Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Brain Res ; 958(1): 210-21, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12468047

ABSTRACT

Progressive neuronal loss and cognitive decline in Alzheimer's disease (AD) might be aggravated by beta-amyloid-enhanced excitotoxicity. Memantine is an uncompetitive NMDA receptor antagonist under clinical development for the treatment of AD. Memantine has neuroprotective actions in several in vitro and in vivo models. In the present study, we determined whether memantine protected against beta-amyloid induced neurotoxicity and learning impairment in rats. Twenty Sprague-Dawley rats received vehicle or vehicle plus memantine (steady-state plasma concentrations of 2.34+/-0.23 microM, n=10) s.c. by osmotic pump for 9 days. After 2 days of treatment, 2 microl of water containing beta-amyloid 1-40 [Abeta(1-40)] were injected into the hippocampal fissure. On the ninth day of treatment, animals were sacrificed, and morphological and immunohistochemical techniques were used to determine the extent of neuronal degeneration and astrocytic and microglial activation in the hippocampus. Psychomotor activity and spatial discrimination were tested on the eighth day of treatment. Abeta(1-40), but not water, injections into hippocampus led to neuronal loss in the CA1 subfield, evidence of widespread apoptosis, and astrocytic and microglial activation and hypertrophy. Memantine treated animals had significant reductions in the amount of neuronal degeneration, pyknotic nuclei, and GFAP immunostaining as compared with vehicle treated animals. These data suggest that memantine, at therapeutically relevant concentrations, can protect against neuronal degeneration induced by beta-amyloid.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Memantine/pharmacology , Peptide Fragments/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Death/drug effects , Cell Death/physiology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Ectodysplasins , Female , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry , Membrane Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microtubule-Associated Proteins/metabolism , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism
2.
Nucl Med Biol ; 29(2): 227-31, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11823128

ABSTRACT

Previous studies in mice and PET investigations in a Rhesus monkey showed that the regional uptake of 18F-memantine could be blocked by pharmacological doses of memantine and (+)-MK-801. In the present study, the binding characteristics of 18F-memantine was examined in five healthy volunteers. In humans, 18F-memantine was homogeneously distributed in gray matter i.e. cortex and basal ganglia regions, as well as the cerebellum. No radioactive metabolites were detected in plasma during the time-frame of the PET studies. The uptake of 18F-memantine in receptor-rich regions such as striatum and frontal cortex could be well described by a 1-tissue compartment model. The DV" values of all gray matter regions were similar and ranged from 15 to 20 ml/ml. The white matter showed lower DV" values of 15 +/- 1.4 ml/ml. These results suggest that 18F-memantine distribution in human brain does not reflect the regional NMDA receptor concentration, and therefore, this radioligand is not suitable for the PET imaging of the NMDA receptors.


Subject(s)
Brain/diagnostic imaging , Memantine/analogs & derivatives , Memantine/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/metabolism , Adult , Brain/metabolism , Humans , Male , Memantine/blood , Radioligand Assay , Radiopharmaceuticals/blood , Receptors, N-Methyl-D-Aspartate/analysis , Reference Values , Tomography, Emission-Computed
3.
J Neural Transm (Vienna) ; 108(2): 167-87, 2001.
Article in English | MEDLINE | ID: mdl-11314771

ABSTRACT

The antiparkinsonian effects of the uncompetitive NMDA antagonists, memantine, amantadine and MK-801, in combination with an acute subthreshold dose of L-Dopa (5 mg/kg) in drug-naive MPTP-treated mice or a suprathreshold dose (20 mg/kg) in L-Dopa tolerant MPTP-treated mice were investigated. In the former case, memantine (locomotion: 3 mg/kg; rearing: 1 mg/kg) and amantadine (locomotion and rearing: 10 mg/kg) injected 60 min before the subthreshold dose of L-Dopa (5 mg/kg), each induced an antiparkinsonian action in hypokinesic MPTP-treated mice that consisted of dose-specific, as opposed to dose-related, elevations of locomotion and rearing behaviour. At the same time, higher doses of memantine reduced further the rearing (10 and 30 mg/kg) and locomotor (30mg/kg) behaviour of the MPTP-treated mice. MK-801 plus L-Dopa elevated locomotion (0.1 mg/kg) but reduced rearing at the 0.3 mg/kg dose. In control, saline-treated mice, memantine (3, 10 and 30 mg/kg) and MK-801 (0.1 and 0.3 mg/kg) increased locomotor behaviour but decreased rearing behaviour, while amantadine produced no effects. Memantine increased locomotor (1 and 3 mg/kg, s.c.; 1 mg/kg dose restored activity) and rearing (0.3 and 3 mg/kg) activity in the L-Dopa tolerant MPTP-treated mice, whereas amantadine (3 and 10 mg/kg) restored both locomotor (30 mg/kg significantly increased locomotion but did not restore the activity level) and rearing (3 mg/kg only) activity. MK-801 (0.1 and 0.3 mg/kg, s.c.) also increased significantly locomotor activity of L-Dopa-tolerant MPTP mice although the antikinetic action was not reversed, thereby precluding a restorative effect of the compound. These results, demonstrating both a synergistic and a restorative effect of the NMDA antagonists in coadministration with L-Dopa, demonstrate a putative antiparkinson action by these compounds in a functional animal model that incorporates the "wearing-off" complications of L-Dopa administration in the disorder.


