Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Toxicol Sci ; 48(7): 375-385, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37394651

RESUMEN

Long-term use of proton pump inhibitors (PPIs) is known to clinically induce hypomagnesemia, increasing the risk toward QT-interval prolongation and lethal ventricular arrhythmias, whereas PPIs can directly modulate cardiac ionic currents in the in vitro experiments. In order to fill the gap between those information, we assessed acute cardiohemodynamic and electrophysiological effects of sub- to supra-therapeutic doses (0.05, 0.5 and 5 mg/kg/10 min) of typical PPIs omeprazole, lansoprazole and rabeprazole, using halothane-anesthetized dogs (n = 6 for each drug). The low and middle doses of omeprazole and lansoprazole increased or tended to increase the heart rate, cardiac output and ventricular contraction, whereas the high dose plateaued and decreased them. Meanwhile, the low and middle doses of omeprazole and lansoprazole decreased the total peripheral vascular resistance, whereas the high dose plateaued and increased it. Rabeprazole decreased the mean blood pressure in a dose-related manner; moreover, its high dose decreased the heart rate and tended to reduce the ventricular contractility. On the other hand, omeprazole prolonged the QRS width. Omeprazole and lansoprazole tended to prolong the QT interval and QTcV, and rabeprazole mildly but significantly prolonged them in a dose-related manner. High dose of each PPI prolonged the ventricular effective refractory period. Omeprazole shortened the terminal repolarization period, whereas lansoprazole and rabeprazole hardly altered it. In effects, PPIs can exert multifarious cardiohemodynamic and electrophysiological actions in vivo, including mild QT-interval prolongation; thus, PPIs should be given with caution to patients with reduced ventricular repolarization reserve.


Asunto(s)
Halotano , Inhibidores de la Bomba de Protones , Perros , Animales , Inhibidores de la Bomba de Protones/toxicidad , Rabeprazol , Omeprazol/toxicidad , Lansoprazol/toxicidad
2.
Pharmacol Rep ; 73(2): 551-562, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33476036

RESUMEN

BACKGROUND: Omeprazole (OME), a most frequently used proton pump inhibitor in gastric acidosis, is evident to show many adverse effects, including genetic instability. This study evaluated toxicogenic effects of OME in Mus musculus. METHODS: For this study, 40 male Swiss mice were divided into 8 groups (n = 5) and treated with OME at doses of 10, 20, and 40 mg/kg and/or treated with the antioxidants retinol palmitate (100 IU/kg) and ascorbic acid (2.0 µM/kg). Cyclophosphamide 50 mg/kg, (cytotoxic agent) and the vehicle were served as positive and negative control group, respectively. After 14 days of treatment, the stomach cells along with the bone marrow and peripheral blood lymphocytes were collected and submitted to the comet assay (alkaline version) and micronucleus test. Additionally, hematological and biochemical parameters of the animals were also determined inspect of vehicle group. RESULTS: The results suggest that OME at all doses induced genotoxicity and mutagenicity in the treated cells. However, in association with the antioxidants, these effects were modulated and/or inhibited along with a DNA repair capacity. CONCLUSIONS: Taken together, antioxidants (such as retinol palmitate and ascorbic acid) may be one of the best options to counteract OME-induced cytogenetic instability.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Diterpenos/farmacología , Omeprazol/toxicidad , Ésteres de Retinilo/farmacología , Animales , Antineoplásicos/farmacología , Ensayo Cometa , Ciclofosfamida/toxicidad , Reparación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Mutagénesis/efectos de los fármacos , Omeprazol/administración & dosificación , Inhibidores de la Bomba de Protones/administración & dosificación , Inhibidores de la Bomba de Protones/toxicidad
3.
Life Sci ; 265: 118818, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33275985

RESUMEN

AIM: Proton pump inhibitors (PPIs) are widely used drugs recently linked to chronic kidney disease. However, the invloved mechanisms remained elusive. Since defective autophagy is identified as a new culprit in the pathogenesis of diabetic nephropathy (DN), we aimed to trace the link of autophagy blockade by PPIs to the progression of DN with and without the standard therapy of metformin and enalapril. MAIN METHODS: Male CD1 albino mice (20-25 g) were randomly assigned to normal control or diabetic mice. Diabetes was induced by intraperitoneal streptozotocin (35 mg/kg) injection combined with high fat diet. DN mice were randomized to receive vehicle, lansoprazole (5 mg/kg), metformin (200 mg/kg), lansoprazole + metformin, metformin + enalapril (0.5 mg/kg) or the three drugs together, orally daily for four weeks. At the study end, albuminuria, fasting plasma glucose, HbA1c, renal functions and malondialdehyde were assessed. Renal tissues were examined microscopically, and autophagic changes were evaluated by immunohistochemical detection of LC3-II and p62. KEY FINDINGS: Consistent with autophagic blockade, lansoprazole increased both LC3II and p62 in the glomerular and tubular cells. This was associated with impaired creatinine clearance and renal functions, enhanced albuminuria, oxidative stress and augmented DN histopathological changes. Opposite effects on autophagy markers were observed by single or combined treatment of metformin with enalapril; which also ameliorated glycemic control and signs of DN. This improvement was mitigated by combination with lansoprazole. SIGNIFICANCE: Autophagy blockade by lansoprazole augmented diabetic nephropathy and opposed the reno-protective effects of metformin and enalapril. The use of PPIs in diabetes should be considered with great caution.


Asunto(s)
Autofagia/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/fisiopatología , Lansoprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Albuminuria/etiología , Animales , Nefropatías Diabéticas/prevención & control , Dieta Alta en Grasa , Enalapril/farmacología , Lansoprazol/administración & dosificación , Masculino , Metformina/farmacología , Ratones , Estrés Oxidativo/efectos de los fármacos , Inhibidores de la Bomba de Protones/administración & dosificación , Estreptozocina
4.
Toxicol Lett ; 333: 97-104, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32763312

RESUMEN

Proton pump inhibitors (PPIs) have wide pleiotropic action in addition to their therapeutic potential in gastroesophageal reflux diseases. Conversely, recent reports revealed a significant incidence of toxic events of PPIs including nephritis, osteoporosis, and cardiac damage. Thus, the study was designed to reconcile the deceptive contraindications. The present investigation targeted to reveal the toxic impact of sub-acute and sub-chronic administration of pantoprazole (PPZ) with different concentrations (low dose 4 mg/kg, medium-dose 8 mg/kg and high dose 16 mg/kg once a day) on normal vascular endothelium and renal tissue of rats. Vascular endothelial dysfunction (VED) was estimated by the contractility of an isolated aortic ring, nitrite/nitrate concentration, oxidative stress, and integrity of the endothelium layer. Moreover, the renal abnormalities were further confirmed by an increased level of serum creatinine, blood urea nitrogen (BUN), the incidence of microproteinuria, and structural alteration. Sub-acute administration of PPZ treatment did not produce any toxicological impact on endothelium and renal tissue. Whereas, sub-chronic administration of PPZ treatment causes moderate VED and renal dysfunction in a dose-dependent manner. Sub-chronic treatment of PPZ also influences the mitigation of NO and elevation of oxidative stress. Collecting all the evidence, it concludes that decreased nitric oxide availability and increased levels of oxidative stress may be a possible underlying mechanism of causing VED and renal abnormalities from high-dose PPZ treatment.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Riñón/efectos de los fármacos , Pantoprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Administración Oral , Animales , Aorta Torácica/inmunología , Aorta Torácica/metabolismo , Aorta Torácica/patología , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Nitratos/metabolismo , Nitritos/metabolismo , Ratas , Ratas Wistar , Vasodilatación/efectos de los fármacos
5.
Cardiovasc Toxicol ; 20(6): 531-538, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32500385

RESUMEN

Recent results from data mining analyses and reports of adverse drug events suggest a QT-prolonging drug-drug interaction resulting from the combination of distinct proton pump inhibitors and cephalosporins. Therefore, this study aimed at investigating the effect of the suspected QT-prolonging combinations of lansoprazole + ceftriaxone and esomeprazole + cefazolin, respectively. 26 hearts of New Zealand White rabbits were retrogradely perfused and paced at different cycle lengths. After generating baseline data, the hearts were assigned to two groups: In group 1, hearts were treated with 5 µM lansoprazole. Thereafter, 200 µM ceftriaxone was infused additionally. Group 2 was perfused with 10 µM esomeprazole followed by 250 µM cefazolin. In group 1, lansoprazole did not significantly alter QT intervals and APD90. Additional treatment with ceftriaxone significantly shortened QT interval, APD90 and slightly reduced dispersion of repolarization compared to sole lansoprazole infusion. In group 2, esomeprazole led to a significant shortening of the QT interval without altering APD90 or dispersion. Additional treatment with the antibiotic cefazolin further shortened QT interval, APD90 and reduced the dispersion of repolarization. Incidence of ventricular arrhythmias was not significantly altered in both groups. This is the first experimental whole-heart study that investigated the impact of a concomitant treatment with proton pump inhibitors and cephalosporins. In contrast to previous reports, the combination of both agents did not cause QT prolongation but instead shortened QT interval and action potential duration. As a consequence, no triggered activity occurred in the presence of a stable dispersion of repolarization.


Asunto(s)
Antibacterianos/toxicidad , Arritmias Cardíacas/inducido químicamente , Ceftriaxona/toxicidad , Esomeprazol/toxicidad , Sistema de Conducción Cardíaco/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Lansoprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Potenciales de Acción , Animales , Arritmias Cardíacas/fisiopatología , Estimulación Cardíaca Artificial , Interacciones Farmacológicas , Femenino , Sistema de Conducción Cardíaco/fisiopatología , Preparación de Corazón Aislado , Conejos , Periodo Refractario Electrofisiológico , Medición de Riesgo , Factores de Tiempo
6.
Toxicol Lett ; 318: 86-91, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31669099

RESUMEN

Proton pump inhibitors (PPIs) have been used worldwide to treat gastrointestinal disorders. A recent study showed that long-term use of PPIs caused iron deficiency; however, it is unclear whether PPIs affect iron metabolism directly. We investigated the effect of PPIs on the peptide hepcidin, an important iron regulatory hormone. First, we used the FDA Adverse Event Reporting System database and analyzed the influence of PPIs. We found that PPIs, as well as H2 blockers, increased the odds ratio of iron-deficient anemia. Next, HepG2 cells were used to examine the action of PPIs and H2 blockers on hepcidin. PPIs augmented hepcidin expression, while H2 blockers did not. In fact, the PPI omeprazole increased hepcidin secretion, and omeprazole-induced hepcidin upregulation was inhibited by gene silencing or the pharmacological inhibition of the aryl hydrocarbon receptor. In mouse experiments, omeprazole also increased hepatic hepcidin mRNA expression and blood hepcidin levels. In mice treated with omeprazole, protein levels of duodenal and splenic ferroportin decreased. Taken together, PPIs directly affect iron metabolism by suppressing iron absorption through the inhibition of duodenal ferroportin via hepcidin upregulation. These findings provide a new insight into the molecular mechanism of PPI-induced iron deficiency.


Asunto(s)
Anemia Ferropénica/inducido químicamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Duodeno/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepcidinas/metabolismo , Absorción Intestinal/efectos de los fármacos , Hierro/sangre , Inhibidores de la Bomba de Protones/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Anemia Ferropénica/sangre , Anemia Ferropénica/fisiopatología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Transporte de Catión/metabolismo , Duodeno/metabolismo , Duodeno/fisiopatología , Células Hep G2 , Hepatocitos/metabolismo , Antagonistas de los Receptores H2 de la Histamina/toxicidad , Humanos , Deficiencias de Hierro , Masculino , Ratones Endogámicos C57BL , Receptores de Hidrocarburo de Aril/genética
7.
Andrologia ; 51(6): e13260, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30854683

RESUMEN

The effect of omeprazole, a commonly used drug belongs to proton--pump inhibitor class, on human sperm function is still undetermined. Here, we hypothesised that addition of omeprazole to the ejaculated human semen may affect sperm parameters, and hence sperm function. Therefore, we assessed the in vitro effect of omeprazole on human sperm motility, viability and DNA integrity. Sixty-six normozoospermic semen samples were collected randomly from men who attended the andrology laboratory at King Abdullah University Hospital. Sperm motility, viability and DNA breaks were assessed in the presence (1-hr incubation at 37°C) of omeprazole at 5, 10, 20 and 50 µM compared to control (0 µM). None of the examined sperm parameters, at any tested omeprazole concentration, showed significant difference (p > 0.05) compared with the control. In conclusion, omeprazole at 5, 10, 20 and 50 µM does not alter human sperm motility, viability or DNA integrity in vitro.


Asunto(s)
Omeprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Espermatozoides/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN/efectos de los fármacos , Humanos , Masculino , Motilidad Espermática/efectos de los fármacos , Pruebas de Toxicidad
8.
Cancer Lett ; 449: 252-262, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30790678

RESUMEN

Prostate cancer (PCa) is one of the most common cancer in men. Although hormone-sensitive PCa responds to androgen-deprivation, there are no effective therapies for castration-resistant PCa. It has been recently suggested that proton pump inhibitors (PPIs) may increase the risk of certain cancers; however, association with PCa remains elusive. Here, we evaluated the tumorigenic activities of PPIs in vitro, in PCa cell lines and epithelial cells from benign prostatic hyperplasia (BPH) and in vivo, in PCa mice xenografts. PPIs increased survival and proliferation, and inhibited apoptosis in LNCaP cells. These effects were attenuated or absent in androgen-insensitive DU-145 and PC3 cells, respectively. Specifically, omeprazole (OME) promoted cell cycle progression, increased c-Myc expression, ErbB2 activity and PSA secretion. Furthermore, OME induced the phosphorylation of MAPK-ERK1/2, PI3K/Akt and GSK-3ß, and blunted the expression and activity of cellular prostatic acid phosphatase. OME also increased survival, proliferation and PSA levels in BPH cells. In vivo, OME promoted tumor growth in mice bearing LNCaP xenografts. Our results indicate that PPIs display tumorigenic activities in PCa cells, suggesting that their long-term administration in patients should be carefully monitored.


Asunto(s)
Fosfatasa Ácida/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Hormono-Dependientes/enzimología , Omeprazol/toxicidad , Fosfatidilinositol 3-Quinasa/metabolismo , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Inhibidores de la Bomba de Protones/toxicidad , Receptor ErbB-2/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Apoptosis/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Hormono-Dependientes/patología , Células PC-3 , Fosforilación , Neoplasias de la Próstata/patología , Transducción de Señal
9.
Front Med ; 13(2): 277-284, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29845582

RESUMEN

The development of new proton-pump inhibitors (PPIs) with less adverse effects by lowering the pKa values of nitrogen atoms in pyrimidine rings has been previously suggested by our group. In this work, we proposed that new PPIs should have the following features: (1) number of ring II = number of ring I + 1; (2) preferably five, six, or seven-membered heteroatomic ring for stability; and (3) 1 < pKa1 < 4. Six molecular scaffolds based on the aforementioned criteria were constructed, and R groups were extracted from compounds in extensive data sources. A virtual molecule dataset was established, and the pKa values of specific atoms on the molecules in the dataset were calculated to select the molecules with required pKa values. Drug-likeness screening was further conducted to obtain the candidates that significantly reduced the adverse effects of long-term PPI use. This study provided insights and tools for designing targeted molecules in silico that are suitable for practical applications.


Asunto(s)
Simulación por Computador , Diseño de Fármacos , Inhibidores de la Bomba de Protones/toxicidad , Humanos , Fenómenos Toxicológicos
10.
J Appl Toxicol ; 38(7): 987-995, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29659034

RESUMEN

Vonoprazan is a new potassium-competitive acid blocker to treat acid-related diseases. However, its safety during pregnancy is unclear. The aim of the study was to investigate the potential reproductive toxicity on the embryo-fetal development of vonoprazan. Vonoprazan acetate was administered by intravenous injection to pregnant rats (0, 2, 6 and 20 mg kg-1 day-1 ) and rabbits (0, 1.2, 3.6 and 12 mg kg-1 day-1 ) during the organogenetic period (gestation day 6-15 [rats] and 6-18 [rabbits]). Maternal reproductive endpoints were evaluated, together with effects on fetal growth and morphological development. In rats, no treatment-related effects were found in the highest dose group (20 mg kg-1 ) and the maternal plasma exposure was ≥50-fold the expected clinical human exposure. However, in rabbits, dose-related clinical signs (soft or liquid feces) occurred in the 12 mg kg-1 group, which was regarded as a maternal toxicity. Besides, decreased maternal weight gain also was considered as a minimal maternal toxicity. At 12 mg kg-1 , delayed fetal ossification was found as evidence of embryo-fetal growth retardation, which was related to decreased fetal and placental weights. There was no maternal and developmental toxicity in the 1.2 and 3.6 mg kg-1 groups. Thus, the no-observed-adverse-effect levels of vonoprazan acetate in rabbits are considered 3.6 mg kg-1 day-1 , which produced plasma exposure that was about 18-fold human clinical exposure.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Feto/efectos de los fármacos , Inhibidores de la Bomba de Protones/toxicidad , Pirroles/toxicidad , Sulfonamidas/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Femenino , Desarrollo Fetal/efectos de los fármacos , Retardo del Crecimiento Fetal/inducido químicamente , Peso Fetal/efectos de los fármacos , Feto/patología , Edad Gestacional , Inyecciones Intravenosas , Exposición Materna , Nivel sin Efectos Adversos Observados , Placenta/efectos de los fármacos , Placenta/patología , Embarazo , Conejos , Ratas Sprague-Dawley , Medición de Riesgo , Especificidad de la Especie
11.
Cell Physiol Biochem ; 43(3): 1037-1051, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28968600

RESUMEN

BACKGROUND/AIMS: Glucose-stimulated insulin secretion (GSIS) of pancreatic ß-cells involves glucose uptake and metabolism, closure of KATP channels and depolarization of the cell membrane potential (Vmem), activation of voltage-activated Ca2+ currents (ICav) and influx of Ca2+, which eventually triggers hormone exocytosis. Beside this classical pathway, KATP-independent mechanisms such as changes in intracellular pH (pHi) or cell volume, which also affect ß-cell viability, can elicit or modify insulin release. In ß-cells the regulation of pHi is mainly accomplished by Na+/H+ exchangers (NHEs). To investigate if other proton extrusion mechanisms than NHEs are involved in pH regulation, we tested for the presence of the non-gastric H+/K+ ATPase in rat insulinoma cells and assessed effects of the H+/K+ ATPase inhibitor SCH-28080 on insulin secretion, cell viability and apoptosis. METHODS: In INS-1E cell cultures, H+/K+ ATPase gene and protein expression was analyzed by reverse transcription PCR and Western blotting. Intracellular pH (pHi) recovery after acute acidic load was measured by NH4Cl prepulsing using BCECF. Insulin secretion was determined by ELISA from the cell culture supernatant. Vmem, K+ and Ca2+ currents were recorded using patch clamp. Overall cell responses were determined using resazurin (viability) and cytotoxicity assays. The mean cell volume (MCV), cell granularity (side-scatter; SSC), phosphatidylserine (PS) exposure, cell membrane integrity, caspase activity and the mitochondrial membrane potential (ΔΨm) were measured by flow cytometry. RESULTS: We found that the α-subunit of the non-gastric H+/K+ ATPase (HKα2) is expressed on mRNA and protein level. However, compared to rat colon tissue, in INS-1E cells mRNA abundance was very low. In NH4Cl prepulsing experiments no K+-dependent pHi recovery was observed under Na+-free extracellular conditions. Nonetheless within 1 h, 20 µM SCH-28080 inhibited GSIS by ∼50%, while basal release was unaffected. The L-type ICav blocker nifedipine caused a full inhibition of GSIS at 10 and 20 µM. At 20 µM, SCH-28080 inhibited ICav comparable to 20 µM nifedipine and in addition augmented IKATP recorded at -60 mV and hyperpolarized Vmem by ∼15 mV. Cell viability 2 and 24 h post treatment with SCH-28080 was dose-dependently inhibited with IC50 values of 22.9 µM and 15.3 µM, respectively. At 20 µM the percentages of Annexin-V+, caspase+ and propidium iodide+ cells were significantly increased after 24 and 48 h. Concurrently, the MCV was significantly decreased (apoptotic volume decrease, AVD) and the SSC signal was increased. At concentrations >40-50 µM, SCH-28080 became progressively cytotoxic causing a steep increase in necrotic cells already 2 h post treatment and a breakdown of ΔΨm within 4 h under 50 and 100 µM while 10 and 20 µM had no effect on ΔΨm within 24 h. CONCLUSION: We demonstrate expression of HKα2 in rat INS-1E cells. However, the pump is apparently non-functional under the given conditions. Nonetheless the H+/K+ ATPase blocker SCH-28080 inhibits insulin secretion and induces cell death. Importantly, we show that SCH-28080 inhibits ICav - and activates KATP channels identifying them as novel "off-targets" of the inhibitor, causing hyperpolarization of Vmem and inhibition of insulin secretion.


Asunto(s)
Apoptosis/efectos de los fármacos , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Imidazoles/toxicidad , Insulina/análisis , Inhibidores de la Bomba de Protones/toxicidad , Animales , Calcio/metabolismo , Línea Celular Tumoral , Tamaño de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colon/metabolismo , Ensayo de Inmunoadsorción Enzimática , Glucosa/farmacología , ATPasa Intercambiadora de Hidrógeno-Potásio/química , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Secreción de Insulina , Insulinoma/metabolismo , Insulinoma/patología , Canales KATP/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Nifedipino/toxicidad , Técnicas de Placa-Clamp , Fosfatidilserinas/farmacología , ARN Mensajero/metabolismo , Ratas
12.
Aquat Toxicol ; 191: 62-72, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28800409

RESUMEN

Omeprazole (OMP) is one of the most commonly used drugs for the treatment of gastro-intestinal disorders. Although it is daily consumed in high quantities and commonly detected in waters worldwide, relatively little is known about its ecotoxicity. The aim of this study was to evaluate the potential acute toxicity of increasing concentrations of OMP on the marine microalga Tetraselmis suecica analysing several cytotoxicity biomarkers by flow cytometry after 24h of exposure. Results showed that OMP caused a decrease in growth and autofluorescence, an increase in cellular volume and intracellular complexity, hyperpolarization of cytoplasmic and mitochondrial membranes and intracellular acidification. In addition, large amounts of reactive oxygen species (ROS) were generated which resulted in a decrease in the percentage of the viable population. However, the viable population showed an increase in their metabolic activity as an early response to overcome the stress. In conclusion, OMP may affect proton pumps in non-target organisms such as microalgae; it disturbed pH homeostasis and provoked an early accumulation of ROS that resulted in a rapid cell death in cells exposed to the highest concentration assayed.


Asunto(s)
Chlorophyta/efectos de los fármacos , Microalgas/efectos de los fármacos , Omeprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Contaminantes Químicos del Agua/toxicidad , Clorofila/metabolismo , Clorofila A , Chlorophyta/crecimiento & desarrollo , Chlorophyta/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Microalgas/crecimiento & desarrollo , Microalgas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
13.
J Mass Spectrom ; 52(7): 459-471, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28544042

RESUMEN

Regulatory agencies from all over the world have set up stringent guidelines with regard to drug degradation products due to their toxic effects or carcinogenicity. Lansoprazole, a proton-pump inhibitor, was subjected to forced degradation studies as per ICH guidelines Q1A (R2). The drug was found to degrade under acidic, basic, neutral hydrolysis and oxidative stress conditions, whereas it was found to be stable under thermal and photolytic conditions. The chromatographic separation of the drug and its degradation products were achieved on a Hiber Purospher, C18 (250 × 4.6 mm, 5 µ) column using 10 mM ammonium acetate and acetonitrile as a mobile phase in a gradient elution mode at a flow rate of 1.0 ml/min. The eight degradation products (DP1-8) were identified and characterized by UPLC/ESI/HRMS with in-source CID experiments combined with accurate mass measurements. DP-1, DP-2 and DP-3 were formed in acidic, DP-4 in basic, DP-5 in neutral and DP-1, DP-6, DP-7 and DP-8 were in oxidation stress condition Among eight degradation products, five were hitherto unknown degradation products. In addition, one of the major degradation products, DP-2, was isolated by using semi preparative HPLC and other two, DP-6 and DP-7 were synthesized. The cytotoxic effect of these degradation products (DP-2, DP-6 and DP-7) were tested on normal human cells such as HEK 293 (embryonic kidney cells) and RWPE-1(normal prostate epithelial cells) by MTT assay. From the results of cytotoxicity, it was found that lansoprazole as well as its degradation products (DP-2, DP-6 and DP-7) were nontoxic up to 50-µM concentrations, and the latter showed slightly higher cytotoxicity when compared with that of lansoprazole. DNA binding studies using spectroscopic techniques indicate that DP-2, DP-6 and DP-7 molecules interact with ctDNA and may bind to its surface. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Lansoprazol/química , Inhibidores de la Bomba de Protones/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión/métodos , Calor , Humanos , Hidrólisis , Lansoprazol/toxicidad , Espectroscopía de Resonancia Magnética/métodos , Oxidación-Reducción , Fotólisis , Inhibidores de la Bomba de Protones/toxicidad , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masas en Tándem/métodos
14.
Dermatitis ; 26(6): 287-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26551609

RESUMEN

Few published reports have described occupational contact dermatitis from proton pump inhibitor (PPI) exposure in the literature. We present an additional case of a 58-year-old male pharmaceutical worker with an occupational airborne allergic contact dermatitis to PPIs confirmed by patch testing. This is a novel report of workplace exposure to dexlansoprazole and esomeprazole PPIs with resultant clinical contact allergy and relevant positive patch test results to these 2 agents. A literature review of all previously reported cases of occupational contact dermatitis to PPI is summarized. The case also emphasizes the importance of even minute exposures when considering workplace accommodation.


Asunto(s)
Contaminantes Ocupacionales del Aire/toxicidad , Dermatitis por Contacto/etiología , Dermatitis Profesional/etiología , Dermatosis Facial/inducido químicamente , Inhibidores de la Bomba de Protones/toxicidad , Párpados , Humanos , Masculino , Persona de Mediana Edad , Cuello
15.
Toxicol Lett ; 235(1): 28-36, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25797602

RESUMEN

The role of the aryl hydrocarbon receptor (AhR) in hemostasis has recently gained increased attention. Here, we demonstrate, by qRT-PCR and western blot, that human platelets express both AhR mRNA and AhR protein. AhR protein levels increase in a dose dependent manner when incubated with either 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or omeprazole. Treatment of platelets with puromycin blocks increased AhR protein synthesis in the presence of AhR activators. Additionally, treatment of platelets with either activator results in phosphorylation of p38MAPK and cPLA2, two key signaling molecules in platelet activation pathways. Using the AhR competitive inhibitors alpha naphthoflavone and CH-223191, we show that phosphorylation of p38MAPK is AhR dependent. Further, inhibition of p38MAPK blocks downstream cPLA2 phosphorylation induced by TCDD or omeprazole. Treatment with AhR activators results in platelet priming, as demonstrated by increased platelet aggregation, which is inhibited by AhR antagonists. Our data support a model of the platelet AhR non-genomic pathway in which treatment with AhR activators results in increased expression of the AhR, phosphorylation of p38MAPK and cPLA2, leading to platelet priming in response to agonist.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Plaquetas/efectos de los fármacos , Omeprazol/toxicidad , Activación Plaquetaria/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Plaquetas/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Fosfolipasas A2 Grupo IV/metabolismo , Humanos , Ligandos , Fragmentos de Péptidos/farmacología , Fosforilación , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Puromicina/farmacología , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Cardiovasc Ther ; 33(2): 27-34, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25677801

RESUMEN

BACKGROUND: The safety of all proton pump inhibitors (PPIs) in patients with intrinsic cardiac disease has not been well studied. In the present study, the effect of PPI pantoprazole on ventricular arrhythmias induced by either ischemia or ischemia-reperfusion (I/R) was studied. METHODS: The left main coronary artery (LAD) was ligated for 30 or 10 min followed by a 30-min reperfusion in anesthetized rats. Rats were pretreated with pantoprazole (1.3 mg/kg) or vehicle by gastric gavage (daily for 3 weeks) before ligation. Serum electrolytes levels were measured by the end of the third week before coronary ligation. Lactate dehydrogenase (LDH) activity and nitric oxide (NO) concentrations were measured at the end of the ischemia and IR injury. RESULTS: Pantoprazole caused significant hyperkalemia by the end of third week compared with vehicle-treated rats. After LAD ligation (30 min), ventricular premature contractions (VPC), ventricular tachycardia (VT) and ventricular fibrillation (VF) were recorded in rats of the vehicle ischemia group. Pantoprazole pretreatment aggravate these arrhythmias and increased mortality. A 10-min period of ischemia followed by a 30-min reperfusion induced high incidence of VT (100%) and VF (80%) in the vehicle-treated group. The group of rats administered pantoprazole had significantly lower incidence and durations of VT and VF together with reduction of mortality rate. Pantoprazole significantly reduced serum LDH activity and NO release from myocardial tissue after both ischemia and I/R injury. CONCLUSION: Pantoprazole aggravated ischemia-induced arrhythmias but had a significant antiarrhythmic effect on I/R-induced ventricular arrhythmias.


Asunto(s)
2-Piridinilmetilsulfinilbencimidazoles/farmacología , Antiarrítmicos/farmacología , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/prevención & control , Sistema de Conducción Cardíaco/efectos de los fármacos , Isquemia Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/complicaciones , Inhibidores de la Bomba de Protones/farmacología , 2-Piridinilmetilsulfinilbencimidazoles/toxicidad , Animales , Antiarrítmicos/toxicidad , Arritmias Cardíacas/sangre , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Sistema de Conducción Cardíaco/fisiopatología , Hiperpotasemia/inducido químicamente , L-Lactato Deshidrogenasa/sangre , Masculino , Isquemia Miocárdica/sangre , Isquemia Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/fisiopatología , Óxido Nítrico/sangre , Pantoprazol , Inhibidores de la Bomba de Protones/toxicidad , Ratas Wistar , Taquicardia Ventricular/inducido químicamente , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/prevención & control , Factores de Tiempo , Fibrilación Ventricular/inducido químicamente , Fibrilación Ventricular/fisiopatología , Fibrilación Ventricular/prevención & control , Complejos Prematuros Ventriculares/inducido químicamente , Complejos Prematuros Ventriculares/fisiopatología , Complejos Prematuros Ventriculares/prevención & control
17.
Am J Physiol Gastrointest Liver Physiol ; 308(9): G785-93, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25721304

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) are widely used to treat a number of conditions, and proton pump inhibitors (PPIs) are often used to prevent NSAID-induced gastric mucosal damage; however, the effects of NSAIDs on intestinal motility are poorly understood. The purpose of the present study is to determine the effects of a prototypical NSAID, indomethacin, either alone or in conjunction with the PPI omeprazole, on intestinal motility. Rats were randomly divided into four groups treated with vehicle, omeprazole, indomethacin, or a combination of indomethacin and omeprazole. Intestinal motility and transit were measured along with inflammatory mediators in the intestinal smooth muscle, markers of mucosal damage, and bacterial counts in the intestinal wall. Indomethacin, but not omeprazole, caused mucosal injury indicated by lower gut bleeding; however, both omeprazole and indomethacin suppressed contractile activity and frequency in the distal part of the small intestine. Cotreatment with omeprazole did not reduce indomethacin-induced intestinal bleeding. Furthermore, although indomethacin caused increased inflammation as indicated by increased edema development and inflammatory mediators, cotreatment with omeprazole did not reduce inflammation in the intestinal smooth muscle or prevent the increased bacterial count in the intestinal wall induced by indomethacin. We conclude that both NSAID and PPI treatment suppressed contractile activity in the distal regions of the small intestine. The suppression of intestinal contractility was associated with increased inflammation in both cases; however, indomethacin and omeprazole appear to affect intestinal motility by different mechanisms.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Motilidad Gastrointestinal/efectos de los fármacos , Íleon/efectos de los fármacos , Indometacina/toxicidad , Yeyuno/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Omeprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Animales , Biomarcadores/metabolismo , Enteritis/inducido químicamente , Enteritis/metabolismo , Enteritis/fisiopatología , Hemorragia Gastrointestinal/inducido químicamente , Hemorragia Gastrointestinal/patología , Hemorragia Gastrointestinal/fisiopatología , Íleon/metabolismo , Íleon/microbiología , Íleon/patología , Íleon/fisiopatología , Mediadores de Inflamación/metabolismo , Yeyuno/metabolismo , Yeyuno/microbiología , Yeyuno/patología , Yeyuno/fisiopatología , Masculino , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Ratas Sprague-Dawley
18.
Chem Res Toxicol ; 28(3): 419-30, 2015 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-25626140

RESUMEN

Proton pump inhibitors (PPIs) are extensively used for the treatment of gastric acid-related disorders. PPIs appear to be well tolerated and almost have no short-term side effects. However, the clinical adverse reactions of long-term PPI usage are increasingly reported in recent years. So far, there is no study that elucidates the side effect mechanisms of PPIs comprehensively and systematically. In this study, a well-defined small molecule perturbed microarray data set of 344 compounds and 1695 samples was analyzed. With this high-throughput data set, a new index (Identity, I) was designed to identify PPI-specific differentially expressed genes. Results indicated that (1) up-regulated genes, such as RETSAT, CYP1A1, CYP1A2, and UGT, enhanced vitamin A's metabolism processes in the cellular retinol metabolism pathway; and that (2) down-regulated genes, such as C1QA, C1QC, C4BPA, C4BPB, CFI, and SERPING1, enriched in the complement and coagulation cascades pathway. In addition, strong association was observed between these PPI-specific differentially expressed genes and the reported side effects of PPIs by the gene-disease association network analysis. One potential toxicity mechanism of PPIs as suggested from this systematic PPI-specific gene expression analysis is that PPIs are enriched in acidic organelles where they are activated and inhibit V-ATPases and acid hydrolases, and consequently block the pathways of antigen presentation, the synthesis and secretion of cytokines, and complement component proteins and coagulation factors. The strategies developed in this work could be extended to studies on other drugs.


Asunto(s)
Expresión Génica/efectos de los fármacos , Inhibidores de la Bomba de Protones/toxicidad , Animales , Simulación por Computador , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas Sprague-Dawley
19.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G390-6, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24699328

RESUMEN

Proton pump inhibitors reduce gastric acid secretion and are commonly utilized in the management of gastroesophageal reflux disease across all ages. Yet a decrease in lower esophageal sphincter tone has been reported in vitro in rats through an unknown mechanism; however, their effect on the gastroesophageal muscle tone early in life was never studied. Hypothesizing that proton pump inhibitors also reduce gastroesophageal muscle contraction in newborn and juvenile rats, we evaluated the in vitro effect of pantoprazole on gastric and lower esophageal sphincter muscle tissue. Electrical field stimulation and carbachol-induced force were significantly (P < 0.01) reduced in the presence of pantoprazole, whereas the drug had no effect on the neuromuscular-dependent relaxation. When administered in vivo, pantoprazole (9 mg/kg) significantly (P < 0.01) reduced gastric emptying time at both ages. To ascertain the signal transduction pathway responsible for the reduction in muscle contraction, we evaluated the tissue ROCK-2 and CPI-17 activity. Pantoprazole reduced myosin light chain phosphatase MYPT-1, but not CPI-17 phosphorylation of gastric and lower esophageal sphincter tissue, strongly suggesting that it is a ROCK-2 inhibitor. To the extent that these findings can be extrapolated to human neonates, the use of pantoprazole may impair gastric and lower sphincter muscle tone and thus paradoxically exacerbate esophageal reflux. Further studies addressing the effect of proton pump inhibitors on gastroesophageal muscle contraction are warranted to justify its therapeutic use in gastroesophageal reflux disease.


Asunto(s)
2-Piridinilmetilsulfinilbencimidazoles/toxicidad , Esfínter Esofágico Inferior/efectos de los fármacos , Vaciamiento Gástrico/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , Estómago/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Carbacol/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Esfínter Esofágico Inferior/enzimología , Esfínter Esofágico Inferior/inervación , Proteínas Musculares/metabolismo , Pantoprazol , Fosfoproteínas/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estómago/enzimología , Estómago/inervación , Factores de Tiempo , Quinasas Asociadas a rho/metabolismo
20.
J Toxicol Sci ; 37(5): 969-85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23038005

RESUMEN

Omeprazole (OPZ) and ß-naphthoflavone (BNF) are cytochrome P450 (CYP)1A inducers and have liver tumor promoting effects. In this study, we investigated the co-promoting and co-initiating effects of OPZ and BNF in rats. In Experiment 1, male rats were subjected to partial hepatectomy (PH), and given oral doses of 138 or 276 mg/kg OPZ, 0.125% or 0.25% BNF or 138 mg/kg OPZ+0.125% BNF (n = 9~12) for 6 weeks after N-diethylnitrosamine (DEN) initiation. In Experiment 2, male rats were treated with oral doses of 138 or 276 mg/kg OPZ, 0.03% or 0.06% BNF or 138 mg/kg OPZ+0.03% BNF (n = 11~12) for 9 days starting 1 week before initiating treatment. As an initiating treatment, 2-Amino-3,4-dimethylimidazo[4,5-f]quinolone (MeIQx) was orally administered 12 hr after PH. The rats were fed a basal diet for 15 days, followed by a diet containing 0.015% 2-acetylaminofluorene for the next 10 days with a single oral dose of carbon tetrachloride. In Experiment 1, the number and area of glutathione S-transferase placental form-positive foci in the OPZ+BNF group were significantly higher than the average values of the High OPZ or the High BNF group. The expression of cyclooxygenase-2 (Cox-2) and COX-2 protein in the liver significantly increased in the OPZ+BNF group. In Experiment 2, liver initiation activity was not enhanced by the co-administration of OPZ+BNF. The results of our studies suggest that the co-administration of OPZ and BNF results in synergistic effects in the liver tumor promotion probably owing to increased COX-2 expression, but no modifying effect in the liver initiation activity of MeIQx in rats.


Asunto(s)
Carcinógenos/toxicidad , Citocromo P-450 CYP1A1/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Omeprazol/toxicidad , Inhibidores de la Bomba de Protones/toxicidad , beta-naftoflavona/toxicidad , Animales , Tetracloruro de Carbono , Carcinógenos/administración & dosificación , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dietilnitrosamina , Sinergismo Farmacológico , Peroxidación de Lípido/efectos de los fármacos , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Omeprazol/administración & dosificación , Omeprazol/sangre , Inhibidores de la Bomba de Protones/administración & dosificación , Inhibidores de la Bomba de Protones/sangre , Quinoxalinas , Ratas , Ratas Endogámicas F344 , beta-naftoflavona/administración & dosificación , beta-naftoflavona/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...