Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 744
1.
World J Gastroenterol ; 30(16): 2258-2271, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38690023

BACKGROUND: Irritable bowel syndrome (IBS) is one of the most frequent and debilitating conditions leading to gastroenterological referrals. However, recommended treatments remain limited, yielding only limited therapeutic gains. Chitin-glucan (CG) is a novel dietary prebiotic classically used in humans at a dosage of 1.5-3.0 g/d and is considered a safe food ingredient by the European Food Safety Authority. To provide an alternative approach to managing patients with IBS, we performed preclinical molecular, cellular, and animal studies to evaluate the role of chitin-glucan in the main pathophysiological mechanisms involved in IBS. AIM: To evaluate the roles of CG in visceral analgesia, intestinal inflammation, barrier function, and to develop computational molecular models. METHODS: Visceral pain was recorded through colorectal distension (CRD) in a model of long-lasting colon hypersensitivity induced by an intra-rectal administration of TNBS [15 milligrams (mg)/kilogram (kg)] in 33 Sprague-Dawley rats. Intracolonic pressure was regularly assessed during the 9 wk-experiment (weeks 0, 3, 5, and 7) in animals receiving CG (n = 14) at a human equivalent dose (HED) of 1.5 g/d or 3.0 g/d and compared to negative control (tap water, n = 11) and positive control (phloroglucinol at 1.5 g/d HED, n = 8) groups. The anti-inflammatory effect of CG was evaluated using clinical and histological scores in 30 C57bl6 male mice with colitis induced by dextran sodium sulfate (DSS) administered in their drinking water during 14 d. HT-29 cells under basal conditions and after stimulation with lipopolysaccharide (LPS) were treated with CG to evaluate changes in pathways related to analgesia (µ-opioid receptor (MOR), cannabinoid receptor 2 (CB2), peroxisome proliferator-activated receptor alpha, inflammation [interleukin (IL)-10, IL-1b, and IL-8] and barrier function [mucin 2-5AC, claudin-2, zonula occludens (ZO)-1, ZO-2] using the real-time PCR method. Molecular modelling of CG, LPS, lipoteichoic acid (LTA), and phospholipomannan (PLM) was developed, and the ability of CG to chelate microbial pathogenic lipids was evaluated by docking and molecular dynamics simulations. Data were expressed as the mean ± SEM. RESULTS: Daily CG orally-administered to rats or mice was well tolerated without including diarrhea, visceral hypersensitivity, or inflammation, as evaluated at histological and molecular levels. In a model of CRD, CG at a dosage of 3 g/d HED significantly decreased visceral pain perception by 14% after 2 wk of administration (P < 0.01) and reduced inflammation intensity by 50%, resulting in complete regeneration of the colonic mucosa in mice with DSS-induced colitis. To better reproduce the characteristics of visceral pain in patients with IBS, we then measured the therapeutic impact of CG in rats with TNBS-induced inflammation to long-lasting visceral hypersensitivity. CG at a dosage of 1.5 g/d HED decreased visceral pain perception by 20% five weeks after colitis induction (P < 0.01). When the CG dosage was increased to 3.0 g/d HED, this analgesic effect surpassed that of the spasmolytic agent phloroglucinol, manifesting more rapidly within 3 wk and leading to a 50% inhibition of pain perception (P < 0.0001). The underlying molecular mechanisms contributing to these analgesic and anti-inflammatory effects of CG involved, at least in part, a significant induction of MOR, CB2 receptor, and IL-10, as well as a significant decrease in pro-inflammatory cytokines IL-1b and IL-8. CG also significantly upregulated barrier-related genes including muc5AC, claudin-2, and ZO-2. Molecular modelling of CG revealed a new property of the molecule as a chelator of microbial pathogenic lipids, sequestering gram-negative LPS and gram-positive LTA bacterial toxins, as well as PLM in fungi at the lowesr energy conformations. CONCLUSION: CG decreased visceral perception and intestinal inflammation through master gene regulation and direct binding of microbial products, suggesting that CG may constitute a new therapeutic strategy for patients with IBS or IBS-like symptoms.


Chitin , Colon , Disease Models, Animal , Glucans , Irritable Bowel Syndrome , Rats, Sprague-Dawley , Visceral Pain , Animals , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/physiopathology , Male , Humans , Colon/drug effects , Colon/pathology , Rats , Visceral Pain/drug therapy , Visceral Pain/physiopathology , Visceral Pain/metabolism , Visceral Pain/etiology , Chitin/pharmacology , Glucans/pharmacology , Glucans/administration & dosage , Mice , Prebiotics/administration & dosage , Trinitrobenzenesulfonic Acid/toxicity , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Colitis/drug therapy , Colitis/chemically induced , Colitis/physiopathology , Colitis/pathology , HT29 Cells
2.
Sci Rep ; 14(1): 11127, 2024 05 15.
Article En | MEDLINE | ID: mdl-38750102

