Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.917
Filtrar
1.
Nat Commun ; 15(1): 8623, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39366940

RESUMEN

Malnutrition is linked to 45% of global childhood mortality, however, the impact of maternal malnutrition on the child's health remains elusive. Previous studies suggested that maternal malnutrition does not affect breast milk composition. Yet, malnourished children often develop a so-called environmental enteropathy, assumed to be triggered by frequent pathogen uptake and unfavorable gut colonization. Here, we show in a murine model that maternal malnutrition induces a persistent inflammatory gut dysfunction in the offspring that establishes during nursing and does not recover after weaning onto standard diet. Early intestinal influx of neutrophils, impaired postnatal development of gut-regulatory functions, and expansion of Enterobacteriaceae were hallmarks of this enteropathy. This gut phenotype resembled those developing under deficient S100a8/a9-supply via breast milk, which is a known key factor for the postnatal development of gut homeostasis. We could confirm that S100a8/a9 is lacking in the breast milk of malnourished mothers and the offspring's intestine. Nutritional supply of S100a8 to neonates of malnourished mothers abrogated the aberrant development of gut mucosal immunity and microbiota colonization and protected them lifelong against severe enteric infections and non-infectious bowel diseases. S100a8 supplementation after birth might be a promising measure to counteract deleterious imprinting of gut immunity by maternal malnutrition.


Asunto(s)
Animales Recién Nacidos , Calgranulina A , Calgranulina B , Desnutrición , Animales , Calgranulina A/metabolismo , Calgranulina A/genética , Calgranulina B/metabolismo , Calgranulina B/genética , Femenino , Ratones , Desnutrición/complicaciones , Desnutrición/metabolismo , Microbioma Gastrointestinal , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Masculino , Suplementos Dietéticos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Embarazo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Inmunidad Mucosa , Humanos , Intestinos/microbiología , Intestinos/patología , Intestinos/inmunología
2.
Cell Host Microbe ; 32(10): 1668-1680, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39389031

RESUMEN

The resident microbiota are a key component of a healthy organism. The vast majority of microbiome studies have focused on bacterial members, which constitute a significant portion of resident microbial biomass. Recent studies have demonstrated how the fungal component of the microbiota, or the mycobiome, influences mammalian biology despite its low abundance compared to other microbes. Fungi are known for their pathogenic potential, yet fungi are also prominent colonizers in healthy states, highlighting their duality. We summarize the characteristics that define the gut mycobiome across life, the factors that can impact its composition, and studies that identify mechanisms of how fungi confer health benefits. The goal of this review is to synthesize our knowledge regarding the composition and function of a healthy mycobiome with a view to inspiring future therapeutic advances.


Asunto(s)
Hongos , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped , Humanos , Hongos/patogenicidad , Hongos/fisiología , Microbioma Gastrointestinal/fisiología , Animales , Micobioma , Intestinos/microbiología , Interacciones Huésped-Patógeno
3.
Front Cell Infect Microbiol ; 14: 1357178, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39391883

RESUMEN

Sepsis, a disease with high incidence, mortality, and treatment costs, has a complex interaction with the gut microbiota. With advances in high-throughput sequencing technology, the relationship between sepsis and intestinal dysbiosis has become a new research focus. However, owing to the intricate interplay between critical illness and clinical interventions, it is challenging to establish a causal relationship between sepsis and intestinal microbiota imbalance. In this review, the correlation between intestinal microecology and sepsis was summarized, and new therapies for sepsis intervention based on microecological target therapy were proposed, and the shortcomings of bacterial selection and application timing in clinical practice were addressed. In conclusion, current studies on metabolomics, genomics and other aspects aimed at continuously discovering potential probiotics are all providing theoretical basis for restoring intestinal flora homeostasis for subsequent treatment of sepsis.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Sepsis , Sepsis/microbiología , Humanos , Probióticos/uso terapéutico , Animales , Metabolómica , Intestinos/microbiología
4.
Sci Rep ; 14(1): 23471, 2024 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-39379397

RESUMEN

Heat stress adversely affects both the productivity and well-being of chickens. Probiotics offer beneficial impacts on the health and growth performance of broilers. The current study investigates the influence of administering of Bacillus (including B. subtilis, B. licheniformis, B. coagulans, and B. indicus) and Lactobacillus (consisting of L. acidophilus, L. plantarum, L. buchneri, and L. rhamnosus) probiotics via drinking water, either singular or combined, on various aspects including growth performance, oxidative stress markers, carcass characteristics, fecal microbial composition, intestinal structure, and intestinal pH in broilers exposed to chronic heat stress. A total of 150 one-day-old broiler chicks were divided into 5 groups: (1) NC, negative control; (2) HS, birds exposed to chronic heat stress; (3) HSpBacil, exposed to chronic heat stress and received Bacillus probiotic; (4) HSpLAB, subjected to chronic heat stress and provided with Lactobacillus probiotic; (5) HSpMix, subjected to chronic heat stress and administered a combined probiotic from Bacillus and Lactobacillus. The HS group exhibited significantly reduced levels of growth performance, carcass traits, and notably affected oxidative stress indices, as well as intestinal pH and histomorphology in the birds. Additionally, the administered probiotics led to increased weight of lymphoid organs, enhanced body weight gain, and improved intestinal histomorphology. Furthermore, the probiotics decreased malondialdehyde and increased total antioxidant capacity in broilers. In conclusion, Bacillus and Lactobacillus probiotics, as single or multi-species, particularly Lactobacillus and combined probiotic, demonstrated potential in alleviating the adverse effects of heat stress in broiler chickens. They could serve as beneficial feed additives and growth enhancers.


Asunto(s)
Antioxidantes , Pollos , Respuesta al Choque Térmico , Intestinos , Estrés Oxidativo , Probióticos , Animales , Pollos/crecimiento & desarrollo , Probióticos/administración & dosificación , Probióticos/farmacología , Antioxidantes/metabolismo , Intestinos/efectos de los fármacos , Intestinos/microbiología , Respuesta al Choque Térmico/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Bacillus/fisiología , Lactobacillus , Microbioma Gastrointestinal/efectos de los fármacos
5.
Gut Microbes ; 16(1): 2393272, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224006

RESUMEN

The intestine is the largest organ in terms of surface area in the human body. It is responsible not only for absorbing nutrients but also for protection against the external world. The gut microbiota is essential in maintaining a properly functioning intestinal barrier, primarily through producing its metabolites: short-chain fatty acids, bile acids, and tryptophan derivatives. Ethanol overconsumption poses a significant threat to intestinal health. Not only does it damage the intestinal epithelium, but, maybe foremostly, it changes the gut microbiome. Those ethanol-driven changes shift its metabolome, depriving the host of the protective effect the physiological gut microbiota has. This literature review discusses the impact of ethanol consumption on the gut, the gut microbiota, and its metabolome, providing a comprehensive overview of the mechanisms through which ethanol disrupts intestinal homeostasis and discussing potential avenues for new therapeutic intervention.


Asunto(s)
Etanol , Microbioma Gastrointestinal , Homeostasis , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Etanol/metabolismo , Etanol/farmacología , Animales , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/microbiología , Intestinos/efectos de los fármacos
6.
Nat Commun ; 15(1): 7733, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231950

RESUMEN

Strains of the Bacillus cereus (Bc) group are sporulating bacteria commonly associated with foodborne outbreaks. Spores are dormant cells highly resistant to extreme conditions. Nevertheless, the pathological processes associated with the ingestion of either vegetative cells or spores remain poorly understood. Here, we demonstrate that while ingestion of vegetative bacteria leads to their rapid elimination from the intestine of Drosophila melanogaster, a single ingestion of spores leads to the persistence of bacteria for at least 10 days. We show that spores do not germinate in the anterior part of the intestine which bears the innate immune defenses. Consequently, spores reach the posterior intestine where they germinate and activate both the Imd and Toll immune pathways. Unexpectedly, this leads to the induction of amidases, which are negative regulators of the immune response, but not to antimicrobial peptides. Thereby, the local germination of spores in the posterior intestine dampens the immune signaling that in turn fosters the persistence of Bc bacteria. This study provides evidence for how Bc spores hijack the intestinal immune defenses allowing the localized birth of vegetative bacteria responsible for the digestive symptoms associated with foodborne illness outbreaks.


Asunto(s)
Bacillus cereus , Drosophila melanogaster , Esporas Bacterianas , Bacillus cereus/inmunología , Esporas Bacterianas/inmunología , Animales , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Intestinos/microbiología , Intestinos/inmunología , Inmunidad Innata , Proteínas de Drosophila/metabolismo , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Receptores Toll-Like/inmunología , Femenino
7.
Curr Protoc ; 4(9): e70013, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39269316

RESUMEN

The study of human intestinal physiology and host-microbe interactions is crucial for understanding gastrointestinal health and disease. Traditional two-dimensional cell culture models lack the complexity of the native intestinal environment, limiting their utility in studying intestinal biology. Here, we present a detailed protocol for the set up and utilization of a three-dimensional (3D) in vitro bioreactor system for human intestinal studies and bacterial co-culture. This article outlines the design and assembly of the bioreactor system, scaffold fabrication, bacterial culture techniques, analysis methods, and troubleshooting tips. By providing step-by-step instructions, the goal is to enable other laboratories to utilize physiologically relevant tissue models of the human intestine, incorporating key features, such as nutrient flow, multiple human cell types, 3D architecture, and microbial communities. The incorporation of commensal bacteria into the bioreactor system allows for the investigation of complex host-microbe interactions, providing insight into gastrointestinal health and pathology. This article serves as a comprehensive resource for scientists seeking to advance their understanding of intestinal biology toward the development of novel therapeutic strategies for gastrointestinal disorders. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Scaffold design Basic Protocol 2: Intestinal cell culture: Caco2 cells Basic Protocol 3: Intestinal cell culture: organoids Basic Protocol 4: Bioreactor design and set up Basic Protocol 5: Bacteria in 3D bioreactor set up Basic Protocol 6: Bacteria and drug dosing.


Asunto(s)
Reactores Biológicos , Técnicas de Cocultivo , Intestinos , Humanos , Reactores Biológicos/microbiología , Técnicas de Cocultivo/métodos , Técnicas de Cocultivo/instrumentación , Intestinos/microbiología , Intestinos/citología , Células CACO-2 , Microbioma Gastrointestinal , Técnicas de Cultivo Tridimensional de Células/métodos , Técnicas de Cultivo Tridimensional de Células/instrumentación
8.
Nutrients ; 16(17)2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39275356

RESUMEN

ATP-binding cassette transporter subfamily G member 2 (ABCG2) is responsible for the excretion of foreign substances, such as uric acid (UA) and indoxyl sulfate (IS), from the body. Given the importance of increased ABCG2 expression in UA excretion, we investigated the enhancement of intestinal ABCG2 expression using Lactiplantibacillus plantarum 06CC2 (LP06CC2). Mice were reared on a potassium oxonate-induced high-purine model at doses of 0.02% or 0.1% LP06CC2 for three weeks. Results showed that LP06CC2 feeding resulted in increased ABCG2 expression in the small intestine. The expression level of large intestinal ABCG2 also showed a tendency to increase, suggesting upregulation of the intestinal excretion transporter ABCG2 by LP06CC2. Overall, LP06CC2 treatment increased fecal UA excretion and showed a trend towards increased fecal excretion of IS, suggesting that LP06CC2 treatment enhanced the expression of intestinal ABCG2, thereby promoting the excretion of UA and other substances from the intestinal tract.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Ácido Úrico , Animales , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Ácido Úrico/metabolismo , Ácido Úrico/orina , Ratones , Masculino , Heces/química , Heces/microbiología , Probióticos , Mucosa Intestinal/metabolismo , Lactobacillus plantarum/metabolismo , Lactobacillaceae/metabolismo , Intestino Delgado/metabolismo , Intestinos/microbiología
9.
Sheng Wu Gong Cheng Xue Bao ; 40(9): 2916-2933, 2024 Sep 25.
Artículo en Chino | MEDLINE | ID: mdl-39319715

RESUMEN

The human gut is a complex ecosystem harboring rich microbes that play a key role in the nutrient absorption, drug metabolism, and immune responses. With the continuous development of microfluidics and organ-on-a-chip, gut-on-a-chip has become a powerful tool for modeling host-microbe interactions. The chip is able to mimic the complex physiological environment of the human gut in vitro, providing a unique platform for studying host-microbe interactions. Firstly, we introduce the physiological characteristics of the human gut. Secondly, we comprehensively summarize the advantages of the microfluidic chip in vitro recapitulating the intestinal system by integrating microenvironmental factors, such as complex cell components, dynamic fluids, oxygen gradients, and mechanical mechanics. Thirdly, we expound the key performance indicators for evaluating the construction performance of gut-on-a-chip. In addition, we review the progress of gut-on-a-chip models in the research on gut microecology, disease modeling, and drug evaluation. Finally, we highlight the challenges and prospects in the applications of the emerging technology. The above is summarized with a view to informing the application of gut-on-a-chip for indepth studies of gut microbe-host interactions.


