Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proteome Sci ; 21(1): 19, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37875866

RESUMEN

OBJECTIVE: Thoracic aortic aneurysm (TAA) is a cardiovascular disease with high morbidity and mortality. However, the causes and mechanisms of TAA are not fully understood. Serum exosomes from mice with TAA were used to explore the markers associated with this disease. METHODS: C57BL/6 mice were divided into three groups and given ordinary drinking water, ordinary drinking water plus a saline osmotic pump, or drinking water containing ß-aminopropionitrile (BAPN) (1 g/kg/d) plus an angiotensin II (Ang II) (1 µg/kg/min) osmotic pump. Haematoxylin and eosin staining of thoracic aortic tissues was performed. The basic characteristics of exosomes were analysed. Differentially expressed proteins (DEPs) were identified by LC‒MS/MS. Protein‒protein networks and enrichment analysis were used to explore possible molecular mechanisms. RESULTS: The present study elucidated the protein expression profile of serum exosomes in mice with TAA induced by BAPN combined with Ang II. In this work, the expression of a total of 196 proteins was significantly dysregulated in serum exosomes of mice with TAA, with 122 proteins significantly upregulated and 74 proteins markedly downregulated. Notably, Haptoglobin (Hp) and Serum amyloid p-component (Sap) identified based on the PPI network were significantly upregulated and have been strongly linked to cardiovascular disease. Interestingly, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that the upregulated and downregulated proteins were involved in the complement and coagulation cascade pathways. CONCLUSIONS: This study showed that the identified DEPs have potential as biomarkers for the diagnosis of TAA and provided a more comprehensive understanding of the pathophysiological mechanisms of TAA.

2.
Front Pharmacol ; 14: 1047863, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36778014

RESUMEN

Many drugs have been shown to be metabolized by the human gut microbiome, but probiotic-driven drug-metabolizing capacity is rarely explored. Here, we developed an integrated metabolomics, culturomics, and bionics framework for systematically studying probiotics-driven drug metabolism. We discovered that 75% (27/36 of the assayed drugs) were metabolized by five selected probiotics, and drugs containing nitro or azo groups were more readily metabolized. As proof-of-principle experiments, we showed that Lacticaseibacillus casei Zhang (LCZ) could metabolize racecadotril to its active products, S-acetylthiorphan and thiorphan, in monoculture, in a near-real simulated human digestion system, and in an ex vivo fecal co-culture system. However, a personalized effect was observed in the racecadotril-metabolizing activity of L. casei Zhang, depending on the individual's host gut microbiome composition. Based on data generated by our workflow, we proposed a possible mechanism of interactions among L. casei Zhang, racecadotril, and host gut microbiome, providing practical guidance for probiotic-drug co-treatment and novel insights into precision probiotics.

3.
iScience ; 25(7): 104472, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35733791

RESUMEN

Multiple pesticide residue accumulations increase the probability of chronic metabolic diseases in humans. Thus, we applied multi-omics techniques to reveal how the gut microbiome responded to pesticide exposure. Then, we explored how probiotic Lactiplantibacillus plantarum P9 (P9) consumption impacted the gut microbiota and immune factors after high pesticide exposure. Multi-omics results indicated frequent exposure to pesticides did not alter the composition of the intestinal microbiota, but it did increase the abundance of Lipopolysaccharide in the gut, which might contribute to chronic inflammation. Supplementation with P9 maintained the homeostasis of the gut microbiota and reduced the abundance of pathogens in the high pesticide-exposed subjects. By detecting metabolites, we observed uridine and 5-oxoproline concentrations increased significantly after P9 consumption. Furthermore, P9 alleviated immune factors disorder and promoted pesticide residue excretion. Our findings provide new insights into the application of probiotics for pesticide detoxification, and suggest probiotics as daily supplements for pesticide exposure prevention.

4.
Anim Nutr ; 9: 100-109, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35509811

RESUMEN

Improving silage production by adding exogenous microorganisms not only maximizes nutrient preservation, but also extends product shelf life. Herein, changes in the quality and quantity of Lactobacillus plantarum PS-8 (PS-8) -inoculated alfalfa (Medicago sativa) during silage fermentation were monitored at d 0, 7, 14, and 28 (inoculum dose of PS-8 was 1 × 105 colony forming units [cfu]/g fresh weight; 50 kg per bag; 10 bags for each time point) by reconstructing metagenomic-assembled genomes (MAG) and Growth Rate InDex (GRiD). Our results showed that the exogenous starter bacterium, PS-8 inoculation, became the most dominating strain by d 7, and possibly played a highly active role throughout the fermentation process. The pH value of the silage decreased greatly, accompanied by the growth of acid-producing microorganisms namely PS-8, which inhibited the growth of harmful microorganisms like molds (4.18 vs. 1.42 log cfu/g) and coliforms (4.95 vs. 0.66 log most probable number [MPN]/g). The content of neutral detergent fiber (NDF) decreased significantly (41.6% vs. 37.6%; dry matter basis). In addition, the abundance and diversity of genes coding microbial carbohydrate-active enzymes (CAZymes) increased significantly and desirably throughout the fermentation, particularly the genes responsible for degrading starch, arabino-xylan, and cellulose. Overall, our results showed that PS-8 was replicating rapidly and consistently during early- and mid-fermentation phases, promoting the growth of beneficial lactic acid bacteria and inhibiting undesirable microbes, ultimately improving the quality of silage.

5.
mSystems ; 7(2): e0010022, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35343796

RESUMEN

Accumulating evidence suggests that gut dysbiosis may play a role in cardiovascular problems like coronary artery disease (CAD). Thus, target steering the gut microbiota/metabolome via probiotic administration could be a promising way to protect against CAD. A 6-month randomized, double-blind, placebo-controlled clinical trial was conducted to investigate the added benefits and mechanism of the probiotic strain, Bifidobacterium lactis Probio-M8, in alleviating CAD when given together with a conventional regimen. Sixty patients with CAD were randomly divided into a probiotic group (n = 36; received Probio-M8, atorvastatin, and metoprolol) and placebo group (n = 24; placebo, atorvastatin, and metoprolol). Conventional treatment significantly improved the Seattle Angina Questionnaire (SAQ) scores of the placebo group after the intervention. However, the probiotic group achieved even better SAQ scores at day 180 compared with the placebo group (P < 0.0001). Moreover, Probio-M8 treatment was more conducive to alleviating depression and anxiety in patients (P < 0.0001 versus the placebo group, day 180), with significantly lower serum levels of interleukin-6 and low-density lipoprotein cholesterol (P < 0.005 and P < 0.001, respectively). In-depth metagenomic analysis showed that, at day 180, significantly more species-level genome bins (SGBs) of Bifidobacterium adolescentis, Bifidobacterium animalis, Bifidobacterium bifidum, and Butyricicoccus porcorum were detected in the probiotic group compared with the placebo group, while the abundances of SGBs representing Flavonifractor plautii and Parabacteroides johnsonii decreased significantly among the Probio-M8 receivers (P < 0.05). Furthermore, significantly more microbial bioactive metabolites (e.g., methylxanthine and malonate) but less trimethylamine-N-oxide and proatherogenic amino acids were detected in the probiotic group than placebo group during/after intervention (P < 0.05). Collectively, we showed that coadministering Probio-M8 synergized with a conventional regimen to improve the clinical efficacy in CAD management. The mechanism of the added benefits was likely achieved via probiotic-driven modulation of the host's gut microbiota and metabolome, consequently improving the microbial metabolic potential and serum metabolite profile. This study highlighted the significance of regulating the gut-heart/-brain axes in CAD treatment. IMPORTANCE Despite recent advances in therapeutic strategies and drug treatments (e.g., statins) for coronary artery disease (CAD), CAD-related mortality and morbidity remain high. Active bidirectional interactions between the gut microbiota and the heart implicate that probiotic application could be a novel therapeutic strategy for CAD. This study hypothesized that coadministration of atorvastatin and probiotics could synergistically protect against CAD. Our results demonstrated that coadministering Probio-M8 with a conventional regimen offered added benefits to patients with CAD compared with conventional treatment alone. Our findings have provided a wide and integrative view of the pathogenesis and novel management options for CAD and CAD-related diseases.


Asunto(s)
Adyuvantes Inmunológicos , Bifidobacterium animalis , Enfermedad de la Arteria Coronaria , Humanos , Adyuvantes Farmacéuticos , Atorvastatina , Encéfalo , Metoprolol
6.
J Dairy Sci ; 103(12): 11025-11038, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33222846

RESUMEN

Studies suggest that probiotics and fermented milk can improve defecation in constipated patients. However, the mechanism of fermented milk containing probiotics on constipation remains poorly understood. Volunteers with chronic constipation symptoms were recruited and given 200 g/d of fermented milk containing Lactobacillus casei Zhang and Bifidobacterium animalis ssp. lactis V9 (PFM) for 4 wk. Clinical symptoms, cytokines, metagenomics, and metabolomics were evaluated in constipated participants before and after PFM intervention. After PFM intervention, we observed significant improvement of constipation symptoms. In the serum samples, the anti-inflammatory cytokine IL-10 increased and the proinflammatory cytokine C-reactive protein and lipopolysaccharides decreased. Metagenomics results showed that the increase of B. animalis was correlated with an increase in defecation frequency. Fatty acid biosynthesis and bile acid biosynthesis in stool samples as well as carnitine shuttle, vitamin E metabolism, and ascorbate and aldarate metabolism were identified as significantly altered metabolic pathways. Acylcarnitine, located on the carnitine shuttle pathway, had a significantly positive correlation with defecation frequency. It was speculated that PFM may contribute to alleviating constipation symptoms through 3 potential mechanisms: fine-tuning gastrointestinal microbiota, fighting inflammation, and regulating metabolic pathways.


Asunto(s)
Bifidobacterium animalis/fisiología , Estreñimiento/terapia , Productos Lácteos Cultivados/microbiología , Microbioma Gastrointestinal/fisiología , Inflamación/terapia , Lacticaseibacillus casei/fisiología , Adulto , Animales , Reactores Biológicos , Citocinas/sangre , Femenino , Humanos , Masculino , Redes y Vías Metabólicas/fisiología , Metabolómica , Metagenómica , Probióticos/uso terapéutico
7.
Mol Med Rep ; 16(5): 7138-7144, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28901498

RESUMEN

The authors' previous study demonstrated that Golgi phosphoprotein 3 (GOLPH3) was significantly overexpressed in esophageal squamous cell carcinoma (ESCC), correlating with poor patient survival. In the present study, GOLPH3 stable overexpression and knockdown KYSE­140 cell lines were constructed. Cell proliferation, colony formation, cell cycle progression and tumorigenesis assays were performed. The results revealed that GOLPH3 promoted ESCC cell growth and proliferation. The effects of GOLPH3 on the mechanistic target of rapamycin (mTOR) and Wnt/ß­catenin signaling pathways were investigated using western blot analyis and dual­luciferase reporter assays, and were observed to be activated in cells with GOLPH3 overexpression. Furthermore, overexpression of GOLPH3 resulted in the downregulation of p21 protein, upregulation of cyclin D1 and increased retinoblastoma­associated protein phosphorylation, consequently leading to accelerated cell cycle progression. In addition, GOLPH3 knockdown resulted in reversed effects. The results of the current study suggest that GOLPH3 serves an important role in promoting tumorigenicity of ESCC via mTOR and Wnt/ß­catenin signaling pathway activation.


Asunto(s)
Proteínas de la Membrana/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Interferencia de ARN , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA