15 results on '"Volberg W"'
Search Results
2. ChemInform Abstract: Synthesis and Structure‐Activity Relationships of 6‐Heterocyclic‐ Substituted Purines as Inactivation Modifiers of Cardiac Sodium Channels.
- Author
-
ESTEP, K. G., primary, JOSEF, K. A., additional, BACON, E. R., additional, CARABATEAS, P. M., additional, RUMNEY, S. IV, additional, PILLING, G. M., additional, KRAFTE, D. S., additional, VOLBERG, W. A., additional, DILLON, K., additional, DUGRENIER, N., additional, BRIGGS, G. M., additional, CANNIFF, P. C., additional, GORCZYCA, W. P., additional, STANKUS, G. P., additional, and EZRIN, A. M., additional
- Published
- 1995
- Full Text
- View/download PDF
3. Chemical synthesis and characterization of ShK toxin: a potent potassium channel inhibitor from a sea anemone
- Author
-
PENNINGTON, M. W., primary, BYRNES, M. E., additional, ZAYDENBERG, I., additional, KHAYTIN, I., additional, DE CHASTONAY, J., additional, KRAFTE, D. S., additional, HILL, R., additional, MAHNIR, V. M., additional, VOLBERG, W .A., additional, GORCZYCA, W., additional, and KEM, W. R., additional
- Published
- 1995
- Full Text
- View/download PDF
4. Characterization of endogenous sodium channel gene expressed in Chinese hamster ovary cells
- Author
-
Lalik, P. H., primary, Krafte, D. S., additional, Volberg, W. A., additional, and Ciccarelli, R. B., additional
- Published
- 1993
- Full Text
- View/download PDF
5. Identification of a specific radioligand for the cardiac rapidly activating delayed rectifier K+ channel.
- Author
-
Chadwick, C C, primary, Ezrin, A M, additional, O'Connor, B, additional, Volberg, W A, additional, Smith, D I, additional, Wedge, K J, additional, Hill, R J, additional, Briggs, G M, additional, Pagani, E D, additional, and Silver, P J, additional
- Published
- 1993
- Full Text
- View/download PDF
6. WIN 17317-3: novel nonpeptide antagonist of voltage-activated K+ channels in human T lymphocytes.
- Author
-
Hill, R J, Grant, A M, Volberg, W, Rapp, L, Faltynek, C, Miller, D, Pagani, K, Baizman, E, Wang, S, and Guiles, J W
- Abstract
We report the in vitro biological characterization of WIN 17317-3 (1-benzyl-7-chloro-4-n-propylimino-1,4-dihydroquinoline hydrochloride), a novel inhibitor of voltage-activated (n-type) K+ channels in human T lymphocytes. WIN 17317-3 inhibits 125I-charybdotoxin binding to n-type K+ channels with an IC50 value of 83 +/- 4 nM. WIN 17317-3 demonstrates competitive inhibition of 125I-charybdotoxin binding by increasing its dissociation constant without changing the total number of channels bound and by having no effect on its dissociation rate constant. WIN 17317-3 inhibits whole-cell, n-type K+ currents with characteristics indicative of open channel block and has an IC50 value of 335 nM. The compound is 150-fold selective for n-type K+ channels, compared with Ca(2+)-activated, charybdotoxin-sensitive K+ channels in smooth muscle. In purified CD4+ T lymphocytes activated with either anti-CD3 plus phorbol ester or anti-CD3 plus anti-CD28, WIN 17317-3 decreases interleukin-2 production with EC50 values of 0.8 microM and 1 microM, respectively. WIN 17317-3 is a novel, potent, and selective nonpeptide n-type K+ channel antagonist that inhibits interleukin-2 production in human T lymphocytes.
- Published
- 1995
7. (3R,4S)-3-[4-(4-Fluorophenyl)-4-hydroxypiperidin-1-yl]chroman-4,7-diol: A Conformationally Restricted Analogue of the NR2B Subtype-Selective NMDA Antagonist (1S,2S)-1-(4-Hydroxyphenyl)-2- (4-hydroxy-4-phenylpiperidino)-1-propanol
- Author
-
Butler, T. W., Blake, J. F., Bordner, J., Butler, P., Chenard, B. L., Collins, M. A., DeCosta, D., Ducat, M. J., Eisenhard, M. E., Menniti, F. S., Pagnozzi, M. J., Sands, S. B., Segelstein, B. E., Volberg, W., White, W. F., and Zhao, D.
- Abstract
(1S,2S)-1-(4-Hydroxyphenyl)-2-(4-hydroxy-4-phenylpiperidino)-1-propanol (CP-101,606,
1 ) is a recently described antagonist of N-methyl-d -aspartate (NMDA) receptors containing the NR2B subunit. In the present study, the optimal orientation of compounds of this structural type for their receptor was explored. Tethering of the pendent methyl group of1 to the phenolic aromatic ring via an oxygen atom prevents rotation about the central portion of the molecule. Several of the new chromanol compounds have high affinity for the racemic [3H]CP-101,606 binding site on the NMDA receptor and protect against glutamate toxicity in cultured hippocampal neurons. The new ring caused a change in the stereochemical preference of the receptor&sbd;cis (erythro) compounds had better affinity for the receptor than the trans isomers. Computational studies suggest that steric interactions between the pendent methyl group and the phenol ring in the acyclic series determine which structures can best fit the receptor. The chromanol analogue, (3R,4S)-3-[4-(4-fluorophenyl)-4-hydroxypiperidin-1-yl]chroman-4,7-diol (12a , CP-283,097), was found to possess potency and selectivity comparable to CP-101,606. Thus12a is a new tool to explore the function of the NR2B-containing NMDA receptors.- Published
- 1998
8. An impedance-based approach using human iPSC-derived cardiomyocytes significantly improves in vitro prediction of in vivo cardiotox liabilities.
- Author
-
Koci B, Luerman G, Duenbostell A, Kettenhofen R, Bohlen H, Coyle L, Knight B, Ku W, Volberg W, Woska JR Jr, and Brown MP
- Subjects
- Arrhythmias, Cardiac metabolism, Arrhythmias, Cardiac pathology, Arrhythmias, Cardiac physiopathology, Cardiotoxicity, Cell Differentiation, Cell Lineage, Cells, Cultured, Dose-Response Relationship, Drug, Electric Impedance, Heart Rate drug effects, High-Throughput Screening Assays, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Molecular Structure, Myocardial Contraction drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Phenotype, Reproducibility of Results, Risk Assessment, Structure-Activity Relationship, Time Factors, Arrhythmias, Cardiac chemically induced, Biological Assay, Drug Discovery methods, Induced Pluripotent Stem Cells drug effects, Myocytes, Cardiac drug effects, Toxicity Tests methods
- Abstract
Current in vitro approaches to cardiac safety testing typically focus on mechanistic ion channel testing to predict in vivo proarrhythmic potential. Outside of the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative, structural and functional cardiotoxicity related to chronic dosing effects are of great concern as these effects can impact compound attrition. Development and implementation of an in vitro cardiotoxicity screening platform that effectively identifies these liabilities early in the discovery process should reduce costly attrition and decrease preclinical development time. Impedence platforms have the potential to accurately identify structural and functional cardiotoxicity and have sufficient throughput to be included in a multi-parametric optimization approach. Human induced pluripotent stem cell cardiomyocytes (hIPSC-CMs) have demonstrated utility in cardiac safety and toxicity screening. The work described here leverages these advantages to assess the predictive value of data generated by two impedance platforms. The response of hIPSC-CMs to compounds with known or predicted cardiac functional or structural toxicity was determined. The compounds elicited cardiac activities and/or effects on "macro" impedance often associated with overt structural or cellular toxicity, detachment, or hypertrophy. These assays correctly predicted in vivo cardiotox findings for 81% of the compounds tested and did not identify false positives. In addition, internal or literature C
max values from in vivo studies correlated within 4 fold of the in vitro observations. The work presented here demonstrates the predictive power of impedance platforms with hIPSC-CMs and provides a means toward accelerating lead candidate selection by assessing preclinical cardiac safety earlier in the drug discovery process., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
9. Electrical alternans and hemodynamics in the anesthetized guinea pig can discriminate the cardiac safety of antidepressants.
- Author
-
Fossa AA, Gorczyca W, Wisialowski T, Yasgar A, Wang E, Crimin K, Volberg W, and Zhou J
- Subjects
- Animals, Blood Pressure drug effects, Cell Line, Drug Evaluation, Preclinical, ERG1 Potassium Channel, Ether-A-Go-Go Potassium Channels analysis, Guinea Pigs, Humans, Ion Channels physiology, Myocytes, Cardiac drug effects, Myocytes, Cardiac physiology, Patch-Clamp Techniques, Venlafaxine Hydrochloride, Action Potentials drug effects, Antidepressive Agents, Second-Generation pharmacology, Citalopram pharmacology, Cyclohexanols pharmacology, Fluoxetine pharmacology, Ion Channels drug effects
- Abstract
Introduction: The arrhythmogenic risk of fluoxetine, citalopram, and venlafaxine were evaluated through preclinical assays measuring hERG, blood pressure and electrical alternans over their respective clinical unbound concentration ranges., Methods: Anesthetized guinea pigs were instrumented with jugular and carotid cannulae for drug infusion and blood pressure monitoring respectively; a thoracotomy was performed for placement of a monophasic action potential probe on the left ventricle and for placement of pacing wires on the left ventricular apex. Drugs were infused as a 5-min loading dose immediately followed by a 10-min maintenance dose to achieve clinically relevant plasma concentrations; blood samples were taken at the end of each maintenance dose. Ventricular pacing was performed twice at baseline and at each dose level as follows: 50 preconditioning-beats at S1=220 (or 240) ms immediately followed by 30 test-beats at S2=200 ms. This S1-S2 protocol was repeated for S2=190 to 140 ms. HERG and calcium current measurements were recorded in HEK-293 cells stably expressing hERG potassium currents and freshly isolated guinea pig cardiac myocytes using the whole-cell configuration of the patch clamp technique., Results: Physiologically relevant inhibition (IC(20)) of hERG occurred at concentrations 22-fold (fluoxetine), 9-fold (citalopram), and 11-fold (venlafaxine) beyond their respective clinically effective concentration (C(eff)). At the highest achievable levels, fluoxetine (20-fold C(eff)) and citalopram (28-fold C(eff)) significantly decreased heart rate and/or blood pressure as well as increasing electrical alternans by 5 and 18 ms respectively. Venlafaxine increased blood pressure at only 1.3-fold C(eff), but did not increase electrical alternans at the highest achievable dose (3.1-fold C(eff))., Discussion: These data suggest that evaluating other dose limiting side effects in relation to a drug's therapeutic range may be crucial for accurate assessment of arrhythmia liability.
- Published
- 2007
- Full Text
- View/download PDF
10. Synthesis and structure-activity relationships of 6-heterocyclic-substituted purines as inactivation modifiers of cardiac sodium channels.
- Author
-
Estep KG, Josef KA, Bacon ER, Carabateas PM, Rumney S 4th, Pilling GM, Krafte DS, Volberg WA, Dillon K, and Dugrenier N
- Subjects
- Animals, Cats, Guinea Pigs, Heterocyclic Compounds chemistry, Heterocyclic Compounds pharmacology, Humans, In Vitro Techniques, Magnetic Resonance Spectroscopy, Male, Myocardial Contraction drug effects, Purines chemistry, Purines pharmacology, Rabbits, Structure-Activity Relationship, Xenopus, Heart drug effects, Heterocyclic Compounds chemical synthesis, Myocardium chemistry, Purines chemical synthesis, Sodium Channel Blockers
- Abstract
Purine-based analogs of SDZ 211-500 (5) were prepared and evaluated as inactivation modifiers of guinea pig or human cardiac sodium (Na) channels expressed in Xenopus oocytes. Substances which remove or slow the Na channel inactivation process in cardiac tissue are anticipated to prolong the effective refractory period and increase inotropy and thus have potential utility as antiarrhythmic agents. Heterocyclic substitution at the 6-position of the purine ring resulted in compounds with increased Na activity and potency, with 5-membered heterocycles being optimal. Only minor modifications to the benzhydrylpiperazine side chain were tolerated. Selected compounds which delayed the inactivation of Na channels were found to increase refractoriness and contractility in a rabbit Langendorff heart model, consistent with the cellular mechanism. Activity in both the oocyte and rabbit heart assays was specific to the S enantiomers. Preliminary in vivo activity has been demonstrated following intravenous infusion. The most promising compound on the basis of in vitro data is the formylpyrrole (S)-74, which is 25-fold more potent than DPI 201-106 (1) in the human heart Na channel assay.
- Published
- 1995
- Full Text
- View/download PDF
11. Novel inhibitors of potassium ion channels on human T lymphocytes.
- Author
-
Michne WF, Guiles JW, Treasurywala AM, Castonguay LA, Weigelt CA, Oconnor B, Volberg WA, Grant AM, Chadwick CC, and Krafte DS
- Subjects
- Charybdotoxin, Computer Simulation, Humans, Iodine Radioisotopes, Kinetics, Membrane Potentials drug effects, Models, Molecular, Potassium Channels metabolism, Scorpion Venoms metabolism, Structure-Activity Relationship, Potassium Channels drug effects, Quinolines chemical synthesis, Quinolines pharmacology, T-Lymphocytes chemistry, T-Lymphocytes drug effects
- Abstract
The in vitro biological characterization of a series of 4-(alkylamino)-1,4-dihydroquinolines is reported. These compounds are novel inhibitors of voltage-activated n-type potassium ion (K+) channels in human T lymphocytes. This series, identified from random screening, was found to inhibit [125I]charybdotoxin binding to n-type K+ channels with IC50 values ranging from 10(-6) to 10(-8) M. These analogs also inhibit whole cell n-type K+ currents with IC50 values from 10(-5) to 10(-7) M. The preparation of a series of new 4-(alkylamino)-1,4-dihydroquinolines is described. Structure-activity relationships are discussed. Naphthyl analog 7c, the best compound prepared, exhibited > 100-fold selectivity for inhibition of [125I]charybdotoxin binding to n-type K+ channels compared with inhibition of [3H]dofetilide binding to cardiac K+ channels. These compounds represent a potent and selective series of n-type K+ channel inhibitors that have the potential for further development as anti-inflammatory agents.
- Published
- 1995
- Full Text
- View/download PDF
12. Stable expression and functional characterization of a human cardiac Na+ channel gene in mammalian cells.
- Author
-
Krafte DS, Volberg WA, Rapp L, Kallen RG, Lalik PH, and Ciccarelli RB
- Subjects
- Amino Acid Sequence, Animals, Antibodies pharmacology, CHO Cells, Cricetinae, Electrophysiology, Gene Transfer Techniques, Humans, Mammals, Molecular Sequence Data, Peptides chemical synthesis, Peptides immunology, RNA, Messenger analysis, Sodium Channels biosynthesis, Sodium Channels chemistry, Sodium Channels metabolism, Myocardium metabolism, Sodium Channels genetics
- Abstract
In order to develop mammalian cell lines expressing a functional human heart Na+ channel gene (hH1), Chinese hamster ovary (CHO-K1) cells and HeLa cells were transfected with the hH1 gene and the bacterial neomycin (neo) resistance gene. In CHO-K1 cells, direct screening for hH1-positive, G418-resistant colonies by functional patch clamp analysis was complicated due to low-level endogenous expression of a brain-type Na+ channel. Therefore, we developed a stepwise strategy for isolation of cell lines expressing functional hH1 Na+ channels: G418-resistant colonies were sequentially analysed for (1) chromosomal integration of hH1 DNA by PCR, (2) specific hH1 mRNA expression by RT-PCR, (3) hH1 protein production by immunoprecipitation with hH1-specific antisera, and (4) hH1 Na+ channel function by patch-clamp analysis. Using this strategy we obtained two CHO-K1 cell lines which express functional human heart Na+ channels. However, using the same strategy, we were unsuccessful in obtaining functional, hH1-positive HeLa cell lines, even though hH1 mRNA and protein was produced in these cells. The two CHO-K1 cell lines stably express human cardiac Na+ channels which retain normal electrophysiological characteristics with respect to activation and inactivation. In addition, the Na+ channels expressed in these cells are blocked by tetrodotoxin with an IC50 value of 2.5 microM; consistent with known cardiac Na+ channel pharmacology. The density of channels is high enough to permit recording of pseudomacroscopic currents in excised outside-out patches of membrane.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1995
- Full Text
- View/download PDF
13. Voltage dependence of cardiac delayed rectifier block by methanesulfonamide class III antiarrhythmic agents.
- Author
-
Krafte DS and Volberg WA
- Subjects
- Action Potentials drug effects, Animals, Dose-Response Relationship, Drug, Electrophysiology, Guinea Pigs, Heart Ventricles cytology, In Vitro Techniques, Phenethylamines pharmacology, Piperidines pharmacology, Procainamide analogs & derivatives, Procainamide pharmacology, Pyridines pharmacology, Sotalol pharmacology, Stereoisomerism, Sulfonamides pharmacology, Ventricular Function, Anti-Arrhythmia Agents pharmacology, Heart Ventricles drug effects
- Abstract
Voltage-clamp experiments were performed on isolated guinea pig ventricular myocytes to examine the voltage dependence of delayed rectifier block by methanesulfonamide channel blockers. Voltage-dependent channel block, in which block decreases as membrane potential is made more positive, could contribute to the phenomenon of reverse use dependence, in which the magnitude of the drug-induced prolongation in action potential duration (APD) is inversely proportional to stimulation rate. To determine whether such a voltage dependence exists, concentration-response curves were generated for dofetilide, E-4031, sematilide, and D,L-sotalol at test potentials ranging from 0 to 60 mV. All agents blocked current in a manner consistent with selective blockade of the rapidly activating component of delayed rectifier current, IKr. The rank order of potency was E-4031 approximately dofetilide > sematilide > sotalol. Block of tail currents by this class of compounds was more potent after test potentials to +60 mV than after those < or = 0-10 mV. These data are inconsistent with voltage-dependent channel block being a contributing factor to reverse use-dependence and suggest that other mechanisms must be responsible for this phenomenon.
