Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 290
Filter
1.
Crit Rev Eukaryot Gene Expr ; 33(5): 17-27, 2023.
Article in English | MEDLINE | ID: mdl-37199311

ABSTRACT

Excess thyroid hormone secretion can cause endocrine metabolic disorders, which can lead to cardiovascular diseases, including heart enlargement, atrial fibrillation (AF), and heart failure. The present study investigated the molecular mechanisms of hyperthyroidism-induced AF. A rabbit susceptibility model of hyperthyroidism-induced AF was constructed, and metoprolol treatment was administered. Norepinephrine levels were determined using enzyme-linked immunosorbent assay; quantitative reverse transcription polymerase chain reaction and immunohistochemistry were used to detect the expression of markers for sympathetic remodeling (growth associated protein 43 and tyrosine hydroxylase in atrial myocardial tissues and stellate ganglia). Primary rabbit cardiomyocytes were cultured and identified by immunofluorescence staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling staining was used to measure cardiomyocyte apoptosis; western blot was used to detect the expression of apoptosis-related proteins, including Bax, Bcl-2, and cleaved caspase-3, as well as to measure the phosphorylation states of p38 mitogen-activated protein kinase (MAPK) pathway proteins. Metoprolol inhibited sympathetic activation and cardiomyocyte apoptosis in the rabbit model by inhibiting the p38 MAPK signaling pathway. Immunofluorescence staining results revealed that the rabbit cardiomyocytes were isolated successfully. Inhibition of p38 MAPK signaling alleviated norepinephrine-induced apoptosis in cardiomyocytes. Sympathetic activation promotes apoptosis in cardiomyocytes with hyperthyroidism-induced AF via the p38 MAPK signaling pathway. The results of the present study provide a novel theoretical basis for the potential clinical treatment of patients with hyperthyroidism and AF.


Subject(s)
Atrial Fibrillation , Hyperthyroidism , Animals , Rabbits , p38 Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Metoprolol/pharmacology , Metoprolol/metabolism , Apoptosis , Signal Transduction , Norepinephrine/pharmacology , Norepinephrine/metabolism , Hyperthyroidism/complications , Hyperthyroidism/metabolism
2.
Environ Sci Technol ; 57(11): 4471-4480, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36877486

ABSTRACT

The increasing use of chiral pharmaceuticals has led to their widespread presence in the environment. However, their toxicokinetics have rarely been reported. Therefore, the tissue-specific uptake and depuration kinetics of two pairs of pharmaceutical enantiomers, S-(-)-metoprolol versus R-(+)-metoprolol and S-(+)-venlafaxine versus R-(-)-venlafaxine, were studied in marine medaka (Oryzias melastigma) during a 28-day exposure and 14-day clearance period. The toxicokinetics of the studied pharmaceuticals, including uptake and depuration rate constants, depuration half-life (t1/2), and bioconcentration factor (BCF), were reported for the first time. The whole-fish results demonstrated a higher S- than R-venlafaxine bioaccumulation potential, whereas no significant difference was observed between S- and R-metoprolol. O-desmethyl-metoprolol (ODM) and α-hydroxy-metoprolol (AHM) were the main metoprolol metabolites identified by suspect screening, and the ratios of ODM to AHM were 3.08 and 1.35 for S- and R-metoprolol, respectively. N,O-Didesmethyl-venlafaxine (NODDV) and N-desmethyl-venlafaxine (NDV) were the main venlafaxine metabolites, and the ratios of NODDV to NDV were 1.55 and 0.73 for S- and R-venlafaxine, respectively. The highest tissue-specific BCFs of the four enantiomers were all found in the eyes, meriting in-depth investigation.


Subject(s)
Oryzias , Animals , Venlafaxine Hydrochloride , Metoprolol/metabolism , Tissue Distribution , Pharmaceutical Preparations
3.
Environ Toxicol ; 38(12): 3006-3017, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37584562

ABSTRACT

Metoprolol, a drug for hypertension and cardiovascular diseases, has become a contaminant of emerging concern because of its frequent detection in various environmental matrices globally. The dwindling in the biodiversity of useful insects owing to increasing presence of environmental chemicals is currently a great interest to the scientific community. In the current research, the toxicological impact of ecologically relevant concentrations of metoprolol at 0, 0.05, 0.1, 0.25, and 0.5 µg/L on Nauphoeta cinerea nymphs following exposure for 42 consecutive days was evaluated. The insects' behavior was analyzed with automated video-tracking software (ANY-maze, Stoelting Co, USA) while biochemical assays were done using the midgut, head and fat body. Metoprolol-exposed nymphs exhibited significant diminutions in the path efficiency, mobility time, distance traveled, body rotation, maximum speed and turn angle cum more episodes, and time of freezing. In addition, the heat maps and track plots confirmed the metoprolol-mediated wane in the exploratory and locomotor fitness of the insects. Compared with control, metoprolol exposure decreased acetylcholinesterase activity in insects head. Antioxidant enzymes activities and glutathione level were markedly decreased whereas indices of inflammation and oxidative injury to proteins and lipids were significantly increased in head, midgut and fat body of metoprolol-exposed insects. Taken together, metoprolol exposure induces neurobehavioral insufficiency and oxido-inflammatory injury in N. cinerea nymphs. These findings suggest the potential health effects of environmental contamination with metoprolol on ecologically and economically important nontarget insects.


Subject(s)
Cockroaches , Metoprolol , Animals , Metoprolol/toxicity , Metoprolol/metabolism , Acetylcholinesterase/metabolism , Oxidative Stress , Antioxidants/metabolism , Cockroaches/metabolism
4.
Chem Res Toxicol ; 35(6): 1059-1069, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35575346

ABSTRACT

As a selective ß1-receptor antagonist, metoprolol tartrate (MTA) is commonly used to treat cardiovascular diseases such as hypertension and angina pectoris. There have been cases of liver injury induced by MTA, but the mechanism of hepatotoxicity induced by MTA is not clear. The purposes of this study were to identify the reactive metabolites of MTA, to determine the pathway for the metabolic activation of MTA, and to define a possible correlation between the metabolic activation and cytotoxicity of MTA. Three oxidative metabolites (M1-M3), a glutathione (GSH) conjugate (M4), and an N-acetyl cysteine (NAC) conjugate (M5) were detected in rat liver microsomal incubations containing MTA and GSH or NAC. M4 was also detected in cultured rat primary hepatocytes and bile of rats given MTA, and M5 was detected in the urine of MTA-treated rats. A quinone methide intermediate may be produced from the metabolic activation process in vitro and in vivo. The metabolite was reactive to glutathione and N-acetyl cysteine. MTA induced marked cytotoxicity in cultured rat primary hepatocytes. Pretreatment of aminobenzotriazole, a nonselective P450 enzyme inhibitor, attenuated the susceptibility of hepatocytes to MTA cytotoxicity.


Subject(s)
Metoprolol , Microsomes, Liver , Animals , Rats , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Glutathione/metabolism , Metoprolol/metabolism , Metoprolol/pharmacology , Microsomes, Liver/metabolism
5.
Int J Mol Sci ; 23(19)2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36232617

ABSTRACT

Heart failure (HF) carries the highest mortality in the western world and ß-blockers [ß-adrenergic receptor (AR) antagonists] are part of the cornerstone pharmacotherapy for post-myocardial infarction (MI) chronic HF. Cardiac ß1AR-activated ßarrestin2, a G protein-coupled receptor (GPCR) adapter protein, promotes Sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2a SUMO (small ubiquitin-like modifier)-ylation and activity, thereby directly increasing cardiac contractility. Given that certain ß-blockers, such as carvedilol and metoprolol, can activate ßarrestins and/or SERCA2a in the heart, we investigated the effects of these two agents on cardiac ßarrestin2-dependent SERCA2a SUMOylation and activity. We found that carvedilol, but not metoprolol, acutely induces ßarrestin2 interaction with SERCA2a in H9c2 cardiomyocytes and in neonatal rat ventricular myocytes (NRVMs), resulting in enhanced SERCA2a SUMOylation. However, this translates into enhanced SERCA2a activity only in the presence of the ß2AR-selective inverse agonist ICI 118,551 (ICI), indicating an opposing effect of carvedilol-occupied ß2AR subtype on carvedilol-occupied ß1AR-stimulated, ßarrestin2-dependent SERCA2a activation. In addition, the amplitude of fractional shortening of NRVMs, transfected to overexpress ßarrestin2, is acutely enhanced by carvedilol, again in the presence of ICI only. In contrast, metoprolol was without effect on NRVMs' shortening amplitude irrespective of ICI co-treatment. Importantly, the pro-contractile effect of carvedilol was also observed in human induced pluripotent stem cell (hIPSC)-derived cardiac myocytes (CMs) overexpressing ßarrestin2, and, in fact, it was present even without concomitant ICI treatment of human CMs. Metoprolol with or without concomitant ICI did not affect contractility of human CMs, either. In conclusion, carvedilol, but not metoprolol, stimulates ßarrestin2-mediated SERCA2a SUMOylation and activity through the ß1AR in cardiac myocytes, translating into direct positive inotropy. However, this unique ßarrestin2-dependent pro-contractile effect of carvedilol may be opposed or masked by carvedilol-bound ß2AR subtype signaling.


Subject(s)
Heart Failure , Induced Pluripotent Stem Cells , Adenosine Triphosphatases/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Carvedilol/pharmacology , Heart Failure/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Metoprolol/metabolism , Metoprolol/pharmacology , Metoprolol/therapeutic use , Myocytes, Cardiac/metabolism , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Ubiquitins/metabolism , beta-Arrestin 2/metabolism
6.
Am J Kidney Dis ; 77(5): 704-712, 2021 05.
Article in English | MEDLINE | ID: mdl-33010357

ABSTRACT

RATIONAL & OBJECTIVE: Beta-blockers are recommended for patients with heart failure (HF) but their benefit in the dialysis population is uncertain. Beta-blockers are heterogeneous, including with respect to their removal by hemodialysis. We sought to evaluate whether ß-blocker use and their dialyzability characteristics were associated with early mortality among patients with chronic kidney disease with HF who transitioned to dialysis. STUDY DESIGN: Retrospective cohort study. SETTING & PARTICIPANTS: Adults patients with chronic kidney disease (aged≥18 years) and HF who initiated either hemodialysis or peritoneal dialysis during January 1, 2007, to June 30, 2016, within an integrated health system were included. EXPOSURES: Patients were considered treated with ß-blockers if they had a quantity of drug dispensed covering the dialysis transition date. OUTCOMES: All-cause mortality within 6 months and 1 year or hospitalization within 6 months after transition to maintenance dialysis. ANALYTICAL APPROACH: Inverse probability of treatment weights using propensity scores was used to balance covariates between treatment groups. Cox proportional hazard analysis and logistic regression were used to investigate the association between ß-blocker use and study outcomes. RESULTS: 3,503 patients were included in the study. There were 2,115 (60.4%) patients using ß-blockers at transition. Compared with nonusers, the HR for all-cause mortality within 6 months was 0.79 (95% CI, 0.65-0.94) among users of any ß-blocker and 0.68 (95% CI, 0.53-0.88) among users of metoprolol at transition. There were no observed differences in all-cause or cardiovascular-related hospitalization. LIMITATIONS: The observational nature of our study could not fully account for residual confounding. CONCLUSIONS: Beta-blockers were associated with a lower rate of mortality among incident hemodialysis patients with HF. Similar associations were not observed for hospitalizations within the first 6 months following transition to dialysis.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Heart Failure/drug therapy , Hospitalization/statistics & numerical data , Kidney Failure, Chronic/therapy , Mortality , Renal Dialysis , Adrenergic beta-Antagonists/metabolism , Aged , Aged, 80 and over , Atenolol/metabolism , Atenolol/therapeutic use , Bisoprolol/metabolism , Bisoprolol/therapeutic use , Carvedilol/metabolism , Carvedilol/therapeutic use , Cause of Death , Cohort Studies , Female , Heart Failure/complications , Humans , Kidney Failure, Chronic/complications , Labetalol/metabolism , Labetalol/therapeutic use , Logistic Models , Male , Metoprolol/metabolism , Metoprolol/therapeutic use , Middle Aged , Nadolol/metabolism , Nadolol/therapeutic use , Proportional Hazards Models , Propranolol/metabolism , Propranolol/therapeutic use , Protective Factors , Retrospective Studies , Risk , Risk Factors
7.
Molecules ; 25(11)2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32531944

ABSTRACT

Water from wastewater treatment plants contains concentrations of pharmaceutically active compounds as high as micrograms per liter, which can adversely affect fish health and behavior, and contaminate the food chain. Here, we tested the ability of the common carp hepatic S9 fraction to produce the main metabolites from citalopram, metoprolol, sertraline, and venlafaxine. Metabolism in fish S9 fractions was compared to that in sheep. The metabolism of citalopram was further studied in fish. Our results suggest a large difference in the rate of metabolites formation between fish and sheep. Fish hepatic S9 fractions do not show an ability to form metabolites from venlafaxine, which was also the case for sheep. Citalopram, metoprolol, and sertraline were metabolized by both fish and sheep S9. Citalopram showed concentration-dependent N-desmethylcitalopram formation with Vmax = 1781 pmol/min/mg and Km = 29.7 µM. The presence of ellipticine, a specific CYP1A inhibitor, in the incubations reduced the formation of N-desmethylcitalopram by 30-100% depending on the applied concentration. These findings suggest that CYP1A is the major enzyme contributing to the formation of N-desmethylcitalopram. In summary, the results from the present in vitro study suggest that common carp can form the major metabolites of citalopram, metoprolol, and sertraline.


Subject(s)
Citalopram/metabolism , Cytochrome P-450 CYP1A1/metabolism , Metoprolol/metabolism , Microsomes, Liver/metabolism , Pharmaceutical Preparations/metabolism , Sertraline/metabolism , Venlafaxine Hydrochloride/metabolism , Animals , Carps , Female , In Vitro Techniques , Male , Sheep
8.
Clin Lab ; 65(5)2019 May 01.
Article in English | MEDLINE | ID: mdl-31115220

ABSTRACT

BACKGROUND: CYP2D6*10 is mainly responsible for the large pharmacokinetic variability of routinely administered metoprolol in middle-aged and elderly Asian patients. Utilizing an efficient method for identifying the CYP2D6*10 genotypes is clinically important for evaluating the pharmacokinetic effect of administration of metoprolol. This study attempted to evaluate the effectiveness of the two methods used to detect the rs1065852 and rs1135840 SNPs of the CYP2D6*10 gene. METHODS: Blood samples were processed for the collection of genomic DNA from 198 subjects across Chinese population, and detection of CYP2D6*10 (rs1065852 and rs1135840) was performed using the PyroMark Q24 pyrose-quencing and matrix-assisted laser desorption/ionization time-of-flight mass-spectrometry (MALDI-TOF MS). The discordant results were further validated with Sanger sequencing. We eventually attempted to assess some features of these two methods including reliability, rapidness, being appropriate, and cost-effectiveness. RESULTS: Genotyping of rs1065852 and rs1135840 detected by MALDI-TOF MS were concordant with those identified by PyroMark Q24 pyrosequencing in all 198 (100%) individuals. The hands-on-time and the turnaround time were shorter in the PyroMark Q24 pyrosequencing method than in the MALDI-TOF MS method for SNP of CYP2D6*10. In terms of being cost-effective and high-throughput, the MALDI-TOF MS method outperformed the PyroMark Q24 pyrosequencing method. CONCLUSIONS: CYP2D6*10 genotypes detected by PyroMark Q24 pyrosequencing and MALDI-TOF-MS showed that both methods were reliable, rapid, appropriate, and cost-effective methods. These methods are valuable for clinical applications.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Genotyping Techniques/methods , High-Throughput Nucleotide Sequencing/methods , Polymorphism, Single Nucleotide , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Aged , Antihypertensive Agents/blood , Antihypertensive Agents/metabolism , Antihypertensive Agents/therapeutic use , Asian People/genetics , China , Cost-Benefit Analysis , Cytochrome P-450 CYP2D6/metabolism , Female , Genotype , Genotyping Techniques/economics , Humans , Hypertension/blood , Hypertension/drug therapy , Hypertension/genetics , Male , Metoprolol/blood , Metoprolol/metabolism , Metoprolol/therapeutic use , Middle Aged , Reproducibility of Results
9.
Mol Pharm ; 15(10): 4677-4688, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29996058

ABSTRACT

The use of in vitro data for the quantitative prediction of transporter-mediated clearance is critical. Central to this evaluation is the use of hepatocytes, since they contain the full complement of transporters and metabolic enzymes. In general, uptake clearance (CLuptake) is evaluated by measuring the appearance of compound in the cell. Passive clearance (CLpd) is often determined by conducting parallel studies at 4 °C or by attempting to saturate uptake pathways. Both approaches have their limitations. Recent studies have proposed the use of Rifamycin-SV (RFV) as a pan-inhibitor of hepatic uptake pathways. In our studies, we confirm that transport activity of all major hepatic uptake transporters is inhibited significantly by RFV at 1 mM (OATP1B1, 1B3, and 2B1 = NTCP (80%), OCT1 (65%), OAT2 (60%)). Under these incubation conditions, we found that the free intracellular concentration of RFV is ∼175 µM and that several major CYPs and UGTs can be reversibly inhibited. Using this approach, we also determined CLuptake and CLpd of nine known OATP substrates across three different lots of human hepatocytes. The scaling factors generated for these compounds at 37 °C with RFV and 4 °C were found to be similar. The CLpd of passively permeable compounds like metoprolol and semagacestat were found to be higher at 37 °C compared to 4 °C, indicating a temperature effect on these compounds. In addition, our data also suggests that incorporation of medium concentrations into CLuptake and CLpd calculations may be critical for highly protein bound and highly lipophilic drugs. Overall, our data indicate that RFV, instead of 4 °C, can be reliably used to measure CLuptake and CLpd of drugs.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , Rifamycins/metabolism , Alanine/analogs & derivatives , Alanine/metabolism , Azepines/metabolism , Biological Transport , Humans , Kinetics , Metoprolol/metabolism
10.
Pharmacoepidemiol Drug Saf ; 27(6): 621-629, 2018 06.
Article in English | MEDLINE | ID: mdl-29575226

ABSTRACT

PURPOSE: Co-prescription of paroxetine/fluoxetine (a strong CYP2D6 inhibitor) in metoprolol (a CYP2D6 substrate) users is common, but data on the clinical consequences of this drug-drug interaction are limited and inconclusive. Therefore, we assessed the effect of paroxetine/fluoxetine initiation on the existing treatment with metoprolol on the discontinuation and dose adjustment of metoprolol among elderly. METHODS: We performed a cohort study using the University of Groningen IADB.nl prescription database (www.IADB.nl). We selected all elderly (≥60 years) who had ever been prescribed metoprolol and had a first co-prescription of paroxetine/fluoxetine, citalopram (weak CYP2D6 inhibitor), or mirtazapine (negative control) from 1994 to 2015. The exposure group was metoprolol and paroxetine/fluoxetine co-prescription, and the other groups acted as controls. The outcomes were early discontinuation and dose adjustment of metoprolol. Logistic regression was applied to estimate adjusted odds ratios (OR) and 95% confidence intervals (CI). RESULTS: Combinations of metoprolol-paroxetine/fluoxetine, metoprolol-citalopram, and metoprolol-mirtazapine were started in 528, 673, and 625 patients, respectively. Compared with metoprolol-citalopram, metoprolol-paroxetine/fluoxetine was not significantly associated with the early discontinuation and dose adjustment of metoprolol (OR = 1.07, 95% CI:0.77-1.48; OR = 0.87, 95% CI:0.57-1.33, respectively). In comparison with metoprolol-mirtazapine, metoprolol-paroxetine/fluoxetine was associated with a significant 43% relative increase in early discontinuation of metoprolol (OR = 1.43, 95% CI:1.01-2.02) but no difference in the risk of dose adjustment. Stratified analysis by gender showed that women have a significantly high risk of metoprolol early discontinuation (OR = 1.62, 95% CI:1.03-2.53). CONCLUSION: Paroxetine/fluoxetine initiation in metoprolol prescriptions, especially for female older patients, is associated with the risk of early discontinuation of metoprolol.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP2D6/metabolism , Fluoxetine/pharmacokinetics , Metoprolol/administration & dosage , Aged , Aged, 80 and over , Citalopram/administration & dosage , Citalopram/pharmacokinetics , Cohort Studies , Cytochrome P-450 CYP2D6 Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Drug Prescriptions/statistics & numerical data , Drug Therapy, Combination , Female , Fluoxetine/administration & dosage , Humans , Male , Metoprolol/metabolism , Metoprolol/pharmacokinetics , Middle Aged , Mirtazapine/administration & dosage , Mirtazapine/pharmacokinetics , Netherlands , Paroxetine
11.
Water Sci Technol ; 77(3-4): 1137-1148, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29488977

ABSTRACT

The effect of granular activated carbon (GAC) addition on the removal of diclofenac, ibuprofen, metoprolol, galaxolide and triclosan in a up-flow anaerobic sludge blanket (UASB) reactor was studied. Prior to the reactor studies, batch experiments indicated that addition of activated carbon to UASB sludge can decrease micropollutant concentrations in both liquid phase and sludge. In continuous experiments, two UASB reactors were operated for 260 days at an HRT of 20 days, using a mixture of source separated black water and sludge from aerobic grey water treatment as influent. GAC (5.7 g per liter of reactor volume) was added to one of the reactors on day 138. No significant difference in COD removal and biogas production between reactors with and without GAC addition was observed. In the presence of GAC, fewer micropollutants were washed out with the effluent and a lower accumulation of micropollutants in sludge and particulate organic matter occurred, which is an advantage in micropollutant emission reduction from wastewater. However, the removal of micropollutants by adding GAC to a UASB reactor would require more activated carbon compared to effluent post-treatment. Additional research is needed to estimate the effect of bioregeneration on the lifetime of activated carbon in a UASB-GAC reactor.


Subject(s)
Bioreactors , Carbon/chemistry , Waste Disposal, Fluid/methods , Water Pollutants, Chemical/chemistry , Anaerobiosis , Benzopyrans/chemistry , Benzopyrans/metabolism , Carbon/metabolism , Diclofenac/chemistry , Diclofenac/metabolism , Ibuprofen/chemistry , Ibuprofen/metabolism , Metoprolol/chemistry , Metoprolol/metabolism , Sewage/chemistry , Triclosan/chemistry , Triclosan/metabolism , Water Pollutants, Chemical/metabolism
12.
Drug Metab Dispos ; 45(8): 896-899, 2017 08.
Article in English | MEDLINE | ID: mdl-28495902

ABSTRACT

A ß-blocker, metoprolol, is one of the in vivo probes for human cytochrome P450 (P450) 2D6. Investigation of nonhuman primate P450 enzymes helps to improve the accuracy of the extrapolation of pharmacokinetic data from animals into humans. Common marmosets (Callithrix jacchus) are a potential primate model for preclinical research, but the detailed roles of marmoset P450 enzymes in metoprolol oxidation remain unknown. In this study, regio- and stereo-selectivity of metoprolol oxidations by a variety of P450 enzymes in marmoset and human livers were investigated in vitro. Although liver microsomes from cynomolgus monkeys and rats preferentially mediated S-metoprolol O-demethylation and R-metoprolol α-hydroxylation, respectively, those from humans, marmosets, minipigs, and dogs preferentially mediated R-metoprolol O-demethylation, in contrast to the slow rates of R- and S-metoprolol oxidation in mouse liver microsomes. R- and S-metoprolol O-demethylation activities in marmoset livers were strongly inhibited by quinidine and ketoconazole, and were significantly correlated with bufuralol 1'-hydroxylation and midazolam 1'-hydroxylation activities and also with P450 2D and 3A4 contents, which is different from the case in human livers that did not have any correlations with P450 3A-mediated midazolam 1'-hydroxylation. Recombinant human P450 2D6 enzyme and marmoset P450 2D6/3A4 enzymes effectively catalyzed R-metoprolol O-demethylation, comparable to the activities of human and marmoset liver microsomes, respectively. These results indicated that the major roles of P450 2D enzymes for the regio- and stereo-selectivity of metoprolol oxidation were similar between human and marmoset livers, but the minor roles of P450 3A enzymes were unique to marmosets.


Subject(s)
Cardiovascular Agents/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Liver/metabolism , Metoprolol/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Callithrix/metabolism , Dogs , Female , Humans , Male , Mice , Microsomes, Liver/metabolism , Middle Aged , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Swine , Swine, Miniature , Young Adult
13.
Luminescence ; 32(6): 942-951, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28233399

ABSTRACT

In the present work, the mechanism of the interaction between a ß1 receptor blocker, metoprolol succinate (MS) and human serum albumin (HSA) under physiological conditions was investigated by spectroscopic techniques, namely fluorescence, Fourier transform infra-red spectroscopy (FT-IR), fluorescence lifetime decay and circular dichroism (CD) as well as molecular docking and cyclic voltammetric methods. The fluorescence and lifetime decay results indicated that MS quenched the intrinsic intensity of HSA through a static quenching mechanism. The Stern-Volmer quenching constants and binding constants for the MS-HSA system at 293, 298 and 303 K were obtained from the Stern-Volmer plot. Thermodynamic parameters for the interaction of MS with HSA were evaluated; negative values of entropy change (ΔG°) indicated the spontaneity of the MS and HSA interaction. Thermodynamic parameters such as negative ΔH° and positive ΔS° values revealed that hydrogen bonding and hydrophobic forces played a major role in MS-HSA interaction and stabilized the complex. The binding site for MS in HSA was identified by competitive site probe experiments and molecular docking studies. These results indicated that MS was bound to HSA at Sudlow's site I. The efficiency of energy transfer and the distance between the donor (HSA) and acceptor (MS) was calculated based on the theory of Fosters' resonance energy transfer (FRET). Three-dimensional fluorescence spectra and CD results revealed that the binding of MS to HSA resulted in an obvious change in the conformation of HSA. Cyclic voltammograms of the MS-HSA system also confirmed the interaction between MS and HSA. Furthermore, the effects of metal ions on the binding of MS to HSA were also studied.


Subject(s)
Metoprolol/chemistry , Serum Albumin, Human/chemistry , Binding Sites , Circular Dichroism , Humans , Hydrophobic and Hydrophilic Interactions , Metoprolol/metabolism , Molecular Docking Simulation , Protein Binding , Serum Albumin, Human/metabolism , Spectroscopy, Fourier Transform Infrared , Thermodynamics
14.
Mol Pharm ; 13(9): 3013-21, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27504798

ABSTRACT

Currently there are only a limited number of determinations of human Peff in the distal small intestine and none in the large intestine. This has hindered the validation of preclinical models with regard to absorption in the distal parts of the intestinal tract, which can be substantial for BCS class II-IV drugs, and drugs formulated into modified-release (MR) dosage forms. To meet this demand, three model drugs (atenolol, metoprolol, and ketoprofen) were dosed in solution intravenously, and into the jejunum, ileum, and colon of 14 healthy volunteers. The Peff of each model drug was then calculated using a validated deconvolution method. The median Peff of atenolol in the jejunum, ileum, and colon was 0.45, 0.15, and 0.013 × 10(-4) cm/s, respectively. The corresponding values for metoprolol were 1.72, 0.72, and 1.30 × 10(-4) cm/s, and for ketoprofen 8.85, 6.53, and 3.37 × 10(-4) cm/s, respectively. This is the first study where the human Peff of model drugs has been determined in all parts of the human intestinal tract in the same subjects. The jejunal values were similar to directly determined values using intestinal single-pass perfusion, indicating that the deconvolution method is a valid approach for determining regional Peff. The values from this study will be highly useful in the validation of preclinical regional absorption models and in silico tools.


Subject(s)
Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Adolescent , Adult , Atenolol/metabolism , Atenolol/pharmacokinetics , Colon/metabolism , Female , Humans , Ileum/metabolism , Intestine, Small/metabolism , Jejunum/metabolism , Ketoprofen/metabolism , Ketoprofen/pharmacokinetics , Male , Metoprolol/metabolism , Metoprolol/pharmacokinetics , Middle Aged , Permeability , Young Adult
15.
Biomed Chromatogr ; 30(8): 1309-17, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26766521

ABSTRACT

A sensitive, accurate and reliable bioanalytical method for the enantioselective determination of metoprolol in plasma and saliva samples utilizing liquid chromatography-electrospray ionization tandem mass spectrometry was developed and validated. Human plasma and saliva samples were pretreated by microextraction by packed sorbent (MEPS) prior to analysis. A new MEPS syringe form with two inputs was used. Metoprolol enantiomers and internal standard pentycaine (IS) were eluted from MEPS sorbent using isopropanol after removal of matrix interferences using aliquots of 5% methanol in water. Complete separation of metoprolol enantiomers was achieved on a Cellulose-SB column (150 × 4.6 mm, 5 µm) using isocratic elution with mobile phase 0.1% ammonium hydroxide in hexane-isopropanol (80:20, v/v) with a flow rate of 0.8 mL/min. A post-column solvent-assisted ionization was applied to enhance metoprolol ionization signal in positive mode monitoring (+ES) using 0.5% formic acid in isopropanol at a flow rate of 0.2 mL/min. The total chromatographic run time was 10 min for each injection. The detection of metoprolol in plasma and saliva samples was performed using triple quadrupole tandem mass spectrometer in +ES under the following mass transitions: m/z 268.08 → 72.09 for metoprolol and m/z 303.3 → 154.3 for IS. The linearity range was 2.5-500 ng/mL for both R- and S-metoprolol in plasma and saliva. The limits of detection and quantitation for both enantiomers were 0.5 and 2.5 ng/mL respectively, in both matrices (plasma and saliva). The intra- and inter-day precisions were presented in terms of RSD values for replicate analysis of quality control samples and were <5%; the accuracy of determinations varied from 96 to 99%. The method was able to determine the therapeutic levels of metoprolol enantiomers in both human plasma and saliva samples successfully, which can aid in therapeutic drug monitoring in clinical laboratories. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Adrenergic beta-Antagonists/metabolism , Chromatography, Liquid/methods , Metoprolol/metabolism , Saliva/metabolism , Tandem Mass Spectrometry/methods , Adrenergic beta-Antagonists/blood , Humans , Limit of Detection , Metoprolol/blood , Reproducibility of Results , Stereoisomerism
16.
Biomed Chromatogr ; 30(10): 1556-72, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27006091

ABSTRACT

The aim of the present study was to evaluate the contribution of metabolites to drug-drug interaction and drug-herb interaction using the inhibition of CYP2D6 and CYP3A4 by metoprolol (MET) and its metabolites. The peak concentrations of unbound plasma concentration of MET, α-hydroxy metoprolol (HM), O-desmethyl metoprolol (ODM) and N-desisopropyl metoprolol (DIM) were 90.37 ± 2.69, 33.32 ± 1.92, 16.93 ± 1.70 and 7.96 ± 0.94 ng/mL, respectively. The metabolites identified, HM and ODM, had a ratio of metabolic area under the concentration-time curve (AUC) to parent AUC of ≥0.25 when either total or unbound concentration of metabolite was considered. In vitro CYP2D6 and CYP3A4 inhibition by MET, HM and ODM study revealed that MET, HM and ODM were not inhibitors of CYP3A4-catalyzed midazolam metabolism and CYP2D6-catalyzed dextromethorphan metabolism. However, DIM only met the criteria of >10% of the total drug related material and <25% of the parent using unbound concentrations. If CYP inhibition testing is solely based on metabolite exposure, DIM metabolite would probably not be considered. However, the present study has demonstrated that DIM contributes significantly to in vitro drug-drug interaction. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors/pharmacology , Cytochrome P-450 CYP2D6/drug effects , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/drug effects , Metoprolol/pharmacology , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Area Under Curve , Cytochrome P-450 CYP2D6 Inhibitors/metabolism , Cytochrome P-450 CYP3A Inhibitors/metabolism , Drug Interactions , Humans , Metoprolol/metabolism
17.
Biopharm Drug Dispos ; 37(9): 511-521, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27662517

ABSTRACT

Drug interactions are one of the commonest causes of side effects, particularly in long-term therapy. The aim of the current study was to investigate the possible effects of metoprolol on the pharmacokinetics of metformin in rats and to clarify the mechanism of drug interaction. In this study, rats were treated with metformin alone or in combination with metoprolol. Plasma, urine and tissue concentrations of metformin were determined by HPLC. Western blotting and real-time qPCR were used to evaluate the expression of rOCTs and rMATE1. The results showed that, after single or 7-day repeated administration, the plasma concentrations of metformin in the co-administration group were significantly decreased compared with that in the metformin group. However, the parameter V/F of metformin in the co-administration group was markedly increased compared with that in the metformin group. The hepatic, renal and muscular Kp of metformin were markedly elevated after co-administration with metoprolol. Consistently, metformin uptake in rat kidney slices was significantly induced by metoprolol. In addition, multiple administrations of metoprolol significantly reduced the expression of rMATE1 in rat kidney as well as the urinary excretion of metformin. Importantly, after long-term administration, lactic acid and uric acid levels in the co-administration group were increased by 25% and 26%, respectively, compared with that in the metformin group. These results indicate that metoprolol can decrease the plasma concentration of metformin via the induction of hepatic, renal and muscular uptake, and long-term co-administration of metformin and metoprolol can cause elevated lactic acid and uric acid levels. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Kidney/metabolism , Liver/metabolism , Metformin/blood , Metoprolol/metabolism , Muscle, Skeletal/metabolism , Adrenergic beta-1 Receptor Antagonists/blood , Adrenergic beta-1 Receptor Antagonists/metabolism , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Drug Interactions/physiology , Hypoglycemic Agents/blood , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Liver/drug effects , Male , Metformin/pharmacology , Metoprolol/blood , Metoprolol/pharmacology , Muscle, Skeletal/drug effects , Organ Culture Techniques , Random Allocation , Rats , Rats, Wistar
18.
Molecules ; 22(1)2016 Dec 27.
Article in English | MEDLINE | ID: mdl-28035999

ABSTRACT

Pathological states in the organism, e.g., renal or hepatic diseases, cataract, dysfunction of coronary artery, diabetes mellitus, and also intensive workout, induce the structural modification of proteins called molecular ageing or N-A isomerization. The aim of this study was to analyze the structural changes of serum albumin caused by alkaline ageing using absorption, spectrofluorescence, and circular dichroism spectroscopy. The N-A isomerization generates significant changes in bovine (BSA) and human (HSA) serum albumin subdomains-the greatest changes were observed close to the tryptophanyl (Trp) and tyrosyl (Tyr) residue regions while a smaller change was observed in phenyloalanine (Phe) environment. Moreover, the changes in the polarity of the Trp neighborhood as well as the impact of the ageing process on α-helix, ß-sheet content, and albumin molecule rotation degree have been analyzed. Based on the spectrofluorescence study, the alterations in metoprolol binding affinity to the specific sites that increase the toxicity of the drug were investigated.


Subject(s)
Aging/metabolism , Metoprolol/metabolism , Serum Albumin/chemistry , Serum Albumin/ultrastructure , Animals , Binding Sites , Cattle , Circular Dichroism , Computer Simulation , Humans , Protein Binding , Protein Structure, Tertiary , Spectrometry, Fluorescence
19.
Drug Metab Dispos ; 43(12): 1960-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26438628

ABSTRACT

1-Aminobenzotriazole (ABT) is regularly used in vivo as a nonspecific and irreversible cytochrome P450 inhibitor to elucidate the role of metabolism on the pharmacokinetic profile of xenobiotics. However, few reports have considered the recent findings that ABT can alter drug absorption or have investigated the possible differential inhibition of ABT on intestinal and hepatic metabolism. To address these uncertainties, pharmacokinetic studies under well controlled and defined ABT pretreatment conditions (50 mg/kg, 1 hour ABT i.v. and 16 hours ABT p.o.) were conducted prior to the oral administration of metoprolol, a permeable P450 probe that undergoes extensive intestinal and hepatic metabolism. The pharmacokinetic profile of metoprolol was affected differently by the two ABT pretreatments. An increase in area under the curve of 16-fold with ABT p.o. and 6.5-fold with ABT i.v. was observed compared with control. Based on in vitro studies, this difference could not be attributed to a differential inhibition of intestinal and hepatic metabolism. In the ABT i.v. pretreatment group, the increase in area under the curve was also associated with a prolonged time at maximal concentration (24-fold versus control), suggesting a delay in absorption. This was further confirmed by the administration of a charcoal meal, which resulted in a 7-fold increase in stomach weights in the 1-hour ABT pretreated groups compared with the untreated or 16-hour ABT pretreated rats. Based on these results, we recommend pretreating rats with ABT p.o. 16 hours before the administration of a test compound to preserve the inhibitory effect on intestinal and hepatic metabolism and avoid the confounding effect on drug absorption.


Subject(s)
Metoprolol/metabolism , Triazoles/metabolism , Animals , Drug Interactions/physiology , Gastric Emptying/drug effects , Gastric Emptying/physiology , Male , Metoprolol/chemistry , Metoprolol/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Triazoles/chemistry , Triazoles/pharmacology
20.
Br J Clin Pharmacol ; 79(4): 605-16, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25291152

ABSTRACT

AIM: To investigate the influence of gestational diabetes mellitus (GDM) on the kinetic disposition and transplacental and amniotic fluid distribution of metoprolol and its metabolites O-desmethylmetoproloic acid and α-hydroxymetoprolol stereoisomers in hypertensive parturients receiving a single dose of the racemic drug. METHODS: The study was conducted on hypertensive parturients with well-controlled GDM (n = 11) and non-diabetic hypertensive parturients (n = 24), all receiving a single 100 mg oral dose of racemic metoprolol tartrate before delivery. Serial maternal blood samples (0-24 h) and umbilical blood and amniotic fluid samples were collected for the quantitation of metoprolol and its metabolite stereoisomers using LC-MS/MS or fluorescence detection. RESULTS: The kinetic disposition of metoprolol and its metabolites was stereoselective in the diabetic and control groups. Well-controlled GDM prolonged tmax for both enantiomers of metoprolol (1.5 vs. 2.5 h R-(+)-MET; 1.5 vs. 2.75 h S-(-)-MET) and O-desmethylmetoproloic acid (2.0 vs. 3.5 h R-(+)-AOMD; 2.0 vs. 3.0 h S-(-)-OAMD), and for the four stereoisomers of α-hydroxymetoprolol (2.0 vs. 3.0 h for 1'S,2R-, 1'R,2R- and 1'R,2S-OHM; 2.0 vs. 3.5 h for 1'S,2S-OHM) and reduced the transplacental distribution of 1'S,2S-, 1'R,2R-, and 1'R,2S-OHM by approximately 20%. CONCLUSIONS: The kinetic disposition of metoprolol was enantioselective, with plasma accumulation of the S-(-)-MET eutomer. Well-controlled GDM prolonged the tmax of metoprolol and O-desmethylmetoproloic acid enantiomers and the α-hydroxymetoprolol stereoisomers and reduced by about 20% the transplacental distribution of 1'S,2S-, 1'R,2R-, and 1'R,2S-OHM. Thus, well-controlled GDM did not change the activity of CYP2D6 and CYP3A involved in metoprolol metabolism.


Subject(s)
Antihypertensive Agents/blood , Diabetes, Gestational/metabolism , Hypertension/drug therapy , Metoprolol/blood , Placenta/metabolism , Adult , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Antihypertensive Agents/therapeutic use , Diabetes, Gestational/blood , Diabetes, Gestational/drug therapy , Female , Fetal Blood/chemistry , Humans , Hypertension/blood , Hypertension/complications , Hypertension/metabolism , Infant, Newborn , Metoprolol/chemistry , Metoprolol/metabolism , Metoprolol/therapeutic use , Parturition , Pregnancy , Stereoisomerism , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL