Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.624
Filter
1.
Sci Rep ; 14(1): 13003, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844563

ABSTRACT

Helicobacter pylori eradication therapy reduces the risk of gastric cancer. However, it is unclear whether the severity of risk factors for gastric cancer such as atrophy and intestinal metaplasia are reduced after eradication in the long term. We aimed to study long-term changes in endoscopic risk factors for gastric cancer up to 20 years post-eradication. The endoscopic severity of gastritis according to the Kyoto Classification of Gastritis in 167 patients was retrospectively evaluated over an average follow-up 15.7 years. A significant improvement in mean total gastric cancer risk score (4.36 ± 1.66 to 2.69 ± 1.07, p < 0.001), atrophy (1.73 ± 0.44 to 1.61 ± 0.49, p = 0.004), and diffuse redness (1.22 ± 0.79 to 0.02 ± 0.13, p < 0.001) was observed compared to baseline in the Eradication group. However, there was no change in the never infection and current infection groups. The frequency of map-like redness increased over time until 15 years (3.6% to 18.7%, p = 0.03). The Cancer group had significantly higher risk scores at all time points. Endoscopic atrophy significantly improved in eradicated patients over long-term, suggested that eradication is one of the key elements in gastric cancer prevention. Individualized surveillance strategies based on endoscopic gastritis severity before eradication may be important for those at risk of gastric cancer.


Subject(s)
Gastric Mucosa , Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Male , Helicobacter Infections/drug therapy , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Female , Helicobacter pylori/drug effects , Middle Aged , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/drug effects , Retrospective Studies , Stomach Neoplasms/pathology , Stomach Neoplasms/microbiology , Aged , Adult , Risk Factors , Gastritis/microbiology , Gastritis/drug therapy , Gastritis/pathology , Gastroscopy , Follow-Up Studies , Anti-Bacterial Agents/therapeutic use
2.
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
3.
Int J Immunopathol Pharmacol ; 38: 3946320241249429, 2024.
Article in English | MEDLINE | ID: mdl-38721971

ABSTRACT

OBJECTIVE: This study investigated the raft-forming suspension of famotidine as an anti-reflux formulation to improve the oral bioavailability of narrow absorption window drugs by enhancing gastric residence time (GRT) and preventing gastro-esophageal reflux disease (GERD). METHOD: Various combinations of raft-forming agents, such as Tragacanth gum (TG), guar gum (GG), and xanthan gum (XG), were evaluated alongside sodium alginate (SA) to develop an effective raft. Preformulation studies and preliminary screening were conducted to identify the most suitable raft-forming agent, and GG was chosen due to its mucilaginous properties. The formulation was optimized using a 32 full factorial design, with the quantities of GG and SA as independent factors and apparent viscosity and in-vitro drug release (%) as dependent factors. The in vivo floating behavior study was performed for optimized and stabilized formulation. RESULTS: Among the tested batches, F6 was selected as the optimized formulation. It exhibited desirable characteristics such as adequate raft weight for extended floating in gastric fluid, improved apparent viscosity, and a significant percentage of drug release at 12 h. A mathematical model was applied to the in-vitro data to gain insights into the drug release mechanism of the formulation. The stability of the suspension was assessed under accelerated conditions, and it demonstrated satisfactory stability. The formulation remains floating in the Rabbit stomach for more than 12 h. CONCLUSION: It concludes that the developed formulation has enhanced bioavailability in the combination of GG and SA. The floating layer of the raft prevents acid reflux, and the famotidine is retained for an extended period of time in the gastric region, preventing excess acid secretion. The developed formulations are effective for stomach ulcers and GERD, with the effect of reducing acid secretion by H2 receptor antagonists.


Subject(s)
Drug Delivery Systems , Famotidine , Galactans , Famotidine/administration & dosage , Famotidine/pharmacokinetics , Animals , Drug Delivery Systems/methods , Drug Liberation , Alginates , Gastroesophageal Reflux/drug therapy , Gastroesophageal Reflux/metabolism , Biological Availability , Mannans/administration & dosage , Plant Gums , Viscosity , Male , Rabbits , Gastric Mucosa/metabolism , Gastric Mucosa/drug effects , Polysaccharides, Bacterial , Drug Stability , Administration, Oral
4.
Nutrients ; 16(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38794668

ABSTRACT

INTRODUCTION: Justicia pectoralis Jacq. is traditionally applied in folk medicine in Brazil and in several Latin American countries. The leaves are used in tea form, especially in the treatment of respiratory disorders, acting as an expectorant. It also has activity in gastrointestinal disorders, and it is anti-inflammatory, antioxidant, sedative, and estrogenic, among others. AIMS: To investigate the gastroprotective activity of the methanol extract of the leaves of Justicia pectoralis Jacq. (MEJP) in different experimental models of gastric ulcers. MATERIALS AND METHODS: The adult leaves of Justicia pectoralis Jacq. were collected and cultivated in beds, with an approximate spacing of 40 × 40 cm, organic fertilization, irrigation with potable water and without shelter from light. The MEJP was prepared from the dried and pulverized leaves and concentrated under reduced pressure in a rotary evaporator. For the experimental model of gastric ulcer, Swiss male albino mice were used. The inputs used in the experiment were MEJP at three different concentrations (250, 500 and 1000 mg/kg p.o.), cimetidine (50 mg/kg p.o.), indomethacin (50 mg/kg s.c.) and vehicle (10 mL/kg p.o.). RESULTS: MEJP (250, 500 and 1000 mg/kg p.o.) demonstrated gastroprotective activity, with levels of protection of 45.65%, 44.80% and 40.22%, respectively, compared to the control (vehicle). Compared with cimetidine (48.29%), MEJP showed similar gastroprotective activity. CONCLUSIONS: This study demonstrated the gastroprotective activity of MEJP and contributes to validate the traditional use the species for gastric disorders and provides a pharmacological basis for its clinical potential.


Subject(s)
Plant Extracts , Plant Leaves , Stomach Ulcer , Animals , Plant Extracts/pharmacology , Mice , Stomach Ulcer/drug therapy , Stomach Ulcer/prevention & control , Plant Leaves/chemistry , Male , Anti-Ulcer Agents/pharmacology , Methanol/chemistry , Justicia/chemistry , Disease Models, Animal , Cimetidine/pharmacology , Acanthaceae/chemistry , Indomethacin , Brazil , Gastric Mucosa/drug effects , Gastric Mucosa/pathology
5.
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
6.
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
7.
World J Gastroenterol ; 30(16): 2220-2232, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38690017

ABSTRACT

Several features of drug-induced mucosal alterations have been observed in the upper gastrointestinal tract, i.e., the esophagus, stomach, and duodenum. These include pill-induced esophagitis, desquamative esophagitis, worsening of gastroesophageal reflux, chemotherapy-induced esophagitis, proton pump inhibitor-induced gastric mucosal changes, medication-induced gastric erosions and ulcers, pseudomelanosis of the stomach, olmesartan-related gastric mucosal inflammation, lanthanum deposition in the stomach, zinc acetate hydrate tablet-induced gastric ulcer, immune-related adverse event gastritis, olmesartan-asso-ciated sprue-like enteropathy, pseudomelanosis of the duodenum, and lanthanum deposition in the duodenum. For endoscopists, acquiring accurate knowledge regarding these diverse drug-induced mucosal alterations is crucial not only for the correct diagnosis of these lesions but also for differential diag-nosis of other conditions. This minireview aims to provide essential information on drug-induced mucosal alterations observed on esophagogastroduodenoscopy, along with representative endoscopic images.


Subject(s)
Endoscopy, Digestive System , Humans , Endoscopy, Digestive System/methods , Gastric Mucosa/pathology , Gastric Mucosa/drug effects , Gastric Mucosa/diagnostic imaging , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/diagnostic imaging , Proton Pump Inhibitors/adverse effects , Esophageal Mucosa/pathology , Esophageal Mucosa/drug effects , Esophageal Mucosa/diagnostic imaging
8.
Molecules ; 29(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731435

ABSTRACT

Self-assembled peptide-based nanobiomaterials exhibit promising prospects for drug delivery applications owing to their commendable biocompatibility and biodegradability, facile tissue uptake and utilization, and minimal or negligible unexpected toxicity. TFF3 is an active peptide autonomously secreted by gastric mucosal cells, possessing multiple biological functions. It acts on the surface of the gastric mucosa, facilitating the repair process of gastric mucosal damage. However, when used as a drug, TFF3 faces significant challenges, including short retention time in the gastric mucosal cavity and deactivation due to degradation by stomach acid. In response to this challenge, we developed a self-assembled short peptide hydrogel, Rqdl10, designed as a delivery vehicle for TFF3. Our investigation encompasses an assessment of its properties, biocompatibility, controlled release of TFF3, and the mechanism underlying the promotion of gastric mucosal injury repair. Congo red/aniline blue staining revealed that Rqdl10 promptly self-assembled in PBS, forming hydrogels. Circular dichroism spectra indicated the presence of a stable ß-sheet secondary structure in the Rqdl10 hydrogel. Cryo-scanning electron microscopy and atomic force microscopy observations demonstrated that the Rqdl10 formed vesicle-like structures in the PBS, which were interconnected to construct a three-dimensional nanostructure. Moreover, the Rqdl10 hydrogel exhibited outstanding biocompatibility and could sustainably and slowly release TFF3. The utilization of the Rqdl10 hydrogel as a carrier for TFF3 substantially augmented its proliferative and migratory capabilities, while concurrently bolstering its anti-inflammatory and anti-apoptotic attributes following gastric mucosal injury. Our findings underscore the immense potential of the self-assembled peptide hydrogel Rqdl10 for biomedical applications, promising significant contributions to healthcare science.


Subject(s)
Gastric Mucosa , Hydrogels , Peptides , Trefoil Factor-3 , Hydrogels/chemistry , Trefoil Factor-3/chemistry , Trefoil Factor-3/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/injuries , Peptides/chemistry , Peptides/pharmacology , Animals , Humans , Drug Delivery Systems , Mice , Wound Healing/drug effects
9.
Bull Exp Biol Med ; 176(5): 581-584, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38724817

ABSTRACT

A bradykinin B1 receptors antagonist PAV-0056, an 1,4-benzodiazepin-2-one derivative, intragastrically administrated to mice at doses of 0.1 and 1 mg/kg causes analgesia in the "formalin test" not inferior to that of diclofenac sodium (10 mg/kg) and tramadol (20 mg/kg). PAV-0056 at doses of 0.1 and 10 mg/kg has no anxiolytic and central muscle relaxant effects in mice and does not damage the gastric mucosa in rats. Based on the results of the conditioned place preference test, PAV-0056 also does not induce addiction in mice.


Subject(s)
Analgesics , Animals , Mice , Rats , Male , Analgesics/pharmacology , Diclofenac/pharmacology , Tramadol/pharmacology , Psychotropic Drugs/pharmacology , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Anti-Anxiety Agents/pharmacology , Bradykinin B1 Receptor Antagonists/pharmacology , Rats, Wistar , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Pain Measurement/drug effects , Pain Measurement/methods
10.
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
11.
Drug Des Devel Ther ; 18: 1651-1672, 2024.
Article in English | MEDLINE | ID: mdl-38774485

ABSTRACT

Background: The Zuojin Pill (ZJP) is widely used for treating chronic atrophic gastritis (CAG) in clinical practice, effectively ameliorating symptoms such as vomiting, pain, and abdominal distension in patients. However, the underlying mechanisms of ZJP in treating CAG has not been fully elucidated. Purpose: This study aimed to clarify the characteristic function of ZJP in the treatment of CAG and its potential mechanism. Methods: The CAG model was established by alternant administrations of ammonia solution and sodium deoxycholate, as well as an irregular diet. Therapeutic effects of ZJP on body weight, serum biochemical indexes and general condition were analyzed. HE staining and AB-PAS staining were analyzed to characterize the mucosal injury and the thickness of gastric mucosa. Furthermore, network pharmacology and molecular docking were used to predict the regulatory mechanism and main active components of ZJP in CAG treatment. RT-PCR, immunohistochemistry, immunofluorescence and Western blotting were used to measure the expression levels of apoptosis-related proteins, gastric mucosal barrier-associated proteins and PI3K/Akt signaling pathway proteins. Results: The results demonstrated that ZJP significantly improved the general state of CAG rats, alleviated weight loss and gastric histological damage and reduced the serum biochemical indicators. Network pharmacology and molecular docking found that ZJP in treating CAG by inhibiting inflammation, suppressing apoptosis, and protecting the gastric mucosal barrier via the PI3K/Akt signaling pathway. Further experiments confirmed that ZJP obviously modulated the expression of key proteins involved in gastric mucosal cell apoptosis, such as Bax, Bad, Apaf-1, cleaved-caspase-3, cleaved-caspase-9, Cytochrome C, Bcl-2, and Bcl-xl. Moreover, ZJP significantly reversed the protein expression of Occludin, ZO-1, Claudin-4 and E-cadherin. Conclusion: Our study revealed that ZJP treats CAG by inhibiting the PI3K/Akt signaling pathway. This research provided a scientific basis for the rational use of ZJP in clinical practice.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal , Gastric Mucosa , Gastritis, Atrophic , Molecular Docking Simulation , Rats, Sprague-Dawley , Animals , Gastritis, Atrophic/drug therapy , Gastritis, Atrophic/pathology , Gastritis, Atrophic/metabolism , Rats , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Male , Chronic Disease , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Apoptosis/drug effects , Network Pharmacology , Proto-Oncogene Proteins c-akt/metabolism
14.
Food Funct ; 15(8): 4079-4094, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38563230

ABSTRACT

Gastritis is a common disease characterized by gastric ulcers and severe bleeding. Excessive daily alcohol consumption can cause acute gastritis, impacting individuals' quality of life. This study aims to explore the protective effects of different ethanol-fractional polysaccharides of Dendrobium officinale (EPDO) on acute alcohol-induced gastric injury in vivo. Results showed that EPDO-80, identified as a ß-glucan, exhibited significant anti-inflammatory properties in pathology. It could reduce the area of gastric mucosal injury and cell infiltration. EPDO-80 had a dose-effect relationship in reducing the levels of malondialdehyde and cyclooxygenase-2 and decreasing the levels of inflammation mediators such as tumor necrosis factor α. More extensively, EPDO-80 could inhibit the activation of the TNFR/IκB/NF-κB signaling pathway, reducing the production of TNF-α mRNA and cell apoptosis in organs. Conversely, EPDO-80 could promote changes in the gut microbiota structure. These findings suggest that EPDO-80 could have great potential in limiting oxidative stress and inflammation mediated by inhibiting the NF-κB signaling pathway, which is highly related to its ß-glucan structure and functions in gut microbiota.


Subject(s)
Dendrobium , Ethanol , Gastritis , NF-kappa B , Polysaccharides , Dendrobium/chemistry , Animals , Polysaccharides/pharmacology , Polysaccharides/chemistry , Gastritis/chemically induced , Gastritis/drug therapy , Male , Mice , NF-kappa B/metabolism , NF-kappa B/genetics , Gastrointestinal Microbiome/drug effects , Signal Transduction/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Plant Extracts/pharmacology , Oxidative Stress/drug effects , Anti-Inflammatory Agents/pharmacology , Protective Agents/pharmacology
15.
Drug Dev Ind Pharm ; 50(5): 460-469, 2024 May.
Article in English | MEDLINE | ID: mdl-38602337

ABSTRACT

OBJECTIVE: Ferulic acid (FA) is a promising nutraceutical molecule which exhibits antioxidant and anti-inflammatory properties, but it suffers from poor solubility and bioavailability. In the presented study, FA nanoemulsions were prepared to potentiate the therapeutic efficacy of FA in prevention of gastric ulcer. METHODS: FA nanoemulsions were prepared, pharmaceutically characterized, and the selected nanoemusion was tested for its ulcer-ameliorative properties in rats after induction of gastric ulcer using ethanol, by examination of stomach tissues, assessment of serum IL-1ß and TNF-α, assessment of nitric oxide, prostaglandin E2, glutathione, catalase and thiobarbituric acid reactive substance in stomach homogenates, as well as histological and immunohistochemical evaluation. RESULTS: Results revealed that the selected FA nanoemulsion showed a particle size of 90.43 nm, sustained release of FA for 8 h, and better in vitro anti-inflammatory properties than FA. Moreover, FA nanoemulsion exhibited significantly better anti-inflammatory and antioxidant properties in vivo, and the gastric tissue treated with FA nanoemulsion was comparable to the normal control upon histological and immunohistochemical evaluation. CONCLUSION: Findings suggest that the prepared ferulic acid nanoemulsion is an ideal anti-ulcer system, which is worthy of further investigations.


Subject(s)
Anti-Ulcer Agents , Antioxidants , Coumaric Acids , Emulsions , Nanoparticles , Stomach Ulcer , Animals , Coumaric Acids/pharmacology , Coumaric Acids/chemistry , Emulsions/chemistry , Stomach Ulcer/drug therapy , Rats , Antioxidants/pharmacology , Antioxidants/chemistry , Male , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/chemistry , Anti-Ulcer Agents/pharmacokinetics , Nanoparticles/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/administration & dosage , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Rats, Wistar , Particle Size , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Solubility , Nitric Oxide/metabolism
16.
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
17.
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
18.
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
19.
Inflammopharmacology ; 32(3): 2049-2060, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38570398

ABSTRACT

Gastric ulcers affect approx. 10% of population. Non-steroidal anti-inflammatory drugs (NSAIDs), including acetylsalicylic acid (ASA) predispose to or impair the physiologically complex healing of pre-existing ulcers. Since H2S is an endogenous cytoprotective molecule, we hypothesized that new H2S-releasing ASA-derivative (ATB-340) could overcome pathological impact of NSAIDs on GI regeneration.Clinically translational gastric ulcers were induced in Wistar rats using state-of-the-art microsurgical model employing serosal application of acetic acid. This was followed by 9 days long i.g. daily treatment with vehicle, ATB-340 (6-24 mg/kg) or equimolar ASA doses (4-14 mg/kg). Ulcer area was assessed macro- and microscopically. Prostaglandin (PG)E2  levels, indicating pharmacological activity of NSAIDs and 8-hydroxyguanozine content, reflecting nucleic acids oxidation in serum/gastric mucosa, were determined by ELISA. Qualitative and/or quantitative pathway-specific alterations at the ulcer margin were evaluated using real-time PCR and mass spectrometry-based proteomics.ASA, unlike ATB-340, dose-dependently delayed/impaired gastric tissue recovery, deregulating 310 proteins at the ulcer margin, including Ras signalling, wound healing or apoptosis regulators. ATB-340 maintained NSAIDs-specific cyclooxygenase-inhibiting capacity on systemic and GI level but in time-dependent manner. High dose of ATB-340 (24 mg/kg daily), but not ASA, decreased nucleic acids oxidation and upregulated anti-oxidative/anti-inflammatory heme oxygenase-1, 24-dehydrocholesterol reductase or suppressor of cytokine signalling (SOCS3) at the ulcer margin.Thus, ASA impairs the physiological healing of pre-existing gastric ulcers, inducing the extensive molecularly functional and proteomic alterations at the wound margin. H2S-releasing ATB-340 maintains the target activity of NSAIDs with limited impact on gastric PGE2 signalling and physiological GI regeneration, enhancing anti-inflammatory and anti-oxidative response, and providing the pharmacological advantage.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Aspirin , Gastric Mucosa , Hydrogen Sulfide , Proteomics , Rats, Wistar , Stomach Ulcer , Wound Healing , Animals , Stomach Ulcer/drug therapy , Stomach Ulcer/metabolism , Aspirin/pharmacology , Rats , Proteomics/methods , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Male , Wound Healing/drug effects , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dinoprostone/metabolism , Chronic Disease , Dose-Response Relationship, Drug , Disease Models, Animal , Naproxen/analogs & derivatives
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...