- Published
- 1994
- Full Text
- View/download PDF
14. Properties of endogenous voltage-dependent sodium currents in Xenopus laevis oocytes.
- Author
-
Krafte DS and Volberg WA
- Subjects
- Animals, Electrophysiology, Microelectrodes, RNA pharmacology, Tetrodotoxin pharmacology, Xenopus laevis, Oocytes metabolism, Sodium Channels drug effects
- Abstract
Endogenous voltage-dependent sodium currents were recorded using standard 2-microelectrode techniques in Xenopus laevis oocytes. Maximal inward current occurred at -10 mV with an average amplitude of -279 +/- 17 nA and steady-state inactivation was half-maximal at a voltage of -38 +/- 0.5 mV. Currents were blocked by low concentrations of tetrodotoxin (TTX) with an IC50 value of 6 nM. These properties make the endogenous sodium current in Xenopus oocytes similar to sodium currents expressed following injection of mammalian brain RNA. While endogenous sodium channels have the potential to complicate analysis when using the oocyte expression system, they are only present at significant levels in rare batches of oocytes (less than 5%). Our results do stress the need, however, to reproduce results from exogenous expression studies across several batches of oocytes from different donors.
- Published
- 1992
- Full Text
- View/download PDF
15. Inhibition of calmodulin and protein kinase C by amiodarone and other class III antiarrhythmic agents.
- Author
-
Silver PJ, Connell MJ, Dillon KM, Cumiskey WR, Volberg WA, and Ezrin AM
- Subjects
- Actomyosin metabolism, Amiodarone analogs & derivatives, Animals, Bepridil pharmacology, Calcium metabolism, Guinea Pigs, Imidazoles pharmacology, Kinetics, Male, Phosphodiesterase Inhibitors pharmacology, Quaternary Ammonium Compounds pharmacology, Sotalol pharmacology, Amiodarone pharmacology, Anti-Arrhythmia Agents pharmacology, Calmodulin antagonists & inhibitors, Protein Kinase C antagonists & inhibitors
- Abstract
Class III antiarrhythmic agents may prolong refractoriness via modulation of ion channels, which may be sensitive to Ca2+ regulatory proteins or enzymes. Accordingly, the purpose of this study was to quantitate the effects of several structurally diverse class III antiarrhythmic agents on calmodulin-regulated enzymes and protein kinase C activity, and to evaluate the ability of these agents and known calmodulin antagonists to prolong cardiac refractoriness in vivo. The rank order of potency (IC50;microM) of selected class III antiarrhythmic agents and reference calmodulin antagonists as inhibitors of calmodulin-regulated phosphodiesterase activity were: calmidazolium (0.12 microM) greater than amiodarone (0.62 microM) greater than desethylamiodarone (1.5 microM) greater than trifluoperazine (4.3 microM), bepridil (5 microM) greater than W-7 (7.5 microM), clofilium (13 microM). Similar concentration-related inhibition was evident in a second calmodulin-regulated system, inhibition of myosin light-chain phosphorylation and superprecipitation of arterial actomyosin. Sotalol and tetraethylammonium were inactive at 100 microM. Protein kinase C activity was also inhibited by some of these agents; desethylamiodarone (IC50 = 11 microM) was more potent than the reference agent, H-7 (IC50 = 79 microM), or amiodarone (38% inhibition at 100 microM) and clofilium (32% inhibition at 100 microM). In vivo, the minimally effective doses required to increase ventricular effective refractory periods in paced guinea pigs were (in mg/kg) bepridil, sotalol [1] greater than clofilium [3] greater than amiodarone [10] greater than W-7, desethylamiodarone [20]. No changes in refractory period were noted with maximum testable doses of calmidazolium or trifluoperazine. These studies show that some, but not all, class III antiarrhythmic agents are effective and potent calmodulin antagonists or protein kinase C inhibitors. Moreover, some calmodulin antagonists are effective at prolonging refractoriness in vivo. However, a lack of correlation between these agents suggests that these mechanisms are not solely responsible for the prolongation of refractoriness of all class III agents.
- Published
- 1989
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.