Subject(s)
Amantadine/pharmacology , Antiparkinson Agents/pharmacology , Memantine/pharmacology , Motor Activity/drug effects , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Dizocilpine Maleate/pharmacology , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Excitatory Amino Acid Antagonists/pharmacology , Levodopa/pharmacology , Male , Mice , Mice, Inbred C57BL , Recovery of Function/drug effects
4.
Antimicrob Agents Chemother ; 45(5): 1360-6, 2001 May.
Article in English | MEDLINE | ID: mdl-11302796

ABSTRACT

We reported recently that the bloodstream form of the African trypanosome, Trypanosoma brucei, is sensitive to the anti-influenza virus drug rimantadine. In the present report we describe the trypanocidal properties of a further 62 aminoadamantane and aminoalkylcyclohexane derivatives. Seventeen of the compounds were found to be more active than rimantadine, with four inhibiting growth in vitro of T. brucei by >90% at concentrations of 1 microM. The most active derivative (1-adamantyl-4-amino-cyclohexane) was about 20 to 25 times more effective than rimantadine. We observed a correlation between structural features of the derivatives and their trypanocidal properties; hydrophobic substitutions to the adamantane or cyclohexane rings generally enhanced activity. As with rimantadine, the activity in vitro varied with the pH. T. brucei was more sensitive in an alkaline environment (including a normal bloodstream pH of 7.4) and less sensitive under acidic conditions. Tests for activity in vivo were carried out with a mouse model of infection with a virulent strain of T. brucei. Although the parasitemia was not eliminated, it could be transiently suppressed by >98% with the most active compounds tested. These results suggest that aminoadamantane derivatives could have potential as a new class of trypanocidal agents.


Subject(s)
Adamantane/analogs & derivatives , Cyclohexylamines/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Adamantane/chemistry , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Cyclohexylamines/chemistry , Cyclohexylamines/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Hydrogen-Ion Concentration , Mice , Structure-Activity Relationship , Trypanocidal Agents/chemistry , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/growth & development , Trypanosomiasis/drug therapy
5.
Neurochem Res ; 25(11): 1497-502, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11071369

ABSTRACT

Subclinical hepatic encephalopathy (SHE) was produced in rats by two intraperitoneal injections of TAA at 24 h intervals and the animals were examined 21 days later. Concentrations of the neuroactive amino acids taurine (Tau), glutamate (Glu) and aspartate (Asp), were measured in the cerebral cortical microdialysates of thioacetamide (TAA)-treated and untreated control rats. During microdialysis some animals were awake while others were anesthetized with ketamine plus xylazine. There was no difference in the water content of cerebral cortical slices isolated from control and SHE rats, indicating a recovery from cerebral cortical edema that accompanies the acute, clinical phase of hepatic encephalopathy in this model. When microdialysis was carried out in awake rats, dialysate concentrations of all the three amino acids were 30% to 50% higher in SHE rats than in control rats. Ketamine anesthesia caused a 2.2% increase of water content of cerebral cortical slices and increased Asp, Glu, and Tau concentration in microdialysates of control rats. In SHE rats, ketamine anesthesia produced a similar degree of cerebral edema, however, it did not alter Asp and Glu concentrations in the microdialysates. These data may reflect on one hand a neuropathological process of excitotoxic neuronal damage related to increased Glu and Asp, on the other hand neuroprotection from neuronal swelling indicated by Tau redistribution in the cerebral cortex. The reduction of the effects of SHE on Glu and Asp content in ketamine-anesthesized rats is likely to be due to interference of ketamine with the NMDA receptor-mediated component of the SHE-evoked excitatory neurotransmitter efflux and/or reuptake of the two amino acids. By contrast, the SHE-related increase of Tau content was not affected by ketamine anesthesia, indicating that the mechanism(s) underlying SHE-evoked accumulation of Tau must be different from the mechanism causing release of excitatory amino acids. The results with ketamine advocate caution when using this anesthetic in studies employing the cerebral microdialysis technique for measurement of extracellular amino acids.


