Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 7(5): 1103-11, 2007 May.
Article in English | MEDLINE | ID: mdl-17456198

ABSTRACT

ATP decreases markedly in small-for-size liver grafts. This study tested if the mitochondrial permeability transition (MPT) underlies dysfunction of small-for-size livers. Half-size livers were implanted into recipients of about twice the donor weight, resulting in quarter-size liver grafts. NIM811 (5 microM), a nonimmunosuppressive MPT inhibitor was added to the storage solutions. Mitochondrial polarization and cell death were assessed by confocal microscopy of rhodamine 123 (Rh123) and propidium iodide (PI), respectively. After quarter-size transplantation, alanine aminotransferase (ALT), serum bilirubin and necrosis all increased. NIM811 blocked these increases by >70%. After 38 h, BrdU labeling, a marker of cell proliferation and graft weight increase were 3% and 5%, respectively, which NIM811 increased to 30% and 42%. NIM811 also increased survival of quarter-size grafts. In sham-operated livers, hepatocytes exhibited punctate Rh123 fluorescence. By contrast, in quarter-size grafts at 18 h after implantation, mitochondria of most hepatocytes did not take up Rh123, indicating mitochondrial depolarization. Nearly all hepatocytes not taking up Rh123 continued to exclude PI at 18 h, indicating that depolarization preceded cell death. NIM811 and free radical-scavenging polyphenols strongly attenuated mitochondrial depolarization. In conclusion, mitochondria depolarized after quarter-size liver transplantation. NIM811 decreased injury and stimulated regeneration, probably by inhibiting free radical-dependent MPT onset.


Subject(s)
Cyclosporine/pharmacology , Liver Transplantation/physiology , Mitochondria, Liver/drug effects , Mitochondria, Liver/physiology , Animals , Apoptosis/drug effects , Electrophysiology , Graft Rejection/physiopathology , Graft Rejection/prevention & control , Hepatocytes/metabolism , Hepatocytes/pathology , Hyperbilirubinemia/physiopathology , Hyperbilirubinemia/prevention & control , Ion Channels/metabolism , Liver Regeneration/drug effects , Liver Transplantation/adverse effects , Liver Transplantation/pathology , Male , Mitochondrial Proteins/metabolism , Necrosis/physiopathology , Necrosis/prevention & control , Organ Size , Permeability , Rats , Rats, Inbred Lew , Uncoupling Protein 2
2.
J Neurosci Res ; 79(1-2): 231-9, 2005.
Article in English | MEDLINE | ID: mdl-15573402

ABSTRACT

Experimental traumatic brain injury (TBI) and spinal cord injury (SCI) result in a rapid and significant necrosis of neuronal tissue at the site of injury. In the ensuing hours and days, secondary injury exacerbates the primary damage, resulting in significant neurologic dysfunction. It is believed that alterations in excitatory amino acids (EAA), increased reactive oxygen species (ROS), and the disruption of Ca(2+) homeostasis are major factors contributing to the ensuing neuropathology. Mitochondria serve as the powerhouse of the cell by maintaining ratios of ATP:ADP that thermodynamically favor the hydrolysis of ATP to ADP + P(i), yet a byproduct of this process is the generation of ROS. Proton-pumping by components of the electron transport system (ETS) generates a membrane potential (DeltaPsi) that can then be used to phosphorylate ADP or sequester Ca(2+) out of the cytosol into the mitochondrial matrix. This allows mitochondria to act as cellular Ca(2+) sinks and to be in phase with changes in cytosolic Ca(2+) levels. Under extreme loads of Ca(2+), however, opening of the mitochondrial permeability transition pore (mPTP) results in the extrusion of mitochondrial Ca(2+) and other high- and low-molecular weight components. This catastrophic event discharges DeltaPsi and uncouples the ETS from ATP production. Cyclosporin A (CsA), a potent immunosuppressive drug, inhibits mitochondrial permeability transition (mPT) by binding to matrix cyclophilin D and blocking its binding to the adenine nucleotide translocator. Peripherally administered CsA attenuates mitochondrial dysfunction and neuronal damage in an experimental rodent model of TBI, in a dose-dependent manner. The underlying mechanism of neuroprotection afforded by CsA is most likely via interaction with the mPTP because the immunosuppressant FK506, which has no effect on the mPT, was not neuroprotective. When CsA was administrated after experimental SCI at the same dosage and regimen used TBI paradigms, however, it had no beneficial neuroprotective effects. This review takes a comprehensive and critical look at the evidence supporting the role for mPT in central nervous system (CNS) trauma and highlights the differential responses of CNS mitochondria to mPT induction and the implications this has for therapeutically targeting the mPT in TBI and SCI.


Subject(s)
Brain Injuries/physiopathology , Mitochondria/physiology , Neurons/physiology , Spinal Cord Injuries/physiopathology , Animals , Brain Injuries/drug therapy , Brain Injuries/metabolism , Calcium/metabolism , Cell Death/drug effects , Cell Death/physiology , Cyclosporine/therapeutic use , Enzyme Inhibitors/therapeutic use , Humans , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/physiopathology , Models, Biological , Neurons/drug effects , Permeability/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Time Factors
3.
Br J Pharmacol ; 131(4): 721-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11030721

ABSTRACT

Apoptosis and mitochondrial dysfunction are thought to be involved in the aetiology of neurodegenerative diseases. We have tested an orally active anti-apoptotic molecule (CGP 3466B) that binds to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in an animal model with motoneuron degeneration, i.e. a mouse mutant with progressive motor neuronopathy (pmn). In pmn/pmn mice, CGP 3466B was administered orally (10 - 100 nmol kg(-1)) at the onset of the clinical symptoms (2 weeks after birth). CGP 3466B slowed disease progression as determined by a 57% increase in life-span, preservation of body weight and motor performance. This improvement was accompanied by a decreased loss of motoneurons and motoneuron fibres as well as an increase in retrograde transport. Electron microscopic analysis showed that CGP 3466B protects mitochondria which appear to be selectively disrupted in the motoneurons of pmn/pmn mice. The data support evaluation of CGP 3466B as a potential treatment for motor neuron disease.


Subject(s)
Apoptosis/drug effects , Disease Models, Animal , Mitochondria/drug effects , Motor Neuron Disease/drug therapy , Nerve Growth Factors , Oxepins/pharmacology , Administration, Oral , Animals , Glial Cell Line-Derived Neurotrophic Factor , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Mice , Motor Neuron Disease/mortality , Motor Neurons/drug effects , Nerve Tissue Proteins/pharmacology , Weight Loss/drug effects
4.
J Neural Transm Suppl ; (60): 197-214, 2000.
Article in English | MEDLINE | ID: mdl-11205140

ABSTRACT

(-)-Deprenyl, used for the treatment of Parkinson's disease, was reported to possess neurorescuing/antiapoptotic effects independent of its MAO-B inhibiting properties. It is metabolized to (-)-desmethyldeprenyl, which seems to be the active principle, and further to (-)-amphetamine and (-)-methamphetamine, which antagonize its rescuing effects. These complications may explain the limited neurorescuing potential of (-)-deprenyl observed clinically. CGP 3466 (dibenzo[b,f]oxepin-10-ylmethyl-methyl-prop-2-ynyl-amine), structurally related to (-)-deprenyl, exhibits virtually no MAO-B nor MAO-A inhibiting properties and is not metabolized to amphetamines. It was shown to bind to glyceraldehyde-3-phosphate dehydrogenase, a glycolytic enzyme with multiple other functions including an involvement in apoptosis, and shows neurorescuing properties qualitatively similar to, but about 100-fold more potent than those of (-)-deprenyl in several in vitro and in vivo paradigms. In concentrations ranging from 10(-13)-10(-5) M, it rescues partially differentiated PC12 cells from apoptosis induced by trophic withdrawal, cerebellar granule cells from apoptosis induced by cytosine arabinoside, rat embryonic mesencephalic dopaminergic cells from death caused by MPP+, and PAJU human neuroblastoma cells from death caused by rotenone. However, it did not affect apoptosis elicited by a variety of agents in rapidly proliferating cells from thymus or skin or in liver or kidney cells. In vivo, it rescued facial motor neuron cell bodies in rat pups after axotomy, rat hippocampal CA1 neurons after transient ischemia/hypoxia, and mouse nigral dopaminergic cell bodies from death induced by MPTP, in doses ranging between 0.0003 and 0.1 mg/kg p.o. or s.c., depending on the model. It also partially prevented the loss of tyrosine hydroxylase immunoreactivity in the substantia nigra of 6-OHDA-lesioned rats and improved motor function in these animals. Moreover, it prolonged the life-span of progressive motor neuronopathy (pmn) mice (a model for ALS), preserved their body weight and improved their motor performance. This was accompanied by a decreased loss of motor neurons and motor neuron fibers, and protection of mitochondria. The active concentration- or dose-ranges in the different in vitro and in vivo paradigms were remarkably similar. In several paradigms, bell-shaped dose-response curves were observed, the rescuing effect being lost above about 1 mg/kg, a fact that must be considered in clinical investigations.


Subject(s)
Cell Survival/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxepins/pharmacology , Parkinson Disease/drug therapy , Selegiline/analogs & derivatives , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Ligands , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neuroprotective Agents/chemistry , Oxepins/chemistry , Rats , Rats, Wistar , Selegiline/adverse effects
5.
Naunyn Schmiedebergs Arch Pharmacol ; 362(6): 526-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11138845

ABSTRACT

The propargylamine derivative CGP 3466 (dibenzo[b,f]oxepin-10-ylmethyl-methyl-prop-2-ynyl-amine) has previously been found to exhibit neurorescuing and antiapoptotic properties in several in vitro and in vivo paradigms. After showing that this compound does not inhibit monoamine oxidase B and only marginally inhibits monoamine oxidase A at concentrations or doses far above those relevant for its reported neuroprotective effects, we investigated it in models considered relevant for Parkinson's disease. CGP 3466 or its hydrogen maleate salt, CGP 3466B, at concentrations between 10(-11) M and 10(-7) M, protected rat embryonic mesencephalic dopaminergic neurons in free-floating or dispersed cell culture from death inflicted by treatment with 1-methyl-4-phenyl pyridinium ion (MPP+) as measured by different readouts such as dopamine uptake, tyrosine hydroxylase activity, and counts of tyrosine hydroxylase-positive cells. Treatment of mice lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 2x30 mg/kg s.c. at a 72-h interval) with CGP 3466 (0.1 mg/kg s.c.) or CGP 3466B (0.014 mg/kg and 0.14 mg/kg p.o.) b.i.d. for 18 days partially prevented the loss of tyrosine hydroxylase-positive cells in the substantia nigra; a lower dose of CGP 3466B (0.0014 mg/kg p.o.) showed a marginal effect, whereas a high dose, i.e. 1.4 mg/kg p.o., was ineffective, suggesting a bell-shaped dose-response relationship which has also been observed in other paradigms. The effect of CGP 3466 on motor function was evaluated in rats that received intrastriatal injections of 6-OHDA unilaterally, according to a four-site injection protocol, and that were subsequently treated b.i.d. with 0.014 mg/kg i.p. CGP 3466B for 3 weeks. After another 3 weeks without treatment, skilled paw use was assessed by means of the staircase test. The results indicated a significant improvement of skilled motor performance as measured by means of the number of eaten pellets. Since due to the long wash-out period a symptomatic effect of CGP 3466B can be ruled out, it is likely that this improvement was related to interference with the course of the degeneration of the dopaminergic neurons. In conclusion, our results indicate that CGP 3466 is able to prevent death of dopaminergic cells in in vitro and in vivo models of Parkinson's disease. In addition, treatment with CGP 3466 resulted in improved skilled motor performance in 6-OHDA-lesioned rats.


Subject(s)
Antiparkinson Agents/pharmacology , Dopamine/metabolism , Neurons/drug effects , Oxepins/pharmacology , Parkinsonian Disorders/pathology , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Brain/drug effects , Brain/enzymology , Cell Culture Techniques , Cell Death/drug effects , Disease Models, Animal , Dopamine/pharmacokinetics , Female , Liver/drug effects , Liver/enzymology , Mesencephalon/cytology , Mesencephalon/drug effects , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neurons/enzymology , Neurons/pathology , Oxidopamine , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/enzymology , Pregnancy , Rats , Selegiline/pharmacology , Substantia Nigra/enzymology , Tritium , Tyrosine 3-Monooxygenase/metabolism
6.
J Cell Biol ; 151(7): 1537-48, 2000 Dec 25.
Article in English | MEDLINE | ID: mdl-11134081

ABSTRACT

Ras is a universal eukaryotic intracellular protein integrating extracellular signals from multiple receptor types. To investigate its role in the adult central nervous system, constitutively activated V12-Ha-Ras was expressed selectively in neurons of transgenic mice via a synapsin promoter. Ras-transgene protein expression increased postnatally, reaching a four- to fivefold elevation at day 40 and persisting at this level, thereafter. Neuronal Ras was constitutively active and a corresponding activating phosphorylation of mitogen-activated kinase was observed, but there were no changes in the activity of phosphoinositide 3-kinase, the phosphorylation of its target kinase Akt/PKB, or expression of the anti-apoptotic proteins Bcl-2 or Bcl-X(L). Neuronal Ras activation did not alter the total number of neurons, but induced cell soma hypertrophy, which resulted in a 14.5% increase of total brain volume. Choline acetyltransferase and tyrosine hydroxylase activities were increased, as well as neuropeptide Y expression. Degeneration of motorneurons was completely prevented after facial nerve lesion in Ras-transgenic mice. Furthermore, neurotoxin-induced degeneration of dopaminergic substantia nigra neurons and their striatal projections was greatly attenuated. Thus, the Ras signaling pathway mimics neurotrophic effects and triggers neuroprotective mechanisms in adult mice. Neuronal Ras activation might become a tool to stabilize donor neurons for neural transplantation and to protect neuronal populations in neurodegenerative diseases.


Subject(s)
Hypertrophy/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Neurodegenerative Diseases/pathology , Neuroprotective Agents/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins p21(ras)/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Axotomy , Brain/drug effects , Brain/enzymology , Brain/pathology , Cell Count , Cell Size , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Enzyme Activation , Hypertrophy/enzymology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Motor Neurons/drug effects , Motor Neurons/enzymology , Mutation/genetics , Neurodegenerative Diseases/enzymology , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Oxidopamine/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
7.
Eur J Pharmacol ; 362(1): 27-34, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9865526

ABSTRACT

The pharmacological properties of morpholin-2-yl-phosphinic acids were evaluated on GABA(B) receptors. In rat neocortical slices maintained in Mg2+-free Krebs medium, baclofen, a GABA(B) receptor agonist, produced a concentration-dependent depression of the frequency of spontaneous discharges with an EC50 of 14 +/- 5.5 microM, which was antagonised reversibly by the morpholin-2-yl-phosphinic derivatives. The order of potency was 3-[(3S,6R)-6-[(cyclohexylmethyl)hydroxyphosphinoylmethyl- morpholin-3-yl]benzoic acid (CGP 76290A) (pA2 = 7.1 +/- 0.05) > its enantiomer 3-[(3R,6S)-6-[(cyclohexylmethyl)hydroxyphosphinoylmethyl]-++ +morpholin-3-yl]benzoic acid (CGP 76291A) (pA2 = 6.8 +/- 0.1) > cyclohexylmethyl-[(2R',5S')-5-(3-nitrophenyl)-morpholin-2-++ +ylmethyl]phosphinic acid (CGP 71978) (pA2 = 6.5 +/- 0.05) > cyclohexylmethyl-[(2R,5S)-5-phenyl-morpholin-2-ylmethyl++ +]phosphinic acid (CGP 71980) (pA2 = 6.3 +/- 0.15) > its enantiomer cyclohexylmethyl-[(2S,5R)-5-phenyl-morpholin-2-ylmethyl++ +]phosphinic acid (CGP 71979) (pA2 = 5.8 +/- 0.1). An open chain analogue of CGP 76290A, CGP 56999A (3-[1(R)-[(3-cyclohexylmethyl-hydroxyphosphinoyl)-2(S)-hydro xypropyl-amino]-ethyl]benzoic acid lithium salt) gave a pA2 of 6.6 +/- 0.2. In GABA(B) receptor binding assays, CGP 71982 (the racemic mixture of CGP 76290A and CGP 76291A), CGP 76290A, CGP 76291A, CGP 71978, CGP 71980 and CGP 71979 had IC50 values against [3H]CGP 27492 binding of 8, 1.85, 69, 124, 326 and 1460 nM, respectively. In electrically-evoked [3H]GABA release from rat cortical slices, CGP 71982, CGP 71978, CGP 71980 and its enantiomer CGP 71979, antagonised GABA(B) autoreceptors with EC150 values of 2.5, 33, 181 and 474 nM, respectively. These compounds form a novel class of potent GABA(B) receptor antagonists.


Subject(s)
GABA Antagonists/pharmacology , GABA-B Receptor Antagonists , Morpholines/pharmacology , Neocortex/drug effects , Phosphinic Acids/pharmacology , Animals , Baclofen/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Protein Binding , Rats , Rats, Sprague-Dawley
8.
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
9.
Nucl Med Biol ; 23(3): 229-34, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8782230

ABSTRACT

Brofaromine (4-5(-methoxy-7-bromobenzofuranyl)-2-piperidine-HCl) is a potent and selective inhibitor of monoamine oxidase (MAO) A. Two methods for its synthesis and a preliminary positron emission tomography (PET) evaluation in monkey brain are described. The first method, at low carrier concentration of CO2, consisted of direct O-methylation of (4-(5-hydroxy-7-bromobenzofuranyl)-2-piperidine). The total radiochemical yield achieved ranged from 30 to 50% (from end of bombardment [EOB] and decay corrected) with an overall synthesis time of 45 min. The second approach, with high carrier amounts of CO2 arising from inherent target problems, was accomplished in a three-step route involving protection of secondary amino functionality, O-methylation and deprotection. The total radiochemical yield was 10% (from EOB and decay corrected) with a total synthesis time of 70 min. For both methods methylation was achieved using the classical methylating agent [11C]CH3I, and radiochemical purity was higher than 98%. PET evaluation of the radioligand in a Rhesus monkey showed a high uptake of radioactivity in the brain. Using the irreversible MAO-A inhibitor clorgyline and reversible MAO-A inhibitors moclobemide and brofaromine, three blockade experiments were designed to determine the extent of specific binding of [11C]brofaromine to MAO-A. No apparent decrease in accumulation of radioactivity in the monkey brain was observed when compared to a baseline scan.


Subject(s)
Brain/metabolism , Carbon Radioisotopes , Monoamine Oxidase Inhibitors , Monoamine Oxidase/analysis , Piperidines/pharmacokinetics , Tomography, Emission-Computed/methods , Animals , Benzamides/pharmacology , Brain/diagnostic imaging , Brain/drug effects , Carbon Radioisotopes/pharmacokinetics , Clorgyline/pharmacology , Female , Hydrocarbons, Iodinated , Indicators and Reagents , Isoenzymes/analysis , Isotope Labeling , Macaca mulatta , Moclobemide , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/pharmacology , Piperidines/chemical synthesis
10.
Biochem Pharmacol ; 51(5): 613-9, 1996 Mar 08.
Article in English | MEDLINE | ID: mdl-8615897

ABSTRACT

An involvement of GABAergic neurons has been suggested in the process of memory consolidation based on anatomical evidence and increasing physiological and biochemical data. With the advent of orally active GABAB antagonists, such as CGP 36742, the question of their therapeutic value, for example in Alzheimer's disease, becomes relevant. Therefore, a new GC/MS method was developed to determine the concentration of CGP 36742 (3-amino-propyl-n-butyl phosphinic acid) in various intra- and extracerebral tissues after different routes of application. The compound was chemically derivatised in a two-step process (acylation of the amino group and esterification of the phosphinic acid). The limit of detection of the method was 0.01 microgram/g tissue and 0.0005 microgram/mL plasma. The time-course after i.p. treatment showed peak levels of CGP 36742 between 30 min and 1 hr after injection. After a dose of 100 mg/kg, the concentration in the brain ranged from 1 to 1.4 microgram/g or 6 to 8 microM, assuming that 1 mg tissue equals 1 microL (i.e., below the IC50 of the interaction with GABAB receptors as measured by [3-3H]-aminopropyl-phosphinic acid binding [35 microM]). These results are discussed in light of the psychopharmacological effects (improvement of cognitive performance of rats) of CGP 36742 observed at very low oral doses.


Subject(s)
Brain/metabolism , GABA Antagonists/analysis , GABA-A Receptor Antagonists , Organophosphorus Compounds/analysis , Administration, Oral , Animals , Gas Chromatography-Mass Spectrometry , Male , Organophosphorus Compounds/pharmacokinetics , Rats
11.
J Neural Transm (Vienna) ; 103(1-2): 217-45, 1996.
Article in English | MEDLINE | ID: mdl-9026374

ABSTRACT

The antidepressant activity of monoamine oxidase inhibitors has been well established for 30 years. Nevertheless, this group of compounds was handled with great care, mainly because of the interaction potential with tyramine-containing foodstuff. With the discovery of reversible and selective inhibitors of monoamine oxidase type A a renaissance of these compounds has begun. In this paper one of these new substances--brofaromine--will be described in detail. Biochemical and pharmacological aspects will be reviewed, showing that brofaromine is a selective and reversible inhibitor of monoamine oxidase type A with additional serotonin reuptake inhibiting properties. Both mechanisms of action may synergize in the antidepressant effect of the compound. The main results of clinical trials in depression and other indication areas will also be covered. Special attention will be put on the side effect profile.


Subject(s)
Depressive Disorder/drug therapy , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Animals , Brain Chemistry/drug effects , Depressive Disorder/psychology , Humans , Monoamine Oxidase Inhibitors/pharmacokinetics , Piperidines/pharmacokinetics
12.
Naunyn Schmiedebergs Arch Pharmacol ; 352(6): 670-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-9053740

ABSTRACT

The alkaloid and medicinal herb constituent, R,R-(-)-daurisoline, was originally reported to be a N-type Ca2+ channel blocker, but newer evidence indicates that it is a blocker of P-type Ca2+ channels. To clarify its specificity with respect to N- and P-channels, we compared its effects on the electrically induced release of endogenous glutamate, 3H-GABA and 3H-noradrenaline, from brain slices with those of omega-agatoxin IVA and omega-conotoxin GVIA. Like omega-agatoxin IVA (but with about 1000-fold lower potency), and unlike omega-conotoxin GVIA, R.R-(-)-daurisoline inhibited the release of 3H-GABA and glutamate, with IC50 values of 8 and 18 microM. However, inhibition particularly of 3H-GABA release was more complete than by omega-agatoxin IVA, indicating interaction with one or more additional voltage-sensitive Ca2+ channels, possibly the Q-type. Its potency to inhibit glutamate release elicited either electrically, by veratrine or by high concentrations of K+ was similar, in contrast to sodium channel blockers. The effects of R,R-(-)-daurisoline on the release of 3H-noradrenaline, 3H-dopamine and 3H-acetylcholine were in agreement with previous knowledge from experiments with omega-agatoxin IVA suggesting an involvement of P-channels. A weak inhibition of 3H-noradrenaline release at 10 microM, similar to that by omega-agatoxin IVA at 0.03 microM, was occluded by alpha 2-antagonistic properties and could be unmasked in presence of rauwolscine. At 10 microM, it also inhibited electrically evoked 3H-dopamine and 3H-5-hydroxytryptamine release and caused a marked spontaneous release of all three monoamines in a reserpine-like manner. Spontaneous and evoked release of 3H-acetylcholine was inhibited by about 25% at 10 microM. In radioligand binding studies, R,R-(-)-daurisoline interacted with alpha 1- and alpha 2-adrenoceptors, 5-HT2 and muscarinic cholinergic receptors with IC50 values close to 1 microM, and with mu opiate receptors even with 0.18 microM. Atropine reduced the weak inhibitory effect of R,R-(-)-daurisoline on 3H-acetylcholine release somewhat, suggesting that it was brought about by both P channel blockade and cholinergic agonist activity. The effect on 3H-GABA release was unaffected by naloxone, indicating that the interaction of R,R-(-)-daurisoline with mu opiate receptors is antagonistic. The pattern of effects on neurotransmitter release observed with R,R-(-)-daurisoline resembles that of omega-agatoxin IVA and supports previous electrophysiological data suggesting that the compound blocks P-type voltage-sensitive Ca2+ channels. However, the more complete blockade of amino acid release by R,R-(-)-daurisoline suggests interaction with additional Ca2+ channel subtypes. Although it does also possess other pharmacological properties, we think that the compound is suitable to test whether blockade of glutamate release via voltage-sensitive Ca2+ channels is a viable concept to obtain novel neuroprotective and/or anticonvulsant compounds.


Subject(s)
Alkaloids/pharmacology , Benzylisoquinolines , Brain/drug effects , Calcium Channel Blockers/pharmacology , Synaptic Transmission/drug effects , Yohimbine/pharmacology , Acetylcholine/metabolism , Alkaloids/metabolism , Animals , Binding, Competitive , Brain/metabolism , Calcium Channel Blockers/metabolism , Dopamine/metabolism , Glutamic Acid/metabolism , Isotope Labeling , Lethal Dose 50 , Male , Norepinephrine/metabolism , Peptides/pharmacology , Radioligand Assay , Rats , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, alpha/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Serotonin/metabolism , Stereoisomerism , gamma-Aminobutyric Acid/metabolism , omega-Conotoxin GVIA
13.
Neurology ; 45(10): 1907-13, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7477991

ABSTRACT

We compared the effects of the antiepileptic drugs carbamazepine, oxcarbazepine, and lamotrigine on the release from rat brain slices of endogenous glutamate, [3H]-GABA, and [3H]-dopamine, elicited by the Na+ channel opener, veratrine, and of the same transmitters as well as [3H]-noradrenaline, [3H]-5-hydroxytryptamine, and [3H]-acetylcholine, elicited by electrical stimulation. The three antiepileptic drugs inhibited veratrine-induced release of endogenous glutamate, [3H]-GABA, and [3H]-dopamine, with IC50 values between 23 and 150 microM, in or near the concentration range in which they interact with Na+ channels, and there was little difference between the compounds. They were five to seven times less potent in inhibiting electrically as compared with veratrine-stimulated release of [3H]-GABA and [3H]-dopamine; similarly, carbamazepine and tetrodotoxin were more potent in inhibiting veratrine-induced as compared with electrically induced release of endogenous glutamate. Carbamazepine, oxcarbazepine, and lamotrigine also inhibited electrically stimulated release of [3H]-5-hydroxytryptamine (IC50 values, 150 to 250 microM) and [3H]-acetylcholine (IC50 values, 50 to 150 microM); [3H]-noradrenaline release was affected to a lesser degree. The active concentration ranges of these drugs with respect to inhibition of veratrine-stimulated neurotransmitter release matched the therapeutic plasma and brain concentrations. It is uncertain whether these effects are relevant in vivo at anticonvulsant doses, because the drugs are markedly less potent in inhibiting the more physiologic release elicited by electrical stimulation. Therefore, the hypothesis that inhibition of glutamate release is the mechanism of anticonvulsant action of lamotrigine (or carbamazepine and oxcarbazepine) is doubtful. Other consequences of Na+ channel blockade may have an important role.


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/analogs & derivatives , Carbamazepine/pharmacology , Glutamic Acid/metabolism , Receptors, Glutamate/metabolism , Triazines/pharmacology , Animals , Lamotrigine , Male , Oxcarbazepine , Rats , Receptors, Dopamine/metabolism , Receptors, GABA/metabolism , Veratrine/analysis
14.
Br J Pharmacol ; 113(4): 1515-21, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7889310

ABSTRACT

1. The effects of a series of nine GABAB receptor antagonists of widely varying potencies on electrically stimulated release from cortical slices of [3H]-GABA in the absence or presence of 10 microM of the GABAB agonist, (-)-baclofen and of endogenous glutamate in the presence of (-)-baclofen were compared. 2. The concentrations of the compounds half maximally increasing [3H]-GABA release (EC50's) at a stimulation frequency of 2 Hz correlated well with the IC50 values obtained from the inhibition of the binding of the agonist, [3H]-CGP 27492, to GABAB receptors in rat brain membranes (rank order of potency: CGP 56999 A > or = CGP 55845 A > CGP 52432 > or = CGP 56433 A > CGP 57034 A > CGP 57070 A > or = CGP 57976 > CGP 51176 > CGP 35348). 3. Likewise, the concentrations causing half-maximal increases of [3H]-GABA in the absence or presence of (-)-baclofen, and of endogenous glutamate in the presence of (-)-baclofen, correlated well with each other. Reports in the literature suggesting the CGP 35348 exhibits a 70 fold preference for inhibition of (-)-baclofen's effects on glutamate over [3H]-GABA release, and that CGP 52432 shows a 100 fold preference in the opposite sense, could not be confirmed in our model. 4. Therefore, our results suggest that, if there are pharmacological differences between GABAB autoreceptors and GABAB heteroreceptors on glutamatergic nerve endings in the rat cortex, they are not revealed by this series of compounds of widely different potencies. 5. In particular, our results with CGP 35348 and CGP 52432 do not support the hypothesis that GABAB autoreceptors and GABAB heteroreceptors on glutamatergic nerve endings represent subtypes with different pharmacology.


Subject(s)
GABA-B Receptor Antagonists , Glutamic Acid/metabolism , Phosphinic Acids , Receptors, Presynaptic/antagonists & inhibitors , gamma-Aminobutyric Acid/metabolism , Animals , Baclofen/pharmacology , Benzylamines/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Electric Stimulation , GABA-A Receptor Antagonists , In Vitro Techniques , Male , Organophosphorus Compounds/pharmacology , Rats , Rats, Inbred Strains , Receptors, GABA-B/metabolism , Receptors, Presynaptic/metabolism
15.
Naunyn Schmiedebergs Arch Pharmacol ; 349(6): 583-7, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7969509

ABSTRACT

Electrically stimulated release of neurotransmitters in brain slices normally displays frequency dependence because of progressive activation of autoreceptors by endogenously released transmitter, which is abolished by blockade of autoreceptors. In consequence, the maximal increase caused by an autoreceptor antagonist in percent of the corresponding controls should be greater at higher than at lower frequencies. In the case of gamma-aminobutyric acid (GABA), we have previously found a marked deviation from this expectation. Among several explanations envisaged, computer simulation suggested only one to be compatible with the experimental data: the release mechanism may not be able to cope with high demand. This hypothesis was tested by investigating the frequency dependence of the release of 3H-GABA in the presence and absence of a high concentration of the potent GABAB antagonist, CGP 55845, using extremely short stimulation periods. To this end, slices were stimulated with groups of 4 POPs (a POP--pseudo-one-pulse--consists of 4 pulses delivered at 100 Hz). The intervals between the POPs within a group were varied from 60-0.5 s, corresponding to frequencies within the POP group of 0.0167-2 Hz. Under such circumstances, the theoretically expected pattern was indeed observed: the GABAB antagonist abolished the frequency dependence. In a second series of experiments, fractional release per POP was determined when 4-32 POPs were delivered at 2 Hz, with and without CGP 55845. The increase of GABA release elicited by the GABAB antagonist gradually subsided with increasing number of POPs.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cerebral Cortex/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Electric Stimulation , GABA-B Receptor Antagonists , In Vitro Techniques , Male , Phosphinic Acids/pharmacology , Propanolamines/pharmacology , Rats , Rats, Inbred Strains , gamma-Aminobutyric Acid/physiology
16.
Naunyn Schmiedebergs Arch Pharmacol ; 348(6): 618-27, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8133904

ABSTRACT

The autoreceptor-mediated control of GABA release was simulated on a personal computer using commercially available software (STELLA/ITHINK). The experimental data to be matched were taken from previous publications. A basic model was able to fairly accurately reproduce frequency dependencies of GABA release in the presence and absence of uptake inhibition as well as concentration-response curves for changes in release produced by the agonist, (-)-baclofen, or by relatively low concentrations of the antagonists, phaclofen and CGP 35348. Obvious mismatch was observed at high concentrations of a potent antagonist, at a stimulation frequency of 2 Hz. Whereas the experimental data indicate a 3-fold increase in release as compared to controls, simulation predicts a 7-fold increase. By adaptation of the model, simulation data were obtained indicating that this mismatch was not due to (a) the autoreceptor occurring as two subtypes with different affinities for antagonists, (b) the occurrence of an agonist and antagonist state of the autoreceptor, with the latter prevailing at low synaptic concentrations of endogenous GABA, and (c) overruling of uptake inhibition by markedly elevated synaptic GABA concentrations. On the other hand, a simple restriction of the amount of transmitter able to be released per time unit produced much better matching data. A refined model assuming a restricted replacement capacity for exocytotically emptied synaptic vesicles at their docking sites gave similar results. As a consequence, we shall attempt to address this possibility experimentally. Simulation can never prove a case in the positive sense. It can, however, help to exclude ill-matching solutions of a problem and to prioritize among possible ones, which then must be experimentally addressed. We found simulation with this user-friendly software extraordinarily useful, also and not least because it necessitates and stimulates very intense dealing with a subject.


Subject(s)
Receptors, GABA/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Baclofen/pharmacology , Computer Simulation , GABA-B Receptor Antagonists , In Vitro Techniques , Kinetics , Neurotransmitter Uptake Inhibitors/pharmacology , Organophosphorus Compounds/pharmacology , Rats , Receptors, GABA/drug effects , Receptors, GABA-B/drug effects , Receptors, GABA-B/metabolism , Software
17.
Naunyn Schmiedebergs Arch Pharmacol ; 348(5): 478-85, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8114946

ABSTRACT

The effect of the new glutamate uptake inhibitor, L-trans-pyrrolidine-2,4-dicarboxylic acid (L-trans-PDC), on the electrically evoked release or, rather, overflow of endogenous glutamate in superfusates from rat cortical slices was compared with that of dihydrokainate. In the absence of these presumed uptake inhibitors, electrical stimulation for 4 min at 1 Hz did not elicit a measurable glutamate overflow over baseline at all. Basal overflow increased concentration-dependently in the presence of 10-100 microM L-trans-PDC, about 5-fold at 100 microM. Also, electrical stimulation caused increases of glutamate overflow over basal levels progressive with increasing concentrations of trans-PDC; a stimulated overflow corresponding to about 50% of basal overflow was obtained at 100 microM. Basal as well as evoked release in the presence of dihydrokainate did not exceed ca. 60% of that obtained with 100 microM L-trans-PDC. In synaptosomes, L-trans-PDC much more than dihydrokainate caused a transient increase of spontaneous glutamate release which was diminished in the absence of Na+, indicating that it is transported into the cytoplasm by the glutamate carrier and induces some efflux of the amino acid from this compartment. Moreover, trans-PDC caused a weak to moderate inhibition of K(+)-evoked glutamate release from synaptosomes at 10-300 microM, without obvious concentration-dependence. Glutamate overflow elicited from rat cortical slices by electrical field stimulation at 1 Hz was Ca(2+)-dependent to about 80%. Tetrodotoxin (0.3 microM) reduced it by about 90%.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cerebral Cortex/metabolism , Dicarboxylic Acids/pharmacology , Glutamates/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Pyrrolidines/pharmacology , Animals , Baclofen/pharmacology , Calcium/physiology , Cerebral Cortex/drug effects , Chromatography, High Pressure Liquid , Electric Stimulation , GABA-B Receptor Antagonists , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Male , Nipecotic Acids/pharmacology , Phosphinic Acids/pharmacology , Potassium/pharmacology , Propanolamines/pharmacology , Rats , Rats, Inbred Strains , Synaptosomes/drug effects , Synaptosomes/metabolism , Tetrodotoxin/pharmacology
18.
Neurochem Res ; 18(11): 1163-7, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8255368

ABSTRACT

CGP 28,014 is a specific inhibitor of catechol-O-methyltransferase (COMT) in vivo. In humans, the inhibition was assessed by measuring urinary excretion of isoquinolines and with the levodopa test. Following administration of CGP 28,014, urinary excretion of isoquinolines was significantly increased. In rats, CGP 28,014 reduced plasma and striatal concentrations of 3-O-methyldopa (30MD) in a dose-dependent manner. Acute and subchronic administration of CGP 28,014 alone or in combination with the peripherally acting decarboxylase inhibitor benserazide decreased plasma 30MD as an index of COMT inhibition by about 50%. There seems to be a close relationship between the time-course of plasma concentrations of CGP 28,014 and the extent of COMT inhibition assessed by the 30MD/DOPA ratio in plasma.


Subject(s)
Amidines/pharmacology , Catechol O-Methyltransferase Inhibitors , Pyridones/pharmacology , Amidines/administration & dosage , Amidines/pharmacokinetics , Animals , Benserazide/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/urine , Dose-Response Relationship, Drug , Homovanillic Acid/urine , Humans , Isoquinolines/urine , Kinetics , Methoxyhydroxyphenylglycol/urine , Norepinephrine/urine , Pyridones/administration & dosage , Pyridones/pharmacokinetics , Rats , Tyrosine/analogs & derivatives , Tyrosine/blood , Tyrosine/metabolism , Vanilmandelic Acid/urine
19.
Biochem Pharmacol ; 45(12): 2417-24, 1993 Jun 22.
Article in English | MEDLINE | ID: mdl-7687131

ABSTRACT

The orally active iron chelator, 1,2-dimethyl-3-hydroxypyridin-4-one (L1, CP20) proposed for reduction of iron overload in hemoglobinopathic patients, was studied in rats with respect to its ability to interfere with dopamine (DA) and serotonin (5-HT) metabolism. At 100 mg/kg i.p., it reduced the levels of DA, 5-HT, 5-hydroxyindoleacetic acid and particularly homovanillic acid in the rat striatum for several hours. These effects were shown to result from concomitant inhibition of catechol-O-methyltransferase (COMT; EC2.1.1.6), tyrosine [tyrosine, tetrahydropteridine: oxygen oxidoreductase (3-hydroxylating) (EC 1.14.16.2)] and tryptophan hydroxylase [tryptophan, tetrahydropteridine: oxygen oxidoreductase (5-hydroxylating) (EC 1.14.16.4)], with similar time-courses. COMT was inhibited with a threshold dose of about 1 mg/kg i.p. and an ED50 of about 10 mg/kg i.p. as determined by the conversion of exogenous L-dihydroxyphenylalanine (L-DOPA) to its O-methylated derivative. Tyrosine and tryptophan hydroxylase activities as measured by the accumulation of DOPA and 5-hydroxytryptophan, respectively, after central decarboxylase inhibition, were inhibited in striatum and cortex, with threshold doses of 3-10 mg/kg and ED50s of about 20-30 mg/kg i.p. or p.o. While COMT inhibition by L1 is probably related to the structural similarity of the latter drug with the normal enzyme substrates, tyrosine and tryptophan hydroxylase inhibition is more likely due to coordination to iron bound to these enzymes. Desferrioxamine at 100 mg/kg i.p. did not show comparable effects. It is not known whether this relates to poor brain and/or cell penetration, or whether multidentate chelators are less suitable as inhibitors of aromatic amino acid hydroxylases.


Subject(s)
Catechol O-Methyltransferase Inhibitors , Corpus Striatum/drug effects , Iron Chelating Agents/pharmacology , Pyridones/pharmacology , Tryptophan Hydroxylase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Administration, Oral , Animals , Corpus Striatum/metabolism , Deferiprone , Deferoxamine/pharmacology , Dopamine/biosynthesis , Dopamine/pharmacology , Female , Hydroxyindoleacetic Acid/metabolism , Male , Rats , Serotonin/biosynthesis , Tyrosine/analogs & derivatives , Tyrosine/metabolism
20.
Naunyn Schmiedebergs Arch Pharmacol ; 347(5): 514-20, 1993 May.
Article in English | MEDLINE | ID: mdl-8391653

ABSTRACT

While the role of GABAB autoreceptors in the regulation of GABA release in synaptosomes and brain slices is well established, little is known about their role in vivo. Doubts have arisen because there is an apparent discrepancy between the frequencies at which GABA neurons fire and the frequency range within which autoreceptor regulation is observed in vitro. To see whether this apparent mismatch could be due to the use of a GABA uptake inhibitor in the release experiments in slices, we have compared the frequency dependencies of GABA release in the presence and absence of uptake inhibition. Before-hand, the previously incomplete frequency curve in the presence of uptake inhibition was extended at the lower end. To achieve this, stimulation was performed by means of groups of 4 pseudo-one-pulses (POP's) at inter-POP intervals corresponding to frequencies of 0.015625-0.5 Hz. It could be shown that activation of the GABAB autoreceptor by endogenously released GABA begins at a stimulation frequency as low as 0.0625 Hz. Experiments with the antagonist, CGP 35348, at inter-POP intervals of 1 min, at which the preceding POP has no longer an effect on GABA release during the next one, showed that basal release alone already substantially activated the autoreceptor. The frequency dependence in the absence as compared to the presence of uptake inhibition was shifted towards higher frequencies by a factor of 4. We do not consider this enough to remove our doubts about the in vivo operativity of GABAB autoreceptors.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Brain/drug effects , GABA-A Receptor Antagonists , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Bicuculline/pharmacology , Brain/metabolism , Electric Stimulation , Male , Muscimol/pharmacology , Nipecotic Acids/pharmacology , Organophosphorus Compounds/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...