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










Publication year range
1.
J Neurosurg Case Lessons ; 7(13)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38531084

ABSTRACT

BACKGROUND: During brain tumor resection, neurophysiological mapping and monitoring help surgeons locate, characterize, and functionally assess eloquent brain areas in real time. The selection of mapping and monitoring targets has implications for successful surgery. Here, the authors compare direct cortical stimulation (DCS) as suggested by median nerve (MN) with posterior tibial nerve (PTN) cortical sensory mapping (SM) during mesial lesion resection. OBSERVATIONS: Recordings from a 6-contact cortical strip served to generate an MN and a PTN sensory map, which indicated the strip was anterior to the central sulcus. Responses exhibited an amplitude gradient with no phase reversal (PR). DCS, elicited through a stimulus probe or contact(s) of the strip, yielded larger responses from the corresponding sensory mapped limb; that is, PTN SM resulted in larger lower limb muscle responses than those suggested by MN SM. LESSONS: SM of the MN and PTN is effective for localizing eloquent cortical areas wherein the PTN is favored in surgery for mesial cortical tumors. The recorded amplitude of the cortical somatosensory evoked potential is a valuable criterion for defining the optimal location for DCS, despite an absent PR. The pathway at risk dictates the specifics of SM, which subsequently defines the optimal location for DCS.

2.
J Neurosurg Spine ; : 1-7, 2022 May 13.
Article in English | MEDLINE | ID: mdl-35561698

ABSTRACT

OBJECTIVE: The human myotome is fundamental to the diagnosis and treatment of neurological disorders. However, this map was largely constructed decades ago, and its breadth, variability, and reliability remain poorly described, limiting its practical use. METHODS: The authors used a novel method to reconstruct the myotome map in patients (n = 42) undergoing placement of dorsal root ganglion electrodes for the treatment of chronic pain. They electrically stimulated nerve roots (n = 79) in the intervertebral foramina at T12-S1 and measured triggered electromyography responses. RESULTS: L4 and L5 stimulation resulted in quadriceps muscle (62% and 33% of stimulations, respectively) and tibialis anterior (TA) muscle (25% and 67%, respectively) activation, while S1 stimulation resulted in gastrocnemius muscle activation (46%). However, L5 and S1 both resulted in abductor hallucis (AH) muscle activation (17% and 31%), L5 stimulation resulted in gastrocnemius muscle stimulation (42%), and S1 stimulation in TA muscle activation (38%). The authors also mapped the breadth of the myotome in individual patients, finding coactivation of adductor and quadriceps, quadriceps and TA, and TA and gastrocnemius muscles under L3, L4, and both L5 and S1 stimulation, respectively. While the AH muscle was commonly activated by S1 stimulation, this rarely occurred together with TA or gastrocnemius muscle activation. Other less common coactivations were also observed throughout T12-S1 stimulation. CONCLUSIONS: The muscular innervation of the lumbosacral nerve roots varies significantly from the classic myotome map and between patients. Furthermore, in individual patients, each nerve root may innervate a broader range of muscles than is commonly assumed. This finding is important to prevent misdiagnosis of radicular pathologies.

3.
Acta Neurochir (Wien) ; 164(3): 903-911, 2022 03.
Article in English | MEDLINE | ID: mdl-34820740

ABSTRACT

OBJECTIVE: Condylar screw fixation is a rescue technique and an alternative to the conventional configuration of occipitocervical fusion. Condylar screws are utilized when previous surgical bone removal along the supraocciput has occurred which makes anchoring of a traditional barplate technically difficult or impossible. However, the challenging dissection of C0-1 necessary for condylar screw fixation and the concerns about possible complications have, thus far, prevented the acquisition of large surgical series utilizing occipital condylar screws. In the largest case series to date, this paper aims to evaluate the safety profile and complications of condylar screw fixation for occipitocervical fusion. METHODS: A retrospective safety and complication-based analysis of occipitocervical fusion via condylar screws fixation was performed. RESULTS: A total of 250 patients underwent occipitocervical fusions using 500 condylar screws between September 2012 and September 2018. No condylar screw pullouts, or vertebral artery impingements were observed in this series. The sacrifice of condylar veins during the dissection at C0-1 did not cause any venous stroke. Hypotrophic condyles were found in 36.4% (91 of the 250) cases and did not prevent the insertion of condylar screws. Two transient hypoglossal deficits occurred at the beginning of this surgical series and were followed by recovery a few months later. Corrective strategies were effective in preventing further hypoglossal injuries. CONCLUSIONS: This surgical series suggests that the use of condylar screws fixation is a relatively safe and reliable option for OC fusion in both adult and pediatric patients. Methodical dissection of anatomical landmarks, intraoperative imaging, and neurophysiologic monitoring allowed the safe execution of the largest series of condylar screws reported to date. Separate contributions will follow in the future to provide details about the long-term clinical outcome of this series.


Subject(s)
Spinal Fusion , Surgeons , Adult , Bone Screws , Cervical Vertebrae/surgery , Child , Humans , Occipital Bone/diagnostic imaging , Occipital Bone/surgery , Retrospective Studies , Spinal Fusion/adverse effects , Spinal Fusion/methods
4.
J Neurosurg Spine ; : 1-10, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34560655

ABSTRACT

OBJECTIVE: This is a retrospective study of a series of occipitocervical fusion procedures with condylar screw fixation in which the authors investigated the utility of electromyography (EMG, free-running and triggered) as a reliable tool in assessing the positioning of condylar screws. This series consisted of 197 patients between 15 and 60 years of age who presented with craniocervical instability, and who were treated between October 2014 and December 2017. METHODS: Intraoperative free-running EMG was observed at the placement of condylar screws, as well as at realigning of the spine. After placement the condylar screws were stimulated electrically, and the thresholds were recorded. CT scans were obtained intraoperatively soon after screw stimulation, and the results were analyzed by the surgeon in real time. Free-running EMG results and triggered EMG thresholds were tabulated, and the minimum acceptable threshold was established. RESULTS: Intraoperative free-running EMG and triggered EMG were able to correlate alerts with condylar screw placement accurately. A triggered EMG threshold of 2.7 mA was found to be a minimum acceptable threshold. A combination criterion of free-running EMG and triggered EMG alerts was found to enable accurate assessment of condylar screw positioning and placement. CONCLUSIONS: Intraoperative free-running EMG and triggered EMG were both found to be invaluable utilities in assessing the placement and positioning of condylar screws. Stimulation thresholds below 2.7 mA correlated with a superior or anterior condylar breach. Thresholds in the 2.7-mA to 9.0-mA range were generally acceptable but warranted additional inspection by the surgeon. Threshold values above 9.0 mA corresponded with solid condylar screw placement.

5.
J Neurol Neurosurg Psychiatry ; 83(2): 182-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21949105

ABSTRACT

BACKGROUND: Mutations of the THAP1 gene were recently shown to underlie DYT6 torsion dystonia. Little is known about the response of this dystonia subtype to deep brain stimulation (DBS) at the internal globus pallidus (GPi). METHODS: Retrospective analysis of the medical records of three DYT6 patients who underwent pallidal DBS by one surgical team. The Burke-Fahn-Marsden Dystonia Rating scale served as the primary outcome measure. Comparison is made to 23 patients with DYT1 dystonia also treated with GPi-DBS by the same team. RESULTS: In contrast with the DYT1 patients who exhibited a robust and sustained clinical response to DBS, the DYT6 patients exhibited more modest gains during the first 2 years of therapy, and some symptom regression between years 2 and 3 despite adjustments to the stimulation parameters and repositioning of one stimulating lead. Microelectrode recordings made during the DBS procedures demonstrated no differences in the firing patterns of GPi neurons from DYT1 and DYT6 patients. DISCUSSION: Discovery of the genetic mutations responsible for the DYT6 phenotype allows for screening and analysis of a new homogeneous group of dystonia patients. DYT6 patients appear to respond less robustly to GPi-DBS than their DYT1 counterparts, most likely reflecting differences in the underlying pathophysiology of these distinct genetic disorders. CONCLUSIONS: While early results of pallidal DBS for DYT6 dystonia are encouraging, further research and additional subjects are needed both to optimise stimulation parameters for this population and to elucidate more accurately their response to surgical treatment.


Subject(s)
Deep Brain Stimulation/methods , Dystonia Musculorum Deformans/therapy , Globus Pallidus/physiology , Adolescent , Adult , Age of Onset , Anti-Dyskinesia Agents/administration & dosage , Anti-Dyskinesia Agents/therapeutic use , Apoptosis Regulatory Proteins/genetics , DNA/genetics , DNA-Binding Proteins/genetics , Data Interpretation, Statistical , Disability Evaluation , Dystonia Musculorum Deformans/drug therapy , Dystonia Musculorum Deformans/genetics , Electrodes, Implanted , Female , Humans , Male , Microelectrodes , Mutation/genetics , Neurosurgical Procedures , Nuclear Proteins/genetics , Retrospective Studies , Treatment Outcome , Young Adult
6.
J Neurosci ; 30(5): 1788-97, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-20130188

ABSTRACT

PARK8/LRRK2 (leucine-rich repeat kinase 2) was recently identified as a causative gene for autosomal dominant Parkinson's disease (PD), with LRRK2 mutation G2019S linked to the most frequent familial form of PD. Emerging in vitro evidence indicates that aberrant enzymatic activity of LRRK2 protein carrying this mutation can cause neurotoxicity. However, the physiological and pathophysiological functions of LRRK2 in vivo remain elusive. Here we characterize two bacterial artificial chromosome (BAC) transgenic mouse strains overexpressing LRRK2 wild-type (Wt) or mutant G2019S. Transgenic LRRK2-Wt mice had elevated striatal dopamine (DA) release with unaltered DA uptake or tissue content. Consistent with this result, LRRK2-Wt mice were hyperactive and showed enhanced performance in motor function tests. These results suggest a role for LRRK2 in striatal DA transmission and the consequent motor function. In contrast, LRRK2-G2019S mice showed an age-dependent decrease in striatal DA content, as well as decreased striatal DA release and uptake. Despite increased brain kinase activity, LRRK2-G2019S overexpression was not associated with loss of DAergic neurons in substantia nigra or degeneration of nigrostriatal terminals at 12 months. Our results thus reveal a pivotal role for LRRK2 in regulating striatal DA transmission and consequent control of motor function. The PD-associated mutation G2019S may exert pathogenic effects by impairing these functions of LRRK2. Our LRRK2 BAC transgenic mice, therefore, could provide a useful model for understanding early PD pathological events.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Motor Skills , Mutation , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Protein Serine-Threonine Kinases/genetics , Aging/metabolism , Animals , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Transgenic , Motor Activity/genetics
7.
J Neurosci ; 29(28): 9002-10, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19605638

ABSTRACT

Hydrogen peroxide (H(2)O(2)) is emerging as a ubiquitous small-molecule messenger in biology, particularly in the brain, but underlying mechanisms of peroxide signaling remain an open frontier for study. For example, dynamic dopamine transmission in dorsolateral striatum is regulated on a subsecond timescale by glutamate via H(2)O(2) signaling, which activates ATP-sensitive potassium (K(ATP)) channels to inhibit dopamine release. However, the origin of this modulatory H(2)O(2) has been elusive. Here we addressed three possible sources of H(2)O(2) produced for rapid neuronal signaling in striatum: mitochondrial respiration, monoamine oxidase (MAO), and NADPH oxidase (Nox). Evoked dopamine release in guinea-pig striatal slices was monitored with carbon-fiber microelectrodes and fast-scan cyclic voltammetry. Using direct fluorescence imaging of H(2)O(2) and tissue analysis of ATP, we found that coapplication of rotenone (50 nM), a mitochondrial complex I inhibitor, and succinate (5 mM), a complex II substrate, limited H(2)O(2) production, but maintained tissue ATP content. Strikingly, coapplication of rotenone and succinate also prevented glutamate-dependent regulation of dopamine release, implicating mitochondrial H(2)O(2) in release modulation. In contrast, inhibitors of MAO or Nox had no effect on dopamine release, suggesting a limited role for these metabolic enzymes in rapid H(2)O(2) production in the striatum. These data provide the first demonstration that respiring mitochondria are the primary source of H(2)O(2) generation for dynamic neuronal signaling.


Subject(s)
Brain/cytology , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Neurons/ultrastructure , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Dopamine/analysis , Drug Interactions , Electric Stimulation/methods , Electrochemistry/methods , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/pharmacology , Glyburide/pharmacology , Guinea Pigs , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Mitochondria/drug effects , Nerve Tissue Proteins/metabolism , Rotenone/pharmacology , Signal Transduction/drug effects , Sodium Channel Blockers/pharmacology , Succinic Acid/pharmacology , Tetrodotoxin/pharmacology
8.
J Neurosci ; 29(20): 6568-79, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19458227

ABSTRACT

Somatodendritic dopamine (DA) release in the substantia nigra pars compacta (SNc) shows a limited dependence on extracellular calcium concentration ([Ca(2+)](o)), suggesting the involvement of intracellular Ca(2+) stores. Here, using immunocytochemistry we demonstrate the presence of the sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2 (SERCA2) that sequesters cytosolic Ca(2+) into the endoplasmic reticulum (ER), as well as inositol 1,4,5-triphosphate receptors (IP(3)Rs) and ryanodine receptors (RyRs) in DAergic neurons. Notably, RyRs were clustered at the plasma membrane, poised for activation by Ca(2+) entry. Using fast-scan cyclic voltammetry to monitor evoked extracellular DA concentration ([DA](o)) in midbrain slices, we found that SERCA inhibition by cyclopiazonic acid (CPA) decreased evoked [DA](o) in the SNc, indicating a functional role for ER Ca(2+) stores in somatodendritic DA release. Implicating IP(3)R-dependent stores, an IP(3)R antagonist, 2-APB, also decreased evoked [DA](o). Moreover, DHPG, an agonist of group I metabotropic glutamate receptors (mGluR1s, which couple to IP(3) production), increased somatodendritic DA release, whereas CPCCOEt, an mGluR1 antagonist, suppressed it. Release suppression by mGluR1 blockade was prevented by 2-APB or CPA, indicating facilitation of DA release by endogenous glutamate acting via mGluR1s and IP(3)R-gated Ca(2+) stores. Similarly, activation of RyRs by caffeine increased [Ca(2+)](i) and elevated evoked [DA](o). The increase in DA release was prevented by a RyR blocker, dantrolene, and by CPA. Importantly, the efficacy of dantrolene was enhanced in low [Ca(2+)](o), suggesting a mechanism for maintenance of somatodendritic DA release with limited Ca(2+) entry. Thus, both mGluR1-linked IP(3)R- and RyR-dependent ER Ca(2+) stores facilitate somatodendritic DA release in the SNc.


Subject(s)
Axons/metabolism , Calcium/metabolism , Dendrites/metabolism , Dopamine/metabolism , Intracellular Fluid/metabolism , Neurons/cytology , Animals , Boron Compounds/pharmacology , Cadmium/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Chelating Agents/pharmacology , Chromones/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation , Electrochemical Techniques/methods , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Guinea Pigs , In Vitro Techniques , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Substantia Nigra/cytology , Tyrosine 3-Monooxygenase/metabolism
9.
J Neurophysiol ; 100(3): 1590-601, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632893

ABSTRACT

Dopamine-glutamate interactions in the striatum are critical for normal basal ganglia-mediated control of movement. Although regulation of glutamatergic transmission by dopamine is increasingly well understood, regulation of dopaminergic transmission by glutamate remains uncertain given the apparent absence of ionotropic glutamate receptors on dopaminergic axons in dorsal striatum. Indirect evidence suggests glutamatergic regulation of striatal dopamine release is mediated by a diffusible messenger, hydrogen peroxide (H2O2), generated downstream from glutamatergic AMPA receptors (AMPARs). The mechanism of H2O2-dependent inhibition of dopamine release involves activation of ATP-sensitive K+ (KATP) channels. However, the source of modulatory H2O2 is unknown. Here, we used whole cell recording, fluorescence imaging of H2O2, and voltammetric detection of evoked dopamine release in guinea pig striatal slices to examine contributions from medium spiny neurons (MSNs), the principal neurons of striatum, and dopamine axons to AMPAR-dependent H2O2 generation. Imaging studies of H2O2 generation in MSNs provide the first demonstration of AMPAR-dependent H2O2 generation in neurons in the complex brain-cell microenvironment of brain slices. Stimulation-induced increases in H2O2 in MSNs were prevented by GYKI-52466, an AMPAR antagonist, or catalase, an H2O2 metabolizing enzyme, but amplified by mercaptosuccinate (MCS), a glutathione peroxidase inhibitor. By contrast, dopamine release evoked by selective stimulation of dopamine axons was unaffected by GYKI-52466 or MCS, arguing against dopamine axons as a significant source of modulatory H2O2. Together, these findings suggest that glutamatergic regulation of dopamine release via AMPARs is mediated through retrograde signaling by diffusible H2O2 generated in striatal cells, including medium spiny neurons, rather than in dopamine axons.


Subject(s)
Corpus Striatum/cytology , Dopamine/metabolism , Hydrogen Peroxide/metabolism , Neurons/drug effects , Receptors, AMPA/physiology , Analysis of Variance , Animals , Axons/metabolism , Benzodiazepines/pharmacology , Catalase/metabolism , Chromatography, High Pressure Liquid/methods , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Neural Pathways/drug effects , Neurons/cytology , Patch-Clamp Techniques/methods , Thiomalates/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Valine/analogs & derivatives , Valine/pharmacology
10.
J Neurosci ; 27(5): 1167-75, 2007 Jan 31.
Article in English | MEDLINE | ID: mdl-17267572

ABSTRACT

Synchronous neural activity causes rapid changes of extracellular pH (pH(e)) in the nervous system. In the CA1 region of the hippocampus, stimulation of the Schaffer collaterals elicits an alkaline pH(e) transient in stratum radiatum that is limited by extracellular carbonic anhydrase (ECA). When interstitial buffering is diminished by inhibition of ECA, the alkalosis is enhanced and NMDA receptor (NMDAR)-mediated postsynaptic currents can be augmented. Accordingly, the dendritic influx of Ca2+ elicited by synaptic excitation may be expected to increase if ECA activity were blocked. We tested this hypothesis in the CA1 stratum radiatum of hippocampal slices from juvenile rats, using extracellular, concentric pH- and Ca2+-selective microelectrodes with response times of a few milliseconds, as well as Fluo-5F imaging of intracellular Ca2+ transients. Brief stimulation of the Schaffer collaterals elicited an alkaline pH(e) transient, a transient decrease in free extracellular Ca2+ concentration ([Ca2+]e), and a corresponding transient rise in free intracellular Ca2+ concentration ([Ca2+]i). Inhibition of ECA with benzolamide caused a marked amplification and prolonged recovery of the pH(e) and [Ca2+]e responses, as well as the dendritic [Ca2+]i transients. The increase in amplitude caused by benzolamide did not occur in the presence of the NMDAR antagonist APV, but the decay of the responses was still prolonged. These results indicate that ECA can shape dendritic Ca2+ dynamics governed by NMDARs by virtue of its regulation of concomitant activity-dependent pH(e) shifts. The data also suggest that Ca2+ transients are influenced by additional mechanisms sensitive to shifts in pH(e).


Subject(s)
Calcium/metabolism , Carbonic Anhydrases/physiology , Excitatory Postsynaptic Potentials/physiology , Extracellular Fluid/enzymology , Hippocampus/enzymology , Pyramidal Cells/enzymology , Animals , Extracellular Fluid/metabolism , Female , Hippocampus/metabolism , Male , Pyramidal Cells/metabolism , Rats
11.
Antioxid Redox Signal ; 9(2): 219-31, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17115944

ABSTRACT

The role of reactive oxygen species (ROS) as signaling agents is increasingly appreciated. Studies of ROS functions in the central nervous system, however, are only in their infancy. Using fast-scan cyclic voltammetry and fluorescence imaging in brain slices, the authors discovered that hydrogen peroxide (H2O2) is an endogenous regulator of dopamine release in the dorsal striatum. Given the key role of dopamine in motor, reward, and cognitive pathways, regulation by H2O2 has implications for normal dopamine function, as well as for dysfunction of dopamine transmission. In this review, data are summarized to show that H2O2 is a diffusible messenger in the striatum, generated downstream from glutamate receptor activation, and an intracellular signal in dopamine neurons of the substantia nigra, generated during normal pacemaker activity. The mechanism by which H2O2 inhibits dopamine release and dopamine cell activity is activation of ATP-sensitive K+ (KATP) channels. Characteristics of the neuronal and glial antioxidant networks required to permit H2O2 signaling, yet prevent oxidative damage, are also considered. Lastly, estimates of physiological H2O2 levels are discussed, and strengths and limitations of currently available methods for H2O2 detection, including fluorescence imaging using dichlorofluorescein (DCF) and the next generation of fluorescent probes, are considered.


Subject(s)
Adenosine Triphosphate/chemistry , Corpus Striatum/metabolism , Dopamine/metabolism , Hydrogen Peroxide/pharmacology , Potassium/chemistry , Animals , Antioxidants/metabolism , Dopamine/chemistry , Fluoresceins/pharmacology , Humans , Microscopy, Fluorescence , Mitochondria/metabolism , Neuroglia/metabolism , Neurons/metabolism , Reactive Oxygen Species , Signal Transduction
12.
J Neurochem ; 96(3): 645-55, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16405515

ABSTRACT

The mechanism underlying somatodendritic release of dopamine (DA) appears to differ from that of axon-terminal release. Specifically, somatodendritic DA release in the substantia nigra pars compacta (SNc) persists in low extracellular Ca2+ concentrations that are insufficient to support axonal release in striatum, suggesting that limited Ca2+ entry is necessary to trigger somatodendritic release. Here, we compared the role of voltage-dependent Ca2+ channels in mediating DA release in striatum versus SNc using specific blockers of N-, P/Q-, T-, R- and L-type Ca2+ channels individually and in combination. Release of DA evoked by a single stimulus pulse in the dorsal striatum and SNc of guinea-pig brain slices was monitored in real time using carbon-fiber microelectrodes with fast-scan cyclic voltammetry. Single-pulse evoked DA release was shown to be independent of regulation by concurrently released glutamate or GABA acting at ionotropic receptors in both regions. Under these conditions, striatal DA release was completely prevented by an N-type channel blocker, omega-conotoxin GVIA (100 nm), and was decreased by 75% by the P/Q-type channel blocker omega-agatoxin IVA (200 nm). Blockade of T-type channels with Ni2+ (100 microm) or R-type channels with SNX-482 (100 nm) decreased axonal release in striatum by 25%, whereas inhibition of L-type channels with nifedipine (20 microm) had no effect. By contrast, none of these Ca2+-channel blockers altered the amplitude of somatodendritic DA release in the SNc. Even a cocktail of all blockers tested did not alter release-signal amplitude in the SNc, although the duration of the release response was curtailed. The limited involvement of voltage-dependent Ca2+ channels in somatodendritic DA release provides further evidence that minimal Ca2+ entry is required to trigger the release process, compared with that required for axon-terminal release.


Subject(s)
Axons/metabolism , Calcium Channels/physiology , Dendrites/metabolism , Dopamine/metabolism , Neurons/cytology , Animals , Axons/drug effects , Benzodiazepines/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Corpus Striatum/cytology , Dendrites/drug effects , Dendrites/radiation effects , Dose-Response Relationship, Radiation , Drug Combinations , Electric Stimulation/methods , Electrochemistry/methods , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Glutamic Acid/pharmacology , Guinea Pigs , In Vitro Techniques , Male , Neurons/drug effects , Neurons/metabolism , Neurons/radiation effects , Substantia Nigra/cytology , gamma-Aminobutyric Acid/pharmacology
13.
J Neurosci ; 25(43): 10029-40, 2005 Oct 26.
Article in English | MEDLINE | ID: mdl-16251452

ABSTRACT

Mitochondrial dysfunction is a potential causal factor in Parkinson's disease. We show here that acute exposure to the mitochondrial complex I inhibitor rotenone (30-100 nM; 30 min) causes concentration-dependent suppression of single-pulse evoked dopamine (DA) release monitored in real time with carbon-fiber microelectrodes in guinea pig striatal slices, with no effect on DA content. Suppression of DA release was prevented by the sulfonylurea glibenclamide, implicating ATP-sensitive K+ (KATP) channels; however, tissue ATP was unaltered. Because KATP channels can be activated by hydrogen peroxide (H2O2), as well as by low ATP, we examined the involvement of rotenone-enhanced H2O2 generation. Confirming an essential role for H2O2, the inhibition of DA release by rotenone was prevented by catalase, a peroxide-scavenging enzyme. Striatal H2O2 generation during rotenone exposure was examined in individual medium spiny neurons using fluorescence imaging with dichlorofluorescein (DCF). An increase in intracellular H2O2 levels followed a similar time course to that of DA release suppression and was accompanied by cell membrane depolarization, decreased input resistance, and increased excitability. Extracellular catalase markedly attenuated the increase in DCF fluorescence and prevented rotenone-induced effects on membrane properties; membrane changes were also largely prevented by flufenamic acid, a blocker of transient receptor potential (TRP) channels. Thus, partial mitochondrial inhibition can cause functional DA denervation via H2O2 and KATP channels, without DA or ATP depletion. Furthermore, amplified H2O2 levels and TRP channel activation in striatal spiny neurons indicate potential sources of damage in these cells. Overall, these novel factors could contribute to parkinsonian motor deficits and neuronal degeneration caused by mitochondrial dysfunction.


Subject(s)
Corpus Striatum/cytology , Dopamine/metabolism , Hydrogen Peroxide/metabolism , Neurons/metabolism , Rotenone/pharmacology , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Catalase/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/radiation effects , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Fluoresceins/metabolism , Glyburide/pharmacology , Guinea Pigs , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Neurons/drug effects , Neurons/radiation effects , Patch-Clamp Techniques/methods , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Time Factors , Tolbutamide/pharmacology , Uncoupling Agents
14.
J Neurosci ; 25(17): 4222-31, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15858048

ABSTRACT

ATP-sensitive K+ (K(ATP)) channels link metabolic state to cell excitability. Here, we examined regulation of K(ATP) channels in substantia nigra dopamine neurons by hydrogen peroxide (H2O2), which is produced in all cells during aerobic metabolism. Blockade of K(ATP) channels by glibenclamide (100 nM) or depletion of intracellular H2O2 by including catalase, a peroxidase enzyme, in the patch pipette increased the spontaneous firing rate of all dopamine neurons tested in guinea pig midbrain slices. Using fluorescence imaging with dichlorofluorescein to visualize intracellular H2O2, we found that moderate increases in H2O2 during partial inhibition of glutathione (GSH) peroxidase by mercaptosuccinate (0.1-0.3 mM) had no effect on dopamine neuron firing rate. However, with greater GSH inhibition (1 mM mercaptosuccinate) or application of exogenous H2O2, 50% of recorded cells showed K(ATP) channel-dependent hyperpolarization. Responsive cells also hyperpolarized with diazoxide, a selective opener for K(ATP) channels containing sulfonylurea receptor SUR1 subunits, but not with cromakalim, a selective opener for SUR2-based channels, indicating that SUR1-based K(ATP) channels conveyed enhanced sensitivity to elevated H2O2. In contrast, when endogenous H2O2 levels were increased after inhibition of catalase, the predominant peroxidase in the substantia nigra, with 3-amino-1,2,4-triazole (1 mM), all dopamine neurons responded with glibenclamide-reversible hyperpolarization. Fluorescence imaging of H2O2 indicated that catalase inhibition rapidly amplified intracellular H2O2, whereas inhibition of GSH peroxidase, a predominantly glial enzyme, caused a slower, smaller increase, especially in nonresponsive cells. Thus, endogenous H2O2 modulates neuronal activity via K(ATP) channel opening, thereby enhancing the reciprocal relationship between metabolism and excitability.


Subject(s)
Adenosine Triphosphate/pharmacology , Dopamine/metabolism , Hydrogen Peroxide/metabolism , Mesencephalon/cytology , Neurons/metabolism , Potassium Channels/physiology , Analysis of Variance , Animals , Cromakalim/pharmacology , Diazoxide/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Glutathione Peroxidase , Glyburide/pharmacology , Guinea Pigs , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Isoquinolines/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Organoplatinum Compounds/pharmacology , Patch-Clamp Techniques/methods , Tetrodotoxin/pharmacology , Thiomalates/pharmacology , Time Factors , Vasodilator Agents/pharmacology
15.
Neuron Glia Biol ; 1(4): 365-76, 2004 Nov.
Article in English | MEDLINE | ID: mdl-18292802

ABSTRACT

Increasing evidence implicates reactive oxygen species, particularly hydrogen peroxide (H(2)O(2)), as intracellular and intercellular messengers in the brain. This raises the question of how the antioxidant network in the brain can be sufficiently permissive to allow messages to be conveyed yet, at the same time, provide adequate protection against oxidative damage. Here we present evidence that this is accomplished in part by differential antioxidant compartmentalization between glia and neurons. Based on the rationale that the glia-to-neuron ratio is higher in guinea-pig brain than in rat brain, we examined the neuroprotective role of the glial antioxidant network by comparing the consequences of elevated H(2)O(2) in guinea-pig and rat brain slices. The effects of exogenously applied H(2)O(2) on evoked population spikes in hippocampal slices and on edema formation in forebrain slices were assessed. In contrast to the epileptiform activity observed in rat hippocampal slices after H(2)O(2) exposure, no pathophysiology was seen in guinea-pig hippocampal slices. Similarly, elevated H(2)O(2) caused edema in rat brain slices, whereas this did not occur in guinea-pig brain tissue. The resistance of guinea-pig brain tissue to H(2)O(2) challenge was lost, however, when glutathione (GSH) synthesis was inhibited (by buthionine sulfoximine), GSH peroxidase activity was inhibited (by mercaptosuccinate), or catalase was inhibited (by 3-amino-1,2,4,-triazole). Strikingly, exogenously applied ascorbate, a predominantly neuronal antioxidant, was able to compensate for loss of any other single component of the antioxidant network. Together, these data imply significant roles for glial antioxidants and neuronal ascorbate in the prevention of pathophysiological consequences of the endogenous neuromodulator, H(2)O(2).

16.
Proc Natl Acad Sci U S A ; 100(20): 11729-34, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-13679582

ABSTRACT

In many cells, ATP-sensitive K+ channels (KATP channels) couple metabolic state to excitability. In pancreatic beta cells, for example, this coupling regulates insulin release. Although KATP channels are abundantly expressed in the brain, their physiological role and the factors that regulate them are poorly understood. One potential regulator is H2O2. We reported previously that dopamine (DA) release in the striatum is modulated by endogenous H2O2, generated downstream from glutamatergic alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-receptor activation. Here we investigated whether H2O2-sensitive KATP channels contribute to DA-release modulation by glutamate and gamma-aminobutyric acid (GABA). This question is important because DA-glutamate interactions underlie brain functions, including motor control and cognition. Synaptic DA release was evoked by using local electrical stimulation in slices of guinea pig striatum and monitored in real time with carbon-fiber microelectrodes and fast-scan cyclic voltammetry. The KATP-channel antagonist glibenclamide abolished the H2O2-dependent increase in DA release usually seen with AMPA-receptor blockade by GYKI-52466 [1-(4-aminophenyl)-4-methyl-7,8-methylenedioxy-5H-2,3-benzodiazepine hydrochloride] and the decrease in DA release seen with GABA-type-A-receptor blockade by picrotoxin. In contrast, 5-hydroxydecanoate, a mitochondrial KATP-channel blocker, was ineffective, as were sulpiride, a D2-receptor antagonist, and tertiapin, a G protein-coupled K+-channel inhibitor. Diazoxide, a sulfonylurea receptor 1 (SUR1)selective KATP-channel opener, prevented DA modulation by H2O2, glutamate, and GABA, whereas cromakalim, a SUR2-selective opener, did not. Thus, endogenous H2O2 activates SUR1-containing KATP channels in the plasma membrane to inhibit DA release. These data not only demonstrate that KATP channels can modulate CNS transmitter release in response to fast-synaptic transmission but also introduce H2O2 as a KATP-channel regulator.


Subject(s)
Adenosine Triphosphate/metabolism , Corpus Striatum/drug effects , Dopamine/pharmacology , Hydrogen Peroxide/pharmacology , Potassium Channels/metabolism , Animals , Corpus Striatum/metabolism , Dopamine/metabolism , Guinea Pigs , In Vitro Techniques , Male , Mitochondria/drug effects , Mitochondria/metabolism , Receptors, Dopamine D2/metabolism
17.
J Neurochem ; 85(6): 1402-11, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787060

ABSTRACT

Decreased cerebral blood flow, hence decreased oxygen and glucose, leads to ischemic brain injury via complex pathophysiological events, including excitotoxicity, mitochondrial dysfunction, increased intracellular Ca2+, and reactive oxygen species (ROS) generation. Each of these could also contribute to cerebral edema, which is the primary cause of patient mortality after stroke. In vitro brain slices are widely used to study ischemia. Here we introduce a slice model to investigate ischemia-induced edema. Significant water gain was induced in coronal slices of rat brain by 5 min of oxygen and glucose deprivation (OGD) at 35 degrees C, with progressive edema formation after return to normoxic, normoglycemic medium. Edema increased with increasing injury severity, determined by OGD duration (5-30 min). Underlying factors were assessed using glutamate-receptor antagonists (AP5/CNQX), blockade of mitochondrial permeability transition [cyclosporin A (CsA) versus FK506], inhibition of Na+/Ca2+ exchange (KB-R7943), and ROS scavengers (ascorbate, Trolox, dimethylthiourea, Tempol). All agents except KB-R7943 and FK506 significantly attenuated edema when applied after OGD; KB-R7943 was effective when applied before OGD. Significantly, complete prevention of ischemia-induced edema was achieved with a cocktail of AP5/CNQX, CsA and Tempo applied after OGD, which demonstrates the involvement of multiple, additive mechanisms. The efficacy of this cocktail further shows the potential value of combination therapies for the treatment of cerebral ischemia.


Subject(s)
2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Brain Edema/prevention & control , Cyclic N-Oxides/pharmacology , Cyclosporine/pharmacology , Hypoxia-Ischemia, Brain/drug therapy , Animals , Antioxidants/pharmacology , Brain Edema/etiology , Brain Edema/metabolism , Cell Hypoxia/drug effects , Drug Therapy, Combination , Excitatory Amino Acid Antagonists/pharmacology , Glucose/deficiency , Glucose/metabolism , Glutamic Acid/metabolism , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/physiopathology , In Vitro Techniques , Male , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Long-Evans , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Sodium-Calcium Exchanger/metabolism , Spin Labels , Water/metabolism
18.
J Neurosci ; 23(7): 2744-50, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12684460

ABSTRACT

How glutamate regulates dopamine (DA) release in striatum has been a controversial issue. Here, we resolve this by showing that glutamate, acting at AMPA receptors, inhibits DA release by a nonclassic mechanism mediated by hydrogen peroxide (H(2)O(2)). Moreover, we show that GABA(A)-receptor activation opposes this process, thereby enhancing DA release. The influence of glutamate and GABA on DA release was assessed in striatal slices using carbon-fiber microelectrodes and fast-scan cyclic voltammetry. Modulation by both transmitters was prevented by H(2)O(2)-metabolizing enzymes. In addition, the influence of GABA(A)-receptor activation was lost when AMPA receptors were blocked with GYKI-52466. Together, these data show that modulation of DA release by glutamate and GABA depends on H(2)O(2) generated downstream from AMPA receptors. This is the first evidence that endogenous glutamate can lead to the generation of reactive oxygen species under physiological conditions. We also show that inhibition of DA release by H(2)O(2) is mediated by sulfonylurea-sensitive K(+) channels: tolbutamide blocked DA modulation by glutamate and by GABA. The absence of ionotropic glutamate or GABA receptors on DA terminals indicates that modulatory H(2)O(2) is generated in non-DA cells. Thus, in addition to its known excitatory actions in striatum, glutamate mediates inhibition by generating H(2)O(2) that must diffuse from postsynaptic sites to inhibit presynaptic DA release via K(+)-channel opening. These findings have significant implications not only for normal striatal function but also for understanding disease states that involve DA and oxidative stress, including disorders as diverse as Parkinson's disease and schizophrenia.


Subject(s)
Benzodiazepines , Dopamine/metabolism , Glutamic Acid/physiology , Hydrogen Peroxide/metabolism , Neostriatum/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Cell Communication , Cells, Cultured , Diffusion , Electric Stimulation , GABA Antagonists/pharmacology , Guinea Pigs , Male , Neostriatum/drug effects , Potassium Channels/physiology , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/physiology , gamma-Aminobutyric Acid/physiology
19.
J Neurophysiol ; 87(6): 2896-903, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12037193

ABSTRACT

Endogenous reactive oxygen species (ROS) can act as modulators of neuronal activity, including synaptic transmission. Inherent in this process, however, is the potential for oxidative damage if the balance between ROS production and regulation becomes disrupted. Here we report that inhibition of synaptic transmission in rat hippocampal slices by H2O2 can be followed by electrical hyperexcitability when transmission returns during H2O2 washout. As in previous studies, H2O2 exposure (15 min) reversibly depressed the extracellular population spike (PS) evoked by Schaffer collateral stimulation. Recovery of PS amplitude, however, was typically accompanied by mild epileptiform activity. Inclusion of ascorbate (400 microM) during H2O2 washout prevented this pathophysiology. No protection was seen with isoascorbate, which is a poor substrate for the stereoselective ascorbate transporter and thus remains primarily extracellular. Epileptiform activity was also prevented by the N-methyl-D-aspartate (NMDA) receptor antagonist, DL-2-amino-5-phosphonopentanoic acid (AP5) during H2O2 washout. Once hyperexcitability was induced, however, AP5 did not reverse it. When present during H2O2 exposure, AP5 did not alter PS depression by H2O2 but did inhibit the recovery of PS amplitude seen during pulse-train stimulation (10 Hz, 5 s) in H2O2. Inhibition of glutamate uptake by l-trans-2,4-pyrrolidine dicarboxylate (PDC; 50 microM) during H2O2 washout markedly enhanced epileptiform activity; coapplication of ascorbate with PDC prevented this. These data indicate that H2O2 exposure can cause activation of normally silent NMDA receptors, possibly via inhibition of redox-sensitive glutamate uptake. When synaptic transmission returns during H2O2 washout, enhanced NMDA receptor activity leads to ROS generation and consequent oxidative damage. These data reveal a pathological cycle that could contribute to progressive degeneration in neurological disorders that involve oxidative stress, including cerebral ischemia.


Subject(s)
Hippocampus/physiopathology , Hydrogen Peroxide/toxicity , Oxidants/toxicity , Receptors, N-Methyl-D-Aspartate/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Epilepsy/pathology , Epilepsy/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Male , Organ Culture Techniques , Rats , Rats, Long-Evans , Reactive Oxygen Species/metabolism
20.
J Neurophysiol ; 87(2): 1155-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11826083

ABSTRACT

We showed previously that dopamine (DA) release in dorsal striatum is inhibited by endogenously generated hydrogen peroxide (H(2)O(2)). Here, we examined whether endogenous H(2)O(2) can also modulate somatodendritic DA release in the substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA), with companion measurements in DA terminal regions. Evoked DA release was monitored in brain slices using carbon-fiber microelectrodes with fast-scan cyclic voltammetry. Exogenous H(2)O(2) decreased DA release by 50-60% in SNc and VTA but only by 35% in nucleus accumbens. Whether endogenous H(2)O(2) also modulated somatodendritic release was examined using the glutathione peroxidase inhibitor, mercaptosuccinate (MCS), which should increase stimulation-evoked H(2)O(2) levels. In the presence of MCS, DA release was suppressed by 30-40% in SNc as well as in dorsal striatum and nucleus accumbens. In striking contrast, DA release in the VTA was unaffected by MCS. These data are consistent with stronger H(2)O(2) regulation or lower H(2)O(2) generation in VTA than in the other regions. Importantly, oxidative stress has been linked causally to Parkinson's disease, in which DA cells in SNc degenerate, but VTA cells are spared. The present data suggest that differences in oxidant regulation or generation between SNc and VTA could contribute to this.


Subject(s)
Dendrites/metabolism , Dopamine/metabolism , Hydrogen Peroxide/metabolism , Substantia Nigra/cytology , Ventral Tegmental Area/cytology , Animals , Glutathione Peroxidase/antagonists & inhibitors , Guinea Pigs , Male , Nucleus Accumbens/cytology , Nucleus Accumbens/metabolism , Organ Culture Techniques , Oxidative Stress/physiology , Substantia Nigra/metabolism , Synaptic Transmission/physiology , Thiomalates/pharmacology , Ventral Tegmental Area/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...