Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 896
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Alzheimers Dis ; : 13872877241289057, 2024 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-39497291

RESUMEN

Recent studies indicate that gut microbiota may play a crucial role in cognitive function. Individuals with cognitive impairment tend to have fewer beneficial gut bacteria and lower microbial diversity. Therefore, gut microbiota could be a potential biomarker for cognitive vulnerability. Further research is needed to understand the mechanisms and lifestyle factors affecting both microbiota composition and cognitive health. While the direct impact of microbiota and diet on cognitive impairment remains unconfirmed, this area holds promise for developing new preventive and treatment strategies.

2.
Front Nutr ; 11: 1491821, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39502877

RESUMEN

The human gut harbors a complex and diverse microbiota essential for maintaining health. Diet is the most significant modifiable factor influencing gut microbiota composition and function, particularly through bioactive compounds like polyphenols, dietary fibers, and carotenoids found in vegetables, fruits, seafood, coffee, and green tea. These compounds regulate the gut microbiota by promoting beneficial bacteria and suppressing harmful ones, leading to the production of key microbiota-derived metabolites such as short-chain fatty acids, bile acid derivatives, and tryptophan metabolites. These metabolites are crucial for gut homeostasis, influencing gut barrier function, immune responses, energy metabolism, anti-inflammatory processes, lipid digestion, and modulation of gut inflammation. This review outlines the regulatory impact of typical bioactive compounds on the gut microbiota and explores the connection between specific microbiota-derived metabolites and overall health. We discuss how dietary interventions can affect disease development and progression through mechanisms involving these metabolites. We examine the roles of bioactive compounds and their metabolites in the prevention and treatment of diseases including inflammatory bowel disease, colorectal cancer, cardiovascular diseases, obesity, and type 2 diabetes mellitus. This study provides new insights into disease prevention and underscores the potential of dietary modulation of the gut microbiota as a strategy for improving health.

3.
Foods ; 13(19)2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39410228

RESUMEN

The gastrointestinal microbiota are important for human health. Dietary intake may modulate the composition and metabolic function of the gut microbiome. We examined how the breakdown of prebiotic dietary fibers by the gut microbiome affects mucin secretion by intestinal epithelial cells. Metagenomic analyses of in vitro gut microbiome consortia revealed taxonomic profiles and genetic diversity of carbohydrate-active enzymes that digest polysaccharides. Two independent consortia exhibited different abilities to produce acetic acid, propionic acid, and butyric acid via the fermentation of polysaccharides derived from dietary fibers of grains and mushrooms. Although acetic acid generally had the highest concentration, the ratios of butyric acid and propionic acid to acetic acid varied depending on the polysaccharide source. These short-chain fatty acids affected morphological differentiation and mucin secretion in HT-29 human intestinal epithelial cells. These results suggest that prebiotic dietary fibers can be digested and metabolized by the gut microbiome to short-chain fatty acids, which can affect gut epithelial cells both directly and indirectly via the modulation of the gut microbiota and their enzymes.

4.
Genes (Basel) ; 15(10)2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39457400

RESUMEN

BACKGROUND/OBJECTIVES: The effect of sodium butyrate (NaB), ß-glucan (ßG) and vitamins in the diet on gut microbiome, cortisol level, lysozyme activity and growth parameters of juvenile hybrid sturgeon (Acipenser gueldenstaedtii♀ × Acipenser baerii♂) was determined. METHODS: Sturgeon hybrids (n = 144) were divided into three groups with enriched feeding (mg/kg of feed): FQV1 (50 NaB; 20 ßG; const. vitamins), FQV2 (150 NaB; 20 ßG; const. vitamins), FQV3 (50 NaB; 60 ßG; const. vitamins) and control (not supplemented), each group in triplicate, 12 fish in each repetition. Rearing was carried out for 30 days in controlled conditions. Gut microbiome was characterized using Next Generation Sequencing (NGS) of DNA samples isolated from intestinal content. Cortisol level was determined using the ELISA test. Lysozyme activity was measured by turbidimetric test. RESULTS: Based on data obtained from NGS, it was determined that the FQV1 group is characterized by the highest values of diversity indices (Shannon, Simpson and Chao-1) and the largest number of ASVs (Amplicon Sequence Variants). The highest abundance of probiotic bacteria (Lactobacillus, Lactococcus) was determined in the FQV1 group. The highest cortisol concentration was determined in the control (33.26 ng/mL), while the lowest was in FQV3 (27.75 ng/mL). The highest lysozyme activity was observed in FQV1 (154.64 U/mL), and the lowest in FQV2 (104.39 U/mL) and control (121.37 U/mL) (p < 0.05). FQV2 was characterized by significantly more favorable values of breeding indicators (p < 0.05). CONCLUSIONS: The obtained results prove that an appropriate composition of NaB, ßG and vitamins can be used in the commercial breeding of juvenile hybrid sturgeons.


Asunto(s)
Ácido Butírico , Suplementos Dietéticos , Peces , Microbioma Gastrointestinal , Secuenciación de Nucleótidos de Alto Rendimiento , Vitaminas , beta-Glucanos , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , beta-Glucanos/farmacología , beta-Glucanos/administración & dosificación , Ácido Butírico/metabolismo , Ácido Butírico/farmacología , Peces/genética , Peces/microbiología , Vitaminas/farmacología , Vitaminas/administración & dosificación , Alimentación Animal/análisis , Masculino , Femenino , Hidrocortisona , Muramidasa/genética
5.
Gut Microbes ; 16(1): 2401654, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39420751

RESUMEN

Diabetes mellitus can be subdivided into several categories based on origin and clinical characteristics. The most common forms of diabetes are type 1 (T1D), type 2 diabetes (T2D) and gestational diabetes mellitus (GDM). T1D and T2D are chronic diseases affecting around 537 million adults worldwide and it is projected that these numbers will increase by 12% over the next two decades, while GDM affects up to 30% of women during pregnancy, depending on diagnosis methods. These forms of diabetes have varied origins: T1D is an autoimmune disease, while T2D is commonly associated with, but not limited to, certain lifestyle patterns and GDM can result of a combination of genetic predisposition and pregnancy factors. Despite some pathogenic differences among these forms of diabetes, there are some common markers associated with their development. For instance, gut barrier impairment and inflammation associated with an unbalanced gut microbiota and their metabolites may be common factors in diabetes development and progression. Here, we summarize the microbial signatures that have been linked to diabetes, how they are connected to diet and, ultimately, the impact on metabolite profiles resulting from host-gut microbiota-diet interactions. Additionally, we summarize recent advances relating to promising preventive and therapeutic interventions focusing on the targeted modulation of the gut microbiota to alleviate T1D, T2D and GDM.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Microbioma Gastrointestinal , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiología , Embarazo , Diabetes Gestacional/metabolismo , Diabetes Gestacional/microbiología , Diabetes Gestacional/prevención & control , Femenino , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/microbiología , Animales , Dieta , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación
6.
J Agric Food Chem ; 72(42): 23211-23223, 2024 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-39393822

RESUMEN

Gut microbiota provides an important insight into clarifying the mechanism of active substances with low bioavailability, but its specific action mechanism varied case by case and remained unclear. Dihydroquercetin (DHQ) is a bioactive flavonoid with low bioavailability, which showed beneficial effects on colitis alleviation and gut microbiota modulation. Herein, we aimed to explore the microbiota-dependent anticolitis mechanism of DHQ in sight of gut microbiota metabolites and their interactions with microRNAs (miRNAs). Dietary supplementation of DHQ alleviated dextran sulfate sodium-induced colitis phenotypes and improved gut microbiota dysbiosis. Fecal microbiota transplantation further revealed that the anticolitis activity of DHQ was mediated by gut microbiota. To clarify how the modulated gut microbiota alleviated colitis in mice, the tandem analyses of the microbiome and targeted metabolome were performed, and altered profiles of metabolite short-chain fatty acids (SCFAs) and bile acids and their producers were observed in DHQ-treated mice. In addition, SCFA treatment showed anticolitis activity compared to that of bile acids, along with the specific inhibition on the phosphoinositide-3-kinase (PI3K)-protein kinase B (Akt) pathway. Subsequently, the colonic miRNA profile of mice receiving SCFA treatment was sequenced, and a differentially expressed miR-10a-5p was identified. Both prediction analysis and dual-luciferase reporter assay indicated that miR-10a-5p directly bind to the 3'-untranslated regions of gene pik3ca, inhibit the PI3K-Akt pathway activation, and lead to colitis alleviation. Together, we proposed that gut microbiota mediated the anticolitis activity of DHQ through the SCFAs/miR-10a-5p/PI3K-Akt axis, and it provided a novel insight into clarifying the microbiota-dependent mechanism via the interaction between metabolites and miRNAs.


Asunto(s)
Colitis , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , MicroARNs , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Quercetina , Transducción de Señal , Animales , MicroARNs/genética , MicroARNs/metabolismo , Quercetina/análogos & derivados , Quercetina/administración & dosificación , Quercetina/metabolismo , Quercetina/farmacología , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colitis/genética , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Microbioma Gastrointestinal/efectos de los fármacos , Ácidos Grasos Volátiles/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Humanos , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Bacterias/efectos de los fármacos
7.
Artículo en Inglés | MEDLINE | ID: mdl-39410870

RESUMEN

In the swine industry, there is a strong need to replace an antibiotic growth promoter (AGP) used as feed additives in weaned piglets to enhance nutrient utilization in their diets and improve growth performance. Lactobacillus sakei HS-1 strain is a microbial preparation isolated from pickles. The study aim is to investigate the effectiveness of heat-treated L. sakei HS-1 strain (HT-LS) as a growth promoter in weaned piglets compared to colistin (CS), a widely used AGP. Eighteen crossbred weaned piglets (Landrace × Yorkshire × Duroc) of 21 days (average body weight [BW]: 7.06 ± 0.59 kg) were divided into three groups: fed the control diet (CT group), fed a diet supplemented with 30 ppm colistin sulphate (CS group), fed a diet supplemented with HT-LS at a concentration of 2.0 × 105 cells/g (LS group) until 49 days. The results indicated that LS group exhibited significantly higher average daily gain (p < 0.05) and higher BW (p < 0.1) compared with CT group, even higher than CS group. CS group showed higher growth performance compared to CT group but the differences were not statistically significant. In addition, LS group had higher (p < 0.05) or tended to higher (p < 0.1) concentrations of several plasma amino acids than the other two groups at 35 and 49 days. Faecal acetate concentration was higher (p < 0.1) in LS group than in CT group at 35 days. Blood immunoglobulin G concentration in LS group was significantly lower (p < 0.05) than in CT group at 35 and 49 days, and blood immunoglobulin A tended to be lower (p < 0.1) at 35 days than in CT group. LS group showed an increased abundance of g_Prevotella 7, g_Streptococcus and g_Lactobacillus (linear discriminant analysis [LDA] score ≥ 2.0). Predictive metagenomic analysis revealed an enrichment of the mixed acid fermentation pathway (LDA score ≥ 2.0). Furthermore, several gut microbes exhibited correlations with plasma amino acids (p < 0.01) and short-chain fatty acids in faeces (p < 0.01). These findings demonstrate that HT-LS improves the growth performance of weaned piglets by enhancing the efficient utilization of nutrients through gut microbiota modification.

8.
Pharmacol Res ; 209: 107445, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39396767

RESUMEN

Gout is characterized by dysregulation of uric acid (UA) metabolism, and the gut microbiota may serve as a regulatory target. This two-month randomized, double-blind, placebo-controlled trial aimed to investigate the additional benefits of coadministering Probio-X alongside febuxostat. A total of 160 patients with gout were randomly assigned to either the probiotic group (n = 120; Probio-X [1 ×1011 CFU/day] with febuxostat) or the placebo group (n = 40; placebo material with febuxostat). Coadministration of Probio-X significantly decreased serum UA levels and the rate of acute gout attacks (P < 0.05). Based on achieving a target sUA level (360 µmol/L) after the intervention, the probiotic group was further subdivided into probiotic-responsive (ProA; n = 54) and probiotic-unresponsive (ProB; n = 66) subgroups. Post-intervention clinical indicators, metagenomic, and metabolomic changes in the ProB and placebo groups were similar, but differed from those in the ProA group, which exhibited significantly lower levels of acute gout attack, gout impact score, serum indicators (UA, XOD, hypoxanthine, and IL-1ß), and fecal gene abundances of UA-producing pathways (KEGG orthologs of K13479 and K01487; gut metabolic modules for formate conversion and lactose and galactose degradation). Additionally, the ProA group showed significantly higher levels (P < 0.05) of gut SCFAs-producing bacteria and UA-related metabolites (xanthine, hypoxanthine, bile acids) after the intervention. Finally, we established a gout metagenomic classifier to predict probiotic responsiveness based on subjects' baseline gut microbiota composition. Our results indicate that probiotic-driven therapeutic responses are highly individual, with the probiotic-responsive cohort benefitting significantly from probiotic coadministration.

9.
Alzheimers Res Ther ; 16(1): 222, 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39396997

RESUMEN

BACKGROUND: Emerging evidence suggested the association between gut dysbiosis and Alzheimer's disease (AD) progression. However, it remained unclear how the gut microbiome and neuroinflammation in the brain mutually interact or how these interactions affect brain functioning and cognition. Here we hypothesized that "gut-brain" axis mediated by microbial derived metabolites was expected to novel breakthroughs in the fields of AD research and development. METHODS: Multiple technologies, such as immunofluorescence, 16s rDNA sequencing, mass spectrometry-based metabolomics (LC-QQQ-MS and GC-MS), were used to reveal potential link between gut microbiota and the metabolism and cognition of the host. RESULTS: Microbial depletion induced by the antibiotics mix (ABX) verified that "gut-brain" can transmit information bidirectionally. Short-chain fatty acid-producing (SCFAs-producing) bacteria and amino acid-producing bacteria fluctuated greatly in 5×FAD mice, especially the reduction sharply of the Bifidobacteriaceae and the increase of the Lachnospiraceae family. Concentrations of several Tryptophan-kynurenine intermediates, lactic acid, CD4+ cell, and CD8+ cells were higher in serum of 5×FAD mice, whilst TCA cycle intermediates and Th1/Th2 were lower. In addition, the levels of iso-butyric acid (IBA) in feces, serum, and brain of 5×FAD mice were increased compared with WT-M mice, especially in serum. And IBA in the brain was positively correlated with Aß and proinflammatory factors. CONCLUSION: Together, our finding highlighted that the alternation in gut microbiota affected the effective communication between the "gut-brain" axis in 5×FAD mice by regulating the immune system, carbohydrate, and energy metabolism.


Asunto(s)
Eje Cerebro-Intestino , Microbioma Gastrointestinal , Enfermedades Neuroinflamatorias , Animales , Microbioma Gastrointestinal/fisiología , Eje Cerebro-Intestino/fisiología , Ratones , Enfermedades Neuroinflamatorias/metabolismo , Enfermedad de Alzheimer/metabolismo , Disbiosis/metabolismo , Encéfalo/metabolismo , Ratones Transgénicos , Medicamentos Herbarios Chinos/farmacología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Medicina Tradicional China/métodos
10.
Eur J Pharmacol ; 984: 177022, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39362390

RESUMEN

Numerous studies conducted over the last ten years have shown a strong correlation between the gut microbiota and the onset and progression of Alzheimer's disease (AD). However, the exact underlying mechanism is still unknown. An ongoing communication mechanism linking the gut and the brain is highlighted by the term "microbiota-gut-brain axis," which was originally coined the "gut-brain axis." Key metabolic, endocrine, neurological, and immunological mechanisms are involved in the microbiota‒gut‒brain axis and are essential for preserving brain homeostasis. Thus, the main emphasis of this review is how the gut microbiota contributes to the development of AD and how various natural products intervene in this disease. The first part of the review provides an outline of various pathways and relationships between the brain and gut microbiota, and the second part provides various mechanisms involved in the gut microbiota and AD. Finally, this review provides knowledge about natural products and their effectiveness in treating gut microbiota-induced AD. AD may be treated in the future by altering the gut microbiota with a customized diet, probiotics/prebiotics, plant products, and natural products. This entails altering the microbiological partners and products (such as amyloid protein) that these partners generate.

