial barrier function. Therefore, probiotics can serve as an adjunct strategy in anticancer treatment together with chemotherapy and immunotherapy. Up to now, the only relevant postbiotics used as interventions in oncological patients remain vitamin K molecules, with few phase-II and III trials available. In fact, postbiotics' levels are strictly dependent on the gut microbiota's composition, which may vary between individuals and can be altered under different physiological and pathological conditions. Therefore, the lack of consistent clinical evidence supporting postbiotics' efficacy is due to their poor bioavailability, short half-life, and fluctuating levels. Synbiotics, a mixture of prebiotics and probiotics, are expected to have a more homogeneous bioavailability with respect to postbiotics and may have greater potential for future development. In this review, we focus on the role of postbiotics as an adjuvant therapy in cancer treatment.


Sujet(s)
Microbiome gastro-intestinal , Tumeurs , Probiotiques , Humains , Tumeurs/thérapie , Probiotiques/usage thérapeutique , Prébiotiques/administration et posologie , Synbiotiques/administration et posologie
20.
BMC Gastroenterol ; 24(1): 283, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39174901

RÉSUMÉ

BACKGROUND: There is a contradiction in the use of microbiota-therapies, including probiotics, prebiotics, and synbiotics, to improve the condition of patients with nonalcoholic fatty liver disease (NAFLD). The aim of this review was to evaluate the effect of microbiota-therapy on liver injury, inflammation, and lipid levels in individuals with NAFLD. METHODS: Using Pubmed, Embase, Cochrane Library, and Web of Science databases were searched for articles on the use of prebiotic, probiotic, or synbiotic for the treatment of patients with NAFLD up to March 2024. RESULTS: Thirty-four studies involving 12,682 individuals were included. Meta-analysis indicated that probiotic, prebiotic, and synbiotic supplementation significantly improved liver injury (hepatic fibrosis, SMD = -0.31; 95% CI: -0.53, -0.09; aspartate aminotransferase, SMD = -0.35; 95% CI: -0.55, -0.15; alanine aminotransferase, SMD = -0.48; 95% CI: -0.71, -0.25; alkaline phosphatase, SMD = -0.81; 95% CI: -1.55, -0.08), lipid profiles (triglycerides, SMD = -0.22; 95% CI: -0.43, -0.02), and inflammatory factors (high-density lipoprotein, SMD = -0.47; 95% CI: -0.88, -0.06; tumour necrosis factor alpha, SMD = -0.86 95% CI: -1.56, -0.56). CONCLUSION: Overall, supplementation with probiotic, prebiotic, or synbiotic had a positive effect on reducing liver enzymes, lipid profiles, and inflammatory cytokines in patients with NAFLD.


Sujet(s)
Stéatose hépatique non alcoolique , Prébiotiques , Probiotiques , Synbiotiques , Humains , Alanine transaminase/sang , Inflammation , Lipides/sang , Foie , Stéatose hépatique non alcoolique/thérapie , Probiotiques/usage thérapeutique , Essais contrôlés randomisés comme sujet , Synbiotiques/administration et posologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE