Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 172(20): 4864-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26227770

ABSTRACT

BACKGROUND AND PURPOSE: As the pathogenesis of postoperative ileus (POI) involves inflammation and oxidative stress, comparable to ischaemia/reperfusion injury which can be ameliorated with nitrite, we investigated whether nitrite can protect against POI and explored the mechanisms involved. EXPERIMENTAL APPROACH: We used intestinal manipulation (IM) of the small intestine to induce POI in C57BL/6J mice. Sodium nitrite (48 nmol) was administered intravenously just before IM. Intestinal transit was assessed using fluorescent imaging. Bethanechol-stimulated jejunal circular muscle contractions were measured in organ baths. Inflammatory parameters, neutrophil infiltration, inducible NOS (iNOS) activity, reactive oxygen species (ROS) levels, mitochondrial complex I activity and cGMP were measured in the intestinal muscularis. KEY RESULTS: Pre-treatment with nitrite markedly improved the delay in intestinal transit and restored the reduced intestinal contractility observed 24 h following IM. This was accompanied by reduced protein levels of TNF-α, IL-6 and the chemokine CCL2, along with reduced iNOS activity and ROS levels. The associated neutrophil influx at 24 h was not influenced by nitrite. IM reduced mitochondrial complex I activity and cGMP levels; treatment with nitrite increased cGMP levels. Pre-treatment with the NO scavenger carboxy-PTIO or the soluble guanylyl cyclase inhibitor ODQ abolished nitrite-induced protective effects. CONCLUSIONS AND IMPLICATIONS: Exogenous nitrite deserves further investigation as a possible treatment for POI. Nitrite-induced protection of POI in mice was dependent on NO and this effect was not related to inhibition of mitochondrial complex I, but did involve activation of soluble guanylyl cyclase.


Subject(s)
Ileus/drug therapy , Postoperative Complications/drug therapy , Sodium Nitrite/therapeutic use , Animals , Chemokine CCL2/metabolism , Cyclic GMP/metabolism , Gastrointestinal Transit/drug effects , Ileus/immunology , Ileus/metabolism , Ileus/physiopathology , Interleukin-6/metabolism , Jejunum/drug effects , Jejunum/metabolism , Jejunum/physiology , Male , Mice, Inbred C57BL , Muscle Contraction/drug effects , Neutrophils/physiology , Nitric Oxide Synthase Type II/metabolism , Postoperative Complications/immunology , Postoperative Complications/metabolism , Postoperative Complications/physiopathology , Reactive Oxygen Species/metabolism , Sodium Nitrite/blood , Sodium Nitrite/pharmacokinetics , Sodium Nitrite/pharmacology , Tumor Necrosis Factor-alpha/metabolism
2.
Neurogastroenterol Motil ; 25(5): e339-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23551931

ABSTRACT

BACKGROUND: Soluble guanylate cyclase (sGC) is the principal target of nitric oxide (NO) to control gastrointestinal motility. The consequence on nitrergic signaling and gut motility of inducing a heme-free status of sGC, as induced by oxidative stress, was investigated. METHODS: sGCß1 (H105F) knock-in (apo-sGC) mice, which express heme-free sGC that has basal activity, but cannot be stimulated by NO, were generated. KEY RESULTS: Diethylenetriamine NONOate did not increase sGC activity in gastrointestinal tissue of apo-sGC mice. Exogenous NO did not induce relaxation in fundic, jejunal and colonic strips, and pyloric rings of apo-sGC mice. The stomach was enlarged in apo-sGC mice with hypertrophy of the muscularis externa of the fundus and pylorus. In addition, gastric emptying and intestinal transit were delayed and whole-gut transit time was increased in the apo-sGC mice, while distal colonic transit time was maintained. The nitrergic relaxant responses to electrical field stimulation at 1-4 Hz were abolished in fundic and jejunal strips from apo-sGC mice, but in pyloric rings and colonic strips, only the response at 1 Hz was abolished, indicating the contribution of other transmitters than NO. CONCLUSIONS & INFERENCES: The results indicate that the gastrointestinal consequences of switching from a native sGC to a heme-free sGC, which cannot be stimulated by NO, are most pronounced at the level of the stomach establishing a pivotal role of the activation of sGC by NO in normal gastric functioning. In addition, delayed intestinal transit was observed, indicating that nitrergic activation of sGC also plays a role in the lower gastrointestinal tract.


Subject(s)
Gastrointestinal Motility/physiology , Gastroparesis/metabolism , Guanylate Cyclase/metabolism , Heme/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Disease Models, Animal , Gastric Mucosa/metabolism , Gene Knock-In Techniques , Guanylate Cyclase/chemistry , Mice , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Oxidative Stress/physiology , Receptors, Cytoplasmic and Nuclear/chemistry , Soluble Guanylyl Cyclase
SELECTION OF CITATIONS
SEARCH DETAIL
...