Subject(s)
Anesthetics, Dissociative/pharmacology , Aspartic Acid/metabolism , Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Ketamine/pharmacology , Liver Failure/chemically induced , Taurine/metabolism , Thioacetamide/toxicity , Animals , Body Water , Extracellular Space/metabolism , Male , Microdialysis , Rats , Rats, Wistar
6.
Amino Acids ; 19(1): 157-66, 2000.
Article in English | MEDLINE | ID: mdl-11026484

ABSTRACT

There is general agreement that moderate affinity uncompetitive NMDA receptor antagonists combine good efficacy and tolerability in animal models of disturbances in glutamatergic transmission. There are several theories on which properties are important for this profile including 1, rapid access to the channel at the start of pathological overactivity 2, rapid, voltage-dependent relief of blockade during physiological synaptic activation and 3, partial untrapping. Merz has developed a series of novel uncompetitive NMDA receptor antagonists based on the cyclohexane structure. In cultured hippocampal neurones MRZ 2/579 (1-amino-1,3,3,5,5-pentamethylcyclohexane) shows similar blocking kinetics to memantine (Kon 10.7 * 10(4)M(-1)sec(-1), Koff 0.20sec(-1) at -70mV) and binds at the same depth in the NMDA receptor channel (delta = 0.8). The potency of MRZ 2/579 assessed as Kd = Koff/Kon = 1.87microM agrees well with the IC50 of 1.29microM against steady-state currents in cultured hippocampal neurones (at -70mV) and with the Ki in [3H]-MK-801 binding of 0.65microM. MRZ 2/579 protected cultured cortical neurones against glutamate toxicity with an IC50 of 2.16microM and was also effective in protecting hippocampal slices against hypoxia/hypoglycaemia-induced reduction of fEPSP amplitude in CA1 with an EC50 of 7.01microM. MRZ 2/579 has similar potency and bio-availability to memantine in vivo assessed using microdialysis, microiontophoresis and MES-induced seizures. Initial characterization in animal models provides strong support for the assumption that MRZ 2/579 could be a useful therapeutic in morphine/alcohol dependence, inhibition of morphine tolerance, chronic pain and as a neuroprotective agent.


Subject(s)
Cyclopentanes/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , In Vitro Techniques , Rats
7.
Amino Acids ; 19(1): 167-72, 2000.
Article in English | MEDLINE | ID: mdl-11026485

ABSTRACT

The previous overviews provided the basis for better therapeutic efficacy/tolerability of low to moderate affinity NMDA channel blockers. This prediction finds support in in vitro studies comparing protective and plasticity impairing effects (therapeutic vs. side-effect) of memantine and (+)MK-801. In fact it turned out that memantine had a far better therapeutic index. But can it be confirmed in the in vivo situation?


Subject(s)
Cyclohexanes/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals
8.
Eur J Med Chem ; 35(6): 555-65, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10906408

ABSTRACT

A series of 1,3,5-alkylsubstituted cyclohexylamines 2 were synthesized as ligands for the N-methyl-D-aspartate (NMDA) receptor phencyclidine (PCP) binding site. Pure diastereomers with defined configuration of amino group 2-ax and 2-eq were obtained. The optimal size of 1,3,5-substituents was determined for cyclohexylamines 2 with an equatorial amino group in the lowest energy conformation using Hansch analysis. According to the data, the lipophilic part of cyclohexylamines 2 does not discriminate between hydrophobic regions of the PCP binding site but rather recognizes this site as a whole lipophilic pocket.


Subject(s)
Cyclohexylamines/chemical synthesis , Cyclohexylamines/metabolism , Phencyclidine/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Amantadine/chemistry , Amantadine/metabolism , Animals , Binding Sites , Dizocilpine Maleate/chemistry , Dizocilpine Maleate/metabolism , Molecular Mimicry , Rats , Structure-Activity Relationship
9.
Life Sci ; 66(12): 1079-83, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10737358

ABSTRACT

The loss of cholinergic neurons within the basal forebrain of patients with Alzheimer's disease (AD) may underlie aspects of the dementia. Excessive activation of N-methyl-D-aspartate (NMDA) receptors may underlie the degeneration of cholinergic cells. New drug therapies have been designed to either enhance cholinergic function by inhibition acetylcholinesterase (AChE), e.g. galanthamine, tetrahydroaminoacridine or donepezil, or by attenuation of NMDA receptor function, e.g. memantine. A combination of these two therapeutic approaches may be more beneficial at slowing the progression of the AD. The current study investigated whether memantine would attenuate the inhibition of AChE produced by these three drugs. The results indicate that these AChE inhibitors do not lose their therapeutic efficacy in combination with memantine. Our in vitro data suggest that the clinical combination of memantine with a reversible AChE inhibitor should be a valuable pharmacotherapeutic approach to dementia.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Corpus Striatum/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Animals , Donepezil , Dose-Response Relationship, Drug , Drug Interactions , Galantamine/pharmacology , Indans/pharmacology , Male , Piperidines/pharmacology , Rats , Rats, Inbred F344 , Tacrine/pharmacology
10.
Eur J Pharm Biopharm ; 49(2): 103-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10704892

ABSTRACT

A novel non-competitive NMDA receptor antagonist MRZ 2/576 is a potent but rather short-acting (5-15 min) anticonvulsant following intravenous administration to mice as estimated by the prevention of maximal electroshock induced convulsions. This is most probably due to a rapid elimination of the drug from the central nervous system by transport processes that are sensitive to probenecid. Intravenous administration of the drug bound to poly(butylcyanoacrylate) nanoparticles coated with polysorbate 80 prolongs the duration of the anticonvulsive activity in mice up to 210 min and after probenecid pre-treatment up to 270 min compared to 150 min with probenecid and MRZ 2/576 alone. The results of this study demonstrate that polysorbate 80 coated poly(butylcyanoacrylate) nanoparticles used so far as a delivery system to the brain for drugs that do not freely penetrate the blood brain barrier can also be used as a parenteral controlled release system to prolong the CNS availability of drugs that have a short duration of action.


Subject(s)
Anticonvulsants/administration & dosage , Drug Delivery Systems , Enbucrilate/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Phthalazines/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Anticonvulsants/pharmacology , Female , Mice , Polysorbates/administration & dosage , Probenecid/pharmacology , Time Factors
11.
Neurotox Res ; 1(4): 299-310, 2000 Apr.
Article in English | MEDLINE | ID: mdl-12835097

ABSTRACT

High doses of NMDA antagonists e.g. (+)MK-801 evoke neurodegeneration in retrosplenial cortex in rodents. To assess functional consequences of such treatment, three paradigms of two-way active avoidance learning (with visual or auditory conditioned stimuli) and additionally a spatial learning paradigm - radial maze - were used. Female rats were treated i.p. with 5 mg/kg of (+)MK-801. Recumbence, severe hypothermia and loss of body weight were observed for 3-7 days. Despite that, there were no statistically significant differences in performance of avoidance reaction between saline and (+)MK-801 treated animals trained 10-40 days after the drug administration. However, in the radial maze test (+)MK-801 impaired reference (but not working) memory in the experiment that started 8 days after the treatment. Similar effect was observed on reversal learning. The clinically used NMDA receptor antagonist memantine at the doses of 20 and 40 mg/kg had also no such long term negative effect on working memory during training (even positive effect was seen at 20 mg/kg) but at 40 mg/kg impaired learning on the first day of reversal. This indicates that (+)MK-801 neurotoxicity in the retrosplenial cortex is connected with subtle alterations in the learning performance that may be seen in some tests only. Moreover, memantine doses greatly exceeding therapeutically relevant range produce minimal functional alteration. An additional experiment revealed that the same dose of memantine results in two fold higher serum levels of the antagonist in female than male rats. Hence, considering that profiling studies are done in male rats, a safety factor of over 16 fold can be calculated for memantine.

12.
Neurotox Res ; 2(2-3): 85-97, 2000.
Article in English | MEDLINE | ID: mdl-16787834

ABSTRACT

The involvement of glutamate mediated neurotoxicity in the pathogenesis of Alzheimer's disease is finding increasingly more acceptance in the scientific community. Central to this hypothesis is the assumption that in particular glutamate receptors of the N-methyl-D-aspartate (NMDA) type are overactivated in a tonic rather than a phasic manner. Such continuous mild activation leads under chronic conditions to neuronal damage. Moreover, one should consider that impairment of plasticity (learning) may result not only from neuronal damage per se but also from continuous activation of NMDA receptors. To investigate this possibility we tested whether overactivation of NMDA receptors using either non-toxic doses/concentrations of a direct NMDA agonist or through an indirect approach--decrease in magnesium concentration--produces deficits in plasticity. In fact NMDA both in vivo (passive avoidance test) and in vitro (LTP in CA1 region) impaired learning and synaptic plasticity. Under these conditions memantine which is an uncompetitive NMDA receptor antagonist with features of "improved magnesium" (voltage dependence, affinity) attenuated the deficit. The more direct proof that memantine can act as a surrogate for magnesium was obtained in LTP experiments under low magnesium conditions. In this case as well, impaired LTP was restored in the presence of therapeutically relevant concentrations of memantine (1 microM). In vivo, doses leading to similar brain/serum levels produce neuroprotection in animal models relevant for neurodegeneration in Alzheimer's disease such as neurotoxicity produced by inflammation in the NBM or beta-amyloid injection to the hippocampus. Hence, we postulate that if in Alzheimer's disease overactivation of NMDA receptors occurs indeed, memantine would be expected to improve both symptoms (cognition) and slow down disease progression because it takes over the physiological function of magnesium.

13.
Hepatology ; 30(3): 636-40, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462368

ABSTRACT

Brain edema sufficient to cause intracranial hypertension and brain herniation remains a major cause of mortality in acute liver failure (ALF). Studies in experimental animal models of ALF suggest a role for ammonia in the pathogenesis of both encephalopathy and brain edema in this condition. As part of a series of studies to evaluate the therapeutic efficacy of ammonia-lowering agents, groups of rats with ALF caused by hepatic devascularization were treated with L-ornithine-L-aspartate (OA), an agent shown previously to be effective in reducing blood ammonia concentrations in both experimental and human chronic liver failure. Treatment of rats in ALF with infusions of OA (0.33 g/kg/h, intravenously) resulted in normalization of plasma ammonia concentrations and in a significant delay in onset of severe encephalopathy. More importantly, brain water content was significantly reduced in OA-treated rats with ALF. These protective effects of OA were accompanied by increased plasma concentrations of several amino acids including glutamate, gamma-aminobutyric acid (GABA), taurine, and alanine, as well as the branched-chain amino acids, leucine, isoleucine, and valine. Increased availability of glutamate following OA treatment provides the substrate for the major ammonia-removal mechanism (glutamine synthetase). Plasma (but not cerebrospinal fluid) glutamine concentrations were increased 2-fold (P <.02) in OA-treated rats, consistent with increased muscle glutamine synthesis. Direct measurement of glutamine synthetase activities revealed a 2-fold increase following OA treatment. These findings demonstrate a significant ammonia-lowering effect of OA together with a protective effect on the development of encephalopathy and brain edema in this model of ALF.


Subject(s)
Ammonia/metabolism , Brain Edema/prevention & control , Dipeptides/therapeutic use , Liver Failure, Acute/drug therapy , Ammonia/blood , Ammonia/cerebrospinal fluid , Animals , Glutamic Acid/metabolism , Liver Failure, Acute/metabolism , Male , Rats , Rats, Sprague-Dawley
14.
Neuropharmacology ; 38(6): 735-67, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10465680

ABSTRACT

N-methyl-D-aspartate (NMDA) receptor antagonists have therapeutic potential in numerous CNS disorders ranging from acute neurodegeneration (e.g. stroke and trauma), chronic neurodegeneration (e.g. Parkinson's disease, Alzheimer's disease, Huntington's disease, ALS) to symptomatic treatment (e.g. epilepsy, Parkinson's disease, drug dependence, depression, anxiety and chronic pain). However, many NMDA receptor antagonists also produce highly undesirable side effects at doses within their putative therapeutic range. This has unfortunately led to the conclusion that NMDA receptor antagonism is not a valid therapeutic approach. However, memantine is clearly an uncompetitive NMDA receptor antagonist at therapeutic concentrations achieved in the treatment of dementia and is essentially devoid of such side effects at doses within the therapeutic range. This has been attributed to memantine's moderate potency and associated rapid, strongly voltage-dependent blocking kinetics. The aim of this review is to summarise preclinical data on memantine supporting its mechanism of action and promising profile in animal models of chronic neurodegenerative diseases. The ultimate purpose is to provide evidence that it is indeed possible to develop clinically well tolerated NMDA receptor antagonists, a fact reflected in the recent interest of several pharmaceutical companies in developing compounds with similar properties to memantine.


Subject(s)
Excitatory Amino Acid Antagonists/adverse effects , Memantine/adverse effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Brain/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Excitatory Amino Acid Antagonists/pharmacokinetics , Humans , Learning/drug effects , Memantine/pharmacokinetics , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics
15.
Neuropharmacology ; 38(1): 85-108, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10193901

ABSTRACT

The present study characterized the in vitro NMDA receptor antagonistic properties of novel amino-alkyl-cyclohexane derivatives and compared these effects with their ability to block excitotoxicity in vitro and MES-induced convulsions in vivo. The 36 amino-alkyl-cyclohexanes tested displaced [3H]-(+)-MK-801 binding to rat cortical membranes with K(i)s between 1.5 and 143 microM. Current responses of cultured hippocampal neurones to NMDA were antagonized by the same compounds with a wide range of potencies (IC50s of 1.3-245 microM, at -70 mV) in a use- and strongly voltage-dependent manner (delta 0.55-0.87). The offset kinetics of NMDA receptor blockade was correlated with equilibrium affinity (Corr Coeff. 0.87 P < 0.0001). As an example, MRZ 2/579 (1-amino-1,3,3,5,5-pentamethyl-cyclohexane HCl) had similar blocking kinetics to those previously reported for memantine (K(on) 10.67 +/- 0.09 x 10(4) M(-1) s(-1), K(off) 0.199 +/- 0.02 s(-1), K(d) = K(off)/K(on) = 1.87 microM c.f. IC50 of 1.29 microM). Most amino-alkyl-cyclohexanes were protective against glutamate toxicity in cultured cortical neurones (e.g. MRZ 2/579 IC50 2.16 +/- 0.03 microM). Potencies in the three in vitro assays showed a relatively strong cross correlation (all corr. coeffs. > 0.72, P < 0.0001). MRZ 2/579 was also effective in protecting hippocampal slices against 7 min. hypoxia/hypoglycaemia-induced reduction of fEPSP amplitude in CA1 with an EC50 of 7.01 +/- 0.24 microM. MRZ 2/579 showed no selectivity between NMDA receptor subtypes expressed in Xenopus oocytes but was somewhat more potent than in patch clamp experiments-IC50s of 0.49 +/- 0.11, 0.56 +/- 0.01 microM, 0.42 +/- 0.04 and 0.49 +/- 0.06 microM on NR1a/2A /2B, /2C and 2/D, respectively. In contrast, memantine and amantadine were both 3-fold more potent at NR1a/2C and NR1a/2D than NR1a/2A receptors. All Merz amino-alkyl-cyclohexane derivatives inhibited MES-induced convulsions in mice with ED50s ranging from 3.6 to 130 mg/kg i.p. The in vivo and in vitro potencies correlated indicating similar access of most compounds to the CNS. MRZ 2/579 administered at 10 mg/kg resulted in peak plasma concentrations of 5.3 and 1.4 microM following i.v. and p.o. administration respectively, which then declined with a half life of around 170-210 min. Analysis of A.U.C. concentrations indicates a p.o./i.v. bioavailability ratio for MRZ 2/579 of 60%. MRZ 2/579 injected i.p. at a dose of 5 mg/kg resulted in peak brain extracellular fluid (ECF) concentrations of 0.78 microM (brain microdialysates). Of the compounds tested MRZ 2/579, 2/615, 2/632, 2/633, 2/639 and 2/640 had affinities, kinetics and voltage-dependency most similar to those of memantine and had good therapeutic indices against MES-induced convulsions. We predict that these amino-alkyl-cyclohexanes, which all had methyl substitutions at R1, R2, and R5, at least one methyl or ethyl at R3 or R4 and a charged amino-containing substitution at R6, could be useful therapeutics in a wide range of CNS disorders proposed to involve disturbances of glutamatergic transmission.


Subject(s)
Amines/pharmacology , Cyclohexanes/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Alkylation , Animals , Binding, Competitive , Cells, Cultured , In Vitro Techniques , Kinetics , Male , Mice , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Rats, Wistar , Time Factors , Xenopus laevis
16.
J Recept Signal Transduct Res ; 19(1-4): 129-41, 1999.
Article in English | MEDLINE | ID: mdl-10071753

ABSTRACT

A synthetic method has been established for preparing [18F]1-amino-3-fluoromethyl-5-methyl-adamantane ([18F]AFA). Biodistribution of the radiotracer in mice showed high brain uptake. The peak uptake (3.7% I.D/g organ) for the brain occurred at 30 min after injection. Accumulation of radioactivity in mouse brain was consistent with the known distribution of the NMDA receptors. The binding of [18F]AFA to the phencyclidine (PCP) binding sites of the NMDA receptor complex and the sigma recognition sites in a Rhesus monkey was also examined using positron emission tomography (PET). The regional brain distribution of [18F]AFA was changed by memantine and by (+)-MK-801, indicating competition for the same binding sites. Treatment with haloperidol caused a marked reduction of radioactivity uptake in all the brain regions examined. (-)-Butaclamol, which has pharmacological specificity for sigma sites, did not have any significant effects.


Subject(s)
Fluorine Radioisotopes , Memantine/analogs & derivatives , Radiopharmaceuticals , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Butaclamol/pharmacology , Female , Fluorine Radioisotopes/pharmacokinetics , Haloperidol/pharmacology , Macaca mulatta , Memantine/chemical synthesis , Memantine/pharmacokinetics , Mice , Mice, Inbred ICR , Phencyclidine/metabolism , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, sigma/drug effects , Receptors, sigma/metabolism , Tissue Distribution , Tomography, Emission-Computed
17.
Exp Toxicol Pathol ; 50(4-6): 501-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9784030

ABSTRACT

Possible antioxidative properties of three N-methyl-D-aspartate (NMDA)-receptor antagonists, the anesthetic ketamine and the antiparkinson drugs memantine and amantadine were investigated in vitro on the microsomal cytochrome P450 (P450) system of rat livers and on rat whole blood chemiluminescence in comparison to nicanartine, a substance with known antiatherosclerotic, hypolipemic and antioxidative capacity. For this purpose, the effects on NADPH- and iron-stimulated lipid peroxidation (LPO), hydrogen peroxide (H2O2) production, and NADPH- and iron-stimulated lucigenin (LC) and luminol (LM) amplified chemiluminescence (CL) were examined using rat liver microsomes. Additionally, the influence on LM amplified whole blood chemiluminescence after zymosan activation of polymorphonuclear leukocytes (WB-CL) was investigated. Furthermore, binding to P450 and effects on P450 mediated monooxygenase function, as measured by the model reactions ethoxyresorufin O-deethylation (EROD), ethoxycoumarin O-deethylation (ECOD), and ethylmorphine N-demethylation (END), were assessed. Nicanartine concentration dependently reduced LPO and H2O2 production already at a concentration of 1 microM, whereas LC and LM amplified CL and WB-CL were not affected. EROD and END were concentration dependently diminished starting at 1 microM, and ECOD already at 0.1 microM. Ketamine decreased LPO, H2O2 production and LM and LC amplified CL, starting at 100 microM. WB-CL was significantly diminished already at 10 microM. EROD and ECOD were inhibited at 10 and 100 microM and END at 100 microM. With memantine a concentration dependent inhibition of LPO and WB-CL was seen at 100 and 1000 microM and a reduction of LC and LM amplified CL only at 1000 microM. H2O2 production was not affected. EROD and ECOD were significantly diminished by a concentration of 100 microM. No effect was observed on END. Amantadine significantly reduced LPO and WB-CL, but only at 1000 microM. H2O2 production and LC and LM amplified CL were not affected. EROD was significantly diminished at 100 microM, whereas no influence was seen on ECOD and END. Nicanartine displayed type II or reverse type I, ketamine, memantine and amantadine type I substrate binding to P450. The highest binding affinity to P450 was seen with nicanartine, followed by ketamine, memantine and then amantadine. These results demonstrate, that all four substances seem to act as radical scavengers and/or as inhibitors of the oxidative function of P450. All four substances seem to interfere with the monooxygenase function of P450. This may result in a possible influence on the biotransformation of endogenous as well as of foreign compounds. The effects of nicanartine were much more pronounced than those of ketamine, memantine, and amantadine.


Subject(s)
Amantadine/pharmacology , Antioxidants/pharmacology , Ketamine/pharmacology , Memantine/pharmacology , Microsomes, Liver/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , 7-Alkoxycoumarin O-Dealkylase/metabolism , Animals , Antiparkinson Agents/pharmacology , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Hydrogen Peroxide/metabolism , Lipid Peroxidation/drug effects , Male , Microsomes, Liver/enzymology , Pyridines/pharmacology , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
18.
Metab Brain Dis ; 13(2): 147-57, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9699922

ABSTRACT

Strategies aimed at the lowering of blood ammonia remain the treatment of choice in portal-systemic encephalopathy (PSE). L-ornithine-L-aspartate (OA) has recently been shown to be effective in the prevention of ammonia-precipitated coma in humans with PSE. These findings prompted the study of mechanisms of the protective effect of OA in portacaval-shunted rats in which reversible coma was precipitated by ammonium acetate administration (3.85 mmol/kg i.p.). OA infusions (300 mg/kg/h, i.v) offered complete protection in 12/12 animals compared to 0/12 saline-infused controls. This protective effect was accompanied by significant reductions of blood ammonia, concomitant increases of urea production and significant increases in blood and cerebrospinal fluid (CSF) glutamate and glutamine. Increased CSF concentrations of leucine and alanine also accompanied the protective effect of OA. These findings demonstrate the therapeutic efficacy of OA in the prevention of ammonia-precipitated coma in portacaval-shunted rats and suggest that this protective effect is both peripherally-mediated (increased urea and glutamine synthesis) and centrally-mediated (increased glutamine synthesis).


Subject(s)
Dipeptides/therapeutic use , Hepatic Encephalopathy/prevention & control , Ammonia/blood , Animals , Dipeptides/pharmacology , Glutamic Acid/biosynthesis , Male , Portacaval Shunt, Surgical , Rats , Rats, Sprague-Dawley , Urea/blood
19.
Nucl Med Biol ; 25(4): 323-30, 1998 May.
Article in English | MEDLINE | ID: mdl-9639292

ABSTRACT

The effect of the fluorinated memantine derivative and NMDA receptor antagonist, 1-amino-3-fluoromethyl-5-methyl-adamantane (19F-MEM), at the NMDA receptor ion channel was studied by patch clamp recording. The results showed that 19F-MEM is a moderate NMDA receptor channel blocker. A procedure for the routine preparation of the 18F-labelled analog 18F-MEM has been developed using a two-step reaction sequence. This involves the no-carrier-added nucleophilic radiofluorination of 1-[N-(tert-butyloxy)carbamoyl]-3-(toluenesulfonyloxy)methyl- 5-methyl-adamantane and the subsequent cleavage of the BOC-protecting group using aqueous HCI. The 18F-MEM was obtained in 22 +/- 7% radiochemical yield (decay-corrected to EOB) in a total synthesis time including HPLC purification of 90 min. A biodistribution study after i.v. injection of 18F-MEM in mice showed a fast clearance of radioactivity from blood and relatively high initial uptake in the kidney and in the lung, which gradually decreased with time. The brain uptake was high (up to 3.6% ID/g, 60 min postinjection) with increasing brain-blood ratios: 2.40, 5.10, 6.33, and 9.27 at 5, 30, 60, and 120 min, respectively. The regional accumulation of the radioactivity in the mouse brain was consistent with the known distribution of the PCP recognition site. Preliminary PET evaluation of the radiotracer in a rhesus monkey demonstrated good uptake and prolonged retention in the brain, with a plateau from 35 min onwards p.i. in the NMDA receptor-rich regions (frontal cortex, striata, and temporal cortex). Delineation of the hippocampus, a region known to contain a high density of NMDA receptors, was not possible owing to the resolution of the PET tomograph. The regional brain uptake of 18F-MEM was changed by memantine and by a pharmacological dose of (+)-MK-801, indicating competition for the same binding sites. In a preliminary experiment, haloperidol, a dopamine D2 and sigma receptor antagonist, decreased the binding of 18F-MEM from the brain regions examined, suggesting that binding was also occurring to the sigma recognition sites.


Subject(s)
Fluorine Radioisotopes/pharmacokinetics , Memantine/analogs & derivatives , Memantine/pharmacokinetics , Radioligand Assay , Radiopharmaceuticals/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Binding, Competitive , Blood-Brain Barrier , Brain/drug effects , Brain/metabolism , Dizocilpine Maleate/pharmacology , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluorine Radioisotopes/pharmacology , Macaca mulatta , Memantine/chemical synthesis , Memantine/pharmacology , Mice , Mice, Inbred ICR , Patch-Clamp Techniques , Radiopharmaceuticals/pharmacology , Tissue Distribution , Tomography, Emission-Computed
20.
Antiviral Res ; 37(2): 83-95, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9588841

ABSTRACT

We tested two approaches to overcoming resistance of influenza A viruses against adamantane derivatives. First, adamantane derivatives that interfere with the ion channel function of the variant M2 protein of amantadine-resistant viruses may prevent drug resistance, if they are used in mixture with amantadine. Second, amantadine acts on the M2 protein (at low concentrations) and indirectly on the hemagglutinin (at concentrations at least 100 times higher). Identifying and using a drug that reacted with both targets at the same concentration might reduce development of resistance, since, in this case, two mutations, one in each target protein would be necessary at once. Such a double mutation is assumed to be a rare event. We evaluated forty adamantane derivatives and two related compounds to determine whether they interfered with plaque formation by influenza A strains, including A/Singapore/1/57 (H2N2). Variants resistant to drugs that interfered at low concentrations (approximately 1 microg/ml; e.g. amantadine) were cross-resistant with each other, but were sensitive to those agents effective at high concentrations (8 microg/ml; e.g. memantine). The former group of compounds act on the ion channel; the corresponding escape mutants tested had amino acid replacements at positions 27, 30 or 31 of the M2 protein. Hemagglutinin was the indirect target of the latter group of compounds. Variants resistant to these agents lacked amino acid replacements within the ion channel of the M2 protein and the mutants tested had amino acid replacements in the hemagglutinin. Although we failed to identify compounds that interacted with the ion channel of amantadine-resistant variants and inhibited their replication, we were able to construct at least two compounds that interfered with both the ion channel and the hemagglutinin at about the same concentration. After passage in the presence of these compounds, we either failed to obtain any drug-resistant mutants or those obtained had amino acid replacements in the ion channel of the M2 protein and the hemagglutinin.


Subject(s)
Adamantane/analogs & derivatives , Antiviral Agents/pharmacology , Influenza A virus/drug effects , Adamantane/chemistry , Animals , Cell Line , Chick Embryo , Dogs , Drug Resistance, Microbial , Genetic Variation , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemolysis , Humans , Hydrogen-Ion Concentration , Influenza A virus/genetics , Influenza A virus/physiology , Molecular Structure , Viral Matrix Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...