Asunto(s)
Microbioma Gastrointestinal , Dispositivos Laboratorio en un Chip , Humanos , Interacciones Microbiota-Huesped , Tracto Gastrointestinal/microbiología , Intestinos/microbiología
10.
Nutrients ; 16(18)2024 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-39339809

RESUMEN

BACKGROUND/OBJECTIVES: Limosilactobacillus (Lm.) reuteri is a widely utilized probiotic, recognized for its significant role in alleviating symptoms associated with gastrointestinal and psychiatric disorders. However, the effectiveness of Lm. reuteri is strain-specific, and its genetic diversity leads to significant differences in phenotypes among different strains. This study aims to identify potential probiotic strains by comparing the strain-specific characteristics of Lm. reuteri to better understand their efficacy and mechanisms in alleviating stress-induced anxiety-like behaviors and gastrointestinal symptoms. METHODS: We cultivated 11 strains of Lm. reuteri from healthy human samples and conducted phenotypic and genomic characterizations. Two strains, WLR01 (=GOLDGUT-LR99) and WLR06, were screened as potential probiotics and were tested for their efficacy in alleviating anxiety-like behavior and intestinal symptoms in mouse models subjected to sleep deprivation (SD) and water avoidance stress (WAS). RESULTS: The results showed that the selected strains effectively improved mouse behaviors, including cognitive impairment and inflammatory response, as well as improving anxiety and regulating gut microbiota composition. The improvements with WLR01 were associated with the regulation of the NLRP3 inflammasome pathway in the SD model mice and were associated with visceral hypersensitivity and intestinal integrity in the WAS model mice. CONCLUSIONS: In summary, this study identified the Lm. reuteri strain WLR01 as having the potential to alleviate anxiety-like behavior and intestinal symptoms through the analysis of Lm. reuteri genotypes and phenotypes, as well as validation in mouse models, thereby laying the foundation for future clinical applications.


Asunto(s)
Ansiedad , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Limosilactobacillus reuteri , Probióticos , Estrés Psicológico , Animales , Limosilactobacillus reuteri/fisiología , Probióticos/uso terapéutico , Ansiedad/terapia , Ratones , Masculino , Humanos , Conducta Animal , Ratones Endogámicos C57BL , Intestinos/microbiología
11.
Sci Rep ; 14(1): 22704, 2024 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-39349558

RESUMEN

This study explores the effects of dietary supplementation with passion fruit peel pectin (Passiflora edulis) and red yeast cell walls (Sporidiobolus pararoseus) on growth performance, immunity, intestinal morphology, gene expression, and gut microbiota of Nile tilapia (Oreochromis niloticus). Nile tilapia with an initial body weight of approximately 15 ± 0.06 g were fed four isonitrogenous (29.09-29.94%), isolipidic (3.01-4.28%), and isoenergetic (4119-4214 Cal/g) diets containing 0 g kg-1 pectin or red yeast cell walls (T1 - Control), 10 g kg-1 pectin (T2), 10 g kg-1 red yeast (T3), and a combination of 10 g kg-1 pectin and 10 g kg-1 red yeast (T4) for 8 weeks. Growth rates and immune responses were assessed at 4 and 8 weeks, while histology, relative immune and antioxidant gene expression, and gut microbiota analysis were conducted after 8 weeks of feeding. The results showed that the combined supplementation (T4) significantly enhanced growth performance metrics, including final weight, weight gain, specific growth rate, and feed conversion ratio, particularly by week 8, compared to T1, T2, and T3 (P < 0.05). Immunological assessments revealed increased lysozyme and peroxidase activities in both skin mucus and serum, with the T4 group showing the most pronounced improvements. Additionally, antioxidant and immune-related gene expression, including glutathione peroxidase (GPX), glutathione reductase (GSR), and interleukin-1 (IL1), were upregulated in the gut, while intestinal morphology exhibited improved villus height and width. Gut microbiota analysis indicated increased alpha and beta diversity, with a notable rise in beneficial phyla such as Actinobacteriota and Firmicutes in the supplemented groups. These findings suggest that the combined use of pectin and red yeast cell walls as prebiotics in aquaculture can enhance the health and growth of Nile tilapia, offering a promising alternative to traditional practices. Further research is needed to determine optimal dosages for maximizing these benefits.


Asunto(s)
Alimentación Animal , Cíclidos , Microbioma Gastrointestinal , Intestinos , Passiflora , Pectinas , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Pectinas/farmacología , Pectinas/administración & dosificación , Cíclidos/crecimiento & desarrollo , Cíclidos/inmunología , Cíclidos/microbiología , Intestinos/efectos de los fármacos , Intestinos/microbiología , Passiflora/química , Suplementos Dietéticos , Basidiomycota/química , Frutas
12.
Int J Mol Sci ; 25(18)2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39337313

RESUMEN

Itaconic acid (IA) is recognized for its potential application in treating intestinal diseases owing to the anti-inflammatory and antioxidant properties. Perfluorooctanoic acid (PFOA) can accumulate in animals and result in oxidative and inflammatory damages to multi-tissue and organ, particularly in the intestinal tract. This study aimed to explore whether IA could mitigate intestinal damage induced by PFOA exposure in laying hens and elucidate its potential underlying mechanisms. The results showed that IA improved the antioxidant capacity of laying hens and alleviated the oxidative damage induced by PFOA, as evidenced by the elevated activities of T-SOD, GSH-Px, and CAT, and the decreased MDA content in both the jejunum and serum. Furthermore, IA improved the intestinal morphological and structural integrity, notably attenuating PFOA-induced villus shedding, length reduction, and microvillus thinning. IA also upregulated the mRNA expression of ZO-1, Occludin, Claudin-1, and Mucin-2 in the jejunum, thereby restoring intestinal barrier function. Compared with the PF group, IA supplementation downregulated the gene expression of Keap1 and upregulated the HO-1, NQO1, SOD1, and GPX1 expression in the jejunum. Meanwhile, the PF + IA group exhibited lower expressions of inflammation-related genes (NF-κB, IL-1ß, IFN-γ, TNF-α, and IL-6) compared to the PF group. Moreover, IA reversed the PFOA-induced imbalance in gut microbiota by reducing the harmful bacteria such as Escherichia-Shigella, Clostridium innocuum, and Ruminococcus torques, while increasing the abundance of beneficial bacteria like Lactobacillus. Correlation analysis further revealed a significant association between gut microbes, inflammatory factors, and the Keap1/Nrf2/HO-1 pathway expression. In conclusion, dietary IA supplementation could alleviate the oxidative and inflammatory damage caused by PFOA exposure in the intestinal tract by reshaping the intestinal microbiota, modulating the Keap1/Nrf2/HO-1 pathway and reducing oxidative stress and inflammatory response, thereby promoting intestinal homeostasis.


Asunto(s)
Caprilatos , Fluorocarburos , Microbioma Gastrointestinal , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Caprilatos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Transducción de Señal/efectos de los fármacos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Pollos , Hemo-Oxigenasa 1/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología , Femenino , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología
13.
Antonie Van Leeuwenhoek ; 118(1): 12, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39340698

RESUMEN

A Gram-staining-negative, dark pink, rod-shaped, amastigote and cellulose-degrading strain, designated H9T, was isolated from intestinal contents of Nipponacmea schrenckii. The isolate was able to grow at 4-42 °C (optimum, 25 °C), at pH 6.5-9.0 (optimum, pH 7.0), and with 0.0-11.0% (w/v) NaCl (optimum, 3.0-5.0%). Phylogenetic analysis of the 16S rRNA gene sequence suggested that isolate H9T belongs to the genus Roseobacter, neighboring Roseobacter insulae YSTF-M11T, Roseobacter cerasinus AI77T and Roseobacter ponti MM-7 T, and the pairwise sequence showed the highest similarity of 99.1% to Roseobacter insulae YSTF-M11T. The major fatty acid was summed feature 8 (C18:1ω7c and/or C18:1ω6c; 81.08%). The predominant respiratory quinone was Q-10. The polar lipids consisted of phosphatidylcholine, phosphatidylglycerol, an unknown lipid, and a small amount of an unknown phospholipid. The genome of strain H9T was 5,351,685 bp in length, and the DNA G + C content was 59.8%. The average amino acid identity (AAI), average nucleotide identity (ANI), and digital DNA hybridization (dDDH) values between strain H9T and closely related strains were 63.4-76.8%, 74.7-78.8%, and 13.4-19.7%, respectively. On the basis of the phenotypic, chemical taxonomic, and phylogenetic data, it is suggested that strain H9T should represent a novel species in the genus Roseobacter, for which the name Roseobacter weihaiensis sp. nov. is proposed. The type strain is H9T (= KCTC 82507 T = MCCC 1K04354T).


Asunto(s)
Composición de Base , Celulosa , ADN Bacteriano , Ácidos Grasos , Filogenia , ARN Ribosómico 16S , Roseobacter , China , ARN Ribosómico 16S/genética , Celulosa/metabolismo , ADN Bacteriano/genética , Ácidos Grasos/metabolismo , Roseobacter/clasificación , Roseobacter/genética , Roseobacter/aislamiento & purificación , Roseobacter/metabolismo , Animales , Técnicas de Tipificación Bacteriana , Análisis de Secuencia de ADN , Genoma Bacteriano , Intestinos/microbiología , Fosfolípidos/análisis
14.
Anim Sci J ; 95(1): e14004, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39327865

RESUMEN

Salmonella Typhimurium (S. Typhimurium) infections can lead to severe intestinal damage and reduce growth performance in broilers. Thus, this study examined the potential mitigating impact of sodium humate (HNa) on intestinal barrier damage resulting from S. Typhimurium infection in broilers. A total of 320 1-day-old Arbor Acres broilers were randomly assigned into 5 treatments with 8 replicates. On d 22-24, broilers in the CON group were challenged with 1 ml of PBS, while broilers in the other groups were challenged with 1 ml of 3 × 109 CFU/ml S. Typhimurium, daily. Dietary administration with 4 g/kg of HNa increased (P < 0.05) the final body weight, jejunal secretory immunoglobulin A (sIgA), total antioxidant capacity (T-AOC), total superoxide dismutase (T-SOD), and catalase (CAT) levels as compared with the MOD group broilers. Furthermore, HNa alleviated intestinal barrier damage by increasing villus height (VH), upregulating protein expression of Occludin, Claudin-1, and zonula occludens-1 (ZO-1), inhibiting toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling pathway activation, and decreasing the secretion of inflammatory cytokines (P < 0.05). Collectively, the present study showed that HNa mitigated intestinal barrier damage induced by S. Typhimurium infection in broilers.


Asunto(s)
Antioxidantes , Pollos , Mucosa Intestinal , FN-kappa B , Enfermedades de las Aves de Corral , Salmonelosis Animal , Salmonella typhimurium , Receptor Toll-Like 4 , Animales , Pollos/microbiología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/prevención & control , FN-kappa B/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Antioxidantes/metabolismo , Salmonelosis Animal/prevención & control , Salmonelosis Animal/microbiología , Receptor Toll-Like 4/metabolismo , Superóxido Dismutasa/metabolismo , Transducción de Señal , Citocinas/metabolismo , Inmunoglobulina A Secretora/metabolismo , Catalasa/metabolismo , Intestinos/microbiología , Claudina-1/metabolismo , Ocludina/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Mediadores de Inflamación/metabolismo , Regulación hacia Arriba
15.
Nat Commun ; 15(1): 8428, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341804

RESUMEN

Excessive accumulation of reactive oxygen and nitrogen species (RONS) and dysbiosis of intestinal microbiota are pivotal symptoms for inflammatory bowel disease (IBD) and its associated complications, such as intestinal fibrosis. This research introduces a probiotic inulin hydrogel loaded with polypyrrole (PPy) nanozymes and antifibrotic drug pirfenidone (PFD) (PPy/PFD@Inulin gel) designed for the concurrent amelioration of IBD and its fibrotic complication. Upon oral administration, the inulin gel matrix could extend the gastrointestinal residence time of PPy nanozymes and PFD, facilitating the efficient reduction of pro-inflammatory cytokine levels and enhancement of the intestinal epithelial barrier repair as well as the suppression of intestinal fibrosis through sustained RONS scavenging, modulation of gut microbiota and attenuation of the TGF-ß/Smad signaling pathway to inhibit fibroblast proliferation. Notably, the PPy/PFD@Inulin gel demonstrated significant prophylactic and therapeutic efficacy in acute and chronic colitis as well as intestinal fibrosis induced by dextran sodium sulfate (DSS) in mouse models. Thus, the engineered ternary PPy/PFD@Inulin gel offered a pioneered paradigm for simultaneous reversal of IBD and its associated complications, such as intestinal fibrosis, in a single therapeutic regimen.


Asunto(s)
Fibrosis , Hidrogeles , Enfermedades Inflamatorias del Intestino , Inulina , Animales , Hidrogeles/química , Inulina/química , Ratones , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/patología , Sulfato de Dextran , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Pirroles/química , Intestinos/patología , Intestinos/efectos de los fármacos , Intestinos/microbiología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
16.
J Agric Food Chem ; 72(39): 21594-21609, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39303156

RESUMEN

The diet in early life is essential for the growth and intestinal health later in life. However, beneficial effects of a diet enriched in branched short-chain fatty acids (BSCFAs) for infants are ambiguous. This study aimed to develop a novel fermented protein food, enriched with BSCFAs and assess the effects of dry and wet ferment products on young pig development, nutrient absorption, intestinal barrier function, and gut microbiota and metabolites. A total of 18 young pigs were randomly assigned to three groups. The dry corn gluten-wheat bran mixture (DFCGW) and wet corn gluten-wheat bran mixture (WFCGW) were utilized as replacements for 10% soybean meal in the basal diet. Our results exhibited that the WFCGW diet significantly increased the growth performance of young pigs, enhanced the expression of tight junction proteins, and regulated associated cytokines expression in the colonic mucosa. Simultaneously, the WFCGW diet led to elevated levels of colonic isobutyric and isovaleric acid, as well as the activation of GPR41 and GPR109A. Furthermore, more potential probiotics including Lactobacillus, Megasphaera, and Lachnospiraceae_ND3007_group were enriched in the WFCGW group and positively associated with the beneficial metabolites such as 5-hydroxyindole-3-acetic acid. Differential metabolite KEGG pathway analysis suggested that WFCGW might exert gut health benefits by modulating tryptophan metabolism. In addition, the WFCGW diet significantly increased ghrelin concentrations in serum and hypothalamus and promoted the appetite of young pigs by activating hypothalamic NPY/AGRP neurons. This study extends the knowledge of BSCFAs and provides a reference for the fermented food application in the infant diet.


Asunto(s)
Alimentación Animal , Bacterias , Ácidos Grasos Volátiles , Alimentos Fermentados , Microbioma Gastrointestinal , Animales , Porcinos/metabolismo , Porcinos/crecimiento & desarrollo , Alimentación Animal/análisis , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/crecimiento & desarrollo , Ácidos Grasos Volátiles/metabolismo , Alimentos Fermentados/análisis , Alimentos Fermentados/microbiología , Fermentación , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Intestinos/metabolismo , Masculino , Zea mays/metabolismo , Zea mays/química
17.
J Agric Food Chem ; 72(39): 21560-21573, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39311539

RESUMEN

Increasing research suggests a connection between gut microbiota and depressive disorders. Targeted changes to the intestinal flora may contribute to alleviating anxiety and depression. This study aimed to identify probiotics that could attenuate stress-induced abnormal behavior and explore potential mechanisms. The administration of LR.KY16 significantly reduced stress-induced abnormal behaviors and physiological dysfunction. The mechanism may be via regulating the structure of the intestinal microbiota in mice, increasing the abundance of Akkermansia muciniphila, prompting enterochromaffin cells to secrete 5-HTP in the gut, which enters the brain through the bloodstream and promotes the synthesis of 5-HT in the brain, and then activates brain-derived neurotrophic factor (BDNF)/tyrosine kinase receptor B (TrkB) through the 5-HT1A receptor. In addition, LR.KY16 also increased the expression of claudin-7, occludin, and zonula occludens-1 (ZO-1) in the colon, inhibited microglial M1 polarization, and inhibited systemic inflammation.


Asunto(s)
Depresión , Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Ratones Endogámicos C57BL , Probióticos , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Ratones , Probióticos/administración & dosificación , Probióticos/farmacología , Masculino , Depresión/metabolismo , Depresión/tratamiento farmacológico , Depresión/microbiología , Lacticaseibacillus rhamnosus/metabolismo , Humanos , Serotonina/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Akkermansia , Intestinos/microbiología , Mucosa Intestinal/metabolismo
18.
Cell Host Microbe ; 32(10): 1787-1804.e9, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39243760

RESUMEN

The role of immunoglobulins produced by IL-10-producing regulatory B cells remains unknown. We found that a particular newborn regulatory B cell population (nBreg) negatively regulates the production of immunoglobulin M (IgM) via IL-10 in an autocrine manner, limiting the intensity of the polyreactive antibody response following innate activation. Based on nBreg scRNA-seq signature, we identify these cells and their repertoire in fetal and neonatal intestinal tissues. By characterizing 205 monoclonal antibodies cloned from intestinal nBreg, we show that newborn germline-encoded antibodies display reactivity against bacteria representing six different phyla of the early microbiota. nBreg-derived antibodies can influence the diversity and the cooperation between members of early microbial communities, at least in part by modulating energy metabolism. These results collectively suggest that nBreg populations help facilitate early-life microbiome establishment and shed light on the paradoxical activities of regulatory B cells in early life.


Asunto(s)
Linfocitos B Reguladores , Microbioma Gastrointestinal , Inmunoglobulina M , Interleucina-10 , Animales , Ratones , Interleucina-10/metabolismo , Interleucina-10/inmunología , Microbioma Gastrointestinal/inmunología , Linfocitos B Reguladores/inmunología , Inmunoglobulina M/inmunología , Bacterias/inmunología , Ratones Endogámicos C57BL , Animales Recién Nacidos , Intestinos/inmunología , Intestinos/microbiología , Anticuerpos Monoclonales/inmunología , Femenino , Microbiota/inmunología , Humanos
19.
Int J Biol Macromol ; 279(Pt 4): 135415, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39245119

RESUMEN

Yam is a dual-purpose crop used in both medicine and food that is commonly used as a dietary supplement in food processing. Since yam proteins are often lost during the production of yam starch, elucidating the functionally active value of yam proteins is an important guideline for fully utilizing yam in industrial production processes. This study aimed to explore the potential protective effect of yam protein (YP) on cyclophosphamide (CTX)-induced immunosuppression in mice. The results showed that YP can reduce immune damage caused by CTX by reversing immunoglobulins (IgA, IgG and IgM), cytokines (TNF-α, IL-6, etc.) in the intestines of mice. Moreover, YPs were found to prevent CTX-induced microbiota dysbiosis by enhancing the levels of beneficial bacteria within the microbiome, such as Lactobacillus, and lowering those of Desulfovibrio_R and Helicobacter_A. Metabolomics analyses showed that YP significantly altered differential metabolites (tryptophan, etc.) and metabolic pathways (ABC transporter protein, etc.) associated with immune responses in the gut. Furthermore, important connections were noted between particular microbiomes and metabolites, shedding light on the immunoprotective effects of YPs by regulating gut flora and metabolism. These findings deepen our understanding of the functional properties of YPs and lay a solid foundation for the utilization of yam.


Asunto(s)
Ciclofosfamida , Dioscorea , Microbioma Gastrointestinal , Ciclofosfamida/farmacología , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Dioscorea/química , Proteínas de Plantas/farmacología , Masculino , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/inmunología , Citocinas/metabolismo , Terapia de Inmunosupresión , Disbiosis/inducido químicamente
20.
J Agric Food Chem ; 72(40): 22336-22348, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39322623

RESUMEN

Prolonged and excessive intake of alcohol results in the onset of alcoholic liver disease, which is marked by oxidative stress, intestinal barrier dysfunction, and disturbance in the intestinal microbiome. Galangin, a potent flavonoid from Alpinia officinarum Hance, has been recognized for its diverse biological properties; however, its ability for protecting against alcohol-stimulated hepatotoxicity remains unexplored in prior research. In the current study, a Gao-Binge mouse model was established to assess the positive role and mechanisms of galangin upon alcohol-induced liver injury. The administration of galangin relieved liver pathological damage, oxidative stress, and NLRP3-mediated inflammation induced by alcohol. In addition, galangin significantly reversed abnormal intestinal histopathological manifestations and damaged the intestinal barrier function. Furthermore, microbiota composition revealed that galangin improved intestinal imbalance by improving the gut microbiota dysbiosis and short-chain fatty acid level. Collectively, this study explored the interactions between phytochemical factors and virulence factors and discovered that galangin powerfully improved alcohol-induced liver disease by repressing the inflammatory cascade via the gut microbiota-mediated gut-liver axis. These results suggested that alcohol-targeted natural products could have potential applications in promoting food safety and human health and offer valuable insights into the possible use of these substances in these important areas.


Asunto(s)
Alpinia , Disbiosis , Flavonoides , Microbioma Gastrointestinal , Hepatopatías Alcohólicas , Hígado , Ratones Endogámicos C57BL , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Flavonoides/administración & dosificación , Flavonoides/farmacología , Masculino , Hígado/efectos de los fármacos , Hígado/metabolismo , Humanos , Hepatopatías Alcohólicas/tratamiento farmacológico , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/microbiología , Disbiosis/tratamiento farmacológico , Disbiosis/microbiología , Alpinia/química , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Etanol/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Intestinos/microbiología , Intestinos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA