Project description:Given the criticle role of gut bacteria involve in number of diseases, the gut microbiota from young and aged people were estimated using 16s rRNA next-generation sequencing. This study will benefit to identify the role of gut bacteria on the pathegenic mechasim of aging relative diseases.
Project description:In this study, we performed a comparative analysis of gut microbiota composition and gut microbiome-derived bacterial extracellular vesicles (bEVs) isolated from patients with solid tumours and healthy controls. After isolating bEVs from the faeces of solid tumour patients and healthy controls, we performed spectrometry analysis of their proteomes and next-generation sequencing (NGS) of the 16S gene. We also investigated the gut microbiomes of faeces from patientsand controls using 16S rRNA sequencing. Machine learning was used to classify the samples into patients and controls based on their bEVs and faecal microbiomes.
Project description:We compared the microbiota of paired mouse caecal contents and faeces by applying a multi-omic approach, including 16S rDNA sequencing, shotgun metagenomics, and shotgun metaproteomics. The aim of the study was to verify whether faecal samples are a reliable proxy for the mouse colonic luminal microbiota, as well as to identify changes in taxonomy and functional activity between caecal and faecal microbial communities, which have to be carefully considered when using stool as sample for mouse gut microbiota investigations.
Project description:Given the gut microbiota involve aging processing, we performed comparative analysis of gut bacteriophage between older and young subjects using next-generation sequencing (NGS). In our previous study, we found that the Ruminococcaceae is higher in aged subjects comparing to young one. To identify the bacteriophage targeting to the Ruminococcaceae, we also access the composition of phage in the Ruminococcaceae (ATCC, TSD-27) after incubated with human stool samples. The Lactobacillus (ATCC, LGG) targeting phage was used as the control. The virome sequencing analysis using NGS indicated that Myoviridae are high enrich in young subjects and predominate in TSD-27 targeting phage.
Project description:Gut microbiota is an unignored target in maintaining intestinal homeostasis due to its regulatory effects on intestinal health through multiple mechanisms, including enhancing intestinal barriers, modulating microbial diversity, secreting various metabolites, etc. Bacteriocins produced by probiotics have been gradually proved vital for intestinal diseases intervention, however, the corresponding mechanisms have received less attention and the whole story of their regulative activities are hard to be fully uncovered. The two-peptide Plantaricin NC8 (PLNC8), coded by gene plnc8, is a bacteriocin ubiquitously produced by Lactobacillus plantarum, has been regarded as the potential vital bacteriocin for the anti-inflammatory effects of Lactobacillus plantarum. This study exploited CRISPR-cas9 and prokaryotic gene overexpression techniques to construct the plnc8 strains for the anti-inflammatory mechanism investigation. Based on the metagenomics, transcriptomics and metabolomics analysis, the anti-enteritis mechanism of PLNC8 systematically in DSS-induced enteritis models were comprehensively revealed. PLNC8 induced alterations in the composition of gut microbiota composition, promoting the alterations of multiple probiotics such as Eubacterium plexicaudatum, Doreasp.5-2, Enterococcus cecorum and Prevotella oulorum. Besides, various metabolites produced by the gut microbiota were influenced, and the key metabolites of xanthine, hypoxanthine, and L-histidine were regulated via purine and histidine metabolic pathways. These metabolites further inhibited p38 MAPK phosphorylation of enterocytes induced by DSS. Ultimately, the intestinal barrier repairment and anti- enteritis were achieved, proving the anti-enteritis effects of PLNC8 via microbe-metabolites-enterocyte axis.
Project description:Human diet emerges as a pivotal determinant of gut microbiota composition and function. Identification of the bacterial taxa targeted by diet derived factors with causal beneficial rather than detrimental effects on therapy and their mechanism of action is challenging but necessary for future clinical progress. The germ free mice colonized with human gut bacteria and four-plants derived nanoparticles uptaking bacteria were sorted with flow cytometry and identified with 16s rRNA next-generation sequencing.
Project description:The mammalian gut harbors a diverse microbial community (gut microbiota) that mainly consists of bacteria. Their combined genomes (the microbiome) provide biochemical and metabolic functions that complement host physiology. Maintaining symbiosis seems to be a key requirement for health as dysbiosis is associated with the development of common diseases. Previous studies indicated that the microbiota and the hostM-bM-^@M-^Ys epithelium signal bidirectional inducing transcriptional responses to fine-tune and maintain symbiosis. However, little is known about the hostM-bM-^@M-^Ys responses to the microbiota along the length of the gut as earlier studies of gut microbial ecology mostly used either colonic or fecal samples. This is of importance as not only function and architecture of the gut varies along its length but also microbial distribution and diversity. Few recent studies have begun to investigate microbiota-induced host responses along the length of the gut. However, these reports used whole tissue samples and therefore do not allow drawing conclusions about specificity of the observed responses. Which cells in the intestinal tissue are responsible for the microbially induced response: epithelial, mesenchymal or immune cells? Where are the responding cells located? Furthermore, the gut microbiota has been implicated in epigenetic regulation of the hostM-bM-^@M-^Ys transcriptional profile. We used using extensive microarray analysis of laser capture microdissection (LCM) harvested ileal and colonic tip and crypt fractions from germ-free mice before and during the time course of colonization with a normal microbiota (on days 1, 3, 5 and 7) to investigate the microbiota-induced transcriptional responses and their kinetics in specific and well-defined cell populations of the hostM-bM-^@M-^Ys epithelium. Ileum and colon segments were dissected from germ-free 10-12 weeks old female C57Bl/6 mice and on day 1, 3, 5 and 7 after colonization, washed and frozen as OCT blocks. Cryosections were prepared from these OCT blocks and tip/crypt fractions isolated using laser capture microdissection. To investigate the microbiota-induced transcriptional responses specific for specific subpopulations of intestinal epithelial cells and their kinetics, tip and crypt fractions of ileal and colonic epithelium of germ-free 10-12 weeks old female C57Bl/6 mice before and during the time course of colonization with a normal microbiota (on days 1, 3, 5 and 7) were harvested using laser capture microdissection and probed in an extensive microarray analysis.