Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
1.
Biotechnol J ; 19(1): e2300250, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38048389

ABSTRACT

As a key synthetic intermediate of the cardiovascular drug diltiazem, methyl (2R,3S)-3-(4-methoxyphenyl) glycidate ((2R,3S)-MPGM) (1) is accessible via the ring closure of chlorohydrin (3S)-methyl 2-chloro-3-hydroxy-3-(4-methoxyphenyl)propanoate ((3S)-2). We report the efficient reduction of methyl 2-chloro-3-(4-methoxyphenyl)-3-oxo-propanoate (3) to (3S)-2 using an engineered enzyme SSCRM2 possessing 4.5-fold improved specific activity, which was obtained through the structure-guided site-saturation mutagenesis of the ketoreductase SSCR by reliving steric hindrance and undesired interactions. With the combined use of the co-expression fine-tuning strategy, a recombinant E. coli (pET28a-RBS-SSCRM2 /pACYCDuet-GDH), co-expressing SSCRM2 and glucose dehydrogenase, was constructed and optimized for protein expression. After optimizing the reaction conditions, whole-cell-catalyzed complete reduction of industrially relevant 300 g L-1 of 3 was realized, affording (3S)-2 with 99% ee and a space-time yield of 519.1 g∙L-1 ∙d-1 , representing the highest record for the biocatalytic synthesis of (3S)-2 reported to date. The E-factor of this biocatalytic synthesis was 24.5 (including water). Chiral alcohol (3S)-2 generated in this atom-economic synthesis was transformed to (2R,3S)-MPGM in 95% yield with 99% ee.


Subject(s)
Diltiazem , Glucose 1-Dehydrogenase , Glucose 1-Dehydrogenase/metabolism , Diltiazem/metabolism , Escherichia coli/metabolism , Propionates/metabolism , Biocatalysis
2.
Curr Drug Metab ; 24(3): 190-199, 2023.
Article in English | MEDLINE | ID: mdl-36694315

ABSTRACT

BACKGROUND: Carboxylesterase 2 (CES2) is mainly distributed in the human liver and gut, and plays an active role in the metabolic activation of many prodrugs and lipid metabolism. Although CES2 is of great significance, there are still few animal models related to CES2. OBJECTIVES: This research aims to construct Ces2c gene knockout (KO) rats and further study the function of CES2. METHODS: CRISPR/Cas9 gene editing technology was used to target and cleave the rat Ces2c gene. Compensatory effects of major CES subtypes both in the liver and small intestine of KO rats were detected at mRNA levels. Meanwhile, diltiazem and aspirin were used as substrates to test the metabolic capacity of Ces2c in KO rats. RESULTS: This Ces2c KO rat model showed normal growth and breeding without off-target effects. The metabolic function of Ces2c KO rats was verified by the metabolic study of CES2 substrates in vitro. The results showed that the metabolic capacity of diltiazem in KO rats was weakened, while the metabolic ability of aspirin did not change significantly. In addition, the serum physiological indexes showed that the Ces2c deletion did not affect the liver function of rats.. CONCLUSION: The Ces2c KO rat model was successfully constructed by CRISPR/Cas9 system. This rat model can not only be used as an important tool to study the drug metabolism mediated by CES2, but also as an important animal model to study the physiological function of CES2.


Subject(s)
CRISPR-Cas Systems , Diltiazem , Rats , Humans , Animals , Gene Knockout Techniques , Diltiazem/metabolism , Liver/metabolism , Aspirin/metabolism
3.
Neurobiol Learn Mem ; 197: 107700, 2023 01.
Article in English | MEDLINE | ID: mdl-36410654

ABSTRACT

Previous findings have indicated that glucocorticoid hormones impair working memory via an interaction with the ß-adrenoceptor-cAMP signaling cascade to rapidly increase cAMP-dependent protein kinase (PKA) activity within the prefrontal cortex (PFC). However, it remains elusive how such activation of PKA can affect downstream cellular mechanisms in regulating PFC cognitive function. PKA is known to activate l-type voltage-gated Ca2+ channels (LTCCs) which regulate a broad range of cellular processes, including neuronal excitability and neurotransmitter release. The present experiments examined whether LTCC activity within the PFC is required in mediating glucocorticoid and PKA effects on spatial working memory. Male Sprague Dawley rats received bilateral administration of the LTCC inhibitor diltiazem together with either the glucocorticoid receptor agonist RU 28362 or PKA activator Sp-cAMPS into the PFC before testing on a delayed alternation task in a T-maze. Both RU 28362 and Sp-cAMPS impaired working memory, whereas the LTCC inhibitor diltiazem fully blocked the working memory impairment induced by either RU 28362 or Sp-cAMPS. Conversely, bilateral administration of the LTCC agonist Bay K8644 into the PFC was sufficient to impair working memory. Thus, these findings provide support for the view that glucocorticoids, via an interaction with the ß-adrenergic signaling cascade and enhanced PKA activity levels, impair working memory by increasing LTCC activity in the PFC.


Subject(s)
Glucocorticoids , Memory, Short-Term , Rats , Animals , Male , Memory, Short-Term/physiology , Glucocorticoids/pharmacology , Calcium Channels, L-Type/metabolism , Rats, Sprague-Dawley , Diltiazem/metabolism , Diltiazem/pharmacology , Memory Disorders , Prefrontal Cortex/physiology
4.
Pharm Res ; 38(10): 1639-1644, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34729703

ABSTRACT

PURPOSE: Previous studies evaluating ticagrelor drug-drug interactions have not differentiated intestinal versus systemic mechanisms, which we do here. METHODS: Using recently published methodologies from our laboratory to differentiate metabolic- from transporter-mediated drug-drug interactions, a critical evaluation of five published ticagrelor drug-drug interactions was carried out to investigate the purported clinical significance of enzymes and transporters in ticagrelor disposition. RESULTS: The suggested CYP3A4 inhibitors, ketoconazole and diltiazem, displayed unchanged mean absorption time (MAT) and time of maximum concentration (Tmax) values as was expected, i.e., the interactions were mainly mediated by metabolic enzymes. The potential CYP3A4/P-gp inhibitor cyclosporine also showed an unchanged MAT value. Further analysis assuming there was no P-gp effect suggested that the increased AUC and unchanged t1/2 for ticagrelor after cyclosporine administration were attributed to the inhibition of intestinal CYP3A4 rather than P-gp. Rifampin, an inducer of CYP3As after multiple dosing, unexpectedly showed decreased MAT and Tmax values, which cannot be completely explained. In contrast, grapefruit juice, an intestinal CYP3A/P-gp/OATP inhibitor, significantly increased MAT and Tmax values for ticagrelor, which may be due to activation of P-gp or inhibition of OATPs expressed in intestine. CONCLUSIONS: This study provides new insight into the role of transporter pathways in ticagrelor intestinal absorption by examining potential MAT and Tmax changes mediated by drug-drug interactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cyclosporine/metabolism , Cytochrome P-450 CYP3A Inhibitors/metabolism , Cytochrome P-450 CYP3A/metabolism , Ticagrelor/metabolism , Citrus paradisi , Cyclosporine/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Diltiazem/metabolism , Drug Interactions , Fruit and Vegetable Juices , Humans , Intestinal Absorption , Intestines , Ketoconazole/metabolism , Rifampin/metabolism , Ticagrelor/pharmacokinetics
5.
Pharmacol Res Perspect ; 9(5): e00879, 2021 10.
Article in English | MEDLINE | ID: mdl-34628723

ABSTRACT

The unbound concentrations of 14 commercial drugs, including five non-efflux/uptake transporter substrates-Class I, five efflux transporter substrates-class II and four influx transporter substrates-Class III, were simultaneously measured in rat liver, muscle, and blood via microanalysis. Kpuu,liver and Kpuu,muscle were calculated to evaluate the membrane transport activity and cell metabolism on the unbound drug concentrations in the skeletal muscle and liver. For Class I compounds, represented by antipyrine, unbound concentrations among liver, muscle and blood are symmetrically distributed when compound hepatic clearance is low. And when compound hepatic clearance is high, unbound concentrations among liver, muscle and blood are asymmetrically distributed, such as Propranolol. For Class II and III compounds, overall, the unbound concentrations among liver, muscle, and blood are asymmetrically distributed due to a combination of hepatic metabolism and efflux and/or influx transporter activity.


Subject(s)
Cell Membrane/metabolism , Liver/metabolism , Membrane Transport Proteins/metabolism , Muscle, Skeletal/metabolism , Pharmaceutical Preparations/metabolism , Animals , Antipyrine/blood , Antipyrine/metabolism , Atenolol/blood , Atenolol/metabolism , Carbamazepine/blood , Carbamazepine/metabolism , Digoxin/blood , Digoxin/metabolism , Diltiazem/blood , Diltiazem/metabolism , Diphenhydramine/blood , Diphenhydramine/metabolism , Drug Elimination Routes , Gabapentin/blood , Gabapentin/metabolism , Lamotrigine/blood , Lamotrigine/metabolism , Memantine/blood , Memantine/metabolism , Microdialysis , Ofloxacin/blood , Ofloxacin/metabolism , Pharmaceutical Preparations/blood , Propranolol/blood , Propranolol/metabolism , Pyrilamine/blood , Pyrilamine/metabolism , Quinidine/blood , Quinidine/metabolism , Rats , Terfenadine/analogs & derivatives , Terfenadine/blood , Terfenadine/metabolism
6.
Eur J Pharmacol ; 895: 173889, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33482177

ABSTRACT

Careful analysis of previously published reports and some new insights into the structure activity studies revealed an important role of Threonine 1143 in drug binding. Substituting T1143 by alanine and other residues significantly reduced channel inhibition by qDil and Dil. Mutation T1143A did not affect channel activation or inactivation while almost completely diminishing channel block by Dil or qDil. These findings support the view that T1143 serves as drug binding determinant. Other mutations in this position than T1143A (T1143L/Y/S/N/C/V/E) diminished channel inhibition by qDil but additionally affected channel activation and inactivation and may therefore affect channel block allosterically. Collectively, our data suggest that T1143 is an essential diltiazem binding determinant.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Diltiazem/pharmacology , Amino Acid Sequence , Binding Sites , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Diltiazem/metabolism , HEK293 Cells , Humans , Hydrogen Bonding , Kinetics , Membrane Potentials , Point Mutation , Protein Binding , Structure-Activity Relationship , Threonine
7.
Biochem Biophys Res Commun ; 527(3): 799-804, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32423809

ABSTRACT

When administrated orally, the vasodilating drug diltiazem can be metabolized into diacetyl diltiazem in the presence of Bacteroides thetaiotaomicron, a human gut microbe. The removal of acetyl group from the parent drug is carried out by the GDSL/SGNH-family hydrolase BT4096. Here the crystal structure of the enzyme was solved by mercury soaking and single-wavelength anomalous diffraction. The protein folds into two parts. The N-terminal part comprises the catalytic domain which is similar to other GDSL/SGNH hydrolases. The flanking C-terminal part is made up of a ß-barrel subdomain and an α-helical subdomain. Structural comparison shows that the catalytic domain is most akin to acetyl-xylooligosaccharide esterase and allows a plausible binding mode of diltiazem to be proposed. The ß-barrel subdomain is similar in topology to the immunoglobulin-like domains, including some carbohydrate-binding modules, of various bacterial glycoside hydrolases. Consequently, BT4096 might originally function as an oligosaccharide deacetylase with additional subdomains that could enhance substrate binding, and it acts on diltiazem just by accident.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides thetaiotaomicron/enzymology , Diltiazem/metabolism , Gastrointestinal Microbiome , Hydrolases/metabolism , Vasodilator Agents/metabolism , Acetylation , Bacterial Proteins/chemistry , Bacteroides thetaiotaomicron/chemistry , Bacteroides thetaiotaomicron/metabolism , Catalytic Domain , Humans , Hydrolases/chemistry , Models, Molecular , Protein Conformation , Substrate Specificity
8.
Int J Mol Sci ; 21(2)2020 Jan 16.
Article in English | MEDLINE | ID: mdl-31963306

ABSTRACT

Alcoholic beverages stimulate pancreatic enzyme secretions by inducing cholecystokinin (CCK) release. CCK is the major stimulatory hormone of pancreatic exocrine secretions, secreted from enteroendocrine I-cells of the intestine. Fermentation products of alcoholic beverages, such as maleic and succinic acids, influence gastric acid secretions. We hypothesize that maleic and succinic acids stimulate pancreatic exocrine secretions during beer and wine ingestion by increasing CCK secretions. Therefore, the effects of maleic and succinic acids on CCK release were studied in duodenal mucosal cells and the enteroendocrine cell line STC-1. Mucosal cells were perfused for 30 min with 5 min sampling intervals, STC-1 cells were studied under static incubation for 15 min, and supernatants were collected for CCK measurements. Succinate and maleate-induced CCK release were investigated. Succinate and maleate doses dependently stimulated CCK secretions from mucosal cells and STC-1 cells. Diltiazem, a calcium channel blocker, significantly inhibited succinate and maleate-induced CCK secretions from mucosal cells and STC-1 cells. Maleate and succinate did not show cytotoxicity in STC-1 cells. Our results indicate that succinate and maleate are novel CCK-releasing factors in fermented alcoholic beverages and could contribute to pancreatic exocrine secretions and their pathophysiology.


Subject(s)
Cholecystokinin/metabolism , Intestinal Mucosa/cytology , Alcoholic Beverages , Animals , Cell Line , Diltiazem/metabolism , Fermentation/physiology , L-Lactate Dehydrogenase/metabolism , Maleates/metabolism , Rats , Succinic Acid/metabolism
9.
Nature ; 570(7762): 462-467, 2019 06.
Article in English | MEDLINE | ID: mdl-31158845

ABSTRACT

Individuals vary widely in their responses to medicinal drugs, which can be dangerous and expensive owing to treatment delays and adverse effects. Although increasing evidence implicates the gut microbiome in this variability, the molecular mechanisms involved remain largely unknown. Here we show, by measuring the ability of 76 human gut bacteria from diverse clades to metabolize 271 orally administered drugs, that many drugs are chemically modified by microorganisms. We combined high-throughput genetic analyses with mass spectrometry to systematically identify microbial gene products that metabolize drugs. These microbiome-encoded enzymes can directly and substantially affect intestinal and systemic drug metabolism in mice, and can explain the drug-metabolizing activities of human gut bacteria and communities on the basis of their genomic contents. These causal links between the gene content and metabolic activities of the microbiota connect interpersonal variability in microbiomes to interpersonal differences in drug metabolism, which has implications for medical therapy and drug development across multiple disease indications.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , Gastrointestinal Microbiome/genetics , Pharmaceutical Preparations/metabolism , Animals , Bacteria/classification , Bacteria/enzymology , Bacteroides thetaiotaomicron/enzymology , Bacteroides thetaiotaomicron/genetics , Bacteroides thetaiotaomicron/metabolism , Diltiazem/metabolism , Female , Gastrointestinal Microbiome/physiology , Genome, Bacterial/genetics , Germ-Free Life , Humans , Male , Mice , Pharmaceutical Preparations/administration & dosage , Substrate Specificity
10.
Chemosphere ; 229: 434-442, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31082711

ABSTRACT

Estuaries routinely receive discharges of contaminants of emerging concern from urban regions. Within these dynamic estuarine systems, salinity and pH can vary across spatial and temporal scales. Our previous research identified bioaccumulation of the calcium channel blocker diltiazem and the antihistamine diphenhydramine in several species of fish residing in multiple urban estuaries along the Gulf of Mexico in Texas, where field-measured observations of diltiazem in fish plasma exceeded human therapeutic plasma doses. However, there remains a limited understanding of pharmaceutical bioaccumulation in estuarine environments. Here, we examined the influence of pH and salinity on bioconcentration of three pharmaceuticals in the Gulf killifish, Fundulus grandis. F. grandis were exposed to low levels of the ionizable pharmaceuticals carbamazepine, diltiazem, and diphenhydramine at two salinities (5 ppt, 20 ppt) and two pH levels (6.7, 8.3). pH influenced bioconcentration of select weak base pharmaceuticals, while salinity did not, suggesting that intestinal uptake via drinking does not appear to be a major exposure route of these pharmaceuticals in killifish. Compared to our previous pH dependent uptake observations with diphenhydramine in the fathead minnow model, killifish apparent volume of distribution values were markedly lower than fatheads, though killifish bioconcentration factors were similar at high pH and four fold higher at low pH than freshwater fish. Advancing an understanding of environmental gradient influences on pharmacokinetics among fish is necessary to improve bioaccumulation assessments and interpretation of toxicological observations for ionizable contaminants.


Subject(s)
Estuaries , Fundulidae/metabolism , Hydrogen-Ion Concentration , Pharmaceutical Preparations/metabolism , Salinity , Animals , Carbamazepine/metabolism , Diltiazem/metabolism , Diphenhydramine/metabolism , Gulf of Mexico , Humans , Pharmacokinetics , Texas , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/metabolism
11.
Drug Deliv ; 24(1): 414-421, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28165822

ABSTRACT

Diltiazem hydrochloride (DTZ) is a calcium channel antagonist depicted by extensive first pass metabolism and low oral bioavailability. The aim of this work was to develop niosomes for potential nasal delivery of DTZ. Niosomes protect hydrophilic drugs inside their core while nasal route offers both rapid onset and evasion of first-pass metabolism. Niosomes were prepared using a combination of Span 60 or Brij-52 with cholesterol (CHOL) in different molar ratios followed by determination of entrapment efficiency, particle size and in vitro drug release. A parallel design was adopted to evaluate the pharmacokinetic performance of DTZ-loaded niosomes in male Wistar rats. Non-compartmental analysis was performed where Cmax, Tmax, t1/2, MRT, area under the release curve (AUC) and Ke were assessed. The prepared niosomes were spherical with mean particle size 0.82-1.59 µm. Span 60-cholesterol niosomes (1:1 molar ratio) showed the highest entrapment and release efficiencies. In vivo study revealed an increase in MRT, t1/2 and AUC with a decrease in Ke. In conclusion, nasal niosomal formulation of DTZ expressed suitable pharmacokinetic parameters and bioavailability through prolonged duration of action inside the body as well as low rate of elimination depicting a promising alternate to the conventional oral route.


Subject(s)
Diltiazem/administration & dosage , Diltiazem/metabolism , Liposomes/administration & dosage , Liposomes/metabolism , Administration, Intranasal , Animals , Biological Availability , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/metabolism , Male , Rats , Rats, Wistar
12.
Article in English | MEDLINE | ID: mdl-27856310

ABSTRACT

INTRODUCTION: Although therapeutically beneficial in the treatment of certain diseases, L-type calcium channel antagonism can result in unwanted off-target pharmacology leading to adverse drug reactions and to the termination of the development of otherwise promising compounds. In the present study three marketed calcium channel inhibitors, nifedipine, verapamil and diltiazem were profiled in a series of in vitro and ex-vivo assays in an effort to determine the ability of these assays to discriminate, between dihydropyridine versus non-dihydropyridine-like compounds, and how well they can predict the cardiovascular effects observed in a conscious telemetered rat model. METHODS: Standard calcium channel antagonists were profiled in radioligand binding, patch clamp and calcium flux assays. In addition, cardiovascular endpoints related to calcium channel activity were also examined in ex vivo tissue bath preparations, including relaxation of pre-constricted rat aorta and the guinea pig Langendorff isolated heart model. The data generated were correlated with in vivo blood pressure and heart rate data from conscious telemetered rats. RESULTS: Our results show that the binding, FLIPR and aorta assays allow differentiation of the compounds in two distinct classes of L-type calcium channel antagonists, and are good predictors of in vivo outcomes. DISCUSSION: These results suggest that in vitro and ex vivo profiling remains a valuable tool in predicting potential in vivo cardiovascular safety issues, and can aid in the selection of novel development compounds that show inherent inhibitory activity against L-type calcium channels.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Translational Research, Biomedical/methods , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cell Line , Diltiazem/metabolism , Diltiazem/pharmacology , Dose-Response Relationship, Drug , Female , Guinea Pigs , Isolated Heart Preparation/methods , Male , Nifedipine/metabolism , Nifedipine/pharmacology , Rabbits , Rats , Rats, Wistar , Verapamil/metabolism , Verapamil/pharmacology
13.
Rapid Commun Mass Spectrom ; 30(9): 1153-62, 2016 May 15.
Article in English | MEDLINE | ID: mdl-27060844

ABSTRACT

RATIONALE: Diltiazem, a calcium channel blocker drug, is widespread in the environment because of its incomplete elimination during water treatment. It can cause negative effects on aquatic organisms; thus, a rapid and sensitive liquid chromatography/mass spectrometry (LC/MS) method to detect its presence was developed. Our approach is based on accurate mass measurements using a hybrid quadrupole-orbital trap mass spectrometer that was used to measure diltiazem and its metabolites in fish tissue. METHODS: Blood plasma, muscle, liver, and kidney tissues of rainbow trout (Oncorhynchus mykiss), exposed for 42 days to 30 µg L(-1) diltiazem, were used for the method development. No metabolite standards were required to identify the diltiazem biotransformation products in the fish tissue. RESULTS: Overall, 17 phase I diltiazem metabolites (including isomeric forms) were detected and tentatively identified using the MassFrontier spectral interpretation software. A semi-quantitative approach was used for organ-dependent comparison of the metabolite concentrations. CONCLUSIONS: These data increase our understanding about diltiazem and its metabolites in aquatic organisms, such as fish. These encompass desmethylation, desacetylation and hydroxylation as well as their combinations. This study represents the first report of the complex diltiazem phase I metabolic pathways in fish.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channel Blockers/metabolism , Diltiazem/chemistry , Diltiazem/metabolism , Fishes/metabolism , Mass Spectrometry/methods , Animals , Chromatography, Liquid/methods , Kidney/chemistry , Kidney/metabolism , Liver/chemistry , Liver/metabolism , Molecular Structure , Muscles/chemistry , Muscles/metabolism , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/metabolism
14.
Environ Technol ; 35(9-12): 1055-63, 2014.
Article in English | MEDLINE | ID: mdl-24701901

ABSTRACT

The fate and removal of six selected endocrine disrupting compounds in a lab-scale anaerobic/aerobic (A/O) sequencing batch reactor (SBR), operating at 5 days, solids retention time (SRT) were investigated. A carbamazepine (CBZ), acetaminophen (ATP), diltiazem (DTZ), butyl benzyl phthalate (BBP), estrone and progesterone mix was spiked as model endocrine disrupting compounds (EDC) into domestic wastewater obtained from a nearby sewage treatment plant. The influent, effluent and sludge samples from the SBR unit were analysed by using an LC/MS/MS instrument equipped with electrospray ionization. More than 80% removal was observed for all the EDCs tested. It was found that biodegradation is the most important mechanism for BBP, ATP and progesterone. Biodegradation constants were calculated according to the simplified Monod model for these compounds. The DTZ seemed to have lower rate of biodegradation. The CBZ appeared totally resistant to biodegradation. However, it presented a high rate of sorption onto the sludge and was thereby treated. This contradicts with the literature studies.


Subject(s)
Bioreactors/microbiology , Endocrine Disruptors/isolation & purification , Water Pollutants, Chemical/isolation & purification , Acetaminophen/analysis , Acetaminophen/isolation & purification , Acetaminophen/metabolism , Adsorption , Carbamazepine/analysis , Carbamazepine/isolation & purification , Carbamazepine/metabolism , Diltiazem/analysis , Diltiazem/isolation & purification , Diltiazem/metabolism , Endocrine Disruptors/analysis , Endocrine Disruptors/metabolism , Estrone/analysis , Estrone/isolation & purification , Estrone/metabolism , Phthalic Acids/analysis , Phthalic Acids/isolation & purification , Phthalic Acids/metabolism , Progesterone/analysis , Progesterone/isolation & purification , Progesterone/metabolism , Sewage/analysis , Wastewater/analysis , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/metabolism
15.
Neurobiol Aging ; 35(8): 1883-91, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24680323

ABSTRACT

Prior work in healthy rats supported a calcium hypothesis of photoreceptor aging, wherein progressive age-related declines in photopic vision are explainable by the extent of earlier escalating d-cis-diltiazem-insensitive increases in photoreceptor L-type calcium channel (LTCC) activity in vivo. Unlike rats, healthy mice have relatively stable photopic vision until after 18 months of age. We therefore hypothesized that photoreceptor LTCC activity in mice would not progressively increase until after 18 months. In 2-5, 10, 18, and 26 months male C57Bl/6J mice, photoreceptor LTCC activity and retinal thickness were evaluated in vivo (manganese-enhanced magnetic resonance imaging) with some groups also treated with d-cis-diltiazem; visual performance was evaluated (optokinetic tracking). Data were calibrated for cone-only responses using mice without rod transducin (GNAT1-/-). Photopic vision was stable until after 18 months without retinal thinning or progressive increases in retinal manganese uptake. We measured an uptake spike at 10 months. This spike, unlike that in the rat, was diltiazem sensitive in the dark and diltiazem insensitive in the light. Between dark and light, uptake in inner retina of older mice was unequal (unlike that in 2-5 months mice); outer retinal uptake was similar to that in 2-5 months mice. Stable murine photopic visual performance and nonescalating photoreceptor LTCC activity before 18 months of age were consistent with a prediction of the calcium hypothesis. Stark differences in the temporal evolution of mouse and rat photoreceptor LTCC activity suggest the need for personalized identification of the retinal mechanisms contributing to declines in photopic vision to ensure success of future treatment efforts.


Subject(s)
Aging/physiology , Calcium Channels, L-Type/physiology , Color Vision/physiology , Photoreceptor Cells, Vertebrate/physiology , Aging/metabolism , Aging/pathology , Animals , Calcium Channels, L-Type/metabolism , Diltiazem/metabolism , Diltiazem/pharmacology , Magnetic Resonance Imaging/methods , Male , Manganese/metabolism , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/metabolism , Rats , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology
16.
Drug Metab Dispos ; 42(1): 126-33, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141856

ABSTRACT

Carboxylesterase 1 (CES1) is the major hydrolase in human liver. The enzyme is involved in the metabolism of several important therapeutic agents, drugs of abuse, and endogenous compounds. However, no studies have described the role of human CES1 in the activation of two commonly prescribed angiotensin-converting enzyme inhibitors: enalapril and ramipril. Here, we studied recombinant human CES1- and CES2-mediated hydrolytic activation of the prodrug esters enalapril and ramipril, compared with the activation of the known substrate trandolapril. Enalapril, ramipril, and trandolapril were readily hydrolyzed by CES1, but not by CES2. Ramipril and trandolapril exhibited Michaelis-Menten kinetics, while enalapril demonstrated substrate inhibition kinetics. Intrinsic clearances were 1.061, 0.360, and 0.02 ml/min/mg protein for ramipril, trandolapril, and enalapril, respectively. Additionally, we screened a panel of therapeutic drugs and drugs of abuse to assess their inhibition of the hydrolysis of p-nitrophenyl acetate by recombinant CES1 and human liver microsomes. The screening assay confirmed several known inhibitors of CES1 and identified two previously unreported inhibitors: the dihydropyridine calcium antagonist, isradipine, and the immunosuppressive agent, tacrolimus. CES1 plays a role in the metabolism of several drugs used in the treatment of common conditions, including hypertension, congestive heart failure, and diabetes mellitus; thus, there is a potential for clinically relevant drug-drug interactions. The findings in the present study may contribute to the prediction of such interactions in humans, thus opening up possibilities for safer drug treatments.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/metabolism , Carboxylic Ester Hydrolases/metabolism , Inactivation, Metabolic/physiology , Carboxylesterase/metabolism , Diltiazem/metabolism , Drug Interactions/physiology , Enalapril/metabolism , Esters/metabolism , Humans , Hydrolysis , Indoles/metabolism , Kinetics , Liver/enzymology , Liver/metabolism , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Nitrophenols/metabolism , Prodrugs/metabolism , Ramipril/metabolism , Recombinant Proteins/metabolism , Verapamil/metabolism
17.
Xenobiotica ; 43(5): 468-78, 2013 May.
Article in English | MEDLINE | ID: mdl-23153054

ABSTRACT

Focusing on the genetic similarity of CYP3A subfamily enzymes (CYP3A4 and CYP3A5) between monkeys and humans, we have attempted to provide a single-species approach to predicting human hepatic clearance (CLh) of CYP3A4 substrates using pharmacokinetic parameters in cynomolgus monkeys following intravenous administrations. 2. Hepatic intrinsic clearance (CLint,h) of six CYP3A4 substrates (alprazolam, clonazepam, diltiazem, midazolam, nifedipine, and quinidine), covering a wide range of clearance, in monkeys correlated well with that cited in literature for humans (R = 0.90) with a simple equation of Y = 0.165X (Y: human CLint,h, X: monkey CLint,h, represented in mL/min/kg). 3. To verify the predictability of human CLint,h, monkey CLint,h of a test set of CYP3A4 substrates cited in literature (dexamethasone, nifedipine, midazolam, quinidine, tacrolimus, and verapamil) was applied to the equation and human CLint,h was calculated. The human CLint,h of all the substrates was predicted within 3-fold error (fold error: 0.35-2.77). 4. The predictability of human CLh by our method was superior to common in vivo prediction methods (allometry and liver blood flow method). These results suggest that human hepatic clearance of CYP3A4 substrates can be predicted by applying cynomolgus monkey CLint,h obtained following intravenous administrations in each laboratory to the simple equation.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Liver/metabolism , Alprazolam/metabolism , Animals , Diltiazem/metabolism , Humans , Macaca fascicularis/metabolism , Midazolam/metabolism , Tacrolimus/metabolism
18.
Biopharm Drug Dispos ; 33(8): 446-54, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22927137

ABSTRACT

This study investigated the effect of piperine on the gene expression of P-glycoprotein (P-gp) as well as pregnane-X-receptor (PXR) activity and also its implication on the bioavailability of diltiazem, a P-gp substrate. The effect of piperine on the systemic exposure of diltiazem was examined in rats after the intravenous and oral administration of diltiazem with/without 2 week pretreatment with piperine. Compared with the control group given diltiazem (20 mg/kg) alone, the pretreatment with piperine (10 or 20 mg/kg, once daily for 2 weeks) decreased the oral exposure of diltiazem by 36-48% in rats. Consequently, the bioavailability of oral diltiazem was significantly lower (p < 0.05) after the 2 week pretreatment with piperine. The pretreatment with piperine for 2 weeks also reduced the systemic exposure of desacetyldiltiazem, a major active metabolite of diltiazem by approximately 73%, accompanied by a significant decrease in the metabolite-parent ratio. In contrast to the oral pharmacokinetics, piperine did not affect the intravenous pharmacokinetics of diltiazem in rats. Immunoblot analysis indicated that the protein expression level of intestinal P-gp was significantly enhanced after the 2 week pretreatment with piperine in rats. In addition, piperine increased the PXR reporter activity in human hepatoma cells. Taken together, the 2 week pretreatment with piperine significantly induced intestinal P-gp expression in conjunction with stimulated PXR activity and decreased the oral exposure of diltiazem and desacetyldiltiazem in rats.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Alkaloids/administration & dosage , Benzodioxoles/administration & dosage , Calcium Channel Blockers/pharmacokinetics , Diltiazem/pharmacokinetics , Piperidines/administration & dosage , Polyunsaturated Alkamides/administration & dosage , Receptors, Steroid/metabolism , Animals , Biological Availability , Calcium Channel Blockers/blood , Diltiazem/analogs & derivatives , Diltiazem/blood , Diltiazem/metabolism , Food-Drug Interactions , Gene Expression/drug effects , Hep G2 Cells , Humans , Male , Pregnane X Receptor , Rats , Rats, Sprague-Dawley
19.
Drug Metab Dispos ; 40(10): 1966-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22798552

ABSTRACT

In vitro metabolite profiling and characterization experiments are widely employed in early drug development to support safety studies. Samples from incubations of investigational drugs with liver microsomes or hepatocytes are commonly analyzed by liquid chromatography/mass spectrometry for detection and structural elucidation of metabolites. Advanced mass spectrometers with accurate mass capabilities are becoming increasingly popular for characterization of drugs and metabolites, spurring changes in the routine workflows applied. In the present study, using a generic full-scan high-resolution data acquisition approach with a time-of-flight mass spectrometer combined with postacquisition data mining, we detected and characterized metabonates (false metabolites) in microsomal incubations of several alkylamine drugs. If a targeted approach to mass spectrometric detection (without full-scan acquisition and appropriate data mining) were employed, the metabonates may not have been detected, hence their formation underappreciated. In the absence of accurate mass data, the metabonate formation would have been incorrectly characterized because the detected metabonates manifested as direct cyanide-trapped conjugates or as cyanide-trapped metabolites formed from the parent drugs by the addition of 14 Da, the mass shift commonly associated with oxidation to yield a carbonyl. This study demonstrates that high-resolution mass spectrometry and the associated workflow is very useful for the detection and characterization of unpredicted sample components and that accurate mass data were critical to assignment of the correct metabonate structures. In addition, for drugs containing an alkylamine moiety, the results suggest that multiple negative controls and chemical trapping agents may be necessary to correctly interpret the results of in vitro experiments.


Subject(s)
Amines/metabolism , Metabolomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Amines/chemistry , Biotransformation , Chromatography, High Pressure Liquid , Data Mining , Diltiazem/metabolism , Humans , Microsomes, Liver , Molecular Structure , Reproducibility of Results , Workflow
20.
Drug Metabol Drug Interact ; 26(3): 119-25, 2011.
Article in English | MEDLINE | ID: mdl-21929464

ABSTRACT

BACKGROUND: In order to identify a suitable rodent model for preclinical study of calcium antagonists, the pharmacokinetics and metabolism of one of the prototypes diltiazem (DTZ) in normotensive Sprague Dawley (SDR) was compared with Wistar Kyoto (WKY) rats and spontaneously hypertensive rats (SHR) following 5 mg/kg twice daily for five doses given by subcutaneous injection. METHODS: Pharmacokinetic data were analyzed by standard procedures assuming a one-compartment model with first-order input using Rstrips(®), and differences between the groups were considered significant when p<0.05. RESULTS: Plasma concentrations of DTZ were higher in the SHR than the normotensive SDR and WKY rats, although the differences did not reach statistical significance (p>0.05). Plasma concentrations of the active metabolites N-desmethyl DTZ (MA), deacetyl DTZ (M1) and deacetyl N-desmethyl DTZ (M2) were significantly higher in the SHR and WKY rats than the SDR, which was attributed to higher DTZ concentrations and also genetic factors. CONCLUSIONS: Although the differences were mainly quantitative and very small, the study has shown for the first time that the metabolism profiles of DTZ in SHR and WKY rats were closer to humans than SDR, and they may be more preferable rat models to study pharmacokinetic and metabolism studies of DTZ or similar agents.


Subject(s)
Calcium Channel Blockers/metabolism , Diltiazem/metabolism , Animals , Area Under Curve , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...