Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Free Radic Biol Med ; 89: 241-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26163002

ABSTRACT

BACKGROUND: Inorganic nitrate from exogenous and endogenous sources is accumulated in saliva, reduced to nitrite by oral bacteria and further converted to nitric oxide (NO) and other bioactive nitrogen oxides in the acidic gastric lumen. To further explore the role of oral microbiota in this process we examined the gastric mucus layer in germ free (GF) and conventional mice given different doses of nitrate and nitrite. METHODS: Mice were given either nitrate (100mg/kg/d) or nitrite (0.55-11 mg/kg/d) in the drinking water for 7 days, with the lowest nitrite dose resembling the levels provided by swallowing of fasting saliva. The gastric mucus layer was measured in vivo. RESULTS: GF animals were almost devoid of the firmly adherent mucus layer compared to conventional mice. Dietary nitrate increased the mucus thickness in conventional animals but had no effect in GF mice. In contrast, nitrite at all doses, restored the mucus thickness in GF mice to the same levels as in conventional animals. The nitrite-mediated increase in gastric mucus thickness was not inhibited by the soluble guanylyl cyclase inhibitor ODQ. Mice treated with antibiotics had significantly thinner mucus than controls. Additional studies on mucin gene expression demonstrated down regulation of Muc5ac and Muc6 in germ free mice after nitrite treatment. CONCLUSION: Oral bacteria remotely modulate gastric mucus generation via bioactivation of salivary nitrate. In the absence of a dietary nitrate intake, salivary nitrate originates mainly from NO synthase. Thus, oxidized NO from the endothelium and elsewhere is recycled to regulate gastric mucus homeostasis.


Subject(s)
Gastric Mucosa/pathology , Mucus/cytology , Nitrates/pharmacology , Nitrites/pharmacology , Saliva/cytology , Animals , Anti-Bacterial Agents/pharmacology , Bacteria , Gastric Mucosa/drug effects , Gastric Mucosa/microbiology , Male , Mice , Mucins/metabolism , Mucus/drug effects , Mucus/microbiology , Nitric Oxide/metabolism , Oxidation-Reduction , Real-Time Polymerase Chain Reaction , Saliva/drug effects , Saliva/microbiology
2.
Redox Biol ; 2: 73-81, 2014.
Article in English | MEDLINE | ID: mdl-24494186

ABSTRACT

BACKGROUND: Inorganic nitrate and nitrite have emerged as alternative substrates for nitric oxide (NO) generation in the gastrointestinal tract, and have shown to be protective against drug-induced gastric injury. The aim of this study was to investigate the preventive and therapeutic effects of nitrate and nitrite in a model of experimental colitis. METHODS: Colitis was induced in mice by administrating dextran sulfate sodium (DSS) with concurrent administration of nitrite (1 mM) or nitrate (10 mM) in the drinking water for 7 days. A therapeutic approach was also investigated by initiating nitrite treatment 3 days after DSS-induced colitis. Clinical and inflammatory markers were assessed and the colonic mucus thickness was measured in vivo. The effect of nitrite on wound healing was evaluated using colon epithelial cells. RESULTS: Concurrent administration of DSS and nitrite (1 mM) alleviated inflammation as determined by reduced disease activity index score (DAI) and increased colon length, while nitrate (10 mM) only reduced the DAI-score. Nitrite also displayed therapeutic effects by ameliorating established colonic inflammation with reduced colonic expression of iNOS and improving histopathology. DSS-induced decrease in colonic mucus thickness was completely prevented by nitrite administration. In addition, goblet cell abundance was lower by DSS treatment, but was increased by addition of nitrite. Further studies using colon epithelial cells revealed an NO-dependent improvement in wound healing with nitrite administration. CONCLUSION: Nitrite exerts both preventive and therapeutic effects in colonic inflammation. The protective effects involve preservation of an intact adherent mucus layer and regulation of epithelial cell restitution.


Subject(s)
Colitis/diet therapy , Colitis/prevention & control , Nitrates/administration & dosage , Nitrites/administration & dosage , Animals , Cell Line , Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Dextran Sulfate , Dietary Supplements , Disease Models, Animal , Drug Administration Schedule , Female , Humans , Mice , Mice, Inbred BALB C , Nitrates/therapeutic use , Nitrites/therapeutic use , Wound Healing/drug effects
3.
Free Radic Biol Med ; 60: 195-200, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23466556

ABSTRACT

Hydrogen sulfide (H2S), generated through various endogenous enzymatic and nonenzymatic pathways, is emerging as a regulator of physiological and pathological events throughout the body. Bacteria in the gastrointestinal tract also produce significant amounts of H2S that regulates microflora growth and virulence responses. However, the impact of the microbiota on host global H2S bioavailability and metabolism remains unknown. To address this question, we examined H2S bioavailability in its various forms (free, acid labile, or bound sulfane sulfur), cystathionine γ-lyase (CSE) activity, and cysteine levels in tissues from germ-free versus conventionally housed mice. Free H2S levels were significantly reduced in plasma and gastrointestinal tissues of germ-free mice. Bound sulfane sulfur levels were decreased by 50-80% in germ-free mouse plasma and adipose and lung tissues. Tissue CSE activity was significantly reduced in many organs from germ-free mice, whereas tissue cysteine levels were significantly elevated compared to conventional mice. These data reveal that the microbiota profoundly regulates systemic bioavailability and metabolism of H2S.


Subject(s)
Biological Availability , Cystathionine gamma-Lyase/metabolism , Gastrointestinal Tract/metabolism , Hydrogen Sulfide/metabolism , Metagenome , Adipose Tissue/metabolism , Animals , Cystathionine beta-Synthase/metabolism , Cysteine/metabolism , Gastrointestinal Tract/microbiology , Humans , Hydrogen Sulfide/blood , Lung/metabolism , Lung/microbiology , Mice
4.
Br J Nutr ; 110(9): 1565-72, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-23531375

ABSTRACT

SCFA are important end products formed during colonic fermentation of dietary fibre (DF). It has been suggested that propionic and butyric acids affect metabolic parameters, low-grade systemic inflammation, insulin resistance and obesity. The aim of the present study was to investigate whether the various SCFA profiles observed after fermentation in the caecum of rats fed pectin, guar gum and fructo-oligosaccharides (FOS) were also represented in hepatic portal and aortic serum. The SCFA in serum were extracted using hollow fibre-supported liquid membrane extraction before GLC analysis. The concentrations of acetic, propionic and butyric acids in caecal content correlated well with those in portal serum (P< 0·001) for all the three diets. A weaker correlation was found for propionic and butyric acids between the caecal content and aortic serum (P< 0·05). Butyric acid concentration in caecal content was also reflected in the aortic serum (P= 0·019) of rats fed FOS. FOS gave rather low amounts of the SCFA, especially butyric acid, but caecal tissue weight was higher with FOS than with the other two diets. This may be explained by rapid fermentation and quick utilisation/absorption of the SCFA. The present study also showed that propionic acid was metabolised/utilised to a higher extent than butyric acid by colonocytes before reaching the liver. We conclude that the formation of propionic and butyric acids in the caecum is reflected by increased concentrations in the aortic blood. This approach may therefore simplify the evaluation and study of SCFA from DF in human subjects.


Subject(s)
Aorta/metabolism , Butyric Acid/metabolism , Cecum/metabolism , Dietary Fiber/metabolism , Liver/metabolism , Portal System/metabolism , Propionates/metabolism , Acetic Acid/blood , Acetic Acid/metabolism , Animals , Butyric Acid/blood , Colon/metabolism , Diet , Fermentation , Fructose/blood , Fructose/metabolism , Galactans/blood , Galactans/metabolism , Male , Mannans/blood , Mannans/metabolism , Oligosaccharides/blood , Oligosaccharides/metabolism , Pectins/blood , Pectins/metabolism , Plant Gums/blood , Plant Gums/metabolism , Propionates/blood , Rats , Rats, Wistar
5.
J Nutr ; 142(12): 2135-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23077197

ABSTRACT

A central event in inflammatory bowel disease is the disruption of the mucosal homeostasis. Trefoil peptides [(TFF)] are emerging as key mediators in the defense and repair of the gastrointestinal mucosa. Here, we demonstrate induction of TFF by CLA with therapeutic antiinflammatory effects in a mouse model of inflammatory bowel disease. SW480 cells were treated with linoleic acid or CLA (0-2.5 µmol/L) in the absence or presence of the PPARγ inhibitor GW9662. Cells treated with CLA showed an upregulation of the intestinal trefoil factor, which was prevented by pretreatment with GW9662. Dextran sulfate sodium (2%) was used to induce colitis in mice and they were simultaneously fed with a standard or a CLA-supplemented (100 mg · kg(-1) · d(-1)) diet for 7 d. The CLA-enriched diet prevented the colon shortening induced by DSS and markedly reduced the disease activity index and the colonic expression of inducible NO synthase and NF-κB. Immunohistochemistry revealed an increase in PPARγ and TFF3 expression after CLA administration. Altogether, these results indicate that dietary CLA protects against DSS-induced colitis in a process involving induction of PPARγ and TFF3.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Colitis/drug therapy , Inflammatory Bowel Diseases/drug therapy , Linoleic Acids, Conjugated/administration & dosage , Mucins/physiology , PPAR gamma/physiology , Animals , Dextran Sulfate , Female , Heme Oxygenase-1/physiology , Linoleic Acids, Conjugated/pharmacology , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mucins/analysis , PPAR gamma/analysis , Trefoil Factor-3
6.
Free Radic Biol Med ; 52(3): 683-692, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22178413

ABSTRACT

Nitric oxide (NO) generated by vascular NO synthases can exert anti-inflammatory effects, partly through its ability to decrease leukocyte recruitment. Inorganic nitrate and nitrite, from endogenous or dietary sources, have emerged as alternative substrates for NO formation in mammals. Bioactivation of nitrate is believed to require initial reduction to nitrite by oral commensal bacteria. Here we investigated the effects of inorganic nitrate and nitrite on leukocyte recruitment in microvascular inflammation and in NSAID-induced small-intestinal injury. We show that leukocyte emigration in response to the proinflammatory chemokine MIP-2 is reduced by 70% after 7 days of dietary nitrate supplementation as well as by acute intravenous nitrite administration. Nitrite also reduced leukocyte adhesion to a similar extent and this effect was inhibited by the soluble guanylyl cyclase inhibitor ODQ, whereas the effect on emigrated leukocytes was not altered by this treatment. Further studies in TNF-α-stimulated endothelial cells revealed that nitrite dose-dependently reduced the expression of ICAM-1. In rats and mice subjected to a challenge with diclofenac, dietary nitrate prevented the increase in myeloperoxidase and P-selectin levels in small-intestinal tissue. Antiseptic mouthwash, which eliminates oral nitrate reduction, markedly blunted the protective effect of dietary nitrate on P-selectin levels. Despite attenuation of the acute immune response, the overall ability to clear an infection with Staphylococcus aureus was not suppressed by dietary nitrate as revealed by noninvasive IVIS imaging. We conclude that dietary nitrate markedly reduces leukocyte recruitment to inflammation in a process involving attenuation of P-selectin and ICAM-1 upregulation. Bioactivation of dietary nitrate requires intermediate formation of nitrite by oral nitrate-reducing bacteria and then probably further reduction to NO and other bioactive nitrogen oxides in the tissues.


Subject(s)
Intestine, Small/pathology , Microvessels/pathology , Neutrophil Infiltration/drug effects , Nitrates/pharmacology , Nitrites/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL2 , Cyclic GMP/metabolism , Diclofenac/adverse effects , Dietary Supplements , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression/drug effects , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Intestine, Small/blood supply , Intestine, Small/immunology , Intestine, Small/metabolism , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Microvessels/drug effects , Mouthwashes/pharmacology , Nitrates/administration & dosage , Nitrates/therapeutic use , Nitrites/administration & dosage , Nitrites/therapeutic use , P-Selectin/genetics , P-Selectin/metabolism , Peroxidase/genetics , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL