Abstract

Lipopolysaccharide (LPS; endotoxin) is a lipophilic pathogenic factor derived from gut Gram‐negative bacteria. Increased serum LPS concentrations are associated with the metabolic syndrome, termed metabolic endotoxemia. Since the origin of circulating LPS is the gut lumen, we studied the mechanisms of LPS transport across the gut mucosa, hypothesizing that transport occurs via known lipid uptake pathways. Since glucagon‐like peptide‐2 (GLP‐2) reduces intestinal solute permeability but enhances fat absorption, we also hypothesized that GLP‐2 affects LPS transport during fat absorption. We thus examined LPS transport mechanisms and the effect of exogenous GLP‐2 on LPS transport in intact intestinal tissues in vitro and in vivo. FITC‐LPS absorption in vivo was quantitated by simultaneously measuring the appearance of FITC‐LPS into the portal vein (PV) and the mesenteric lymph, with or without luminal oleic acid with taurocholate (OA/TCA). We also examined fluorescent dextran 4000 (FD4) or LPS transport from mucosal to serosal solution (m‐to‐s) in Ussing chambered muscle‐stripped intestinal tissues.Bolus intraduodenal infusion of FITC‐LPS (50 μg/ml) with OA/TCA rapidly increased FITC‐LPS appearance into PV, followed by a gradual increase into the lymph, whereas FITC‐LPS alone did not appear on PV or lymph. In Ussing chambered jejunum, luminally‐applied LPS (10 μg/ml) rapidly appeared in the serosal solution with luminal OA/TCA, whereas LPS alone or LPS+TCA was not transported. OA/TCA‐induced LPS m‐to‐s transport in the jejunum was inhibited by luminal pretreatment with the lipid raft inhibitor methyl‐β‐cyclodextrin and the CD36 inhibitor sulfosuccinimidyl oleate, but was enhanced by serosal carbachol and luminal application of the competitive alkaline phosphatase inhibitor glycerol phosphate. Furthermore, FD4 m‐to‐s movement was also enhanced by luminal OA/TCA in the jejunum, whereas lesser effects were observed in the duodenum or ileum. Pretreatment with serosal GLP‐2 (100 nM) reduced OA/TCA‐induced LPS transport, whereas carbachol‐induced LPS transport was not affected.These results suggest that luminal LPS crosses the gut barrier physiologically during fat absorption via lipid raft‐ and CD36‐mediated transport mechanisms, similar to chylomicron transport, and possibly through emptied goblet cells. By reducing lipid raft‐, CD36‐, and chylomicron‐mediated LPS transport, GLP‐2 treatment may be a new therapy for metabolic endotoxemia.Support or Funding InformationSupported by VA Merit Review, NIH R01 DK54221

Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call