Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Science ; 313(5794): 1760-3, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16917025

ABSTRACT

Carbon supercapacitors, which are energy storage devices that use ion adsorption on the surface of highly porous materials to store charge, have numerous advantages over other power-source technologies, but could realize further gains if their electrodes were properly optimized. Studying the effect of the pore size on capacitance could potentially improve performance by maximizing the electrode surface area accessible to electrolyte ions, but until recently, no studies had addressed the lower size limit of accessible pores. Using carbide-derived carbon, we generated pores with average sizes from 0.6 to 2.25 nanometer and studied double-layer capacitance in an organic electrolyte. The results challenge the long-held axiom that pores smaller than the size of solvated electrolyte ions are incapable of contributing to charge storage.

2.
Fitoterapia ; 75(3-4): 309-14, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15158987

ABSTRACT

The decoction of Mimosa pudica leaves given intraperitoneally at dose of 1000-4000 mg/kg protected mice against pentylentetrazol and strychnine-induced seizures. M. pudica had no effect against picrotoxin-induced seizures It also antagonized N-methyl-D-aspartate- induced turning behavior. These properties could explain its use in African traditional medicine.


Subject(s)
Anticonvulsants/pharmacology , Mimosa , Phytotherapy , Plant Extracts/pharmacology , Seizures/prevention & control , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/therapeutic use , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Medicine, African Traditional , Mice , Pentylenetetrazole , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Plant Leaves , Seizures/chemically induced , Strychnine
3.
J Neurosci ; 21(22): 8734-45, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698585

ABSTRACT

To study the role of mGlu7 receptors (mGluR7), we used homologous recombination to generate mice lacking this metabotropic receptor subtype (mGluR7(-/-)). After the serendipitous discovery of a sensory stimulus-evoked epileptic phenotype, we tested two convulsant drugs, pentylenetetrazole (PTZ) and bicuculline. In animals aged 12 weeks and older, subthreshold doses of these drugs induced seizures in mGluR7(-/-), but not in mGluR7(+/-), mice. PTZ-induced seizures were inhibited by three standard anticonvulsant drugs, but not by the group III selective mGluR agonist (R,S)-4-phosphonophenylglycine (PPG). Consistent with the lack of signs of epileptic activity in the absence of specific stimuli, mGluR7(-/-) mice showed no major changes in synaptic properties in two slice preparations. However, slightly increased excitability was evident in hippocampal slices. In addition, there was slower recovery from frequency facilitation in cortical slices, suggesting a role for mGluR7 as a frequency-dependent regulator in presynaptic terminals. Our findings suggest that mGluR7 receptors have a unique role in regulating neuronal excitability and that these receptors may be a novel target for the development of anticonvulsant drugs.


Subject(s)
Genetic Predisposition to Disease , Glycine/analogs & derivatives , Receptors, Metabotropic Glutamate/deficiency , Seizures/genetics , Animals , Anticonvulsants/pharmacology , Bicuculline , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Convulsants , Drug Resistance/genetics , Electroencephalography , Excitatory Amino Acid Agonists/pharmacology , Gene Targeting , Glycine/pharmacology , Hippocampus/drug effects , Hippocampus/physiopathology , Homozygote , In Vitro Techniques , Mice , Mice, Knockout , Pentylenetetrazole , Phenotype , Physical Chromosome Mapping , Receptors, Metabotropic Glutamate/genetics , Seizures/chemically induced , Seizures/physiopathology , Seizures/prevention & control
4.
J Ethnopharmacol ; 76(2): 145-50, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390127

ABSTRACT

The methanolic extract of rhizomes of Cyperus articulatus, a plant used in traditional medicine in Africa and Latin America for many diseases, possesses anticonvulsant activity in mice. This extract protected mice against maximal electroshock (MES)- and pentylenetetrazol (PTZ)-induced seizures. It also delayed the onset of seizures induced by isonicotinic acid hydrazide and strongly antagonized N-methyl-D-aspartate-induced turning behavior. The ED(50) for protection against seizures was 306 (154-541) mg/kg intraperitoneally (i.p.) for the PTZ test and 1005 (797-1200) mg/kg i.p. for the MES test. The ED(50) of methanolic extract against N-methyl-D-aspartate-induced turning behavior was 875 (623-1123) mg/kg i.p. C. articulatus L. methanolic extract protected 54% of mice from seizures induced by strychnine at the dose of 1000 mg/kg i.p. but had no or a moderate effect only against picrotoxin- or bicuculline-induced seizures. With these effects, the rhizome of C. articulatus L. possesses anticonvulsant properties in animals that might explain its use as a traditional medicine for epilepsy in Africa.


Subject(s)
Anticonvulsants/therapeutic use , Medicine, Traditional , Plant Extracts/therapeutic use , Seizures/prevention & control , Animals , Anticonvulsants/isolation & purification , Behavior, Animal/drug effects , Convulsants/toxicity , Electroshock , Male , Mice , N-Methylaspartate/antagonists & inhibitors , Pentylenetetrazole/toxicity , Plant Extracts/isolation & purification , Seizures/chemically induced
5.
J Pharmacol Exp Ther ; 289(3): 1678-87, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10336568

ABSTRACT

Group III metabotropic glutamate receptors (mGluRs) are thought to modulate neurotoxicity of excitatory amino acids, via mechanisms of presynaptic inhibition, such as regulation of neurotransmitter release. Here, we describe (R,S)-4-phosphonophenylglycine (PPG) as a novel, potent, and selective agonist for group III mGluRs. In recombinant cell lines expressing the human receptors hmGluR4a, hmGluR6, hmGluR7b, or hmGluR8a, EC50 values for (R,S)-PPG of 5.2 +/- 0.7 microM, 4.7 +/- 0.9 microM, 185 +/- 42 microM, and 0.2 +/- 0.1 microM, respectively, were measured. The compound showed EC50 and IC50 values of >/=200 microM at group I and II hmGluRs and was inactive at cloned human N-methyl-D-aspartate, alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionate, and kainate receptors (>300 microM). On the other hand, it showed micromolar affinity for a Ca2+/Cl--dependent L-glutamate binding site in rat brain, similar to other phosphono-substituted amino acids like L-2-amino-4-phosphonobutyrate. In cultured cortical neurons, (R, S)-PPG provided protection against a toxic pulse of N-methyl-D-aspartate (EC50 = 12 microM), which was reversed by the group III mGluR antagonist (R,S)-alpha-methylserine-O-phosphate but not by the group II antagonist (2S)-alpha-ethylglutamate. Moreover, (R,S)-PPG protected against N-methyl-D-aspartate- and quinolinic acid-induced striatal lesions in rats and was anticonvulsive in the maximal electroshock model in mice. In contrast to the group III mGluR agonists L-2-amino-4-phosphonobutyrate and L-serine-O-phosphate, (R,S)-PPG showed no proconvulsive effects (2200 nmol i.c.v.). These data provide novel in vivo evidence for group III mGluRs as attractive targets for neuroprotective and anticonvulsive therapy. Also, (R,S)-PPG represents an attractive tool to analyze the roles of group III mGluRs in nervous system physiology and pathology.


Subject(s)
Anticonvulsants/pharmacology , Brain/metabolism , Glycine/analogs & derivatives , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Animals , Cell Line , Cell Membrane/metabolism , Colforsin/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiology , Cyclic AMP/metabolism , Electroshock , Glutamic Acid/metabolism , Glycine/chemistry , Glycine/pharmacology , Humans , Kinetics , Male , Mice , Mice, Inbred Strains , N-Methylaspartate/toxicity , Phosphatidylinositols/metabolism , Protein Isoforms/agonists , Protein Isoforms/genetics , Quinolinic Acid/toxicity , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Recombinant Proteins/agonists , Second Messenger Systems/drug effects , Seizures/physiopathology , Seizures/prevention & control , Structure-Activity Relationship
6.
J Neurosci ; 17(12): 4688-99, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9169529

ABSTRACT

Protease nexin-1 (PN-1), a member of the serpin superfamily, controls the activity of extracellular serine proteases and is expressed in the brain. Mutant mice overexpressing PN-1 in brain under the control of the Thy-1 promoter (Thy 1/PN-1) or lacking PN-1 (PN-1-/-) were found to develop epileptic activity in vivo and in vitro. Theta burst-induced long-term potentiation (LTP) and NMDA receptor-mediated synaptic transmission in the CA1 field of hippocampal slices were augmented in Thy 1/PN-1 mice and reduced in PN-1-/- mice. Compensatory changes in GABA-mediated inhibition in Thy 1/PN-1 mice suggest that altered brain PN-1 levels lead to an imbalance between excitatory and inhibitory synaptic transmission.


Subject(s)
Carrier Proteins/physiology , Epilepsy/physiopathology , Hippocampus/physiology , Long-Term Potentiation , Neurons/physiology , Amyloid beta-Protein Precursor , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Epilepsy/genetics , Hippocampus/physiopathology , Kinetics , Mice , Mice, Knockout , Mice, Neurologic Mutants , Mice, Transgenic , Promoter Regions, Genetic , Protease Nexins , Receptors, Cell Surface , Recombinant Fusion Proteins/biosynthesis , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/physiology , Synaptic Transmission , Thy-1 Antigens/biosynthesis , Thy-1 Antigens/genetics , Time Factors
7.
Naunyn Schmiedebergs Arch Pharmacol ; 354(2): 164-72, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8857593

ABSTRACT

Lamotrigine, carbamazepine and oxcarbazepine inhibit veratrine-induced neurotransmitter release from rat brain slices in concentrations corresponding to those reached in plasma or brain in experimental animals or humans after anticonvulsant doses, presumably due to their sodium channel blocking properties. Microdialysis measurements of extracellular glutamate and aspartate were carried out in conscious rats in order to investigate whether corresponding effects occur in vivo Veratridine (10 microM) was applied via the perfusion medium to the cortex and the corpus striatum in the presence of the glutamate uptake inhibitor L-trans-pyrrolidine-2,4-dicarboxylic acid (1 mM in perfusion medium). Maximally effective anticonvulsant doses of carbamazepine (30 mg/kg), oxcarbazepine (60 mg/kg) and lamotrigine (15 mg/kg) were given orally. The uptake inhibitor increased extracellular glutamate and aspartate about 2-fold in striatum and about 7-fold and 3-fold, respectively, in cortex. Veratridine caused a further 2-3-fold increase in extracellular glutamate in striatum and cortex, respectively, but its effect on extracellular aspartate was less marked in both areas. None of the anticonvulsant compounds affected the veratridine-induced increases in extracellular glutamate or aspartate in the striatum which were, however, markedly inhibited by tetrodotoxin (1 microM) and thus are sensitive to sodium channel blockade. In the cortex the same drugs at the same doses did cause about 50% inhibition of the veratridine-induced increase in extracellular glutamate. Carbamazepine and to a lesser extent lamotrigine, but not oxcarbazepine, also inhibited the veratridine-induced increase in extracellular aspartate in the cortex. Although our results might seem to support the view that inhibition of glutamate and aspartate release is responsible for the anticonvulsant effects of lamotrigine, carbamazepine and oxcarbazepine, two complementary findings argue against this interpretation. First, as previously shown, inhibition of electrically induced released of glutamate requires 5 to 7 times higher concentrations of these compounds than release elicited by veratrine. Second, the present study indicates that doses totally suppressing convulsions caused no inhibition in the striatum and at best a 50% inhibition in the brain cortex. From this we conclude that the doses used here, although to some extent effective against veratridine, did not suppress the release of GLU and ASP elicited by the normal ongoing electrical activity of the glutamatergic and aspartatergic neurons and that the mechanism of the suppression of convulsions must be sought elsewhere.


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/analogs & derivatives , Carbamazepine/pharmacology , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Glutamic Acid/metabolism , Triazines/pharmacology , Veratridine/pharmacology , Animals , Aspartic Acid/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Lamotrigine , Male , Microdialysis , Oxcarbazepine , Rats
8.
Eur J Pharmacol ; 271(2-3): 301-8, 1994 Dec 27.
Article in English | MEDLINE | ID: mdl-7705430

ABSTRACT

The anticonvulsant compound oxcarbazepine and its principal 10-monohydroxy metabolite protected potently against electroshock-induced tonic hindlimb extension. Maximal plasma concentrations depended on dose and were reached < or = 1 h after an oral dose of oxcarbazepine and < 2 h after monohydroxy derivative. In mice, the ED50 was 14 mg/kg for oxcarbazepine and 20.5 mg/kg for the monohydroxy derivative, p.o. In rats, the ED50 was 13.5 mg/kg for oxcarbazepine and 17.0 mg/kg for monohydroxy derivative, p.o. This protective effect compared favorably with the efficacy of carbamazepine, phenytoin, phenobarbital and diazepam in the same test. As observed previously, valproate and ethosuximide were markedly less potent. The effect of oxcarbazepine and its monohydroxy derivative on sustained high frequency repetitive firing of sodium-dependent action potentials of mouse spinal cord neurons in cell culture was also examined using intracellular recording techniques. Both compounds reduced the percentage of neurons capable of sustained action potential firing in concentration-dependent manner. The EC50 for oxcarbazepine was 5 x 10(-8) M and that for monohydroxy derivative was 2 x 10(-8) M (P > 0.05 vs. oxcarbazepine). For comparison, the EC50 for carbamazepine was significantly higher (6 x 10(-7) M, P < 0.001 vs. oxcarbazepine and monohydroxy derivative). Limitation of firing by oxcarbazepine and the monohydroxy derivative depended on firing frequency and membrane potential and was enhanced by depolarization. Input resistance and resting membrane potential were not altered by either drug. The in vitro effect on action potential firing frequency occurred at concentrations below plasma levels of oxcarbazepine and monohydroxy derivative which protected animals against electroshock and were therapeutically effective in patients.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/analogs & derivatives , Seizures/prevention & control , Action Potentials/drug effects , Animals , Carbamazepine/pharmacology , Cells, Cultured , Male , Mice , Oxcarbazepine , Rats , Sodium/metabolism , Spinal Cord/drug effects , Spinal Cord/physiology
9.
Epilepsia ; 35 Suppl 3: S5-9, 1994.
Article in English | MEDLINE | ID: mdl-8156978

ABSTRACT

The antiepileptic drug (AED) oxcarbazepine (OCBZ) and its rapidly formed 10-monohydroxy metabolite (MHD) protect against electroshock-induced tonic hindlimb extension in rodents (ED50 14-21 mg/kg p.o.). Both stereoisomers of MHD also protect. As with carbamazepine (CBZ), these findings suggest clinical efficacy against generalized tonic-clonic and, to some extent, partial seizures. OCBZ (IC50 5 x 10(-8) M), MHD (IC50 2 x 10 (-8) M), and CBZ (IC50 6 x 10(-7) M) limit the frequency of firing of sodium-dependent action potentials by cultured mouse central neurons and reduce Vmax progressively in a use-dependent manner at concentrations below therapeutic plasma concentrations in OCBZ-treated patients. This suggests that blockade of voltage-sensitive sodium channels could contribute to the antiepileptic efficacy of OCBZ. Blockade of penicillin-induced epileptiform discharges in hippocampal slices by MHD and its stereoisomers was diminished when the potassium channel blocker 4-aminopyridine was added to the bath fluid. This indicates that additional mechanisms of action, e.g., an effect on potassium channels, might be clinically important. In addition, both stereoisomers are equally responsible for the antiepileptic activity of the racemate, i.e., MHD, and are therefore likely to play a therapeutic role. Such actions could confer broad clinical utility on OCBZ.


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/analogs & derivatives , Action Potentials/drug effects , Animals , Anticonvulsants/therapeutic use , Carbamazepine/pharmacology , Carbamazepine/therapeutic use , Cells, Cultured , Disease Models, Animal , Electroshock , Hippocampus/cytology , Hippocampus/drug effects , Humans , Neural Conduction/drug effects , Neurons/drug effects , Neurons/physiology , Oxcarbazepine , Rodentia , Seizures/prevention & control , Spinal Cord/cytology
10.
Antimicrob Agents Chemother ; 36(5): 1115-8, 1992 May.
Article in English | MEDLINE | ID: mdl-1510402

ABSTRACT

The pharmacokinetics and tissue penetration of cefotetan were studied after a single injection of 2 g given intravenously for antimicrobial prophylaxis to 16 consecutive patients undergoing colorectal surgery. Concentrations in tissue greater than or equal to the MIC for 90% of the main pathogens tested were considered adequate. The elimination half-life at beta phase was 4.6 +/- 1.4 h, the total body clearance was 0.75 +/- 0.19 ml/kg/min, and the volume of distribution was 260 +/- 71 ml/kg. At the time of incision (33 +/- 16 min after the injection), cefotetan concentrations were 14.2 +/- 7 micrograms/g in abdominal-wall fat, 16.4 +/- 1 micrograms/g in epiploic fat, and 163 +/- 62 mg/liter in serum. At the time of surgical anastomosis (151 +/- 54 min), cefotetan concentrations were 33.3 +/- 6 micrograms/g in the colonic wall and 73 +/- 34 mg/liter in serum. Upon closure of the abdomen (216 +/- 76 min), cefotetan concentrations were 6.3 +/- 3 micrograms/g in abdominal-wall fat, 6.1 +/- 4 micrograms/g in epiploic fat, and 64 +/- 38 mg/liter in serum. Cefotetan tissue penetration was 10% into abdominal and epiploic fat and 46% into the colonic wall. Levels in tissue were compared with the MIC for 90% of the most frequently encountered pathogenic germs (Staphylococcus aureus, Bacteroides fragilis, and Escherichia coli). Adequate concentrations in tissue were obtained up to anastomosis but not upon closure. The authors therefore recommend the injection of an additional dose of 1 g before closure in order to ensure optimal efficacy throughout the surgical procedure.


Subject(s)
Cefotetan/pharmacokinetics , Colorectal Surgery , Premedication , Cefotetan/administration & dosage , Cefotetan/blood , Humans , Injections, Intravenous , Microbial Sensitivity Tests , Postoperative Complications/prevention & control , Tissue Distribution
12.
Neuroscience ; 47(1): 63-8, 1992.
Article in English | MEDLINE | ID: mdl-1315938

ABSTRACT

The effect of the GABAB receptor blocker CGP 35348 on epileptic processes in vitro and in vivo was studied. In hippocampal slices of the rat maintained in vitro, CGP 35348 (100 microM) induced a moderate increase in the frequency of extracellularly recorded spontaneous epileptiform burst discharges induced in CA3 by penicillin (1.2 mM), bicuculline (5 microM) and low Mg(2+) (0.1 mM). This effect was observed in 50-75% of the slices. A similar but less consistent increase was also observed in CA1 in bicuculline and low Mg2+. Data obtained by intracellular recordings from CA1 pyramidal cells in the presence of bicuculline (10 microM) demonstrated that CGP 35348 (100 microM) increased the duration of the paroxysmal depolarization underlying an evoked epileptiform burst and reduced the early component of the after hyperpolarization which followed the burst. In mice pretreated with isoniazid, CGP 35348 (300 mg/kg, i.p.) significantly increased the number of convulsing mice. However, convulsions induced by submaximal doses of pentylenetetrazol, picrotoxin or strychnine were not facilitated by CGP 35348. We conclude that GABAB receptors appear to exert a suppressant effect on various kinds of epileptiform discharges of hippocampal neurons in vitro. In vivo, however, the role of GABAB receptors in regulating convulsions is less prominent since only isoniazid-induced convulsions were facilitated by GABAB receptor blockade.


Subject(s)
Epilepsy/chemically induced , Organophosphorus Compounds/pharmacology , Receptors, GABA-A/physiology , Animals , Baclofen/pharmacology , Convulsants , Epilepsy/physiopathology , GABA-A Receptor Antagonists , Hippocampus/drug effects , Hippocampus/physiopathology , In Vitro Techniques , Isoniazid/pharmacology , Male , Mice , Mice, Inbred Strains , Rats , Rats, Inbred Strains
13.
Pathol Biol (Paris) ; 39(5): 507-10, 1991 May.
Article in French | MEDLINE | ID: mdl-1881684

ABSTRACT

Pharmacokinetics and tissue penetration of netilmicin were studied after the use of a single dose (6 mg/kg) given for antibioprophylaxis in colo-rectal surgery. Thirteen patients, scheduled for elective surgery, were given 6 mg/kg IV netilmicin over 30 min, together with 1000 mg IV ornidazole. Netilmicin peak serum concentration (10 min after end of infusion) was 24.4 +/- 3.4 mg/l and trough level (24 h) was 0.9 +/- 0.5 mg/l. Plasma elimination half-life was 409 +/- 70 min, le volume apparent volume of distribution was 38 +/- 101 and total body clearance was 0.07 +/- 0.02 ml/min. Adequate netilmicin levels (5 greater than or equal to CMI 90 of involved pathogens Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus) were obtained in 100 per cent of patients in abdominal wall and epiploid fat, at time of opening, and in colonic wall at time of anastomosis. Adequate levels were obtained at time of closure in abdominal wall and epiploid fat in 92 to 100 per cent of patients. In situation of allergy to beta-lactam antibiotics, the use of netilmicin in combination with ornidazole may be recommended.


Subject(s)
Bacterial Infections/prevention & control , Colonic Diseases/surgery , Netilmicin/pharmacokinetics , Rectal Diseases/surgery , Surgical Wound Infection/microbiology , Aged , Female , Fluorescence Polarization , Humans , Male , Middle Aged , Netilmicin/blood , Netilmicin/therapeutic use , Preoperative Care , Surgical Wound Infection/prevention & control
14.
Pathol Biol (Paris) ; 39(5): 515-9, 1991 May.
Article in French | MEDLINE | ID: mdl-1881686

ABSTRACT

The aim of the study was to evaluate, in clinical conditions, the penetration of ciprofloxacin into bronchial secretions. Eight patients were included in the study. They presented with nosocomial pneumonia and were under controlled mechanical ventilation. Ciprofloxacin was given at a dose of 3 mg/kg over 30 min. Serial bronchial and blood samples were obtained over a 12 hour period on day 2 and 4. Ciprofloxacin was measured by HPLC. Serum peak levels were 2.95 +/- 1 mg/l on day 2, and 2.43 +/- 0.7 mg/l on day 4. Bronchial peak and through levels were 0.95 +/- 0.51 mg/l and 0.21 +/- 0.12 mg/l, respectively, on day 2, and 0.76 +/- 0.17 mg/l and 0.18 +/- 0.14 mg/l, respectively, on day 4. The ratio of bronchial/serum peak was 0.32 +/- 0.11 and 0.33 +/- 0.06 on day 2 and 4, respectively. The ratio of AUC 0-12 h in the bronchial secretions/AUC 0-12 h in the serum samples was 0.66 +/- 0.04 and 0.55 +/- 0.27 on day 2 and 4, respectively.


Subject(s)
Bronchial Diseases/drug therapy , Ciprofloxacin/pharmacokinetics , Cross Infection/drug therapy , Sputum/drug effects , Aged , Aged, 80 and over , Bronchial Diseases/metabolism , Bronchial Diseases/microbiology , Ciprofloxacin/blood , Ciprofloxacin/therapeutic use , Cross Infection/metabolism , Cross Infection/microbiology , Diffusion , Female , Humans , Male , Middle Aged , Prospective Studies , Respiration, Artificial , Sputum/metabolism
15.
Naunyn Schmiedebergs Arch Pharmacol ; 342(1): 61-6, 1990 Jul.
Article in English | MEDLINE | ID: mdl-1976233

ABSTRACT

Anticonvulsant properties of CGP 37849 and CGP 39551, two novel phosphono-amino acids which are competitive NMDA receptor antagonists, were examined in rodents. At optimal pretreatment times CGP 37849 suppressed electroshock-induced seizures in mice and rats with ED50s ranging from 8 to 22 mg/kg after oral administration, and 0.4 to 2.4 mg/kg after i.v. and i.p. injection. Relative to CGP 37849, CGP 39551 was more potent after p.o. (ED50 3.7-8.1 mg/kg), and less potent after i.v. or i.p. treatment (ED50 2.7-8.7 mg/kg). Following oral treatment, the duration of action of CGP 37849 was about 8 h, while CGP 39551 still showed good activity after 24 h (ED50 8.7 mg/kg, mouse; 21 mg/kg, rat). Both compounds were anticonvulsant at doses below those at which overt behavioural side effects were apparent. CGP 39551 delayed the development of kindling in rats at doses of 10 mg/kg p.o. and above, and showed weak anticonvulsant activity against pentylenetetrazol-evoked seizures. CGP 37849 and CGP 39551 are the first competitive NMDA antagonists to show oral anti-convulsant properties in a therapeutically-useful dose-range, and hence are interesting candidates for novel antiepileptic therapy in man.


Subject(s)
2-Amino-5-phosphonovalerate/analogs & derivatives , Anticonvulsants , Receptors, Neurotransmitter/antagonists & inhibitors , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Electroshock , Kindling, Neurologic/drug effects , Male , Mice , Mice, Inbred Strains , Pentylenetetrazole/antagonists & inhibitors , Postural Balance/drug effects , Rats , Rats, Inbred Strains , Receptors, N-Methyl-D-Aspartate
16.
Br J Pharmacol ; 99(4): 791-7, 1990 Apr.
Article in English | MEDLINE | ID: mdl-1972895

ABSTRACT

1. The pharmacological properties of CGP 37849 (DL-(E)-2-amino-4-methyl-5-phosphono-3-pentenoic acid; 4-methyl-APPA) and its carboxyethylester, CGP 39551, novel unsaturated analogues of the N-methyl-D-aspartate (NMDA) receptor antagonist, 2-amino-5-phosphonopentanoate (AP5), were evaluated in rodent brain in vitro and in vivo. 2. Radioligand binding experiments demonstrated that CGP 37849 potently (Ki 220 nM) and competitively inhibited NMDA-sensitive L-[3H]-glutamate binding to postsynaptic density (PSD) fractions from rat brain. It inhibited the binding of the selective NMDA receptor antagonist, [3H]-((+/-)-3-(2-carboxypiperazin-4-yl)propyl-1-phosphonate (CPP), with a Ki of 35 nM, and was 4, 5 and 7 fold more potent than the antagonists [+/-)-cis-4-phosphonomethylpiperidine-2-carboxylic acid) (CGS 19755), CPP and D-AP5, respectively. Inhibitory activity was associated exclusively with the trans configuration of the APPA molecule and with the D-stereoisomer. CGP 39551 showed weaker activity at NMDA receptor recognition sites and both compounds were weak or inactive at 18 other receptor binding sites. 3. CGP 37849 and CGP 39551 were inactive as inhibitors of L-[3H]-glutamate uptake into rat brain synaptosomes and had no effect on the release of endogenous glutamate from rat hippocampal slices evoked by electrical field stimulation. 4. In the hippocampal slice in vitro, CGP 37849 selectively and reversibly antagonized NMDA-evoked increases in CA1 pyramidal cell firing rate. In slices bathed in medium containing low Mg2+ levels, concentrations of CGP 37849 up to 10 microM suppressed burst firing evoked in CAl neurones by stimulation of Schaffer collateral-commissural fibres without affecting the magnitude of the initial population spike; CGP 39551 exerted the same effect but was weaker. In vivo, oral administration to rats of either CGP 37849 or CGP 39551 selectively blocked firing in hippocampal neurones induced by ionophoreticallyapplied NMDA, without affecting the responses to quisqualate or kainate. 5. CGP 37849 and CGP 39551 suppressed maximal electroshock-induced seizures in mice with ED50 s of 21 and 4 mg kg'- p.o., respectively. 6. CGP 37849 and CGP 39551 are potent and competitive NMDA receptor antagonists which show significant central effects following oral administration to animals. As such, they may find value as tools to elucidate the roles of NMDA receptors in brain function, and potentially as therapeutic agents for the treatment of neurological disorders such as epilepsy and ischaemic brain damage in man.


Subject(s)
2-Amino-5-phosphonovalerate/analogs & derivatives , Receptors, Neurotransmitter/antagonists & inhibitors , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Anticonvulsants , Binding, Competitive/drug effects , Electrophysiology , Electroshock , Glutamates/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , In Vitro Techniques , Kinetics , Male , Neurotransmitter Agents/metabolism , Radioligand Assay , Rats , Rats, Inbred Strains , Receptors, N-Methyl-D-Aspartate , Receptors, Neurotransmitter/metabolism , Sodium/metabolism , Valine/analogs & derivatives , Valine/pharmacology
17.
Neurosci Lett ; 107(1-3): 184-8, 1989 Dec 15.
Article in English | MEDLINE | ID: mdl-2575726

ABSTRACT

CGP 31358, a novel triazole, inhibited the binding of L-[3H]glutamate and [3H]MK-801 to the N-methyl-D-aspartate (NMDA) receptor complex in rat brain synaptic membrane fractions, and showed anticonvulsant activity in mice. It had no effect on the strychnine-insensitive binding of [3H]glycine. Saturation and Hill analyses indicated that CGP 31358 binds to a site on the NMDA receptor which is separate from, but coupled to, both the transmitter recognition site and the channel domain. Available data indicate that this site is distinct from those with which tricyclic antidepressants and ifenprodil interact. CGP 31358 is a new chemical entity with a novel mechanism of action at the NMDA receptor, and as such may form a tool for understanding the molecular pharmacology of this receptor-channel complex.


Subject(s)
Anticonvulsants/pharmacology , Brain/metabolism , Epilepsy/drug therapy , Receptors, Neurotransmitter/metabolism , Triazoles/pharmacology , Animals , Binding, Competitive , Brain/drug effects , Dibenzocycloheptenes/metabolism , Dizocilpine Maleate , Epilepsy/metabolism , Glutamates/metabolism , Glutamic Acid , Mice , Rats , Receptors, N-Methyl-D-Aspartate , Subcellular Fractions/metabolism , Triazoles/therapeutic use
18.
J Neural Transm ; 71(1): 11-27, 1988.
Article in English | MEDLINE | ID: mdl-3343593

ABSTRACT

CGS 8216, a benzodiazepine-receptor ligand with inverse agonistic properties, and CGS 9896, which possesses partial agonistic or mixed agonist-antagonist properties were compared in a number of epilepsy models. The effect of CGS 9896 on the decrease in GABA levels induced by isoniazid was also investigated. CGS 9896 inhibited the kindling process in rats in that it delayed the development of overt seizures and the increase in the duration of afterdischarges. In a genetic rat model characterized by absence-like EEG patterns, CGS 9896 dose-dependently suppressed these spontaneously occurring discharges, while CGS 8216 had no effect. However, CGS 8216 antagonized the anticonvulsant action of CGS 9896. CGS 9896 protected mice against seizures induced by beta-vinyllactic acid, whereas CGS 8216 shortened the latency period before convulsions occurred. CGS 9896 retarded the onset of convulsive fits caused by isoniazid without preventing the decrease in GABA levels produced by that drug. These results confirm the anticonvulsant activity of CGS 9896 and demonstrate the inverse agonistic activity of CGS 8216. The profile of CGS 9896 in the above tests suggests that it might be an effective anticonvulsant, primarily in absence-type seizures.


Subject(s)
Anticonvulsants/therapeutic use , Brain/metabolism , Epilepsy/drug therapy , Pyrazoles/therapeutic use , gamma-Aminobutyric Acid/metabolism , Animals , Anticonvulsants/pharmacology , Brain/drug effects , Brain/physiopathology , Diazepam , Electric Stimulation , Epilepsy/chemically induced , Epilepsy/metabolism , Male , Pyrazoles/pharmacology , Rats , Rats, Inbred Strains
19.
J Neural Transm ; 72(3): 213-33, 1988.
Article in English | MEDLINE | ID: mdl-3418334

ABSTRACT

Biochemical and pharmacological effects of gamma-vinyl GABA (Vigabatrin, GVG), and irreversible enzyme-activated inhibitor of 4-aminobutyrate: 2-oxoglutarate aminotransferase (EC 2.6.1.19; GABA-T), were measured in mice. This anticonvulsant produced a time- and dose-dependent elevation of the GABA, phenylalanine and lysine contents of cortical tissue and simultaneously decreased glutamate, aspartate and alanine levels. In addition, GVG caused a biphasic change in glutamine concentrations (a decline 1-4 hours after administration, followed 20 hours later by an increase). Moreover, we found a new, as yet unidentified amino acid in the brain eluting with the same retention time as alpha-aminoadipic acid from an HPLC cation-exchange column. The level of this novel chemical entity was greatly increased by GVG 20 hours after injection of the drug. At all tested intervals between 1 and 60 hours after injection, GVG was ineffective against maximal electroshock. The GABA-T inhibitor dose-dependently protected mice against isoniazid-induced seizures, simultaneously causing an increase in brain GABA concentrations. However, this apparent correlation applied only until 4 hours after treatment. To better define the anticonvulsant profile of GVG, groups of mice were treated, 1, 2, 4, and 24 hours prior to challenge with convulsant doses of strychnine, pentetrazole (PTZ), and picrotoxin, and brain amino acid levels, including brain concentrations of GVG, were measured. In all instances, the time dependency of the anticonvulsant effects of GVG and of increases in brain GABA levels differed. Amino acid concentrations in animals treated only with GVG were similar to those in animals given GVG and a chemical convulsant. GVG showed no selectivity for seizures produced by impairment of GABA-ergic neurotransmission. Although GVG is an effective GABA-T inhibitor, it apparently affects several other pyridoxal-phosphate-dependent cerebral enzymes and/or interacts with other neurotransmitter systems as well.


Subject(s)
Aminocaproates/pharmacology , Anticonvulsants/pharmacology , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Amino Acids/metabolism , Aminocaproates/pharmacokinetics , Animals , Brain/metabolism , Injections , Isoniazid , Mice , Osmolar Concentration , Pentylenetetrazole , Picrotoxin , Seizures/chemically induced , Seizures/drug therapy , Vigabatrin , gamma-Aminobutyric Acid/metabolism
20.
J Neural Transm ; 63(2): 169-89, 1985.
Article in English | MEDLINE | ID: mdl-3930661

ABSTRACT

Amino acid concentrations were measured in the cortex, cerebellum and hippocampus of the mouse brain before and during seizures induced by isoniazid (250 mg/kg i.p.), an inhibitor of L-glutamate-1-decarboxylase (EC 4.1.1.15: GAD). Valproate sodium and diazepam dose-dependently delay the onset of convulsive fits caused by isoniazid. However, neither diazepam nor valproate prevented the decrease in GABA concentrations produced by isoniazid alone. Also, these antiepileptic drugs did not modify the rate of GABA depletion elicited by isoniazid. These results, observed in four different brain structures, strengthen those first obtained with beta-vinyllactic acid, another inhibitor of GAD.


Subject(s)
Diazepam/therapeutic use , Isoniazid/pharmacology , Seizures/prevention & control , Valproic Acid/therapeutic use , gamma-Aminobutyric Acid/metabolism , Amino Acids/metabolism , Animals , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred Strains , Seizures/chemically induced , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...