Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Gastroenterology ; 141(4): 1314-22, 1322.e1-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745447

ABSTRACT

BACKGROUND & AIMS: Proton pump inhibitors (PPIs) and nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most commonly used classes of drugs, with the former frequently coprescribed to reduce gastroduodenal injury caused by the latter. However, suppression of gastric acid secretion by PPIs is unlikely to provide any protection against the damage caused by NSAIDs in the more distal small intestine. METHODS: Rats were treated with antisecretory doses of omeprazole or lanzoprazole for 9 days, with concomitant treatment with anti-inflammatory doses of naproxen or celecoxib on the final 4 days. Small intestinal damage was blindly scored, and changes in hematocrit were measured. Changes in small intestinal microflora were evaluated by denaturing gradient gel electrophoresis and reverse-transcription polymerase chain reaction. RESULTS: Both PPIs significantly exacerbated naproxen- and celecoxib-induced intestinal ulceration and bleeding in the rat. Omeprazole treatment did not result in mucosal injury or inflammation; however, there were marked shifts in numbers and types of enteric bacteria, including a significant reduction (∼80%) of jejunal Actinobacteria and Bifidobacteria spp. Restoration of small intestinal Actinobacteria numbers through administration of selected (Bifidobacteria enriched) commensal bacteria during treatment with omeprazole and naproxen prevented intestinal ulceration/bleeding. Colonization of germ-free mice with jejunal bacteria from PPI-treated rats increased the severity of NSAID-induced intestinal injury, as compared with mice colonized with bacteria from vehicle-treated rats. CONCLUSIONS: PPIs exacerbate NSAID-induced intestinal damage at least in part because of significant shifts in enteric microbial populations. Prevention or reversal of this dysbiosis may be a viable option for reducing the incidence and severity of NSAID enteropathy.


Subject(s)
Actinobacteria/drug effects , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Bifidobacterium/drug effects , Gastrointestinal Hemorrhage/chemically induced , Jejunum/drug effects , Peptic Ulcer/chemically induced , Proton Pump Inhibitors/toxicity , 2-Pyridinylmethylsulfinylbenzimidazoles/toxicity , Actinobacteria/genetics , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Bifidobacterium/genetics , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Celecoxib , Colon/drug effects , Colon/microbiology , Denaturing Gradient Gel Electrophoresis , Disease Models, Animal , Drug Interactions , Gastrointestinal Hemorrhage/microbiology , Gastrointestinal Hemorrhage/pathology , Gastrointestinal Hemorrhage/prevention & control , Hematocrit , Jejunum/microbiology , Jejunum/pathology , Lansoprazole , Male , Naproxen/toxicity , Omeprazole/toxicity , Peptic Ulcer/microbiology , Peptic Ulcer/pathology , Peptic Ulcer/prevention & control , Probiotics/pharmacology , Pyrazoles/toxicity , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/toxicity , Time Factors
2.
Gastroenterology ; 137(2): 569-78, 578.e1, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19375422

ABSTRACT

BACKGROUND & AIMS: Hydrogen sulfide (H(2)S) is an endogenous gaseous mediator of mucosal defense with antiinflammatory effects that promote ulcer healing. The effects of H(2)S during the pathogenesis of colitis have not been established. We analyzed the contribution of H(2)S to inflammation and ulceration of the colon in a rat model of colitis. METHODS: Colitis was induced by intracolonic administration of trinitrobenzene sulfonic acid. The ability of the colon to synthesize H(2)S was studied over the course of the resolution of the colitis. Expression of 2 enzymes involved in the synthesis of H(2)S and the effects of inhibitors of these enzymes were examined. We also examined the effects of H(2)S donors on the resolution of colitis. RESULTS: The capacity for the colon to produce H(2)S increased markedly over the first days after induction of colitis and then declined toward control levels as the colitis was resolved. Inhibition of colonic H(2)S synthesis markedly exacerbated the colitis, resulting in significant mortality. Inhibition of H(2)S synthesis in healthy rats resulted in inflammation and mucosal injury in the small intestine and colon along with down-regulation of cyclooxygenase-2 messenger RNA expression and prostaglandin synthesis. Intracolonic administration of H(2)S donors significantly reduced the severity of colitis and reduced colonic expression of messenger RNA for the proinflammatory cytokine tumor necrosis factor alpha. CONCLUSIONS: In rats, H(2)S modulates physiological inflammation and contributes to the resolution of colitis.


Subject(s)
Colitis/drug therapy , Colitis/pathology , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Analysis of Variance , Animals , Biomarkers/metabolism , Biopsy, Needle , Colitis/enzymology , Colitis/mortality , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Glyburide/pharmacology , Immunohistochemistry , Male , Pinacidil/pharmacology , Probability , Random Allocation , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Statistics, Nonparametric , Survival Rate , Treatment Outcome , Trinitrobenzenesulfonic Acid/pharmacology , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism
3.
FASEB J ; 21(14): 4070-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17634391

ABSTRACT

Hydrogen sulfide is an endogenous mediator that relaxes vascular smooth muscle, exhibits several antiinflammatory activities, and contributes to gastric mucosal defense. This study was performed to examine the role of hydrogen sulfide in the resolution of injury; specifically, the healing of gastric ulcers. Ulcers were induced in rats by serosal application of acetic acid. This elicited a marked increase in gastric expression of the two key enzymes in hydrogen sulfide synthesis (cystathionine-beta-synthase and cystathionine-gamma-lyase) and in hydrogen sulfide synthesis. Twice-daily treatment for a week with hydrogen sulfide donors significantly increased the extent of healing of gastric ulcers as compared to vehicle-treatment. Similar treatment with L-cysteine, a precursor for hydrogen sulfide, also accelerated healing of the ulcers, and the effect was abolished by cotreatment with an inhibitor of cystathionine-gamma-lyase. The beneficial effects of hydrogen sulfide on ulcer healing were not dependent on nitric oxide synthesis, nor did they appear to occur through activation of ATP-sensitive K+ channels. These results suggest that hydrogen sulfide is produced in the gastric mucosa in response to injury and acts to promote healing. The results further suggest that drugs releasing hydrogen sulfide could be employed to accelerate healing of gastric ulcers, and possibly of other wounds.


Subject(s)
Hydrogen Sulfide/pharmacology , Stomach Ulcer/metabolism , Wound Healing/drug effects , Acetic Acid/administration & dosage , Acetic Acid/toxicity , Animals , Hydrogen Sulfide/metabolism , Intubation, Gastrointestinal , Male , Organothiophosphorus Compounds/pharmacology , Rats , Rats, Wistar , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/enzymology , Thioamides/pharmacology , Wound Healing/physiology
4.
Br J Pharmacol ; 148(3): 274-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16565732

ABSTRACT

1. Platelets contain an array of growth factors that can modulate healing processes, including both pro- (e.g., vascular endothelial growth factor (VEGF)) and antiangiogenic (e.g., endostatin) factors. Previous studies have shown that circulating platelets contribute significantly to gastric ulcer healing, acting as a delivery system for these growth factors to the site of injury. In this study, we examined the effects of orally administered human platelets on the healing of gastric ulcers in rats, and determined the contribution of VEGF and endostatin to healing in this model. 2. Twice-daily administration of human platelets significantly accelerated ulcer healing, but platelet-poor plasma (PPP), lysed platelets and serum failed to produce this effect. There was no correlation between ulcer healing and the levels of VEGF or endostatin in serum, PPP or platelet-rich plasma (PRP). 3. Accelerated ulcer healing could not be produced by oral administration of the angiogenic factors themselves, at concentrations matching those in PRP. 4. The accelerated healing induced by platelets could be reversed by immuno-neutralization of VEGF. In contrast, immuno-neutralization of endostatin did not affect PRP-induced ulcer healing. 5. These studies indicate that VEGF released from platelets accounts for the accelerated healing of gastric ulcers. However, as intact (rather than lysed) platelets were required for the accelerated healing, the presentation of VEGF by the platelet at the site of injury appears to be crucial for enhancement of the healing process.


Subject(s)
Blood Platelets/physiology , Stomach Ulcer/therapy , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Blood Platelets/chemistry , Drug Delivery Systems , Endostatins/administration & dosage , Endostatins/pharmacology , Humans , Male , Platelet Transfusion , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/pharmacology
5.
Proc Natl Acad Sci U S A ; 102(1): 216-20, 2005 Jan 04.
Article in English | MEDLINE | ID: mdl-15615851

ABSTRACT

The roles of proteinase-activated receptors (PARs) in platelet functions other than aggregation are not well understood. Among these is the release of factors that regulate the process of angiogenesis, such as endostatin and VEGF, which, respectively, inhibit and promote angiogenesis. PAR1 and PAR4 are expressed on the surface of human platelets and can be activated by thrombin. In the present study, we have attempted to determine the roles of PAR1 and PAR4 in regulating release of endostatin and VEGF from human platelets. Aggregation and endostatin release could be elicited by a specific PAR4 agonist (AYPGKF-NH(2)). The PAR4 agonist concentration dependently suppressed VEGF release. A selective PAR1 agonist (TFLLR-NH(2)) induced platelet aggregation and VEGF release but suppressed endostatin release. Thrombin did not affect endostatin or VEGF release. However, in the presence of a selective PAR1 antagonist (SCH79797), thrombin stimulated endostatin release and suppressed VEGF release. Conversely, in the presence of a selective PAR4 antagonist (transcinnamoyl-YPGKF-NH(2)), thrombin stimulated VEGF release. In vivo, treatment of rats with established gastric ulcers with a PAR1 antagonist each day for 1 wk resulted in a significant retardation of healing. We conclude that PAR1 and PAR4 counter-regulate the release of endostatin and VEGF from platelets. These protease-activated receptors could therefore play a crucial role in regulating angiogenesis and in turn could regulate the processes of wound healing and tumor growth.


Subject(s)
Blood Platelets/metabolism , Endostatins/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Humans , Receptors, Thrombin/agonists
6.
Am J Physiol Gastrointest Liver Physiol ; 286(1): G76-81, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14665439

ABSTRACT

Aceylation of cyclooxygenase (COX)-2 by aspirin can trigger the formation of 15(R)-epilipoxin A4, or aspirin-triggered lipoxin (ATL). ATL exerts protective effects in the stomach. Selective COX-2 inhibitors block ATL synthesis and exacerbate aspirin-induced gastric damage. Nitric oxide-releasing aspirins, including NCX-4016, have antiplatelet effects similar to aspirin but do not cause gastric damage. In the present study, we examined whether or not NCX-4016 triggers ATL synthesis and/or upregulates gastric COX-2 expression and the effects of coadministration of NCX-4016 with a selective COX-2 inhibitor on gastric mucosal injury and inflammation. Rats were given aspirin or NCX-4016 orally and either vehicle or a selective COX-2 inhibitor (celecoxib) intraperitoneally. Gastric damage was blindly scored, and granulocyte infiltration into gastric tissue was monitored through measurement of myeloperoxidase activity. Gastric PG and ATL synthesis was measured as was COX-2 expression. Whereas celecoxib inhibited gastric ATL synthesis and increased the severity of aspirin-induced gastric damage and inflammation, coadministration of celecoxib and NCX-4016 did not result in damage or inflammation. NCX-4016 did not upregulate gastric COX-2 expression nor did it trigger ATL synthesis (in contrast to aspirin). Daily administration of aspirin for 5 days resulted in significantly less gastric damage than that seen with a single dose, as well as augmented ATL synthesis. Celecoxib reversed this effect. In contrast, repeated administration of NCX-4016 failed to cause gastric damage, whether given alone or with celecoxib. These studies support the notion that NCX-4016 may be an attractive alternative to aspirin for indications such as cardioprotection, including in individuals also taking selective COX-2 inhibitors.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Gastric Mucosa/pathology , Gastritis/pathology , Isoenzymes/metabolism , Nitric Oxide/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Aspirin/analogs & derivatives , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Isoenzymes/biosynthesis , Lipoxins/biosynthesis , Male , Neutropenia/metabolism , Neutrophil Infiltration/drug effects , Peroxidase/metabolism , Prostaglandin-Endoperoxide Synthases/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
7.
Gastroenterology ; 123(5): 1598-606, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12404234

ABSTRACT

BACKGROUND & AIMS: Cyclooxygenase-2 (COX-2) has been implicated as contributing to mucosal defense. Acetylation of COX-2 by aspirin can result in production of an antiinflammatory substance, 15(R)-epi-LXA4. We determined whether aspirin-triggered lipoxin (LX) production via COX-2 diminishes aspirin-induced damage in the rat stomach. METHODS: Rats were treated with aspirin plus or minus celecoxib or rofecoxib. Gastric generation of LXA4 was measured. Effect of exogenous LXA4 or an LXA4 receptor antagonist on gastric resistance to aspirin-induced damage was examined. Aspirin-induced leukocyte adherence in mesenteric venules, and the effects of LXA4, were examined by intravital microscopy. RESULTS: Celecoxib and rofecoxib significantly increased the severity of aspirin-induced gastric damage. Aspirin rapidly up-regulated COX-2 expression in the stomach and caused a significant increase in gastric 15(R)-epi-LXA4 production, which was abolished by celecoxib. LXA4 dose dependently (0.25-2.5 microg/kg, intraperitoneally) reduced the severity of aspirin-induced gastric damage and suppressed aspirin-induced leukocyte adherence, whereas an LXA4 antagonist had the opposite effects. CONCLUSIONS: Aspirin administration results in elevated production of 15(R)-epi-LXA4 via COX-2. LXA4 exerts very potent protective actions on the gastric mucosa. Co-administration of aspirin and a selective COX-2 inhibitor results in substantially more severe gastric injury than is produced with either agent alone.


Subject(s)
Aspirin/adverse effects , Cyclooxygenase Inhibitors/adverse effects , Hydroxyeicosatetraenoic Acids/physiology , Isoenzymes/metabolism , Lipoxins , Prostaglandin-Endoperoxide Synthases/metabolism , Stomach/drug effects , Animals , Aspirin/pharmacology , Celecoxib , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Drug Resistance/physiology , Gastric Mucosa/metabolism , Hydroxyeicosatetraenoic Acids/biosynthesis , Hydroxyeicosatetraenoic Acids/pharmacology , Isoenzymes/antagonists & inhibitors , Lactones/pharmacology , Leukocytes/drug effects , Leukocytes/physiology , Male , Membrane Proteins , Pyrazoles , Rats , Rats, Wistar , Stomach/pathology , Sulfonamides/pharmacology , Sulfones
SELECTION OF CITATIONS
SEARCH DETAIL
...