Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 18015, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269775

ABSTRACT

Aberrant integration of newborn hippocampal granule cells is hypothesized to contribute to the development of temporal lobe epilepsy. To test this hypothesis, we used a diphtheria toxin receptor expression system to selectively ablate these cells from the epileptic mouse brain. Epileptogenesis was initiated using the pilocarpine status epilepticus model in male and female mice. Continuous EEG monitoring was begun 2-3 months after pilocarpine treatment. Four weeks into the EEG recording period, at a time when spontaneous seizures were frequent, mice were treated with diphtheria toxin to ablate peri-insult generated newborn granule cells, which were born in the weeks just before and after pilocarpine treatment. EEG monitoring continued for another month after cell ablation. Ablation halted epilepsy progression relative to untreated epileptic mice; the latter showing a significant and dramatic 300% increase in seizure frequency. This increase was prevented in treated mice. Ablation did not, however, cause an immediate reduction in seizures, suggesting that peri-insult generated cells mediate epileptogenesis, but that seizures per se are initiated elsewhere in the circuit. These findings demonstrate that targeted ablation of newborn granule cells can produce a striking improvement in disease course, and that the treatment can be effective when applied months after disease onset.


Subject(s)
Hippocampus/surgery , Neurons/physiology , Neurosurgical Procedures/methods , Status Epilepticus/surgery , Animals , Disease Models, Animal , Disease Progression , Electroencephalography , Female , Hippocampus/physiopathology , Male , Mice , Mice, Transgenic , Pilocarpine , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Treatment Outcome
2.
Epilepsia ; 58(12): e162-e166, 2017 12.
Article in English | MEDLINE | ID: mdl-29105060

ABSTRACT

The endocannabinoid system has gained attention as an important modulator of activity in the central nervous system. Initial studies focused on cannabinoid receptor 1 (CB1), which is widely expressed in the brain, but recent work also implicates cannabinoid receptor 2 (CB2) in modulating neuronal activity. Both receptors are capable of reducing neuronal activity, generating interest in cannabinoid receptor agonists as potential anticonvulsants. CB1 (Cnr1) and CB2 (Cnr2) single-knockout mice have been generated, with the former showing heightened seizure sensitivity, but not overt seizures. Given overlapping and complementary functions of CB1 and CB2 receptors, we queried whether double-knockout mice would show an exacerbated neurological phenotype. Strikingly, 30% of double-knockout mice exhibited provoked behavioral seizures, and 80% were found to be epileptic following 24/7 video-electroencephalographic monitoring. Single-knockout animals did not exhibit seizures. These findings highlight the importance of the endocannabinoid system for maintaining network stability.


Subject(s)
Epilepsy/genetics , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Animals , Behavior, Animal , Electroencephalography , Epilepsy/etiology , Handling, Psychological , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Net/physiopathology , Phenotype , Seizures/etiology , Seizures/genetics , Seizures/psychology
3.
Neurobiol Dis ; 108: 339-351, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28855130

ABSTRACT

Deletion of the mTOR pathway inhibitor PTEN from postnatally-generated hippocampal dentate granule cells causes epilepsy. Here, we conducted field potential, whole cell recording and single cell morphology studies to begin to elucidate the mechanisms by which granule cell-specific PTEN-loss produces disease. Cells from both male and female mice were recorded to identify sex-specific effects. PTEN knockout granule cells showed altered intrinsic excitability, evident as a tendency to fire in bursts. PTEN knockout granule cells also exhibited increased frequency of spontaneous excitatory synaptic currents (sEPSCs) and decreased frequency of inhibitory currents (sIPSCs), further indicative of a shift towards hyperexcitability. Morphological studies of PTEN knockout granule cells revealed larger dendritic trees, more dendritic branches and an impairment of dendrite self-avoidance. Finally, cells from both female control and female knockout mice received more sEPSCs and more sIPSCs than corresponding male cells. Despite the difference, the net effect produced statistically equivalent EPSC/IPSC ratios. Consistent with this latter observation, extracellularly evoked responses in hippocampal slices were similar between male and female knockouts. Both groups of knockouts were abnormal relative to controls. Together, these studies reveal a host of physiological and morphological changes among PTEN knockout cells likely to underlie epileptogenic activity. SIGNIFICANCE STATEMENT: Hyperactivation of the mTOR pathway is associated with numerous neurological diseases, including autism and epilepsy. Here, we demonstrate that deletion of the mTOR negative regulator, PTEN, from a subset of hippocampal dentate granule impairs dendritic patterning, increases excitatory input and decreases inhibitory input. We further demonstrate that while granule cells from female mice receive more excitatory and inhibitory input than males, PTEN deletion produces mostly similar changes in both sexes. Together, these studies provide new insights into how the relatively small number (≈200,000) of PTEN knockout granule cells instigates the development of the profound epilepsy syndrome evident in both male and female animals in this model.


Subject(s)
Hippocampus/metabolism , Hippocampus/pathology , Neurons/metabolism , Neurons/pathology , PTEN Phosphohydrolase/deficiency , Sex Characteristics , Animals , Cell Count , Epilepsy/metabolism , Epilepsy/pathology , Female , Immunohistochemistry , Male , Membrane Potentials/physiology , Mice, Knockout , Microscopy, Confocal , Neural Inhibition/physiology , PTEN Phosphohydrolase/genetics , Patch-Clamp Techniques , Tissue Culture Techniques
4.
Neurochem Res ; 42(7): 2024-2032, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28462450

ABSTRACT

Status epilepticus is a common manifestation of nerve agent toxicity and represents a serious medical emergency with high rates of mortality and neurologic injury in those that survive. The aim of the current study was to determine if targeting oxidative stress with the catalytic antioxidant, AEOL10150, would reduce pilocarpine-induced mortality and attenuate neuronal death and neuroinflammation. We found that treatment with AEOL10150 in conjunction with scopolamine and diazepam following pilocarpine-induced SE was able to significantly reduce mortality compared to treatment with just scopolamine and diazepam. Mortality was further reduced when AEOL10150 was used in conjunction with atropine and diazepam which is considered the standard of care for nerve agent exposures. Both treatment paradigms offered significant protection against SE-induced oxidative stress. Additionally, treatment with scopolamine, AEOL10150 and diazepam attenuated SE-induced neuronal loss and neuroinflammation. Taken together, the data suggest that pharmacological targeting of oxidative stress can improve survival and attenuate secondary neurological damage following SE induced by the nerve agent surrogate pilocarpine.


Subject(s)
Anticonvulsants/therapeutic use , Antioxidants/therapeutic use , Hippocampus/metabolism , Oxidative Stress/physiology , Status Epilepticus/metabolism , Status Epilepticus/mortality , Animals , Anticonvulsants/pharmacology , Antioxidants/pharmacology , Hippocampus/drug effects , Male , Oxidative Stress/drug effects , Pilocarpine/toxicity , Rats , Rats, Sprague-Dawley , Status Epilepticus/drug therapy
5.
Cell Rep ; 17(1): 37-45, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27681419

ABSTRACT

Seizures are bursts of excessive synchronized neuronal activity, suggesting that mechanisms controlling brain excitability are compromised. The voltage-gated potassium channel Kv4.2, a major mediator of hyperpolarizing A-type currents in the brain, is a crucial regulator of neuronal excitability. Kv4.2 expression levels are reduced following seizures and in epilepsy, but the underlying mechanisms remain unclear. Here, we report that Kv4.2 mRNA is recruited to the RNA-induced silencing complex shortly after status epilepticus in mice and after kainic acid treatment of hippocampal neurons, coincident with reduction of Kv4.2 protein. We show that the microRNA miR-324-5p inhibits Kv4.2 protein expression and that antagonizing miR-324-5p is neuroprotective and seizure suppressive. MiR-324-5p inhibition also blocks kainic-acid-induced reduction of Kv4.2 protein in vitro and in vivo and delays kainic-acid-induced seizure onset in wild-type but not in Kcnd2 knockout mice. These results reveal an important role for miR-324-5p-mediated silencing of Kv4.2 in seizure onset.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , MicroRNAs/genetics , Seizures/genetics , Shal Potassium Channels/genetics , Status Epilepticus/genetics , Animals , Antagomirs/genetics , Antagomirs/metabolism , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Seizures/chemically induced , Seizures/pathology , Seizures/prevention & control , Shal Potassium Channels/metabolism , Signal Transduction , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Status Epilepticus/prevention & control
6.
eNeuro ; 3(2)2016.
Article in English | MEDLINE | ID: mdl-27066534

ABSTRACT

Altered metabolism is an important feature of many epileptic syndromes but has not been reported in Dravet syndrome (DS), a catastrophic childhood epilepsy associated with mutations in a voltage-activated sodium channel, Nav1.1 (SCN1A). To address this, we developed novel methodology to assess real-time changes in bioenergetics in zebrafish larvae between 4 and 6 d postfertilization (dpf). Baseline and 4-aminopyridine (4-AP) stimulated glycolytic flux and mitochondrial respiration were simultaneously assessed using a Seahorse Biosciences extracellular flux analyzer. Scn1Lab mutant zebrafish showed a decrease in baseline glycolytic rate and oxygen consumption rate (OCR) compared to controls. A ketogenic diet formulation rescued mutant zebrafish metabolism to control levels. Increasing neuronal excitability with 4-AP resulted in an immediate increase in glycolytic rates in wild-type zebrafish, whereas mitochondrial OCR increased slightly and quickly recovered to baseline values. In contrast, scn1Lab mutant zebrafish showed a significantly slower and exaggerated increase of both glycolytic rates and OCR after 4-AP. The underlying mechanism of decreased baseline OCR in scn1Lab mutants was not because of altered mitochondrial DNA content or dysfunction of enzymes in the electron transport chain or tricarboxylic acid cycle. Examination of glucose metabolism using a PCR array identified five glycolytic genes that were downregulated in scn1Lab mutant zebrafish. Our findings in scn1Lab mutant zebrafish suggest that glucose and mitochondrial hypometabolism contribute to the pathophysiology of DS.


Subject(s)
Epilepsies, Myoclonic/physiopathology , Glycolysis/genetics , Mitochondria/metabolism , Oxygen Consumption/genetics , 4-Aminopyridine/pharmacology , Animals , Animals, Genetically Modified , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Diet, Ketogenic/methods , Disease Models, Animal , Epilepsies, Myoclonic/diet therapy , Epilepsies, Myoclonic/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycolysis/drug effects , Histocompatibility Antigens/metabolism , Larva , Mitochondria/drug effects , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Oxygen Consumption/drug effects , Potassium Channel Blockers/pharmacology , Statistics, Nonparametric , Zebrafish
7.
Neurobiol Dis ; 82: 289-297, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26184893

ABSTRACT

Cognitive dysfunction is an important comorbidity of temporal lobe epilepsy (TLE). However, no targeted therapies are available and the mechanisms underlying cognitive impairment, specifically deficits in learning and memory associated with TLE remain unknown. Oxidative stress is known to occur in the pathogenesis of TLE but its functional role remains to be determined. Here, we demonstrate that oxidative stress and resultant processes contribute to cognitive decline associated with epileptogenesis. Using a synthetic catalytic antioxidant, we show that pharmacological removal of reactive oxygen species (ROS) prevents 1) oxidative stress, 2) deficits in mitochondrial oxygen consumption rates, 3) hippocampal neuronal loss and 4) cognitive dysfunction without altering the intensity of the initial status epilepticus (SE) or epilepsy development in a rat model of SE-induced TLE. Moreover, the effects of the catalytic antioxidant on cognition persisted beyond the treatment period suggestive of disease-modification. The data implicate oxidative stress as a novel mechanism by which cognitive dysfunction can arise during epileptogenesis and suggest a potential disease-modifying therapeutic approach to target it.


Subject(s)
Cognition Disorders/metabolism , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/psychology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Disease Models, Animal , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nootropic Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pilocarpine , Random Allocation , Rats, Sprague-Dawley , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Status Epilepticus/pathology , Status Epilepticus/psychology
8.
Neurobiol Dis ; 75: 151-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25600213

ABSTRACT

Metabolic alterations have been implicated in the etiology of temporal lobe epilepsy (TLE), but whether or not they have a functional impact on cellular energy producing pathways (glycolysis and/or oxidative phosphorylation) is unknown. The goal of this study was to determine if alterations in cellular bioenergetics occur using real-time analysis of mitochondrial oxygen consumption and glycolytic rates in an animal model of TLE. We hypothesized that increased steady-state levels of reactive oxygen species (ROS) initiated by epileptogenic injury result in impaired mitochondrial respiration. We established methodology for assessment of bioenergetic parameters in isolated synaptosomes from the hippocampus of Sprague-Dawley rats at various times in the kainate (KA) model of TLE. Deficits in indices of mitochondrial respiration were observed at time points corresponding with the acute and chronic phases of epileptogenesis. We asked if mitochondrial bioenergetic dysfunction occurred as a result of increased mitochondrial ROS and if it could be attenuated in the KA model by pharmacologically scavenging ROS. Increased steady-state ROS in mice with forebrain-specific conditional deletion of manganese superoxide dismutase (Sod2(fl/fl)NEX(Cre/Cre)) in mice resulted in profound deficits in mitochondrial oxygen consumption. Pharmacological scavenging of ROS with a catalytic antioxidant restored mitochondrial respiration deficits in the KA model of TLE. Together, these results demonstrate that mitochondrial respiration deficits occur in experimental TLE and ROS mechanistically contribute to these deficits. Furthermore, this study provides novel methodology for assessing cellular metabolism during the entire time course of disease development.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Glycolysis/physiology , Hippocampus/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Acute Disease , Animals , Antioxidants/pharmacology , Cell Respiration/drug effects , Cell Respiration/physiology , Chronic Disease , Disease Models, Animal , Female , Hippocampus/drug effects , Kainic Acid , Male , Mice, Knockout , Mitochondria/drug effects , Rats, Sprague-Dawley , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics
9.
Free Radic Biol Med ; 62: 121-131, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23411150

ABSTRACT

A role for mitochondria and oxidative stress is emerging in acquired epilepsies such as temporal lobe epilepsy (TLE). TLE is characterized by chronic unprovoked seizures arising from an inciting insult with a variable seizure-free "latent period." The mechanism by which inciting injury induces chronic epilepsy, known as epileptogenesis, involves multiple cellular, molecular, and physiological changes resulting in altered hyperexcitable circuitry. Whether mitochondrial and redox mechanisms contribute to epileptogenesis remains to be fully clarified. Mitochondrial impairment is revealed in studies from human imaging and tissue analysis from TLE patients. The collective data from animal models suggest that steady-state mitochondrial reactive oxygen species and resultant oxidative damage to cellular macromolecules occur during different phases of epileptogenesis. This review discusses evidence for the role of mitochondria and redox changes occurring in human and experimental TLE. Potential mechanisms by which mitochondrial energetic and redox mechanisms contribute to increased neuronal excitability and therapeutic approaches to target TLE are delineated.


Subject(s)
Energy Metabolism , Epilepsy, Temporal Lobe/metabolism , Mitochondria/metabolism , Oxidative Stress , Animals , Disease Progression , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/physiopathology , Humans , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism
10.
Neurobiol Dis ; 45(3): 1068-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22200564

ABSTRACT

Epileptic seizures are a common feature associated with inherited mitochondrial diseases. This study investigated the role of mitochondrial oxidative stress in epilepsy resulting from mitochondrial dysfunction using cross-bred mutant mice lacking mitochondrial manganese superoxide dismutase (MnSOD or SOD2) and a lipophilic metalloporphyrin catalytic antioxidant. Video-EEG monitoring revealed that in the second to third week of postnatal life (P14-P21) B6D2F2 Sod2(-/-) mice exhibited frequent spontaneous motor seizures providing evidence that oxidative stress-induced mitochondrial dysfunction may contribute to epileptic seizures. To confirm the role of mitochondrial oxidative stress in epilepsy a newly developed lipophilic metalloporphyrin, AEOL 11207, with high potency for catalytic removal of endogenously generated reactive oxygen species was utilized. AEOL 11207-treated Sod2(-/-) mice showed a significant decrease in both the frequency and duration of spontaneous seizures but no effect on seizure severity. A significant increase in the average lifespan of AEOL 11207-treated Sod2(-/-) mice compared to vehicle-treated Sod2(-/-) mice was also observed. Indices of mitochondrial oxidative stress and damage (aconitase inactivation, 3-nitrotyrosine formation, and depletion of reduced coenzyme A) and ATP levels affecting neuronal excitability were significantly attenuated in the brains of AEOL 11207-treated Sod2(-/-) mice compared to vehicle-treated Sod2(-/-) mice. The occurrence of epileptic seizures in Sod2(-/-) mice and the ability of catalytic antioxidant therapy to attenuate seizure activity, mitochondrial dysfunction, and ATP levels suggest that ongoing mitochondrial oxidative stress can contribute to epilepsy associated with mitochondrial dysfunction and disease.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy , Metalloporphyrins/therapeutic use , Mitochondria/drug effects , Oxidative Stress/drug effects , Superoxide Dismutase/deficiency , Aconitate Hydratase/metabolism , Adenine Nucleotides/metabolism , Analysis of Variance , Animals , Animals, Newborn , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Chromatography, High Pressure Liquid , Coenzyme A/metabolism , Disease Models, Animal , Electroencephalography , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy/pathology , Fumarate Hydratase/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD/metabolism , Oxidative Stress/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
Synapse ; 63(2): 147-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19021208

ABSTRACT

The abuse of methamphetamine (METH) is a serious public health problem because METH can cause persistent dopaminergic deficits in the brains of both animal models and humans. Surprisingly, adolescent postnatal day (PND)40 rats are resistant to these METH-induced deficits, whereas young adult PND90 rats are not. Studies described in this report used rotating disk electrode voltammetry and western blotting techniques to investigate whether there are age-dependent differences in monoamine transporter function in PND38-42 and PND88-92 rats that could contribute to this phenomenon. The initial velocities of dopamine (DA) transport into, METH-induced DA efflux from, and DA transporter (DAT) immunoreactivity in striatal suspensions are greater in PND38-42 rats than in PND88-92 rats. DA transport velocities into vesicles that cofractionate with synaptosomal membranes after osmotic lysis are also greater in PND38-42 rats. However, there is no difference in vesicular monoamine transporter-2 (VMAT-2) immunoreactivity between the two age groups in this fraction. This suggests that younger rats have a greater capacity to sequester cytoplasmic DA into membrane-associated vesicles due to kinetically upregulated VMAT-2 and also have increased levels of functionally active DAT. In the presence of METH, these may provide additional routes of cellular efflux for DA that is released from vesicles into the cytoplasm and thereby prevent cytoplasmic DA concentrations in younger rats from rising to neurotoxic levels after drug administration. These findings provide novel insight into the age-dependent physiological regulation of neuronal DA sequestration and may advance the treatment of disorders involving abnormal DA disposition including substance abuse and Parkinson's disease.


Subject(s)
Central Nervous System Stimulants/toxicity , Dopamine Plasma Membrane Transport Proteins/drug effects , Methamphetamine/toxicity , Neurons/drug effects , Vesicular Monoamine Transport Proteins/drug effects , Age Factors , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Vesicular Monoamine Transport Proteins/metabolism
13.
J Pharmacol Exp Ther ; 327(1): 161-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18591219

ABSTRACT

Methylphenidate (MPD) administration alters the subcellular distribution of vesicular monoamine transporter-2 (VMAT-2)-containing vesicles in rat striatum. This report reveals previously undescribed pharmacological features of MPD by elucidating its receptor-mediated effects on VMAT-2-containing vesicles that cofractionate with synaptosomal membranes after osmotic lysis (referred to herein as membrane-associated vesicles) and on striatal dopamine (DA) release. MPD administration increased DA transport into, and decreased the VMAT-2 immunoreactivity of, the membrane-associated vesicle subcellular fraction. These effects were mimicked by the D2 receptor agonist quinpirole and blocked by the D2 receptor antagonist eticlopride. Both MPD and quinpirole increased vesicular DA content. However, MPD increased, whereas quinpirole decreased, K(+)-stimulated DA release from striatal suspensions. Like MPD, the muscarinic receptor agonist, oxotremorine, increased K(+)-stimulated DA release. Both eticlopride and the muscarinic receptor antagonist scopolamine blocked MPD-induced increases in K(+)-stimulated DA release, whereas the N-methyl-d-aspartate receptor antagonist (-)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801) was without effect. This suggests that D2 receptors mediate both the MPD-induced redistribution of vesicles away from synaptosomal membranes and the MPD-induced up-regulation of vesicles remaining at the membrane. This results in a redistribution of DA within the striatum from the cytoplasm into vesicles, leading to increased DA release. However, D2 receptor activation alone is not sufficient to mediate the MPD-induced increases in striatal DA release because muscarinic receptor activation is also required. These novel findings provide insight into the mechanism of action of MPD, regulation of DA sequestration/release, and treatment of disorders affecting DA disposition, including attention-deficit hyperactivity disorder, substance abuse, and Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Methylphenidate/pharmacology , Receptors, Dopamine D2/physiology , Receptors, Muscarinic/physiology , Animals , Male , Oxotremorine/pharmacology , Potassium/pharmacology , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Scopolamine/pharmacology , Vesicular Monoamine Transport Proteins/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...