Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3253-3267, 2023 11.
Article in English | MEDLINE | ID: mdl-37231170

ABSTRACT

Due to the role of astrocytes and microglia in the pathophysiology of epilepsy and limited studies of antiseizure medication (ASM) effects on glial cells, we studied tiagabine (TGB) and zonisamide (ZNS) in an astrocyte-microglia co-culture model of inflammation. Different concentrations of ZNS (10, 20, 40, 100 µg/ml) or TGB (1, 10, 20, 50 µg/ml) were added to primary rat astrocytes co-cultures with 5-10% (M5, physiological conditions) or 30-40% (M30, pathological inflammatory conditions) microglia for 24 h, aiming to study glial viability, microglial activation, connexin 43 (Cx43) expression and gap-junctional coupling. ZNS led to the reduction of glial viability by only 100 µg/ml under physiological conditions. By contrast, TGB revealed toxic effects with a significant, concentration-dependent reduction of glial viability under physiological and pathological conditions. After the incubation of M30 co-cultures with 20 µg/ml TGB, the microglial activation was significantly decreased and resting microglia slightly increased, suggesting possible anti-inflammatory features of TGB under inflammatory conditions. Otherwise, ZNS caused no significant changes of microglial phenotypes. The gap-junctional coupling was significantly decreased after the incubation of M5 co-cultures with 20 and 50 µg/ml TGB, which can be related to its anti-epileptic activity under noninflammatory conditions. A significant decrease of Cx43 expression and cell-cell coupling was found after the incubation of M30 co-cultures with 10 µg/ml ZNS, suggesting additional anti-seizure effects of ZNS with the disruption of glial gap-junctional communication under inflammatory conditions. TGB and ZNS differentially regulated the glial properties. Developing novel ASMs targeting glial cells may have future potential as an "add-on" therapy to classical ASMs targeting neurons.


Subject(s)
Astrocytes , Microglia , Rats , Animals , Coculture Techniques , Tiagabine/metabolism , Tiagabine/pharmacology , Connexin 43/metabolism , Zonisamide/pharmacology , Zonisamide/metabolism , Cell Communication , Neuroglia/metabolism , Inflammation/pathology
2.
Nature ; 606(7915): 820-826, 2022 06.
Article in English | MEDLINE | ID: mdl-35676483

ABSTRACT

γ-Aminobutyric acid (GABA) transporter 1 (GAT1)1 regulates neuronal excitation of the central nervous system by clearing the synaptic cleft of the inhibitory neurotransmitter GABA upon its release from synaptic vesicles. Elevating the levels of GABA in the synaptic cleft, by inhibiting GABA reuptake transporters, is an established strategy to treat neurological disorders, such as epilepsy2. Here we determined the cryo-electron microscopy structure of full-length, wild-type human GAT1 in complex with its clinically used inhibitor tiagabine3, with an ordered part of only 60 kDa. Our structure reveals that tiagabine locks GAT1 in the inward-open conformation, by blocking the intracellular gate of the GABA release pathway, and thus suppresses neurotransmitter uptake. Our results provide insights into the mixed-type inhibition of GAT1 by tiagabine, which is an important anticonvulsant medication. Its pharmacodynamic profile, confirmed by our experimental data, suggests initial binding of tiagabine to the substrate-binding site in the outward-open conformation, whereas our structure presents the drug stalling the transporter in the inward-open conformation, consistent with a two-step mechanism of inhibition4. The presented structure of GAT1 gives crucial insights into the biology and pharmacology of this important neurotransmitter transporter and provides blueprints for the rational design of neuromodulators, as well as moving the boundaries of what is considered possible in single-particle cryo-electron microscopy of challenging membrane proteins.


Subject(s)
GABA Plasma Membrane Transport Proteins , GABA Uptake Inhibitors , gamma-Aminobutyric Acid , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Cryoelectron Microscopy , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/metabolism , GABA Plasma Membrane Transport Proteins/ultrastructure , GABA Uptake Inhibitors/chemistry , GABA Uptake Inhibitors/pharmacology , Humans , Neurotransmitter Agents/metabolism , Protein Conformation/drug effects , Tiagabine/chemistry , Tiagabine/metabolism , Tiagabine/pharmacology , gamma-Aminobutyric Acid/metabolism
3.
Molecules ; 26(12)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207748

ABSTRACT

Tiagabine is an antiepileptic drug used for the treatment of partial seizures in humans. Recently, this drug has been found useful in several non-epileptic conditions, including anxiety, chronic pain and sleep disorders. Since tachycardia-an impairment of cardiac rhythm due to cardiac ion channel dysfunction-is one of the most commonly reported non-neurological adverse effects of this drug, in the present paper we have undertaken pharmacological and numerical studies to assess a potential cardiovascular risk associated with the use of tiagabine. A chemical interaction of tiagabine with a model of human voltage-gated ion channels (VGICs) is described using the molecular docking method. The obtained in silico results imply that the adverse effects reported so far in the clinical cardiological of tiagabine could not be directly attributed to its interactions with VGICs. This is also confirmed by the results from the isolated organ studies (i.e., calcium entry blocking properties test) and in vivo (electrocardiogram study) assays of the present research. It was found that tachycardia and other tiagabine-induced cardiac complications are not due to a direct effect of this drug on ventricular depolarization and repolarization.


Subject(s)
Calcium Channels, L-Type/chemistry , ERG1 Potassium Channel/antagonists & inhibitors , Epilepsy/drug therapy , Heart/drug effects , NAV1.5 Voltage-Gated Sodium Channel/chemistry , Tiagabine/pharmacology , Action Potentials , Animals , Anticonvulsants/adverse effects , Calcium Channels, L-Type/metabolism , Computer Simulation , ERG1 Potassium Channel/metabolism , Epilepsy/complications , Epilepsy/metabolism , Humans , Male , Molecular Docking Simulation/methods , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Rats , Rats, Wistar , Tiagabine/adverse effects
4.
Molecules ; 25(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081136

ABSTRACT

The human gamma aminobutyric acid transporter subtype 1 (hGAT1) located in the nerve terminals is known to catalyze the neuronal function by the electrogenic reuptake of γ-aminobutyric acid (GABA) with the co-transport of Na+ and Cl- ions. In the past, there has been a major research drive focused on the dysfunction of hGAT1 in several neurological disorders. Thus, hGAT1 of the GABAergic system has been well established as an attractive target for such diseased conditions. Till date, there are various reports about stereo selectivity of -COOH group of tiagabine, a Food and Drug Administration (FDA)-approved hGAT1-selective antiepileptic drug. However, the effect of the stereochemistry of the protonated -NH group of tiagabine has never been scrutinized. Therefore, in this study, tiagabine has been used to explore the binding hypothesis of different enantiomers of tiagabine. In addition, the impact of axial and equatorial configuration of the-COOH group attached at the meta position of the piperidine ring of tiagabine enantiomers was also investigated. Further, the stability of the finally selected four hGAT1-tiagabine enantiomers namely entries 3, 4, 6, and 9 was evaluated through 100 ns molecular dynamics (MD) simulations for the selection of the best probable tiagabine enantiomer. The results indicate that the protonated -NH group in the R-conformation and the -COOH group of Tiagabine in the equatorial configuration of entry 4 provide maximum strength in terms of interaction within the hGAT1 binding pocket to prevent the change in hGAT1 conformational state, i.e., from open-to-out to open-to-in as compared to other selected tiagabine enantiomers 3, 6, and 9.


Subject(s)
GABA Agents/chemistry , GABA Plasma Membrane Transport Proteins/chemistry , Tiagabine/chemistry , gamma-Aminobutyric Acid/chemistry , GABA Agents/pharmacology , Humans , Molecular Dynamics Simulation , Protein Binding/drug effects , Stereoisomerism , Tiagabine/pharmacology
5.
J Neurosci ; 40(8): 1640-1649, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31915255

ABSTRACT

To bridge the gap between preclinical cellular models of disease and in vivo imaging of human cognitive network dynamics, there is a pressing need for informative biophysical models. Here we assess dynamic causal models (DCM) of cortical network responses, as generative models of magnetoencephalographic observations during an auditory oddball roving paradigm in healthy adults. This paradigm induces robust perturbations that permeate frontotemporal networks, including an evoked 'mismatch negativity' response and transiently induced oscillations. Here, we probe GABAergic influences in the networks using double-blind placebo-controlled randomized-crossover administration of the GABA reuptake inhibitor, tiagabine (oral, 10 mg) in healthy older adults. We demonstrate the facility of conductance-based neural mass mean-field models, incorporating local synaptic connectivity, to investigate laminar-specific and GABAergic mechanisms of the auditory response. The neuronal model accurately recapitulated the observed magnetoencephalographic data. Using parametric empirical Bayes for optimal model inversion across both drug sessions, we identify the effect of tiagabine on GABAergic modulation of deep pyramidal and interneuronal cell populations. We found a transition of the main GABAergic drug effects from auditory cortex in standard trials to prefrontal cortex in deviant trials. The successful integration of pharmaco- magnetoencephalography with dynamic causal models of frontotemporal networks provides a potential platform on which to evaluate the effects of disease and pharmacological interventions.SIGNIFICANCE STATEMENT Understanding human brain function and developing new treatments require good models of brain function. We tested a detailed generative model of cortical microcircuits that accurately reproduced human magnetoencephalography, to quantify network dynamics and connectivity in frontotemporal cortex. This approach identified the effect of a test drug (GABA-reuptake inhibitor, tiagabine) on neuronal function (GABA-ergic dynamics), opening the way for psychopharmacological studies in health and disease with the mechanistic precision afforded by generative models of the brain.


Subject(s)
Auditory Cortex/diagnostic imaging , Frontal Lobe/diagnostic imaging , Models, Neurological , Nerve Net/diagnostic imaging , Neurons/physiology , Aged , Auditory Cortex/drug effects , Cross-Over Studies , Double-Blind Method , Female , Frontal Lobe/drug effects , GABA Uptake Inhibitors/pharmacology , Humans , Magnetoencephalography/methods , Male , Middle Aged , Nerve Net/drug effects , Neurons/drug effects , Tiagabine/pharmacology
6.
Neuroimage ; 209: 116462, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31857204

ABSTRACT

Neuroimaging studies of the psychedelic state offer a unique window onto the neural basis of conscious perception and selfhood. Despite well understood pharmacological mechanisms of action, the large-scale changes in neural dynamics induced by psychedelic compounds remain poorly understood. Using source-localised, steady-state MEG recordings, we describe changes in functional connectivity following the controlled administration of LSD, psilocybin and low-dose ketamine, as well as, for comparison, the (non-psychedelic) anticonvulsant drug tiagabine. We compare both undirected and directed measures of functional connectivity between placebo and drug conditions. We observe a general decrease in directed functional connectivity for all three psychedelics, as measured by Granger causality, throughout the brain. These data support the view that the psychedelic state involves a breakdown in patterns of functional organisation or information flow in the brain. In the case of LSD, the decrease in directed functional connectivity is coupled with an increase in undirected functional connectivity, which we measure using correlation and coherence. This surprising opposite movement of directed and undirected measures is of more general interest for functional connectivity analyses, which we interpret using analytical modelling. Overall, our results uncover the neural dynamics of information flow in the psychedelic state, and highlight the importance of comparing multiple measures of functional connectivity when analysing time-resolved neuroimaging data.


Subject(s)
Cerebral Cortex/drug effects , Connectome , Hallucinogens/pharmacology , Ketamine/pharmacology , Lysergic Acid Diethylamide/pharmacology , Magnetoencephalography/drug effects , Nerve Net/drug effects , Psilocybin/pharmacology , Adult , Anticonvulsants/pharmacology , Cerebral Cortex/physiology , Female , Hallucinogens/administration & dosage , Humans , Ketamine/administration & dosage , Lysergic Acid Diethylamide/administration & dosage , Male , Nerve Net/physiology , Psilocybin/administration & dosage , Tiagabine/pharmacology , Young Adult
7.
Plant Mol Biol ; 101(6): 561-574, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31621006

ABSTRACT

KEY MESSAGE: We investigated the functions of two cyanobacterial HemY protoporphyrinogen IX oxidase (PPO) genes with in vitro and in vivo assays and evaluated their applicability as resistance traits to PPO-inhibiting herbicides. We isolated HemY-type protoporphyrinogen IX oxidase (PPO) genes from cyanobacteria, OnPPO gene from Oscillatoria nigro-viridis PCC7112 and HaPPO gene from Halothece sp. PCC7418. The alignment of amino acid sequences as well as phylogenetic analyses conducted showed that OnPPO and HaPPO are classified as HemY-type PPO and are more closely related to plastidic PPOs than to mitochondrial PPOs. The PPO-deficient Escherichia coli BT3 strain, which requires heme supplementation, could obtain normal growth in the absence of heme supplementation when complemented with OnPPO and HaPPO. The enzyme assays of OnPPO, HaPPO, and Arabidopsis thaliana PPO1 (AtPPO1) proteins each revealed different kinetic properties in terms of catalytic efficiency, substrate affinity, and the degree of inhibition by PPO inhibitors. In particular, the catalytic efficiencies (kcat/Km) of OnPPO and HaPPO were approximately twofold higher than that of AtPPO1. The elution profiles of all three PPOs, acquired by size-exclusion chromatography, showed only a single peak with a molecular weight of approximately 52-54 kDa, which corresponds to a monomeric form. Moreover, functional complementation with OnPPO and HaPPO in AtPPO1-silenced Arabidopsis resulted in restored growth, whereas AtPPO1-silenced wild type Arabidopsis suffered necrotic death. In addition, we observed that overexpression of OnPPO and HaPPO in Arabidopsis conferred resistance to the PPO-inhibiting herbicides tiafenacil and saflufenacil. These results suggest that two HemY-type PPOs of cyanobacteria can functionally substitute for plastidic PPO activity in Arabidopsis and can enhance resistance to tiafenacil and saflufenacil.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/drug effects , Arabidopsis/enzymology , Protoporphyrinogen Oxidase/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Herbicide Resistance/genetics , Herbicide Resistance/physiology , Protoporphyrinogen Oxidase/genetics , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Tiagabine/pharmacology
8.
Neuropharmacology ; 158: 107733, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31377197

ABSTRACT

Cannabidiol and cannabidiol-enriched products have recently attracted much attention as an add-on therapy for epilepsy, especially drug-resistant seizures. It should be, however, remembered that concomitant use of cannabidiol and antiepileptic drugs may pose a risk of interactions between them. For this reason, the aim of our study was to assess the effect of cannabidiol on the activity of selected new antiepileptic drugs in the electrically-induced seizure models in mice. We studied the effect of cannabidiol on the anticonvulsant action of topiramate, oxcarbazepine, lamotrigine, and pregabalin in the maximal electroshock-induced seizure test as well as on the activity of levetiracetam, tiagabine, lacosamide, and gabapentin in the 6 Hz seizure test in mice. We showed that cannabidiol increased the activity of topiramate, oxcarbazepine, pregabalin, tiagabine, and gabapentin. It did not affect the anticonvulsant effect of lamotrigine and lacosamide. Interestingly, cannabidiol attenuated the anticonvulsant activity of levetiracetam. Co-administration of antiepileptic drugs with cannabidiol did not cause adverse effects such as impairment of motor coordination, changes in neuromuscular strength or potentiation of the cannabidiol-induced hypolocomotion. Serum and brain levels of antiepileptic drugs and cannabidiol were determined by using HPLC in order to ascertain any pharmacokinetic contribution to the observed behavioral effects. Only interaction with levetiracetam was purely pharmacodynamic in nature because no changes in serum and brain concentration of either levetiracetam or cannabidiol were observed. Increased anticonvulsant activity of topiramate, oxcarbazepine, pregabalin, tiagabine, and gabapentin could be, at least in part, related to pharmacokinetic interactions with cannabidiol because there were changes in serum and/or brain concentrations of antiepileptic drugs and/or cannabidiol. Pharmacokinetic interactions cannot be also excluded between lacosamide and cannabidiol because cannabidiol increased brain concentration of lacosamide and lacosamide increased brain concentration of cannabidiol. Further pharmacokinetic studies are required to evaluate the type of interactions between cannabidiol and novel antiepileptic drugs.


Subject(s)
Anticonvulsants/pharmacology , Cannabidiol/pharmacology , Seizures/drug therapy , Animals , Brain/metabolism , Chromatography, High Pressure Liquid , Disease Models, Animal , Drug Interactions , Drug Resistant Epilepsy/drug therapy , Electric Stimulation , Gabapentin/pharmacology , Lacosamide/pharmacology , Lamotrigine/pharmacology , Levetiracetam/pharmacology , Male , Mice , Oxcarbazepine/pharmacology , Pregabalin/pharmacology , Tiagabine/pharmacology , Topiramate/pharmacology
9.
Bioorg Med Chem ; 27(5): 822-831, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30718063

ABSTRACT

To discover new, potent, and selective inhibitors for the murine gamma-aminobutyric acid transporter 4 (mGAT4), the structure-activity relationship (SAR) study of a new cis-alkene analog family based on DDPM-1457 [(S)-2], which previously showed promising inhibitory potency at and subtype selectivity for mGAT4, was conducted. To uncover the importance of the differences between the trans- and the cis-alkene moiety in the spacer, the present publication describes the synthesis of the new compounds via catalytic hydrogenation with Lindlar's catalyst. The biological results collected by the SAR study revealed that analog rac-7j characterized by a four-instead of a three-carbon atom spacer with a cis double bond applying to the majority of the studied compounds displays a surprisingly high potency at mGAT1 (pIC50 = 6.00 ±â€¯0.04) and at the same time a reasonable potency at mGAT4 (pIC50 = 4.82).


Subject(s)
Alkenes/pharmacology , GABA Uptake Inhibitors/pharmacology , Nipecotic Acids/pharmacology , Alkenes/chemical synthesis , Alkenes/chemistry , Animals , Drug Design , GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/chemical synthesis , GABA Uptake Inhibitors/chemistry , HEK293 Cells , Humans , Mice , Nipecotic Acids/chemical synthesis , Nipecotic Acids/chemistry , Stereoisomerism , Structure-Activity Relationship , Tiagabine/pharmacology
10.
Hum Brain Mapp ; 40(4): 1276-1289, 2019 03.
Article in English | MEDLINE | ID: mdl-30549127

ABSTRACT

Brain responses to transcranial magnetic stimulation (TMS) recorded by electroencephalography (EEG) are emergent noninvasive markers of neuronal excitability and effective connectivity in humans. However, the underlying physiology of these TMS-evoked EEG potentials (TEPs) is still heavily underexplored, impeding a broad application of TEPs to study pathology in neuropsychiatric disorders. Here we tested the effects of a single oral dose of three antiepileptic drugs with specific modes of action (carbamazepine, a voltage-gated sodium channel (VGSC) blocker; brivaracetam, a ligand to the presynaptic vesicle protein VSA2; tiagabine, a gamma-aminobutyric acid (GABA) reuptake inhibitor) on TEP amplitudes in 15 healthy adults in a double-blinded randomized placebo-controlled crossover design. We found that carbamazepine decreased the P25 and P180 TEP components, and brivaracetam the N100 amplitude in the nonstimulated hemisphere, while tiagabine had no effect. Findings corroborate the view that the P25 represents axonal excitability of the corticospinal system, the N100 in the nonstimulated hemisphere propagated activity suppressed by inhibition of presynaptic neurotransmitter release, and the P180 late activity particularly sensitive to VGSC blockade. Pharmaco-physiological characterization of TEPs will facilitate utilization of TMS-EEG in neuropsychiatric disorders with altered excitability and/or network connectivity.


Subject(s)
Anticonvulsants/pharmacology , Cerebral Cortex/drug effects , Evoked Potentials/drug effects , Transcranial Magnetic Stimulation/drug effects , Adult , Carbamazepine/pharmacology , Cerebral Cortex/physiology , Cross-Over Studies , Double-Blind Method , Electroencephalography/drug effects , Electroencephalography/methods , Electromyography/drug effects , Electromyography/methods , Evoked Potentials/physiology , Healthy Volunteers , Humans , Male , Pyrrolidinones/pharmacology , Tiagabine/pharmacology , Transcranial Magnetic Stimulation/methods , Young Adult
11.
Nat Commun ; 9(1): 3235, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30104642

ABSTRACT

Premature infants are more likely to develop locomotor disorders than term infants. In a chronic sub-lethal hypoxia (Hx) mouse model of neonatal brain injury, we recently demonstrated the presence of cellular and physiological changes in the cerebellar white matter. We also observed Hx-induced delay in Purkinje cell (PC) arborization. However, the behavioral consequences of these cellular alterations remain unexplored. Using the Erasmus Ladder to study cerebellar behavior, we report the presence of locomotor malperformance and long-term cerebellar learning deficits in Hx mice. Optogenetics experiments in Hx mice reveal a profound reduction in spontaneous and photoevoked PC firing frequency. Finally, treatment with a gamma-aminobutyric acid (GABA) reuptake inhibitor partially rescues locomotor performance and improves PC firing. Our results demonstrate a long-term miscoordination phenotype characterized by locomotor malperformance and cerebellar learning deficits in a mouse model of neonatal brain injury. Our findings also implicate the developing GABA network as a potential therapeutic target for prematurity-related locomotor deficits.


Subject(s)
Brain Injuries/pathology , Brain Injuries/physiopathology , Cerebellum/pathology , Cerebellum/physiopathology , Learning , Purkinje Cells/pathology , Action Potentials/drug effects , Animals , Animals, Newborn , Behavior, Animal , Cerebellum/drug effects , Female , GABA Uptake Inhibitors/pharmacology , Integrases/metabolism , Learning/drug effects , Male , Mice , Motor Activity/drug effects , Purkinje Cells/drug effects , Tiagabine/pharmacology
12.
Neuroimage ; 179: 582-595, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29959047

ABSTRACT

Neurophysiological recordings are dominated by arhythmical activity whose spectra can be characterised by power-law functions, and on this basis are often referred to as reflecting scale-free brain dynamics (1/fß). Relatively little is known regarding the neural generators and temporal dynamics of this arhythmical behaviour compared to rhythmical behaviour. Here we used Irregularly Resampled AutoSpectral Analysis (IRASA) to quantify ß, in both the high (5-100 Hz, ßhf) and low frequency bands (0.1-2.5 Hz, ßlf) in MEG/EEG/ECoG recordings and to separate arhythmical from rhythmical modes of activity, such as, alpha rhythms. In MEG/EEG/ECoG data, we demonstrate that oscillatory alpha power dynamically correlates over time with ßhf and similarly, participants with higher rhythmical alpha power have higher ßhf. In a series of pharmaco-MEG investigations using the GABA reuptake inhibitor tiagabine, the glutamatergic AMPA receptor antagonist perampanel, the NMDA receptor antagonist ketamine and the mixed partial serotonergic agonist LSD, a variety of effects on both ßhf and ßlf were observed. Additionally, strong modulations of ßhf were seen in monkey ECoG data during general anaesthesia using propofol and ketamine. We develop and test a unifying model which can explain, the 1/f nature of electrophysiological spectra, their dynamic interaction with oscillatory rhythms as well as the sensitivity of 1/f activity to drug interventions by considering electrophysiological spectra as being generated by a collection of stochastically perturbed damped oscillators having a distribution of relaxation rates.


Subject(s)
Brain/drug effects , Brain/physiology , Models, Neurological , Adult , Animals , Brain Mapping/methods , Electrocorticography/drug effects , Electroencephalography/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Female , GABA Uptake Inhibitors/pharmacology , Haplorhini , Humans , Hypnotics and Sedatives/pharmacology , Ketamine/pharmacology , Lysergic Acid Diethylamide/pharmacology , Magnetoencephalography/drug effects , Male , Nitriles , Propofol/pharmacology , Pyridones/pharmacology , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin Antagonists/pharmacology , Tiagabine/pharmacology , Young Adult
13.
Photochem Photobiol ; 94(1): 173-178, 2018 01.
Article in English | MEDLINE | ID: mdl-28881432

ABSTRACT

5-aminolevulinic acid (5-ALA)-based photodynamic therapy (PDT) has been successfully used in the treatment of cancers. However, the mechanism of 5-ALA transportation into cancer cells is still not fully elucidated. Previous studies have confirmed that the efficiency of 5-ALA-PDT could be affected by the membrane skeleton protein 4.1R. In this study, we investigated the role of 4.1R in the transport of 5-ALA into cells. Wild-type (4.1R+/+ ) and 4.1R gene knockout (4.1R-/- ) mouse embryonic fibroblast (MEF) cells were incubated with 1 mm 5-ALA and different concentrations of specific inhibitors of GABA transporters GAT (1-3). Our results showed that the inhibition of GAT1 and GAT2 in particular markedly attenuated the intracellular PpIX production, reactive oxygen species (ROS) level and 5-ALA-induced photodamage. However, the inhibition of GAT3 did not show such effects. Further research showed that 4.1R-/- MEF cells had a lower expression of GAT1 and GAT2 than 4.1R+/+ MEF cells. Additionally, 4.1R directly bound to GAT1 and GAT2. Taken together, GAT1 and GAT2 transporters are involved in the uptake of 5-ALA in MEF cells. 4.1R plays an important role in transporting 5-ALA into cells via at least partly interaction with GAT1 and GAT2 transporters in 5-ALA-PDT.


Subject(s)
Aminolevulinic Acid/metabolism , GABA Plasma Membrane Transport Proteins/metabolism , Microfilament Proteins/metabolism , Photosensitizing Agents/metabolism , Analysis of Variance , Animals , Cell Line , Fibroblasts/metabolism , GABA Plasma Membrane Transport Proteins/genetics , GABA Uptake Inhibitors/pharmacology , Mice , Mice, Knockout , Microfilament Proteins/genetics , Protoporphyrins/metabolism , Reactive Oxygen Species/metabolism , Tiagabine/pharmacology
14.
Epilepsy Res ; 140: 1-7, 2018 02.
Article in English | MEDLINE | ID: mdl-29227795

ABSTRACT

Animal models are valuable tools for screening novel therapies for patients who suffer from epilepsy. However, a wide array of models are necessary to cover the diversity of human epilepsies. In humans, neonatal hypoxia (or hypoxia-ischemia) is one of the most common causes of epilepsy early in life. Hypoxia-induced seizures (HS) during the neonatal period can also lead to spontaneous seizures in adulthood. This phenomenon, i.e., early-life hypoxia leading to adult epilepsy - is also seen in experimental models, including rats. However, it is not known which anti-seizure medications are most effective at managing adult epilepsy resulting from neonatal HS. Here, we examined the efficacy of three anti-seizure medications against spontaneous seizures in adult rats with a history of neonatal HS: (1) phenobarbital (PHB), the oldest epilepsy medicine still in use today; (2) levetiracetam (LEV); and (3) tiagabine (TGB). Both LEV and TGB are relatively new anticonvulsant drugs that are ineffective in traditional seizure models, but strikingly effective in other models. We found that PHB and LEV decreased seizures in adult rats with a history of HS, whereas TGB exacerbated seizures. These divergent drug effects indicate that the HS model may be useful for differentiating the clinical efficacy of putative epilepsy therapies.


Subject(s)
Anticonvulsants/pharmacology , Hypoxia/complications , Levetiracetam/pharmacology , Seizures/drug therapy , Seizures/etiology , Tiagabine/pharmacology , Animals , Animals, Newborn , Brain/drug effects , Brain/physiopathology , Disease Models, Animal , Hypoxia/physiopathology , Male , Phenobarbital/pharmacology , Prohibitins , Rats, Long-Evans , Seizures/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...