Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jose H. Woodhead is active.

Publication


Featured researches published by Jose H. Woodhead.


Epilepsy Research | 1997

Topiramate enhances GABA-mediated chloride flux and GABA-evoked chloride currents in murine brain neurons and increases seizure threshold

H. Steve White; S.Douglas Brown; Jose H. Woodhead; Skeen Ga; Harold H. Wolf

The anticonvulsant topiramate is effective in laboratory animals against maximal electroshock seizures, amygdala kindling, and spike-wave discharges and has demonstrated efficacy in humans for the treatment of complex partial seizures. However, its mechanism of action has yet to be clearly elucidated. When the chloride-sensitive fluorescent probe N-(ethoxycarbonylmethyl)-6-methoxyquinolinium bromide (MQAE) was used as a tool for estimating the effect of anticonvulsant drugs on GABA receptor function, topiramate was observed to enhance GABA-stimulated chloride (Cl-) flux. At a therapeutic concentration, topiramate (10 microM) enhanced GABA-stimulated (10 microM) Cl- influx into cerebellar granule neurons but did not significantly increase Cl- influx alone. Phenytoin (10 microM) and acetazolamide (300 microM) did not enhance GABA-stimulated Cl- influx. In patch-clamp electrophysiological studies, topiramate also enhanced GABA-evoked whole cell Cl- currents in mouse cerebral cortical neurons in culture. In vivo anticonvulsant studies confirmed that topiramate, like phenytoin, is primarily effective against tonic extension seizures induced by maximal electroshock and is ineffective against clonic seizures induced by the subcutaneously administered chemoconvulsants pentylenetetrazol (PTZ), bicuculline (Bic), and picrotoxin (Pic). In contrast to phenytoin, topiramate, at a dose equivalent to the MES median effective dose (ED50), was found to elevate seizure threshold as estimated by the intravenous PTZ seizure threshold test. Taken together these results support the conclusion that enhancement of GABA-mediated Cl- flux may represent one mechanism that contributes to the anticonvulsant activity of topiramate.


Epilepsia | 2000

Topiramate Modulates GABA-Evoked Currents in Murine Cortical Neurons by a Nonbenzodiazepine Mechanism

White Hs; Brown Sd; Jose H. Woodhead; Skeen Ga; Harold H. Wolf

Purpose: These studies further investigate the ability of topiramate (TPM) to enhance γ‐aminobutyric acid (GABA)‐mediated inhibition through a benzodiazepine‐insensitive pathway.


Journal of Pharmaceutical Sciences | 2000

A Self‐Complementary, Self‐Assembling Microsphere System: Application for Intravenous Delivery of the Antiepileptic and Neuroprotectant Compound Felbamate

Huiling Lian; Solomon S. Steiner; R. Duane Sofia; Jose H. Woodhead; Harold H. Wolf; H. Steve White; Gregory S. Shen; Christopher A. Rhodes; R. Tyler McCabe

Felbamate (FBM) is a novel antiepileptic drug (AED) and neuroprotectant (NP) compound that interacts with strychnine-insensitive (SI) glycine receptors in brain (IC(50) = 374 microM). FBM concentrations required to interact with SI glycine receptors are consistent with brain levels following oral and intraperitoneal administration of AED and NP doses. Because of the solubility limits of FBM, an intravenous (iv) form has not been developed. Nevertheless, an iv form could be important for the treatment of disorders such as status epilepticus and neuronal damage due to hypoxic/ischemic events. Substituted diketopiperazines precipitate in acid to form microspherical particles of uniform size ( approximately 2 microm). The microsphere system entraps drugs on precipitation and dissolves near physiological pH to release the drug cargo. Therefore, microspheres were used to produce an iv formulation of FBM. Mice were administered the FBM/microsphere (20-60 mg/kg FBM) and tested for protection against tonic extension seizures using maximal electroshock. The FBM/microsphere was effective in a time- and dose-dependent manner following iv administration. The median effective dose (ED(50)) for protection against MES seizures at 30 min was 27.2 mg/kg [95% confidence interval (CI) = 20.8-33.4, slope = 6.5]. The ED(50) for minimal motor impairment at 30 min was 167 mg/kg (95% CI = 155-177, slope = 28.1). Thus, the feasibility of encapsulating FBM or similar aqueous insoluble compounds in a microsphere system with delivery by the iv route for treatment of epilepsy and various central nervous system disorders has been clearly demonstrated. Studies were performed in accordance with the Guide for the Care and Use of Laboratory Animals.


Epilepsia | 2002

Anticonvulsant profile and teratogenicity of N-methyl-tetramethylcyclopropyl carboxamide: A new antiepileptic drug

Nina Isoherranen; H. Steve White; Richard H. Finnell; Boris Yagen; Jose H. Woodhead; Gregory D. Bennett; Karen S. Wilcox; Matthew E. Barton; Meir Bialer

Summary:  Purpose: The studies presented here represent our efforts to investigate the anticonvulsant activity of N‐methyl‐tetramethylcyclopropyl carboxamide (M‐TMCD) and its metabolite tetramethylcyclopropyl carboxamide (TMCD) in various animal (rodent) models of human epilepsy, and to evaluate their ability to induce neural tube defects (NTDs) and neurotoxicity.


Epilepsia | 2001

Anticonvulsant Profile of Valrocemide (TV1901): A New Antiepileptic Drug

Nina Isoherranen; Jose H. Woodhead; H. Steve White; Meir Bialer

Summary:  Purpose: We sought to investigate the anticonvulsant activity of the new antiepileptic drug (AED), valrocemide or TV1901 (VGD) in various animal (rodent) models of human epilepsy to determine its anticonvulsant profile and safety margin.


Epilepsy Research | 1990

Preclinical profile of remacemide: A novel anticonvulsant effective against maximal electroshock seizures in mice

Mary L. Stagnitto; Gene C. Palmer; J.Mark Ordy; Ronald C. Griffith; James J. Napier; C.N. Becker; Robert J. Gentile; George E. Garske; Jerry M. Frankenheim; Jose H. Woodhead; H. Steve White; Ewart A. Swinyard

Anticonvulsant tests in mice revealed specific, potent actions of remacemide for protection of mice against maximal electroshock seizures (MES). Comparisons of oral efficacy to reference compounds yielded the following ED50 values (expressed as mg/kg): remacemide = 33, phenytoin = 11, phenobarbital = 20, carbamazepine = 13 and valproate = 631. The duration for protection by remacemide was longer than carbamazepine or valproate, but shorter than phenytoin or phenobarbital. In neural impairment tests (inverted screen or rotorod) to determine the oral toxic dose 50 (TD50) the following therapeutic indices (TD50/ED50) were obtained: (1) inverted screen--remacemide = 17.6, phenytoin = 57.4, phenobarbital = 5.1, carbamazepine = 10.2, and valproate = greater than 3; and (2) rotorod--remacemide = 5.6, phenytoin = 9.6, phenobarbital 4.8, and valproate = 1.9. Remacemide was devoid of sedative actions and possessed a favorable 28.1 margin of safety value (median estimated lethal dose/ED50 for MES). An intermediate potency against either audiogenic- or N-methyl-D-aspartate-induced seizures was exhibited by remacemide. Tolerance to MES was not apparent after 5 days of oral daily dosing of remacemide. Remacemide was inactive in vitro against gamma-aminobutyrate or benzodiazepine receptors and adenosine uptake mechanisms. Therapeutic utility for generalized tonic/clonic seizures is predicted for remacemide.


Pharmaceutical Research | 2003

Pharmacokinetic-Pharmacodynamic Relationships of (2S,3S)-Valnoctamide and Its Stereoisomer (2R,3S)-Valnoctamide in Rodent Models of Epilepsy

Nina Isoherranen; H. Steve White; Brian D. Klein; Michael Roeder; Jose H. Woodhead; Volker Schurig; Boris Yagen; Meir Bialer

AbstractPurpose. Racemic valnoctamide (VCD) is a central nervous system- active drug commercially available in Europe. VCD possesses two chiral centers and, therefore, it exists as a mixture of four stereoisomers. The purpose of this study was to evaluate the anticonvulsant activity of two VCD stereoisomers in comparison with VCD (racemate), valpromide (VPD), and valproic acid (VPA) and to study their pharmacokinetic-pharmacodynamic relationships. Methods. The ability of racemic VCD, (2S,3S)-VCD, (2R,3S)-VCD and VPD to block partial seizures was studied in the 6Hz psychomotor seizure model in mice and in the hippocampal kindled rat. The ability of (2S,3S)-VCD and (2R,3S)-VCD to prevent generalized seizures was evaluated in the maximum electroshock (MES) and subcutaneous metrazole (sc Met) seizure tests. The PK of (2S,3S)-VCD, (2R,3S)-VCD, and VPD was studied in the mice utilized in the 6Hz model. Results. All of the tested compounds were effective in the models tested. No significant difference in ED50 values was observed but the plasma and brain EC50 values of (2R,3S)-VCD in the 6Hz model at 32 mA stimulation were 2-fold higher than the EC50 values of (2S,3S)-VCD. An excellent pharmacokinetic-pharmacodynamic correlation was found between the plasma and brain concentrations of the VCD stereoisomers and their anticonvulsant effect in mice. Stereoselectivity was observed in clearance, volume of distribution, and in brain-to-plasma AUC ratio at a dose of 25 mg/kg, but the difference disappeared at higher doses as the clearance of the stereoisomers decreased and their half-life increased. For (2R,3S)-VCD the brain-to-plasma AUC ratio doubled at the tested dose range, while it remained constant for (2S,3S)-VCD. Conclusions. Racemic VCD, VPD, (2R,3S)-VCD, and (2S,3S)-VCD are effective anticonvulsants in animal models of partial seizures and are more potent than VPA. The more favorable brain penetration of (2S,3S)-VCD and its lower EC50 value in the 6Hz test provides one advantage over (2R,3S)-VCD as a new antiepileptic drug.


British Journal of Pharmacology | 2003

Characterization of the anticonvulsant profile and enantioselective pharmacokinetics of the chiral valproylamide propylisopropyl acetamide in rodents

Nina Isoherranen; Boris Yagen; Jose H. Woodhead; Ofer Spiegelstein; Simcha Blotnik; Karen S. Wilcox; Richard H. Finnell; Gregory D. Bennett; H. Steve White; Meir Bialer

Propylisopropyl acetamide (PID) is a new chiral amide derivative of valproic acid. The purpose of this study was to evaluate the anticonvulsant activity of PID in rodent models of partial, secondarily generalized and sound‐induced generalized seizures which focus on different methods of seizure induction, both acute stimuli, and following short‐term plastic changes as a result of kindling, and to assess enantioselectivity and enantiomer–enantiomer interactions in the pharmacokinetics and pharmacodynamics of racemic PID and its pure enantiomers in rodents. Anticonvulsant activity of (S)‐PID, (R)‐PID and racemic PID was evaluated in the 6 Hz psychomotor seizure model in mice, in the hippocampal kindled rat, and in the Frings audiogenic seizure susceptible mouse. The pharmacokinetics of (S)‐PID and (R)‐PID was studied in mice and rats. In mice (S)‐PID, (R)‐PID and racemic PID were effective in preventing the 6 Hz seizures with (R)‐PID being significantly (P<0.05) more potent (ED50 values 11 mg kg−1, 46 mg kg−1 and 57 mg kg−1 at stimulation intensities of 22, 32 and 44 mA, respectively) than (S)‐PID (ED50 values 20 mg kg−1, 73 mg kg−1 and 81 mg kg−1 at stimulation intensities of 22, 32 and 44 mA, respectively). (S)‐PID, (R)‐PID and racemic PID also blocked generalized seizures in the Frings mice (ED50 values 16 mg kg−1, 20 mg kg−1 and 19 mg kg−1 respectively). In the hippocampal kindled rat a dose of 40 mg kg−1 of (R)‐ and (S)‐PID prevented the secondarily generalized seizure, whereas racemic PID also blocked the expression of partial seizures following an i.p. dose of 40 mg kg−1. Racemic PID also significantly increased the seizure threshold in this model. Mechanistic studies showed that PID did not affect voltage‐sensitive sodium channels or kainate‐, GABA‐ or NMDA‐ evoked currents. The pharmacokinetics of PID was enantioselective following i.p. administration of individual enantiomers to mice, with (R)‐PID having lower clearance and longer half‐life than (S)‐PID. In rats and mice, no enantioselectivity in the pharmacokinetics of PID was observed following administration of the racemate, which may be due to enantiomer–enantiomer interaction. This study demonstrated that PID has both enantioselective pharmacokinetics and pharmacodynamics. The better anticonvulsant potency of (R)‐PID in comparison to (S)‐PID may be due to its more favorable pharmacokinetic profile. The enhanced efficacy of the racemate over the individual enantiomers in the kindled rat may be explained by a pharmacokinetic enantiomer–enantiomer interaction in rats. This study also showed the importance of studying the pharmacokinetics and pharmacodynamics of chiral drugs following administration of the individual enantiomers as well as the racemic mixture.


Epilepsia | 1987

The Effect of Chronic Felbamate Administration on Anticonvulsant Activity and Hepatic Drug-Metabolizing Enzymes in Mice and Rats

Ewart A. Swinyard; Jose H. Woodhead; Michael R. Franklin; R. Duane Sofia; Harvey J. Kupferberg

Summary: The possibility of tolerance development from chronic administration of felbamate (FBM) was investigated in mice and rats. Chronic administration (15 days) of FBM (150 mg/kg i.p.) in mice had no significant effect on either intravenous pentylenetetrazol (PTZ) seizure threshold or hexobarbital sleep time; however, hexobarbital sleep time was significantly increased after a single dose. Chronic administration (5–7 days) of FBM (48 or 95 mg/kg orally) in rats also had no significant effect on either maximal electroshock seizure activity or hexobarbital sleep time. Chronic administration of FBM at 238 mg/kg slightly decreased anti‐subcutaneous PTZ activity in chronically treated rats (one of eight protected) as compared with those receiving only a single dose (three of eight protected), but there was no significant change in hexobarbital sleep time. Chronic treatment of rats for 7 days with 48 mg/kg had no significant effect on any hepatic parameters. However, 95 or 238 mg/kg of FBM significantly increased p‐nitroanisole O‐demethylase activity. It is concluded that the increased hexobarbital sleep time induced by an acute dose of FBM reflects the CNS‐depressant effect of the substance. The increased p‐nitroanisole O‐demethylase activity observed after chronic administration may be indicative of some liver microsomal induction. Overall, FBM in doses ranging from 48 to 238 mg/kg appears to have minimal potential for tolerance development.


Epilepsy Research | 1991

Preclinical profile of stereoisomers of the anticonvulsant remacemide in mice

Gene C. Palmer; Mary L. Stagnitto; J.Mark Ordy; Ronald C. Griffith; James J. Napiera; Robert J. Gentile; Jose H. Woodhead; H. Steve White; Ewart A. Swinyard

Stereoisomers of remacemide (racemate form) were compared for anticonvulsant efficacy and safety in mice. In the maximal electroshock seizure (MES) test for oral efficacy, the (-) stereoisomer, FPL 14145, was more potent than the racemate or the (+) stereoisomer, FPL 14144. Respective ED50 values (expressed as mg/kg) were: remacemide, 58; FPL 14145, 45; FPL 14144, 79. In 2 of 3 tests for neural impairment, FPL 14145 yielded significantly better therapeutic indices (toxic dose 50/ED50) than the racemate. The margin of safety (estimated median lethal dose ED50) was more favorable for FPL 14144: remacemide, 15.1; FPL 14144, 18.9; FPL 14145, 15.7. The duration of protection against MES indicated the stereoisomers were longer acting than the racemate. After intravenous administration the order of potency against MES was similar: FPL 14145 greater than remacemide greater than FPL 14144. Following daily administration of the oral ED98 for 4 days, with a dose response curve run on day 5, the MES ED50 values for all compounds were increased. The test indicates tolerance. In the pentylenetetrazol infusion test the racemate and FPL 14144 demonstrated more proconvulsant properties than FPL 14145. Intraperitoneal administration of 50 mg/kg or more produced changes in behavior with all compounds. At higher doses the racemate and FPL 14145 elicited more severe symptoms with death at 200 mg/kg.

Collaboration


Dive into the Jose H. Woodhead's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meir Bialer

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Nina Isoherranen

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Boris Yagen

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge