Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 23(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36362193

ABSTRACT

The inhibition of synaptic glutamate release to maintain glutamate homeostasis contributes to the alleviation of neuronal cell injury, and accumulating evidence suggests that natural products can repress glutamate levels and associated excitotoxicity. In this study, we investigated whether eupatilin, a constituent of Artemisia argyi, affected glutamate release in rat cortical nerve terminals (synaptosomes). Additionally, we evaluated the effect of eupatilin in an animal model of kainic acid (KA) excitotoxicity, particularly on the levels of glutamate and N-methyl-D-aspartate (NMDA) receptor subunits (GluN2A and GluN2B). We found that eupatilin decreased depolarization-evoked glutamate release from rat cortical synaptosomes and that this effect was accompanied by a reduction in cytosolic Ca2+ elevation, inhibition of P/Q-type Ca2+ channels, decreased synapsin I Ca2+-dependent phosphorylation and no detectable effect on the membrane potential. In a KA-induced glutamate excitotoxicity rat model, the administration of eupatilin before KA administration prevented neuronal cell degeneration, glutamate elevation, glutamate-generating enzyme glutaminase increase, excitatory amino acid transporter (EAAT) decrease, GluN2A protein decrease and GluN2B protein increase in the rat cortex. Taken together, the results suggest that eupatilin depresses glutamate exocytosis from cerebrocortical synaptosomes by decreasing P/Q-type Ca2+ channels and synapsin I phosphorylation and alleviates glutamate excitotoxicity caused by KA by preventing glutamatergic alterations in the rat cortex. Thus, this study suggests that eupatilin can be considered a potential therapeutic agent in the treatment of brain impairment associated with glutamate excitotoxicity.


Subject(s)
Artemisia , Neurotoxicity Syndromes , Rats , Animals , Glutamic Acid/metabolism , Synapsins/metabolism , Artemisia/metabolism , 4-Aminopyridine/pharmacology , Rats, Sprague-Dawley , Cerebral Cortex/metabolism , Calcium/metabolism , Synaptosomes/metabolism , Exocytosis , Kainic Acid/pharmacology , Neurotoxicity Syndromes/metabolism
2.
Cell Rep ; 35(3): 109007, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882305

ABSTRACT

Parkinson's disease is characterized by both hypokinetic and hyperkinetic symptoms. While increased subthalamic burst discharges have a direct causal relationship with the hypokinetic manifestations (e.g., rigidity and bradykinesia), the origin of the hyperkinetic symptoms (e.g., resting tremor and propulsive gait) has remained obscure. Neuronal burst discharges are presumed to be autonomous or less responsive to synaptic input, thereby interrupting the information flow. We, however, demonstrate that subthalamic burst discharges are dependent on cortical glutamatergic synaptic input, which is enhanced by A-type K+ channel inhibition. Excessive top-down-triggered subthalamic burst discharges then drive highly correlative activities bottom-up in the motor cortices and skeletal muscles. This leads to hyperkinetic behaviors such as tremors, which are effectively ameliorated by inhibition of cortico-subthalamic AMPAergic synaptic transmission. We conclude that subthalamic burst discharges play an imperative role in cortico-subcortical information relay, and they critically contribute to the pathogenesis of both hypokinetic and hyperkinetic parkinsonian symptoms.


Subject(s)
Globus Pallidus/physiopathology , Hyperkinesis/physiopathology , Motor Cortex/physiopathology , Parkinson Disease, Secondary/physiopathology , Subthalamic Nucleus/physiopathology , Tremor/physiopathology , 4-Aminopyridine/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Globus Pallidus/drug effects , Globus Pallidus/metabolism , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Humans , Hyperkinesis/metabolism , Male , Membrane Potentials/drug effects , Mice, Inbred C57BL , Motor Cortex/drug effects , Motor Cortex/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Optogenetics/methods , Parkinson Disease, Secondary/metabolism , Rats , Rats, Wistar , Subthalamic Nucleus/drug effects , Subthalamic Nucleus/metabolism , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Synaptic Transmission , Tremor/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
3.
Eur J Pharmacol ; 865: 172781, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31706856

ABSTRACT

Inhibiting glutamate release can reduce neuronal excitability and is recognized as a key mechanism of anti-epileptic drugs. In this study, by using isolated nerve terminal (synaptosome) and slice preparations, we investigated the effect of asiatic acid, a triterpene isolated from Centella asiatica with antiepileptic activity, on glutamate release in the hippocampus of rats. In hippocampal synaptosomes, application of asiatic acid resulted in a concentration-dependent inhibition of 4-aminopyridine-evoked glutamate release. This inhibitory action was dependent on extracellular calcium, blocked by inhibiting the vesicular transporter, but was unaffected by inhibiting the glutamate transporter. In addition, asiatic acid decreased the 4-aminopyridine-induced increase in the intraterminal calcium and failed to alter the synaptosomal potential. Furthermore, the asiatic acid-mediated release inhibition was significantly suppressed by the N- and P/Q-type calcium channel inhibitor ω-conotoxin MVIIC or protein kinase C inhibitor GF109203X. Western blotting data in synaptosomes also revealed that asiatic acid reduced 4-aminopyridine-induced phosphorylation of protein kinase C. In hippocampal slices, asiatic acid decreased the frequencies of spontaneous excitatory postsynaptic currents without changing their amplitudes and glutamate-activated currents in CA3 pyramidal neurons. We also observed that asiatic acid significantly suppressed 4-aminopyridine-induced burst firing. These data suggest that, in rat hippocampal nerve terminals, asiatic acid attenuates the calcium influx via N- and P/Q-type calcium channels, subsequently suppressing protein kinase C activity and decreasing glutamate release.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/drug effects , Pentacyclic Triterpenes/pharmacology , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Centella , Hippocampus/metabolism , Hippocampus/physiology , Indoles/pharmacology , Male , Maleimides/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism , omega-Conotoxins/pharmacology
4.
Insect Biochem Mol Biol ; 87: 75-80, 2017 08.
Article in English | MEDLINE | ID: mdl-28668511

ABSTRACT

The goal of this research was to express receptors and ion channels in hormone-treated insect cell lines. Treatment of Anopheles gambiae Sua1B cells with 20-hydroxyecdysone showed an inhibition of cell growth over a time course of three days, with no change in cellular morphology. The effect of 20-hydroxyecdysone was enhanced in the presence of the potassium channel blocker 4-aminopyridine, but not tetraethylammonium. Concentration-response curves of 4-aminopyridine in the presence of 42 µM (1 mg/ml) 20-hydroxyecdysone showed similar IC50 values (6-10 µM) across 3 day exposures. Whole cell patch clamp confirmed the expression of delayed-rectifier (Kv2) potassium channels in hormone-supplemented Sua1B cells, whereas untreated Sua1B cells showed no evidence of Kv2 expression. The hormone-induced expression of Kv2 channels occurred in as little as 4 h after treatment, but were not observed after 24 h of exposure to 20-hydroxyecdysone, suggesting they played a role in cell death. The expressed channels had current-voltage relationships diagnostic for the Kv2 subtype, and were inhibited with an IC50 = 13 mM of tetraethylammonium. Overall, these parameters were similar to Anopheles gambiae Kv2 potassium channels expressed in HEK-293 cells. The induced presence of ion channels (and possibly receptors) in these cells has potential utility for high throughput screening and basic neuroscience research.


Subject(s)
Anopheles/drug effects , Ecdysterone/pharmacology , Shab Potassium Channels/metabolism , 4-Aminopyridine/pharmacology , Animals , Anopheles/cytology , Anopheles/metabolism , Cell Line , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Tetraethylammonium/pharmacology
5.
Epilepsia ; 58(9): 1637-1644, 2017 09.
Article in English | MEDLINE | ID: mdl-28691204

ABSTRACT

OBJECTIVE: Sudden unexplained death in epilepsy is the leading cause of death in young adult epilepsy patients, typically occurring during the early postictal period, presumably resulting from brainstem and cardiorespiratory dysfunction. We hypothesized that ictal discharges in the brainstem disrupt the cardiorespiratory network, causing mortality. To study this hypothesis, we chose an animal model comprising focal unilateral hippocampal injection of 4-aminopyridine (4-AP), which produced focal recurrent hippocampal seizures with secondary generalization in awake, behaving rats. METHODS: We studied ictal and interictal intracranial electrographic activity (iEEG) in 23 rats implanted with a custom electrode array into the hippocampus, the contralateral cortex, and brainstem. The hippocampal electrodes contained a cannula to administer the potassium channel blocker and convulsant (4-AP). iEEG was recorded continuously before, during, and after seizures induced by 4-AP infusion into the hippocampus. RESULTS: The control group (n = 5) was monitored for 2-3 months, and the weekly baseline iEEG recordings showed long-term stability. The low-dose group (1 µL 4-AP, 40 mm, n = 5) exhibited local electrographic seizures without spread to the contralateral cerebral cortex or brainstem. The high-dose group (5 µL 4-AP, 40 mm, n = 3) had several hippocampal electrographic seizures, which spread contralaterally and triggered brainstem discharges within 40 min, and were associated with violent motor seizures followed by dyspnea and respiratory arrest, with cortical and hippocampal iEEG flattening. The group that received high-dose 4-AP without brainstem implantation (n = 5) had similar seizure-related respiratory difficulties. Finally, five rats that received high-dose 4-AP without EEG recording also developed violent motor seizures with postictal respiratory arrest. Following visualized respiratory arrest in groups III, IV, and V, manual respiratory resuscitation was successful in five of 13 animals. SIGNIFICANCE: These studies show that hippocampal seizure activity can spread or trigger brainstem epileptiform discharges that may cause mortality, possibly mediated by respiratory network dysfunction.


Subject(s)
4-Aminopyridine/pharmacology , Brain Stem/drug effects , Hippocampus/drug effects , Seizures/chemically induced , Animals , Electroencephalography/drug effects , Male , Rats , Rats, Wistar , Recurrence , Seizures/mortality
6.
J Neuroimmunol ; 305: 72-74, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28284349

ABSTRACT

We investigated the effect of 4-aminopyridine (4-AP) on experimental autoimmune neuritis (EAN) using a 4-AP-treated group in which 4-AP was administered in the diet, and a control group (n=10 per group). Electrophysiological and pathological assessment was performed in the sciatic nerve. The EAN clinical scores were significantly lower in the 4-AP-treated group than in the control group (p<0.05). The motor conductance velocity two weeks post-immunization was significantly higher in the 4-AP-treated group (p<0.05). Finally, 4-AP did not lead to pathological changes. Thus, 4-AP might be a potential therapeutic agent in demyelinating neuropathy.


Subject(s)
4-Aminopyridine/therapeutic use , Neuritis, Autoimmune, Experimental/drug therapy , Potassium Channel Blockers/therapeutic use , 4-Aminopyridine/pharmacology , Animals , Disease Models, Animal , Electromyography , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Freund's Adjuvant/toxicity , Male , Myelin Proteins/toxicity , Neural Conduction/drug effects , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/immunology , Potassium Channel Blockers/pharmacology , Rats , Rats, Inbred Lew , Statistics, Nonparametric , Vaccination/adverse effects
7.
Neurorehabil Neural Repair ; 31(4): 387-396, 2017 04.
Article in English | MEDLINE | ID: mdl-28107804

ABSTRACT

BACKGROUND: 4-Aminopyridine (4-AP) is a Food and Drug Administration-approved drug to improve motor function in people with multiple sclerosis. Preliminary results suggest the drug may act on intact neural circuits and not just on demyelinated ones. OBJECTIVE: To determine if 4-AP at clinically relevant levels alters the excitability of intact motor circuits. METHODS: In anesthetized rats, electrodes were placed over motor cortex and the dorsal cervical spinal cord for electrical stimulation, and electromyogram electrodes were inserted into biceps muscle to measure responses. The motor responses to brain and spinal cord stimulation were measured before and for 5 hours after 4-AP administration both in uninjured rats and rats with a cut lesion of the pyramidal tract. Blood was collected at the same time as electrophysiology to determine drug plasma concentration with a goal of 20 to 100 ng/mL. RESULTS: We first determined that a bolus infusion of 0.32 mg/kg 4-AP was optimal: it produced on average 61.5 ± 1.8 ng/mL over the 5 hours after infusion. This dose of 4-AP increased responses to spinal cord stimulation by 1.3-fold in uninjured rats and 3-fold in rats with pyramidal tract lesion. Responses to cortical stimulation also increased by 2-fold in uninjured rats and up to 4-fold in the injured. CONCLUSION: Clinically relevant levels of 4-AP strongly augment physiological responses in intact circuits, an effect that was more robust after partial injury, demonstrating its broad potential in treating central nervous system injuries.


Subject(s)
4-Aminopyridine/pharmacology , Central Nervous System Agents/pharmacology , Cervical Cord/drug effects , Motor Cortex/drug effects , Pyramidal Tracts/drug effects , Spinal Cord Injuries/drug therapy , 4-Aminopyridine/blood , 4-Aminopyridine/pharmacokinetics , Animals , Central Nervous System Agents/blood , Central Nervous System Agents/pharmacokinetics , Cervical Cord/injuries , Cervical Cord/physiology , Cervical Cord/physiopathology , Drug Evaluation, Preclinical , Electric Stimulation , Electromyography , Evoked Potentials, Motor/drug effects , Evoked Potentials, Motor/physiology , Female , Forelimb/drug effects , Forelimb/physiology , Forelimb/physiopathology , Microelectrodes , Motor Cortex/physiology , Motor Cortex/physiopathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Pyramidal Tracts/injuries , Pyramidal Tracts/physiology , Pyramidal Tracts/physiopathology , Random Allocation , Rats, Sprague-Dawley , Spinal Cord Injuries/physiopathology
8.
Fish Shellfish Immunol ; 60: 426-435, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27744058

ABSTRACT

Potassium ion channels are one of the most diversely and widely distributed channels, which are involved in all kinds of physiological functions in both excitable and non-excitable cells. The expression of voltage-gated potassium ion (Kv) channels is highly variable according to the state of macrophages activation. Macrophages have an important function in innate immunity against intruding pathogens. They produce a variety of inflammatory and immunoactive molecules that modulate imflammatory responses. Here we show that blockade of K+ channels by non-selective Kv channel inhibitor tetraethylammonium chloride (TEA), and 4-aminopyridine (4-AP) inhibited proinflammatory cytokines expression, cell proliferation, and reactive oxygen species (ROS) production in LPS-stimulated macrophages of Sea perch (Lateolabrax japonicas). Then we isolated four Kv channels genes (spKv1.1, spKv1.2, spKv1.5 and spKv3.1) in LPS-activated fish macrophages. These channels genes were up-regulated after LPS stimulation except spKv3.1, which remained unchanged during the test. The results of this study indicate that Kv channels could be required for modulating the immune function of fish macrophages.


Subject(s)
Cytokines/genetics , Fish Proteins/genetics , Macrophage Activation/drug effects , Perciformes/genetics , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/genetics , Reactive Oxygen Species/metabolism , 4-Aminopyridine/pharmacology , Amino Acid Sequence , Animals , Cloning, Molecular , Cytokines/immunology , Cytokines/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Fish Proteins/chemistry , Fish Proteins/metabolism , Immunity, Innate/drug effects , Immunity, Innate/immunology , Lipopolysaccharides/pharmacology , Perciformes/immunology , Perciformes/metabolism , Phylogeny , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary , Tetraethylammonium/pharmacology
9.
Sci Rep ; 6: 34749, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703262

ABSTRACT

4-aminopyridine is commonly used to stimulate neurotransmitter release resulting from sustained plasma membrane depolarization and Ca2+-influx from the extracellular space. This paper elucidated unconventional mechanism of 4-aminopyridine-stimulated glutamate release from neurons and non-neuronal cells which proceeds in the absence of external Ca2+. In brain nerve terminals, primary neurons and platelets 4-aminopyridine induced the exocytotic release of glutamate that was independent of external Ca2+ and was triggered by the sequestration of Ca2+ from intracellular stores. The initial level of 4-aminopyridine-stimulated glutamate release from neurons in the absence or presence of external Ca2+ was subequal and the difference was predominantly associated with subsequent tonic release of glutamate in Ca2+-supplemented medium. The increase in [Ca2+]i and the secretion of glutamate stimulated by 4-aminopyridine in Ca2+-free conditions have resulted from Ca2+ efflux from endoplasmic reticulum and were abolished by intracellular free Ca2+ chelator BAPTA. This suggests that Ca2+ sequestration plays a profound role in the 4-aminopyridine-mediated stimulation of excitable and non-excitable cells. 4-Aminopyridine combines the properties of depolarizing agent with the ability to sequester intracellular Ca2+. The study unmasks additional mechanism of action of 4-aminopyridine, an active substance of drugs for treatment of multiple sclerosis and conditions related to reduced Ca2+ efflux from intracellular stores.


Subject(s)
4-Aminopyridine/pharmacology , Calcium/metabolism , Neurons/drug effects , Potassium Channel Blockers/pharmacology , Animals , Cells, Cultured , Endoplasmic Reticulum/metabolism , Exocytosis , Glutamic Acid/metabolism , Male , Mice , Neurons/cytology , Neurons/metabolism , Rabbits , Rats
10.
J Neurophysiol ; 116(6): 2550-2563, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27605536

ABSTRACT

The coding of sound level by ensembles of neurons improves the accuracy with which listeners identify how loud a sound is. In the auditory system, the rate at which neurons fire in response to changes in sound level is shaped by local networks. Voltage-gated conductances alter local output by regulating neuronal firing, but their role in modulating responses to sound level is unclear. We tested the effects of L-type calcium channels (CaL: CaV1.1-1.4) on sound-level coding in the central nucleus of the inferior colliculus (ICC) in the auditory midbrain. We characterized the contribution of CaL to the total calcium current in brain slices and then examined its effects on rate-level functions (RLFs) in vivo using single-unit recordings in awake mice. CaL is a high-threshold current and comprises ∼50% of the total calcium current in ICC neurons. In vivo, CaL activates at sound levels that evoke high firing rates. In RLFs that increase monotonically with sound level, CaL boosts spike rates at high sound levels and increases the maximum firing rate achieved. In different populations of RLFs that change nonmonotonically with sound level, CaL either suppresses or enhances firing at sound levels that evoke maximum firing. CaL multiplies the gain of monotonic RLFs with dynamic range and divides the gain of nonmonotonic RLFs with the width of the RLF. These results suggest that a single broad class of calcium channels activates enhancing and suppressing local circuits to regulate the sensitivity of neuronal populations to sound level.


Subject(s)
Action Potentials/physiology , Calcium Channels, L-Type/metabolism , Inferior Colliculi/cytology , Neurons/physiology , Sound , 4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/pharmacology , Acoustic Stimulation , Action Potentials/drug effects , Amifampridine , Animals , Biophysical Phenomena/drug effects , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Mice , Mice, Inbred CBA , Nimodipine/pharmacology , Potassium Channel Blockers/pharmacology , Quinoxalines/pharmacology , Wakefulness , omega-Conotoxin GVIA/pharmacology
11.
Article in English | MEDLINE | ID: mdl-26635537

ABSTRACT

A fundamental question in vision neuroscience is how parallel processing of Retinal Ganglion Cell (RGC) signals is integrated at the level of the visual thalamus. It is well-known that parallel ON-OFF pathways generate output signals from the retina that are conveyed to the dorsal lateral geniculate nucleus (dLGN). However, it is unclear how these signals distribute onto thalamic cells and how these two pathways interact. Here, by electrophysiological recordings and c-Fos expression analysis, we characterized the effects of pharmacological manipulations of the retinal circuit aimed at inducing either a selective activation of a single pathway, OFF RGCs [intravitreal L-(+)-2-Amino-4-phosphonobutyric, L-AP4] or an unregulated activity of all classes of RGCs (intravitreal 4-Aminopyridine, 4-AP). In in vivo experiments, the analysis of c-Fos expression in the dLGN showed that these two manipulations recruited active cells from the same area, the lateral edge of the dLGN. Despite this similarity, the unregulated co-activation of both ON and OFF pathways by 4-AP yielded a much stronger recruitment of GABAergic interneurons in the dLGN when compared to L-AP4 pure OFF activation. The increased activation of an inhibitory thalamic network by a high level of unregulated discharge of ON and OFF RGCs might suggest that cross-inhibitory pathways between opposing visual channels are presumably replicated at multiple levels in the visual pathway, thus increasing the filtering ability for non-informative or noisy visual signals.


Subject(s)
GABAergic Neurons/physiology , Retinal Ganglion Cells/physiology , Thalamus/physiology , Visual Pathways/physiology , Visual Perception/physiology , 4-Aminopyridine/pharmacology , Action Potentials , Aminobutyrates/pharmacology , Animals , Evoked Potentials, Visual , Excitatory Amino Acid Agonists/pharmacology , Interneurons/physiology , Male , Models, Neurological , Monte Carlo Method , Photic Stimulation , Potassium Channel Blockers/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Signal Processing, Computer-Assisted , Tissue Culture Techniques , Visual Pathways/drug effects , Visual Perception/drug effects
12.
Neurotoxicology ; 50: 157-69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26342684

ABSTRACT

The citrus flavonoid hesperidin exerts neuroprotective effects and could cross the blood-brain barrier. Given the involvement of glutamate neurotoxicity in the pathogenesis of neurodegenerative disorders, this study was conducted to evaluate the potential role of hesperidin in glutamate release and glutamate neurotoxicity in the hippocampus of rats. In rat hippocampal nerve terminals (synaptosomes), hesperidin inhibited the release of glutamate and elevation of cytosolic free Ca(2+) concentration evoked by 4-aminopyridine (4-AP), but did not alter 4-AP-mediated depolarization. The inhibitory effect of hesperidin on evoked glutamate release was prevented by chelating the extracellular Ca(2+) ions and blocking the activity of Cav2.2 (N-type) and Cav2.1 (P/Q-type) channels or protein kinase C. In hippocampal slice preparations, whole-cell patch clamp experiments showed that hesperidin reduced the frequency of spontaneous excitatory postsynaptic currents without affecting their amplitude, indicating the involvement of a presynaptic mechanism. In addition, intraperitoneal (i.p.) injection of kainic acid (KA, 15 mg/kg) elevated the extracellular glutamate levels and caused considerable neuronal loss in the hippocampal CA3 area. These KA-induced alterations were attenuated by pretreatment with hesperidin (10 or 50 mg/kg, i.p.) before administering the KA. These results demonstrate that hesperidin inhibits evoked glutamate release in vitro and attenuates in vivo KA-induced neuronal death in the hippocampus. Our findings indicate that hesperidin may be a promising candidate for preventing or treating glutamate excitotoxicity related brain disorders such as neurodegenerative diseases.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/metabolism , Hesperidin/therapeutic use , Hippocampus/metabolism , Kainic Acid/toxicity , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/etiology , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Disease Models, Animal , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/ultrastructure , Male , Membrane Potentials/drug effects , Neurotoxicity Syndromes/pathology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism , Tetrodotoxin/pharmacology
13.
J Neurophysiol ; 113(10): 3930-42, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25761950

ABSTRACT

Despite its widespread use, high-resolution imaging with multiphoton microscopy to record neuronal signals in vivo is limited to the surface of brain tissue because of limited light penetration. Moreover, most imaging studies do not simultaneously record electrical neural activity, which is, however, crucial to understanding brain function. Accordingly, we developed a photometric patch electrode (PME) to overcome the depth limitation of optical measurements and also enable the simultaneous recording of neural electrical responses in deep brain regions. The PME recoding system uses a patch electrode to excite a fluorescent dye and to measure the fluorescence signal as a light guide, to record electrical signal, and to apply chemicals to the recorded cells locally. The optical signal was analyzed by either a spectrometer of high light sensitivity or a photomultiplier tube depending on the kinetics of the responses. We used the PME in Oregon Green BAPTA-1 AM-loaded avian auditory nuclei in vivo to monitor calcium signals and electrical responses. We demonstrated distinct response patterns in three different nuclei of the ascending auditory pathway. On acoustic stimulation, a robust calcium fluorescence response occurred in auditory cortex (field L) neurons that outlasted the electrical response. In the auditory midbrain (inferior colliculus), both responses were transient. In the brain-stem cochlear nucleus magnocellularis, calcium response seemed to be effectively suppressed by the activity of metabotropic glutamate receptors. In conclusion, the PME provides a powerful tool to study brain function in vivo at a tissue depth inaccessible to conventional imaging devices.


Subject(s)
Brain Mapping , Brain/cytology , Brain/physiology , Fluorescence , Neurons/physiology , 4-Aminopyridine/pharmacology , Acoustic Stimulation , Action Potentials/drug effects , Action Potentials/physiology , Animals , Animals, Newborn , Auditory Pathways/physiology , Calcium/metabolism , Chickens , Electric Stimulation , GABA Antagonists/pharmacology , In Vitro Techniques , Neurons/drug effects , Photometry , Potassium Channel Blockers/pharmacology , Pyridazines/pharmacology , Tetraethylammonium Compounds/pharmacology , Transfection
14.
J Neurochem ; 130(6): 770-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903676

ABSTRACT

In the present study, the effects of the two classical anti-epileptic drugs, carbamazepine and valproic acid, and the non-classical anti-seizure drug vinpocetine were investigated on the expression of the pro-inflammatory cytokines IL-1ß and TNF-α in the hippocampus of rats by PCR or western blot after the administration of one or seven doses. Next, the effects of the anti-seizure drugs were investigated on the rise in cytokine expression induced by lipopolysaccharides (LPS) inoculation in vivo. To validate our methods, the changes induced by the pro-convulsive agents 4-aminopyridine, pentylenetetrazole and pilocarpine were also tested. Finally, the effect of the anti-seizure drugs on seizures and on the concomitant rise in pro-inflammatory cytokine expression induced by 4-aminopyridine was explored. Results show that vinpocetine and carbamazepine reduced the expression of IL-1ß and TNF-α from basal conditions, and the increase in both pro-inflammatory cytokines induced by LPS. In contrast, valproic acid failed to reduce both the expression of the cytokines from basal conditions and the rise in IL-1ß and TNF-α expression induced by LPS. Tonic-clonic seizures induced either by 4-aminopyridine, pentylenetetrazole or pilocarpine increased the expression of IL-1ß and TNF-α markedly. 4-aminopyridine-induced changes were reduced by all the tested anti-seizure drugs, although valproic acid was less effective. We conclude that the anti-seizure drugs, vinpocetine and carbamazepine, whose mechanisms of action involve a decrease in ion channels permeability, also reduce cerebral inflammation. The mechanism of action of anti-seizure drugs like vinpocetine and carbamazepine involves a decrease in Na(+) channels permeability. We here propose that this mechanism of action also involves a decrease in cerebral inflammation.


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/pharmacology , Hippocampus/metabolism , Interleukin-1beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Valproic Acid/pharmacology , Vinca Alkaloids/pharmacology , 4-Aminopyridine/antagonists & inhibitors , 4-Aminopyridine/pharmacology , Animals , Behavior, Animal/drug effects , Blotting, Western , Convulsants/pharmacology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Epilepsy, Tonic-Clonic/chemically induced , Epilepsy, Tonic-Clonic/physiopathology , Hippocampus/drug effects , Interleukin-1beta/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Polymerase Chain Reaction , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/antagonists & inhibitors
15.
Circulation ; 130(3): 224-34, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24895457

ABSTRACT

BACKGROUND: New drugs are routinely screened for IKr blocking properties thought to predict QT prolonging and arrhythmogenic liability. However, recent data suggest that chronic (hours) drug exposure to phosphoinositide 3-kinase inhibitors used in cancer can prolong QT by inhibiting potassium currents and increasing late sodium current (INa-L) in cardiomyocytes. We tested the extent to which IKr blockers with known QT liability generate arrhythmias through this pathway. METHODS AND RESULTS: Acute exposure to dofetilide, an IKr blocker without other recognized electropharmacologic actions, produced no change in ion currents or action potentials in adult mouse cardiomyocytes, which lack IKr. By contrast, 2 to 48 hours of exposure to the drug generated arrhythmogenic afterdepolarizations and ≥15-fold increases in INa-L. Including phosphatidylinositol 3,4,5-trisphosphate, a downstream effector for the phosphoinositide 3-kinase pathway, in the pipette inhibited these effects. INa-L was also increased, and inhibitable by phosphatidylinositol 3,4,5-trisphosphate, with hours of dofetilide exposure in human-induced pluripotent stem cell-derived cardiomyocytes and in Chinese hamster ovary cells transfected with SCN5A, encoding sodium current. Cardiomyocytes from dofetilide-treated mice similarly demonstrated increased INa-L and afterdepolarizations. Other agents with variable IKr-blocking potencies and arrhythmia liability produced a range of effects on INa-L, from marked increases (E-4031, d-sotalol, thioridazine, and erythromycin) to little or no effect (haloperidol, moxifloxacin, and verapamil). CONCLUSIONS: Some but not all drugs designated as arrhythmogenic IKr blockers can generate arrhythmias by augmenting INa-L through the phosphoinositide 3-kinase pathway. These data identify a potential mechanism for individual susceptibility to proarrhythmia and highlight the need for a new paradigm to screen drugs for QT prolonging and arrhythmogenic liability.


Subject(s)
Drug Evaluation, Preclinical/methods , NAV1.5 Voltage-Gated Sodium Channel/drug effects , NAV1.5 Voltage-Gated Sodium Channel/physiology , Potassium Channel Blockers/pharmacology , Torsades de Pointes/epidemiology , 4-Aminopyridine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques , Phenethylamines/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Risk Factors , Signal Transduction/physiology , Sulfonamides/pharmacology , Torsades de Pointes/physiopathology , Transfection
16.
Planta Med ; 80(6): 465-72, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24710898

ABSTRACT

Quercetin is one of the most common flavonoids in the human daily diet. Its affects the coronary artery, especially L-type voltage-gated Ca2+ channels and voltage-gated K+ channels in the arterial smooth muscle cells, which are poorly understood. The present experiments were designed to study the myogenic effect of quercetin and its possible underlying mechanisms in the rat coronary artery. A wire myograph was used to observe the myogenic effects. Arterial smooth muscle cells were freshly isolated from the rat coronary artery and the intracellular free Ca2+ concentration was measured with molecular probe fluo-4-AM. The effects of quercetin on L-type voltage-gated Ca2+ channels and voltage-gated K+ channels were studied using a whole-cell patch clamp. Quercetin (3-30 µM) produced a depression and relaxation on the contraction induced by KCl or the thromboxane A2 analog 9,11-Dideoxy-9α,11α-methanoepoxy prostaglandin F 2α . The vasorelaxation was attenuated by 4-aminopyridine, a specific voltage-gated K+ channel inhibitor, but was not affected by the NG-nitro-L-arginine methylester ester (a nitric oxide synthesis inhibitor), glibenclamide (a ATP-activated K+ channel inhibitor), iberiotoxin (a Ca2+-activated K+ channel inhibitor), BaCl2 (an inward rectifier K+ channel inhibitor), or by endothelium denudation. At the same concentrations, quercetin reduced the KCl-induced elevation of the intracellular free Ca2+ concentration, inhibited the inward Ca2+ currents through L-type voltage-gated Ca2+ channels, and increased the outward K+ currents through voltage-gated K+ channels in the rat coronary artery smooth muscle cells. Collectively, our results demonstrate that quercetin possesses vasospasmolytic effects in RCA and suggest that depression of the Ca2+ influx through L-type voltage-gated Ca2+ channels and augmentation of voltage-gated K+ channel activity in the myocytes may underlie coronary relaxation.


Subject(s)
Calcium Channels, L-Type/metabolism , Coronary Vessels/drug effects , Muscle, Smooth, Vascular/drug effects , Potassium Channels, Voltage-Gated/metabolism , Quercetin/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , 4-Aminopyridine/pharmacology , Aniline Compounds , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Coronary Vessels/cytology , Coronary Vessels/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Patch-Clamp Techniques , Plant Extracts/pharmacology , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Xanthenes
17.
PLoS One ; 9(2): e88644, 2014.
Article in English | MEDLINE | ID: mdl-24520409

ABSTRACT

An excessive release of glutamate is considered to be a molecular mechanism associated with several neurological diseases that causes neuronal damage. Therefore, searching for compounds that reduce glutamate neurotoxicity is necessary. In this study, the possibility that the natural flavone acacetin derived from the traditional Chinese medicine Clerodendrum inerme (L.) Gaertn is a neuroprotective agent was investigated. The effect of acacetin on endogenous glutamate release in rat hippocampal nerve terminals (synaptosomes) was also investigated. The results indicated that acacetin inhibited depolarization-evoked glutamate release and cytosolic free Ca(2+) concentration ([Ca(2+)]C) in the hippocampal nerve terminals. However, acacetin did not alter synaptosomal membrane potential. Furthermore, the inhibitory effect of acacetin on evoked glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker known as ω-conotoxin MVIIC. In a kainic acid (KA) rat model, an animal model used for excitotoxic neurodegeneration experiments, acacetin (10 or 50 mg/kg) was administrated intraperitoneally to the rats 30 min before the KA (15 mg/kg) intraperitoneal injection, and subsequently induced the attenuation of KA-induced neuronal cell death and microglia activation in the CA3 region of the hippocampus. The present study demonstrates that the natural compound, acacetin, inhibits glutamate release from hippocampal synaptosomes by attenuating voltage-dependent Ca(2+) entry and effectively prevents KA-induced in vivo excitotoxicity. Collectively, these data suggest that acacetin has the therapeutic potential for treating neurological diseases associated with excitotoxicity.


Subject(s)
Flavones/pharmacology , Glutamic Acid/metabolism , Kainic Acid/toxicity , Neurons/metabolism , Neurons/pathology , Neurotoxins/toxicity , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cell Death/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Membrane Potentials/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nerve Endings/drug effects , Nerve Endings/metabolism , Neurons/drug effects , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
18.
PLoS One ; 8(11): e78727, 2013.
Article in English | MEDLINE | ID: mdl-24236040

ABSTRACT

Nitric oxide (NO) is an unconventional membrane-permeable messenger molecule that has been shown to play various roles in the nervous system. How NO modulates ion channels to affect neuronal functions is not well understood. In gastropods, NO has been implicated in regulating the feeding motor program. The buccal motoneuron, B19, of the freshwater pond snail Helisoma trivolvis is active during the hyper-retraction phase of the feeding motor program and is located in the vicinity of NO-producing neurons in the buccal ganglion. Here, we asked whether B19 neurons might serve as direct targets of NO signaling. Previous work established NO as a key regulator of growth cone motility and neuronal excitability in another buccal neuron involved in feeding, the B5 neuron. This raised the question whether NO might modulate the electrical activity and neuronal excitability of B19 neurons as well, and if so whether NO acted on the same or a different set of ion channels in both neurons. To study specific responses of NO on B19 neurons and to eliminate indirect effects contributed by other cells, the majority of experiments were performed on single cultured B19 neurons. Addition of NO donors caused a prolonged depolarization of the membrane potential and an increase in neuronal excitability. The effects of NO could mainly be attributed to the inhibition of two types of calcium-activated potassium channels, apamin-sensitive and iberiotoxin-sensitive potassium channels. NO was found to also cause a depolarization in B19 neurons in situ, but only after NO synthase activity in buccal ganglia had been blocked. The results suggest that NO acts as a critical modulator of neuronal excitability in B19 neurons, and that calcium-activated potassium channels may serve as a common target of NO in neurons.


Subject(s)
Motor Neurons/physiology , Nitric Oxide/physiology , Potassium Channels, Calcium-Activated/metabolism , 4-Aminopyridine/pharmacology , Action Potentials , Animals , Apamin/pharmacology , Calcium Channels/metabolism , Cells, Cultured , Ganglia, Autonomic/cytology , Growth Cones/physiology , Helix, Snails , Nitric Oxide Donors/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/agonists , Tetraethylammonium/pharmacology
19.
J Pharmacol Sci ; 123(2): 147-58, 2013.
Article in English | MEDLINE | ID: mdl-24096832

ABSTRACT

To develop a simple screening system for blockers of voltage-gated Kv1.3 and Kv1.5 channels, new cell lines co-expressing mutated Nav1.5 (IFM/Q3), Kir2.1 (Kir), and Kv1.3 or Kv1.5 were introduced as IFM/Q3+Kir+Kv1.3 and IFM/Q3+Kir+Kv1.5, respectively. Electrical stimulation (ES) of a cell line, IFM/Q3+Kir, induced prolonged action potentials due to the slow inactivation of IFM/Q3 and subsequent cell death. Additional co-expression of Kv1.3 or Kv1.5 to IFM/Q3+Kir shortened the evoked action potentials and prevented cell death. In the presence of margatoxin, a selective Kv1.3-blocker, ES induced cell death in IFM/Q3+Kir+Kv1.3, but not in IFM/Q3+Kir+Kv1.5. In the presence of 4-aminopyridine, a non-selective Kv-channel blocker, ES application elicited cell death in both cell lines. The IC50s of acacetin, a Kv1.5-blocker, was 10.2 µM in IFM/Q3+Kir+Kv1.3 and almost identical to that in IFM/Q3+Kir+Kv1.5 (7.6 µM). The IC50s of citalopram, a 5-HT uptake-inhibitor, were 1.8 µM in IFM/Q3+Kir+Kv1.3 and 1.5 µM in IFM/Q3+Kir+Kv1.5, respectively. These IC50s were comparable to those determined electrophysiologically. In conclusion, acacetin and citalopram block both Kv1.3 and Kv1.5 without selectivity. The Kv1.3 or Kv1.5 channel inhibition assay using these new cell lines may be applicable to high-throughput screening because of its simplicity, accuracy, and high cost-performance.


Subject(s)
Action Potentials/drug effects , Cell Death/drug effects , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , 4-Aminopyridine/pharmacology , Cell Survival/drug effects , Citalopram/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation , Evoked Potentials/drug effects , Flavones/pharmacology , HEK293 Cells , Humans , Mutation , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Scorpion Venoms/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology
20.
J Ethnopharmacol ; 147(2): 488-96, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23542145

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Danshen is a commonly used traditional Chinese medicine and has received considerable attention due to their beneficial effects on the health, including prevention of cardiovascular disease, and cancer. Tanshinone IIA, a major active constituent of Danshen, has been reported to have a neuroprotective profile. AIM OF THE STUDY: An excessive release of glutamate is considered to be related to neuropathology of several neurological diseases. In this study, we investigated whether tanshinone IIA could affect endogenous glutamate release and explored the possible mechanism. MATERIALS AND METHODS: The experimental model was the isolated nerve terminals (synaptosomes) purified from the rat cerebral cortex. The release of glutamate was evoked by the K(+) channel blocker 4-aminopyridine (4-AP) and measured by one-line enzyme-coupled fluorometric assay. We also used a membrane potential-sensitive dye to assay nerve terminal excitability and depolarization, and a Ca(2+) indicator, Fura-2-acetoxymethyl ester, to monitor cytosolic Ca(2+) concentrations ([Ca(2+)]C). RESULTS: Tanshinone IIA inhibited the release of glutamate evoked by 4-AP in a concentration-dependent manner. Inhibition of glutamate release by tanshinone IIA was prevented by the chelating the extracellular Ca(2+) ions, and by the vesicular transporter inhibitor bafilomycin A1. However, the glutamate transporter inhibitor DL-threo-beta-benzyl-oxyaspartate did not have any effect on the action of tanshinone IIA. Tanshinone IIA decreased the depolarization-induced increase in [Ca(2+)]C, whereas it did not alter the resting synaptosomal membrane potential or 4-AP-mediated depolarization. Furthermore, the effect of tanshinone IIA on evoked glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC, but not by the ryanodine receptor blocker dantrolene or the mitochondrial Na(+)/Ca(2+) exchanger blocker CGP37157. Mitogen-activated protein kinase (MEK) inhibition also prevented the inhibitory effect of tanshinone IIA on evoked glutamate release. CONCLUSION: These results show that tanshinone IIA inhibits glutamate release from cortical synaptosomes in rats through the suppression of presynaptic voltage-dependent Ca(2+) entry and MEK signaling cascade.


Subject(s)
Abietanes/pharmacology , Cerebral Cortex/cytology , Glutamic Acid/metabolism , Synaptosomes/metabolism , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Cell Membrane/physiology , Drugs, Chinese Herbal , Male , Membrane Potentials , Mitogen-Activated Protein Kinases/metabolism , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Salvia miltiorrhiza
SELECTION OF CITATIONS
SEARCH DETAIL