Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Alzheimers Dement (N Y) ; 2(1): 13-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-29067291

ABSTRACT

INTRODUCTION: Gln-1062 (Memogain) is a pharmacologically inactive prodrug of galantamine. Owing to its lipophilic nature, it preferentially enters the brain, where it is cleaved into active galantamine. Gln-1062 is expected to have fewer peripheral side effects than other cholinesterase inhibitors, with improved effectiveness. METHODS: This was a double-blind, comparator and placebo-controlled, sequential cohort, single ascending dose study in 58 healthy subjects with Gln-1062 in doses of 5.5, 11, 22, 33, and 44 mg, compared with oral galantamine 16 mg and donepezil 10 mg. Safety, tolerability, pharmacokinetics, and pharmacodynamics were assessed. RESULTS: Gln-1062 doses up to 33 mg were well tolerated and induced a dose-dependent increase in the plasma concentrations of Gln-1062 and galantamine. Gln-1062 had a dose-dependent positive effect on verbal memory and attention, mainly in the first hours after drug administration. DISCUSSION: Gln-1062 was better tolerated than galantamine in doses with the same molarity and led to improved effects in cognitive tests. This is most likely caused by the more favorable distribution ratio between peripheral and central cholinesterase inhibition. These results give reason for further exploration of this compound.

2.
J Alzheimers Dis ; 46(1): 123-36, 2015.
Article in English | MEDLINE | ID: mdl-25720404

ABSTRACT

The plant alkaloid galantamine is an established symptomatic drug treatment for Alzheimer's disease (AD), providing cognitive and global relief in human patients. However, as an acetylcholinesterase inhibitor, gastrointestinal side effects limit the dosage and duration of treatment. Memogain (Gln-1062), a pro-drug, liberates galantamine on cleavage by a carboxyesterase in the brain. The possibility to deliver Memogain intranasally may further circumvent side effects, allowing higher dosing compared to galantamine. In this study, the 5X Familial Alzheimer's Disease (5XFAD) mouse model was used to investigate the effect of chronic Memogain treatment on behavior and amyloid-ß (Aß) plaque deposition in the brain. Chronic intranasal dosage of 6 mg/kg body weight twice daily was tolerated well, whereas the double dose caused body weight loss in males and was less effective in some behavioral tests. 8 weeks of chronic treatment resulted in improved performance in behavioral tests, such as open field and light-dark avoidance, and in fear conditioning already at mildly affected stages at the age of 18 weeks compared to untreated controls. Furthermore, after treatment a significantly lower plaque density in the brain, i.e., in the entorhinal cortex (reduction 20% females, 40% males) and the hippocampus (19% females, 31% males) at the age of 18 weeks was observed. These results show that nasal application of Memogain effectively delivers the drug to the brain with the potential to retard plaque deposition and improve behavioral symptoms in AD similar to the approved galantamine.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Behavior, Animal/drug effects , Cholinesterase Inhibitors/administration & dosage , Galantamine/analogs & derivatives , Prodrugs/administration & dosage , Administration, Intranasal , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Disease Models, Animal , Fear/drug effects , Fear/physiology , Galantamine/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Muscle Strength/drug effects , Muscle Strength/genetics , Mutation/genetics , Plaque, Amyloid/drug therapy , Plaque, Amyloid/etiology , Presenilin-1/genetics
3.
PLoS One ; 9(2): e89454, 2014.
Article in English | MEDLINE | ID: mdl-24586789

ABSTRACT

The plant alkaloid galantamine is an established symptomatic drug treatment for Alzheimer's disease (AD), providing temporary cognitive and global relief in human patients. In this study, the 5X Familial Alzheimer's Disease (5XFAD) mouse model was used to investigate the effect of chronic galantamine treatment on behavior and amyloid ß (Aß) plaque deposition in the mouse brain. Quantification of plaques in untreated 5XFAD mice showed a gender specific phenotype; the plaque density increased steadily reaching saturation in males after 10 months of age, whereas in females the density further increased until after 14 months of age. Moreover, females consistently displayed a higher plaque density in comparison to males of the same age. Chronic oral treatment with galantamine resulted in improved performance in behavioral tests, such as open field and light-dark avoidance, already at mildly affected stages compared to untreated controls. Treated animals of both sexes showed significantly lower plaque density in the brain, i.e., the entorhinal cortex and hippocampus, gliosis being always positively correlated to plaque load. A high dose treatment with a daily uptake of 26 mg/kg body weight was tolerated well and produced significantly larger positive effects than a lower dose treatment (14 mg/kg body weight) in terms of plaque density and behavior. These results strongly support that galantamine, in addition to improving cognitive and behavioral symptoms in AD, may have disease-modifying and neuroprotective properties, as is indicated by delayed Aß plaque formation and reduced gliosis.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Cholinesterase Inhibitors/therapeutic use , Galantamine/therapeutic use , Maze Learning/drug effects , Plaque, Amyloid/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Behavior, Animal/drug effects , Brain/pathology , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Female , Galantamine/pharmacology , Male , Mice , Neurons/drug effects , Neurons/pathology , Plaque, Amyloid/pathology , Sex Factors
4.
J Recept Signal Transduct Res ; 30(6): 469-83, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21062106

ABSTRACT

Galantamine is an approved drug treatment for Alzheimer's disease. Initially identified as a weak cholinesterase inhibitor, we have established that galantamine mainly acts as an 'allosterically potentiating ligand (APL)' of nicotinic acetylcholine receptors (nAChR). Meanwhile other 'positive allosteric modulators (PAM)' of nAChR channel activity have been discovered, and for one of them a binding site within the transmembrane domain has been proposed. Here we show, by performing site-directed mutagenesis studies of ectopically expressed chimeric chicken α7/mouse 5-hydroxytryptamine 3 receptor-channel complex, in combination with whole-cell current measurements, in the presence and absence of galantamine, that the APL binding site is different from the proposed PAM binding site. We demonstrate that residues T197, I196, and F198 of ß-strand 10 represent major elements of the galantamine binding site. Residue K123, earlier suggested as being 'close to' the APL binding site, is not part of this site but rather appears to play a role in coupling of agonist binding to channel opening and closing. Our data confirm our earlier results that the galantamine binding site is different from the ACh binding site. Both sites are in close proximity and hence may influence each other in a synergistic fashion. Other interesting areas identified in the present study are a 'hinge' region around and containing residues F122, K123, and K143 possibly being involved in relaying the signal of agonist binding to gating of the transmembrane channel, and a 'folding centre', with P119 as the dominating residue, that crucially positions the agonist binding site with respect to the hinge region.


Subject(s)
Cholinesterase Inhibitors/metabolism , Galantamine/metabolism , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Allosteric Regulation , Animals , Binding Sites , Cell Line , Chickens , Dose-Response Relationship, Drug , Humans , Ligands , Mice , Models, Molecular , Patch-Clamp Techniques , Receptors, Serotonin, 5-HT3/chemistry , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT3/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , alpha7 Nicotinic Acetylcholine Receptor
5.
J Biol Chem ; 285(51): 40180-91, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20947502

ABSTRACT

Reduction of brain amyloid-ß (Aß) has been proposed as a therapeutic target for Alzheimer disease (AD), and microglial Aß phagocytosis is noted as an Aß clearance system in brains. Galantamine is an acetylcholinesterase inhibitor approved for symptomatic treatment of AD. Galantamine also acts as an allosterically potentiating ligand (APL) for nicotinic acetylcholine receptors (nAChRs). APL-binding site is located close to but distinct from that for acetylcholine on nAChRs, and FK1 antibody specifically binds to the APL-binding site without interfering with the acetylcholine-binding site. We found that in human AD brain, microglia accumulated on Aß deposits and expressed α7 nAChRs including the APL-binding site recognized with FK1 antibody. Treatment of rat microglia with galantamine significantly enhanced microglial Aß phagocytosis, and acetylcholine competitive antagonists as well as FK1 antibody inhibited the enhancement. Thus, the galantamine-enhanced microglial Aß phagocytosis required the combined actions of an acetylcholine competitive agonist and the APL for nAChRs. Indeed, depletion of choline, an acetylcholine-competitive α7 nAChR agonist, from the culture medium impeded the enhancement. Similarly, Ca(2+) depletion or inhibition of the calmodulin-dependent pathways for the actin reorganization abolished the enhancement. These results suggest that galantamine sensitizes microglial α7 nAChRs to choline and induces Ca(2+) influx into microglia. The Ca(2+)-induced intracellular signaling cascades may then stimulate Aß phagocytosis through the actin reorganization. We further demonstrated that galantamine treatment facilitated Aß clearance in brains of rodent AD models. In conclusion, we propose a further advantage of galantamine in clinical AD treatment and microglial nAChRs as a new therapeutic target.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Cholinesterase Inhibitors/pharmacology , Galantamine/pharmacology , Microglia/metabolism , Receptors, Nicotinic/metabolism , Aged , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Antibodies, Monoclonal , Binding Sites , Calcium Signaling/drug effects , Calmodulin/metabolism , Cells, Cultured , Choline/metabolism , Choline/pharmacology , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Microglia/pathology , Nootropic Agents/pharmacology , Phagocytosis/drug effects , Rats , Rats, Wistar
6.
J Mol Neurosci ; 40(1-2): 135-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19669943

ABSTRACT

Memogain (Gln-1062) is an inactive pro-drug of galantamine, the latter being a plant alkaloid approved for the treatment of mild to moderate Alzheimer's disease. Memogain has more than 15-fold higher bioavailability in the brain than the same doses of galantamine. In the brain, Memogain is enzymatically cleaved to galantamine, thereby regaining its pharmacological activity as a cholinergic enhancer. In animal models of drug-induced amnesia, Memogain produced several fold larger cognitive improvement than the same doses of galantamine, without exhibiting any significant levels of gastrointestinal side effects that are typical for the unmodified drug and other inhibitors of cholinesterases, such as donepezil and rivastigmin. In the ferret, dramatically reduced emetic and behavioral responses were observed when Memogain was administered instead of galantamine. Based on these and other preclinical data, Memogain may represent an advantageous drug treatment for Alzheimer's disease, combining much lesser gastrointestinal side effects and considerably higher potency in enhancing cognition, as compared to presently available drugs.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Cholinesterase Inhibitors/pharmacology , Cognition Disorders/drug therapy , Galantamine/analogs & derivatives , Galantamine/pharmacology , Acetylcholine/agonists , Animals , Brain/physiopathology , Brain Chemistry/drug effects , Cholinesterase Inhibitors/adverse effects , Cognition/drug effects , Cognition Disorders/chemically induced , Cognition Disorders/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Ferrets , Galantamine/adverse effects , Gastrointestinal Diseases/chemically induced , Humans , Mice , Scopolamine/antagonists & inhibitors , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Treatment Outcome
7.
ChemMedChem ; 4(11): 1874-82, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19739198

ABSTRACT

Current treatments of Alzheimer's disease include the allosteric potentiation of nicotinic acetylcholine receptor (nAChR) response. The location of the binding site for allosteric potentiating ligands (APLs) within the receptor is not yet fully understood. Based on homology models for the ligand binding domain of human alpha7, human alpha4beta2, and chicken alpha7 receptors, as well as blind docking experiments with galanthamine, physostigmine, codeine, and 5HT, we identified T197 as an essential element of the APL binding site at the outer surface of the ligand binding domain (LBD) of nAChR. We also found the previously known galanthamine binding site in the region of K123 at the inside of the receptor funnel, which, however, was shown to not be part of the APL site. Our results are verified by site-directed mutagenesis and electrophysiological experiments, and suggest that APL and ACh bind to different sites on nicotinic receptors and that allosteric potentiation may arise from a direct interplay between both these sites.


Subject(s)
Ligands , Receptors, Nicotinic/chemistry , Allosteric Regulation , Amino Acid Sequence , Animals , Binding Sites , Chickens , Computer Simulation , Humans , Molecular Sequence Data , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/pharmacology , Protein Structure, Tertiary , Receptors, Nicotinic/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
8.
J Mol Neurosci ; 30(1-2): 227-32, 2006.
Article in English | MEDLINE | ID: mdl-17192682

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive impairments that become severe enough to interfere with the daily activities of patients and eventually lead to death (Chung and Cummings, 2000). Arecent study reports that approx 24 million people suffer from dementia worldwide. If the mortality rate does not change and no curative or preventive treatment is developed, this number is expected to double every 20 yr worldwide (Ferri et al., 2005). Although the causes of AD remain obscure, it has been reported that incremental loss of cholinergic neurons and of nicotinic receptor (nAChR) function/expression in specific brain regions correlates well with the severity of the symptoms at early stages of the disease (Hellström-Lindahl et al., 1999; Nordberg, 2001; Perry et al., 2001; Wevers et al., 1999). In patients with more advanced stages of AD, such a correlation between the magnitude of nAChR loss and of cognitive decline does not appear to exist (Sabbagh et al., 2001). The nicotinic cholinergic system plays a central role in modulating different forms of associative learning known to be impaired in AD patients, including the eyeblink classical conditioning (Woodruff-Pak, 2001), and in maintaining neuronal viability. Neuroprotection and cognitive improvement result from increasing the activity of different nAChR subtypes, including those bearing the alpha7 subunit (Carlson et al., 1998; Hejmadi et al., 2003; Kihara et al., 1997; Levin et al., 2006). Thus, increasing nAChR activity in the brain was proposed as a mechanism to slow down the progression of the disease (Maelicke and Albuquerque, 1996).


Subject(s)
Galantamine/analogs & derivatives , Galantamine/pharmacology , Hippocampus/physiology , Neurons/physiology , Receptors, Nicotinic/physiology , Animals , Cell Culture Techniques , Choline/pharmacology , Evoked Potentials/drug effects , Ligands , Neurons/drug effects , Patch-Clamp Techniques , Receptors, Nicotinic/drug effects , alpha7 Nicotinic Acetylcholine Receptor
9.
J Pharmacol Exp Ther ; 312(3): 1195-205, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15522999

ABSTRACT

The N-methyl-d-aspartate (NMDA) receptor antagonist memantine is an approved drug for treatment of Alzheimer's disease (AD). Other such treatments are cholinesterase inhibitors and nicotinic acetylcholine receptor (nAChR)-sensitizing agents such as galantamine. The present study was designed to test whether memantine exerts any effect on the cholinergic system, in particular the Ca(2+)-conducting alpha7(*) nAChR, in cultured hippocampal neurons. Memantine caused a concentration-dependent reduction of the amplitudes of whole-cell currents evoked by the alpha7(*) nAChR-selective agonist choline (10 mM) or by N-methyl-d-aspartate (NMDA) (50 muM) plus glycine (10 muM). It also inhibited tonically activated NMDA receptors. Memantine was more potent in inhibiting alpha7(*) nAChRs than NMDA receptors; at -60 mV, the IC(50) values for memantine were 0.34 and 5.1 muM, respectively. Consistent with an open-channel blocking mechanism, memantine-induced NMDA receptor inhibition was voltage and use-dependent; the Hill coefficient (n(H)) was approximately 1. Memantine-induced alpha7(*) nAChR inhibition had an n(H) < 1 and showed a variable voltage dependence; the effect was voltage-independent at 0.1 muM, becoming voltage-dependent at >/=1 muM. Thus, memantine interacts with more than one class of sites on the alpha7(*) nAChRs. One is voltage-sensitive and therefore likely to be within the receptor channel. The other is voltage-insensitive and therefore likely to be in the extracellular domain of the receptor. It is suggested that blockade of alpha7(*) nAChRs by memantine could decrease its effectiveness for treatment of AD, particularly at early stages when the degrees of nAChR dysfunction and of cognitive decline correlate well.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Memantine/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Nicotinic/drug effects , Animals , Dose-Response Relationship, Drug , Membrane Potentials , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor
10.
Biochem Biophys Res Commun ; 325(3): 976-82, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15541385

ABSTRACT

Galantamine is a plant alkaloid that is used in the treatment of Alzheimer's disease. We have studied the effects of galantamine on beta-amyloid-enhanced glutamate toxicity using primary rat cultured cortical neurons. Nicotine and galantamine alone, and in combination, protected neurons against this neurotoxicity. The protection was not blocked by alpha4beta2 nicotinic acetylcholine receptor (nAChR) antagonists, but was partially blocked by alpha7 nAChR antagonists. Galantamine induced phosphorylation of Akt, an effector of phosphatidylinositol 3-kinase (PI3K), while PI3K inhibitors blocked the protective effect and Akt phosphorylation. The antibody FK1, which selectively blocks the allosterically potentiating ligand site on nAChR, significantly reduced the galantamine-induced protection and Akt phosphorylation. Furthermore, suppression of alpha7 nAChR using an RNA interference technique reduced Akt phosphorylation induced by galantamine. Our data suggest that neuroprotection by galantamine is mediated, at least in part, by alpha7 nAChR-PI3K cascade.


Subject(s)
Amyloid beta-Peptides/pharmacology , Galantamine/pharmacology , Glutamic Acid/pharmacology , Neurons/cytology , Neurons/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Animals , Cell Survival/drug effects , Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Neurons/drug effects , Neuroprotective Agents/pharmacology , Rats
11.
Mol Endocrinol ; 18(11): 2714-26, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15297607

ABSTRACT

The germ cell nuclear factor (GCNF) is essential for normal embryonic development and gametogenesis. To test the prediction that GCNF is additionally required for neuronal differentiation, we used the mouse embryonal carcinoma cell line PCC7-Mz1, which represents an advantageous model to study neuronal cells from the stage of fate choice until the acquirement of functional competence. We generated stable transfectants that express gcnf sense or antisense RNA under the control of a tetracycline-regulated promoter. After retinoic acid-induced withdrawal from the cell cycle, sense clones developed a neuron network with changed properties, and the time course of neuron maturation was shortened. Consistent with these data, differentiation of neuronal precursor cells was impaired in antisense cultures. This involved a delay in 1) the down-regulation of nestin, a marker for undifferentiated neuroepithelial cells and stem cells of the central nervous system, and 2) up-regulation of the somatodendritic protein microtubule-associated protein 2 and the synaptic vesicle protein synaptophysin. Neuronal cells in the antisense cultures acquired functional competence, although with a significant delay. Our data propose that the level of GCNF is critical for differentiation and maturation of neuronal precursor cells.


Subject(s)
DNA-Binding Proteins/metabolism , Neurons/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Cycle/drug effects , Cell Differentiation/genetics , Cell Polarity , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Down-Regulation , GAP-43 Protein/analysis , GAP-43 Protein/biosynthesis , GAP-43 Protein/genetics , Gene Expression , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Nuclear Receptor Subfamily 6, Group A, Member 1 , Patch-Clamp Techniques , RNA, Antisense/genetics , Receptors, Cytoplasmic and Nuclear/analysis , Receptors, Cytoplasmic and Nuclear/genetics , Synaptophysin/analysis , Synaptophysin/biosynthesis , Synaptophysin/genetics , Tretinoin/pharmacology , Up-Regulation
12.
Bioorg Med Chem Lett ; 14(8): 1879-87, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15050620

ABSTRACT

In HEK293 cells stably expressing alpha4beta2 nAChRs, naltrexone, but not naloxone, blocked alpha4beta2 nAChRs via an open-channel blocking mechanism. In primary hippocampal cultures, naltrexone inhibited alpha7 nAChRs up-regulated by nicotine, and in organotypic hippocampal cultures naltrexone caused a time-dependent up-regulation of functional alpha7 nAChRs that was detected after removal of the drug. These results indicate that naltrexone could be used as a smoking cessation aid.


Subject(s)
Naloxone/pharmacology , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Up-Regulation/drug effects , Cell Line , Gene Expression/drug effects , Humans , Neurons/metabolism , Nicotine/antagonists & inhibitors , Nicotine/pharmacology , Patch-Clamp Techniques , Receptors, Nicotinic/genetics , Sensitivity and Specificity , Smoking Cessation/methods , Structure-Activity Relationship , Time Factors , Up-Regulation/physiology
13.
FEBS Lett ; 554(1-2): 219-23, 2003 Nov 06.
Article in English | MEDLINE | ID: mdl-14596943

ABSTRACT

The Xenopus laevis oocyte expression system was used to determine the activities of alpha-conotoxins EpI and the ribbon isomer of AuIB, on defined nicotinic acetylcholine receptors (nAChRs). In contrast to previous findings on intracardiac ganglion neurones, alpha-EpI showed no significant activity on oocyte-expressed alpha3beta4 and alpha3beta2 nAChRs but blocked the alpha7 nAChR with an IC50 value of 30 nM. A similar IC50 value (103 nM) was obtained on the alpha7/5HT3 chimeric receptor stably expressed in mammalian cells. Ribbon AuIB maintained its selectivity on oocyte-expressed alpha3beta4 receptors but unlike in native cells, where it was 10-fold more potent than native alpha-AuIB, had 25-fold lower activity. These results indicate that as yet unidentified factors influence alpha-conotoxin pharmacology at native versus oocyte-expressed nAChRs.


Subject(s)
Conotoxins/pharmacology , Receptors, Nicotinic/drug effects , Animals , Inhibitory Concentration 50 , Nicotinic Antagonists/pharmacology , Oocytes , Protein Subunits/drug effects , Rats , Receptors, Nicotinic/genetics , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Substrate Specificity , Transfection , Xenopus laevis
14.
J Pharmacol Exp Ther ; 305(3): 1024-36, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12649296

ABSTRACT

Galantamine (Reminyl), an approved treatment for Alzheimer's disease (AD), is a potent allosteric potentiating ligand (APL) of human alpha 3 beta 4, alpha 4 beta 2, and alpha 6 beta 4 nicotinic receptors (nAChRs), and of the chicken/mouse chimeric alpha 7/5-hydroxytryptamine3 receptor, as was shown by whole-cell patch-clamp studies of human embryonic kidney-293 cells stably expressing a single nAChR subtype. Galantamine potentiates agonist responses of the four nAChR subtypes studied in the same window of concentrations (i.e., 0.1-1 microM), which correlates with the cerebrospinal fluid concentration of the drug at the recommended daily dosage of 16 to 24 mg. At concentrations >10 microM, galantamine acts as an nAChR inhibitor. The other presently approved AD drugs, donepezil and rivastigmine, are devoid of the nicotinic APL action; at micromolar concentrations they also block nAChR activity. Using five CHO-SRE-Luci cell lines, each of them expressing a different human muscarinic receptor, and a reporter gene assay, we show that galantamine does not alter the activity of M1-M5 receptors, thereby confirming that galantamine modulates selectively the activity of nAChRs. These studies support our previous proposal that the therapeutic action of galantamine is mainly produced by its sensitizing action on nAChRs rather than by general cholinergic enhancement due to cholinesterase inhibition. Galantamine's APL action directly addresses the nicotinic deficit in AD.


Subject(s)
Galantamine/pharmacology , Neurons/drug effects , Phenylcarbamates , Receptors, Muscarinic/metabolism , Receptors, Nicotinic/metabolism , Allosteric Regulation , Animals , Carbamates/pharmacology , Cells, Cultured , Cholinesterase Inhibitors/pharmacology , Donepezil , Humans , Indans/pharmacology , Mice , Neurons/metabolism , Piperidines/pharmacology , Receptors, Nicotinic/drug effects , Recombinant Fusion Proteins/drug effects , Rivastigmine , Tacrine/pharmacology , Trichlorfon/pharmacology
15.
J Recept Signal Transduct Res ; 22(1-4): 283-95, 2002.
Article in English | MEDLINE | ID: mdl-12503622

ABSTRACT

Digital expression pattern display (DEPD) is an open, automated, PCR-based system of gene expression profiling that is capable of resolving as many as 100,000 transcripts from a single brain tissue cDNA sample. It has a detection sensitivity of better than 1 in 750,000 and it can reliably detect differences in RNA expression levels of less than two-fold. Digital expression pattern display presently is the most sensitive and therefore the only expression profiling method available that is capable of monitoring, in a semi-quantitative fashion, the expression of even the rarest of transcripts found in human brain tissue. Biofrontera applies this proprietary technique, together with state-of-the-art bioinformatics, for the purposes of elucidating pathology pathways of major brain diseases, of analysing the target profiles of drugs presently applied or in development, and of identifying novel targets for drug action.


Subject(s)
Brain/drug effects , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/analysis , Animals , Brain/metabolism , Computational Biology , Drug Delivery Systems , Drug Design , Humans , Pharmaceutical Preparations , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
16.
J Neurobiol ; 53(4): 479-500, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12436414

ABSTRACT

Evidence gathered from epidemiologic and behavioral studies have indicated that neuronal nicotinic receptors (nAChRs) are intimately involved in the pathogenesis of a number of neurologic disorders, including Alzheimer's disease, Parkinson's disease, and schizophrenia. In the mammalian brain, neuronal nAChRs, in addition to mediating fast synaptic transmission, modulate fast synaptic transmission mediated by the major excitatory and inhibitory neurotransmitters glutamate and GABA, respectively. Of major interest, however, is the fact that the activity of the different subtypes of neuronal nAChR is also subject to modulation by substances of endogenous origin such as choline, the tryptophan metabolite kynurenic acid, neurosteroids, and beta-amyloid peptides and by exogenous substances, including the so-called nicotinic allosteric potentiating ligands, of which galantamine is the prototype, and psychotomimetic drugs such as phencyclidine and ketamine. The present article reviews and discusses the effects of unconventional ligands on nAChR activity and briefly describes the potential benefits of using some of these compounds in the treatment of neuropathologic conditions in which nAChR function/expression is known to be altered.


Subject(s)
Ligands , Receptors, Nicotinic/metabolism , Amyloid beta-Peptides/metabolism , Anesthetics/pharmacology , Animals , Choline/metabolism , Galantamine/pharmacology , Hallucinogens/metabolism , Humans , Kynurenic Acid/metabolism , Serotonin/metabolism , Steroids/metabolism
18.
Mol Pharmacol ; 61(5): 1222-34, 2002 May.
Article in English | MEDLINE | ID: mdl-11961141

ABSTRACT

In this study, the patch-clamp technique was used to determine the effects of galantamine, a cholinesterase inhibitor and a nicotinic allosteric potentiating ligand (APL) used for treatment of Alzheimer's disease, on synaptic transmission in brain slices. In rat hippocampal and human cerebral cortical slices, 1 microM galantamine, acting as a nicotinic APL, increased gamma-aminobutyric acid (GABA) release triggered by 10 microM acetylcholine (ACh). Likewise, 1 microM galantamine, acting as an APL on presynaptically located nicotinic receptors (nAChRs) that are tonically active, potentiated glutamatergic or GABA-ergic transmission between Schaffer collaterals and CA1 neurons in rat hippocampal slices. The cholinesterase inhibitors rivastigmine, donepezil, and metrifonate, which are devoid of nicotinic APL action, did not affect synaptic transmission. Exogenous application of ACh indicated that high and low levels of nAChR activation in the Schaffer collaterals inhibit and facilitate, respectively, glutamate release onto CA1 neurons. The finding then that the nAChR antagonists methyllycaconitine and dihydro-beta-erythroidine facilitated glutamatergic transmission between Schaffer collaterals and CA1 neurons indicated that in a single hippocampal slice, the inhibitory action of strongly, tonically activated nAChRs in some glutamatergic fibers prevails over the facilitatory action of weakly, tonically activated nAChRs in other glutamatergic fibers synapsing onto a given neuron. Galantamine is known to sensitize nAChRs to activation by low, but not high agonist concentrations. Therefore, at 1 microM, galantamine is likely to increase facilitation of synaptic transmission by weakly, tonically activated nAChRs just enough to override inhibition by strongly, tonically activated nAChRs. In conclusion, the nicotinic APL action can be an important determinant of the therapeutic effectiveness of galantamine.


Subject(s)
Central Nervous System/drug effects , Galantamine/pharmacology , Nicotine/pharmacology , Nootropic Agents/pharmacology , Synaptic Transmission/drug effects , Action Potentials/drug effects , Allosteric Regulation/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Central Nervous System/physiology , Cholinesterases/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , In Vitro Techniques , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cholinergic/metabolism , Receptors, Glutamate/metabolism , Time Factors , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...