Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 2 de 2
1.
Nutrients ; 14(18)2022 Sep 16.
Article En | MEDLINE | ID: mdl-36145205

Exposure to probiotics in early life contributes to host intestinal development and prevention of necrotizing enterocolitis (NEC). Cronobacter sakazakii (C. sakazakii), an opportunistic pathogen, can cause NEC, bacteremia, and meningitis in neonates, but the research of probiotics against C. sakazakii is limited relative to other enteropathogens. Here, the protective effect and mechanism of a novel probiotic Ligilactobacillus salivarius (L. salivarius) YL20 isolated from breast milk on C. sakazakii-induced intestinal injury were explored by using two in vitro models, including an C. sakazakii-infected intestinal organoid model and intestinal barrier model, as well as an in vivo experimental animal model. Our results revealed that L. salivarius YL20 could promote epithelial cell proliferation in intestinal organoids, rescue budding-impaired organoids, prevent the decrease of mRNA levels of leucine-rich repeat containing G protein-coupled receptor 5 (Lgr5), zonula occludens-1 (Zo-1) and Occludin, and reverse C. sakazakii-induced low level of Mucin 2 (MUC2) in intestinal organoids. Additionally, YL20 could inhibit C. sakazakii invasion, increase the expression of ZO-1 and occludin in C. sakazakii-infected HT-29 cells, and reverse TEER decrease and corresponding permeability increase across C. sakazakii-infected Caco-2 monolayers. Furthermore, YL20 administration could alleviate NEC in C. sakazakii-infected neonatal mice by increasing the mice survival ratio, decreasing pathology scores, and downregulating pro-inflammatory cytokines. Meanwhile, YL20 could also enhance intestinal barrier function in vivo by increasing the number of goblet cells, the level of MUC-2 and the expression of ZO-1. Our overall findings demonstrated for the first time the beneficial effects of L. salivarius YL20 against C. sakazakii-induced NEC by improving intestinal stem cell function and enhancing intestinal barrier integrity.


Cronobacter sakazakii , Enterocolitis, Necrotizing , Infant, Newborn, Diseases , Ligilactobacillus salivarius , Probiotics , Animals , Caco-2 Cells , Cytokines/metabolism , Disease Models, Animal , Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/prevention & control , Humans , Infant, Newborn , Intestinal Mucosa/metabolism , Ligilactobacillus salivarius/genetics , Leucine/metabolism , Mice , Mucin-2/genetics , Mucin-2/metabolism , Occludin/metabolism , Probiotics/pharmacology , RNA, Messenger/metabolism
2.
Mar Drugs ; 20(2)2022 Jan 24.
Article En | MEDLINE | ID: mdl-35200626

Chitosan oligosaccharides (COS) have been shown to have potential protective effects against colitis, but the mechanism underlying this effect has not been fully elucidated. In this study, COS were found to significantly attenuate dextran sodium sulfate-induced colitis in mice by decreasing disease activity index scores, downregulating pro-inflammatory cytokines, and upregulating Mucin-2 levels. COS also significantly inhibited the levels of nitric oxide (NO) and IL-6 in lipopolysaccharide-stimulated RAW 264.7 cells. Importantly, COS inhibited the activation of the NF-κB signaling pathway via activating PPARγ and SIRT1, thus reducing the production of NO and IL-6. The antagonist of PPARγ could abolish the anti-inflammatory effects of COS in LPS-treated cells. COS also activated SIRT1 to reduce the acetylation of p65 protein at lysine 310, which was reversed by silencing SIRT1 by siRNA. Moreover, COS treatment increased the diversity of intestinal microbiota and partly restored the Firmicutes/Bacteroidetes ratio. COS administration could optimize intestinal microbiota composition by increasing the abundance of norank_f_Muribaculaceae, Lactobacillus and Alistipes, while decreasing the abundance of Turicibacte. Furthermore, COS could also increase the levels of propionate and butyrate. Overall, COS can improve colitis by regulating intestinal microbiota and the PPARγ/SIRT1-mediated NF-κB pathway.


Chitosan/pharmacology , Colitis/drug therapy , Gastrointestinal Microbiome/drug effects , Oligosaccharides/pharmacology , Animals , Colitis/microbiology , Disease Models, Animal , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , PPAR gamma/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Sirtuin 1/metabolism
...