Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.811
Filter
1.
Carbohydr Polym ; 344: 122535, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39218555

ABSTRACT

Oat ß-(1 â†’ 3, 1 â†’ 4)-d-glucan (OBG), a linear polysaccharide primarily found in oat bran, has been demonstrated to possess immunomodulatory properties and regulate gut microbiota. This study aimed to investigate the impact of low molecular weight (Mw) OBG (155.2 kDa) on colonic injury and allergic symptoms induced by food allergy (FA), and to explore its potential mechanism. In Experiment 1, results indicated that oral OBG improved colonic inflammation and epithelial barrier, and significantly relieved allergy symptoms. Importantly, the OBG supplement altered the gut microbiota composition, particularly increasing the abundance of Lachnospiraceae and its genera, and promoted the production of short-chain fatty acids, especially butyrate. However, in Experiment 2, the gut microbial depletion eliminated these protective effects of OBG on the colon in allergic mice. Further, in Experiment 3, fecal microbiota transplantation and sterile fecal filtrate transfer directly validated the role of OBG-mediated gut microbiota and its metabolites in relieving FA and its induced colonic injury. Our findings suggest that low Mw OBG can alleviate FA-induced colonic damage by increasing Lachnospiraceae abundance and butyrate production, and provide novel insights into the health benefits and mechanisms of dietary polysaccharide intervention for FA.


Subject(s)
Avena , Butyrates , Colon , Food Hypersensitivity , Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/drug effects , Mice , Colon/pathology , Colon/drug effects , Colon/metabolism , Butyrates/metabolism , Avena/chemistry , Clostridiales , beta-Glucans/pharmacology , beta-Glucans/chemistry , Mice, Inbred BALB C , Male , Glucans/pharmacology , Glucans/chemistry , Fatty Acids, Volatile/metabolism , Fecal Microbiota Transplantation
2.
J Nutr Sci Vitaminol (Tokyo) ; 70(4): 311-317, 2024.
Article in English | MEDLINE | ID: mdl-39218692

ABSTRACT

Chronic inflammation in adipose tissue is thought to contribute to insulin resistance, which involves the gut microbiota. Our previous studies have demonstrated that ingestion of 1-kestose can alter the gut microbiota composition, increase cecal butyrate levels, and improve insulin resistance in Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Additionally, we found that 1-kestose supplementation ameliorated insulin resistance in obese rat models fed a high-fat diet (HFD), although the effects of 1-kestose on the abundance of inflammation-related gene in adipose tissue and gut microbiota composition in these rats were not explored. This study aimed to investigate the impact of 1-kestose on these parameters in HFD-fed rats, compared to OLETF rats. Male Sprague-Dawley rats were divided into two dietary groups, control or HFD, for 19 wk. Each group was further subdivided to receive either tap water or tap water supplemented with 2% (w/v) 1-kestose throughout the study. We evaluated gene expression in adipose tissue, as well as short-chain fatty acids (SCFAs) levels and microbial composition in the cecum contents. 1-Kestose intake restored the increased relative abundance of tumor necrosis factor (Tnf) mRNA in adipose tissue and the reduced level of butyrate in the cecum contents of HFD-fed rats to those observed in control diet-fed rats. Additionally, 1-kestose consumption changed the composition of the gut microbiota, increasing Butyricicoccus spp., decreasing UGC-005 and Streptococcus spp., in the cecum contents of HFD-fed rats. Our findings suggest that 1-kestose supplementation reduces adipose tissue inflammation and increases butyrate levels in the gut of HFD-fed rats, associated with changes in the gut microbiota composition, distinct from those seen in OLETF rats.


Subject(s)
Adipose Tissue , Cecum , Diet, High-Fat , Fatty Acids, Volatile , Gastrointestinal Microbiome , Inflammation , RNA, Messenger , Rats, Sprague-Dawley , Animals , Gastrointestinal Microbiome/drug effects , Male , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Inflammation/metabolism , RNA, Messenger/metabolism , Rats , Fatty Acids, Volatile/metabolism , Cecum/microbiology , Cecum/metabolism , Insulin Resistance , Rats, Inbred OLETF , Obesity/metabolism , Obesity/microbiology , Dietary Supplements , Butyrates/metabolism
3.
Theranostics ; 14(12): 4622-4642, 2024.
Article in English | MEDLINE | ID: mdl-39239516

ABSTRACT

Rationale: Consumption of a high-fat diet (HFD) has been implicated in cognitive deficits and gastrointestinal dysfunction in humans, with the gut microbiota emerging as a pivotal mediator of these diet-associated pathologies. The introduction of plant-based polysaccharides into the diet as a therapeutic strategy to alleviate such conditions is gaining attention. Nevertheless, the mechanistic paradigm by which polysaccharides modulate the gut microbiota remains largely undefined. This study investigated the mechanisms of action of Eucommiae cortex polysaccharides (EPs) in mitigating gut dysbiosis and examined their contribution to rectifying diet-related cognitive decline. Methods: Initially, we employed fecal microbiota transplantation (FMT) and gut microbiota depletion to verify the causative role of changes in the gut microbiota induced by HFD in synapse engulfment-dependent cognitive impairments. Subsequently, colonization of the gut of chow-fed mice with Escherichia coli (E. coli) from HFD mice confirmed that inhibition of Proteobacteria by EPs was a necessary prerequisite for alleviating HFD-induced cognitive impairments. Finally, supplementation of HFD mice with butyrate and treatment of EPs mice with GW9662 demonstrated that EPs inhibited the expansion of Proteobacteria in the colon of HFD mice by reshaping the interactions between the gut microbiota and colonocytes. Results: Findings from FMT and antibiotic treatments demonstrated that HFD-induced cognitive impairments pertaining to neuronal spine loss were contingent on gut microbial composition. Association analysis revealed strong associations between bacterial taxa belonging to the phylum Proteobacteria and cognitive performance in mice. Further, introducing E. coli from HFD-fed mice into standard diet-fed mice underscored the integral role of Proteobacteria proliferation in triggering excessive synaptic engulfment-related cognitive deficits in HFD mice. Crucially, EPs effectively counteracted the bloom of Proteobacteria and subsequent neuroinflammatory responses mediated by microglia, essential for cognitive improvement in HFD-fed mice. Mechanistic insights revealed that EPs promoted the production of bacteria-derived butyrate, thereby ameliorating HFD-induced colonic mitochondrial dysfunction and reshaping colonocyte metabolism. This adjustment curtailed the availability of growth substrates for facultative anaerobes, which in turn limited the uncontrolled expansion of Proteobacteria. Conclusions: Our study elucidates that colonocyte metabolic disturbances, which promote Proteobacteria overgrowth, are a likely cause of HFD-induced cognitive deficits. Furthermore, dietary supplementation with EPs can rectify behavioral dysfunctions associated with HFD by modifying gut microbiota-colonocyte interactions. These insights contribute to the broader understanding of the modulatory effects of plant prebiotics on the microbiota-gut-brain axis and suggest a potential therapeutic avenue for diet-associated cognitive dysfunction.


Subject(s)
Cognitive Dysfunction , Diet, High-Fat , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Mice, Inbred C57BL , Polysaccharides , Gastrointestinal Microbiome/drug effects , Animals , Diet, High-Fat/adverse effects , Mice , Cognitive Dysfunction/therapy , Polysaccharides/pharmacology , Male , Dysbiosis/therapy , Colon/microbiology , Escherichia coli , Butyrates/metabolism , Proteobacteria/isolation & purification , Proteobacteria/drug effects , Disease Models, Animal
4.
Microb Biotechnol ; 17(9): e70006, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39235453

ABSTRACT

Feedstock variability represents a challenge in lignocellulosic biorefineries, as it can influence both lignocellulose deconstruction and microbial conversion processes for biofuels and biochemicals production. The impact of feedstock variability on microbial performance remains underexplored, and predictive tools for microbial behaviour are needed to mitigate risks in biorefinery scale-up. Here, twelve batches of corn stover were deconstructed via deacetylation, mechanical refining, and enzymatic hydrolysis to generate lignin-rich and sugar streams. These batches and their derived streams were characterised to identify their chemical components, and the streams were used as substrates for producing muconate and butyrate by engineered Pseudomonas putida and wildtype Clostridium tyrobutyricum, respectively. Bacterial performance (growth, product titers, yields, and productivities) differed among the batches, but no strong correlations were identified between feedstock composition and performance. To provide metabolic insights into the origin of these differences, we evaluated the effect of twenty-three isolated chemical components on these microbes, including three components in relevant bioprocess settings in bioreactors, and we found that growth-inhibitory concentrations were outside the ranges observed in the streams. Overall, this study generates a foundational dataset on P. putida and C. tyrobutyricum performance to enable future predictive models and underscores their resilience in effectively converting fluctuating lignocellulose-derived streams into bioproducts.


Subject(s)
Clostridium tyrobutyricum , Lignin , Metabolic Engineering , Pseudomonas putida , Zea mays , Pseudomonas putida/genetics , Pseudomonas putida/metabolism , Lignin/metabolism , Zea mays/microbiology , Clostridium tyrobutyricum/metabolism , Clostridium tyrobutyricum/genetics , Biotransformation , Bioreactors/microbiology , Sugars/metabolism , Butyrates/metabolism
5.
Nutrients ; 16(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125336

ABSTRACT

Postbiotics could exert different metabolic activities in animal models of non-alcoholic fatty liver disease (NAFLD) and in humans affected by metabolic syndrome. This is a randomized, double-blind, placebo-controlled, parallel-group clinical trial that enrolled a sample of 50 Caucasian healthy individuals with NAFLD, defined as liver steatosis, and metabolic syndrome. After a 4-week run-in, the enrolled individuals were randomized to take a food for special medical purposes with functional release, one tablet a day, containing calcium butyrate (500 mg/tablet), zinc gluconate (zinc 5 mg/tablet), and vitamin D3 (500 IU/tablet), or an identical placebo for 3 months. Liver and metabolic parameters were measured at baseline and at the end of the study. No subject experienced any adverse events during the trial. In both groups, a significant decrease in total cholesterol (TC) and triglycerides (TG) plasma levels was observed at the randomization visit vs. pre-run-in visit (p < 0.05). Regarding liver parameters, after treatment, the fatty liver index (FLI) improved significantly vs. baseline values (p < 0.05) and vs. placebo group (p < 0.05) in the active treatment group, and the hepatic steatosis index (HSI) improved significantly vs. baseline values (p < 0.05). Moreover, after active treatment, TC, TG, and gamma-glutamyl transferase (gGT) improved significantly vs. baseline values (p < 0.05), and TC and TG improved vs. placebo group (p < 0.05), as well. In the placebo group, liver parameters remained unchanged after treatment; only TG improved significantly vs. baseline values (p < 0.05). In our study, we observed that the butyrate-based formula improved FLI and plasma lipid patterns in individuals affected by liver steatosis and metabolic syndrome.


Subject(s)
Butyrates , Dietary Supplements , Metabolic Syndrome , Non-alcoholic Fatty Liver Disease , Humans , Double-Blind Method , Metabolic Syndrome/drug therapy , Male , Female , Middle Aged , Adult , Non-alcoholic Fatty Liver Disease/drug therapy , Triglycerides/blood , Liver/metabolism , Liver/drug effects , Cholesterol/blood , Gluconates/administration & dosage , Treatment Outcome
6.
Cells ; 13(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39120272

ABSTRACT

Sulphated glycosaminoglycans (GAGs) such as heparin are a major component of mast cell granules and form the matrix within which biogenic mediators are stored. Since GAGs released from mast cells also play an important role in helminth expulsion, understanding GAG storage can offer new insights into mast cell function. Sodium butyrate (NaBu), a short-chain fatty acid, causes ultrastructural changes within the granules of human mast cells (HMC-1) and increases their histamine content. Therefore, we hypothesized that NaBu treatment would also modify the storage of polysaccharides such as GAGs. NaBu (1 mM) significantly increased GAG content and granularity in a time- and concentration-dependent manner without affecting cell viability and metabolic activity. NaBu increased the expression of enzymes associated with heparin biosynthesis (GLCE, NDST1, NDST2, HS6ST1, and GALT1) in a time-dependent manner. A cholesteryl butyrate emulsion (CholButE) increased heparin content after 24 and 48 h and modestly altered the expression of genes involved in heparin biosynthesis. Similar to NaBu, CholButE reduced cell proliferation without significantly altering viability or metabolic activity. These data show that butyrate increases the synthesis and storage of heparin in human mast cells, perhaps by altering their metabolic pathways.


Subject(s)
Heparin , Mast Cells , Humans , Mast Cells/metabolism , Mast Cells/drug effects , Heparin/pharmacology , Heparin/metabolism , Cell Survival/drug effects , Butyrates/pharmacology , Butyrates/metabolism , Cell Proliferation/drug effects , Butyric Acid/pharmacology , Cell Line , Cholesterol Esters/metabolism
7.
Microbiologyopen ; 13(4): e1430, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39115291

ABSTRACT

The gut microbiota significantly contributes to human health and well-being. The aim of this study was to evaluate the stability and resilience of a consortium composed of three next-generation probiotics (NGPs) candidates originally found in the human gut. The growth patterns of Akkermansia muciniphila, Bacteroides thetaiotaomicron, and Faecalibacterium prausnitzii were studied both individually and consortium. The growth kinetics of Akkermansia muciniphila (A. muciniphila), Bacteroides thetaiotaomicron (B. thetaiotaomicron), and Faecalibacterium prausnitzii (F. prausnitzii) were characterized both individually and in consortium using isothermal microcalorimetry and 16S ribosomal RNA next-generation sequencing. The consortium reached stability after three passages and demonstrated resilience to changes in its initial composition. The concentration of butyrate produced was nearly twice as high in the consortium compared to the monoculture of F. prausnitzii. The experimental conditions and methodologies used in this article are a solid foundation for developing further complex consortia.


Subject(s)
Calorimetry , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Gastrointestinal Microbiome/physiology , RNA, Ribosomal, 16S/genetics , Faecalibacterium prausnitzii/genetics , Akkermansia/growth & development , Akkermansia/physiology , Microbial Consortia/physiology , Microbial Consortia/genetics , High-Throughput Nucleotide Sequencing , Butyrates/metabolism , Probiotics , Verrucomicrobia/genetics , Verrucomicrobia/growth & development , Bacteroides/genetics , Bacteroides/growth & development , DNA, Bacterial/genetics
8.
Clin Sci (Lond) ; 138(17): 1039-1054, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39136693

ABSTRACT

Maternal high-fat diet intake has profound effects on the long-term health of offspring, predisposing them to a higher susceptibility to obesity and metabolic dysfunction-associated steatotic liver disease. However, the detailed mechanisms underlying the role of a maternal high-fat diet in hepatic lipid accumulation in offspring, especially at the weaning age, remain largely unclear. In this study, female C57BL/6J mice were randomly assigned to either a high-fat diet or a control diet, and lipid metabolism parameters were assessed in male offspring at weaning. Gut microbiota analysis and targeted metabolomics of short-chain fatty acids (SCFAs) in these offspring were further performed. Both in vivo and in vitro studies were conducted to explore the role of butyrate in hepatic cholesterol excretion in the liver and HepG2 cells. Our results showed that maternal high-fat feeding led to obesity and dyslipidemia, and exacerbated hepatic lipid accumulation in the livers of offspring at weaning. We observed significant decreases in the abundance of the Firmicutes phylum and the Allobaculum genus, known as producers of SCFAs, particularly butyrate, in the offspring of dams fed a high-fat diet. Additionally, maternal high-fat diet feeding markedly decreased serum butyrate levels and down-regulated ATP-binding cassette transporters G5 (ABCG5) in the liver, accompanied by decreased phosphorylated AMP-activated protein kinase (AMPK) and histone deacetylase 5 (HADC5) expressions. Subsequent in vitro studies revealed that butyrate could induce ABCG5 activation and alleviate lipid accumulation via the AMPK-pHDAC5 pathway in HepG2 cells. Moreover, knockdown of HDAC5 up-regulated ABCG5 expression and promoted cholesterol excretion in HepG2 cells. In conclusion, our study provides novel insights into how maternal high-fat diet feeding inhibits hepatic cholesterol excretion and down-regulates ABCG5 through the butyrate-AMPK-pHDAC5 pathway in offspring at weaning.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5 , Butyrates , Cholesterol , Diet, High-Fat , Gastrointestinal Microbiome , Liver , Mice, Inbred C57BL , Animals , Diet, High-Fat/adverse effects , Female , Butyrates/metabolism , Humans , Liver/metabolism , Hep G2 Cells , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , Male , Cholesterol/metabolism , Cholesterol/blood , Pregnancy , Mice , Lipid Metabolism , Prenatal Exposure Delayed Effects/metabolism , Maternal Nutritional Physiological Phenomena , Obesity/metabolism , Obesity/microbiology , Dyslipidemias/metabolism , Dyslipidemias/microbiology , Dyslipidemias/etiology , Lipoproteins
9.
Gut Microbes ; 16(1): 2389319, 2024.
Article in English | MEDLINE | ID: mdl-39182227

ABSTRACT

Alterations in the gut-microbiome-brain axis are increasingly being recognized to be involved in Alzheimer's disease (AD) pathogenesis. However, the functional consequences of enteric dysbiosis linking gut microbiota and brain pathology in AD progression remain largely undetermined. The present work investigated the causal role of age-associated temporal decline in butyrate-producing bacteria and butyrate in the etiopathogenesis of AD. Longitudinal metagenomics, neuropathological, and memory analyses were performed in the 3×Tg-AD mouse model. Metataxonomic analyses showed a significant temporal decline in the alpha diversity marked by a decrease in butyrate-producing bacterial communities and a concurrent reduction in cecal butyrate production. Inferred metagenomics analysis identified the bacterial acetyl-CoA pathway as the main butyrate synthesis pathway impacted. Concomitantly, there was an age-associated decline in the transcriptionally permissive acetylation of histone 3 at lysines 9 and 14 (H3K9/K14-Ac) in hippocampal neurons. Importantly, these microbiome-gut-brain changes preceded AD-related neuropathology, including oxidative stress, tau hyperphosphorylation, memory deficits, and neuromuscular dysfunction, which manifest by 17-18 months. Initiation of oral administration of tributyrin, a butyrate prodrug, at 6 months of age mitigated the age-related decline in butyrate-producing bacteria, protected the H3K9/K14-Ac status, and attenuated the development of neuropathological and cognitive changes associated with AD pathogenesis. These data causally implicate age-associated decline in butyrate-producing bacteria as a key pathogenic feature of the microbiome-gut-brain axis affecting the onset and progression of AD. Importantly, the regulation of butyrate-producing bacteria and consequent butyrate synthesis could be a significant therapeutic strategy in the prevention and treatment of AD.


Subject(s)
Alzheimer Disease , Bacteria , Butyrates , Disease Models, Animal , Dysbiosis , Gastrointestinal Microbiome , Memory Disorders , Animals , Butyrates/metabolism , Mice , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Memory Disorders/microbiology , Memory Disorders/metabolism , Memory Disorders/pathology , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Bacteria/isolation & purification , Dysbiosis/microbiology , Hippocampus/metabolism , Hippocampus/pathology , Mice, Transgenic , Male , Disease Progression , Brain-Gut Axis/physiology , Brain/metabolism , Brain/pathology
10.
PLoS One ; 19(8): e0309742, 2024.
Article in English | MEDLINE | ID: mdl-39213333

ABSTRACT

Novel antimicrobial strategies are necessary to tackle using antibiotics during the suckling and weaning period of piglets, often characterized by E. coli-induced diarrhea. In the last decades, acetate, propionate, and butyrate, all short-chain fatty acids (SCFAs), have been proposed as an alternative to antibiotics. SCFAs are instrumental in promoting the proliferation of enterocytes, preserving intestinal integrity, and modulating the microbial community by suppressing the growth of pathogenic bacteria in pigs. The effect of individual SCFAs (proprionate, acetate and butyrate) on the regenerative capacity of intestinal cells was investigated via an optimized wound-healing assay in IPEC-J2 cells, a porcine jejunal epithelial cell line. IPEC-J2 cells proved a good model as they express the free fatty acid receptor 2 (FFAR2), an important SCFA receptor with a high affinity for proprionate. Our study demonstrated that propionate (p = 0.005) and acetate (p = 0.037) were more effective in closing the wound than butyrate (p = 0.190). This holds promise in using SCFA's per os as an alternative to antibiotics.


Subject(s)
Cell Movement , Cell Proliferation , Fatty Acids, Volatile , Animals , Cell Proliferation/drug effects , Swine , Fatty Acids, Volatile/pharmacology , Fatty Acids, Volatile/metabolism , Cell Movement/drug effects , Cell Line , Butyrates/pharmacology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Propionates/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism
11.
Food Res Int ; 192: 114801, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39147503

ABSTRACT

This study aimed to evaluate the effects of fermented beetroot ketchup enriched with Lactobacillus johnsonii K4 and non-fermented beetroot ketchup on pooled fecal microbiota from healthy adults in in vitro colon model. The research focused on how these products influenced the composition and functionality of the gut microbiota, as well as metabolite production, using the validated dynamic in vitro colon model, TNO Intestinal Model (TIM-2). After an initial starvation phase, a single 60 g dose of predigested and freeze-dried ketchup was introduced into the model. The potential probiotic strain Lactobacillus johnsonii K4 was added over three days. A carbohydrate mixture of standard ileal effluent medium (SIEM) served as the control. Our analysis identified 21 bacterial taxa that were significantly modulated (q-value < 0.2) when comparing ketchup samples to control samples. Notably, the ketchup samples led to an increase in butyrate-producing taxa, including Faecalibacterium, Blautia, Ruminococcaceae, Ruminiclostridium 6, and Anaerostipes. Conversely, there was a reduction in potentially pathogenic genera Desulfovibrio and Escherichia-Shigella. Distinct profiles of short-chain fatty acids (SCFA) were observed among the fermented ketchup, non-fermented ketchup, and control samples. Non-fermented ketchup resulted in higher proportions of acetate, propionate, and butyrate compared to the other interventions. This may be related to the fermentation with lactic acid bacteria in fermented samples with lower levels of substrate for SCFAs production. However, fermented ketchup sample has higher relative abundance of beneficial bacteria like Lactobacillus, Weissella and Dorea in gut microbiota. These findings suggest that beetroot ketchup, can positively influence gut microbiota composition and function, highlighting its potential benefits for human health.


Subject(s)
Colon , Fatty Acids, Volatile , Feces , Fermentation , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Colon/microbiology , Colon/metabolism , Humans , Feces/microbiology , Fatty Acids, Volatile/metabolism , Probiotics , Beta vulgaris/microbiology , Beta vulgaris/chemistry , Adult , Lactobacillus/metabolism , Fermented Foods/microbiology , Bacteria/metabolism , Bacteria/classification , Butyrates/metabolism , Male
12.
Int J Mol Sci ; 25(16)2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39201684

ABSTRACT

Parkinson's Disease (PD) is a prevalent neurodegenerative disorder characterized by motor and non-motor symptoms. Emerging evidence suggests that gut microbiota alterations, specifically involving short-chain fatty acids (SCFAs) like butyrate, may influence PD pathogenesis and symptomatology. This Systematic Review aims to synthesize current research on the role of butyrate in modulating motor symptoms and its neuroprotective effects in PD, providing insights into potential therapeutic approaches. A systematic literature search was conducted in April 2024 across databases, including ScienceDirect, Scopus, Wiley, and Web of Science, for studies published between 2000 and 2024. Keywords used were "neuroprotective effects AND butyrate AND (Parkinson disease OR motor symptoms)". Four authors independently screened titles, abstracts, and full texts, applying inclusion criteria focused on studies investigating butyrate regulation and PD motor symptoms. A total of 1377 articles were identified, with 40 selected for full-text review and 14 studies meeting the inclusion criteria. Data extraction was performed on the study population, PD models, methodology, intervention details, and outcomes. Quality assessment using the SYRCLE RoB tool highlighted variability in study quality, with some biases noted in allocation concealment and blinding. Findings indicate that butyrate regulation has a significant impact on improving motor symptoms and offers neuroprotective benefits in PD models. The therapeutic modulation of gut microbiota to enhance butyrate levels presents a promising strategy for PD symptom management.


Subject(s)
Butyrates , Gastrointestinal Microbiome , Neuroprotective Agents , Parkinson Disease , Signal Transduction , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Humans , Butyrates/metabolism , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Signal Transduction/drug effects , Animals
13.
J Clin Psychopharmacol ; 44(5): 457-461, 2024.
Article in English | MEDLINE | ID: mdl-39146178

ABSTRACT

BACKGROUND: Preliminary evidence suggests that people with schizophrenia have decreased relative abundance of butyrate-producing bacteria in the gut microbiota. Butyrate plays a critical role in maintaining the integrity of the gut-blood barrier and has a number of anti-inflammatory effects. This proof-of-concept study was designed to assess whether the addition of the oligofructose-enriched inulin (OEI) prebiotic: Prebiotin could increase the production of butyrate. METHODS: Twenty-seven people who met the criteria for either Diagnostic and Statistical Manual of Mental Disorders, 5th Edition, schizophrenia or schizoaffective disorder were entered into a 10-day, double-blind, placebo-controlled, randomized clinical trial. The study was conducted on an inpatient unit to standardize the participant diet and environment. Participants were randomized to either OEI (4 g, 3 times a day) or a placebo (4 g of maltodextrin, 3 times a day). In order to assess the effect of OEI treatment on butyrate levels, participants underwent pretreatment and posttreatment OEI challenges. The primary outcome measure was relative change in postchallenge plasma butyrate levels after 10 days of OEI treatment. RESULTS: In both the intent-to-treat and completer analyses, OEI treatment was associated with a greater number of participants who met the OEI challenge responder criteria than those treated with placebo. OEI treatment was also associated with an increase in baseline butyrate levels (effect size for the group difference in the change of baseline butyrate levels was 0.58). CONCLUSIONS: We were able to demonstrate that treatment with the prebiotic OEI selectively increased the level of plasma butyrate in people with schizophrenia.Trial registration:ClinicalTrials.gov identifier NCT03617783.


Subject(s)
Butyrates , Oligosaccharides , Prebiotics , Schizophrenia , Humans , Schizophrenia/drug therapy , Schizophrenia/blood , Prebiotics/administration & dosage , Double-Blind Method , Male , Female , Adult , Middle Aged , Oligosaccharides/administration & dosage , Oligosaccharides/pharmacology , Inulin/administration & dosage , Inulin/pharmacology , Gastrointestinal Microbiome/drug effects , Proof of Concept Study , Psychotic Disorders/drug therapy , Psychotic Disorders/diet therapy , Psychotic Disorders/blood , Young Adult
14.
Vet Med Sci ; 10(4): e1538, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38985126

ABSTRACT

BACKGROUND: Selenium (Se) is a rare essential element that plays a vital role in the health and performance of animals. By interfering in the production of antioxidant enzymes such as glutathione peroxidase, thioredoxin reductase and methionine sulfoxide, Se plays a role in reducing the effects of oxidative stress and animal performance. OBJECTIVES: This study aimed to investigate the effect of hydroxy-selenomethionine (OH-SeMet) in the diet of broiler breeder and old broiler breeder roosters on productive performance, reproduction and sperm quality parameters. METHODS: For this purpose, 260 broiler breeders of the Ross 308 strain were used in a completely randomized design with four treatments and five replications (13 hens and one rooster in each replication). Experimental treatments included: (1) a basal diet without OH-SeMet (T1:control), (2) a broiler breeder diet without OH-SeMet and a rooster diet containing 0.1 mg/kg OH-SeMet (T2), (3) broiler breeder diet containing 0.1 mg/kg OH-SeMet and rooster diet without OH-SeMet (T3) and (4) broiler breeder and rooster diet contained 0.1 mg/kg OH-SeMet (T4). RESULTS: The results showed that T3 and T4 treatments improved egg production, egg weight, egg mass and feed conversion ratio (FCR) compared to the control treatment (p < 0.05). The fertility and hatchability percentages of T4 and T2 treatments increased compared to T1 and T3 treatments (p < 0.05). The rate of embryonic losses in T1 was higher than in other treatments. However, grade one chickens were higher in T4 than in other treatments (p < 0.05). Total motility and viability of sperms were significantly higher in T2 and T4 treatments than in T1 and T3 treatments. The sperm abnormality percentage and sperm MDA concentration decreased in T2 and T4 treatments. CONCLUSIONS: Therefore, using OH-SeMet may be a practical approach to help old broiler breeders' production and reproduction performance.


Subject(s)
Animal Feed , Chickens , Diet , Dietary Supplements , Reproduction , Selenomethionine , Animals , Chickens/physiology , Selenomethionine/pharmacology , Selenomethionine/administration & dosage , Diet/veterinary , Male , Animal Feed/analysis , Female , Dietary Supplements/analysis , Reproduction/drug effects , Random Allocation , Butyrates , Selenium Compounds
15.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39000417

ABSTRACT

Metabolites resulting from the bacterial fermentation of dietary fibers, such as short-chain fatty acids, especially butyrate, play important roles in maintaining gut health and regulating various biological effects in the skin. However, butyrate is underutilized due to its unpleasant odor. To circumvent this organoleptic unfavorable property, phenylalanine butyramide (PBA), a butyrate precursor, has been synthesized and is currently available on the market. We evaluated the inhibition of mushroom tyrosinase by butyrate and PBA through in vitro assays, finding IC50 values of 34.7 mM and 120.3 mM, respectively. Docking calculations using a homology model of human tyrosinase identified a putative binding mode of PBA into the catalytic site. The anti-aging and anti-spot efficacy of topical PBA was evaluated in a randomized, double-blind, parallel-arm, placebo-controlled clinical trial involving 43 women affected by photo-damage. The results of this study showed that PBA significantly improved skin conditions compared to the placebo and was well tolerated. Specifically, PBA demonstrated strong skin depigmenting activity on both UV and brown spots (UV: -12.7% and -9.9%, Bs: -20.8% and -17.7% after 15 and 30 days, respectively, p < 0.001). Moreover, PBA brightened and lightened the skin (ITA°: +12% and 13% after 15 and 30 days, respectively, p < 0.001). Finally, PBA significantly improved skin elasticity (Ua/Uf: +12.4% and +32.3% after 15 and 30 days, respectively, p < 0.001) and firmness (Uf: -3.2% and -14.9% after 15 and 30 days, respectively, p < 0.01).


Subject(s)
Monophenol Monooxygenase , Phenylalanine , Skin Aging , Skin Pigmentation , Adult , Female , Humans , Middle Aged , Agaricales/enzymology , Butyrates/chemistry , Butyrates/pharmacology , Double-Blind Method , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Monophenol Monooxygenase/antagonists & inhibitors , Phenylalanine/chemistry , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Skin Aging/drug effects , Skin Pigmentation/drug effects
16.
Lipids Health Dis ; 23(1): 216, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003477

ABSTRACT

BACKGROUND: The regulation of the circadian clock genes, which coordinate the activity of the immune system, is disturbed in inflammatory bowel disease (IBD). Emerging evidence suggests that butyrate, a short-chain fatty acid produced by the gut microbiota is involved in the regulation of inflammatory responses as well as circadian-clock genes. This study was conducted to investigate the effects of sodium-butyrate supplementation on the expression of circadian-clock genes, inflammation, sleep and life quality in active ulcerative colitis (UC) patients. METHODS: In the current randomized placebo-controlled trial, 36 active UC patients were randomly divided to receive sodium-butyrate (600 mg/kg) or placebo for 12-weeks. In this study the expression of circadian clock genes (CRY1, CRY2, PER1, PER2, BMAl1 and CLOCK) were assessed by real time polymerase chain reaction (qPCR) in whole blood. Gene expression changes were presented as fold changes in expression (2^-ΔΔCT) relative to the baseline. The faecal calprotectin and serum level of high-sensitivity C-reactive protein (hs-CRP) were assessed by enzyme-linked immunosorbent assay method (ELIZA). Moreover, the sleep quality and IBD quality of life (QoL) were assessed by Pittsburgh sleep quality index (PSQI) and inflammatory bowel disease questionnaire-9 (IBDQ-9) respectively before and after the intervention. RESULTS: The results showed that sodium-butyrate supplementation in comparison with placebo significantly decreased the level of calprotectin (-133.82 ± 155.62 vs. 51.58 ± 95.57, P-value < 0.001) and hs-CRP (-0.36 (-1.57, -0.05) vs. 0.48 (-0.09-4.77), P-value < 0.001) and upregulated the fold change expression of CRY1 (2.22 ± 1.59 vs. 0.63 ± 0.49, P-value < 0.001), CRY2 (2.15 ± 1.26 vs. 0.93 ± 0.80, P-value = 0.001), PER1 (1.86 ± 1.77 vs. 0.65 ± 0.48, P-value = 0.005), BMAL1 (1.85 ± 0.97 vs. 0.86 ± 0.63, P-value = 0.003). Also, sodium-butyrate caused an improvement in the sleep quality (PSQI score: -2.94 ± 3.50 vs. 1.16 ± 3.61, P-value < 0.001) and QoL (IBDQ-9: 17.00 ± 11.36 vs. -3.50 ± 6.87, P-value < 0.001). CONCLUSION: Butyrate may be an effective adjunct treatment for active UC patients by reducing biomarkers of inflammation, upregulation of circadian-clock genes and improving sleep quality and QoL.


Subject(s)
Colitis, Ulcerative , Dietary Supplements , Sleep Quality , Humans , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Male , Female , Adult , Double-Blind Method , Middle Aged , Inflammation/genetics , Inflammation/drug therapy , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Quality of Life , Circadian Clocks/genetics , Circadian Clocks/drug effects , Leukocyte L1 Antigen Complex/genetics , Leukocyte L1 Antigen Complex/metabolism , Gene Expression Regulation/drug effects , Butyrates , Butyric Acid
17.
Sci Rep ; 14(1): 15095, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956125

ABSTRACT

Nanogels offer hope for precise drug delivery, while addressing drug delivery hurdles is vital for effective prostate cancer (PCa) management. We developed an injectable elastin nanogels (ENG) for efficient drug delivery system to overcome castration-resistant prostate cancer (CRPC) by delivering Decursin, a small molecule inhibitor that blocks Wnt/ßcatenin pathways for PCa. The ENG exhibited favourable characteristics such as biocompatibility, flexibility, and low toxicity. In this study, size, shape, surface charge, chemical composition, thermal stability, and other properties of ENG were used to confirm the successful synthesis and incorporation of Decursin (DEC) into elastin nanogels (ENG) for prostate cancer therapy. In vitro studies demonstrated sustained release of DEC from the ENG over 120 h, with a pH-dependent release pattern. DU145 cell line induces moderate cytotoxicity of DEC-ENG indicates that nanomedicine has an impact on cell viability and helps strike a balance between therapeutics efficacy and safety while the EPR effect enables targeted drug delivery to prostate tumor sites compared to free DEC. Morphological analysis further supported the effectiveness of DEC-ENG in inducing cell death. Overall, these findings highlight the promising role of ENG-encapsulated decursin as a targeted drug delivery system for CRPC.


Subject(s)
Elastin , Nanogels , Prostatic Neoplasms, Castration-Resistant , Male , Elastin/chemistry , Humans , Cell Line, Tumor , Nanogels/chemistry , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Drug Delivery Systems , Cell Survival/drug effects , Drug Liberation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Benzopyrans , Butyrates
18.
Microbiome ; 12(1): 131, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030599

ABSTRACT

BACKGROUND: The average daily gain (ADG) of preweaning calves significantly influences their adult productivity and reproductive performance. Gastrointestinal microbes are known to exert an impact on host phenotypes, including ADG. The aim of this study was to investigate the mechanisms by which gastrointestinal microbiome regulate ADG in preweaning calves and to further validate them by isolating ADG-associated rumen microbes in vitro. RESULTS: Sixteen Holstein heifer calves were selected from a cohort with 106 calves and divided into higher ADG (HADG; n = 8) and lower ADG (LADG; n = 8) groups. On the day of weaning, samples of rumen contents, hindgut contents, and plasma were collected for rumen metagenomics, rumen metabolomics, hindgut metagenomics, hindgut metabolomics, and plasma metabolomics analyses. Subsequently, rumen contents of preweaning Holstein heifer calves from the same dairy farm were collected to isolate ADG-associated rumen microbes. The results showed that the rumen microbes, including Pyramidobacter sp. C12-8, Pyramidobacter sp. CG50-2, Pyramidobacter porci, unclassified_g_Pyramidobacter, Pyramidobacter piscolens, and Acidaminococcus fermentans, were enriched in the rumen of HADG calves (LDA > 2, P < 0.05). Enrichment of these microbes in HADG calves' rumen promoted carbohydrate degradation and volatile fatty acid production, increasing proportion of butyrate in the rumen and ultimately contributing to higher preweaning ADG in calves (P < 0.05). The presence of active carbohydrate degradation in the rumen was further suggested by the negative correlation of the rumen microbes P. piscolens, P. sp. C12-8 and unclassified_g_Pyramidobacter with the rumen metabolites D-fructose (R < - 0.50, P < 0.05). Widespread positive correlations were observed between rumen microbes (such as P. piscolens, P. porci, and A. fermentans) and beneficial plasma metabolites (such as 1-pyrroline-5-carboxylic acid and 4-fluoro-L-phenylalanine), which were subsequently positively associated with the growth rate of HADG calves (R > 0.50, P < 0.05). We succeeded in isolating a strain of A. fermentans from the rumen contents of preweaning calves and named it Acidaminococcus fermentans P41. The in vitro cultivation revealed its capability to produce butyrate. In vitro fermentation experiments demonstrated that the addition of A. fermentans P41 significantly increased the proportion of butyrate in the rumen fluid (P < 0.05). These results further validated our findings. The relative abundance of Bifidobacterium pseudolongum in the hindgut of HADG calves was negatively correlated with hindgut 4-hydroxyglucobrassicin levels, which were positively correlated with plasma 4-hydroxyglucobrassicin levels, and plasma 4-hydroxyglucobrassicin levels were positively correlated with ADG (P < 0.05). CONCLUSIONS: This study's findings unveil that rumen and hindgut microbes play distinctive roles in regulating the preweaning ADG of Holstein heifer calves. Additionally, the successful isolation of A. fermentans P41 not only validated our findings but also provided a valuable strain resource for modulating rumen microbes in preweaning calves. Video Abstract.


Subject(s)
Gastrointestinal Microbiome , Rumen , Weaning , Animals , Cattle , Rumen/microbiology , Rumen/metabolism , Bacteria/classification , Bacteria/isolation & purification , Bacteria/metabolism , Bacteria/genetics , Female , Fermentation , Metagenomics/methods , Metabolomics , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Weight Gain , Butyrates/metabolism
19.
Mediators Inflamm ; 2024: 6263447, 2024.
Article in English | MEDLINE | ID: mdl-39015676

ABSTRACT

Group 2 innate lymphoid cells (ILC2) strongly modulate COPD pathogenesis. However, the significance of microbiota in ILC2s remains unelucidated. Herein, we investigated the immunomodulatory role of short-chain fatty acids (SCFAs) in regulating ILC2-associated airway inflammation and explores its associated mechanism in COPD. In particular, we assessed the SCFA-mediated regulation of survival, proliferation, and cytokine production in lung sorted ILC2s. To elucidate butyrate action in ILC2-driven inflammatory response in COPD models, we administered butyrate to BALB/c mice via drinking water. We revealed that SCFAs, especially butyrate, derived from dietary fiber fermentation by gut microbiota inhibited pulmonary ILC2 functions and suppressed both IL-13 and IL-5 synthesis by murine ILC2s. Using in vivo and in vitro experimentation, we validated that butyrate significantly ameliorated ILC2-induced inflammation. We further demonstrated that butyrate suppressed ILC2 proliferation and GATA3 expression. Additionally, butyrate potentially utilized histone deacetylase (HDAC) inhibition to enhance NFIL3 promoter acetylation, thereby augmenting its expression, which eventually inhibited cytokine production in ILC2s. Taken together, the aforementioned evidences demonstrated a previously unrecognized role of microbial-derived SCFAs on pulmonary ILC2s in COPD. Moreover, our evidences suggest that metabolomics and gut microbiota modulation may prevent lung inflammation of COPD.


Subject(s)
Butyrates , Dietary Fiber , Lymphocytes , Mice, Inbred BALB C , Pulmonary Disease, Chronic Obstructive , Animals , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Mice , Butyrates/pharmacology , Lymphocytes/metabolism , Dietary Fiber/pharmacology , Dietary Fiber/therapeutic use , Fatty Acids, Volatile/metabolism , Inflammation/metabolism , Gastrointestinal Microbiome , Male , Cytokines/metabolism , Humans , GATA3 Transcription Factor/metabolism
20.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000076

ABSTRACT

The gut microbiota is a diverse bacterial community consisting of approximately 2000 species, predominantly from five phyla: Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia. The microbiota's bacterial species create distinct compounds that impact the host's health, including well-known short-chain fatty acids. These are produced through the breakdown of dietary fibers and fermentation of undigested carbohydrates by the intestinal microbiota. The main short-chain fatty acids consist of acetate, propionate, and butyrate. The concentration of butyrate in mammalian intestines varies depending on the diet. Its main functions are use as an energy source, cell differentiation, reduction in the inflammatory process in the intestine, and defense against oxidative stress. It also plays an epigenetic role in histone deacetylases, thus helping to reduce the risk of colon cancer. Finally, butyrate affects the gut-brain axis by crossing the brain-blood barrier, making it crucial to determine the right concentrations for both local and peripheral effects. In recent years, there has been a significant amount of attention given to the role of dietary polyphenols and fibers in promoting human health. Polyphenols and dietary fibers both play crucial roles in protecting human health and can produce butyrate through gut microbiota fermentation. This paper aims to summarize information on the key summits related to the negative correlation between intestinal microbiota diversity and chronic diseases to guide future research on determining the specific activity of butyrate from polyphenols and dietary fibers that can carry out these vital functions.


Subject(s)
Butyrates , Dietary Fiber , Gastrointestinal Microbiome , Polyphenols , Gastrointestinal Microbiome/drug effects , Dietary Fiber/metabolism , Dietary Fiber/pharmacology , Humans , Polyphenols/pharmacology , Butyrates/metabolism , Animals , Fatty Acids, Volatile/metabolism , Fermentation
SELECTION OF CITATIONS
SEARCH DETAIL