Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.767
Filter
1.
PLoS One ; 19(5): e0302015, 2024.
Article in English | MEDLINE | ID: mdl-38728332

ABSTRACT

Nature has proven to be a treasure resource of bioactive metabolites. In this regard, Tamarix aphylla (F. Tamaricaceae) leaves crude extract was investigated for its gastroprotective effect against indomethacin-induced damage to the gastric mucosa. Additionally, phytochemical investigation of the methanolic extract afforded eight flavonoids' derivatives (1-8). On pharmacology networking study, the isolated compounds identified 123 unique targets where only 45 targets were related to peptic ulcer conditions, these 45 targets include 11 targets specifically correlate to gastric ulcer. The protein-protein interaction defined the PTGS2 gene as one of the highly interacted genes and the complete pharmacology network defined the PTGS2 gene as the most represented gene. The top KEGG signaling pathways according to fold enrichment analysis was the EGFR tyrosine kinase inhibitor resistance pathway. As a result, these findings highlighted the significance of using T. aphylla leaves crude extract as an anti-gastric ulcer candidate, which provides a safer option to chemical antisecretory medicines, which are infamous for their negative side effects. Our findings have illuminated the potent anti-inflammatory and antioxidant effects of T. aphylla, which are likely mediated by suppressing IL-1ß, IL-6, TNF-α, and MAPK signaling pathways, without compromising gastric acidity.


Subject(s)
Indomethacin , MAP Kinase Signaling System , Oxidative Stress , Plant Extracts , Stomach Ulcer , Tamaricaceae , Stomach Ulcer/drug therapy , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Animals , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Indomethacin/adverse effects , Indomethacin/toxicity , Rats , Tamaricaceae/chemistry , MAP Kinase Signaling System/drug effects , Male , Plant Leaves/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/chemically induced , Rats, Sprague-Dawley , Network Pharmacology , Gastric Mucosa/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Anti-Ulcer Agents/chemistry , Flavonoids/pharmacology , Flavonoids/chemistry
2.
Pak J Pharm Sci ; 37(2): 315-320, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38767098

ABSTRACT

The present study was designed to assess Tradescantia spathacea's antidiabetic ability, as well as the antiulcer activity of the entire plant extract. The diabetic condition was evaluated using Streptozotocin's oral glucose tolerance test, diabetes-alloxan and diabetes-models. Antiulcer activities were observed in rats where gastric ulcers were either caused by oral administration of ethanol, or pyloric ligation. Standards include ranitidine, glibenclamide and sucralfate. In all models, the blood glucose levels of animals treated with the test extract were found to be significantly lower compared to diabetic care. Similarly, in all models, the ulcer index in the animals treated with the test extract was found to be significantly lower relative to the animals under vehicle supervision. Our findings say T. Spathacea extract has essential anti-diabetic properties, as well as antiulcer properties.


Subject(s)
Anti-Ulcer Agents , Blood Glucose , Diabetes Mellitus, Experimental , Hypoglycemic Agents , Plant Extracts , Rats, Wistar , Stomach Ulcer , Animals , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/isolation & purification , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/isolation & purification , Diabetes Mellitus, Experimental/drug therapy , Stomach Ulcer/drug therapy , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Stomach Ulcer/chemically induced , Male , Rats , Blood Glucose/drug effects , Blood Glucose/metabolism , Methanol/chemistry , Glucose Tolerance Test , Solvents/chemistry , Phytotherapy
3.
BMC Gastroenterol ; 24(1): 187, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811868

ABSTRACT

BACKGROUND: Proton-pump inhibitors (PPIs) prevent aspirin-associated gastric and duodenal mucosal damage. However, long-term use of PPIs can lead to various adverse reactions, such as gastric polyps and enterochromaffin-like cell hyperplasia. Current research indicates that the abovementioned adverse reactions are mainly related to hypergastrinemia. We investigated whether low-frequency administration of omeprazole could effectively repair aspirin-induced mucosal damage and reduce the increase in gastrin levels associated with long-term use of PPIs. METHODS: Sprague‒Dawley rats were divided into four treatment groups: daily aspirin, daily aspirin and omeprazole once every day (qd), daily aspirin and omeprazole once every other day (qod), and daily aspirin and omeprazole once every three days (1/d3). After 15 days of feeding, blood samples were collected, and the stomachs of sacrificed rats were subjected to macroscopic, histological, and immunohistochemical studies. Moreover, in clinical practice, patients with peptic ulcers caused by aspirin took a standard dose of omeprazole (20 mg) every other day. Two months later, gastroscopy was performed to examine the healing of the ulcers. RESULTS: Both the omeprazole qd and omeprazole qod administrations effectively prevented aspirin-induced gastric peptic ulcers, with no significant difference between the two groups in the inhibition of parietal cell secretion of gastric acid and cell apoptosis. However, omeprazole 1/d3 failed to completely prevent aspirin-induced gastric mucosal injury. Notably, the gastrin levels, cell proliferation ability and cholecystokinin B receptor expression of the omeprazole qd group were significantly higher than those of the omeprazole qod group. In clinical work, patients with peptic ulcers caused by aspirin were given a standard dose of omeprazole every other day, and their ulcers healed after 2 months, as observed by gastroscopy. CONCLUSIONS: Omeprazole administration once every other day can effectively prevent aspirin-induced peptic ulcers and reduce hypergastrinemia, which may reduce the long-term adverse effects of PPI treatment.


