Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Life Sci ; 275: 119370, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-33744322

ABSTRACT

BACKGROUND: This study aimed to investigate the gastroprotective effect of chlorogenic acid (CGA) against Indomethacin (IND)-induced gastric ulcer (GU) in rats and its underlying mechanism, especially through autophagic and apoptotic pathways. METHODS: Seventy-five rats were divided into five groups; control, IND (50 mg/kg, p.o.), CGA (100 mg/kg, p.o., 14 days), IND pretreated with CGA (50 mg/kg or 100 mg/kg, p.o., 14 days). The stomach tissues were examined to calculate the ulcer index and analyze markers of autophagy (beclin-1, LC3-II/LC3-I and p62), lysosomal function (cathepsin-D) and apoptosis (Bcl-2, Bax and caspase-3), along with expression of Akt/mTOR pathway using western blot or ELISA techniques. In addition, viability of gastric mucosal cells was detected by flowcytometry. Structural changes were assessed histologically, while autophagic and apoptotic changes of gastric mucosa were observed by transmission electron microscopy. RESULTS: CGA exhibited a dose-dependent gastroprotective effect by reversing IND-induced accumulation of autophagic vacuoles, significant reduction in beclin-1, LC3-II/LC3-I, and p62 levels, and down-regulation of p-Akt/p-mTOR expression. CGA100 also restored normal autolysosomal function by modulation of cathepsin-D levels. Furthermore, pretreatment with CGA100 was significantly associated with an increase in antiapoptotic protein Bcl-2 along with a decrease in proapoptotic Bax and caspase-3 proteins in such a way that impairs IND-induced apoptosis. This was confirmed by CGA-induced significant decrease in annexin V+ cells. CONCLUSIONS: The natural compound CGA offers a novel gastroprotective intervention against IND-induced GU through restoration of normal autophagic flux, impairment of apoptosis in a crosstalk mechanism mediated by Akt/mTOR pathway reactivation, and alleviation of IND-induced lysosomal dysfunction.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Chlorogenic Acid/therapeutic use , Indomethacin/adverse effects , Stomach Ulcer/chemically induced , Animals , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Indomethacin/antagonists & inhibitors , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Stomach Ulcer/pathology , Stomach Ulcer/prevention & control , TOR Serine-Threonine Kinases/metabolism
2.
Antioxid Redox Signal ; 32(11): 766-784, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31830804

ABSTRACT

Aims: Nonsteroidal anti-inflammatory drugs (NSAIDs), among the most commonly used drugs worldwide, are associated with gastrointestinal (GI) complications that severely limit the clinical utility of this essential class of pain medications. Here, we mechanistically dissect the protective impact of a natural product, malabaricone C (Mal C), on NSAID-induced gastropathy. Results: Mal C dose dependently diminished erosion of the stomach lining and inflammation in mice treated with NSAIDs with the protective impact translating to improvement in survival. By decreasing oxidative and nitrative stress, Mal C treatment prevented NSAID-induced mitochondrial dysfunction and cell death; nuclear factor κ-light-chain enhancer of activated B cell induction, release of proinflammatory cytokines and neutrophil infiltration; and disruptions in the vascular endothelial growth factor/endostatin balance that contributes to mucosal autohealing. Importantly, Mal C failed to impact the therapeutic anti-inflammatory properties of multiple NSAIDs in a model of acute inflammation. In all assays tested, Mal C proved as or more efficacious than the current first-line therapy for NSAID-dependent GI complications, the proton pump inhibitor omeprazole. Innovation: Given that omeprazole-mediated prophylaxis is, itself, associated with a shift in NSAID-driven GI complications from the upper GI to the lower GI system, there is a clear and present need for novel therapeutics aimed at ameliorating NSAID-induced gastropathy. Mal C provided significant protection against NSAID-induced gastric ulcerations impacting multiple critical signaling cascades contributing to inflammation, cell loss, extracellular matrix degradation, and angiogenic autohealing. Conclusion: Thus, Mal C represents a viable lead compound for the development of novel gastroprotective agents.


Subject(s)
Indomethacin/antagonists & inhibitors , Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Protective Agents/pharmacology , Resorcinols/pharmacology , Stomach Ulcer/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Death/drug effects , Dose-Response Relationship, Drug , Indomethacin/administration & dosage , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/pathology , Oxidative Stress/drug effects , Protective Agents/administration & dosage , Protective Agents/metabolism , Resorcinols/administration & dosage , Resorcinols/metabolism , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Wound Healing/drug effects
3.
Arch Pharm Res ; 41(10): 1019-1031, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30109575

ABSTRACT

Indomethacin is a non-steroidal anti-inflammatory drug with clearly known side effects on the gastrointestinal tract. The purpose of the present study was to investigate whether eupatilin inhibit cell injury induced by indomethacin in cultured feline esophageal epithelial cells (EECs). EECs were used to investigate the ability of eupatilin to induce the expression of heat shock proteins (HSP27 and HSP70) and analyze its cytoprotective effect against indomethacin-induced damage. The treatment of EECs with indomethacin for 8 h decreased cell viability. Western blot analysis showed that the levels of HSPs gradually decreased in cells treated with indomethacin, while eupatilin treatment increased the levels of HSPs. When treated with both indomethacin and eupatilin, the levels of HSPs increased rapidly, and were maintained at 130-140%. In addition, treatment with the specific inhibitors of PTK, PKC, PLC, p38 MAPK, JNKs, and PI3K attenuated the eupatilin-induced expression of HSPs. Pretreatment of EECs with the inhibitors of protein synthesis, actinomycin D or cycloheximide, attenuated the cytoprotective effect of eupatilin on indomethacin-induced cell damage. Reactive oxygen species production was upregulated by indomethacin, but downregulated by eupatilin. Taken together, it was suggested that HSPs were partly responsible for the eupatilin-mediated cytoprotective activity against the indomethacin-induced damage in EECs.


Subject(s)
Cytoprotection/drug effects , Epithelial Cells/drug effects , Esophagus/cytology , Flavonoids/pharmacology , HSP27 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/biosynthesis , Indomethacin/antagonists & inhibitors , Animals , Benzophenanthridines/pharmacology , Cats , Cell Survival/drug effects , Cells, Cultured , Estrenes/pharmacology , Female , Flavonoids/antagonists & inhibitors , Fluorescent Antibody Technique , HSP27 Heat-Shock Proteins/analysis , HSP27 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/analysis , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Indomethacin/pharmacology , Male , Pyrrolidinones/pharmacology , Tyrphostins/pharmacology
4.
Chem Biol Interact ; 273: 266-272, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28648817

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) consumption has been commonly associated with gastric mucosal lesions including gastric ulcer. Silymarin (SM) is a flavonoid mixture with anti-oxidant and anti-inflammatory activities which explain its protective role against hepatic and renal injuries. However, its impact on gastric ulcer has not yet been elucidated. Thus we went further to investigate the potential protective effects of SM against indomethacin-induced gastric injury in rats. Pretreatment with SM (50 mg/kg orally) attenuated the severity of gastric mucosal damage as evidenced by decreasing ulcer index (UI) and ulcer score, improvement of disturbed histopathologicl features to be insignificant with those induced by the reference anti-ulcer drug. Pretreatment with SM also suppressed gastric inflammation by decreasing myeloperoxidase activity, tumer necrosis factor-α (TNF- α) and interleukin 6 (IL6) levels along with nuclear factor kappa B p65 (NF-κB) expression. Meanwhile, SM prevent gastric oxidative stress via inhibition of lipid peroxides formation, enhancement of glutathione peroxidase, superoxide dismutase activities and up-regulation of nuclear factor-erythroid-2-related factor 2 (Nrf2), the redox-sensitive master regulator of oxidative stress signaling. In conclusion, the results herein revealed that SM has a gastro-protective effect which is mediated via suppression of gastric inflammation, oxidative stress, increased the anti-oxidant and the cyto-protective defense mechanisms.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Inflammation/drug therapy , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Silymarin/pharmacology , Stomach Ulcer/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Indomethacin/antagonists & inhibitors , Inflammation/metabolism , Inflammation/pathology , Silybum marianum/chemistry , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Silymarin/administration & dosage , Silymarin/isolation & purification , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism
5.
Mem Inst Oswaldo Cruz ; 109(5): 668-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25141285

ABSTRACT

Eicosanoids affect the immunity of several pathogen/insect models, but their role on the Anopheles gambiae response to Plasmodium is still unknown. Plasmodium berghei-infected mosquitoes were injected with an eicosanoid biosynthesis inhibitor, indomethacin (IN), or a substrate, arachidonic acid (AA), at day 7 or day 12 post-infection (p.i.). Salivary gland invasion was evaluated by sporozoite counts at day 21 p.i. IN promoted infection upon sporozoite release from oocysts, but inhibited infection when sporozoites were still maturing within the oocysts, as observed by a reduction in the number of sporozoites reaching the salivary glands. AA treatment had the opposite effect. We show for the first time that An. gambiae can modulate parasite survival through eicosanoids by exerting an antagonistic or agonistic effect on the parasite, depending on its stage of development.


Subject(s)
Anopheles/parasitology , Eicosanoids/pharmacology , Insect Vectors/parasitology , Oocysts/drug effects , Plasmodium berghei/drug effects , Salivary Glands/parasitology , Animals , Anopheles/drug effects , Arachidonic Acid/pharmacology , Indomethacin/antagonists & inhibitors , Indomethacin/pharmacology , Oocysts/growth & development , Plasmodium berghei/physiology
6.
Toxicol Mech Methods ; 24(8): 560-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25137224

ABSTRACT

Escin, a natural mixture of triterpenoid saponin isolated from the seed of the horse chestnut, is reported to have a potent antiulcer activity against ethanol-induced gastric mucosal lesions. This study investigated the possible mechanisms underlying the gastroprotective effect of escin against indomethacin-induced gastric ulcer in mice. Gastric ulceration was induced by a single intragastric administration of indomethacin (18 mg/kg). The mice underwent intragastric treatment with escin at doses of 0.45, 0.9 or 1.8 mg/kg. Gastric lesion was estimated morphometrically and histopathologically 6 h after the indomethacin administration. The antioxidative parameters in gastric mucosa were measured. Moreover, the activity of myeloperoxidase and the contents of TNF-α, P-selectin and VCAM-1 in gastric tissues were determined. The results showed that escin protected gastric tissues against indomethacin-induced gastropathy as demonstrated from a reduction in the ulcer index and an attenuation of histopathologic changes. Escin caused significant reductions of the contents of malondialdehyde, TNF-α, P-selectin, VCAM-1 and myeloperoxidase activity. The altered activities of superoxide dismutase, catalase and glutathione peroxidase in the stomach tissues were also ameliorated by escin treatment. The present study demonstrated that escin had a protective effect against indomethacin-induced gastric ulcer in mice, not only by virtue of its antioxidant potential, but also due to its anti-inflammatory effect.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Ulcer Agents/therapeutic use , Escin/therapeutic use , Gastric Mucosa/drug effects , Indomethacin/antagonists & inhibitors , Protective Agents/therapeutic use , Stomach Ulcer/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Ulcer Agents/administration & dosage , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Dose-Response Relationship, Drug , Escin/administration & dosage , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , H(+)-K(+)-Exchanging ATPase/chemistry , H(+)-K(+)-Exchanging ATPase/metabolism , Indomethacin/adverse effects , Lipid Peroxidation/drug effects , Mice , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , P-Selectin/antagonists & inhibitors , P-Selectin/chemistry , P-Selectin/metabolism , Protective Agents/administration & dosage , Random Allocation , Severity of Illness Index , Stomach/drug effects , Stomach/immunology , Stomach/pathology , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/chemistry , Vascular Cell Adhesion Molecule-1/metabolism
7.
Mem. Inst. Oswaldo Cruz ; 109(5): 668-671, 19/08/2014. graf
Article in English | LILACS | ID: lil-720428

ABSTRACT

Eicosanoids affect the immunity of several pathogen/insect models, but their role on the Anopheles gambiae response to Plasmodium is still unknown. Plasmodium berghei-infected mosquitoes were injected with an eicosanoid biosynthesis inhibitor, indomethacin (IN), or a substrate, arachidonic acid (AA), at day 7 or day 12 post-infection (p.i.). Salivary gland invasion was evaluated by sporozoite counts at day 21 p.i. IN promoted infection upon sporozoite release from oocysts, but inhibited infection when sporozoites were still maturing within the oocysts, as observed by a reduction in the number of sporozoites reaching the salivary glands. AA treatment had the opposite effect. We show for the first time that An. gambiae can modulate parasite survival through eicosanoids by exerting an antagonistic or agonistic effect on the parasite, depending on its stage of development.


Subject(s)
Animals , Anopheles/parasitology , Eicosanoids/pharmacology , Insect Vectors/parasitology , Oocysts/drug effects , Plasmodium berghei/drug effects , Salivary Glands/parasitology , Anopheles/drug effects , Arachidonic Acid/pharmacology , Indomethacin/antagonists & inhibitors , Indomethacin/pharmacology , Oocysts/growth & development , Plasmodium berghei/physiology
8.
J Agric Food Chem ; 61(22): 5291-7, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23668856

ABSTRACT

The aim of this study was to determine the protective effect of quercetin, epigallocatechingallate, resveratrol, and rutin against the disruption of epithelial integrity induced by indomethacin in Caco-2 cell monolayers. Indomethacin decreased the transepithelial electrical resistance and increased the permeability of the monolayers to fluorescein-dextran. These alterations were abolished by all the tested polyphenols but rutin, with quercetin being the most efficient. The protective effect of quercetin was associated with its capacity to inhibit the redistribution of ZO-1 protein induced in the tight junction by indomethacin or rotenone, a mitochondrial complex-I inhibitor, and to prevent the decrease of ZO-1 and occludin expression induced by indomethacin. The fact that the antioxidant polyphenols assayed in this study differ in their protective capacity against the epithelial damage induced by indomethacin suggests that this damage is due to the ability of this agent to induce not only oxidative stress but also mitochondrial dysfunction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Antioxidants/metabolism , Enterocytes/metabolism , Functional Food/analysis , Indomethacin/antagonists & inhibitors , Occludin/metabolism , Zonula Occludens-1 Protein/metabolism , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antioxidants/analysis , Caco-2 Cells , Catechin/analogs & derivatives , Catechin/analysis , Catechin/metabolism , Enterocytes/drug effects , Gene Expression Regulation/drug effects , Humans , Indomethacin/adverse effects , Occludin/agonists , Occludin/antagonists & inhibitors , Occludin/genetics , Permeability/drug effects , Polyphenols/analysis , Polyphenols/metabolism , Protein Transport/drug effects , Quercetin/analysis , Quercetin/metabolism , Resveratrol , Stilbenes/analysis , Stilbenes/metabolism , Tight Junctions/drug effects , Zonula Occludens-1 Protein/agonists , Zonula Occludens-1 Protein/antagonists & inhibitors , Zonula Occludens-1 Protein/genetics
9.
Life Sci ; 90(1-2): 8-12, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22036620

ABSTRACT

AIMS: Recent evidence has shown that systemic administration of sulfonylureas and biguanides block the diclofenac-induced antinociception, but not the effect produced by indomethacin. However, there are no reports about the peripheral interaction between analgesics and the biguanides metformin and phenformin. Therefore, this work was undertaken to determine whether glibenclamide and glipizide and the biguanides metformin and phenformin have any effect on the peripheral antinociception induced by diclofenac and indomethacin. MAIN METHODS: Diclofenac and indomethacin were administered locally in the formalin-injured rat paw, and the antinociceptive effect was evaluated using the 1% formalin test. To determine whether peripheral antinociception induced by diclofenac or indomethacin was mediated by either the ATP-sensitive K(+) channels or biguanides-induced mechanisms, the effect of pretreatment with the appropriates vehicles or glibenclamide, glipizide, metformin and phenformin on the antinociceptive effect induced by local peripheral diclofenac and indomethacin was assessed. KEY FINDINGS: Local peripheral injections of diclofenac (50-200 µg/paw) and indomethacin (200-800 µg/paw) produced a dose-dependent antinociception during the second phase of the test. Local pretreatment with glibenclamide, glipizide, metformin and phenformin blocked the diclofenac-induced antinociception. On the other hand, the pretreatment with glibenclamide and glipizide did not prevent the local antinociception produced by indomethacin. Nonetheless, metformin and phenformin reversed the local antinociception induced by indomethacin. SIGNIFICANCE: Data suggest that diclofenac could activate the K(+) channels and biguanides-dependent mechanisms to produce its peripheral antinociceptive effects in the formalin test. Likewise, a biguanides-dependent mechanism could be activated by indomethacin consecutively to generate its peripheral antinociceptive effect.


Subject(s)
Diclofenac/antagonists & inhibitors , Diclofenac/pharmacology , Indomethacin/antagonists & inhibitors , Indomethacin/pharmacology , Metformin/pharmacology , Pain Measurement/drug effects , Phenformin/pharmacology , Animals , Dose-Response Relationship, Drug , KATP Channels/antagonists & inhibitors , KATP Channels/physiology , Male , Pain Measurement/methods , Rats , Rats, Wistar
10.
J Pharmacol Sci ; 116(3): 274-82, 2011.
Article in English | MEDLINE | ID: mdl-21691039

ABSTRACT

We have reported that nicotine and the specific α7AChR agonist ameliorate indomethacin-induced intestinal lesions in mice by activating α7 nicotinic acetylcholine receptors (α7nAChR). Dopamine D2-receptor antagonists, such as domperidone and metoclopramide, enhance the release of ACh from vagal efferent nerves. The present study examined the effects of domperidone and metoclopramide on indomethacin-induced small intestinal ulceration in mice, focusing on the α7AChR. Male C57BL/6 mice were administered indomethacin (10 mg/kg, s.c.) and sacrificed 24 h later. Domperidone (0.1-10 mg/kg) and metoclopramide (0.03-0.3 mg/kg) were administered i.p. twice, at 0.5 h before and 8 h after indomethacin treatment, while methyllycaconitine (a selective antagonist of α7nAChR, 30 mg/kg) was administered twice, at 0.5 h before each domperidone treatment. Indomethacin caused severe hemorrhagic lesions in the small intestine, mostly to the jejunum and ileum, with a concomitant increase in myeloperoxidase (MPO) activity. Domperidone suppressed the severity of lesions and the increase in MPO activity at low doses (0.1-3 mg/kg), but not at a high dose (10 mg/kg). Similar effects were also observed by metoclopramide. The protective effects of domperidone and metoclopramide were totally abolished by prior administration of methyllycaconitine. Indomethacin treatment markedly enhanced inducible nitric oxide synthase and chemokine mRNA expression in the small intestine, but these responses were all significantly attenuated by either domperidone or metoclopramide. These findings suggest that dopamine D2-receptor antagonists ameliorate indomethacin-induced small intestinal ulceration through the activation of endogenous anti-inflammatory pathways mediated by α7nAChR.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Intestinal Diseases/prevention & control , Intestine, Small/drug effects , Receptors, Nicotinic/metabolism , Ulcer/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/agonists , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/adverse effects , Domperidone/administration & dosage , Domperidone/adverse effects , Domperidone/therapeutic use , Dopamine Antagonists/administration & dosage , Dopamine Antagonists/adverse effects , Dose-Response Relationship, Drug , Gastrointestinal Agents/agonists , Gastrointestinal Agents/antagonists & inhibitors , Gastrointestinal Agents/toxicity , Gastrointestinal Motility/drug effects , Gene Expression Regulation/drug effects , Indomethacin/agonists , Indomethacin/antagonists & inhibitors , Indomethacin/toxicity , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Metoclopramide/administration & dosage , Metoclopramide/adverse effects , Metoclopramide/therapeutic use , Mice , Mice, Inbred C57BL , Nicotinic Antagonists/toxicity , Rats , Rats, Wistar , Receptors, Nicotinic/chemistry , Severity of Illness Index , Ulcer/metabolism , Ulcer/pathology , alpha7 Nicotinic Acetylcholine Receptor
11.
J Pineal Res ; 51(1): 61-74, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21342246

ABSTRACT

Collagenases are key proteases involved in inflammation and injury. We addressed whether collagenases have an association with the susceptibility of gastric injury under diabetes as well as the effect of melatonin on collagenases in ulcerated gastric tissues. Diabetes was induced in rats by a single dose of streptozotocin (STZ) followed by gastric ulceration using indomethacin, and melatonin's action was studied by its application prior to indomethacin exposure. Ulcer indices and damage were elevated significantly in gastric tissues of diabetic compared with nondiabetic rats. Melatonin reversed the effect of indomethacin during protection of gastric ulcers in diabetic rats. Matrix metalloproteinase (MMP)-13 (i.e., collagenase-3) was upregulated in diabetic gastric mucosa and enhanced further upon ulceration while melatonin ameliorated their activity. In addition, gastric tissues showed enhanced expression of both MMP-1 (i.e., collagenases-1) and -13 significantly in diabetic rats compared with nondiabetic animals and more so during ulceration while tissue inhibitors of metalloproteinase-1 (TIMP-1) showed an opposite trend. MMP-2 activities exhibited a ∼50% downregulation during gastric ulceration which were rescued by melatonin. Moreover, increased expression of both MMP-1 and -13 was mediated by activator protein-1 activation via extracellular signal-regulated kinase 1/2 which were parallel to upregulation of tumor necrosis factor-α, interleukin-1ß, and heat shock protein-70 during ulceration. Melatonin arrested collagenase expression by downregulation of these signaling molecules thereby halting the progression of the disease. We conclude that diabetic gastric tissues are susceptible to ulceration and associated with MMP-1 and -13 upregulation in indomethacin-induced injury. Additionally, melatonin protects the gastric damage under diabetes via regulation of both MMP-1 and -13.


Subject(s)
Collagenases/metabolism , Diabetes Mellitus, Experimental/metabolism , Matrix Metalloproteinase 13/metabolism , Melatonin/pharmacology , Stomach Ulcer/metabolism , Analysis of Variance , Animals , Collagenases/genetics , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Drug Interactions , Gelatinases/biosynthesis , Gelatinases/genetics , Histocytochemistry , Immunohistochemistry , Indomethacin/antagonists & inhibitors , Male , Matrix Metalloproteinase 13/genetics , Rats , Rats, Sprague-Dawley , Stomach Ulcer/chemically induced , Stomach Ulcer/enzymology , Stomach Ulcer/pathology , Up-Regulation/drug effects
12.
Eur J Pharmacol ; 614(1-3): 114-21, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19445918

ABSTRACT

The clinical utility of non-steroidal anti-inflammatory drugs (NSAIDs) is limited by their gastrointestinal and renal toxicities. Indomethacin (an NSAID commonly used in toxicity studies) has been shown to induce significant oxidative stress and mitochondrial dysfunction in the rat kidney. The current study was designed to assess the potential of zinc, a known antioxidant, to protect the kidney against these drug-induced effects. Male Wistar rats were pre-treated with zinc sulphate (50 mg/kg) and dosed with indomethacin (20 mg/kg) by oral gavage. Rats were sacrificed 24 h after the dose of indomethacin. Parameters of oxidative stress, mitochondrial function and lipid content of the mitochondrial membranes were measured in the kidneys of these animals. It was found that zinc significantly attenuated indomethacin-induced oxidative stress, mitochondrial dysfunction and changes in the lipids in mitochondrial membranes in the kidney. The content of metallothionein, a cysteine-rich zinc-binding protein, was also determined in the tissue. There was no significant induction of metallothionein in the kidney in zinc-treated animals. Estimation of serum creatinine showed that zinc seemed to hasten functional recovery of the kidney following indomethacin administration. We conclude that pretreatment with zinc is effective in protecting against indomethacin-induced changes in the rat kidney. This protective effect does not appear to be mediated by metallothionein.


Subject(s)
Antioxidants/pharmacology , Indomethacin/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Mitochondria/drug effects , Oxidative Stress/drug effects , Zinc/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Enzymes/metabolism , Humans , Indomethacin/antagonists & inhibitors , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lipid Metabolism/drug effects , Male , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
13.
Biol Pharm Bull ; 32(4): 589-96, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19336889

ABSTRACT

We previously reported that eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) extracted from Artemisia asiaitica, augmented the cellular antioxidant defense capacity through induction of the antioxidant protein heme oxygenase-1 (HO-1), thereby protecting ileal smooth muscle cells from nonsteroidal anti-inflammatory drug (NSAID)-induced intestinal toxicity. In the present study, we used cultured feline esophageal epithelial cells (EEC) to investigate the ability of eupatilin to induce expression of HO-1 and to analyze its cytoprotective effect against indomethacin-induced damage, since NSAID users have a higher risk of esophageal ulcers or esophagitis than non-NSAID users. A culture of EEC from cat was prepared. The identity of the cultures was confirmed by immunocytochemistry using cytokeratin antibodies. Western blot analysis showed a concentration- and time- dependent expression of HO-1 in response to eupatilin. Phosphorylation of extracellular regulating protein kinase (ERKs) and Akt, and nuclear translocation of nuclear related factor 2 (Nrf2) were induced by 150 microM eupatilin in a time-dependent manner. Eupatilin-induced HO-1 expression and Nrf2 were partly attenuated by MEK inhibitor PD98059 and almost completely by phosphatidyl-inactiol 3 kinase (PI3K) inhibitor LY294002, but not by c-Jun N-terminal kinase (JNK) inhibitor SP600125 or p38 mitogen activated protein kinase (MAPK) inhibitor SB202190. MTT assay showed that treatment with 2 mM indomethacin for 2 h decreased cell viability to about 41%. Pre-treatment of cells with eupatilin resulted in the dose-dependent inhibition of indomethacin-induced cell damage. We confirmed that ZnPP, an HO-1 inhibitor, repressed eupatilin-induced HO-1 activity and showed the protective effect of eupatilin against indomethacin-induced cell injury. The data suggested that HO-1 was partly responsible for the eupatilin-mediated protective action of esophageal epithelial cells against indomethacin via both ERKs and PI3K/Akt pathways as well as Nrf2 translocation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Epithelial Cells/drug effects , Esophagus/cytology , Esophagus/drug effects , Flavonoids/pharmacology , Heme Oxygenase-1/biosynthesis , Indomethacin/antagonists & inhibitors , Indomethacin/toxicity , Protective Agents , Animals , Blotting, Western , Cats , Cell Survival/drug effects , Cells, Cultured , Culture Media, Serum-Free , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Indicators and Reagents , Nuclear Proteins/metabolism , Rats , Rats, Sprague-Dawley
14.
Arch Toxicol ; 82(7): 471-81, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18060385

ABSTRACT

Oxidative stress and mitochondrial dysfunction have been implicated in the pathogenesis of indomethacin-induced enteropathy. We evaluated the potential of curcumin, a known cytoprotectant, as an agent to protect against such effects. Rats were pretreated with curcumin (40 mg/kg by intra-peritoneal injection) before administration of indomethacin (20 mg/kg by gavage). One hour later, the small intestine was isolated and used for assessment of parameters of oxidative stress. Mitochondria, brush border membranes (BBM) and surfactant-like particles (SLP) were also isolated from the tissue. Mitochondria were used for assessment of functional integrity, estimation of products of lipid peroxidation and lipid content. BBM were used for estimation of products of lipid peroxidation and lipid content, while the SLP were used for measurement of lipid content. The results showed that oxidative stress and mitochondrial dysfunction occurred in the small intestine of indomethacin-treated rats. Pre-treatment with curcumin was found to ameliorate these drug-induced changes. Significant changes were seen in some of the lipids in the mitochondria, BBM and SLP in response to indomethacin. However, curcumin did not have any significant effect on these drug-induced changes. We conclude that curcumin, by attenuating oxidative stress and mitochondrial dysfunction, holds promise as an agent that can potentially reduce NSAID-induced adverse effects in the small intestine.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Curcumin/pharmacology , Indomethacin/antagonists & inhibitors , Indomethacin/toxicity , Mitochondrial Diseases/drug therapy , Oxidative Stress/drug effects , Alkaline Phosphatase/metabolism , Animals , Intestinal Diseases/chemically induced , Intestinal Diseases/prevention & control , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Male , Microvilli/drug effects , Microvilli/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/prevention & control , Rats , Rats, Wistar , Surface-Active Agents/pharmacology
15.
Free Radic Res ; 41(8): 861-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17654042

ABSTRACT

Indomethacin is one of non-steroidal anti-inflammatory drugs that are commonly used clinically and often cause gastric mucosal injury as a side effect. Generation of reactive oxygen species (ROS) and activation of apoptotic signaling are involved in the pathogenesis of indomethacin-induced gastric mucosal injury. Thioredoxin-1 (Trx-1) is a small redox-active protein with anti-oxidative activity and redox-regulating functions. The aim of this study was to investigate the protective effect of Trx-1 against indomethacin-induced gastric mucosal injury. Trx-1 transgenic mice displayed less gastric mucosal damage than wild type (WT) C57BL/6 mice after intraperitoneal administration of indomethacin. Administration of recombinant human Trx-1 (rhTrx-1) or transfection of the Trx-1 gene reduced indomethacin-induced cytotoxicity in rat gastric epithelial RGM-1 cells. Pretreatment with rhTrx-1 suppressed indomethacininduced ROS production and downregulation of phosphorylated Akt in RGM-1 cells. Survivin, a member of inhibitors of apoptosis proteins family, was downregulated by indomethacin, which was suppressed in Trx-1 transgenic mice or by administration of rhTrx-1 in RGM-1 cells. Trx-1 inhibits indomethacin-induced apoptotic signaling and gastric ulcer formation, suggesting that it may have a preventive and therapeutic potential against indomethacin-induced gastric injury.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Gastric Mucosa/drug effects , Indomethacin/antagonists & inhibitors , Thioredoxins/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Gastric Mucosa/metabolism , Indomethacin/toxicity , Mice , Mice, Transgenic , Rats , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Thioredoxins/genetics , Thioredoxins/metabolism
16.
Eur J Gastroenterol Hepatol ; 19(3): 245-50, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17301652

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drug (NSAID) use is associated with an elevated risk of gastrointestinal damage. As adenosine 5'-triphosphate (ATP) may play a protective role in the small intestine, our objective was to determine the local effect of ATP on small intestinal permeability changes induced by short-term challenge of the NSAID indomethacin in healthy humans. METHODS: Mucosal permeability of the small intestine was assessed by the lactulose/rhamnose permeability test, that is, ingestion of a test drink containing 5 g lactulose and 0.5 g L-rhamnose followed by total urine collection for 5 h. Urinary excretion of lactulose and L-rhamnose was determined by fluorescent detection high-pressure liquid chromatography (HPLC). Basal small intestinal permeability was assessed as a control condition. As a model of increased small intestinal permeability, two doses of indomethacin were ingested before ingestion of the test drink (75 mg and 50 mg at 10 h and 1 h before the test drink, respectively). Concomitantly with indomethacin ingestion, placebo or 30 mg/kg ATP was administered through a naso-intestinal tube. RESULTS: Median urinary lactulose/rhamnose ratio (g/g) in the control condition was 0.023 (interquartile range: 0.013-0.041). Compared with the control condition, urinary lactulose/rhamnose ratio after ingestion of indomethacin and administration of placebo was significantly increased [0.042 (0.028-0.076); P<0.01]. In contrast, urinary lactulose/rhamnose ratio after indomethacin ingestion plus ATP administration [0.027 (0.020-0.046)] was significantly lower than the lactulose/rhamnose ratio in the placebo condition (P<0.01). CONCLUSIONS: Topical ATP administration into the small intestine during short-term challenge of the NSAID indomethacin attenuates the NSAID-induced increase in small intestinal permeability in healthy humans.


Subject(s)
Adenosine Triphosphate/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Indomethacin/antagonists & inhibitors , Intestinal Absorption/drug effects , Intestine, Small/drug effects , Adolescent , Adult , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cross-Over Studies , Double-Blind Method , Female , Humans , Indomethacin/pharmacology , Intestine, Small/metabolism , Lactulose/urine , Male , Permeability/drug effects , Rhamnose/urine
17.
J Pharmacol Sci ; 103(1): 40-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17202747

ABSTRACT

We examined the effect of tacrolimus (FK506), an immunosuppressive drug, on indomethacin-induced small intestinal ulceration in rats. Animals were given indomethacin (10 mg/kg, s.c.), killed 24 h later, and myeloperoxidase (MPO) activity and thiobarbituric acid reactants (TBARS) were evaluated in intestinal lesions. Tacrolimus (0.3 - 3 mg/kg) was administered p.o. twice 0.5 h before and 6 h after indomethacin injection. The expression of inducible nitric oxide synthase (iNOS) mRNA was determined by a TaqMan real-time RT-PCR, while the activity of nuclear factor (NF)-kappaB DNA-binding was analyzed by electrophoresis mobility shift assays (EMSA) 6 h after indomethacin treatment. Indomethacin provoked severe hemorrhagic lesions in the small intestine, mainly in the jejunum and ileum, accompanied with increases in MPO activity and TBARS. Oral administration of tacrolimus reduced the severity of indomethacin-induced intestinal lesions in a dose-dependent manner. The increases in MPO activity and TBARS were also significantly attenuated by tacrolimus. The expression of iNOS mRNA was markedly enhanced when examined 6 h after indomethacin administration, and this response was counteracted by tacrolimus. Indomethacin also activated NF-kappaB in a tacrolimus-preventable manner. These results suggest that tacrolimus prevents indomethacin-induced small intestinal ulceration in the rat. This effect may be due to inhibition of iNOS induction through suppression of NF-kappaB activation.


Subject(s)
Immunosuppressive Agents/pharmacology , Indomethacin/antagonists & inhibitors , Intestinal Diseases/prevention & control , Nitric Oxide Synthase Type II/antagonists & inhibitors , Tacrolimus/pharmacology , Ulcer/prevention & control , Animals , Intestinal Diseases/chemically induced , Lipid Peroxidation/drug effects , Male , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Peroxidase/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Ulcer/chemically induced
18.
Biol Pharm Bull ; 28(12): 2206-10, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16327150

ABSTRACT

Indomethacin is used as an anti-inflammatory drug and a nonselective cyclooxygenase inhibitor. When indomethacin in methanol was photo-irradiated with an Hg lamp, methyl ester, ethyl ester, and gamma-lactone derivatives of indomethacin were produced. In the present study, we found that the methyl ester derivative of indomethacin (M-IN) could more potently inhibit prostaglandin E(2) (PGE(2)) and nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX 2) protein expression from lipopolysaccharide (LPS)-stimulated RAW 264.7 cells than indomethacin, similar to the effect of a non-steroidal anti-inflammatory drugs (NSAID). On the other hand, the results showed that M-IN with an IC(50) value maintained at 36.9 microg/ml for 12 h exhibited stronger cytotoxicity than ethyl ester, gamma-lactone derivatives of indomethacin, and indomethacin in promyelocytic leukemia HL-60 cells. Moreover, a series of biochemical analyses determined that M-IN caused apoptotic bodies, DNA fragmentation, and enhanced PARP and pro-caspase 3 degradation in HL-60 cells. These above results indicate that the photosynthesized product, M-IN, had stronger anti-inflammatory effects in LPS-stimulated RAW 264.7 cells and cytotoxicity effects in HL-60 cells than the parent drug, indomethacin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Indomethacin/analogs & derivatives , Indomethacin/therapeutic use , Inflammation/prevention & control , Leukemia, Promyelocytic, Acute/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Apoptosis/genetics , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Survival/drug effects , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA Fragmentation/drug effects , DNA Fragmentation/genetics , Dinoprostone/antagonists & inhibitors , Dinoprostone/genetics , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Formazans , G1 Phase/drug effects , Gene Expression/drug effects , Gene Expression/genetics , HL-60 Cells , Humans , Indomethacin/antagonists & inhibitors , Inflammation/drug therapy , Inhibitory Concentration 50 , Lipopolysaccharides/pharmacology , Methods , Mice , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Tetrazolium Salts
19.
Yao Xue Xue Bao ; 40(5): 402-5, 2005 May.
Article in Chinese | MEDLINE | ID: mdl-16220780

ABSTRACT

AIM: To study the vasodilation effect of atropine and its mechanism. METHODS: Isometric tension was recorded in isolated rat super mesenteric arteries precontracted by noradrenaline (NE) to study the vasodilation effect of atropine, and to investigate the role of endothelial cell and vascular smooth muscle cell on vasodilation. RESULTS: Atropine was shown to significantly dilate the endothelium-intact and endothelium-denuded arteries precontracted by NE. Nomega-Nitro-L-arginine methyl ester (L-NAME, nitric oxide synthase inhabitor), indomethacin (cyclooxygenase inhibitor), propranolol (general beta adrenoceptor antagonist) and glibenclamide (ATP sensitive potassium channel inhibitor) showed no effect on vasodilation of atropine. Atropine did not affect the concentration-contraction curve of K+. However, atropine suppressed the contraction induced by NE and CaCl2, but not that by caffeine in the Ca+ -free Krebs solution. CONCLUSION: Atropine showed significant vasodilation effect which may derive, in part, from endothelium. Besides, atropine could inhibit the receptor-mediated Ca2+ -influx and Ca2+ -release, which was inferred to the mechanism of atropine on vasodilation.


Subject(s)
Atropine/pharmacology , Mesenteric Artery, Superior/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Calcium/metabolism , Calcium Chloride/antagonists & inhibitors , Cyclooxygenase Inhibitors/pharmacology , Endothelial Cells/physiology , Female , In Vitro Techniques , Indomethacin/antagonists & inhibitors , Male , NG-Nitroarginine Methyl Ester/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Norepinephrine/antagonists & inhibitors , Potassium Chloride/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
20.
Biochemistry ; 43(49): 15439-45, 2004 Dec 14.
Article in English | MEDLINE | ID: mdl-15581355

ABSTRACT

Cyclooxygenases (COXs) are the therapeutic targets of nonsteroidal antiinflammatory drugs. Indomethacin (INDO) was one of the first nonsteroidal antiinflammatory drugs to be characterized as a time-dependent, functionally irreversible inhibitor, but the molecular basis of this phenomenon is uncertain. In the crystal structure of INDO bound to COX-2, a small hydrophobic pocket was identified that surrounds the 2'-methyl group of INDO. The pocket is formed by the residues Ala-527, Val-349, Ser-530, and Leu-531. The contribution of this pocket to inhibition was evaluated by altering its volume by mutagenesis of Val-349. The V349A mutation expanded the pocket and increased the potency of INDO, whereas the V349L mutation reduced the size of the pocket and decreased the potency of INDO. Particularly striking was the reversibility of INDO inhibition of the V349L mutant. The 2'-des-methyl analogue of INDO (DM-INDO) was synthesized and tested against wild-type COX-1 and COX-2, as well as the Val-349 mutants. DM-INDO bound to all enzymes tested, but only inhibited wt mCOX-2 and the V349I enzyme. Without the 2'-methyl group anchoring DM-INDO in the active site, the compound was readily competed off of the enzyme by arachidonic acid. The kinetics of inhibition were comparable to the kinetics of binding as evaluated by fluorescence quenching. These results highlight binding of the 2'-methyl of INDO in the hydrophobic pocket as an important determinant of its time-dependent inhibition of COX enzymes.


Subject(s)
Cyclooxygenase Inhibitors/chemistry , Indomethacin/analogs & derivatives , Indomethacin/chemistry , Prostaglandin-Endoperoxide Synthases/metabolism , Amino Acid Substitution/genetics , Animals , Binding Sites , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/metabolism , Indomethacin/antagonists & inhibitors , Indomethacin/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Male , Mice , Prostaglandin-Endoperoxide Synthases/genetics , Sheep , Spectrometry, Fluorescence , Substrate Specificity/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...