Nutraceutical interventions supporting microbiota and eliciting clinical improvements in metabolic diseases have grown significantly. Chronic stress, gut dysbiosis, and metainflammation have emerged as key factors intertwined with sleep disorders, consequently exacerbating the decline in quality of life. This study aimed to assess the effects of two nutraceutical formulations containing prebiotics (fructooligosaccharides (FOS), galactooligosaccharides (GOS), yeast ß-glucans), minerals (Mg, Se, Zn), and the herbal medicine Silybum marianum L. Gaertn., Asteraceae (Milk thistle or Silymarin). These formulations, namely NSupple (without silymarin) and NSupple_Silybum (with silymarin) were tested over 180 days in overweight/obese volunteers from Brazil's southeastern region. We accessed fecal gut microbiota by partial 16S rRNA sequences; cytokines expression by CBA; anthropometrics, quality of life and sleep, as well as metabolic and hormonal parameters, at baseline (T0) and 180 days (T180) post-supplementation. Results demonstrated gut microbiota reshaping at phyla, genera, and species level post-supplementation. The Bacteroidetes phylum, Bacteroides, and Prevotella genera were positively modulated especially in the NSupple_Silybum group. Gut microbiota modulation was associated with improved sleep patterns, quality-of-life perception, cytokines expression, and anthropometric parameters post-supplementation. Our findings suggest that the nutraceutical blends positively enhance cardiometabolic and inflammatory markers. Particularly, NSupple_Silybum modulated microbiota composition, underscoring its potential significance in ameliorating metabolic dysregulation. Clinical trial registry number: NCT04810572. 23/03/2021.


Cytokines , Dietary Supplements , Gastrointestinal Microbiome , Quality of Life , Humans , Gastrointestinal Microbiome/drug effects , Male , Brazil , Female , Double-Blind Method , Adult , Cytokines/metabolism , Middle Aged , Prebiotics/administration & dosage , Feces/microbiology , Silymarin/pharmacology , Minerals/pharmacology , Obesity/microbiology , Oligosaccharides/pharmacology , Oligosaccharides/administration & dosage
3.
Nutr Diabetes ; 14(1): 25, 2024 May 10.
Article En | MEDLINE | ID: mdl-38729941

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a significant risk factor for non-alcoholic fatty liver disease (NAFLD). Increased fasting blood sugar (FBS), fasting insulin (FI), and insulin resistance (HOMA-IR) are observed in patients with NAFLD. Gut microbial modulation using prebiotics, probiotics, and synbiotics has shown promise in NAFLD treatment. This meta-umbrella study aimed to investigate the effects of gut microbial modulation on glycemic indices in patients with NAFLD and discuss potential mechanisms of action. METHODS: A systematic search was conducted in PubMed, Web of Science, Scopus, and Cochrane Library until March 2023 for meta-analyses evaluating the effects of probiotics, prebiotics, and synbiotics on patients with NAFLD. Random-effect models, sensitivity analysis, and subgroup analysis were employed. RESULTS: Gut microbial therapy significantly decreased HOMA-IR (ES: -0.41; 95%CI: -0.52, -0.31; P < 0.001) and FI (ES: -0.59; 95%CI: -0.77, -0.41; P < 0.001). However, no significant effect was observed on FBS (ES: -0.17; 95%CI: -0.36, 0.02; P = 0.082). Subgroup analysis revealed prebiotics had the most potent effect on HOMA-IR, followed by probiotics and synbiotics. For FI, synbiotics had the most substantial effect, followed by prebiotics and probiotics. CONCLUSION: Probiotics, prebiotics, and synbiotics administration significantly reduced FI and HOMA-IR, but no significant effect was observed on FBS.


Gastrointestinal Microbiome , Glycemic Index , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Prebiotics , Probiotics , Synbiotics , Humans , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/diet therapy , Prebiotics/administration & dosage , Probiotics/therapeutic use , Probiotics/administration & dosage , Synbiotics/administration & dosage , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/therapy , Insulin/blood
4.
Gut Microbes ; 16(1): 2356279, 2024.
Article En | MEDLINE | ID: mdl-38778521

Repeated exposure to antibiotics and changes in the diet and environment shift the gut microbial diversity and composition, making the host susceptible to pathogenic infection. The emergence and ongoing spread of AMR pathogens is a challenging public health issue. Recent evidence showed that probiotics and prebiotics may play a role in decolonizing drug-resistant pathogens by enhancing the colonization resistance in the gut. This review aims to analyze available evidence from human-controlled trials to determine the effect size of probiotic interventions in decolonizing AMR pathogenic bacteria from the gut. We further studied the effects of prebiotics in human and animal studies. PubMed, Embase, Web of Science, Scopus, and CINAHL were used to collect articles. The random-effects model meta-analysis was used to pool the data. GRADE Pro and Cochrane collaboration tools were used to assess the bias and quality of evidence. Out of 1395 citations, 29 RCTs were eligible, involving 2871 subjects who underwent either probiotics or placebo treatment to decolonize AMR pathogens. The persistence of pathogenic bacteria after treatment was 22%(probiotics) and 30.8%(placebo). The pooled odds ratio was 0.59(95% CI:0.43-0.81), favoring probiotics with moderate certainty (p = 0.0001) and low heterogeneity (I2 = 49.2%, p = 0.0001). The funnel plot showed no asymmetry in the study distribution (Kendall'sTau = -1.06, p = 0.445). In subgroup, C. difficile showed the highest decolonization (82.4%) in probiotics group. Lactobacillus-based probiotics and Saccharomyces boulardii decolonize 71% and 77% of pathogens effectively. The types of probiotics (p < 0.018) and pathogens (p < 0.02) significantly moderate the outcome of decolonization, whereas the dosages and regions of the studies were insignificant (p < 0.05). Prebiotics reduced the pathogens from 30% to 80% of initial challenges. Moderate certainty of evidence suggests that probiotics and prebiotics may decolonize pathogens through modulation of gut diversity. However, more clinical outcomes are required on particular strains to confirm the decolonization of the pathogens. Protocol registration: PROSPERO (ID = CRD42021276045).


Bacteria , Gastrointestinal Microbiome , Prebiotics , Probiotics , Probiotics/administration & dosage , Probiotics/therapeutic use , Probiotics/pharmacology , Humans , Prebiotics/administration & dosage , Gastrointestinal Microbiome/drug effects , Bacteria/classification , Bacteria/isolation & purification , Animals , Treatment Outcome , Anti-Bacterial Agents/pharmacology , Bacterial Infections/microbiology , Bacterial Infections/prevention & control , Gastrointestinal Tract/microbiology
5.
ACS Nano ; 18(21): 13583-13598, 2024 May 28.
Article En | MEDLINE | ID: mdl-38740518

A significant gap exists in the demand for safe and effective drugs for inflammatory bowel disease (IBD), and its associated intestinal fibrosis. As oxidative stress plays a central role in the pathogenesis of IBD, astaxanthin (AST), a good antioxidant with high safety, holds promise for treating IBD. However, the application of AST is restricted by its poor solubility and easy oxidation. Herein, different protein-based nanoparticles (NPs) are fabricated for AST loading to identify an oral nanovehicle with potential clinical applicability. Through systematic validation via molecular dynamics simulation and in vitro characterization of properties, whey protein isolate (WPI)-driven NPs using a simple preparation method without the need for cross-linking agents or emulsifiers were identified as the optimal carrier for oral AST delivery. Upon oral administration, the WPI-driven NPs, benefiting from the intrinsic pH sensitivity and mucoadhesive properties, effectively shielded AST from degradation by gastric juices and targeted release of AST at intestinal lesion sites. Additionally, the AST NPs displayed potent therapeutic efficacy in both dextran sulfate sodium (DSS)-induced acute colitis and chronic colitis-associated intestinal fibrosis by ameliorating inflammation, oxidative damage, and intestinal microecology. In conclusion, the AST WPI NPs hold a potential therapeutic value in treating inflammation and fibrosis in IBD.


Inflammatory Bowel Diseases , Nanoparticles , Prebiotics , Reactive Oxygen Species , Whey Proteins , Whey Proteins/chemistry , Whey Proteins/pharmacology , Animals , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Reactive Oxygen Species/metabolism , Administration, Oral , Nanoparticles/chemistry , Prebiotics/administration & dosage , Fibrosis/drug therapy , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Mice , Xanthophylls/pharmacology , Xanthophylls/chemistry , Xanthophylls/administration & dosage , Dextran Sulfate , Mice, Inbred C57BL , Male , Antioxidants/chemistry , Antioxidants/pharmacology , Humans
6.
Cell Commun Signal ; 22(1): 268, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745207

Ulcerative colitis (UC) is increasingly common, and it is gradually become a kind of global epidemic. UC is a type of inflammatory bowel disease (IBD), and it is a lifetime recurrent disease. UC as a common disease has become a financial burden for many people and has the potential to develop into cancer if not prevented or treated. There are multiple factors such as genetic factors, host immune system disorders, and environmental factors to cause UC. A growing body of research have suggested that intestinal microbiota as an environmental factor play an important role in the occurrence and development of UC. Meanwhile, evidence to date suggests that manipulating the gut microbiome may represent effective treatment for the prevention or management of UC. In addition, the main clinical drugs to treat UC are amino salicylate and corticosteroid. These clinical drugs always have some side effects and low success rate when treating patients with UC. Therefore, there is an urgent need for safe and efficient methods to treat UC. Based on this, probiotics and prebiotics may be a valuable treatment for UC. In order to promote the wide clinical application of probiotics and prebiotics in the treatment of UC. This review aims to summarize the recent literature as an aid to better understanding how the probiotics and prebiotics contributes to UC while evaluating and prospecting the therapeutic effect of the probiotics and prebiotics in the treatment of UC based on previous publications.


Colitis, Ulcerative , Gastrointestinal Microbiome , Prebiotics , Probiotics , Humans , Colitis, Ulcerative/therapy , Colitis, Ulcerative/microbiology , Probiotics/therapeutic use , Probiotics/administration & dosage , Prebiotics/administration & dosage , Animals
7.
Sci Rep ; 14(1): 10960, 2024 05 14.
Article En | MEDLINE | ID: mdl-38744950

The relationship between gut microbiota and obesity has recently been an important subject for research as the gut microbiota is thought to affect body homeostasis including body weight and composition, intervening with pro and prebiotics is an intelligent possible way for obesity management. To evaluate the effect of hypo caloric adequate fiber regimen with probiotic supplementation and physical exercise, whether it will have a good impact on health, body composition, and physique among obese Egyptian women or has no significant effect. The enrolled 58 women, in this longitudinal follow-up intervention study; followed a weight loss eating regimen (prebiotic), including a low-carbohydrate adequate-fiber adequate-protein dietary pattern with decreased energy intake. They additionally received daily probiotic supplements in the form of yogurt and were instructed to exercise regularly for 3 months. Anthropometric measurements, body composition, laboratory investigations, and microbiota analysis were obtained before and after the 3 months weight loss program. Statistically highly significant differences in the anthropometry, body composition parameters: and obesity-related biomarkers (Leptin, ALT, and AST) between the pre and post-follow-up measurements at the end of the study as they were all decreased. The prebiotic and probiotic supplementation induced statistically highly significant alterations in the composition of the gut microbiota with increased relative abundance of Lactobacillus, Bifidobacteria, and Bacteroidetes and decreased relative abundance of Firmicutes and Firmicutes/Bacteroidetes Ratio. Hypo caloric adequate fiber regimen diet with probiotics positively impacts body composition and is effective for weight loss normalizing serum Leptin and AST.


Body Composition , Gastrointestinal Microbiome , Obesity , Prebiotics , Probiotics , Humans , Probiotics/administration & dosage , Female , Prebiotics/administration & dosage , Adult , Longitudinal Studies , Obesity/therapy , Obesity/diet therapy , Obesity/microbiology , Weight Reduction Programs/methods , Weight Loss , Middle Aged , Exercise
8.
Nutrients ; 16(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38674901

The consumption of functional foods in a daily diet is a promising approach for the maintenance of cognitive health. The present study examines the effects of water-soluble prebiotic dietary-fiber, partially hydrolyzed guar gum (PHGG), on cognitive function and mental health in healthy elderly individuals. Participants consumed either 5 g/day of PHGG or a placebo daily for 12 weeks in this randomized, double-blind, placebo-controlled, and parallel-group study. An assessment of cognitive functions, sleep quality, and subjective mood evaluations was performed at baseline and after 8 and 12 weeks of either PHGG or placebo intake. The visual memory scores in cognitive function tests and sleepiness on rising scores related to sleep quality were significantly improved in the PHGG group compared to the placebo group. No significant differences were observed in mood parameters between the groups. Vigor-activity scores were significantly improved, while the scores for Confusion-Bewilderment decreased significantly in the PHGG group when compared to the baseline. In summary, supplementation with PHGG was effective in improving cognitive functions, particularly visual memory, as well as enhancing sleep quality and vitality in healthy elderly individuals (UMIN000049070).


Cognition , Galactans , Mannans , Plant Gums , Humans , Galactans/pharmacology , Mannans/pharmacology , Mannans/administration & dosage , Plant Gums/pharmacology , Double-Blind Method , Cognition/drug effects , Aged , Male , Female , Sleep/drug effects , Prebiotics/administration & dosage , Sleep Quality , Dietary Fiber/pharmacology , Dietary Fiber/administration & dosage , Hydrolysis , Memory/drug effects , Dietary Supplements , Middle Aged , Healthy Volunteers , Affect/drug effects
9.
Expert Rev Anti Infect Ther ; 22(5): 297-306, 2024 May.
Article En | MEDLINE | ID: mdl-38676422

INTRODUCTION: Strong clinical data demonstrate that inflammatory bowel disease (IBD) is an independent risk factor for Clostridiodes difficile infection (CDI) and suggest a globally increased prevalence and severity of C. difficile coinfection in IBD patients (CDI-IBD). In addition to elderly individuals, children are also at higher risk of CDI-IBD. Rapid diagnosis is essential since the clinical manifestations of active IBD and CDI-IBD are indistinguishable. Antibiotics have been well established in the treatment of CDI-IBD, but they do not prevent recurrence. AREAS COVERED: Herein, the authors focus on reviewing recent research advances on the new therapies of CDI-IBD. The novel therapies include gut microbiota restoration therapies (such as prebiotics, probiotics and FMT), immunotherapy (such as vaccines and monoclonal antibodies) and diet strategies (such as groningen anti-inflammatory diet and mediterranean diet). Future extensive prospective and placebo-controlled studies are required to evaluate their efficacy and long-term safety. EXPERT OPINION: Available studies show that the prevalence of CDI-IBD is not optimistic. Currently, potential treatment options for CDI-IBD include a number of probiotics and novel antibiotics. This review updates the knowledge on the management of CDI in IBD patients, which is timely and important for GI doctors and scientists.


Anti-Bacterial Agents , Clostridium Infections , Inflammatory Bowel Diseases , Probiotics , Humans , Clostridium Infections/therapy , Clostridium Infections/microbiology , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/microbiology , Probiotics/administration & dosage , Anti-Bacterial Agents/administration & dosage , Risk Factors , Fecal Microbiota Transplantation , Clostridioides difficile/isolation & purification , Gastrointestinal Microbiome , Prebiotics/administration & dosage , Coinfection , Immunotherapy/methods , Child , Prevalence , Severity of Illness Index , Age Factors , Aged
10.
Calcif Tissue Int ; 114(5): 513-523, 2024 May.
Article En | MEDLINE | ID: mdl-38656326

Previously, we demonstrated that prebiotics may provide a complementary strategy for increasing calcium (Ca) absorption in adolescents which may improve long-term bone health. However, not all children responded to prebiotic intervention. We determine if certain baseline characteristics of gut microbiome composition predict prebiotic responsiveness. In this secondary analysis, we compared differences in relative microbiota taxa abundance between responders (greater than or equal to 3% increase in Ca absorption) and non-responders (less than 3% increase). Dual stable isotope methodologies were used to assess fractional Ca absorption at the end of crossover treatments with placebo, 10, and 20 g/day of soluble corn fiber (SCF). Microbial DNA was obtained from stool samples collected before and after each intervention. Sequencing of the 16S rRNA gene was used to taxonomically characterize the gut microbiome. Machine learning techniques were used to build a predictive model for identifying responders based on baseline relative taxa abundances. Model output was used to infer which features contributed most to prediction accuracy. We identified 19 microbial features out of the 221 observed that predicted responsiveness with 96.0% average accuracy. The results suggest a simplified prescreening can be performed to determine if a subject's bone health may benefit from a prebiotic. Additionally, the findings provide insight and prompt further investigation into the metabolic and genetic underpinnings affecting calcium absorption during pubertal bone development.


Calcium , Gastrointestinal Microbiome , Prebiotics , Adolescent , Child , Female , Humans , Male , Calcium/metabolism , Cross-Over Studies , Feces/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Microbiome/genetics , Pilot Projects , Prebiotics/administration & dosage
11.
Gut Microbes ; 16(1): 2347021, 2024.
Article En | MEDLINE | ID: mdl-38685762

Inulin, an increasingly studied dietary fiber, alters intestinal microbiota. The aim of this study was to assess whether inulin decreases intestinal colonization by multidrug resistant E. coli and to investigate its potential mechanisms of action. Mice with amoxicillin-induced intestinal dysbiosis mice were inoculated with extended spectrum beta-lactamase producing E. coli (ESBL-E. coli). The combination of inulin and pantoprazole (IP) significantly reduced ESBL-E. coli fecal titers, whereas pantoprazole alone did not and inulin had a delayed and limited effect. Fecal microbiome was assessed using shotgun metagenomic sequencing and qPCR. The efficacy of IP was predicted by increased abundance of 74 taxa, including two species of Adlercreutzia. Preventive treatments with A. caecimuris or A. muris also reduced ESBL-E. coli fecal titers. Fecal microbiota of mice effectively treated by IP was enriched in genes involved in inulin catabolism, production of propionate and expression of beta-lactamases. They also had increased beta-lactamase activity and decreased amoxicillin concentration. These results suggest that IP act through production of propionate and degradation of amoxicillin by the microbiota. The combination of pantoprazole and inulin is a potential treatment of intestinal colonization by multidrug-resistant E. coli. The ability of prebiotics to promote propionate and/or beta-lactamase producing bacteria may be used as a screening tool to identify potential treatments of intestinal colonization by multidrug resistant Enterobacterales.


Amoxicillin , Drug Resistance, Multiple, Bacterial , Escherichia coli , Feces , Gastrointestinal Microbiome , Inulin , Pantoprazole , Animals , Inulin/pharmacology , Inulin/metabolism , Mice , Gastrointestinal Microbiome/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Feces/microbiology , Amoxicillin/pharmacology , Pantoprazole/pharmacology , beta-Lactamases/metabolism , beta-Lactamases/genetics , Dysbiosis/microbiology , Dysbiosis/drug therapy , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Female , Prebiotics/administration & dosage
12.
Gut Microbes ; 16(1): 2338946, 2024.
Article En | MEDLINE | ID: mdl-38656273

Synbiotics combine the concepts of probiotics and prebiotics to synergistically enhance the health-associated effects of both components. Previously, we have shown that the intestinal persistence of inulin-utilizing L. plantarum Lp900 is significantly increased in rats fed an inulin-supplemented, high-calcium diet. Here we employed a competitive population dynamics approach to demonstrate that inulin and GOS can selectively enrich L. plantarum strains that utilize these substrates for growth during in vitro cultivation, but that such enrichment did not occur during intestinal transit in rats fed a GOS or inulin-supplemented diet. The intestinal persistence of all L. plantarum strains increased irrespective of their prebiotic utilization phenotype, which was dependent on the calcium level of the diet. Analysis of fecal microbiota and intestinal persistence decline rates indicated that prebiotic utilization capacity did not selectively stimulate intestinal persistence in prebiotic supplemented diets. Moreover, microbiota and organic acid profile analyses indicate that the prebiotic utilizing probiotic strains are vastly outcompeted by the endogenous prebiotic-utilizing microbiota, and that the collective enhanced persistence of all L. plantarum strains is most likely explained by their well-established tolerance to organic acids.


Feces , Gastrointestinal Microbiome , Inulin , Prebiotics , Animals , Prebiotics/administration & dosage , Inulin/metabolism , Inulin/administration & dosage , Rats , Feces/microbiology , Lactobacillus plantarum/metabolism , Lactobacillus plantarum/physiology , Male , Probiotics/administration & dosage , Synbiotics/administration & dosage , Rats, Sprague-Dawley
13.
Braz J Med Biol Res ; 57: e13205, 2024.
Article En | MEDLINE | ID: mdl-38656071

Acute diarrhea is the second leading cause of morbidity and mortality attributed to infections in children under five years of age worldwide, with 1.7 million annual estimated cases and more than 500,000 deaths. Although hydroelectrolytic replacement is the gold standard in treating diarrhea, it does not interfere with the restoration of the intestinal microbiota. Several studies have searched for an adequate alternative in restructuring intestinal homeostasis, finding that treatments based on probiotics, prebiotics, and synbiotics are effective, which made such treatments increasingly present in clinical practice by reducing illness duration with minimal side effects. However, there are still controversies regarding some unwanted reactions in patients. The diversity of strains and the peculiarities of the pathogens that cause diarrhea require further studies to develop effective protocols for prevention and treatment. Here, we provide a descriptive review of childhood diarrhea, emphasizing treatment with probiotics, prebiotics, and synbiotics.


Diarrhea , Prebiotics , Probiotics , Synbiotics , Humans , Probiotics/therapeutic use , Synbiotics/administration & dosage , Prebiotics/administration & dosage , Diarrhea/microbiology , Diarrhea/therapy , Diarrhea/prevention & control , Child , Gastrointestinal Microbiome/physiology , Child, Preschool
14.
J Microbiol ; 62(3): 201-216, 2024 Mar.
Article En | MEDLINE | ID: mdl-38635003

The application of microbiome-based therapies in various areas of human disease has recently increased. In chronic respiratory disease, microbiome-based clinical applications are considered compelling options due to the limitations of current treatments. The lung microbiome is ecologically dynamic and affected by various conditions, and dysbiosis is associated with disease severity, exacerbation, and phenotype as well as with chronic respiratory disease endotype. However, it is not easy to directly modulate the lung microbiome. Additionally, studies have shown that chronic respiratory diseases can be improved by modulating gut microbiome and administrating metabolites. Although the composition, diversity, and abundance of the microbiome between the gut and lung are considerably different, modulation of the gut microbiome could improve lung dysbiosis. The gut microbiome influences that of the lung via bacterial-derived components and metabolic degradation products, including short-chain fatty acids. This phenomenon might be associated with the cross-talk between the gut microbiome and lung, called gut-lung axis. There are multiple alternatives to modulate the gut microbiome, such as prebiotics, probiotics, and postbiotics ingestion and fecal material transplantation. Several studies have shown that high-fiber diets, for example, present beneficial effects through the production of short-chain fatty acids. Additionally, genetically modified probiotics to secrete some beneficial molecules might also be utilized to treat chronic respiratory diseases. Further studies on microbial modulation to regulate immunity and potentiate conventional pharmacotherapy will improve microbiome modulation techniques, which will develop as a new therapeutic area in chronic respiratory diseases.


Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Probiotics , Humans , Probiotics/administration & dosage , Probiotics/therapeutic use , Dysbiosis/therapy , Dysbiosis/microbiology , Lung/microbiology , Chronic Disease , Prebiotics/administration & dosage , Microbiota , Animals , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics
15.
Crit Rev Oncol Hematol ; 197: 104328, 2024 May.
Article En | MEDLINE | ID: mdl-38490281

In recent years, cancer research has highlighted the role of disrupted microbiota in carcinogenesis and cancer recurrence. However, microbiota may also interfere with drug metabolism, influencing the efficacy of cancer drugs, especially immunotherapy, and modulating the onset of adverse events. Intestinal micro-organisms can be altered by external factors, such as use of antibiotics, proton pump inhibitors treatment, lifestyle and the use of prebiotics or probiotics. The aim of our review is to provide a picture of the current evidence about preclinical and clinical data of the role of gut and local microbiota in malignancies and its potential clinical role in cancer treatments. Standardization of microbiota sequencing approaches and its modulating strategies within prospective clinical trials could be intriguing for two aims: first, to provide novel potential biomarkers both for early cancer detection and for therapeutic effectiveness; second, to propose personalized and "microbiota-tailored" treatment strategies.


Gastrointestinal Microbiome , Neoplasms , Humans , Gastrointestinal Microbiome/drug effects , Neoplasms/microbiology , Neoplasms/therapy , Probiotics/therapeutic use , Prebiotics/administration & dosage , Microbiota/drug effects
16.
Infect Disord Drug Targets ; 24(5): e160124225675, 2024.
Article En | MEDLINE | ID: mdl-38317473

BACKGROUND: A potential limelight is flashed on the Gut Microbiota (GM) in the human body, which confers additional psychological as well as physiological attributes to health. Other than just occupying a wide portion of the gastrointestinal tract, it also plays numerous functions in the systems of the body. Gut Microbiota is largely responsible for a considerably vast array of conditions such as obesity, diabetes ,other metabolic disorders, and cardiovascular disorders. Strategies targeting the gut microbiota have been proposed as a promising approach for the management of these disorders. OBJECTIVE: This review aims to summarize the different strategies targeting the gut microbiota for the management of several disorders and to highlight the importance of a sustainable approach. METHODS: A comprehensive literature search was conducted using various databases between 2008 and 2022 that focused on the use of prebiotics, probiotics, synbiotics, postbiotics, fecal microbiota transplantation, dietary interventions, and antibiotics. RESULTS: Different strategies targeting the gut microbiota for the management of several disorders were identified, including probiotics, prebiotics, synbiotics, postbiotics, fecal microbiota transplantation, and dietary interventions. Modification in diet and lifestyle, allowing favorable microbiota growth in the stomach, intake of prebiotics and probiotics, and fecal microbiota transplantation are amongst the widely accepted recent approaches allowing the application of GM in the field of treatment. CONCLUSION: Although considerable steps in enhancing and understanding the mechanism of treatment with the help of gut microbiota are under progress, much diversified and elaborate research must be conducted in order to enhance and implement the use of GM with high effectiveness.


Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Prebiotics , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Probiotics/therapeutic use , Prebiotics/administration & dosage , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Synbiotics/administration & dosage , Gastrointestinal Tract/microbiology , Obesity/microbiology , Obesity/therapy
17.
Reprod Sci ; 31(6): 1508-1520, 2024 Jun.
Article En | MEDLINE | ID: mdl-38228976

Polycystic ovary syndrome (PCOS) is a multifaceted disease with an intricate etiology affecting reproductive-aged women. Despite attempts to unravel the pathophysiology, the molecular mechanism of PCOS remains unknown. There are no effective or suitable therapeutic strategies available to ameliorate PCOS; however, the symptoms can be managed. In recent years, a strong association has been found between the gut microbiome and PCOS, leading to the formulation of novel ideas on the genesis and pathological processes of PCOS. Further, gut microbiome dysbiosis involving microbial metabolites may trigger PCOS symptoms via many mechanistic pathways including those associated with carbohydrates, short-chain fatty acids, lipopolysaccharides, bile acids, and gut-brain axis. We present the mechanistic pathways of PCOS-related microbial metabolites and therapeutic opportunities available to treat PCOS, such as prebiotics, probiotics, and fecal microbiota therapy. In addition, the current review highlights the emerging treatment strategies available to alleviate the symptoms of PCOS.


Dysbiosis , Gastrointestinal Microbiome , Polycystic Ovary Syndrome , Probiotics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/microbiology , Polycystic Ovary Syndrome/therapy , Humans , Female , Gastrointestinal Microbiome/physiology , Dysbiosis/therapy , Probiotics/therapeutic use , Probiotics/administration & dosage , Prebiotics/administration & dosage , Brain-Gut Axis/physiology , Fecal Microbiota Transplantation , Animals
18.
J Assoc Physicians India ; 71(9): 75-81, 2023 Sep.
Article En | MEDLINE | ID: mdl-38700306

The human gut microbiota fosters the development of a dynamic group of microorganisms impacted by diverse variables that include genetics, diet, infection, stress, ingested drugs, such as antibiotics and small intestine bacterial overgrowth (SIBO) as well as the gut microbiota itself. These factors may influence the change in microbial composition, which results in dysbiosis (microbial imbalance) and exposes the gut to pathogenic insults. Dysbiosis is incidental to the etiology of inflammatory diseases such as irritable bowel syndrome (IBS) and metabolic diseases, including type 2 diabetes and obesity. IBS exhibits different symptoms like abdominal pain or discomfort, distention/bloating, and flatulence. To treat IBS, modification of dysregulated gut microbiota can be done using treatment strategies like a low-fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet, antibiotics that cannot be absorbed like rifaximin and neomycin, probiotics and prebiotics, and fecal microbiota transplantation (FMT). The remedial modalities in the existing literature have been demonstrated to be efficacious in the prevention and mitigation of IBS. Additionally, newer curative approaches with serum-derived bovine immunoglobulin (SBI) are an effective option. The focal point of the review paper is the pathophysiology of IBS, mainly due to dysbiosis and the various factors that advance dysbiosis. Here, we have also discussed the different treatment strategies targeting dysbiosis that effectively treat IBS. How to cite this article: Abraham P, Pratap N. Dysbiosis in Irritable Bowel Syndrome. J Assoc Physicians India 2023;71(9):75-81.


Dysbiosis , Gastrointestinal Microbiome , Irritable Bowel Syndrome , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/therapy , Dysbiosis/therapy , Humans , Gastrointestinal Microbiome/physiology , Anti-Bacterial Agents/therapeutic use , Probiotics/therapeutic use , Fecal Microbiota Transplantation/methods , Prebiotics/administration & dosage
19.
Nutr. hosp ; 39(3): 663-677, may. - jun. 2022. ilus, tab, graf
Article Es | IBECS | ID: ibc-209949

Antecedentes: los trastornos gastrointestinales (TGI) son comorbilidades comunes en los pacientes con trastornos del espectro autista (TEA); los tratamientos con dietas libres de gluten y caseína (LGLC) o suplementos de prebióticos/probióticos podrían reducir la severidad de los TGI. Objetivo: integrar y discutir la evidencia sobre la efectividad de las terapias con dietas LGLC y suplementos de prebióticos/probióticos sobre los TGI en pacientes con TEA. Metodología: se utilizaron las guías para la publicación de revisiones sistemáticas y metaanálisis (PRISMA). Se analizaron las características de los participantes, las intervenciones dietéticas, la administración de suplementos de prebióticos/prebióticos, los efectos de las intervenciones sobre los TGI, el riesgo de sesgo de los estudios y la seguridad de los tratamientos. Resultados: se analizaron quince investigaciones; la prevalencia de los TGI entre los pacientes con TEA fue alta (58 %; rango, 27-83 %). En más del 20 % de los pacientes intervenidos con dietas LGLC o suplementos disminuyó la severidad de los TGI (principalmente estreñimiento, diarrea y dolor abdominal). Se reportaron aumentos en los conteos de bacterias benéficas y una disminución de la proporción de bacterias patógenas tras el uso de los suplementos. Sin embargo, todas estas investigaciones presentaron sesgos metodológicos importantes. Conclusiones: aunque se han encontrado reducciones en la frecuencia y severidad de algunos TGI, la efectividad de estos tratamientos aún no se ha comprobado. Dadas las diferencias metodológicas de las investigaciones, se justifica el diseño de estudios rigurosos para evaluar los efectos terapéuticos de estos tratamientos sobre la salud gastrointestinal en pacientes con TEA (AU)


Background: gastrointestinal disorders (GIDs) are common comorbidities in patients with autism spectrum disorders (ASD); treatments with gluten- and casein-free (LGLC) diets or prebiotic/probiotic supplements may reduce the severity of GIDs. Objective: to integrate and discuss the evidence on the effectiveness of LGLC diet therapies and prebiotic/probiotic supplements on GIDs in patients with ASD. Methodology: the guidelines for the publication of systematic reviews and meta-analyses (PRISMA) were used. Participant characteristics, dietary interventions, prebiotic/prebiotic supplementation, effects of interventions on GIDs, risk of bias, and safety of treatments were analyzed. Results: fifteen investigations were analyzed; the prevalence of GIDs among patients with ASD was high (58 %; range, 27-83 %). In more than 20 % of the patients managed with LGLC diets or supplements GID severity decreased (mainly constipation, diarrhea, and abdominal pain). Increases in the counts of beneficial bacteria and a decrease in the proportion of pathogenic bacteria were reported after supplement use. However, all these investigations had significant methodological biases. Conclusions: although reductions in the frequency and severity of some GIDs have been found, the effectiveness of these treatments has not been proven yet. Given the methodological differences in the investigations, the design of rigorous studies to evaluate the therapeutic effects of these treatments on gastrointestinal health in patients with ASD is warranted (AU)


Humans , Autism Spectrum Disorder/complications , Gastrointestinal Diseases/diet therapy , Gastrointestinal Diseases/etiology , Functional Food , Prebiotics/administration & dosage , Probiotics/administration & dosage
20.
Pharm Biol ; 60(1): 437-450, 2022 Dec.
Article En | MEDLINE | ID: mdl-35188051

CONTEXT: Ocimum sanctum Linn (Labiatae) (OS), Zingiber officinale Rose (Zingiberaceae) (ZO), and Piper nigrum Linn (Piperaceae) (PN) are used in traditional medicine as immunomodulator, anti-inflammatory, and bioavailability enhancer agents. OBJECTIVE: Active phytoconstituents of OS, ZO, PN hydro-alcoholic extracts and their effects on gut microbiota, basal inflammation and lipid profile were investigated in rats. MATERIALS AND METHODS: Active phytoconstituents of extracts were analysed using HPLC and GC-MS. SD rats were supplemented with individual/combined extracts (OS-850; ZO-500; PN-100 mg/kg Bw) and Fructooligosaccharide (standard prebiotic-5g/kg-Bw), orally for 30 days. Haematology, lipid profile, LPS, CRP, IL-6, insulin and histology of vital organs were analysed. Caecal bacterial levels were assessed by RT-PCR. RESULTS: High content of phenolic compounds luteolin-7-O-glucoside (430 ± 2.3 mg/100g), gallic acid (84.13 ± 1.2 mg/100 g) and flavones (88.18 ± 1.8 mg/100 g) were found in OS, ZO, and PN, respectively. Combined extract was rich in luteolin-7-O-glucoside (266.0 ± 1.80 mg/100 g). Essential oils including methyleugenol (13.96%), 6-shogaol (11.00%), piperine (18.26%), and cyclopentasiloxane (10.06%) were higher in OS, ZO, PN and combined extract. Higher levels of caecal Lactobacillus (1.7-3.4-fold), Bifidobacterium (5.89-28.4-fold), and lower levels of Firmicutes (0.04-0.91-fold), Bacteroides (0.69-0.88-fold) were noted among extracts and FOS supplemented rats. Significant (p < 0.05) decrease in plasma lipid profile and LPS was noted in all supplemented rats. DISCUSSION AND CONCLUSIONS: The current study could be first of its kind in exploring prebiotic potential of OS, ZO, PN and their effect on native gut bacterial population.


Gastrointestinal Microbiome/drug effects , Inflammation/drug therapy , Oils, Volatile/pharmacology , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Female , Zingiber officinale/chemistry , Lipids/blood , Medicine, Traditional , Ocimum sanctum/chemistry , Oils, Volatile/isolation & purification , Piper nigrum/chemistry , Prebiotics/administration & dosage , Rats , Rats, Sprague-Dawley
...