Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.088
Filter
1.
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
2.
Recent Pat Biotechnol ; 18(4): 344-357, 2024.
Article in English | MEDLINE | ID: mdl-38566382

ABSTRACT

BACKGROUND: There are patents available related to fermented food and beverages which enhance to human health. Citrus limetta (Mosambi) has a high content of flavonoids and exhibits antioxidant activity, which could stimulate the digestive system and be useful for gastroprotective activity. It supports digestion by neutralizing the acidic digestive juices and reducing gastric acidity. OBJECTIVE: This study explored the potential of using waste peel extract from Citrus limetta to prevent ulcers. The study specifically sought to assess the anti-ulcer properties of fermented and non-fermented extracts and compare them. Further, the study looked at the potential benefits of treating or preventing ulcers with Citrus limetta waste peels and whether fermentation affected the efficacy of the treatment. METHODS: Thirty female Wistar albino rats were equally distributed into five different groups. Group 1 received distilled water (20 ml/kg/b.w); Group 2 received indomethacin (mg/kg/b.w); Group 3 received omeprazole (20 mg/kg/b.w); Group 4 received aqueous extract of Mosambi peel (400 mg/kg/b.w) and Group 5 received fermented product of extract of Mosambi peel (400 mg/kg/b.w). RESULTS: Findings explored that, compared to non-fermented citrus fruit juice, biofermented exhibited less gastric volume (1.58 ± 0.10 ml vs. 1.8 ± 0.14 ml), reduced MDA levels (355.23 ± 100.70 µmol/mg protein vs. 454.49 ± 155.88 µmol/mg protein), and low ulcer index (0.49 ± 0.07 vs. 0.72 ± 0.14). CONCLUSION: The results suggest that the bio-fermented product of Citrus limetta peel has better anti-ulcer potential against peptic ulcer induced by indomethacin in Wistar albino rats compared to non-fermented.


Subject(s)
Anti-Ulcer Agents , Citrus , Fermentation , Plant Extracts , Rats, Wistar , Stomach Ulcer , Animals , Citrus/chemistry , Female , Rats , Plant Extracts/pharmacology , Plant Extracts/chemistry , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/chemistry , Stomach Ulcer/drug therapy , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Patents as Topic , Indomethacin/metabolism , Fruit/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Omeprazole/pharmacology
3.
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
4.
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
5.
J Mol Med (Berl) ; 102(5): 679-692, 2024 05.
Article in English | MEDLINE | ID: mdl-38453697

ABSTRACT

Chronic kidney disease (CKD) is the 16th leading cause of mortality worldwide. Clinical studies have raised that long-term use of omeprazole (OME) is associated with the morbidity of CKD. OME is commonly used in clinical practice to treat peptic ulcers and gastroesophageal reflux disease. However, the mechanism underlying renal failure following OME treatment remains mostly unknown and the rodent model of OME-induced CKD is yet to be established. We described the process of renal injury after exposure to OME in mice; the early renal injury markers were increased in renal tubular epithelial cells (RTECs). And after long-term OME treatment, the OME-induced CKD mice model was established. Herein, aryl hydrocarbon receptor (AHR) translocation appeared after exposure to OME in HK-2 cells. Then for both in vivo and in vitro, we found that Ahr-knockout (KO) and AHR small interfering RNA (siRNA) substantially alleviated the OME-induced renal function impairment and tubular cell damage. Furthermore, our data demonstrate that antagonists of AHR and CYP1A1 could attenuate OME-induced tubular cell impairment in HK-2 cells. Taken together, these data indicate that OME induces CKD through the activation of the AHR-CYP axis in RTECs. Our findings suggest that blocking the AHR-CYP1A1 pathway acts as a potential strategy for the treatment of CKD caused by OME. KEY MESSAGES: We provide an omeprazole-induced chronic kidney disease (CKD) mice model. AHR activation and translocation process was involved in renal tubular damage and promoted the occurrence of CKD. The process of omeprazole nephrotoxicity can be ameliorated by blockade of the AHR-CYP1A1 axis.


Subject(s)
Cytochrome P-450 CYP1A1 , Mice, Inbred C57BL , Mice, Knockout , Omeprazole , Receptors, Aryl Hydrocarbon , Renal Insufficiency, Chronic , Animals , Humans , Male , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A1/genetics , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Omeprazole/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/chemically induced , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics
6.
Clin Pharmacokinet ; 63(4): 469-481, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38393578

ABSTRACT

BACKGROUND: We investigated the effect of a 5-day low-dose ritonavir therapy, as it is used in the treatment of COVID-19 with nirmatrelvir/ritonavir, on the pharmacokinetics of three factor Xa inhibitors (FXaI). Concurrently, the time course of the activities of the cytochromes P450 (CYP) 3A4, 2C19, and 2D6 was assessed. METHODS: In an open-label, fixed sequence clinical trial, the effect and duration of a 5-day oral ritonavir (100 mg twice daily) treatment on the pharmacokinetics of three oral microdosed FXaI (rivaroxaban 25 µg, apixaban 25 µg, and edoxaban 50 µg) and microdosed probe drugs (midazolam 25 µg, yohimbine 50 µg, and omeprazole 100 µg) was evaluated in eight healthy volunteers. The plasma concentrations of all drugs were quantified using validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) methods and pharmacokinetics were analysed using non-compartmental analyses. RESULTS: Ritonavir increased the exposure of apixaban, edoxaban, and rivaroxaban, but to a different extent the observed area under the plasma concentration-time curve (geometric mean ratio 1.29, 1.46, and 1.87, respectively). A strong CYP3A4 inhibition (geometric mean ratio > 10), a moderate CYP2C19 induction 2 days after ritonavir (0.64), and no alteration of CYP2D6 were observed. A CYP3A4 recovery half-life of 2.3 days was determined. CONCLUSION: This trial with three microdosed FXaI suggests that at most the rivaroxaban dose should be reduced during short-term ritonavir, and only in patients receiving high maintenance doses. Thorough time series analyses demonstrated differential effects on three different drug-metabolising enzymes over time with immediate profound inhibition of CYP3A4 and only slow recovery after discontinuation. CLINICAL TRIAL REGISTRATION: EudraCT number: 2021-006643-39.


Subject(s)
Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6 , Cytochrome P-450 CYP3A , Drug Interactions , Factor Xa Inhibitors , Healthy Volunteers , Pyridones , Ritonavir , Humans , Ritonavir/administration & dosage , Ritonavir/pharmacokinetics , Ritonavir/pharmacology , Male , Adult , Factor Xa Inhibitors/pharmacokinetics , Factor Xa Inhibitors/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Pyridones/pharmacokinetics , Pyridones/administration & dosage , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP2C19/genetics , Administration, Oral , Female , Rivaroxaban/pharmacokinetics , Rivaroxaban/administration & dosage , Young Adult , Pyridines/pharmacokinetics , Pyridines/administration & dosage , Pyridines/pharmacology , Pyrazoles/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Thiazoles/pharmacokinetics , Thiazoles/administration & dosage , Thiazoles/pharmacology , Midazolam/pharmacokinetics , Midazolam/administration & dosage , Omeprazole/pharmacokinetics , Omeprazole/administration & dosage , Omeprazole/pharmacology
7.
Inflammopharmacology ; 32(2): 1519-1529, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38227096

ABSTRACT

AIMS: Putative beneficial effects of neuropeptide W (NPW) in the early phase of gastric ulcer healing process and the involvement of cyclooxygenase (COX) enzymes were investigated in an acetic acid-induced gastric ulcer model. MAIN METHODS: In anesthetized male Sprague-Dawley rats, acetic acid was applied surgically on the serosa and then a COX-inhibitor (COX-2-selective NS-398, COX-1-selective ketorolac, or non-selective indomethacin; 2 mg/kg/day, 3 mg/kg/day or 5 mg/kg/day; respectively) or saline was injected intraperitoneally. One h after ulcer induction, omeprazole (20 mg/kg/day), NPW (0.1 µg/kg/day) or saline was intraperitoneally administered. Injections of NPW, COX-inhibitors, omeprazole or saline were continued for the following 2 days until rats were decapitated at the end of the third day. KEY FINDINGS: NPW treatment depressed gastric prostaglandin (PG) I2 level, but not PGE2 level. Similar to omeprazole, NPW treatment significantly reduced gastric and serum tumor necrosis factor-alpha and interleukin-1 beta levels and depressed the upregulation of nuclear factor kappa B (NF-κB) and COX-2 expressions due to ulcer. In parallel with the histopathological findings, treatment with NPW suppressed ulcer-induced increases in myeloperoxidase activity and malondialdehyde level and replenished glutathione level. However, the inhibitory effect of NPW on myeloperoxidase activity and NPW-induced increase in glutathione were not observed in the presence of COX-1 inhibitor ketorolac or the non-selective COX-inhibitor indomethacin. SIGNIFICANCE: In conclusion, NPW facilitated the healing of gastric injury in rats via the inhibition of pro-inflammatory cytokine production, oxidative stress and neutrophil infiltration as well as the downregulation of COX-2 protein and NF-κB gene expressions.


Subject(s)
Neuropeptides , Signal Transduction , Stomach Ulcer , Animals , Male , Rats , Acetates/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Gastric Mucosa , Glutathione/metabolism , Indomethacin/therapeutic use , Ketorolac/adverse effects , Neuropeptides/therapeutic use , NF-kappa B/metabolism , Omeprazole/pharmacology , Omeprazole/therapeutic use , Peroxidase/metabolism , Rats, Sprague-Dawley , Stomach Ulcer/drug therapy , Ulcer/metabolism , Ulcer/pathology
8.
Phytother Res ; 38(3): 1610-1622, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38296262

ABSTRACT

Gastroesophageal reflux disease (GERD) is the most common foregut disease, affecting about 20% of the adult population. Esophageal epithelial barrier plays a fundamental role in the pathophysiology of GERD; however, pharmacological therapies mainly aim to reduce the acidity of the gastroesophageal environment rather than to protect esophageal tissue integrity. This study aims to evaluate the efficacy of an oral solution containing xyloglucan and pea proteins (XP) in reestablishing gastroesophageal tissue integrity and biochemical markers. To induce GERD, C57BL/6 mice were alternatively overfed and fasted for 56 days and then treated with XP, sodium alginate, omeprazole, or omeprazole+XP twice daily for 7 days. Gastric pain and inflammatory markers were evaluated after 3 and 7 days of treatment. After sacrifice, the esophagi and stomachs were surgically removed for macroscopic and histological examination. Gastric pain was significantly reduced at days 3 and 7 by XP, omeprazole, and omeprazole+XP, while alginates were ineffective at day 3. XP was able to diminish gastric macroscopic damage and demonstrated the same efficacy as omeprazole in reducing esophageal damage. XP significantly reduced histological damage, with an efficacy comparable to that of omeprazole, but superior to alginates. Inflammatory markers were significantly reduced by XP, with superior efficacy compared with alginates at day 7. Interestingly, XP was also able to significantly increase gastric pH. This study demonstrated that XP restored gastric homeostasis, improved esophageal integrity, and decreased inflammation and pain with a similar efficacy to omeprazole and greater than alginates.


Subject(s)
Gastroesophageal Reflux , Glucans , Pea Proteins , Xylans , Animals , Mice , Pea Proteins/therapeutic use , Disease Models, Animal , Mice, Inbred C57BL , Gastroesophageal Reflux/drug therapy , Omeprazole/pharmacology , Omeprazole/therapeutic use , Pain/drug therapy
9.
J Equine Vet Sci ; 133: 105015, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38281606

ABSTRACT

Feedstuffs are often recommended to mitigate potential damage from acid associated with equine squamous gastric disease (ESGD). In acidic conditions, pectin alters its structure to one like mucus and binds the stomach mucosa, whilst alfalfa has a strong intrinsic acid buffering capacity. The study aimed to determine whether feeding a commercial beet pulp/alfalfa/oat fibre mix aids ESGD healing and/or prevention of recurrence. Ten adult horses with naturally occurring ESGD were included. All animals were treated with omeprazole as per the attending veterinarian's recommendation and randomly allocated to also be fed a commercial beet pulp/alfalfa/oat fibre mix (1Kg/horse divided into 2 meals/day; n=5) or no additional feed (n=5) for one month. Gastroscopy was then repeated to assess response to therapy. If the ESGD had healed, omeprazole therapy was discontinued, and the commercial feed given to all horses for a further month. Gastroscopy was repeated to determine ESGD recurrence. The mean (±SD) age of the horses was 11.6 (±3.8) years; 4 mares and 6 geldings; various breeds were represented; and the median (range) initial ESGD grade was 2 (2-4). ESGD had healed (grade 0/4) in all animals after one month. After a further month, ESGD had recurred in significantly (p=0.04) more animals that did not receive the commercial feed initially (3/5; 60%; mean [range] ESGD grade 3 [0,4]) compared to those that did (0/5; 0%; mean [range] ESGD grade 0 [0,0]). Thus, the commercial beet pulp/alfalfa/oat fibre mix aided prevention of ESGD recurrence when fed during the healing and prevention phases.


Subject(s)
Carcinoma, Squamous Cell , Horse Diseases , Stomach Diseases , Horses , Animals , Female , Male , Plant Breeding , Stomach Diseases/veterinary , Omeprazole/pharmacology , Omeprazole/therapeutic use , Horse Diseases/drug therapy , Horse Diseases/prevention & control , Medicago sativa , Carcinoma, Squamous Cell/veterinary
10.
Eur J Pharm Sci ; 194: 106653, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38006986

ABSTRACT

As a widely used antidepressant that works by inhibiting the reuptake of serotonin, sertraline exerts an antidepressant effect depending on its concentration in the brain, which might be limited by the blood-brain barrier (BBB). It is highly possible to combine proton pump inhibitors (PPIs) with sertraline in clinical trials. Nevertheless, the role played by PPIs in regulating the transport of sertraline across the BBB remains unclear. Here, the impact of PPIs on the distribution of sertraline in the brain and the mechanisms involved were investigated. A mouse brain distribution study showed that Omeprazole (OME), Pantoprazole (PAN), Ilaprazole (ILA), and Esomeprazole (ESO) increased the area under the brain concentration-time curves (AUC) for sertraline by 2.02-, 3.18-, 3.04-, and 4.21-fold, respectively, after the 14-day administration of PPIs. Besides, PPIs significantly increased the permeability of sertraline in brain perfusion experiments, with PAN having the highest rank order, followed by ILA, OME, and ESO. In the tail suspension test (TST), co-administration PPI groups showed significantly shorter immobility time than the control group. In vitro, four PPIs inhibited sertraline efflux in breast cancer resistance protein (BCRP)-overexpressing MDCKII cells, and showed a mixed inhibition type. In this study, PPIs were further found to inhibit the mRNA and protein expression of brain BCRP. To sum up, the findings of this study revealed that PPIs could enhance the brain distribution and antidepressant effect of sertraline, which may be attributed to the inhibition of BCRP expression at the BBB by PPIs.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles , Proton Pump Inhibitors , Sertraline , Animals , Mice , Proton Pump Inhibitors/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Sertraline/pharmacology , Blood-Brain Barrier/metabolism , Neoplasm Proteins/metabolism , Omeprazole/pharmacology , Esomeprazole , Antidepressive Agents/pharmacology , Pantoprazole/pharmacology
11.
CPT Pharmacometrics Syst Pharmacol ; 13(2): 247-256, 2024 02.
Article in English | MEDLINE | ID: mdl-38130031

ABSTRACT

Proton pump inhibitors (PPIs) can affect the release of drugs from their dosage forms in vivo by elevating the gastric pH. Our recent clinical study has demonstrated that drug-drug interactions (DDIs) exist between a PPI, omeprazole, and nifedipine extended-release formulations, where systemic exposure of nifedipine was increased in subjects after multiple-dose pretreatment of omeprazole. However, the mechanism of the observed DDIs between omeprazole and nifedipine has not been well-understood, as the DDI may also be mediated through CYP3A4 enzyme inhibition in addition to the elevated gastric pH caused by omeprazole. This study used physiologically-based pharmacokinetic (PBPK) modeling and simulations to investigate the underlying mechanism of these complex DDIs. A formulation exhibiting differences in in vitro dissolution across physiological pH range and another formulation where pH does not impact dissolution appreciably (e.g., an osmotic pump) were chosen to characterize the potential impact of pH. The PBPK models incorporated two-stage in vitro release profiles via US Pharmacopeia 2 apparatus. PBPK simulations suggest that the elevated gastric pH following multiple-dose administration of omeprazole has a minimal effect on nifedipine pharmacokinetics (PKs), whereas CYP3A4-mediated DDI is likely the main driver to the observed change of nifedipine PKs in the presence of omeprazole. Compared to the osmotic formulation, the slightly increased exposure of nifedipine can be accounted for by the enhanced drug release in the pH-dependent formulation. The reported model-based approach may be useful in DDI risk assessments, product formulation designs, and bioequivalence evaluations.


Subject(s)
Nifedipine , Omeprazole , Humans , Nifedipine/chemistry , Nifedipine/pharmacokinetics , Omeprazole/pharmacology , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Drug Liberation , Administration, Oral
12.
J Matern Fetal Neonatal Med ; 36(2): 2272577, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37884440

ABSTRACT

OBJECTIVE: To investigate the correlation between the aryl hydrocarbon receptor (AhR) and reactive oxygen species (ROS) in peripheral blood mononuclear cells (PBMCs) of premature infants, to demonstrate the protective role of AhR against hyperoxia-induced oxidative stress in premature infants and to provide a rational basis for the use of omeprazole (OM) as a new treatment for bronchopulmonary dysplasia (BPD). METHODS: From January 2021 to June 2021, 1-3 ml of discarded peripheral blood was collected from premature infants of gestational age less than 32 weeks who were not taking inhaled oxygen and were admitted to the Department of Neonatology of the Affiliated Hospital of Southwest Medical University. Using a random number table, the PBMCs were randomly assigned to each of the following groups: the control group, air + OM group, hyperoxia group, and hyperoxia + OM group. After 48 h of in vitro modeling and culture, PBMCs and the culture medium of each group were collected. Immunofluorescence analysis was used to examine ROS levels in PBMCs. A full-spectrum spectrophotometer was used to examine malondialdehyde (MDA) levels in the culture medium. Enzyme-linked immunosorbent assay (ELISA) was used to examine monocyte chemotactic protein 1 (MCP-1) levels in culture medium. Immunofluorescence analysis was used to examine the intracellular localization of AhR. Western blotting was used to examine the expression level of AhR in PBMCs. RESULTS: Compared with those in the control group, the levels of ROS, MDA, and MCP-1 and the cytoplasm-nuclear translocation rate of AhR in the air + OM group did not change significantly (p > 0.05), but the expression level of AhR increased significantly (p < 0.05). The levels of ROS, MDA, and MCP-1 and the cytoplasm-nuclear translocation rate of AhR significantly increased in the hyperoxia group (p < 0.05), and the expression level of AhR was significantly reduced (p < 0.05). Compared with those in the hyperoxia group, the levels of ROS, MDA, and MCP-1 in the hyperoxia + OM group were significantly reduced (p < 0.05), and the cytoplasm-nuclear translocation rate of AhR and the expression level of AhR were significantly increased (p < 0.05), but did not reach the level of the control group (p < 0.05). CONCLUSION: OM can activate AhR to inhibit hyperoxia-induced oxidative stress in the PBMCs from premature infants.


Subject(s)
Hyperoxia , Humans , Infant, Newborn , Infant , Hyperoxia/complications , Reactive Oxygen Species/metabolism , Omeprazole/pharmacology , Omeprazole/therapeutic use , Receptors, Aryl Hydrocarbon/metabolism , Leukocytes, Mononuclear/metabolism , Infant, Premature , Oxidative Stress , Lung/metabolism
13.
Vet Med Sci ; 9(6): 2594-2599, 2023 11.
Article in English | MEDLINE | ID: mdl-37872836

ABSTRACT

BACKGROUND: Fungal and yeast infections, including those caused by Malassezia spp., are becoming increasingly difficult to treat, likely due to the occurrence of drug resistance. OBJECTIVES: This study aimed to evaluate the antifungal effects of omeprazole (OME), a proton pump inhibitor, against antifungal-resistant Malassezia pachydermatis and to investigate the potential synergistic effects between OME and other antifungal compounds. METHODS: In total, 15 samples of M. pachydermatis isolated from the skin of dogs were tested. The susceptibility of M. pachydermatis to itraconazole, ketoconazole, miconazole, terbinafine and OME was assessed using a modified broth microdilution (BM) method. The in vitro efficacy of OME alone and in combination with other antifungal compounds was evaluated for all isolates using the BM chequerboard method. The data obtained were analysed using the fractional inhibitory concentration index (FICI). RESULTS: The minimum inhibitory concentration (MIC) values of antifungal compounds and OME against quality control strain (M. pachydermatis CBS1879) were lower than the MIC90 values of same drugs against clinically collected strains. There was no significant difference in MIC values between drugs alone and combination. According to the analysis by the FICI method, no interaction was observed with OME and antifungal compounds. CONCLUSIONS: Most M. pachydermatis strains were resistant to azole antifungal compounds. OME exerted antifungal effects against Malassezia spp. and even showed good effects on antifungal-resistant strains. No synergistic effects were observed between the antifungal compounds and OME.


Subject(s)
Antifungal Agents , Malassezia , Animals , Dogs , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Omeprazole/pharmacology , Drug Resistance, Fungal
14.
Mol Biol Rep ; 50(11): 9085-9098, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37741810

ABSTRACT

BACKGROUND: A gastric ulcer is a painful lesion of the gastric mucosa that can be debilitating or even fatal. The effectiveness of several plant extracts in the therapy of this illness has been demonstrated in traditional pharmacopoeias. AIM: this study was aimed to see if propolis, ginseng in normal or nano form, and amygdalin might help in preventing the ulcerative effects of absolute ethanol. METHODS: Gastroprotective properties of pretreatments before ethanol gavage in rats were compared to omeprazole. The ulcer and stomach parameters (ulcerated regions) were measured (mm2), ulcer inhibition percentage, the stomachs were assessed macroscopically with gastric biopsy histological examinations. RESULTS: Amygdalin, normal and nano ginseng, nano propolis followed by propolis all showed great efficacy in protecting the cyto-architecture and function of the gastric mucosa. The number of ulcerated sites was greatly reduced, and the percentage of stomach protection was increased. Histopathological examination had confirmed great protective effects of the nanoformulations followed by amygdalin. The protection and healing rate was completed to about 100% in all tested materials while ulcer areas were still partially unhealed in normal propolis and omeprazole. Quantitative assay of the m-RNA levels Enothelin 1(ET-1), leukotriene4 (LT-4), and caspase 3(Cas-3) genes and Histamine were done and revealed significant up-regulations in ethanol group and the maximum protective effect was reported with ginseng nano, moreover the histamine content was significantly decreased with nano- formulated extracts. CONCLUSION: Amygdalin and the nanoformulated ginseng and propolis had exhibited a marked protective effect against the ulcerative toxic effects of ethanol.


Subject(s)
Amygdalin , Anti-Ulcer Agents , Propolis , Stomach Ulcer , Rats , Animals , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology , Ulcer/drug therapy , Ulcer/pathology , Propolis/pharmacology , Amygdalin/pharmacology , Histamine/pharmacology , Histamine/therapeutic use , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Gastric Mucosa , Omeprazole/pharmacology , Ethanol/adverse effects
15.
Indian J Gastroenterol ; 42(5): 686-693, 2023 10.
Article in English | MEDLINE | ID: mdl-37665542

ABSTRACT

BACKGROUND: Long-term use of proton pump inhibitors (PPIs) can increase the risk of gastric cancer in Helicobacter pylori-infected patients; nevertheless, there is no data about their impact on the pathogenicity of H. pylori. This study aimed at investigating the transcriptional alteration of key gene mediators of cytotoxin-associated gene-pathogenicity island (cag-PAI) among clinical H. pylori isolates in response to omeprazole at different pH levels. METHODS: Accordingly, H. pylori isolates with the same virulence genotypes selected from the gastric biopsies of patients and transcriptional alteration in the cag-PAI genes studied in the presence or absence of omeprazole (2 mg/mL) at pH 2.0, 4.0 and 7.0 after 30 and 90 minutes of the treatment. Relative changes in the transcriptional levels were recorded in each assay, separately. RESULTS: Of 18 H. pylori isolates, the cag-PAI empty site was detected in four strains, while the presence of cagA, cagL and cagY was characterized in 77.7%, 83.3% and 83.3% of the cag-PAI-positive strains, respectively. Transcriptional analysis of the selected strains showed up-regulation of cagA and cagL, mainly at pH 2.0 and 4.0 after 30 and 90-minute exposure. A diversity in the expression levels of cag-PAI genes was seen among the strains at the extent and time of induction. CONCLUSION: Our results showed that omeprazole could increase the expression of H. pylori cagA and cagL at acidic pH. Heterogeneity among the strains probably has an impact on the extent of their interplay with PPIs. Further studies are needed to establish this correlation.


Subject(s)
Helicobacter pylori , Proton Pump Inhibitors , Humans , Proton Pump Inhibitors/adverse effects , Helicobacter pylori/genetics , Genomic Islands/genetics , Omeprazole/pharmacology , Hydrogen-Ion Concentration
16.
Mol Biol Rep ; 50(10): 8615-8622, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37648947

ABSTRACT

BACKGROUND: Alpha-pinene (α-pinene) is a monoterpene with gastroprotective activity. We evaluated the gastroprotective effect of α-pinene in the gastric damage model with ethanol. METHODS: We evaluated the macroscopic evaluation of the stomach cavity, alteration in pH, mRNA expression of nuclear factor erythroid 2- related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), total antioxidant capacity (TAC) levels, and histopathological changes. RESULTS: Pretreatment with α-pinene (10, 50 and 100 mg/kg i.p.) before oral administration of ethanol reduced gastric mucosal damage by increasing the percentage of ulcer inhibition. Alpha-pinene also increased gastric pH similar to omeprazole. In addition, the histopathological examination showed that in the groups pretreated with α-pinene 50 and 100 mg/kg, and omeprazole20 mg/kg, the lesions were less than the control group. Moreover, α- pinene 10, 50, 100, and omeprazole 20 mg/kg upregulated the NRF2 and HO1. CONCLUSIONS: Our results show that pretreatment with α-pinene is effective in reducing ethanol-induced gastric damage through regulation of Nrf2/HO-1.


Subject(s)
Heme Oxygenase-1 , NF-E2-Related Factor 2 , Animals , Rats , Heme Oxygenase-1/genetics , Stomach , Signal Transduction , Ethanol , Omeprazole/pharmacology
17.
J Vet Intern Med ; 37(4): 1537-1543, 2023.
Article in English | MEDLINE | ID: mdl-37390114

ABSTRACT

BACKGROUND: Rebound gastric hyperacidity (RGH) secondary to hypergastrinemia has been suggested to contribute to the rapid recurrence of equine squamous gastric disease (ESGD) in horses after discontinuation of omeprazole. HYPOTHESIS/OBJECTIVES: To evaluate changes in serum gastrin and chromogranin A (CgA) concentrations in response to medium-term (57-day) omeprazole treatment and after omeprazole discontinuation. ANIMALS: Fourteen mature Thoroughbred racehorses in simulated race training. METHODS: Horses received 2.28 g of oral omeprazole PO q24h for 57 days within a 61-day period, excluding a withholding period applied mid-protocol during which treatment was stopped as part of a concurrent study. Serum samples were collected on day 0 before omeprazole treatment, on day 1 of each week of the treatment period, and for an additional 5 weeks after discontinuation of treatment. Serum gastrin and CgA concentrations were analyzed using radioimmunoassay (RIA) and ELISA, respectively. RESULTS: Median serum gastrin concentrations increased 2.5-fold from baseline to day 7 (P < .001) but did not increase further during the omeprazole treatment period. Median serum gastrin concentrations returned to baseline within 2 to 4 days after administration of the last dose of omeprazole. No effect of treatment or discontinuation was seen in serum CgA concentrations. CONCLUSIONS AND CLINICAL IMPORTANCE: Serum gastrin concentrations increased in response to omeprazole treatment but returned to baseline within 2 to 4 days after the last dose of omeprazole. No effect of treatment or discontinuation was seen in serum CgA concentrations. Our results do not support the use of tapering protocols in horses.


Subject(s)
Anti-Ulcer Agents , Omeprazole , Horses , Animals , Omeprazole/therapeutic use , Omeprazole/pharmacology , Chromogranin A , Anti-Ulcer Agents/therapeutic use , Anti-Ulcer Agents/pharmacology , Gastrins , Stomach
18.
Int Immunopharmacol ; 120: 110335, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37201406

ABSTRACT

This study aimed to elucidate the gastro-protective effect of fucoidan against ethanol-induced gastric ulcer mediated via NLRP3-induced pyroptosis as an underlying mechanism, not yet assessed in prior research. Forty-eight male Albino mice were divided into six groups: Group I (normal control), group II (Ulcer/ethanol control), group III (Omeprazole + ethanol), group IV (fucoidan 25 mg + ethanol), group V (fucoidan 50 mg + ethanol) and group VI (fucoidan only). Fucoidan was administered orally for seven consecutive days followed by ulcer induction by a single oral dose of ethanol. Using colorimetric analysis, ELISA, qRT-PCR, histological assessment, and immunohistochemical studies, the results revealed that ethanol-induced ulcer exhibited an ulcer score of 42.5 ± 5.1 and a significant increase (p < 0.05) in malondialdehyde (MDA), nuclear factor kappa B (NF-κB), and interleukin 6 (IL-6) with a significant decrease in the gastro-protective mediators, prostaglandin E2 (PGE2), superoxide dismutase (SOD) and glutathione (GSH), accompanied with an increase in NLRP3, interleukin 1ß (IL-1ß), interleukin 18 (IL-18), caspase 1, caspase 11, gasdermin D, and toll-like receptor 4 (TLR4), compared with the normal control. Pre-treatment with fucoidan showed a comparable result with omeprazole. Additionally, pre-treatments elevated the levels of the gastro-protective mediators and lessened oxidative stress, relative to the positive control findings. Conclusively, fucoidan has a promising gastro-protective role by inhibiting inflammation and pyroptosis.


Subject(s)
Stomach Ulcer , Mice , Animals , Male , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Stomach Ulcer/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ulcer/metabolism , Pyroptosis , Gastric Mucosa , Oxidative Stress , Inflammation/metabolism , Glutathione/metabolism , Omeprazole/therapeutic use , Omeprazole/pharmacology , Ethanol/metabolism , NF-kappa B/metabolism
19.
Cell Mol Biol (Noisy-le-grand) ; 69(1): 48-53, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-37213156

ABSTRACT

Gastric ulcer is a chronic condition that occurs when the mucosa of the stomach is broken. There is a physiological equilibrium between aggressive factors and mucosal defense. The purpose of this research was to compare the prevention level and efficiency of herbal medicinal plants (Punica granatum) to the omeprazole drug. Many groups were prepared from Albino male rats, the first control group (inoculate with H. pylori and fed with standard pellet), the Second group, rats inoculated by H. pylori and prevented with Punica granatum aqueous extracts (PGAE) in two dosages (250mg/kg, 500mg/kg), and last group inoculated by H. pylori and prevented with standard drug omeprazole at the dose (20mg/kg). The results showed that the Ulcer Inhibition % of Punica granatum with a high dose of 500mg/kg and a low dose of 250mg/kg was 84.60±5.48 and 42.87±7.14, respectively. While in the omeprazole treatment group, Ulcer Inhibition % was 24.50±6.35 and this Ulcer Inhibition %  in the Punica granatum treatment groups was significant compared to the omeprazole treatment group and the control group (P=0.0001). PGAE displayed a significant lessening in stomach index and infectious cell proliferation with much cell damage. Although the result of the current study improves, a high dosage of aqueous extracts of plants has more effectiveness than a low dosage of aqueous extracts plants.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Pomegranate , Stomach Ulcer , Rats , Animals , Stomach Ulcer/drug therapy , Stomach Ulcer/prevention & control , Ulcer/drug therapy , Omeprazole/pharmacology , Omeprazole/therapeutic use , Helicobacter Infections/drug therapy , Amoxicillin/therapeutic use
20.
Antimicrob Agents Chemother ; 67(6): e0022123, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37223895

ABSTRACT

Clostridioides difficile infection (CDI) is associated with high recurrence rates that have substantial effects on patients' quality of life. To investigate the risk factors and potential mechanisms contributing to recurrent CDI (rCDI), a total of 243 cases were enrolled in this study. The history of omeprazole (OME) medication and ST81 strain infection were considered the two independent risks with the highest odds ratios in rCDI. In the presence of OME, we detected concentration-dependent increases in the MIC values of fluoroquinolone antibiotics against ST81 strains. Mechanically, OME facilitated ST81 strain sporulation and spore germination by blocking the pathway of purine metabolism and also promoted an increase in cell motility and toxin production by turning the flagellar switch to the ON state. In conclusion, OME affects several biological processes during C difficile growth, which have fundamental impacts on the development of rCDI caused by ST81 strains. Programmed OME administration and stringent surveillance of the emerging ST81 genotype are matters of considerable urgency and significance in rCDI prevention.


Subject(s)
Clostridioides difficile , Clostridium Infections , Humans , Omeprazole/pharmacology , Omeprazole/therapeutic use , Quality of Life , Clostridioides difficile/genetics , Clostridium Infections/prevention & control , Fluoroquinolones/pharmacology , Recurrence , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...