Subject(s)
Aspirin , Gastric Mucosa , Gastrins , Omeprazole , Proton Pump Inhibitors , Rats, Sprague-Dawley , Animals , Aspirin/adverse effects , Aspirin/administration & dosage , Omeprazole/pharmacology , Omeprazole/administration & dosage , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/administration & dosage , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastrins/blood , Male , Rats , Drug Administration Schedule , Humans , Peptic Ulcer/prevention & control , Peptic Ulcer/chemically induced , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Stomach Ulcer/prevention & control , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology
4.
Biomaterials ; 309: 122599, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38703409

ABSTRACT

Development of bioadhesives that can be facilely delivered by endoscope and exhibit instant and robust adhesion with gastric tissues to promote gastric ulcer healing remains challenging. In this study, an advanced bioadhesive is prepared through free radical polymerization of ionized N-acryloyl phenylalanine (iAPA) and N-[tris (hydroxymethyl) methyl] acrylamide (THMA). The precursory polymer solution exhibits low viscosity with the capability for endoscope delivery, and the hydrophilic-hydrophobic transition of iAPA upon exposure to gastric acid can trigger gelation through phenyl groups assisted multiple hydrogen bonds formation and repel water molecules on tissue surface to establish favorable environment for interfacial interactions between THMA and functional groups on tissues. The in-situ formed hydrogel features excellent stability in acid environment (14 days) and exhibits firm wet adhesion to gastric tissue (33.4 kPa), which can efficiently protect the wound from the stimulation of gastric acid and pepsin. In vivo studies reveal that the bioadhesive can accelerate the healing of ulcers by inhibiting inflammation and promoting capillary formation in the acetic acid-induced gastric ulcer model in rats. Our work may provide an effective solution for the treatment of gastric ulcers clinically.


Subject(s)
Stomach Ulcer , Wound Healing , Animals , Stomach Ulcer/drug therapy , Stomach Ulcer/chemically induced , Wound Healing/drug effects , Hydrogen-Ion Concentration , Rats , Rats, Sprague-Dawley , Male , Hydrogels/chemistry , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology , Phenylalanine/chemistry
5.
Biomed Pharmacother ; 175: 116647, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703503

ABSTRACT

OBJECTIVE: To improve the biological and toxicological properties of Mefenamic acid (MA), the galactosylated prodrug of MA named MefeGAL was included in polymeric solid dispersions (PSs) composed of poly(glycerol adipate) (PGA) and Pluronic® F68 (MefeGAL-PS). MefeGAL-PS was compared with polymeric solid formulations of MA (MA-PS) or a mixture of equal ratio of MefeGAL/MA (Mix-PS). METHODS: The in vitro and in vivo pharmacological and toxicological profiles of PSs have been investigated. In detail, we evaluated the anti-inflammatory (carrageenan-induced paw edema test), analgesic (acetic acid-induced writhing test) and ulcerogenic activity in mice after oral treatment. Additionally, the antiproliferative activity of PSs was assessed on in vitro models of colorectal and non-small cell lung cancer. RESULTS: When the PSs were resuspended in water, MefeGAL's, MA's and their mixture's apparent solubilities improved due to the interaction with the polymeric formulation. By comparing the in-vivo biological performance of MefeGAL-PS with that of MA, MefeGAL and MA-PS, it was seen that MefeGAL-PS exhibited the same sustained and delayed analgesic and anti-inflammatory profile as MefeGAL but did not cause gastrointestinal irritation. The pharmacological effect of Mix-PS was present from the first hours after administration, lasting about 44 hours with only slight gastric mucosa irritation. In-vitro evaluation indicated that Mix-PS had statistically significant higher cytotoxicity than MA-PS and MefeGAL-PS. CONCLUSIONS: These preliminary data are promising evidence that the galactosylated prodrug approach in tandem with a polymer-drug solid dispersion formulation strategy could represent a new drug delivery route to improve the solubility and biological activity of NSAIDs.


Subject(s)
Drug Delivery Systems , Mefenamic Acid , Animals , Mefenamic Acid/pharmacology , Mefenamic Acid/administration & dosage , Mice , Humans , Male , Edema/drug therapy , Edema/chemically induced , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Prodrugs/pharmacology , Prodrugs/administration & dosage , Analgesics/pharmacology , Analgesics/administration & dosage , Analgesics/toxicity , Cell Proliferation/drug effects , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology , Poloxamer/chemistry
6.
Toxicol Appl Pharmacol ; 486: 116950, 2024 May.
Article in English | MEDLINE | ID: mdl-38701902

ABSTRACT

Antidepressant duloxetine has been shown protective effect on indomethacin-induced gastric ulcer, which was escorted by inflammation in the gastric mucosa. Cytokines are the principal mediators of inflammation. Thus, by screening the differential expression of cytokines in the gastric mucosa using cytokine array at 3 h after indomethacin exposure, when the gastric ulcer began to format, we found that indomethacin increased cytokines which promoted inflammation responses, whereas duloxetine decreased pro-inflammatory cytokines increased by indomethacin and increased RANTES expression. RANTES was consistently increased by pretreated with both 5 mg/kg and 20 mg/kg duloxetine at 3 h and 6 h after indomethacin exposure in male rats. Selective blockade of RANTES-CCR5 axis by a functional antagonist Met-RANTES or a CCR5 antagonist maraviroc suppressed the protection of duloxetine. Considering the pharmacologic action of duloxetine on reuptake of monoamine neurotransmitters, we examined the serotonin (5-HT), norepinephrine and dopamine contents in the blood and discovered 20 mg/kg duloxetine increased 5-HT levels in platelet-poor plasma, while treatment with 5-HT promoted expression of RANTES in the gastric mucosa and alleviated the indomethacin-induced gastric injury. Furthermore, duloxetine activated PI3K-AKT-VEGF signaling pathway, which was regulated by RANTES-CCR5, and selective inhibitor of VEGF receptor axitinib blocked the prophylactic effect of duloxetine. Furthermore, duloxetine also protected gastric mucosa from indomethacin in female rats, and RANTES was increased by duloxetine after 6 h after indomethacin exposure too. Together, our results identified the role of cytokines, particularly RANTES, and the underlying mechanisms in gastroprotective effect of duloxetine against indomethacin, which advanced our understanding in inflammatory modulation by monoamine-based antidepressants.


Subject(s)
Chemokine CCL5 , Duloxetine Hydrochloride , Gastric Mucosa , Indomethacin , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Serotonin , Signal Transduction , Stomach Ulcer , Vascular Endothelial Growth Factor A , Animals , Duloxetine Hydrochloride/pharmacology , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Male , Indomethacin/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Chemokine CCL5/metabolism , Signal Transduction/drug effects , Rats , Vascular Endothelial Growth Factor A/metabolism , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Stomach Ulcer/metabolism , Serotonin/metabolism , Phosphatidylinositol 3-Kinases/metabolism
7.
Mol Biol Rep ; 51(1): 684, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796650

ABSTRACT

BACKGROUND: Indomethacin is an anti-inflammatory drug that causes ulcers on the gastric mucosa due to its use. Probiotic bacteria are live microorganisms, and it has been stated by various studies that these bacteria have antioxidant and anti-inflammatory effects. In this study, we investigated the possible protective effect of various types of probiotic bacteria (Lactobacillus rhamnosus, Lactobacillus fermentum, and Lactobacillus brevis) against acute gastric mucosal damage caused by indomethacin. METHODS: Control group - Physiological saline was administered daily for 10 days. Indo group-Physiological saline was administered daily for 10 days. Ranitidine + Indo group 5 mg/kg ranitidine dose was administered daily for 5 days. On day 11, a single dose of 100 mg/kg of indomethacin was given to the same group. Probiotic + Indo group 1 ml/kg of oral probiotic bacteria was administered daily for 10 days. On day 11, a single 100 mg/kg dose of indomethacin was given. After the application, the rats were anesthetized with ketamine xylazine, killed under appropriate conditions, the abdominal cavity was opened and the stomach tissues were removed. The obtained gastric tissues were used in the biochemical and histopathological analyses discussed below. All data were statistically evaluated by one-way ANOVA using SPSS 20.00, followed by Duncan Post hoc test. The data were expressed as mean ± SD. P < 0.05 was considered statistically significant. RESULTS: As a result, the administration of indomethacin caused gastric damage, stimulating oxidative stress, inflammation, and apoptosis. We found that the use of probiotic bacteria reduces oxidative stress (TOC), increases the activity of antioxidant enzymes (TAC), suppresses inflammation (IL-6 and Tnf-α), and inhibits apoptosis (Bax and Bcl-2) (P < 0.05). CONCLUSION: Probiotic treatment can mitigate gastric damage and apoptosis caused by indomethacin-induced gastric damage in rats. Probiotic also enhances the restoration of biochemical oxidative enzymes as it has anti-inflammatory, antioxidant, and antiapoptotic properties.


Subject(s)
Apoptosis , Gastric Mucosa , Indomethacin , Inflammation , Oxidative Stress , Probiotics , Stomach Ulcer , Indomethacin/adverse effects , Probiotics/pharmacology , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Stomach Ulcer/metabolism , Oxidative Stress/drug effects , Apoptosis/drug effects , Rats , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Inflammation/metabolism , Male , Rats, Wistar , Antioxidants/metabolism , Antioxidants/pharmacology
8.
Biomed Pharmacother ; 174: 116544, 2024 May.
Article in English | MEDLINE | ID: mdl-38599058

ABSTRACT

The current study was designed to investigate the potential of a synthetic therapeutic agent for better management of pain and inflammation, exhibiting minimal to non-existent ulcerogenic effects. The effect of 1-(2-chlorobenzoyl)-3-(2,3-dichlorophenyl) thiourea was assessed through model systems of nociception and anti-inflammatory activities in mice. In addition, the ulcerogenic potential was evaluated in rats using the NSAID-induced pyloric ligation model, followed by histopathological and biochemical analysis. The test was conducted on eight groups of albino rats, comprising of group I (normal saline), groups II and III (aspirin® at doses of 100 mg/kg and 150 mg/kg, respectively), groups IV and V (indomethacin at doses of 100 mg/kg and 150 mg/kg, respectively), and groups VI, VII, and VIII (lead-compound at 15 mg/kg, 30 mg/kg and 45 mg/kg doses, respectively). Furthermore, molecular docking analyses were performed to predict potential molecular target site interactions. The results showed that the lead-compound, administered at doses of 15, 30, and 45 mg/kg, yielded significant reductions in chemically and thermally induced nociceptive pain, aligning with the levels observed for aspirin® and tramadol. The compound also effectively suppressed inflammatory response in the carrageenan-induced paw edema model. As for the ulcerogenic effects, the compound groups displayed no considerable alterations compared to the aspirin® and indomethacin groups, which displayed substantial increases in ulcer scores, total acidity, free acidity, and gastric juice volume, and a decrease in gastric juice pH. In conclusion, these findings suggest that our test compound exhibits potent antinociceptive, anti-inflammatory properties and is devoid of ulcerogenic effects.


Subject(s)
Inflammation , Molecular Docking Simulation , Nociception , Stomach Ulcer , Thiourea , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Stomach Ulcer/drug therapy , Thiourea/analogs & derivatives , Thiourea/pharmacology , Male , Nociception/drug effects , Mice , Inflammation/drug therapy , Inflammation/pathology , Rats , Rats, Wistar , Analgesics/pharmacology , Analgesics/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Computer Simulation , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Indomethacin/pharmacology , Pain/drug therapy , Pain/chemically induced , Pain/pathology , Anti-Inflammatory Agents/pharmacology
9.
Biotech Histochem ; 99(3): 147-156, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38644776

ABSTRACT

The purpose of this study was to evaluate the effects of syringic acid, an anti-oxidant, on indomethacin induced gastric ulcers in rats. Experimental groups were control, ulcer, ulcer treated with 20 mg/kg esomeprazole (a proton pump inhibitor that reduces acid secretion), and ulcer treated with 100 mg/kg syringic acid. Rats were pretreated with esomeprazole or syringic acid two weeks before ulcer induction. Our histopathological observations showed that either syringic acid or esomeprazole attenuated the severity of gastric mucosal damage. Moreover, syringic acid and esomeprazole pretreatments alleviated indomethacin-induced damage by regulating oxidative stress, inflammatory response, the level of transforming growth factor-ß (TGF-ß), expressions of COX and prostaglandin E2, cell proliferation, apoptosis and regulation of the NF-κB signaling pathway. We conclude that either esomeprazole or syringic acid administration protected the gastric mucosa from harmful effects of indomethacin. Syringic acid might, therefore be a potential therapeutic agent for preventing and treating indomethacin-induced gastric damage.


Subject(s)
Apoptosis , Gallic Acid , Indomethacin , Inflammation , Oxidative Stress , Stomach Ulcer , Animals , Indomethacin/pharmacology , Indomethacin/toxicity , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology , Oxidative Stress/drug effects , Apoptosis/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Male , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Esomeprazole/pharmacology
10.
Inflammopharmacology ; 32(3): 1961-1982, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38652367

ABSTRACT

Gastric ulcer (GU) is one of the most common diseases of the upper gastrointestinal tract that affects millions of people worldwide. This study aimed to investigate the possible alleviating effect of a combined treatment of pantoprazole (PANTO) and adipose tissue-derived mesenchymal stem cells (ADSCs) in comparison with each treatment alone on the healing process of the experimentally induced GU in rats, and to uncover the involved pathways. Rats were divided into five groups: (1) Control, (2) GU, (3) PANTO, (4) ADSCs and (5) ADSCs + PANTO. Markers of oxidative stress, inflammation and apoptosis were assessed. The current data indicated that PANTO-, ADSCs- and ADSCs + PANTO-treated groups showed significant drop (p < 0.05) in serum advanced oxidation protein products (AOPPs) and advanced glycation end products (AGEPs) along with significant elevation (p < 0.05) in serum TAC versus the untreated GU group. Moreover, the treated groups (PANTO, ADSCs and ADSCs + PANTO) displayed significant down-regulation (p < 0.05) in gastric nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumor necrosis factor alpha (TNF-α), cyclooxygenase-2 (COX-2), intercellular adhesion molecule-1 (ICAM-1), matrix metallopeptidase 9 (MMP-9) and caspase-3 along with significant up-regulation (p < 0.05) in vascular endothelial growth factor (VEGF) and peroxisome proliferator-activated receptor gamma (PPARγ) genes expression compared to the untreated GU group. Immunohistochemical examination of gastric tissue for transforming growth factor ß1 (TGF-ß1), epidermal growth factor (EGF) and proliferating cell nuclear antigen (PCNA) showed moderate to mild and weak immune reactions, respectively in the PANTO-, ADSCs- and ADSCs + PANTO-treated rat. Histopathological investigation of gastric tissue revealed moderate to slight histopathological alterations and almost normal histological features of the epithelial cells, gastric mucosal layer, muscularis mucosa and submucosa in PANTO-, ADSCs- and ADSCs + PANTO-treated rats, respectively. Conclusively, the co-treatment with ADSCs and PANTO evidenced sententious physiological protection against GU by suppressing oxidative stress, inhibiting inflammation and reducing apoptosis with consequent acceleration of gastric tissue healing process.


Subject(s)
Apoptosis , Inflammation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Oxidative Stress , Pantoprazole , Stomach Ulcer , Animals , Oxidative Stress/drug effects , Stomach Ulcer/chemically induced , Rats , Apoptosis/drug effects , Pantoprazole/pharmacology , Inflammation/metabolism , Inflammation/drug therapy , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Rats, Wistar , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/administration & dosage , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Combined Modality Therapy
11.
Int J Pharm ; 657: 124143, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38663641

ABSTRACT

Gastric ulcer, a significant health issue characterized by the degradation of the gastric mucosa, often arises from excessive gastric acid secretion and poses a challenge in current medical treatments due to the limited efficacy and side effects of first-line drugs. Addressing this, our study develops a novel therapeutic strategy leveraging gas therapy, specifically targeting the release of hydrogen sulfide (H2S) in the treatment of gastric ulcers. We successfully developed a composite nanoparticle, named BSA·SH-DATS, through a two-step process. Initially, bovine serum albumin (BSA) was sulfhydrated to generate BSA·SH nanoparticles via a mercaptosylation method. Subsequently, these nanoparticles were further functionalized by incorporating diallyltrisulfide (DATS) through a precise Michael addition reaction. This sequential modification resulted in the creation of BSA·SH-DATS nanoparticles. Our comprehensive in vitro and in vivo investigations demonstrate that these nanoparticles possess an exceptional ability for site-specific action on gastric mucosal cells under the controlled release of H2S in response to endogenous glutathione (GSH), markedly diminishing the production of pro-inflammatory cytokines, thereby alleviating inflammation and apoptosis. Moreover, the BSA·SH-DATS nanoparticles effectively regulate critical inflammatory proteins, including NF-κB and Caspase-3. Our study underscores their potential as a transformative approach for gastric ulcer treatment.


Subject(s)
Allyl Compounds , Ethanol , Gastric Mucosa , Hydrogen Sulfide , Nanoparticles , Serum Albumin, Bovine , Stomach Ulcer , Sulfides , Animals , Sulfides/chemistry , Sulfides/administration & dosage , Sulfides/pharmacology , Nanoparticles/chemistry , Ethanol/chemistry , Allyl Compounds/chemistry , Allyl Compounds/pharmacology , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Gastric Mucosa/metabolism , Gastric Mucosa/drug effects , Hydrogen Sulfide/chemistry , Serum Albumin, Bovine/chemistry , Male , Apoptosis/drug effects , Glutathione/metabolism , Mice , Cytokines/metabolism , Humans , NF-kappa B/metabolism
12.
J Agric Food Chem ; 72(14): 7933-7942, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38546719

ABSTRACT

Ethanol (EtOH) has been identified as a potential pathogenic factor in gastric ulcer development primarily due to its association with gastric injury and excessive production of reactive oxygen species. Magnolol (Mag), the principal active compound in Magnolia officinalis extract, is well studied for its notable anti-inflammatory and antioxidant properties. However, its limited solubility, propensity for agglomeration, and low absorption and utilization rates significantly restrict its therapeutic use. This study aims to overcome these challenges by developing a Mag nanoparticle system targeting the treatment and prevention of EtOH-induced gastric ulcers in mice. Utilizing a click chemistry approach, we successfully synthesized this system by reacting thiolated bovine serum albumin (BSA·SH) with Mag. The in vitro analysis revealed effective uptake of the BSA·SH-Mag nanoparticle system by human gastric epithelial cells (GES-1), showcasing its antioxidant and anti-inflammatory capabilities. Additionally, BSA·SH-Mag exhibited gradual disintegration and release in simulated gastric fluid, resulting in a notable reduction of oxidative stress in gastric tissues and mucosal tissue repair and effectively reducing inflammatory expression. Furthermore, BSA·SH-Mag attenuated EtOH-induced gastric inflammation by decreasing the level of NOX4 protein expression and augmenting the level of Nrf2 protein expression. In conclusion, our findings indicate that BSA·SH-Mag represents a promising candidate as an oral therapeutic for gastric ulcer treatment.


Subject(s)
Biphenyl Compounds , Lignans , Nanoparticles , Stomach Ulcer , Mice , Humans , Animals , Ethanol/adverse effects , Ethanol/metabolism , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Antioxidants/metabolism , Anti-Inflammatory Agents/pharmacology , Gastric Mucosa/metabolism
13.
Toxicol Appl Pharmacol ; 484: 116880, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38447874

ABSTRACT

Gastric ulcer (GU) is a serious upper gastrointestinal tract disorder that affects people worldwide. The drugs now available for GU treatment have a high rate of relapses and drug interactions, as well as mild to severe side effects. As a result, new natural therapeutic medications for treating GU with fewer negative side effects are desperately needed. Because of quercetin's (QCT) diverse pharmacological effects and unique structural features, we decided to semi-synthesize new QCT derivatives and test them for antiulcer activity. Docking assays were performed on the synthesized compounds to determine their affinity for TLR-4/MD-2, MyD88/TIR, and NF-κB domains, an important inflammatory pathway involved in GU development and progression. Mice were given oral famotidine (40 mg/kg/day), QCT, QCT pentamethyl (QPM), or QCT pentaacetyl (QPA) (50 mg/kg/day) for 5 days before GU induction by a single intraperitoneal injection of indomethacin (INDO; 18 mg/kg). QPM and QPA have a stronger binding affinity for TLR-4/MD-2, MyD88/TIR and NF-κB domains than QCT. In comparison, they demonstrated the greatest reduction in ulcer score and index, gastric MDA and nitric oxide (NO) contents, MyD88 and NF-κB expressions, and gastric TLR-4 immunostaining. They also enhanced the levels of GSH, CAT, COX-1, and COX-2 in the gastric mucosa, as well as HO-1 and Nrf2 expression, with histological regression in gastric mucosal lesions, with QPA-treated mice demonstrating the best GU healing. QPA is safe against all of the target organs and adverse pathways studied, with good ADME properties. However, further in vitro experiments are necessary to demonstrate the inhibitory effects of QPM and QPA on the protein targets of interest. In addition, preclinical research on its bioavailability and safety is essential before clinical management can be undertaken. Overall, the new QPA derivative could one day serve as the basis for a new class of potential antiulcer drugs.


Subject(s)
Indomethacin , Stomach Ulcer , Humans , Mice , Animals , Indomethacin/toxicity , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology , Quercetin/pharmacology , Quercetin/therapeutic use , Molecular Docking Simulation , Ulcer/metabolism , Ulcer/pathology , NF-kappa B/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology
14.
J Ethnopharmacol ; 327: 117970, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38428660

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Solenostemma argel is widely distributed in Africa & Asia with traditional usage in alleviating abdominal colic, aches, & cramps. This plant is rich in phytochemicals, which must be explored for its pharmacological effects. PURPOSE: Peptic Ulcer Disease (PUD) is the digestion of the digestive tube. PUD not only interferes with food digestion & nutrient absorption, damages one of the largest defensive barriers against pathogenic micro-organisms, but also impedes drug absorption & bioavailability, rendering the oral route, the most convenient way, ineffective. Omeprazole, one of the indispensable cost-effective proton-pump inhibitors (PPIs) extensively prescribed to control PUD, is showing growing apprehensions toward multiple drug interactions & side effects. Hence, finding a natural alternative with Omeprazole-like activity & limited side effects is a medical concern. STUDY DESIGN: Therefore, we present Stemmoside C as a new gastroprotective phytochemical agent isolated from Solenostemma argel to be tested in upgrading doses against ethanol-induced gastric ulcers in mice compared to negative, positive, & reference Omeprazole groups. METHODS: We carried out in-depth pharmacological & histopathological studies to determine the possible mechanistic pathway. RESULTS: Our results showed that Stemmoside C protected the stomach against ethanol-induced gastric ulcers parallel to Omeprazole. Furthermore, the mechanistic studies revealed that Stemmoside C produced its effect using an orchestrated array of different mechanisms. Stemmoside C stimulates stomach defense by increasing COX-2, PGE-2, NO, & TFF-1 healing factors, IL-10 anti-inflammatory cytokine, & Nrf-2 & HO-1 anti-oxidant pathways. It also suppresses stomach ulceration by inhibiting leucocyte recruitment, especially neutrophils, leading to subsequent inhibition of NF-κBp65, TNF-α, IL-1ß, & iNOS pro-inflammatory cytokines & JAK-1/STAT-3 inflammation-induced carcinogenicity cascade in addition to MMP-9 responsible for tissue degradation. CONCLUSION: These findings cast light on Stemmoside C's clinical application against gastric ulcer progression, recurrence, & tumorigenicity & concurrently with chemotherapy.


Subject(s)
Anti-Ulcer Agents , Stomach Ulcer , Mice , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Extracts/metabolism , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Omeprazole/pharmacology , Omeprazole/therapeutic use , Ethanol/pharmacology , Cytokines/metabolism , Gastric Mucosa
15.
Br J Nutr ; 131(11): 1844-1851, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38443203

ABSTRACT

The primary goal of the investigation was to analyse the anti-inflammatory and antioxidant properties of Gamma-linolenic acid (GLA) on rats with indomethacin (IND)-induced gastric ulcers. Thirty rats were divided into five groups: Control, IND (50 mg/kg, p.o.), IND pretreated with GLA 100 mg/kg (p.o. for 14 d), IND pretreated with GLA 150 mg/kg (p.o. for 14 d) and IND pretreated with omeprazole (20 mg/kg, p.o. for 14 d). The stomach tissues were examined to calculate the ulcer index and pH and analyse biochemical markers (prostaglandin E2 (PGE2), cyclooxygenase 1 (COX1), TNF-1, IL-6 and intercellular adhesion molecule-1 (ICAM1)) and oxidative stress parameters (malondialdehyde: (MDA), superoxide dismutase (SOD), glutathione (GSH) and CAT (catalase)) as well as undergo histopathological assessment. GLA 100 and 150 mg/kg showed a protective effect against IND-induced gastric damage. It reduced levels of COX1, TNF-1, IL-6 and ICAM and increased PGE2 levels. GLA also normalised antioxidant function by modulating MDA, SOD, GSH and CAT. GLA intervention protects against IND-induced gastric ulcers by restoring oxidant/antioxidant balance and reducing inflammation.


Subject(s)
Antioxidants , Dinoprostone , Indomethacin , Oxidative Stress , Rats, Wistar , Stomach Ulcer , gamma-Linolenic Acid , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/drug therapy , Indomethacin/adverse effects , Antioxidants/pharmacology , Rats , Oxidative Stress/drug effects , gamma-Linolenic Acid/pharmacology , Male , Dinoprostone/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Interleukin-6/metabolism , Intercellular Adhesion Molecule-1/metabolism , Superoxide Dismutase/metabolism , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Glutathione/metabolism , Tumor Necrosis Factor-alpha/metabolism , Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 1/metabolism , Malondialdehyde/metabolism , Omeprazole/pharmacology
16.
Dig Dis Sci ; 69(4): 1156-1168, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448762

ABSTRACT

BACKGROUND/AIMS: We examined the involvement of cholecystokinin (CCK) in the exacerbation of indomethacin (IND)-induced gastric antral ulcers by gastroparesis caused by atropine or dopamine in mice. METHODS: Male mice were fed for 2 h (re-feeding) following a 22-h fast. Indomethacin (IND; 10 mg/kg, s.c.) was administered after re-feeding; gastric lesions were examined 24 h after IND treatment. In another experiment, mice were fed for 2 h after a 22-h fast, after which the stomachs were removed 1.5 h after the end of the feeding period. Antral lesions, the amount of gastric contents, and the gastric luminal bile acids concentration were measured with or without the administration of the pro- and antimotility drugs CCK-octapeptide (CCK-8), atropine, dopamine, SR57227 (5-HT3 receptor agonist), apomorphine, lorglumide (CCK1 receptor antagonist), ondansetron, and haloperidol alone and in combination. RESULTS: IND produced severe lesions only in the gastric antrum in re-fed mice. CCK-8, atropine, dopamine, SR57227 and apomorphine administered just after re-feeding increased bile reflux and worsened IND-induced antral lesions. These effects were significantly prevented by pretreatment with lorglumide. Although atropine and dopamine also increased the amount of gastric content, lorglumide had no effect on the delayed gastric emptying provoked by atropine and dopamine. Both ondansetron and haloperidol significantly inhibited the increase of bile reflux and the exacerbation of antral lesions induced by atropine and dopamine, respectively, but did not affect the effects of CCK-8. CONCLUSIONS: These results suggest that CCK-CCK1 receptor signal increases bile reflux during gastroparesis induced by atropine and dopamine, exacerbating IND-induced antral ulcers.


Subject(s)
Bile Reflux , Gastroparesis , Stomach Ulcer , Mice , Male , Animals , Indomethacin , Ulcer , Receptor, Cholecystokinin A , Sincalide/adverse effects , Apomorphine/adverse effects , Dopamine , Haloperidol/adverse effects , Ondansetron , Stomach Ulcer/chemically induced , Cholecystokinin/adverse effects , Receptors, Cholecystokinin , Atropine/adverse effects
17.
Sci Rep ; 14(1): 6193, 2024 03 14.
Article in English | MEDLINE | ID: mdl-38486044

ABSTRACT

Gastric ulcers are a type of digestive disease that can severely affect a person's quality of life. Our study aimed to investigate the effects of fish oil on ethanol-induced gastric ulcers in rats, with the purpose of providing more comprehensive information on the topic. The study looked at various factors such as gastric ulcer index, and nitric oxide (NO) levels in stomach tissue. To investigate apoptosis, the mRNA levels of Bax, Bcl-2, and Caspase 3 were analyzed. The results showed that fish oil can reduce gastric acidity and the gastric ulcer index in cases of ethanol-induced gastric ulcers. It was found that fish oil can increase NO levels and improve the anti-apoptotic system by increasing the expression of Bcl-2 while decreasing the expression of Bax and Caspase 3. In general, the study demonstrates that fish oil can protect the stomach from ethanol-induced damage by reducing the apoptosis pathway via nitric oxide.


Subject(s)
Stomach Ulcer , Humans , Rats , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/metabolism , Caspase 3/metabolism , Gastric Mucosa/metabolism , Nitric Oxide/metabolism , Ethanol/toxicity , Ethanol/metabolism , Fish Oils/adverse effects , Quality of Life , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Apoptosis
18.
PeerJ ; 12: e17062, 2024.
Article in English | MEDLINE | ID: mdl-38435992

ABSTRACT

Background: Obesity leads to an elevated risk of developing gastrointestinal disease such as gastric ulcers. Callistemon citrinus leaf extract has shown antioxidant, antimicrobial, hepatoprotective, and chemoprotective effects against colon cancer. The aim of this study is to evaluate the gastroprotective effect of C. citrinus leaf extract on indomethacin-induced gastric ulcers in obese rats. Methods: Gastric ulcers were induced in female obese Wistar rats using a single oral dose of indomethacin (IND). In the first stage, the rats were fed with a high fat sugar diet (HFSD) for 15 weeks to induce obesity and, at the same time, the diet of the other group of animals included daily administration of ethanolic C. citrinus leaf extract (250 mg/kg) in addition to HFSD. In the second stage, gastric ulcers were induced with IND (30 mg/kg). The gastroprotective activity of C. citrinus, the inflammatory enzyme activities, and cytokines in the stomach were determined. Results: C. citrinus produced a reduction of gastric lesions caused by IND. Myeloperoxidase (MPO), cyclooxygenase-2 (COX-2), and 5-lipoxygenase (5-LOX) activities also decreased. Although inflammatory biomarkers such as TNFα, IL-6, AOPP, and leptin were significantly decreased by C. citrinus, adiponectin levels increased. Moreover, C. citrinus decreased weight gain and morphological and biochemical parameters. Conclusion: The use of indomethacin in rats fed with a high fat-sugar diet increased gastric ulcers. Gastroprotective effect of C. citrinus in obese rats is attributed to the reduction of pro-inflammatory cytokines and the inflammatory enzymes.


Subject(s)
Indomethacin , Stomach Ulcer , Female , Rats , Animals , Stomach Ulcer/chemically induced , Rats, Wistar , Anti-Inflammatory Agents , Obesity/complications , CD36 Antigens , Sugars , Cytokines , Plant Extracts/pharmacology
19.
Mol Biol Rep ; 51(1): 401, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38457071

ABSTRACT

BACKGROUND: Gastric ulcer (GU) is a common gastrointestinal tract illness. Aloe vera has anti-inflammatory, antioxidant, and healing characteristics. This research sought to explore the therapeutic impact of Aloe vera gel on ethanol-provoked GU in rats and to elucidate the underlying mechanisms involved. METHODS: An ethanol-induced GU rat model was constructed using forty male Wistar rats distributed at random into four groups: control, ulcer, pantoprazole, and Aloe vera. Gross evaluation of the stomach, ulcer index (UI), inhibition index, and gastric pH estimation were analyzed. Gastric malondialdehyde (MDA) and reduced glutathione (GSH) were determined using the spectrophotometric method, and serum gastrin level was measured by an enzyme-linked immunosorbent assay. Gastric nucleotide-binding domain, leucine-rich repeat, and pyrin domain PYD containing protein 3 (NLRP3) and gasdermin D (GSDMD) mRNA expression levels were estimated by quantitative real-time PCR. Finally, the histopathological examination of the glandular part of stomach tissue was done. RESULTS: The ulcer group revealed a significant increase in MDA, gastrin, NLRP3, and GSDMD and a decrease in gastric pH and GSH compared to the control group. Gross investigations of the ulcer group revealed a hemorrhagic lesion in the stomach and an increase in UI. Also, histopathological results for this group showed severe epithelial loss, haemorrhage, inflammatory cell infiltration, and blood vessel congestion. However, Aloe vera treatment improved the gross, biochemical, molecular, and histopathological alterations induced by ethanol when compared to the ulcer group. CONCLUSIONS: Aloe vera exerted antiulcer activities through modulation of oxidant/antioxidant status, anti-secretory properties, and mitigation of pyroptosis.


Subject(s)
Plant Preparations , Stomach Ulcer , Rats , Male , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Antioxidants/pharmacology , Antioxidants/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein , Ethanol/adverse effects , Ulcer/drug therapy , Gastrins/therapeutic use , Pyroptosis , Rats, Wistar , Plant Extracts/pharmacology , Signal Transduction
20.
Microbiol Spectr ; 12(4): e0389423, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38488394

ABSTRACT

Recently, Enterococcus has been shown to have gastric protective functions, and the mechanisms by which Enterococcus modulates gastric function are still being investigated. Herein, we investigated how Enterococcus faecium (Efm) and E. faecium-derived extracellular vesicles (EVs) (EfmEVs) exert protective effect against ethanol-induced gastric injury by investigating the effect of EfmEVs on gastric mucosal ulcer scoring, histological lesion, mucosal glycoprotein production, acidity, anti-oxidative function, and inflammatory responses in rat. Pretreatment with Efm showed significant reduction of ethanol-induced gastric injury, as evidenced by the lowering of ulcer index, histological lesion, gastric pH, and inflammatory responses and the enhancement of mucosal glycoprotein production and anti-oxidative function. Further functional studies on three bioactive components [inactivated Efm, EfmEVs (EVs), and EV-free supernatants] of the bacterial culture showed that EVs are mostly responsible for the gastroprotective effect. Moreover, EV secretion is beneficial for the gastroprotective effect of Efm. Hence, EVs mediated the protective effect of Efm against ethanol-induced gastric injury by lowering inflammatory responses and enhancing anti-oxidative function and may be a potent anti-inflammatory and anti-oxidative strategy to alleviate hyperinflammatory gastrointestinal tract conditions.IMPORTANCEThis study indicated that Enterococcus faecium provided a protective effect against rat gastric injury, which involved improvement of the mucosal glycoprotein production, anti-oxidative function, and inflammatory responses. Furthermore, we confirmed that three bioactive components (inactivated Efm, extracellular vesicles, and EV-free supernatants) of E. faecium culture also contributed to the gastroprotective effect. Importantly, E. faecium-derived EVs showed an effective impact for the gastroprotective effect.


Subject(s)
Enterococcus faecium , Stomach Ulcer , Rats , Animals , Oxidative Stress , Ulcer , Ethanol/toxicity , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Stomach Ulcer/pathology , Glycoproteins
SELECTION OF CITATIONS
SEARCH DETAIL
...