Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 203(2): 512-20, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17049515

ABSTRACT

Parkinson's disease (PD) is associated with loss of total glutathione (GSH) which may contribute to progressive cell death. Peripheral GSH administration has been used clinically with reported benefits. Despite this, there is little specific information to characterize its cellular uptake or clearance, brain elevation with peripheral delivery or neuroprotective efficacy in PD models. The current study was carried out to provide this information using in vitro and in vivo approaches. In rat mesencephalic culture, the monoethyl ester of GSH (GEE), but not GSH (1-10 mM, 24 h) produced a dose-dependent elevation in GSH. The half-life for clearance was 10.14 h and was not different in cells depleted of GSH prior to loading. Elevation of GSH with GEE protected neurons from oxidative stress with H2O2 or metabolic stress with the complex I and II inhibitors MPP+ and malonate, respectively. To determine if peripheral administration of GEE could elevate brain GSH levels, rats were administered 0.1-50 mg/kg/day GEE via osmotic minipump either subcutaneously (sc) or via a cannula placed into the left cerebral ventricle (icv) for 28 days. Only central delivery of GEE resulted in significant elevations of brain GSH. Elevation of brain GSH by icv infusion of GEE was examined for its neuroprotective effects against chronic central delivery of MPP+. Infusion of 0.142 mg/kg/day MPP+ for 28 days caused a selective ipsilateral loss of striatal dopamine. Co-infusion of MPP+ with 10 mg/kg/day GEE significantly protected against striatal dopamine loss. These findings show that the ethyl ester of GSH but not GSH per se can elevate intracellular GSH, that brain elevation of GSH requires central delivery of the ethyl ester and that this elevation provides neuroprotection against oxidative stress or chronic mitochondrial impairment.


Subject(s)
Brain Chemistry/drug effects , Glutathione/analogs & derivatives , Glutathione/metabolism , Glutathione/pharmacology , Neurons/metabolism , Parkinson Disease/metabolism , Animals , Cell Count , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Dopamine/metabolism , Female , Half-Life , Injections, Intraventricular , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism
2.
Exp Neurol ; 200(1): 172-83, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16546169

ABSTRACT

Mitochondrial dysfunction is observed in sporadic Parkinson's disease (PD) and may contribute to progressive neurodegeneration. While acute models of mitochondrial dysfunction have been used for many years to investigate PD, chronic models may better replicate the cellular disturbances caused by long-standing mitochondrial derangements and may represent a better model for neurotherapeutic testing. This study sought to develop a chronic model of PD that has the advantages of continuous low level toxin delivery, low mortality, unilateral damage to minimize aphagia and adipsia as well as minimal animal handling to reduce stress-related confounds. Infusion by osmotic minipump of the complex I toxin, 1-methyl-4-phenylpyridinium (MPP+), for 28 days into the left cerebral ventricle in rats caused a selective ipsilateral loss of nigral tyrosine hydroxylase immunoreactive somata (35% loss). In animals that were sacrificed 14 days after the chronic MPP+ administration, there was an even greater loss of nigral tyrosine hydroxylase cells (65% loss). Lewy-body-like structures that stained positive for ubiquitin and alpha-synuclein were found in striatal neurons near the infusion site but were not observed in nigral neurons. At the electron microscope level, however, swollen and abnormal mitochondria were observed in the nigral dopamine neurons, which may represent the early formation of an inclusion body. There were no animal deaths with the chronic treatment regimen that was utilized, and the magnitude of nigrostriatal neuronal loss was relatively consistent among the animals. This model of progressive neurodegeneration of nigrostriatal dopamine neurons may be useful for studying neuroprotective therapeutic agents for PD.


Subject(s)
1-Methyl-4-phenylpyridinium/administration & dosage , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Injections, Intraventricular , Male , Parkinson Disease, Secondary/mortality , Rats , Rats, Sprague-Dawley , Survival Rate
3.
J Neurochem ; 79(1): 152-60, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595767

ABSTRACT

The neurotoxic actions of methamphetamine (METH) may be mediated in part by reactive oxygen species (ROS). Methamphetamine administration leads to increases in ROS formation and lipid peroxidation in rodent brain; however, the extent to which proteins may be modified or whether affected brain regions exhibit similar elevations of lipid and protein oxidative markers have not been investigated. In this study we measured concentrations of TBARs, protein carbonyls and monoamines in various mouse brain regions at 4 h and 24 h after the last of four injections of METH (10 mg/kg/injection q 2 h). Substantial increases in TBARs and protein carbonyls were observed in the striatum and hippocampus but not the frontal cortex nor the cerebellum of METH-treated mice. Furthermore, lipid and protein oxidative markers were highly correlated within each brain region. In the hippocampus and striatum elevations in oxidative markers were significantly greater at 24 h than at 4 h. Monoamine levels were maximally reduced within 4 h (striatal dopamine [DA] by 95% and serotonin [5-HT] in striatum, cortex and hippocampus by 60-90%). These decrements persisted for 7 days after METH, indicating effects reflective of nerve terminal damage. Interestingly, NE was only transiently depleted in the brain regions investigated (hippocampus and cortex), suggesting a pharmacological and non-toxic action of METH on the noradrenergic nerve terminals. This study provides the first evidence for concurrent formation of lipid and protein markers of oxidative stress in several brain regions of mice that are severely affected by large neurotoxic doses of METH. Moreover, the differential time course for monoamine depletion and the elevations in oxidative markers indicate that the source of oxidative stress is not derived directly from DA or 5HT oxidation.


Subject(s)
Biomarkers/analysis , Brain/drug effects , Brain/metabolism , Lipid Peroxidation , Methamphetamine/pharmacology , Nerve Tissue Proteins/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Biogenic Monoamines/metabolism , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydroxyindoleacetic Acid/metabolism , Male , Mice , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Serotonin/metabolism , Thiobarbituric Acid Reactive Substances/analysis
4.
J Pharmacol Exp Ther ; 298(3): 925-33, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504786

ABSTRACT

Disturbance in phosphorylation/dephosphorylation can trigger apoptosis. Little is known as to its effects on mesencephalic dopamine neurons, the major neurons lost in Parkinson's disease. In this study, okadaic acid (OKA), a phosphatase 1 and 2A inhibitor, with greater potency toward 2A, was toxic to mesencephalic dopamine and gamma-aminobutyric acid (GABA) neurons, however, dopamine neurons were 4-fold more sensitive. The EC(50) for dopamine versus GABA toxicity was 1.5 versus 6.5 nM, respectively, and was consistent with an inhibition of phosphatase 2A. Dopamine neurons were also more sensitive to calyculin-A, a phosphatase inhibitor equipotent toward 1 and 2A. OKA-methyl-ester, which lacks phosphatase inhibitory activity, was without effect. DNA laddering typical of apoptosis was observed in cultures at a concentration that was specifically toxic to dopamine neurons (5 nM). In contrast to the sensitivity of mesencephalic neurons to phosphatase inhibition, inhibition of protein kinase activity with staurosporine or K252a showed little toxicity and protected neurons from OKA. Consistent with in vitro findings, infusion of 32 to 320 pmol of OKA into the left striatum of rats caused a dose-dependent loss of striatal dopamine without any loss of GABA 1 week following infusion. Acutely, OKA increased tyrosine hydroxylase activity, a phosphatase 2A substrate, and increased dopamine turnover. The above-mentioned findings demonstrate that dysregulation of phosphatase activity is detrimental to mesencephalic neurons, with dopamine neurons, in vitro and in vivo, being relatively more sensitive to phosphatase 2A inhibition. Disturbances in the phosphorylation control of proteins unique to dopamine neurons may contribute to their enhanced vulnerability to OKA exposure.


Subject(s)
Enzyme Inhibitors/pharmacology , Mesencephalon/cytology , Mesencephalon/drug effects , Neurons/drug effects , Phosphoprotein Phosphatases/antagonists & inhibitors , Animals , Cell Count , Cells, Cultured , DNA Fragmentation , Dopamine/metabolism , Immunohistochemistry , Injections , Male , Neostriatum/physiology , Okadaic Acid/administration & dosage , Okadaic Acid/pharmacology , Phosphorylation , Protein Phosphatase 1 , Protein Phosphatase 2 , Rats , Rats, Sprague-Dawley , Staurosporine/administration & dosage , Staurosporine/pharmacology , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
5.
Brain Res ; 910(1-2): 116-25, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11489261

ABSTRACT

The vesicular monoamine transporter 2 (VMAT2) has sequence homology with bacterial multidrug transporters which in turn share homology with mammalian P-glycoprotein (P-GP). Both VMAT2 and P-GP can detoxify cells. 1-Methyl-4-phenylpyridinium (MPP(+)), the toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), is a substrate for VMAT2 that has several structural features in common with P-GP substrates and inhibitors. The present studies investigated whether P-GP is responsible for the elimination of MPP(+) from the brain. Additionally, VMAT2 and P-GP are inhibited by many of the same compounds. Thus we also investigated whether VMAT2 inhibitors could block P-GP in vitro and vice versa whether P-GP inhibitors could block VMAT2 mediated transport of [3H]-DA into synaptic vesicles. In mice treated with MPTP and a P-GP inhibitor (quinidine, trans-flupentixol or cyclosporine A), the elimination of MPP(+) from the striatum was significantly delayed. However, in experiments using various cell lines expressing either mouse or human P-GP, MPP(+) did not reverse the P-GP mediated resistance to vincristine, suggesting that MPP(+) is a poor substrate for P-GP. Additional experiments were performed using mdr1a/b double knockout mice which lack functional P-GP encoded by these two genes. Data from mdr1a/b knockout mice treated with MPTP also suggest that MPP(+) is not extruded from the brain by P-GP. In other studies, we demonstrated that the VMAT2 inhibitors tetrabenazine and Ro 4-1284 inhibit P-GP and that the P-GP inhibitors trans-flupentixol and quinidine inhibit VMAT2. Thus, several new drugs can be added to the list of compounds that are able to inhibit both VMAT2 and P-GP, providing further evidence of the similarity between these two transporters.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics , 1-Methyl-4-phenylpyridinium/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Brain/drug effects , Drug Interactions/physiology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Transport Proteins , Neuropeptides , Parkinsonian Disorders/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenylpyridinium/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Brain/metabolism , Brain/physiopathology , Calcium Channel Blockers/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Parkinsonian Disorders/physiopathology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Tumor Cells, Cultured , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
6.
J Neurosci ; 21(10): RC143, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11319241

ABSTRACT

Recent epidemiological studies have established an association between the common consumption of coffee or other caffeinated beverages and a reduced risk of developing Parkinson's disease (PD). To explore the possibility that caffeine helps prevent the dopaminergic deficits characteristic of PD, we investigated the effects of caffeine and the adenosine receptor subtypes through which it may act in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxin model of PD. Caffeine, at doses comparable to those of typical human exposure, attenuated MPTP-induced loss of striatal dopamine and dopamine transporter binding sites. The effects of caffeine were mimicked by several A(2A) antagonists (7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH 58261), 3,7-dimethyl-1-propargylxanthine, and (E)-1,3-diethyl-8 (KW-6002)-(3,4-dimethoxystyryl)-7-methyl-3,7-dihydro-1H-purine-2,6-dione) (KW-6002) and by genetic inactivation of the A(2A) receptor, but not by A(1) receptor blockade with 8-cyclopentyl-1,3-dipropylxanthine, suggesting that caffeine attenuates MPTP toxicity by A(2A) receptor blockade. These data establish a potential neural basis for the inverse association of caffeine with the development of PD, and they enhance the potential of A(2A) antagonists as a novel treatment for this neurodegenerative disease.


Subject(s)
Caffeine/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/drug therapy , Purinergic P1 Receptor Antagonists , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Catechols/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Immunity, Innate/genetics , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/genetics , Purines/administration & dosage , Pyrimidines/administration & dosage , Receptor, Adenosine A2A , Receptors, Purinergic P1/deficiency , Receptors, Purinergic P1/genetics , Theobromine/administration & dosage , Theobromine/analogs & derivatives , Triazoles/administration & dosage , Xanthines/administration & dosage
7.
Neuroscience ; 101(4): 1063-9, 2000.
Article in English | MEDLINE | ID: mdl-11113355

ABSTRACT

The vesicular monoamine transporter in the brain can sequester the neurotoxin 1-methyl-4-phenylpyridinium into synaptic vesicles and protect catecholamine-containing neurons from degeneration. Mouse nigrostriatal dopaminergic neurons, and to a lesser extent locus coeruleus noradrenergic neurons, are vulnerable to toxicity produced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. The present study sought to determine whether pharmacological inactivation of the vesicular monoamine transporter in the brain would enhance the degeneration of substantia nigra dopaminergic neurons and locus coeruleus noradrenergic neurons in 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine-treated animals. Mice were treated subacutely with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine alone, or in combination with vesicular monoamine transporter inhibitors (tetrabenazine or Ro4-1284), and 10-24 days later striatal dopamine and cortical norepinephrine levels were measured using chromatographic methods. In the same animals, substantia nigra and locus coeruleus catecholaminergic neurons were counted using tyrosine hydroxylase immunohistochemical staining with computer imaging techniques. Mice in which pharmacological blockage of the vesicular monoamine transporter enhanced the effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity in the depletion of striatal dopamine concentrations also exhibited enhanced degeneration of substantia nigra dopaminergic neurons. In the same animals, however, vesicular monoamine transporter blockade did not enhance the effects of 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine in the locus coeruleus noradrenergic system. These data are consistent with the hypothesis that the vesicular monoamine transporter can protect catecholamine-containing neurons from 1-methyl-4-phenylpyridinium-induced degeneration by sequestration of the toxin within brain vesicular monoamine transporter-containing synaptic vesicles. Since the amount of vesicular monoamine transporter in locus coeruleus neurons is more than in substantia nigra neurons, and because 1-methyl-4-phenylpyridinium is sequestered within locus coeruleus neurons to a far greater extent than within substantia nigra neurons, it may be that a greater amount of vesicular monoamine transporter inhibition is required for 1-methyl-4-phenylpyridinium to be toxic to locus coeruleus neurons than to substantia nigra dopaminergic neurons.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Transport Proteins , Mesencephalon/physiopathology , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/physiology , Neuropeptides , 2H-Benzo(a)quinolizin-2-ol, 2-Ethyl-1,3,4,6,7,11b-hexahydro-3-isobutyl-9,10-dimethoxy-/pharmacology , Animals , Cell Count , Drug Synergism , Locus Coeruleus/physiology , Male , Mesencephalon/pathology , Mice , Nerve Degeneration/pathology , Neurons/pathology , Norepinephrine/physiology , Tetrabenazine/pharmacology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
8.
J Neurochem ; 74(5): 2217-20, 2000 May.
Article in English | MEDLINE | ID: mdl-10800969

ABSTRACT

[3H]Dihydrotetrabenazine ([3H]DTBZ), a specific ligand for the vesicular monoamine transporter (VMAT2), has been used to characterize the integrity of monoaminergic nerve terminals in experimental animals and humans. The purpose of the present studies was to compare the loss of VMAT2 binding with the loss of other neurochemical markers of the dopamine (DA) nerve terminals in mice treated with neurotoxic doses of methamphetamine (METH) or MPTP. Profound decreases (> or =70%) in DA content, tyrosine hydroxylase activity, and PH]carbomethoxy-3-(4-fluorophenyl)tropane binding to the DA transporter were observed in striatal homogenates at both 1 and 6 days after exposure to the neurotoxins. It is surprising that no significant loss of [3H]DTBZ binding in the homogenates was observed at 1 day after exposure, although a significant loss (-50%) was apparent 6 days later. However, in isolated vesicle preparations, [3H]DTBZ binding and active [3H]DA uptake were markedly reduced (>70%) at 1 day. These observations indicate that vesicle function is compromised at an early time point after exposure to neurotoxic insult. Furthermore, the changes in [H]DTBZ binding in homogenates may not be a sensitive indicator of early damage to synaptic vesicles, although homogenate binding reliably identifies a loss of VMAT2 at later times.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Central Nervous System Stimulants/pharmacology , Dopamine Agents/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Methamphetamine/pharmacology , Neuropeptides , Animals , Corpus Striatum/metabolism , Dopamine/metabolism , Histological Techniques , Male , Mice , Synaptic Vesicles/metabolism , Synaptosomes/metabolism , Tetrabenazine/analogs & derivatives , Tetrabenazine/pharmacology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
9.
J Pharmacol Exp Ther ; 293(2): 336-42, 2000 May.
Article in English | MEDLINE | ID: mdl-10773000

ABSTRACT

Dopamine neurons from various animal species differ in sensitivity to the neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium (MPP(+)). Compared with striatal vesicles isolated from mice, those from rats have a higher density of the brain vesicular monoamine transporter (VMAT2) and a greater ability to sequester MPP(+), suggesting a larger storage capacity for MPP(+) in rat vesicles. In the present study, we examined whether striatal VMAT2-containing vesicles might provide protection against the neurotoxic effects of MPP(+) in vivo. Dose-response curves for striatally infused MPP(+) were determined in animals pretreated with or without a VMAT2 inhibitor. Ro 4-1284 administration (10 mg/kg i.p.; VMAT2 inhibitor) produced a 5-fold leftward shift in the MPP(+) dose-response curve and a significant lowering of the EC(50) concentration for MPP(+)-induced damage. These findings provide evidence for a substantial accumulation of MPP(+) in VMAT2-containing vesicles in vivo in the rat striatum and support the hypothesis that MPP(+) sequestration in vesicles can provide protection against its toxic actions. In mice, VMAT2 inhibition did not reliably enhance toxicity produced by a striatal infusion of MPP(+) or by systemic administration of MPTP. These data suggest that vesicular sequestration of MPP(+) may be of less importance in mice than in rats as relates to protection from the toxin. The present results also reveal that although VMAT2 inhibition enhanced striatal MPP(+) toxicity in the rat, the potency of MPP(+) in the rat striatum was less than that in mouse striatum. This implies that there are other factors that either exacerbate MPP(+) toxicity in the mouse or attenuate MPP(+) toxicity in rats.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Brain Chemistry/drug effects , Membrane Glycoproteins/antagonists & inhibitors , Membrane Transport Proteins , Neostriatum/metabolism , Neuropeptides , Neurotoxins/toxicity , Neurotransmitter Agents/antagonists & inhibitors , 2H-Benzo(a)quinolizin-2-ol, 2-Ethyl-1,3,4,6,7,11b-hexahydro-3-isobutyl-9,10-dimethoxy-/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Mice , Microdialysis , Neostriatum/drug effects , Rats , Rats, Sprague-Dawley , Species Specificity , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Time Factors , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
10.
J Pharmacol Exp Ther ; 293(2): 329-35, 2000 May.
Article in English | MEDLINE | ID: mdl-10772999

ABSTRACT

Significant differences exist in the sensitivity of mice and rats to the neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) that cannot be explained by differences in exposure to or uptake of 1-methyl-4-phenylpyridinium (MPP(+)) into dopamine (DA) neurons. MPP(+) is also a substrate for the brain vesicular monoamine transporter (VMAT2), and sequestration into synaptic vesicles may be one mechanism of protection against MPP(+) toxicity. A greater sequestration of MPP(+) into vesicles of DA neurons in rats versus mice could explain the lower vulnerability of DA neurons in the rat to MPP(+) toxicity. To test this hypothesis, the kinetics of uptake for [(3)H]MPP(+) and [(3)H]DA as well as [(3)H]dihydrotetrabenazine binding to VMAT2 were compared in vesicles isolated from the striata of rats and mice. The K(m) value of [(3)H]MPP(+) transport was similar in the two species. In contrast, the maximal transport rate (V(max)) was 2-fold greater in vesicles from rats than in those from mice. Likewise, the K(m) value for [(3)H]DA transport was similar in both preparations, but the V(max) value was 2-fold greater in rat than in mouse vesicles. The B(max) value for [(3)H]dihydrotetrabenazine binding was also 2-fold greater in striatal vesicles from rats than in those from mice. Electron micrographs demonstrated that vesicles isolated from rats and mice were approximately the same size. Based on these observations, we propose that striatal vesicles from rats have more VMAT2 than vesicles from mice and that this species difference in VMAT2 density may help explain the reduced vulnerability of rat DA neurons to MPP(+) neurotoxicity.


Subject(s)
1-Methyl-4-phenylpyridinium/metabolism , Dopamine Agents/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Neostriatum/metabolism , Neuropeptides , Neurotoxins/metabolism , Neurotransmitter Agents/metabolism , Synaptic Vesicles/metabolism , Animals , Cocaine/analogs & derivatives , Cocaine/metabolism , Dopamine Uptake Inhibitors/metabolism , Indicators and Reagents , Kinetics , Male , Mice , Microscopy, Electron , Neostriatum/drug effects , Neostriatum/ultrastructure , Rats , Rats, Sprague-Dawley , Species Specificity , Synaptic Vesicles/drug effects , Synaptic Vesicles/ultrastructure , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
11.
J Neurochem ; 74(4): 1656-65, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10737624

ABSTRACT

Defects in mitochondrial energy metabolism have been implicated in the pathology of several neurodegenerative disorders. In addition, the reactive metabolites generated from the metabolism and oxidation of the neurotransmitter dopamine (DA) are thought to contribute to the damage to neurons of the basal ganglia. We have previously demonstrated that infusions of the metabolic inhibitor malonate into the striata of mice or rats produce degeneration of DA nerve terminals. In the present studies, we demonstrate that an intrastriatal infusion of malonate induces a substantial increase in DA efflux in awake, behaving mice as measured by in vivo microdialysis. Furthermore, pretreatment of mice with tetrabenazine (TBZ) or the TBZ analogue Ro 4-1284 (Ro-4), compounds that reversibly inhibit the vesicular storage of DA, attenuates the malonate-induced DA efflux as well as the damage to DA nerve terminals. Consistent with these findings, the damage to both DA and GABA neurons in mesencephalic cultures by malonate exposure was attenuated by pretreatment with TBZ or Ro-4. Treatment with these compounds did not affect the formation of free radicals or the inhibition of oxidative phosphorylation resulting from malonate exposure alone. Our data suggest that DA plays an important role in the neurotoxicity produced by malonate. These findings provide direct evidence that inhibition of succinate dehydrogenase causes an increase in extracellular DA levels and indicate that bioenergetic defects may contribute to the pathogenesis of chronic neurodegenerative diseases through a mechanism involving DA.


Subject(s)
Corpus Striatum/cytology , Dopamine/physiology , Malonates/pharmacology , Membrane Transport Proteins , Neurons/metabolism , Neuropeptides , 2H-Benzo(a)quinolizin-2-ol, 2-Ethyl-1,3,4,6,7,11b-hexahydro-3-isobutyl-9,10-dimethoxy-/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Biological Transport/drug effects , Biological Transport/physiology , Carbon Radioisotopes , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/pharmacokinetics , Energy Metabolism/drug effects , Energy Metabolism/physiology , Free Radicals/metabolism , Lactic Acid/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Microdialysis , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Neurons/drug effects , Parkinson Disease/metabolism , Presynaptic Terminals/drug effects , Synaptic Vesicles/metabolism , Tetrabenazine/pharmacology , Tritium , Vesicular Biogenic Amine Transport Proteins , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacokinetics
12.
Exp Neurol ; 161(2): 638-46, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10686083

ABSTRACT

Defects in energy metabolism have been detected in patients with Parkinson's disease and have been proposed as a contributing factor in the disease. Previous in vitro studies showed that NMDA receptors contribute to the loss of dopamine neurons caused by the metabolic inhibitor malonate. In vivo, it is not known whether this interaction occurs through a postsynaptic action on the cell body in the substantia nigra or through a presynaptic action at the dopamine terminal in the striatum. So we could discern the anatomical level of NMDA receptor involvement, rats were infused with malonate, either into the left striatum or into the left substantia nigra. NMDA receptors were locally blocked by an intranigral or intrastriatal coinfusion of malonate plus MK-801 followed by a second infusion of MK-801 3 h later. Animals were examined at 1 week for striatal and nigral dopamine and GABA levels. Intranigral infusion of malonate (0.5 micromol) produced an approximate 50% loss of both nigral dopamine and GABA. MK-801 (0.1 micromol) provided significant protection against both nigral dopamine and GABA loss and against anterograde damage to dopamine terminals in the striatum. Intrastriatal administration of malonate (2 micromol) produced a 68 and 35% loss of striatal dopamine and GABA, respectively. In contrast to intranigral administration, intrastriatal blockade of NMDA receptors did not protect against striatal dopamine loss, although GABA loss was significantly attenuated. Core body temperature monitored several hours throughout the experiment was unchanged. Consistent with a lack of effect of NMDA antagonists on malonate-induced toxicity to dopamine neurons in striatum, intrastriatal infusion of NMDA had a pronounced effect on long-term GABA toxicity with little effect of dopamine loss. These findings are consistent with a postsynaptic action of NMDA receptors on mediating toxicity to dopamine neurons during impaired energy metabolism.


Subject(s)
Corpus Striatum/physiology , Dizocilpine Maleate/pharmacology , Malonates/pharmacology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Substantia Nigra/physiology , Animals , Body Temperature , Corpus Striatum/drug effects , Dizocilpine Maleate/administration & dosage , Dopamine/metabolism , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality , Infusions, Parenteral , Male , Malonates/administration & dosage , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , gamma-Aminobutyric Acid/metabolism
13.
Brain Res ; 790(1-2): 264-9, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9593931

ABSTRACT

The neuroprotective effects of lowering body temperature have been well documented in various models of neuronal injury. The present study investigated the effects a lower ambient or core body temperature would have on damage to striatal dopamine (DA) neurons produced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Mice received systemic MPTP treatment at two different temperatures, 4 degrees C and 22 degrees C. MPTP-treated mice maintained at 4 degrees C demonstrated (1) a greater hypothermic response, (2) a significant reduction in striatal DA content and tyrosine hydroxylase (TH) activity, and (3) significantly greater striatal 1-methyl-4-phenylpyridinium (MPP+) levels, as compared to mice dosed with MPTP at room temperature. Parallel studies with methamphetamine (METH) were conducted since temperature appears to play a pivotal role in the mediation of damage to DA neurons by this CNS stimulant in rodents. As previously reported, METH-induced hyperthermia and the subsequent loss of striatal DA content were attenuated in animals dosed at 4 degrees C. We also evaluated the effects a hypothermic state induced by pharmacological agents would have on striatal neurochemistry and MPP+ levels following MPTP treatment. Concurrent administration of MK-801 or 8-OHDPAT increased the striatal MPP+ levels following MPTP treatment. However, only 8-OHDPAT potentiated the MPTP-induced decrements of striatal DA content and TH activity; MK-801 did not affect MPTP decreases in these striatal markers of dopaminergic damage. Altogether, these findings indicate that temperature has a profound effect on striatal MPP+ levels and MPTP-induced damage to DA neurons in mice.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Body Temperature/drug effects , Corpus Striatum/metabolism , Dopamine Agents/toxicity , MPTP Poisoning , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacokinetics , 1-Methyl-4-phenylpyridinium/pharmacokinetics , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Corpus Striatum/cytology , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Dopamine Agents/pharmacokinetics , Excitatory Amino Acid Antagonists/pharmacology , Methamphetamine/pharmacology , Mice , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Neurons/chemistry , Neurons/enzymology , Neurotoxins/pharmacokinetics , Serotonin Receptor Agonists/pharmacology , Tyrosine 3-Monooxygenase/metabolism
14.
Neuroscience ; 84(4): 1177-85, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9578404

ABSTRACT

The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine produces a parkinsonian syndrome in man and experimental animals. The toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, 1-methyl-4-phenylpyridinium, exhibits high-affinity uptake by plasma membrane monoamine transporters and also by the vesicular monoamine transporter. Using autoradiographic and immunohistochemical methods in mice, we demonstrate the accumulation of [3H]1-methyl-4-phenylpyridinium within neurons that contain the vesicular monoamine transporter, following systemic administration of [3H]1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Within 1-24 h following the intraperitoneal administration of 10 microg/kg of [3H]1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, [3H]1-methyl-4-phenylpyridine labelling was found within such regions as the locus coeruleus, dorsal, medial, and pallidal raphe nuclei, substantia nigra pars compacta, ventral tegmental area, and paraventricular nucleus of the hypothalamus. These regions all contain monoaminergic somata as defined by immunohistochemical staining with an antibody against the vesicular monoamine transporter. There was a positive relationship between the density of [3H]1-methyl-4-phenylpyridinium label and the density of vesicular monoamine transporter immunoreactivity: the highest densities of both were found in the locus coeruleus and lowest densities in the midbrain dopaminergic neurons. In addition, [3H]1-methyl-4-phenylpyridinium labelling was detected in the bed nucleus of the stria terminalis and paraventricular nucleus of the thalamus, which also contained vesicular monoamine transporter immunoreactive nerve terminals. The present data indicate that low doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine cause a significant accumulation of 1-methyl-4-phenylpyridinium within monoaminergic somata in parallel with the amount of vesicular monoamine transporter in the neuron. Since nuclei with intense labelling are not damaged by doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine that are toxic to midbrain dopaminergic neurons, these data are consistent with the hypothesis that sequestration of 1-methyl-4-phenylpyridinium within monoaminergic synaptic vesicles can protect the neurons from degeneration caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine.


Subject(s)
1-Methyl-4-phenylpyridinium/metabolism , Dopamine Agents/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Neurons/metabolism , Neuropeptides , Neurotoxins/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Autoradiography , Brain/pathology , Dopamine Agents/pharmacology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurotoxins/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
15.
Amino Acids ; 14(1-3): 69-74, 1998.
Article in English | MEDLINE | ID: mdl-9871444

ABSTRACT

Although controversial, studies with methamphetamine and MPTP suggest a link between glutamate-mediated excitotoxicity and degeneration of dopamine cells. Both compounds are thought to create a metabolic stress. To further explore glutamate actions in DA degeneration, we investigated the effects of other metabolic inhibitors. In mesencephalic cultures, DA cell loss produced by 3-NPA or malonate was potentiated by NMDA and prevented by MK-801. In vivo, striatal DA loss produced by intranigral infusions of malonate was also potentiated by intranigral NMDA and prevented by systemic MK-801. In contrast, systemic MK-801 did not prevent DA loss produced by intrastriatal malonate. Intrastriatal MK-801 or CGS 19755 did attenuate DA loss in METH-treated mice, but was confounded by the findings that METH-induced hyperthermia, an important component in toxicity, was also attenuated. Taken together, the data support the hypothesis of NMDA receptor involvement in degeneration of DA neurons. Furthermore, the data also suggest that this interaction is likely to occur in the substantia nigra rather than in the striatum.


Subject(s)
Dopamine/metabolism , Glutamic Acid/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Disease Models, Animal , Mice , Parkinson Disease/pathology
16.
Brain Res ; 773(1-2): 223-6, 1997 Oct 31.
Article in English | MEDLINE | ID: mdl-9409726

ABSTRACT

This study examined whether damage to dopamine (DA) nerve terminals via inhibition of energy metabolism in the striatum would result in the retrograde loss of cell bodies in the substantia nigra. Infusion of 2 micromol malonate into the left striatum of rats resulted in a 67% loss of striatal DA and a 40% loss of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra. No change in the number of Nissl-positive-TH-negative neurons was observed. These findings demonstrate the retrograde destruction of DA cell bodies in the substantia nigra resulting from energy impairment at their terminal projection site.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Energy Metabolism/physiology , Malonates/toxicity , Substantia Nigra/pathology , Animals , Corpus Striatum/drug effects , Corpus Striatum/pathology , Energy Metabolism/drug effects , Functional Laterality , Infusions, Parenteral , Male , Malonates/administration & dosage , Nerve Endings/drug effects , Nerve Endings/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/analysis
17.
Neuroreport ; 8(15): 3327-31, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9351666

ABSTRACT

The neurotoxin MPTP kills only certain midbrain dopaminergic (DA) neurons to produce a model of Parkinson's disease. The dopamine transporter (DAT) is important to MPTP toxicity because to be neurotoxic, an MPTP metabolite must first gain access to the DA neuron via the DAT. Also, MPTP is less toxic to DA neurons that contain the putative neuroprotective calcium-binding protein calbindin-D28k (CB). The present study examined the relative importance of DAT activity and CB for cellular vulnerability to MPTP-induced degeneration in the C57BL/6 mouse. Cells that were vulnerable to MPTP were found to contain high levels of DAT mRNA, whereas cells that were not vulnerable contained low levels. Also, the few substantia nigra cells remaining after a toxic dose of MPTP contained only low levels of DAT mRNA. However, there was not a strong relationship between cellular resistance to MPTP toxicity and cells containing CB. These data provide in vivo evidence for a direct correlation between midbrain cellular vulnerability to MPTP toxicity and the activity of the DAT.


Subject(s)
Carrier Proteins/biosynthesis , MPTP Poisoning , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins , Mesencephalon/cytology , Mesencephalon/metabolism , Nerve Tissue Proteins , Neurons/metabolism , Neurotoxins/toxicity , RNA, Messenger/biosynthesis , Animals , Calbindin 1 , Calbindins , Dopamine Plasma Membrane Transport Proteins , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Male , Mesencephalon/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , S100 Calcium Binding Protein G/metabolism
18.
J Pharmacol Exp Ther ; 280(3): 1432-44, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9067333

ABSTRACT

Lobeline is currently being developed as a substitution therapy for tobacco smoking cessation. Activation of CNS dopamine (DA) systems results in the reinforcing properties of nicotine. The present study compared the effects of lobeline and nicotine on rat striatum. Both lobeline and nicotine evoked [3H]overflow from striatal slices superfused in the presence of pargyline and nomifensine in the buffer. Marked DA depletion (42-67%) and a concomitant 2-fold increase in dihydroxyphenylacetic acid (DOPAC) in slices superfused with high concentrations (30-100 microM) of lobeline were observed. The effect of nicotine (10 microM) was inhibited in a concentration-dependent manner by mecamylamine (1-100 microM). However, lobeline (0.1-100 microM)-evoked [3H]overflow was calcium-independent, and was not antagonized by mecamylamine (1-100 microM), suggesting a mechanism of action other than stimulation of nicotinic receptors. Lobeline inhibited [3H]DA uptake into synaptosomes (IC50 = 80 +/- 12 microM) and vesicles (IC50 = 0.88 +/- 0.001 microM), whereas nicotine (< or =100 microM) did not inhibit synaptosomal or vesicular [3H]DA uptake. In the absence of pargyline and nomifensine in the buffer, endogenous DA was detected in superfusate only in those slices exposed to the highest concentration (100 microM) of lobeline. However, endogenous DOPAC concentration was increased in a concentration-dependent manner, indicating that lobeline exposure resulted in increased cytosolic DA which was rapidly metabolized to DOPAC. Under these conditions, lobeline (10-100 microM) also significantly depleted (66-85%) DA content; however, no change in DOPAC content was observed. The results suggest that, unlike nicotine, lobeline increases DA release by potent inhibition of DA uptake into synaptic vesicles, and a subsequent alteration in presynaptic DA storage.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Lobeline/pharmacology , Nicotine/pharmacology , Synaptic Vesicles/drug effects , Synaptosomes/drug effects , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Calcium/metabolism , Corpus Striatum/metabolism , Dopamine/administration & dosage , In Vitro Techniques , Male , Mecamylamine/pharmacology , Nicotine/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism , Synaptosomes/metabolism , Tritium
19.
Eur J Pharmacol ; 320(2-3): 111-9, 1997 Feb 12.
Article in English | MEDLINE | ID: mdl-9059843

ABSTRACT

In vitro studies indicate that mesencephalic dopamine neurons are more vulnerable than other neurons to impairment of energy metabolism. Such findings may have bearing on the loss of dopamine neurons in Parkinson's disease, in which mitochondrial deficiencies have been identified, but would only be relevant if the selective vulnerability were maintained in vivo. To examine this, rats were stereotaxically administered various concentrations of the succinate dehydrogenase inhibitor, malonate (0.25-4 mumol), either into the left substantia nigra or striatum. One week following injection, dopamine and gamma-aminobutyric acid (GABA) levels in the mesencephalon and striatum were measured. Intranigral injection of malonate caused nigral dopamine and GABA to be comparably reduced at all doses tested. The 50% dose level for malonate vs. dopamine and GABA loss was 0.39 and 0.42 mumol, respectively. Tyrosine hydroxylase immunocytochemistry of the midbrains of rats which received an intranigral injection of malonate showed normal staining with 0.25 mumol malonate, but almost complete loss of tyrosine hydroxylase positive nigral pars compacta cells with 1 mumol malonate. Intrastriatal injection of malonate produced a loss of both tyrosine hydroxylase activity and dopamine. In contrast to what was seen in substantia nigra, there was a greater loss of dopamine than GABA in striatal regions nearest the injection site. In striatal regions most distal to the injection site, and which received the lowest concentration of malonate due to diffusion, dopamine levels were significantly reduced with all doses of malonate (0.5-4 mumol), whereas GABA levels were unaffected. Intrastriatal coinfusion of succinate along with malonate completely prevented the loss of dopamine and GABA indicating that succinate dehydrogenase inhibition was the cause of toxicity. These findings indicate that dopamine terminals in the striatum of adult rats are selectively more vulnerable than are the GABA neurons to a mild energy impairment.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Malonates/pharmacology , Neurons/metabolism , Substantia Nigra/metabolism , Animals , Axons/drug effects , Axons/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Energy Metabolism/drug effects , Male , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity , Substantia Nigra/cytology , Substantia Nigra/drug effects , gamma-Aminobutyric Acid/metabolism
20.
J Neurochem ; 68(1): 426-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8978755

ABSTRACT

The role of the glutathione system in protecting dopamine neurons from a mild impairment of energy metabolism imposed by the competitive succinate dehydrogenase inhibitor, malonate, was investigated in vitro and in vivo. Treatment of mesencephalic cultures with 10 microM buthionine sulfoxamine for 24 h reduced total glutathione levels in the cultures by 68%. Reduction of cellular glutathione per se was not toxic to the dopamine population, but potentiated toxicity when the cultures were exposed to malonate. In contrast, transgenic mice overexpressing glutathione peroxidase (hGPE) that received an intrastriatal infusion of malonate (3 mumol) into the left side had significantly less loss of striatal dopamine than their hGPE-negative littermates when assayed 1 week following infusion. These studies demonstrate that manipulation of the glutathione system influences susceptibility of dopamine neurons to damage due to energy impairment. The findings may provide insight into the loss of dopamine neurons in Parkinson's disease in which defects in both energy metabolism and the glutathione system have been identified.


Subject(s)
Dopamine/metabolism , Energy Metabolism , Glutathione Peroxidase/metabolism , Mesencephalon/metabolism , Oxidative Stress/physiology , Animals , Buthionine Sulfoximine/pharmacology , Cells, Cultured , Energy Metabolism/drug effects , Glutathione Peroxidase/genetics , Humans , Malonates/pharmacology , Mesencephalon/cytology , Mice , Mice, Transgenic/genetics , Neurons/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...