11.
Nutrients ; 16(19)2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39408351

RESUMEN

Degenerative joint disease osteoarthritis (OA) is characterized by the degeneration of cartilage, synovial inflammation and low-grade systemic inflammation in association with microbial dysbiosis and intestinal barrier defects. Butyrate is known for its anti-inflammatory and barrier protective effects and might benefit OA patients. In a double-blind placebo-controlled randomized trial, the effects of four to five weeks of oral treatment with sustained-release (SR) butyrate tablets (600 mg/day) on systemic inflammation and immune function were studied in hand OA patients. Serum markers for systemic inflammation and lipopolysaccharide (LPS) leakage were measured and ex vivo stimulation of whole blood or peripheral blood mononuclear cells (PBMCs) was performed at baseline and after treatment. Butyrate treatment did not affect the serum markers nor the cytokine release of ex vivo LPS-stimulated whole blood or PBMCs nor the phenotype of restimulated monocytes. By contrast, butyrate treatment reduced the percentage of activated T helper (Th) cells and the Th17/Treg ratio in αCD3/CD28-activated PBMCs, though cytokine release upon stimulation remained unaffected. Nevertheless, the percentage of CD4+IL9+ cells was reduced by butyrate as compared to the placebo. In both groups, the frequency of Th1, Treg, Th17, activated Th17, CD4+IFNγ+ and CD4+TNFα+ cells was reduced. This study shows a proof of principle of some immunomodulatory effects using a SR butyrate treatment in hand OA patients. The inflammatory phenotype of Th cells was reduced, as indicated by a reduced percentage of Th9 cells, activated Th cells and improved Th17/Treg balance in ex vivo αCD3/CD28-activated PBMCs. Future studies are warranted to further optimize the butyrate dose regime to ameliorate inflammation in OA patients.


Asunto(s)
Butiratos , Preparaciones de Acción Retardada , Osteoartritis , Linfocitos T Colaboradores-Inductores , Comprimidos , Humanos , Método Doble Ciego , Masculino , Osteoartritis/tratamiento farmacológico , Femenino , Persona de Mediana Edad , Butiratos/farmacología , Butiratos/administración & dosificación , Anciano , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Citocinas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación
12.
Int J Mol Sci ; 25(19)2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39408727

RESUMEN

Cardiovascular disease is the main cause of mortality in industrialized countries, with over 500 million people affected worldwide. In this work, the roles of low-molecular-weight metabolites originating from the gut microbiome, such as short-chain fatty acids, hydrogen sulfide, trimethylamine, phenylacetic acid, secondary bile acids, indoles, different gases, neurotransmitters, vitamins, and complex lipids, are discussed in relation to their CVD-promoting or preventing activities. Molecules of mixed microbial and human hepatic origin, such as trimethylamine N-oxide and phenylacetylglutamine, are also presented. Finally, dietary agents with cardioprotective effects, such as probiotics, prebiotics, mono- and poly-unsaturated fatty acids, carotenoids, and polyphenols, are also discussed. A special emphasis is given to their gut microbiota-modulating properties.


Asunto(s)
Enfermedades Cardiovasculares , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/metabolismo , Animales , Probióticos , Peso Molecular , Ácidos Grasos Volátiles/metabolismo , Prebióticos
13.
Prev Nutr Food Sci ; 29(3): 270-278, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39371515

RESUMEN

The development of inflammatory bowel disease (IBD) is closely linked to inflammatory damage and dysbiosis. Recently, probiotics are being increasingly used to improve intestinal health. Probiotic-based therapies can prevent IBD by restoring the balance of gastrointestinal microbiota, reducing gut inflammation, and increasing the concentration of short-chain fatty acids (SCFAs). The present study aimed to investigate the protective effects of Lactiplantibacillus plantarum subsp. plantarum Dad-13, a novel probiotic strain derived from dadih (Indonesian curd from buffalo milk), on 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis in BALB/c mice. The results showed that probiotic Dad-13 supplementation at a dose of 107 or 109 CFU/mL improved the clinical symptoms of IBD and enhanced the production of SCFAs, particularly propionate and butyrate. Moreover, probiotic Dad-13 supplementation significantly decreased the levels of pro-inflammatory cytokines [tumor necrosis factor-α, interleukin (IL)-6, and IL-1ß] and significantly increased the levels of anti-inflammatory cytokines (IL-10). These findings show that L. plantarum Dad-13 can effectively prevent TNBS-induced colitis by modulating SCFA production and inflammatory cytokines.

14.
Poult Sci ; 103(12): 104337, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39388980

RESUMEN

In a 21-d study, 480 Cobb 500 (off-sex) male broiler chicks were used to investigate the effects of feeding different sources and levels of resistant starches (RS) on growth performance, nutrient and energy utilization, and intestinal health in broiler chickens. The birds were allocated to 10 dietary treatments in a 3 × 3 + 1 factorial arrangement. The factors were 3 RS-sources (RSS): banana starch (BS), raw potato starch (RPS), and high-amylose corn starch (HCS); each at 3 levels (RSL) 25, 50, or 100 g/kg plus a corn-soybean meal control diet. Birds and feed were weighed on d 0, 8, and 21. On d 21, samples of jejunal tissue and digesta were collected for chemical analysis. Data were analyzed using the mixed model procedure of JMP with factor levels nested with the control. In the 0 to 21 phase, the birds fed the RPS diets had higher (P = 0.011) FI than those fed HCS or control diets, and FCR was greater (P = 0.030) in birds that received BS diets than in other diets. RSS × RSL was significant (P < 0.05) for total tract nutrient retention, AME, and AMEn on d 21. The starch digestibility was higher (P < 0.001) in birds that received the control diet than in RS diets, and decreased as RS levels increased, except for HCS. The apparent metabolizable energy (AME) and nitrogen-corrected AME (AMEn) were higher (P < 0.001) in birds fed 100 g/kg HCS diet, with both decreasing with increasing levels of BS and RPS, except for HCS. Relative ileal oligosaccharides profile showed significant (P < 0.05) RSS × RSL with a higher relative abundance of Hex(3) (P = 0.01) and Pent(3) (P = 0.001) in HCS diets. In conclusion, RS may influence gut health and growth performance in broiler chickens through modulation of cecal SCFA and nutrient digestion, but these depend largely on the botanical origin and concentrations of individual RS.

15.
Chin Med ; 19(1): 142, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39394615

RESUMEN

BACKGROUND: Slow transit constipation (STC) is highly prevalent and has rising incidence. Shouhui Tongbian capsule (SHTB) is a traditional Chinese Medicine formula with extensive and highly efficacious usage in STC treatment, however, its mechanism of action, especially the regulation of microbiome and lipid metabolites, remains unclear. METHODS: After quality control of SHTB using LC‒MS to obtain its material basis, we tried to elucidate the cohesive modulatory network of SHTB against STC using hyphenated methods from microbiomics, lipidomics, mass spectrometry imaging (MSI) and molecular methods. RESULTS: SHTB could repair intestinal barrier damage, reduce systemic inflammation and increase intestinal motility in a diphenoxylate-induced STC rat model. Based on 16S rDNA sequencing results, SHTB rehabilitated the abnormal changes in Alloprevotella, Coprococcus, Marvinbryantia, etc., which were associated with STC symptoms. Meanwhile, microbial functional prediction showed that lipid metabolism was improved with SHTB administration. The differential lipids, including fatty acids, lysophosphatidylcholine, phosphatidylcholine, sphingomyelin triglyceride and ceramide, that are closely related to STC disease and SHTB efficacy. Furthermore, SHTB significantly reversed the abnormal expression of these key target enzymes in colon samples, including CTP-phosphocholine cytidylyltransferase, CTP-phosphoethanolamine cytidylyltransferase, phosphatidic acid phosphatase, acid sphingomyelinase etc. CONCLUSIONS: Combined analysis demonstrated that SHTB reducing lipid accumulation and recovery of intestinal microbial homeostasis was the critical mechanism by which SHTB treats STC.

16.
J Ethnopharmacol ; 337(Pt 2): 118874, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39362332

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Cerebral ischemic stroke (CIS) is one of the most important factors leading to death and disability, which seriously threaten the survival and health of patients. The intentional flora and its derived metabolites are demonstrated to play vital roles in the physiology and onset of CIS. Shouhui Tongbian Capsules (SHTB), a Traditional Chinese Medicine, could regulate gut microbiota and metabolites. Study has found that SHTB has protective effect on CIS, but the mechanism is still unclear. AIM OF STUDY: This study was designed to evaluate the preventive effects and the mechanism of SHTB on CIS injury. MATERIALS AND METHODS: The rats were pretreated with SHTB for 5 days, then the middle cerebral artery occlusion/reperfusion (MCAO/R) was established. Neurological deficit score, TTC staining, brain water content, H&E and Nissl staining were preformed to evaluate the preventive effects of SHTB on CIS. The Occludin and ZO-1 were analyzed to evaluate the blood-brain barrier (BBB). 16S rDNA sequencing and LC-ESI-MS/MS-based metabolomics profiling were performed to analyze the gut microbiota composition and short chain fatty acids (SCFAs) profile in gut. Serum lipopolysaccharide specific IgA antibody (LPS-SIgA) and diamine oxidase (DAO), as well as colon Claudin 5 and ZO-1 were analyzed to evaluate the intestinal barrier. Proteomics was used to evaluated the proteins profile in brain. Lipidomics were used to evaluate the brain SCFAs as well as medium and long chain fatty acids (MCFAs and LCFAs). Malondialdehyde (MDA), Total Superoxide dismutase (T-SOD), Glutathione (GSH), Glutathione peroxidase (GSH-Px), Catalase (CAT) and reactive oxygen species (ROS) were assayed to evaluate the oxidative stress in brain. Western blot was performed to evaluate the expression of PPARγ, Nrf2, SLC3A2, SCL7A11, GPX4, ACSL4 and LOX. RESULTS: SHTB prevented rats from MCAO/R injury, which was confirmed by lower cerebral infarct rate, brain water content, neurological deficit score and nissl body loss, and improved brain pathology. Meanwhile, SHTB upregulated the expression of ZO-1 and Occludin to maintain the integrity of BBB. 16S rDNA sequencing and LC-ESI-MS/MS-based targeted metabolomics found that SHTB increased the abundance of gut microbiota, regulated the numbers of intestinal bacteria to increase the production of Acetic acid, Propionic acid, and Butyric acid, as well as decrease the production of Valeric acid and Hexanoic acid in the gut. Meanwhile, SHTB improved the intestinal barrier by upregulating the protein levels of Claudin 5 and ZO-1, which was confirmed by low concentrations of LPS-SIgA and DAO in serum. Multi omics and spearman correlation analysis indicated that SHTB regulated the abundance of Escherichia-Shigella and Lactobacillus to increase Acetic acid, Propionic acid, and Butyric acid to induce the expression of PPARγ, thereby regulating fatty acid metabolism and degradation, improving lipid metabolism disorders, downregulating lipid oxidative stress, inhibiting ferroptosis, and alleviating brain injury. CONCLUSION: This study confirmed that SHTB improved the disturbance of fatty acid metabolism in brain tissue by regulating gut microbiota and the production of fecal SCFAs to inhibit ferroptosis caused by lipid oxidative stress and prevent CIS injury, which provided a potential candidate drug for the prevention of CIS.

17.
BMC Microbiol ; 24(1): 404, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39390387

RESUMEN

BACKGROUND: Preventing post-weaning diarrhea (PWD) in weaned piglets is a crucial challenge in the swine production industry. The stress of weaning, dietary shifts from maternal milk to solid feed, and environmental changes lead to decreased microbial diversity, increased pathogen abundance, and compromised intestinal integrity. We have previously identified Lactiplantibacillus argentoratensis AGMB00912 (LA) in healthy porcine feces, which demonstrated antimicrobial activity against pathogens and enhanced short-chain fatty acid production. This research aimed to evaluate the efficacy of LA strain supplementation as a strategy to inhibit PWD and enhance overall growth performance in weaned piglets. RESULTS: LA supplementation in weaned piglets significantly increased body weight gain, average daily gain, and average daily feed intake. It also alleviated diarrhea symptoms (diarrhea score and incidence). Notably, LA was found to enrich beneficial microbial populations (Lactobacillus, Anaerobutyricum, Roseburia, Lachnospiraceae, and Blautia) while reducing the abundance of harmful bacteria (Helicobacter and Campylobacter). This not only reduces the direct impact of pathogens but also improves the overall gut microbiota structure, thus enhancing the resilience of weaned piglets. LA treatment also promotes the growth of the small intestinal epithelial structure, strengthens gut barrier integrity, and increases short-chain fatty acid levels in the gut. CONCLUSIONS: The study findings demonstrate the promising potential of LA in preventing PWD. Supplementation with the LA strain offers a promising feed additive for improving intestinal health and growth in piglets during the weaning transition, with the potential to significantly reduce the incidence and severity of PWD.


Asunto(s)
Alimentación Animal , Diarrea , Microbioma Gastrointestinal , Probióticos , Enfermedades de los Porcinos , Destete , Animales , Porcinos , Diarrea/microbiología , Diarrea/veterinaria , Diarrea/prevención & control , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos/administración & dosificación , Alimentación Animal/análisis , Heces/microbiología , Lactobacillaceae/genética , Lactobacillaceae/crecimiento & desarrollo , Aumento de Peso/efectos de los fármacos , Suplementos Dietéticos
18.
Int J Biol Macromol ; 281(Pt 1): 136343, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39374720

RESUMEN

Acanthopanax senticosus has proven health benefits, particularly for liver damage. The objective of this study was to elucidate the protective effects and the underlying mechanisms of action of A. senticosus against metabolic dysfunction-associated fatty liver disease (MAFLD). A novel homogeneous water-soluble polysaccharide, CQ-1, was successfully isolated and purified from A. senticosus root. The main chain structure of CQ-1 was identified as →2)-α-L-Rha-(1 â†’ 4)-α-D-GalAp-(1 â†’ 6)-ß-D-Galp-(1→. Additionally, branched chains comprising an arabinosyl residue, galactosyl residue, and galacturonic acidic residue were identified as being attached to →2,4)-α-L-Rha-(1→, →3,6)-ß-D-Galp-(1→, and →3,4)-α-D-GalAp-(1→, respectively. CQ-1 exhibited antioxidant and prebiotic activities in vitro. CQ-1 increased antioxidant capacity and reduced serum pro-inflammatory cytokines in mice. Additionally, CQ-1 has been shown to enhance the diversity and composition of the gut microbiota, thereby facilitating the restoration of gut function. These include improving intestinal barrier function and increasing short-chain fatty acid levels in mice. Our study has shown that CQ-1 has a hepatoprotective effect in MAFLD mice, and we have proposed that CQ-1 may be a promising strategy for the treatment of MAFLD.

19.
Bioresour Technol ; 415: 131725, 2024 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-39477159

RESUMEN

Efficiently and economically recovering volatile fatty acids (VFAs) from sludge anaerobic fermentation (AF) poses a significant challenge. This study discovered a synergistic enhancement effect on VFAs production and membrane fouling control by combining polyethylene (PE) biocarriers and dynamic membrane technology (DM) in an anaerobic bioreactor. The reduced sludge particle size and enhanced hydrolysis efficiency led to a VFAs yield of 1200 mg/L, which is 2.4 times higher than that of traditional AF processes and 1.7 times greater than using the DM module alone. The introduction of PE promoted the enrichment of hydrolytic bacteria, particularly the Christensenellaceae_R-7_group, and facilitated the biotransformation of organic matter. The frictional properties of PE significantly reduced DM fouling, maintaining the transmembrane pressure drop below 30 kPa throughout operation without the need for DM module replacement or cleaning. This study presents a novel approach for resource recovery from sludge through AF, offering new opportunities in the field.

20.
J Hazard Mater ; 480: 136326, 2024 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-39476687

RESUMEN

An investigation was conducted by researchers on how dibutyl phthalate (DBP) and polystyrene microplastics (PS-MP) influence the development of pneumonia using a mouse model. For a duration of five weeks, the mice were subjected to exposure of DBP (30 mg/kg/day) and PS-MP (0.1 mg/day). The findings indicated notable pathological alterations in airway tissues, increased oxidative stress levels, and intensified inflammation, thereby establishing a connection between plastic pollution and pneumonia. Further examination indicated the involvement of ferroptosis and oxidative stress in the progression of the disease. Administration of deferoxamine (DFO) (100 mg/kg) resulted in symptom relief and reduced pathological alterations, as validated by metabolomic investigations. Increased levels of reactive oxygen species (ROS) triggered a Th2-mediated eosinophilic inflammatory response, marked by elevated IL-4 and reduced IFN-γ via the NFκB pathway. Moreover, analyses of the gut microbiome and metabolomics demonstrated that PPD modifies microbial populations and pulmonary metabolism, linking its effects on pneumonia through the gut-lung axis. This research highlights the health hazards associated with plastic pollution and proposes a framework for tackling these issues.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA