Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Free Radic Biol Med ; 52(9): 1680-91, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22343417

ABSTRACT

The spice-derived phenolic, malabaricone C (mal C), has recently been shown to accelerate healing of the indomethacin-induced gastric ulceration in mice. In this study, we explored its anti-inflammatory activity and investigated the underlying mechanism of the action. Mal C suppressed the microvascular permeability and the levels of tumor necrosis factor-α, interleukin-1ß, and nitric oxide in the lipopolysaccharide (LPS)-administered mice. At a dose of 10 mg/kg, it showed anti-inflammatory activity comparable to that of omeprazole (5 mg/kg) and dexamethasone (50 mg/kg). It also reduced the expression and activities of inducible nitric oxide synthase, cyclooxygenase-2, as well as the pro- vs anti-inflammatory cytokine ratio in the LPS-treated RAW macrophages. Mal C was found to inhibit LPS-induced NF-kB activation in RAW 264.7 cells by blocking the MyD88-dependent pathway. Mal C suppressed NF-κB activation and iNOS promoter activity, which correlated with its inhibitory effect on IκB phosphorylation and degradation, and NF-κB nuclear translocation, in the LPS-stimulated macrophages. It also inhibited LPS-induced phosphorylation of p38 and JNK, which are also upstream activators of NF-κB, without affecting Akt phosphorylation. Mal C also effectively blocked the PKR-mediated activation of NF-κB. These findings indicate that mal C exerts an anti-inflammatory effect through NF-κB-responsive inflammatory gene expressions by inhibiting the p38 and JNK-dependent canonical NF-κB pathway as well as the PKR pathway, and is a potential therapeutic agent against acute inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Resorcinols/pharmacology , Animals , Base Sequence , Cell Line , DNA Primers , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Phosphorylation , Tumor Necrosis Factor-alpha/metabolism
3.
Int Immunopharmacol ; 9(4): 491-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19291837

ABSTRACT

The role of the arginine-metabolism in the healing action of the Myristica malabarica phenol malabaricone B (mal B) and omeprazole against indomethacin-induced stomach ulceration in mouse was investigated. Indomethacin (18 mg kg- 1) was found to induce maximum stomach ulceration in Swiss albino mice on the 3rd day of its administration, which was associated with reduced arginase activity (30.8%, P < 0.01), eNOS expression, along with increased iNOS expression, total NOS activity (5.55 folds, P < 0.001), NO generation (2.19 folds, P < 0.001), and ratio of pro-/anti-inflammatory cytokines. Besides providing comparable healing as omeprazole (3 mg kg- 1 x 3 days), mal B (10 mg kg- 1 x 3 days, p. o.) shifted the iNOS/NO axis to the arginase/polyamine axis as revealed from the increased arginase activity (51.6%, P < 0.001), eNOS expression, and reduced iNOS expression, total NOS activity (approximately 75%, P < 0.001), and NO level (50.6%, P < 0.01). These could be attributed to a favourable anti/pro inflammatory cytokines ratio, generated by mal B. The healing by omeprazole was however, not significantly associated with those parameters.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Cytokines/biosynthesis , Nitric Oxide/biosynthesis , Omeprazole/therapeutic use , Resorcinols/therapeutic use , Stomach Ulcer/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Arginase/drug effects , Arginase/metabolism , Cytokines/blood , Indomethacin/toxicity , Male , Mice , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/blood , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Resorcinols/chemistry , Stomach Ulcer/chemically induced
4.
J Radiat Res ; 46(2): 165-71, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15988134

ABSTRACT

The radioprotective activity of Piper betel ethanolic extract (PE) has been studied using rat liver mitochondria and pBR 322 plasmid DNA as two model in vitro systems. The extract effectively prevented gamma-ray induced lipid peroxidation as assessed by measuring thiobarbituric acid reactive substrates, lipid hydroperoxide and conjugated diene. Likewise, it prevented radiation-induced DNA strand breaks in a concentration dependent manner. The radioprotective activity of PE could be attributed to its hydroxyl and superoxide radicals scavenging property along with its lymphoproliferative activity. The radical scavenging capacity of PE was primarily due to its constituent phenolics, which were isolated and identified as chevibetol and allyl pyrocatechol.


Subject(s)
DNA Damage/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/radiation effects , Piper betle/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Plasmids/drug effects , Plasmids/radiation effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Ethanol/chemistry , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Male , Radiation Dosage , Radiation Tolerance/drug effects , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...