Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Toxicology ; 264(1-2): 96-103, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19647029

ABSTRACT

Clinical studies suggest that colonic luminal hydrogen sulfide (H(2)S), produced by sulfate-reducing bacteria or through other pathways, might be involved in the pathogenesis of inflammatory bowel disease (IBD). Nonetheless, this hypothesis has been poorly investigated by basic studies using laboratory animals. We thus focused on two enzymes, cystathionine-gamma-lyase (CSE) that generates H(2)S from l-cysteine, and rhodanese that directly or indirectly detoxifies H(2)S, particularly in relation to the colitis induced by dextran sulfate sodium (DSS) in mice. CSE was a major H(2)S-forming enzyme in colonic and renal homogenates from mice and rats, and the rhodanese activity was also detectable in both tissues. Colitis-related symptoms including decreased body weight gain, diarrhea, hematochezia and shortening of colon length were observed in the mice drinking DSS. Those symptoms were not or only slightly attenuated by repeated administration of a CSE inhibitor. CSE activity and protein levels in the colonic tissue did not notably change in the mice with colitis. In contrast, the activity and protein/mRNA levels of rhodanese in the colon, but not kidney, significantly decreased nearly in parallel with the development of colitis, followed by elevation of rhodanese activity in red blood cells (RBCs). These data show that rhodanese, but not CSE, is associated with DSS-induced colitis in mice, leading to a hypothesis that impaired detoxification of H(2)S due to down-regulation or suppression of colonic rhodanese is involved in IBD. The delayed enhancement of rhodanese activity in RBCs, a possible compensative event, might be available as a disease marker for IBD.


Subject(s)
Colitis/chemically induced , Colitis/enzymology , Colon/metabolism , Cystathionine gamma-Lyase/metabolism , Dextran Sulfate/toxicity , Sulfides/metabolism , Sulfides/toxicity , Thiosulfate Sulfurtransferase/metabolism , Anemia/blood , Animals , Blotting, Western , Colitis/pathology , Colon/pathology , Cystathionine gamma-Lyase/blood , Erythrocytes/drug effects , Erythrocytes/enzymology , Hydrogen Sulfide/metabolism , Inactivation, Metabolic , Male , Mice , RNA/biosynthesis , RNA/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Thiosulfate Sulfurtransferase/blood , Weight Gain/drug effects
2.
Toxicology ; 241(1-2): 11-8, 2007 Nov 20.
Article in English | MEDLINE | ID: mdl-17825973

ABSTRACT

We investigated effect of hydrogen sulfide (H(2)S) on oxidative stress-caused cell death in gastric mucosal epithelial cells. In rat normal gastric epithelial RGM1 cells, NaHS, a H(2)S donor, at 1.5mM strongly suppressed hydrogen peroxide (H(2)O(2))-caused cell death, while it slightly augmented the H(2)O(2) toxicity at 0.5-1mM. The protective effect of NaHS was abolished by inhibitors of MEK or JNK, but not of p38 MAP kinase. NaHS at 1.5mM actually phosphorylated ERK and JNK in RGM1 cells. Glibenclamide, an ATP-sensitive K(+) (K(ATP)(+)) channel inhibitor, did not affect the protective effect of NaHS, although mRNAs for K(ATP)(+) channel subunits, Kir6.1 and SUR1, were detected in RGM1 cells. In anesthetized rats, oral administration of NaHS protected against gastric mucosal lesion caused by ischemia-reperfusion. These results suggest that NaHS/H(2)S may protect gastric mucosal epithelial cells against oxidative stress through stimulation of MAP kinase pathways, a therapeutic dose range being very narrow.


Subject(s)
Air Pollutants/pharmacology , Epithelial Cells/drug effects , Gastric Mucosa/pathology , Hydrogen Sulfide/pharmacology , Oxidative Stress/drug effects , ATP-Binding Cassette Transporters/metabolism , Animals , Apoptosis/drug effects , Bisbenzimidazole , Cell Death/drug effects , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Gastric Mucosa/drug effects , Glyburide/pharmacology , Hydrogen Peroxide/toxicity , Hypoglycemic Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , KATP Channels/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Oxidants/toxicity , Potassium Channels, Inwardly Rectifying/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptors, Drug , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonylurea Receptors
3.
Life Sci ; 78(9): 950-7, 2006 Jan 25.
Article in English | MEDLINE | ID: mdl-16188279

ABSTRACT

Proteinase-activated receptor (PAR)-1 or -2 modulates gastrointestinal transit in vivo. To clarify the underlying mechanisms, we characterized contraction/relaxation caused by TFLLR-NH2 and SLIGRL-NH2, PAR-1- and -2-activating peptides, respectively, in gastric and small intestinal (duodenal, jejunal and ileal) smooth muscle isolated from wild-type and PAR-2-knockout mice. Either SLIGRL-NH2 or TFLLR-NH2 caused both relaxation and contraction in the gastrointestinal preparations from wild-type animals. Apamin, a K+ channel inhibitor, tended to enhance the peptide-evoked contraction in some of the gastrointestinal preparations, whereas it inhibited relaxation responses to either peptide completely in the stomach, but only partially in the small intestine. Indomethacin reduced the contraction caused by SLIGRL-NH2 or TFLLR-NH2 in both gastric and ileal preparations, but unaffected apamin-insensitive relaxant effect of either peptide in ileal preparations. Repeated treatment with capsaicin suppressed the contractile effect of either peptide in the stomach, but not clearly in the ileum, whereas it enhanced the apamin-insensitive relaxant effect in ileal preparations. In any gastrointestinal preparations from PAR-2-knockout mice, SLIGRL-NH2 produced no responses. Thus, the inhibitory component in tension modulation by PAR-1 and -2 involves both apamin-sensitive and -insensitive mechanisms in the small intestine, but is predominantly attributable to the former mechanism in the stomach. The excitatory component in the PAR-1 and -2 modulation may be mediated, in part, by activation of capsaicin-sensitive sensory nerves and/or endogenous prostaglandin formation. Our study thus clarifies the multiple mechanisms for gastrointestinal motility modulation by PAR-1 and -2, and also provides ultimate evidence for involvement of PAR-2.


Subject(s)
Gastrointestinal Motility/drug effects , Receptor, PAR-1/drug effects , Receptor, PAR-2/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apamin/pharmacology , Calcium Channel Blockers/pharmacology , Capsaicin/pharmacology , Female , In Vitro Techniques , Indomethacin/pharmacology , Intestines/drug effects , Isometric Contraction/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Relaxation/drug effects , Neurons, Afferent/drug effects , Oligopeptides/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Prostaglandins/pharmacology , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-2/agonists , Receptor, PAR-2/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Stomach/drug effects
4.
Br J Pharmacol ; 144(2): 212-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15655521

ABSTRACT

1. Proteinase-activated receptor-2 (PAR(2)), expressed in capsaicin-sensitive sensory neurons, plays a protective role in gastric mucosa. The present study evaluated gastric mucosal cytoprotective effect of 2-furoyl-LIGRL-NH(2), a novel highly potent PAR(2) agonist, in ddY mice and in wild-type and PAR(2)-knockout mice of C57BL/6 background. 2. Gastric mucosal injury was created by oral administration of HCl/ethanol solution in the mice. The native PAR(2)-activating peptide SLIGRL-NH(2), administered intraperitoneally (i.p.) at 0.3-1 micromol kg(-1) in combination with amastatin, an aminopeptidase inhibitor, but not alone, revealed gastric mucosal protection in ddY mice, which was abolished by ablation of capsaicin-sensitive sensory neurons. 3. I.p. administration of 2-furoyl-LIGRL-NH(2) at 0.1 micromol kg(-1), without combined treatment with amastatin, exhibited gastric mucosal cytoprotective activity in ddY mice, the potency being much greater than SLIGRL-NH(2) in combination with amastatin. This effect was also inhibited by capsaicin pretreatment. 4. Oral administration of 2-furoyl-LIGRL-NH(2) at 0.003-0.03 micromol kg(-1) also protected against gastric mucosal lesion in a capsaicin-reversible manner in ddY mice. 5. I.p. 2-furoyl-LIGRL-NH(2) at 0.1-0.3 micromol kg(-1) caused prompt salivation in anesthetized mice, whereas its oral administration at 0.003-1 micromol kg(-1) was incapable of eliciting salivation. 6. In wild-type, but not PAR(2)-knockout, mice of C57BL/6 background, i.p. administration of 2-furoyl-LIGRL-NH(2) caused gastric mucosal protection. 7. Thus, 2-furoyl-LIGRL-NH(2) is considered a potent and orally available gastric mucosal protective agent. Our data also substantiate a role for PAR(2) in gastric mucosal protection and the selective nature of 2-furoyl-LIGRL-NH(2).


Subject(s)
Cytoprotection/drug effects , Gastric Mucosa/drug effects , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Animals , Cytoprotection/physiology , Dose-Response Relationship, Drug , Female , Gastric Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, PAR-2/physiology
5.
J Pharmacol Exp Ther ; 309(3): 1098-107, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14976227

ABSTRACT

To develop potent and metabolically stable agonists for protease-activated receptor-2 (PAR-2), we prepared 2-furoylated (2f) derivatives of native PAR-2-activating peptides, 2f-LIGKV-OH, 2f-LIGRL-OH, 2f-LIGKV-NH(2), and 2f-LIGRL-NH(2), and systematically evaluated their activity in PAR-2-responsive cell lines and tissues. In both HCT-15 cells and NCTC2544 cells overexpressing PAR-2, all furoylated peptides increased cytosolic Ca(2+) levels with a greater potency than the corresponding native peptides, although a similar maximum response was recorded. The absolute potency of each peptide was greater in NCTC2544, possibly due to a higher level of receptor expression. Furthermore, the difference in potency between the 2-furoylated peptides and the native peptides was enhanced when evaluated in the rat superior mesenteric artery and further increased when measuring PAR-2-mediated salivation in ddY mice in vivo. The potency of 2f-LIGRL-NH(2), the most powerful peptide, relative to SLIGKV-OH, was about 100 in the cultured cell Ca(2+) signaling assays, 517 in the vasorelaxation assay, and 1100 in the salivation assay. Amastatin, an aminopeptidase inhibitor, augmented salivation caused by native peptides, but not furoylated peptides. The PAR-2-activating peptides, including the furoylated derivatives, also produced salivation in the wild-type C57BL/6 mice, but not the PAR-2-deficient mice. Our data thus demonstrate that substitution of the N-terminal serine with a furoyl group in native PAR-2-activating peptides dramatically enhances the agonistic activity and decreases degradation by aminopeptidase, leading to development of 2f-LIGRL-NH(2), the most potent peptide. Furthermore, the data from PAR-2-deficient mice provide ultimate evidence for involvement of PAR-2 in salivation and the selective nature of the 2-furoylated peptides.


Subject(s)
Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Aminopeptidases/metabolism , Animals , Calcium Signaling/physiology , Cell Line , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Male , Mesenteric Arteries/drug effects , Mice , Mice, Inbred C57BL , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Tumor Cells, Cultured , Vasoconstriction/drug effects , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...