Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
J Med Chem ; 63(10): 5488-5500, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32337993

ABSTRACT

Neprilysin (NEP) and angiotensin-converting enzyme (ACE) are two key zinc-dependent metallopeptidases in the natriuretic peptide and kinin systems and renin-angiotensin-aldosterone system, respectively. They play an important role in blood pressure regulation and reducing the risk of heart failure. Vasopeptidase inhibitors omapatrilat and sampatrilat possess dual activity against these enzymes by blocking the ACE-dependent conversion of angiotensin I to the potent vasoconstrictor angiotensin II while simultaneously halting the NEP-dependent degradation of vasodilator atrial natriuretic peptide. Here, we report crystal structures of omapatrilat, sampatrilat, and sampatrilat-ASP (a sampatrilat analogue) in complex with NEP at 1.75, 2.65, and 2.6 Å, respectively. A detailed analysis of these structures and the corresponding structures of ACE with these inhibitors has provided the molecular basis of dual inhibitor recognition involving the catalytic site in both enzymes. This new information will be very useful in the design of safer and more selective vasopeptidase inhibitors of NEP and ACE for effective treatment in hypertension and heart failure.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/metabolism , Drug Design , Mesylates/metabolism , Neprilysin/metabolism , Peptidyl-Dipeptidase A/metabolism , Pyridines/metabolism , Thiazepines/metabolism , Tyrosine/analogs & derivatives , Angiotensin-Converting Enzyme Inhibitors/chemistry , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Crystallography, X-Ray/methods , Mesylates/chemistry , Neprilysin/chemistry , Peptidyl-Dipeptidase A/chemistry , Protein Binding/physiology , Protein Structure, Secondary , Pyridines/chemistry , Thiazepines/chemistry , Tyrosine/chemistry , Tyrosine/metabolism
2.
J Med Chem ; 61(22): 10141-10154, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30372620

ABSTRACT

Omapatrilat was designed as a vasopeptidase inhibitor with dual activity against the zinc metallopeptidases angiotensin-1 converting enzyme (ACE) and neprilysin (NEP). ACE has two homologous catalytic domains (nACE and cACE), which exhibit different substrate specificities. Here, we report high-resolution crystal structures of omapatrilat in complex with nACE and cACE and show omapatrilat has subnanomolar affinity for both domains. The structures show nearly identical binding interactions for omapatrilat in each domain, explaining the lack of domain selectivity. The cACE complex structure revealed an omapatrilat dimer occupying the cavity beyond the S2 subsite, and this dimer had low micromolar inhibition of nACE and cACE. These results highlight residues beyond the S2 subsite that could be exploited for domain selective inhibition. In addition, it suggests the possibility of either domain specific allosteric inhibitors that bind exclusively to the nonprime cavity or the potential for targeting specific substrates rather than completely inhibiting the enzyme.


Subject(s)
Drug Design , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/metabolism , Pyridines/metabolism , Thiazepines/metabolism , Amino Acid Sequence , Catalytic Domain , Humans , Ligands , Models, Molecular
3.
Naunyn Schmiedebergs Arch Pharmacol ; 391(2): 185-196, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29230490

ABSTRACT

Tianeptine is an atypical antidepressant with a unique mechanism of action and recently it has been also reported that its major metabolite, compound MC5, possesses pharmacological activity similar to that of the parent drug. The current study aims to investigate the pharmacokinetics (PK) of both tianeptine and MC5 after intravenous or intraperitoneal administration of the parent drug as well as the metabolic ratio of MC5 in rats. To achieve these goals an LC-MS/MS method using the small sample volume for the quantitation of tianeptine and its active metabolite MC5 in rat plasma and liver perfusate has been developed and validated. Following an intravenous administration of tianeptine pharmacokinetic parameters were calculated by non-compartmental analysis. The average tianeptine volume of distribution at steady state was 2.03 L/kg and the systemic clearance equaled 1.84 L/h/kg. The mean elimination half-lives of tianeptine and MC5 metabolite were 1.16 and 7.53 h, respectively. The hepatic clearance of tianeptine determined in the isolated rat liver perfusion studies was similar to the perfusate flow rate despite the low metabolic ratio of MC5. Mass spectrometric analysis of rat bile indicated that tianeptine and MC5 metabolite are eliminated with bile as glucuronide and glutamine conjugates. Bioavailability of tianeptine after its intraperitoneal administration was 69%. The PK model with a metabolite compartment developed in this study for both tianeptine and MC5 metabolite after two routes of administration may facilitate tianeptine dosage selection for the prospective pharmacological experiments.


Subject(s)
Tandem Mass Spectrometry/methods , Thiazepines/administration & dosage , Thiazepines/metabolism , Thiazepines/pharmacokinetics , Animals , Antidepressive Agents, Tricyclic/administration & dosage , Antidepressive Agents, Tricyclic/analysis , Antidepressive Agents, Tricyclic/pharmacokinetics , Chromatography, Liquid/methods , Drug Administration Routes , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Wistar , Thiazepines/analysis
4.
Bioorg Med Chem Lett ; 27(14): 3048-3054, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28554871

ABSTRACT

A series of new of furan derivatised [1,4] benzothiazepine analogues were synthesized starting from 1-(furan-2-yl)ethanone. 1-(Furan-2-yl)ethanone was converted into chalcones by its reaction with various aromatic aldehydes, then were reacted with 2-aminobenzenethiol in acidic conditions to obtain the title compounds in good yields. The synthesized new compounds were characterized by 1H NMR, 13C NMR, Mass spectral studies and elemental analyses. All the new compounds were evaluated for their in vitro VRV-PL-8a and H+/K+ ATPase inhibitor properties. Preliminary studies revealed that, some molecules amongst the designed series showed promising VRV-PL-8a and H+/K+ ATPase inhibitor properties. Further, rigid body docking studies were performed to understand possible docking sites of the molecules on the target proteins and the mode of binding. This finding presents a promising series of lead molecules that can serve as prototypes for the treatment of inflammatory related disorder that can mitigate the ulcer inducing side effect shown by other NSAIDs.


Subject(s)
Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Group II Phospholipases A2/antagonists & inhibitors , H(+)-K(+)-Exchanging ATPase/chemistry , Thiazepines/chemical synthesis , Thiazepines/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Binding Sites , Chalcones/chemistry , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Furans/chemistry , Group II Phospholipases A2/metabolism , H(+)-K(+)-Exchanging ATPase/metabolism , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Molecular Conformation , Molecular Docking Simulation , Protein Structure, Tertiary , Proton Pump Inhibitors/chemical synthesis , Proton Pump Inhibitors/chemistry , Proton Pump Inhibitors/metabolism , Proton Pump Inhibitors/pharmacology , Thiazepines/chemistry , Thiazepines/metabolism
5.
Neuropsychopharmacology ; 42(10): 2052-2063, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28303899

ABSTRACT

Depression is a debilitating chronic illness that affects around 350 million people worldwide. Current treatments, such as selective serotonin reuptake inhibitors, are not ideal because only a fraction of patients achieve remission. Tianeptine is an effective antidepressant with a previously unknown mechanism of action. We recently reported that tianeptine is a full agonist at the mu opioid receptor (MOR). Here we demonstrate that the acute and chronic antidepressant-like behavioral effects of tianeptine in mice require MOR. Interestingly, while tianeptine also produces many opiate-like behavioral effects such as analgesia and reward, it does not lead to tolerance or withdrawal. Furthermore, the primary metabolite of tianeptine (MC5), which has a longer half-life, mimics the behavioral effects of tianeptine in a MOR-dependent fashion. These results point to the possibility that MOR and its downstream signaling cascades may be novel targets for antidepressant drug development.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Receptors, Opioid, mu/metabolism , Thiazepines/pharmacology , Analgesics, Opioid/pharmacology , Animals , Antidepressive Agents, Tricyclic/metabolism , Antidepressive Agents, Tricyclic/pharmacokinetics , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Dose-Response Relationship, Drug , Drug Tolerance , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Morphine/pharmacology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Thiazepines/metabolism , Thiazepines/pharmacokinetics
6.
Chem Commun (Camb) ; 52(34): 5880-3, 2016 Apr 30.
Article in English | MEDLINE | ID: mdl-27052411

ABSTRACT

As a mechanism of defense against pathogens and other types of stress, watercress plants produce a variety of elicited chemical defenses generally known as phytoalexins. Herein the chemical structure, synthesis, biosynthesis and antifungal activity of cyclonasturlexin, the most intriguing indolyl phytoalexin isolated to date, are reported.


Subject(s)
Indoles/pharmacology , Nasturtium/metabolism , Thiazepines/pharmacology , Alternaria/drug effects , Cyclization , Fungicides, Industrial/chemical synthesis , Fungicides, Industrial/metabolism , Fungicides, Industrial/pharmacology , Indoles/chemical synthesis , Indoles/metabolism , Thiazepines/chemical synthesis , Thiazepines/metabolism , Thiocarbamates/metabolism
7.
J Cardiovasc Pharmacol ; 65(5): 494-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25636078

ABSTRACT

INTRODUCTION: K201, a 1,4-benzodiazepine derivative, acts on multiple cardiac ion channels and the ryanodine receptor. We tested whether administration of M-II, the main metabolite of K201, would terminate induced atrial flutter (AFL) or atrial fibrillation (AF) in the canine sterile pericarditis model. METHODS: In 6 dogs, electrophysiologic studies were performed at baseline and after drug administration, measuring atrial effective refractory period (AERP), and conduction time from 3 sites during pacing at cycle lengths (400, 300, and 200 milliseconds) on postoperative days 1-4. In 12 induced episodes of sustained AF/AFL (2/10, respectively), M-II was administered intravenously to test efficacy. Five of the AFL episodes were studied in the open chest state during simultaneous multisite atrial mapping. RESULTS: M-II terminated 2/2 AF and 8/10 AFL episodes, prolonged AERP (P < 0.05), significantly increased atrial pacing capture thresholds but did not significantly change atrial conduction time. AFL CL prolongation was largely explained by prolonged conduction in an area of slow conduction in the reentrant circuit. AFL terminated with block in the area of slow conduction. CONCLUSIONS: M-II was very effective in terminating AFL/AF in the canine sterile pericarditis model. AFL terminated due to block in the area of slow conduction of the reentrant circuit.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Flutter/drug therapy , Heart Conduction System/drug effects , Pericarditis/complications , Thiazepines/pharmacology , Thiazolidinediones/pharmacology , Animals , Anti-Arrhythmia Agents/metabolism , Atrial Fibrillation/diagnosis , Atrial Fibrillation/etiology , Atrial Fibrillation/physiopathology , Atrial Flutter/diagnosis , Atrial Flutter/etiology , Atrial Flutter/physiopathology , Biotransformation , Cardiac Pacing, Artificial , Disease Models, Animal , Dogs , Electrocardiography , Electrophysiologic Techniques, Cardiac , Heart Conduction System/physiopathology , Thiazepines/metabolism , Thiazolidinediones/metabolism , Time Factors
8.
J Org Chem ; 78(24): 12726-34, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24256447

ABSTRACT

A synthesis of the benzothiazepine phosphonic acid 3, employing both enzymatic and transition metal catalysis, is described. The quaternary chiral center of 3 was obtained by resolution of ethyl (2-ethyl)norleucinate (4) with porcine liver esterase (PLE) immobilized on Sepabeads. The resulting (R)-amino acid (5) was converted in two steps to aminosulfate 7, which was used for construction of the benzothiazepine ring. Benzophenone 15, prepared in four steps from trimethylhydroquinone 11, enabled sequential incorporation of phosphorus (Arbuzov chemistry) and sulfur (Pd(0)-catalyzed thiol coupling) leading to mercaptan intermediate 18. S-Alkylation of 18 with aminosulfate 7 followed by cyclodehydration afforded dihydrobenzothiazepine 20. Iridium-catalyzed asymmetric hydrogenation of 20 with the complex of [Ir(COD)2BArF] (26) and Taniaphos ligand P afforded the (3R,5R)-tetrahydrobenzothiazepine 30 following flash chromatography. Oxidation of 30 to sulfone 31 and phosphonate hydrolysis completed the synthesis of 3 in 12 steps and 13% overall yield.


Subject(s)
Esterases/metabolism , Iridium/chemistry , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Thiazepines/pharmacology , Animals , Catalysis , Crystallography, X-Ray , Esterases/chemistry , Humans , Liver/enzymology , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Swine , Thiazepines/chemistry , Thiazepines/metabolism
9.
J Med Chem ; 56(12): 5094-114, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23678871

ABSTRACT

The apical sodium-dependent bile acid transporter (ASBT) transports bile salts from the lumen of the gastrointestinal (GI) tract to the liver via the portal vein. Multiple pharmaceutical companies have exploited the physiological link between ASBT and hepatic cholesterol metabolism, which led to the clinical investigation of ASBT inhibitors as lipid-lowering agents. While modest lipid effects were demonstrated, the potential utility of ASBT inhibitors for treatment of type 2 diabetes has been relatively unexplored. We initiated a lead optimization effort that focused on the identification of a potent, nonabsorbable ASBT inhibitor starting from the first-generation inhibitor 264W94 (1). Extensive SAR studies culminated in the discovery of GSK2330672 (56) as a highly potent, nonabsorbable ASBT inhibitor which lowers glucose in an animal model of type 2 diabetes and shows excellent developability properties for evaluating the potential therapeutic utility of a nonabsorbable ASBT inhibitor for treatment of patients with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Discovery , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Methylamines/chemistry , Methylamines/pharmacology , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Thiazepines/chemistry , Thiazepines/pharmacology , Animals , Bile Acids and Salts/metabolism , Dogs , Drug Stability , HEK293 Cells , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Male , Methylamines/metabolism , Methylamines/therapeutic use , Mice , Rats , Solubility , Thiazepines/metabolism , Thiazepines/therapeutic use
10.
PLoS One ; 8(1): e54208, 2013.
Article in English | MEDLINE | ID: mdl-23349825

ABSTRACT

Activation of the skeletal muscle ryanodine receptor (RyR1) complex results in the rapid release of Ca(2+) from the sarcoplasmic reticulum and muscle contraction. Dissociation of the small FK506 binding protein 12 subunit (FKBP12) increases RyR1 activity and impairs muscle function. The 1,4-benzothiazepine derivative JTV519, and the more specific derivative S107 (2,3,4,5,-tetrahydro-7-methoxy-4-methyl-1,4-benzothiazepine), are thought to improve skeletal muscle function by stabilizing the RyR1-FKBP12 complex. Here, we report a high degree of nonspecific and specific low affinity [(3)H]S107 binding to SR vesicles. SR vesicles enriched in RyR1 bound ∼48 [(3)H]S107 per RyR1 tetramer with EC(50) ∼52 µM and Hillslope ∼2. The effects of S107 and FKBP12 on RyR1 were examined under conditions that altered the redox state of RyR1. S107 increased FKBP12 binding to RyR1 in SR vesicles in the presence of reduced glutathione and the NO-donor NOC12, with no effect in the presence of oxidized glutathione. Addition of 0.15 µM FKBP12 to SR vesicles prevented FKBP12 dissociation; however, in the presence of oxidized glutathione and NOC12, FKBP12 dissociation was observed in skeletal muscle homogenates that contained 0.43 µM myoplasmic FKBP12 and was attenuated by S107. In single channel measurements with FKBP12-depleted RyR1s, in the absence and presence of NOC12, S107 augmented the FKBP12-mediated decrease in channel activity. The data suggest that S107 can reverse the harmful effects of redox active species on SR Ca(2+) release in skeletal muscle by binding to RyR1 low affinity sites.


Subject(s)
Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Protein 1A/metabolism , Thiazepines/pharmacology , Animals , Binding, Competitive/drug effects , Calcium/metabolism , Glutathione/pharmacology , Immunoblotting , Kinetics , Lipid Bilayers/metabolism , Nitroso Compounds/pharmacology , Protein Binding/drug effects , Protein Stability/drug effects , Rabbits , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism , Tacrolimus/pharmacology , Thiazepines/metabolism , Tritium
11.
Cancer Chemother Pharmacol ; 71(2): 331-44, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23108699

ABSTRACT

PURPOSE: Protein kinase D (PKD) mediates diverse biological responses including cell growth and survival. Therefore, PKD inhibitors may have therapeutic potential. We evaluated the in vitro cytotoxicity of two PKD inhibitors, kb-NB142-70 and its methoxy analogue, kb-NB165-09, and examined their in vivo efficacy and pharmacokinetics. METHODS: The in vitro cytotoxicities of kb-NB142-70 and kb-NB165-09 were evaluated by MTT assay against PC-3, androgen-independent prostate cancer cells, and CFPAC-1 and PANC-1, pancreatic cancer cells. Efficacy studies were conducted in mice bearing either PC-3 or CPFAC-1 xenografts. Tumor-bearing mice were euthanized between 5 and 1,440 min after iv dosing, and plasma and tissue concentrations were measured by HPLC-UV. Metabolites were characterized by LC-MS/MS. RESULTS: kb-NB142-70 and kb-NB165-09 inhibited cellular growth in the low-mid µM range. The compounds were inactive when administered to tumor-bearing mice. In mice treated with kb-NB142-70, the plasma C (max) was 36.9 nmol/mL, and the PC-3 tumor C (max) was 11.8 nmol/g. In mice dosed with kb-NB165-09, the plasma C (max) was 61.9 nmol/mL, while the PANC-1 tumor C (max) was 8.0 nmol/g. The plasma half-lives of kb-NB142-70 and kb-NB165-09 were 6 and 14 min, respectively. Both compounds underwent oxidation and glucuronidation. CONCLUSIONS: kb-NB142-70 and kb-NB165-09 were rapidly metabolized, and concentrations in tumor were lower than those required for in vitro cytotoxicity. Replacement of the phenolic hydroxyl group with a methoxy group increased the plasma half-life of kb-NB165-09 2.3-fold over that of kb-NB142-70. Rapid metabolism in mice suggests that next-generation compounds will require further structural modifications to increase potency and/or metabolic stability.


Subject(s)
Heterocyclic Compounds, 3-Ring/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Thiazepines/pharmacology , Animals , Chromatography, High Pressure Liquid , Female , Heterocyclic Compounds, 3-Ring/metabolism , Humans , Mice , Mice, SCID , Protein Binding , Protein Kinase Inhibitors/metabolism , Tandem Mass Spectrometry , Thiazepines/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
12.
Mol Pharmacol ; 81(1): 53-62, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21989257

ABSTRACT

New drugs with enhanced electron donor properties that target the ryanodine receptor from skeletal muscle sarcoplasmic reticulum (RyR1) are shown to be potent inhibitors of single-channel activity. In this article, we synthesize derivatives of the channel activator 4-chloro-3-methyl phenol (4-CmC) and the 1,4-benzothiazepine channel inhibitor 4-[-3{1-(4-benzyl) piperidinyl}propionyl]-7-methoxy-2,3,4,5-tetrahydro-1,4-benzothiazepine (K201, JTV519) with enhanced electron donor properties. Instead of activating channel activity (~100 µM), the 4-methoxy analog of 4-CmC [4-methoxy-3-methyl phenol (4-MmC)] inhibits channel activity at submicromolar concentrations (IC(50) = 0.34 ± 0.08 µM). Increasing the electron donor characteristics of K201 by synthesizing its dioxole congener results in an approximately 16 times more potent RyR1 inhibitor (IC(50) = 0.24 ± 0.05 µM) compared with K201 (IC(50) = 3.98 ± 0.79 µM). Inhibition is not caused by an increased closed time of the channel but seems to be caused by an open state block of RyR1. These alterations to chemical structure do not influence the ability of these drugs to affect Ca(2+)-dependent ATPase activity of sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase type 1. Moreover, the FKBP12 protein, which stabilizes RyR1 in a closed configuration, is shown to be a strong electron donor. It seems as if FKBP12, K201, its dioxole derivative, and 4-MmC inhibit RyR1 channel activity by virtue of their electron donor characteristics. These results embody strong evidence that designing new drugs to target RyR1 with enhanced electron donor characteristics results in more potent channel inhibitors. This is a novel approach to the design of new, more potent drugs with the aim of functionally modifying RyR1 single-channel activity.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/metabolism , Drug Discovery , Ryanodine Receptor Calcium Release Channel/metabolism , Thiazepines/chemistry , Thiazepines/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/chemical synthesis , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Drug Discovery/methods , Electron Transport/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Protein Stability/drug effects , Rabbits , Ryanodine Receptor Calcium Release Channel/chemical synthesis
13.
Eur J Pharm Sci ; 45(5): 559-69, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22207039

ABSTRACT

1-(4-Methoxyphenylethyl)-11H-benzo[f]-1,2-dihydro-pyrido[3,2,c][1,2,5]oxathiazepine 5,5 dioxide (BZN) is a cytotoxic derivative with very promising in vitro activity. Regulatory authority for registration of pharmaceuticals for human use requires to evaluate the stability of active compound under various stress conditions. Forced degradation of BZN was investigated under hydrolytic (0.1M NaOH, 0.1M HCl, neutral), oxidative (3.3% H(2)O(2)), photolytic (visible light) and thermal (25 °C, 70 °C) settings. Relevant degradation took place under thermal acidic (0.1M HCl, 70 °C) and oxidative (3.3% H(2)O(2)) conditions. Liquid chromatography-mass spectrometry (LC-MS) analyses revealed the presence of ten degradation products whose structures were characterized by electrospray ionization-orbitrap mass spectrometry. The full scan accurate mass analysis of degradation products was confirmed or refuted using three tools furnished by the MS software: (1) predictive chemical formula and corresponding mass error; (2) double bond equivalent (DBE) calculation; and (3) accurate mass product ion spectra of degradation products. The structural elucidation showed that the tricycle moiety was unstable under thermal acidic and oxidative conditions since four degradation products possess an opened oxathiazepine ring. Then, a simple and fast HPLC-UV method was developed and validated for the determination of the degradation kinetic of BZN under acidic and oxidative conditions. The method was linear in the 5-100 µg mL(-1) concentration range with a good precision (RSD=2.2% and 2.7% for the repeatability and the intermediate precision, respectively) and a bias which never exceeded 1.6%, whatever the quality control level. With regards to the BZN concentration, a first-order degradation process was determined, with t(1/2)=703 h and 1140 h, under oxidative and acidic conditions, respectively.


Subject(s)
Antimitotic Agents/chemistry , Thiazepines/chemistry , Acids/chemistry , Antimitotic Agents/metabolism , Chromatography, Liquid/methods , Drug Stability , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Oxidation-Reduction , Photolysis , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/methods , Stress, Physiological , Thiazepines/metabolism
14.
J Pharm Sci ; 100(9): 3985-94, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21618543

ABSTRACT

The intestinal absorption mechanism of temocapril, an ester-type prodrug of temocaprilat, was evaluated using Caco-2 cell monolayers with or without active carboxylesterase (CES)-mediated hydrolysis. The inhibition of CES-mediated hydrolysis was achieved by pretreatment of the monolayers with bis-p-nitrophenyl phosphate (BNPP), which inhibited 94% of the total hydrolysis of temocapril in the Caco-2 cells. The remaining 6% hydrolysis was due to the presence of serine esterases, other than CES, on the cell membranes. Transport experiments under CES-inhibited conditions showed temocapril not to be a substrate for peptide transporter 1 (PEPT1) or organic anion transporting polypeptides (OATPs), but to be an inhibitor of PEPT1; P-glycoprotein (P-gp) and breast-cancer-resistant protein (BCRP) were responsible for the efflux of temocapril, which was mainly absorbed by passive diffusion at low apical pH. In Caco-2 cell monolayers with CES-mediated hydrolysis intact, temocaprilat derived from temocapril, was 2.5-fold more rapidly transported into the apical compartment than into the basolateral compartment due to the presence of microvilli on the apical membrane. In contrast, temocaprilat at low intracellular concentrations, was preferentially transported across the basolateral membrane under CES-inhibited conditions.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacokinetics , Carboxylesterase/metabolism , Prodrugs/pharmacokinetics , Thiazepines/pharmacokinetics , Angiotensin-Converting Enzyme Inhibitors/metabolism , Biotransformation , Caco-2 Cells , Chromatography, High Pressure Liquid , Humans , Hydrolysis , Prodrugs/metabolism , Thiazepines/metabolism
15.
J Mass Spectrom ; 46(2): 112-30, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21254313

ABSTRACT

The metabolic fate of the emerging drug candidate S107, possessing the potential for misuse as performance-enhancing agent in sports, was investigated by in vitro phase I and II experiments with human microsomal and S9 liver enzymes. The metabolites were identified by liquid chromatography-mass spectrometry with electrospray ionisation in positive mode (LC-ESI-MS/MS). Their collision-induced dissociation behaviour was studied by high-resolution/high accuracy Orbitrap MS(n) analysis, supported by stable isotope labelling, H/D-exchange experiments and density functional theory calculations. Monooxygenation accounted for the main phase I metabolic transformation due to N- and S-oxidation of the 1,4-benzothiazepine core, as substantiated by chemical synthesis, selective reduction methods and characteristic APCI in source fragmentation behaviour of the metabolites. Another dominant metabolic pathway was demethylation, yielding the N- and O-demethylated metabolite, respectively. The latter was further conjugated by glucuronidation as well as sulfonation in subsequent phase II metabolic reactions, whereas the N-demethylated metabolite was not amenable to conjugation. The active drug molecule itself was converted to two glucuronic acid conjugates, which are proposed to consist of two quaternary S107-N(+)-glucuronide isomers. All glucuronides were susceptible to enzymatic hydrolysis with ß-glucuronidase (Escherichia coli). A comprehensive LC-ESI-MS(/MS)-based detection method for urine was developed and its fitness for purpose was assessed. The assay can serve as a potential screening and/or confirmation method for S107 in clinical drug testing and doping control analysis in the future.


Subject(s)
Chromatography, Liquid/methods , Doping in Sports/prevention & control , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Thiazepines/metabolism , Female , Humans , Male , Thiazepines/chemistry , Thiazepines/urine
16.
Proc Natl Acad Sci U S A ; 107(1): 436-41, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20018762

ABSTRACT

Mitochondrial Ca(2+) efflux is linked to numerous cellular activities and pathophysiological processes. Although it is established that an Na(+)-dependent mechanism mediates mitochondrial Ca(2+) efflux, the molecular identity of this transporter has remained elusive. Here we show that the Na(+)/Ca(2+) exchanger NCLX is enriched in mitochondria, where it is localized to the cristae. Employing Ca(2+) and Na(+) fluorescent imaging, we demonstrate that mitochondrial Na(+)-dependent Ca(2+) efflux is enhanced upon overexpression of NCLX, is reduced by silencing of NCLX expression by siRNA, and is fully rescued by the concomitant expression of heterologous NCLX. NCLX-mediated mitochondrial Ca(2+) transport was inhibited, moreover, by CGP-37157 and exhibited Li(+) dependence, both hallmarks of mitochondrial Na(+)-dependent Ca(2+) efflux. Finally, NCLX-mediated mitochondrial Ca(2+) exchange is blocked in cells expressing a catalytically inactive NCLX mutant. Taken together, our results converge to the conclusion that NCLX is the long-sought mitochondrial Na(+)/Ca(2+) exchanger.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium/metabolism , Animals , Brain/cytology , Brain/metabolism , Clonazepam/analogs & derivatives , Clonazepam/metabolism , Homeostasis , Humans , Mice , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Myocardium/cytology , Myocardium/metabolism , Rats , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Thiazepines/metabolism
17.
Chem Biodivers ; 6(11): 2092-100, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19937843

ABSTRACT

Temocapril is a prodrug whose hydrolysis by carboxylesterase 1 (CES1) yields the active ACE inhibitor temocaprilat. This molecular-dynamics (MD) study uses a resolved structure of the human CES1 (hCES1) to investigate some mechanistic details of temocapril hydrolysis. The ionization constants of temocapril (pK1 and pK3) and temocaprilat (pK1, pK2, and pK3) were determined experimentally and computationally using commercial algorithms. The constants so obtained were in good agreement and revealed that temocapril exists mainly in three ionic forms (a cation, a zwitterion, and an anion), whereas temocaprilat exists in four major ionic forms (a cation, a zwitterion, an anion, and a dianion). All these ionic forms were used as ligands in 5-ns MS simulations. While the cationic and zwitterionic forms of temocapril were involved in an ion-pair bond with Glu255 suggestive of an inhibitor behavior, the anionic form remained in a productive interaction with the catalytic center. As for temocaprilat, its cation appeared trapped by Glu255, while its zwitterion and anion made a slow departure from the catalytic site and a partial egress from the protein. Only its dianion was effectively removed from the catalytic site and attracted to the protein surface by Lys residues. A detailed mechanism of product egress emerges from the simulations.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/metabolism , Carboxylesterase/metabolism , Thiazepines/chemistry , Thiazepines/metabolism , Algorithms , Biotransformation , Computer Simulation , Electrochemistry , Ligands , Models, Biological , Models, Molecular , Potentiometry , Software , X-Ray Diffraction
18.
Rapid Commun Mass Spectrom ; 23(15): 2363-70, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19575395

ABSTRACT

New insights into the biochemistry of cardiac arrhythmia and skeletal muscle fatigue have yielded new drug candidates to counteract these phenomena. Major biological targets have become ryanodine receptor (RyR)-based Ca(2+)-release channels, which tend to 'leak' under various circumstances including strenuous exercise and, thus, cause aberrant calcium sparks that entail impaired muscle function. Therapeutics, which are referred to as rycals, are currently being developed to treat cardiac arrhythmia via enhancement of calstabin-ryanodine affinities that causes a stabilization of the RyR. These therapeutics possess potential for misuse in sports, and an early implementation of target analytes such as the benzothiazepine derivatives S-107 and JTV-519 or putative metabolites into doping control screening procedures is recommended. Reference compounds, deuterated analogues, and a putative metabolic product were synthesized, and electron ionization mass spectra of these products were studied and dissociation pathways elucidated by means of tandem mass spectrometry (MS/MS) and accurate mass measurements. The characterized analytes were incorporated into existing sports drug testing assays based on liquid-liquid extraction and subsequent gas chromatography/mass spectrometry (GC/MS) analysis, and specificity, lower limit of detection (4-6 ng/mL), intraday and interday precision (1.5-17.2%), as well as recovery (63-66%) were determined. The established procedure proved suitable for routine doping control analysis to detect a potential misuse of the drug candidate S-107 in elite sport.


Subject(s)
Gas Chromatography-Mass Spectrometry/methods , Spectrometry, Mass, Electrospray Ionization/methods , Substance Abuse Detection/methods , Thiazepines/chemical synthesis , Thiazepines/urine , Female , Humans , Male , Molecular Structure , Sensitivity and Specificity , Thiazepines/metabolism
19.
J Mass Spectrom ; 44(4): 442-60, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19373874

ABSTRACT

The number of compounds and doping methods in sports is in a state of constant flux. In addition to 'traditional' doping agents, such as anabolic androgenic steroids or erythropoietin, new therapeutics and emerging drugs have considerable potential for misuse in elite sport. Such compounds are commonly based on new chemical structures, and the mechanisms underlying their modes of action represent new therapeutic approaches arising from recent advances in medical research; therefore, sports drug testing procedures need to be continuously modified and complementary methods developed, preferably based on mass spectrometry, to enable comprehensive doping controls. This tutorial not only discusses emerging drugs that can be categorized as anabolic agents (selective androgen receptor modulators, SARMs), gene doping [hypoxia-inducible factor stabilizers, peroxisome-proliferator-activated receptor (PPAR)delta-agonists] and erythropoietin-mimetics (Hematide) but also compounds with potentially performance-enhancing properties that are not classified in the current list of the World Anti-Doping Agency. Compounds such as ryanodine-calstabin-complex modulators (benzothiazepines) are included, their mass spectrometric properties discussed, and current approaches in sports drug testing outlined.


Subject(s)
Anabolic Agents/analysis , Erythropoietin/analysis , Mass Spectrometry/methods , PPAR delta/agonists , Peptides/analysis , Polyethylene Glycols/analysis , Substance Abuse Detection/methods , Anabolic Agents/metabolism , Androgen Antagonists/analysis , Androgen Antagonists/metabolism , Erythropoietin/metabolism , Humans , Hypoxia-Inducible Factor 1/analysis , Hypoxia-Inducible Factor 1/metabolism , Mass Spectrometry/instrumentation , PPAR delta/analysis , PPAR delta/metabolism , Peptides/metabolism , Polyethylene Glycols/metabolism , Receptors, Androgen/analysis , Receptors, Androgen/metabolism , Recombinant Proteins , Thiazepines/analysis , Thiazepines/metabolism
20.
Circulation ; 117(6): 762-72, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18227387

ABSTRACT

BACKGROUND: We previously demonstrated that defective interdomain interaction between N-terminal (0 to 600) and central regions (2000 to 2500) of ryanodine receptor 2 (RyR2) induces Ca2+ leak in failing hearts and that K201 (JTV519) inhibits the Ca2+ leak by correcting the defective interdomain interaction. In the present report, we identified the K201-binding domain and characterized the role of this novel domain in the regulation of the RyR2 channel. METHODS AND RESULTS: An assay using a quartz-crystal microbalance technique (a very sensitive mass-measuring technique) revealed that K201 specifically bound to recombinant RyR2 fragments 1741 to 2270 and 1981 to 2520 but not to other RyR2 fragments from the 1 to 2750 region (1 to 610, 494 to 1000, 741 to 1260, 985 to 1503, 1245 to 1768, 2234 to 2750). By further analysis of the fragment(1741-2270), K201 was found to specifically bind to its subfragment(2114-2149). With the use of the peptide matching this subfragment (DP(2114-2149)) as a carrier, the RyR2 was fluorescently labeled with methylcoumarin acetate (MCA) in a site-directed manner. After tryptic digestion, the major MCA-labeled fragment of RyR2 (155 kDa) was detected by an antibody raised against the central region (Ab(2132)). Moreover, of several recombinant RyR2 fragments, only fragment(2234-2750) was specifically MCA labeled; this suggests that the K201-binding domain(2114-2149) binds with domain(2234-2750). Addition of DP(2114-2149) to the MCA-labeled sarcoplasmic reticulum interfered with the interaction between domain(2114-2149) and domain(2234-2750), causing domain unzipping, as evidenced by an increased accessibility of the bound MCA to a large-size fluorescence quencher. In failing cardiomyocytes, the frequency of spontaneous Ca2+ spark was markedly increased compared with normal cardiomyocytes, whereas incorporation of DP(2114-2149) markedly decreased the frequency of spontaneous Ca2+ spark. CONCLUSIONS: We first identified the K201-binding site as domain(2114-2149) of RyR2. Interruption of the interdomain interaction between the domain(2114-2149) and central domain(2234-2750) seems to mediate stabilization of RyR2 in failing hearts, which may lead to a novel therapeutic strategy against heart failure and perhaps lethal arrhythmia.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Sequence , Animals , Annexin A5/chemistry , Annexin A5/metabolism , Binding Sites , Disease Models, Animal , Dogs , Heart Failure/metabolism , Linear Models , Molecular Sequence Data , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum , Sequence Homology, Amino Acid , Thiazepines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...