Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Eur J Cell Biol ; 102(2): 151317, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37099936

ABSTRACT

Avulsion injury results in motoneuron death due to the increased excitotoxicity developing in the affected spinal segments. This study focused on possible short and long term molecular and receptor expression alterations which are thought to be linked to the excitotoxic events in the ventral horn with or without the anti-excitotoxic riluzole treatment. In our experimental model the left lumbar 4 and 5 (L4, 5) ventral roots of the spinal cord were avulsed. Treated animals received riluzole for 2 weeks. Riluzole is a compound that acts to block voltage-activated Na+ and Ca2+ channels. In control animals the L4, 5 ventral roots were avulsed without riluzole treatment. Expression of astrocytic EAAT-2 and that of KCC2 in motoneurons on the affected side of the L4 spinal segment were detected after the injury by confocal and dSTORM imaging, intracellular Ca2+ levels in motoneurons were quantified by electron microscopy. The KCC2 labeling in the lateral and ventrolateral parts of the L4 ventral horn was weaker compared with the medial part of L4 ventral horn in both groups. Riluzole treatment dramatically enhanced motoneuron survival but was not able to prevent the down-regulation of KCC2 expression in injured motoneurons. In contrast, riluzole successfully obviated the increase of intracellular calcium level and the decrease of EAAT-2 expression in astrocytes compared with untreated injured animals. We conclude that KCC2 may not be an essential component for survival of injured motoneurons and riluzole is able to modulate the intracellular level of calcium and expression of EAAT-2.


Subject(s)
Riluzole , Symporters , Animals , Riluzole/pharmacology , Riluzole/metabolism , Calcium/metabolism , Spinal Nerve Roots/injuries , Spinal Nerve Roots/metabolism , Spinal Cord/metabolism , Symporters/genetics , Symporters/metabolism
2.
Biol Psychiatry ; 94(5): 378-392, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36906501

ABSTRACT

BACKGROUND: Unbalanced activity of medium spiny neurons (MSNs) of the direct and indirect pathways mediates reward-related behaviors induced by addictive drugs. Prelimbic (PL) input to MSNs in the nucleus accumbens core (NAcC) plays a key role in cocaine-induced early locomotor sensitization (LS). However, the adaptive plastic changes at PL-to-NAcC synapses underlying early LS remain unclear. METHODS: Using transgenic mice and retrograde tracing, we identified NAcC-projecting pyramidal neurons (PNs) in the PL cortex based on the expression of dopamine receptor types (D1R or D2R). To examine cocaine-induced alterations in PL-to-NAcC synapses, we measured excitatory postsynaptic current amplitudes evoked by optostimulation of PL afferents to MSNs. Riluzole was chosen to test the effects of PL excitability on cocaine-induced changes of PL-to-NAcC synapses. RESULTS: NAcC-projecting PNs were segregated into D1R- and D2R-expressing PNs (D1- and D2-PNs, respectively), and their excitability was opposingly regulated by respective dopamine agonists. Both D1- and D2-PNs exhibited balanced innervation of direct MSNs and indirect MSNs in naïve animals. Repeated cocaine injections resulted in biased synaptic strength toward direct MSNs through presynaptic mechanisms in both D1- and D2-PNs, although D2R activation reduced the D2-PN excitability. Under group 1 metabotropic glutamate receptors coactivation, however, D2R activation enhanced the D2-PN excitability. The cocaine-induced rewiring accompanied LS, and both rewiring and LS were precluded by PL infusion of riluzole, which reduced the intrinsic excitability of PL neurons. CONCLUSIONS: These findings indicate that cocaine-induced rewiring of PL-to-NAcC synapses correlates well with early behavioral sensitization and that rewiring and LS can be prevented by riluzole-induced reduction of excitability of PL neurons.


Subject(s)
Cocaine , Mice , Animals , Cocaine/pharmacology , Cocaine/metabolism , Nucleus Accumbens , Riluzole/metabolism , Riluzole/pharmacology , Receptors, Dopamine D2/metabolism , Mice, Transgenic , Receptors, Dopamine D1/metabolism
3.
Mol Psychiatry ; 28(4): 1557-1570, 2023 04.
Article in English | MEDLINE | ID: mdl-36750736

ABSTRACT

Dysregulated neurite outgrowth and synapse formation underlie many psychiatric disorders, which are also manifested by wolfram syndrome (WS). Whether and how the causative gene WFS1 deficiency affects synapse formation remain elusive. By mirroring human brain development with cerebral organoids, WFS1-deficient cerebral organoids not only recapitulate the neuronal loss in WS patients, but also exhibit significantly impaired synapse formation and function associated with reduced astrocytes. WFS1 deficiency in neurons autonomously delays neuronal differentiation with altered expressions of genes associated with psychiatric disorders, and impairs neurite outgrowth and synapse formation with elevated cytosolic calcium. Intriguingly, WFS1 deficiency in astrocytes decreases the expression of glutamate transporter EAAT2 by NF-κB activation and induces excessive glutamate. When co-cultured with wildtype neurons, WFS1-deficient astrocytes lead to impaired neurite outgrowth and increased cytosolic calcium in neurons. Importantly, disrupted synapse formation and function in WFS1-deficient cerebral organoids and impaired neurite outgrowth affected by WFS1-deficient astrocytes are efficiently reversed with Riluzole treatment, by restoring EAAT2 expression in astrocytes. Furthermore, Riluzole rescues the depressive-like behavior in the forced swimming test and the impaired recognition and spatial memory in the novel object test and water maze test in Wfs1 conditional knockout mice. Altogether, our study provides novel insights into how WFS1 deficiency affects synapse formation and function, and offers a strategy to treat this disease.


Subject(s)
Human Embryonic Stem Cells , Wolfram Syndrome , Animals , Mice , Humans , Wolfram Syndrome/drug therapy , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Riluzole/pharmacology , Riluzole/metabolism , Calcium/metabolism , Human Embryonic Stem Cells/metabolism , Neurons/metabolism , Mice, Knockout , Synapses/metabolism
4.
J Healthc Eng ; 2022: 8313415, 2022.
Article in English | MEDLINE | ID: mdl-35432830

ABSTRACT

Neuropathic pain since early diabetes swamps patients' lives, and diabetes mellitus has become an increasingly worldwide epidemic. No agent, so far, can terminate the ongoing diabetes. Therefore, strategies that delay the process and the further complications are preferred, such as diabetic neuropathic pain (DNP). Dysfunction of ion channels is generally accepted as the central mechanism of diabetic associated neuropathy, of which hyperpolarization-activated cyclic nucleotide-gated 2 (HCN2) ion channel has been verified the involvement of neuropathic pain in dorsal root ganglion (DRG) neurons. Riluzole is a benzothiazole compound with neuroprotective properties on intervention to various ion channels, including hyperpolarization-activated voltage-dependent channels. To investigate the effect of riluzole within lumbar (L3-5) DRG neurons from DNP models, streptozocin (STZ, 70 mg/kg) injection was recruited subcutaneously followed by paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL), which both show significant reduction, whilst relieved by riluzole (4 mg/kg/d) administration, which was performed once daily for 7 consecutive days for 14 days. HCN2 expression was also decreased in line with alleviated behavioral tests. Our results indicate riluzole as the alleviator to STZ-induced DNP with involvement of downregulated HCN2 in lumbar DRG by continual systemic administration in rats.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Neuralgia , Animals , Diabetic Neuropathies/complications , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Ganglia, Spinal/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neuralgia/complications , Neuralgia/drug therapy , Neuralgia/metabolism , Neurons , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Potassium Channels/metabolism , Potassium Channels/pharmacology , Rats , Riluzole/metabolism , Riluzole/pharmacology , Riluzole/therapeutic use , Streptozocin/metabolism , Streptozocin/pharmacology
5.
Biochem Biophys Res Commun ; 610: 140-146, 2022 06 25.
Article in English | MEDLINE | ID: mdl-35462095

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder leading to memory loss and impaired cognition. Despite several decades of research, AD therapeutic is not available. In this study, we have investigated the impact of a chronic intervention of riluzole on memory and neurometabolism in the AßPP-PS1 mouse model of AD. The 10-month-old AßPP-PS1 mice were administered 30 doses of riluzole (6 mg/kg, intragastrically) on an alternate day for two months. The memory was assessed using Morris Water Maze, while neurometabolism was evaluated by 1H-[13C]-NMR spectroscopy together with an intravenous infusion of [1,6-13C2]glucose. The normal saline-treated AßPP-PS1 mice exhibited a decrease in learning and memory that were restored to the control level following riluzole treatment. Most interestingly, the reduced 13C labeling of GluC4 and AspC3 from [1,6-13C]glucose in the AßPP-PS1 mice was restored to the control level following riluzole intervention. As a consequence, chronic riluzole treatment improved metabolic activity of glutamatergic neurons in AßPP-PS1 mice. Together these data suggest that riluzole may be useful for improving cognition in AD.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Energy Metabolism , Glucose/metabolism , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/metabolism , Riluzole/metabolism , Riluzole/pharmacology , Riluzole/therapeutic use
6.
Sci Rep ; 11(1): 6849, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767237

ABSTRACT

This study aimed to investigate four of the eight PFN-1 mutations that are located near the actin-binding domain and determine the structural changes due to each mutant and unravel how these mutations alter protein structural behavior. Swapaa's command in UCSF chimera for generating mutations, FTMAP were employed and the data was analyzed by RMSD, RMSF graphs, Rg, hydrogen bonding analysis, and RRdisMaps utilizing Autodock4 and GROMACS. The functional changes and virtual screening, structural dynamics, and chemical bonding behavior changes, molecular docking simulation with two current FDA-approved drugs for ALS were investigated. The highest reduction and increase in Rg were found to exist in the G117V and M113T mutants, respectively. The RMSF data consistently shows changes nearby to this site. The in silico data described indicate that each of the mutations is capable of altering the structure of PFN-1 in vivo. The potential effect of riluzole and edaravone two FDA approved drugs for ALS, impacting the structural deviations and stabilization of the mutant PFN-1 is evaluated using in silico tools. Overall, the analysis of data collected reveals structural changes of mutant PFN-1 protein that may explain the neurotoxicity and the reason(s) for possible loss and gain of function of PFN-1 in the neurotoxic model of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Computer Simulation , Edaravone/metabolism , Mutant Proteins/metabolism , Mutation , Profilins/metabolism , Riluzole/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Edaravone/chemistry , Humans , Molecular Docking Simulation , Mutant Proteins/chemistry , Mutant Proteins/genetics , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Profilins/chemistry , Profilins/genetics , Protein Conformation , Riluzole/chemistry
7.
Eur J Pharm Sci ; 120: 30-39, 2018 Jul 30.
Article in English | MEDLINE | ID: mdl-29704642

ABSTRACT

Riluzole is currently one of two approved medications for the treatment of amyotrophic lateral sclerosis (ALS). However, brain disposition of riluzole, as a substrate of P-glycoprotein (P-gp), is limited by the efflux transporters at the blood-brain barrier (BBB). We propose to develop a liposomal co-delivery system that could effectively transport riluzole to brain cells by reducing efflux pumps with a P-gp inhibitor, verapamil. Riluzole and verapamil cocktail liposomes were prepared by lipid film hydration. The average particle size of cocktail liposomes was 194.3 ±â€¯6.0 nm and their polydispersity index (PDI) was 0.272 ±â€¯0.017. The encapsulation efficiencies of verapamil and riluzole in the cocktail liposomes were 86.0 ±â€¯1.4% and 85.6 ±â€¯1.1%, respectively. The drug release from cocktail liposomes after 8 h in PBS at 37 °C was 78.4 ±â€¯6.2% of riluzole and 76.7 ±â€¯3.8% of verapamil. The average particle size of liposomes did not show significant changes at 4 °C after three months. Verapamil cocktail liposomes inhibited P-gp levels measured by western blotting in dose and time-dependent manners in brain endothelial bEND.3 cells. Increased drug efflux transporters were detected in bEND.3 and astrocytes C8D1A cells, promoted by tumor necrosis factor (TNF-α) or hydrogen peroxide (H2O2). Restored accumulations of riluzole and fluorescent dye rhodamine 123 were observed in bEND.3 cells after treatments with cocktail liposomes. It indicated that inhibitory potential of co-delivery liposome system towards P-gp could mediate the transport of both P-gp substrates. Verapamil and riluzole co-loaded liposomes may be used to overcome pharmacoresistance of riluzole for improving ALS therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Amyotrophic Lateral Sclerosis/drug therapy , Astrocytes/drug effects , Brain/drug effects , Drug Resistance/drug effects , Endothelial Cells/drug effects , Neuroprotective Agents/pharmacology , Riluzole/pharmacology , Verapamil/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cell Line , Dose-Response Relationship, Drug , Drug Combinations , Drug Liberation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kinetics , Liposomes , Mice , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Particle Size , Riluzole/administration & dosage , Riluzole/metabolism , Solubility , Verapamil/administration & dosage
8.
Structure ; 26(4): 533-544.e3, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29576321

ABSTRACT

Small conductance potassium (SK) ion channels define neuronal firing rates by conducting the after-hyperpolarization current. They are key targets in developing therapies where neuronal firing rates are dysfunctional, such as in epilepsy, Parkinson's, and amyotrophic lateral sclerosis (ALS). Here, we characterize a binding pocket situated at the intracellular interface of SK2 and calmodulin, which we show to be shared by multiple small-molecule chemotypes. Crystallization of this complex revealed that riluzole (approved for ALS) and an analog of the anti-ataxic agent (4-chloro-phenyl)-[2-(3,5-dimethyl-pyrazol-1-yl)-pyrimidin-4-yl]-amine (CyPPA) bind to and allosterically modulate via this site. Solution-state nuclear magnetic resonance demonstrates that riluzole, NS309, and CyPPA analogs bind at this bipartite pocket. We demonstrate, by patch-clamp electrophysiology, that both classes of ligand interact with overlapping but distinct residues within this pocket. These data define a clinically important site, laying the foundations for further studies of the mechanism of action of riluzole and related molecules.


Subject(s)
Calmodulin/chemistry , Indoles/chemistry , Oximes/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Riluzole/chemistry , Small-Conductance Calcium-Activated Potassium Channels/chemistry , Allosteric Regulation , Amino Acid Motifs , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Binding Sites , Calmodulin/genetics , Calmodulin/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Indoles/metabolism , Models, Molecular , Oximes/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Pyrazoles/metabolism , Pyrimidines/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Riluzole/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism
9.
Transl Psychiatry ; 7(5): e1137, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28534874

ABSTRACT

Currently, there are no effective pharmacologic treatments for the core symptoms of autism spectrum disorder (ASD). There is, nevertheless, potential for progress. For example, recent evidence suggests that the excitatory (E) glutamate and inhibitory (I) GABA systems may be altered in ASD. However, no prior studies of ASD have examined the 'responsivity' of the E-I system to pharmacologic challenge; or whether E-I modulation alters abnormalities in functional connectivity of brain regions implicated in the disorder. Therefore, we used magnetic resonance spectroscopy ([1H]MRS) to measure prefrontal E-I flux in response to the glutamate and GABA acting drug riluzole in adult men with and without ASD. We compared the change in prefrontal 'Inhibitory Index'-the GABA fraction within the pool of glutamate plus GABA metabolites-post riluzole challenge; and the impact of riluzole on differences in resting-state functional connectivity. Despite no baseline differences in E-I balance, there was a significant group difference in response to pharmacologic challenge. Riluzole increased the prefrontal cortex inhibitory index in ASD but decreased it in controls. There was also a significant group difference in prefrontal functional connectivity at baseline, which was abolished by riluzole within the ASD group. Our results also show, for we believe the first time in ASD, that E-I flux can be 'shifted' with a pharmacologic challenge, but that responsivity is significantly different from controls. Further, our initial evidence suggests that abnormalities in functional connectivity can be 'normalised' by targeting E-I, even in adults.


Subject(s)
Autism Spectrum Disorder/physiopathology , Brain/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Prefrontal Cortex/physiopathology , Riluzole/pharmacology , Adult , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/drug therapy , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Brain Mapping/methods , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Functional Neuroimaging/methods , Glutamic Acid/metabolism , Glutamic Acid/physiology , Humans , Magnetic Resonance Spectroscopy/methods , Male , Neural Pathways/physiopathology , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Riluzole/administration & dosage , Riluzole/metabolism , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
10.
Mol Psychiatry ; 22(2): 296-305, 2017 02.
Article in English | MEDLINE | ID: mdl-27021815

ABSTRACT

Alzheimer's disease (AD) and age-related cognitive decline represent a growing health burden and involve the hippocampus, a vulnerable brain region implicated in learning and memory. To understand the molecular effects of aging on the hippocampus, this study characterized the gene expression changes associated with aging in rodents using RNA-sequencing (RNA-seq). The glutamate modulator, riluzole, which was recently shown to improve memory performance in aged rats, prevented many of the hippocampal age-related gene expression changes. A comparison of the effects of riluzole in rats against human AD data sets revealed that many of the gene changes in AD are reversed by riluzole. Expression changes identified by RNA-Seq were validated by qRT-PCR open arrays. Riluzole is known to increase the glutamate transporter EAAT2's ability to scavenge excess glutamate, regulating synaptic transmission. RNA-seq and immunohistochemistry confirmed an increase in EAAT2 expression in hippocampus, identifying a possible mechanism underlying the improved memory function after riluzole treatment.


Subject(s)
Cognition/drug effects , Excitatory Amino Acid Transporter 2/drug effects , Riluzole/therapeutic use , Age Factors , Aging/genetics , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cognitive Aging/physiology , Disease Models, Animal , Glutamic Acid/metabolism , Hippocampus/metabolism , Male , Memory/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Riluzole/metabolism , Synaptic Transmission/physiology , Transcriptome/genetics
11.
Toxicol Appl Pharmacol ; 315: 80-89, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27939241

ABSTRACT

Loss of pericytes, considered an early hallmark of diabetic retinopathy, is thought to involve abnormal activation of protein kinase C (PKC). We previously showed that the anti-amyotrophic lateral sclerosis (ALS) drug riluzole functions as a PKC inhibitor. Here, we examined the effects of riluzole on pathological changes in diabetic retinopathy. Pathological endpoints examined in vivo included the number of pericytes and integrity of retinal vessels in streptozotocin (STZ)-induced diabetic mice. In addition, PKC activation and the induction of monocyte chemotactic protein (MCP1) were assessed in diabetic mice and in human retinal pericytes exposed to advanced glycation end product (AGE) or modified low-density lipoprotein (mLDL). The diameter of retinal vessels and the number of pericytes were severely reduced, and the levels of MCP1 and PKC were increased in STZ-induced diabetic mice. Administration of riluzole reversed all of these changes. Furthermore, the increased expression of MCP1 in AGE- or mLDL-treated cultured retinal pericytes was inhibited by treatment with riluzole or the PKC inhibitor GF109203X. In silico modeling showed that riluzole fits well within the catalytic pocket of PKC. Taken together, our results demonstrate that riluzole attenuates both MCP1 induction and pericyte loss in diabetic retinopathy, likely through its direct inhibitory effect on PKC.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Diabetic Retinopathy/drug therapy , Pericytes/drug effects , Riluzole/pharmacology , Animals , Diabetic Retinopathy/pathology , Male , Mice , Mice, Inbred C57BL , Protein Kinase C beta/metabolism , Riluzole/metabolism , Riluzole/therapeutic use , Streptozocin
12.
J Pharm Biomed Anal ; 117: 453-63, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26454338

ABSTRACT

Cyclodextrin-related host-guest encapsulation is fundamental to modulate the solubility of riluzole (RLZ), promoting its potential pharmaceutical applications. The supramolecular interaction of RLZ and hydroxypropyl-ß-cyclodextrin (HP-ß-CD) was examined through FT-IR spectroscopy, DSC-TGA, PXRD, (1)HNMR, 2D ROESY, ssNMR, and SEM. The HP-ß-CD/RLZ inclusion complex was formed at a molar ratio of 1:1. The stability constant (K=2327 M(-1)) and the corresponding thermodynamic parameters were ascertained through phase solubility studies. The water solubility and dissolution rate of RLZ notably increased in the presence of HP-ß-CD, whereas the inclusion complex did not increase the RLZ toxicity toward the LO2 cell line. The influence of HP-ß-CD on RLZ-human serum albumin (HSA) binding was investigated via fluorescence spectroscopy. Fluorescence quenching of HSA by RLZ in the presence and absence of HP-ß-CD were both static quenching. Data analysis showed that the addition of HP-ß-CD weakened the quenching and binding of RLZ with HSA but did not affect the binding site and binding force between RLZ and HSA. Furthermore, molecular models were generated to determine the binding site between HSA and RLZ, and these models were consistent with the experimental data.


Subject(s)
Cytotoxins/metabolism , Models, Molecular , Riluzole/metabolism , Serum Albumin/metabolism , beta-Cyclodextrins/metabolism , 2-Hydroxypropyl-beta-cyclodextrin , Cytotoxins/chemistry , Humans , Protein Binding/physiology , Riluzole/chemistry , Serum Albumin/chemistry , Solubility , beta-Cyclodextrins/chemistry
13.
J Theor Biol ; 315: 53-63, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-22995823

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a common neurodegenerative disorder characterized mainly by a progressive loss of motor neurons. Glutamate excitotoxicity is likely the main cause of neuronal death, and Riluzole interferes with glutamate-mediated transmission. Thus, in such independent pathway, these effects may be partly due to inactivation of voltage-dependent sodium channels. Here we predict the structural model of the interaction and report the possible binding sites of Riluzole on Nav1.6 channel. The docked complexes were subjected to minimization and we further investigated the key interacting residues, binding free energies, pairing bridge determination, folding pattern, hydrogen bounding formation, hydrophobic contacts and flexibilities. Our results demonstrate that Riluzole interacts with the Nav1.6 channel, more specifically in the key residues TYR 1787, LEU 1843 and GLN 1799, suggesting possible cellular implications driven by these amino acids on Riluzole-Nav1.6 interaction, which may serve as an important output for a more specific and experimental drug design therapy against ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Computational Biology/methods , Molecular Docking Simulation/methods , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Riluzole/metabolism , Riluzole/therapeutic use , Amino Acid Sequence , Binding Sites , Cell Membrane/metabolism , Humans , Molecular Sequence Data , NAV1.6 Voltage-Gated Sodium Channel/chemistry , Protein Structure, Secondary , Protein Subunits/chemistry , Reproducibility of Results , Riluzole/chemistry , Sequence Alignment , Software , Solvents , Structural Homology, Protein
14.
Bioorg Med Chem ; 20(18): 5642-8, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22892214

ABSTRACT

Riluzole (1) is an approved therapeutic for the treatment of ALS and has also demonstrated anti-melanoma activity in metabotropic glutamate GRM1 positive cell lines, a mouse xenograft assay and human clinical trials. Highly variable drug exposure following oral administration among patients, likely due to variable first pass effects from heterogeneous CYP1A2 expression, hinders its clinical use. In an effort to mitigate effects of this clearance pathway and uniformly administer riluzole at efficacious exposure levels, several classes of prodrugs of riluzole were designed, synthesized, and evaluated in multiple in vitro stability assays to predict in vivo drug levels. The optimal prodrug would possess the following profile: stability while transiting the digestive system, stability towards first pass metabolism, and metabolic lability in the plasma releasing riluzole. (S)-O-Benzyl serine derivative 9 was identified as the most promising therapeutically acceptable prodrug.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Drug Design , Melanoma/drug therapy , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Riluzole/metabolism , Riluzole/pharmacology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1A2/metabolism , Drug Stability , Humans , Melanoma/metabolism , Mice , Microsomes, Liver/metabolism , Molecular Structure , Prodrugs/chemistry , Prodrugs/metabolism , Riluzole/blood , Riluzole/chemical synthesis
15.
Eur J Pharmacol ; 677(1-3): 87-92, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22206816

ABSTRACT

We have recently demonstrated that the glutamate transporter activator riluzole paradoxically enhanced glutamate-induced glutamate release from cultured astrocytes. We further showed that both riluzole and the α(2)δ subunit ligand gabapentin activated descending inhibition in rats by increasing glutamate receptor signaling in the locus coeruleus and hypothesized that these drugs share common mechanisms to enhance glutamate release from astrocytes. In the present study, we examined the effects of riluzole and gabapentin on glutamate uptake and release and glutamate-induced Ca(2+) responses in primary cultures of astrocytes. Riluzole and gabapentin facilitated glutamate-induced glutamate release from astrocytes and significantly increased glutamate uptake, the latter being completely blocked by the non-selective glutamate transporter blocker DL-threo-ß-benzyloxyaspartic acid (DL-TBOA). Riluzole and gabapentin also enhanced the glutamate-induced increase in intracellular Ca(2+) concentrations. Some α(2)δ subunit ligands, pregabalin and L-isoleucine, enhanced the glutamate-induced Ca(2+) response, whereas another, 3-exo-aminobicyclo[2.2.1]heptane-2-exo-carboxylic acid (ABHCA), did not. The enhancement of glutamate-induced intracellular Ca(2+) response by riluzole and gabapentin was blocked by the DL-TBOA and an inhibitor of Na(+)/Ca(2+) exchange, 2-[2-[4-(4-nitrobenzyloxy)phenyl]ethyl]isothiurea (KB-R7943). Gabapentin's enhancement of Ca(2+) increase was specific to glutamate stimulation, as it was not mimicked with stimulation by ATP. These results suggest that riluzole and gabapentin enhance Na(+)-glutamate co-transport through glutamate transporters, induce subsequent Ca(2+) influx via the reverse mode of Na(+)/Ca(2+) exchange, and thereby facilitate Ca(2+)-dependent glutamate release by glutamate in astrocytes. The present study also demonstrates a novel target of gabapentinoid action in astrocytes other than α(2)δ subunits in neurons.


Subject(s)
Amines/pharmacology , Amino Acid Transport System X-AG/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Riluzole/pharmacology , gamma-Aminobutyric Acid/pharmacology , Amines/metabolism , Animals , Astrocytes/cytology , Biological Transport/drug effects , Calcium/metabolism , Cells, Cultured , Cyclohexanecarboxylic Acids/metabolism , Gabapentin , Intracellular Space/drug effects , Intracellular Space/metabolism , Ligands , Protein Subunits/metabolism , Rats , Riluzole/metabolism , Sodium/metabolism , gamma-Aminobutyric Acid/metabolism
16.
Acta Physiol (Oxf) ; 202(2): 185-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21306568

ABSTRACT

AIM: Earlier studies have shown that TREK-1 and TREK-2 (TREKs), members of the two-pore domain K(+) (K(2P)) channel family that are highly expressed under pathological conditions, are activated by neuroprotective agents. Baicalein and wogonin, oriental flavonoids originating from the root of the medicinal herb Scutellaria baicalensis, are known to have beneficial effects for neuroprotection. However, little is known about the effects of baicalein and wogonin on ion channels including TREKs. We investigated whether baicalein and wogonin modulate the TREK-2 channel, which has been less studied than TREK-1. METHODS: Single-channel recordings were performed in COS-7 cells transfected with rat TREK-2 and analyzed baicalein- or wogonin-induced channel activity. RESULTS: We found that baicalein and wogonin activated the TREK-2 current by increasing the opening frequency (channel activity: from 0.05 ± 0.01 to 0.17 ± 0.06 in baicalein treatment and from 0.03 ± 0.01 to 0.29 ± 0.09 in wogonin treatment, P < 0.05), while leaving the single-channel conductance and mean open time unchanged. Baicalein continuously activated TREK-2, whereas wogonin transiently activated TREK-2. Application of baicalein and wogonin activated TREK-2 in both cell attached and excised patches, suggesting that baicalein and wogonin may modulate TREK-2 either directly or indirectly with different mechanisms. CONCLUSION: These results suggest that baicalein- and wogonin-induced TREK-2 activation help set the resting membrane potential of cells exposed to pathological conditions and thus may give beneficial effects in neuroprotection.


Subject(s)
Flavanones/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Flavanones/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/genetics , Rats , Riluzole/metabolism , Riluzole/pharmacology
17.
Neurosci Lett ; 472(3): 166-70, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20138122

ABSTRACT

The efflux pumps located at the blood-brain barrier (BBB) prevent drugs entering the brain. As such, efflux pumps are a major obstacle to drug brain distribution. Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with little therapeutics available: riluzole is the only drug approved in its treatment. The lack of response to treatment in ALS may be, at least in part, due to increased activities of efflux pumps in relation to disease, leading to subtherapeutic brain concentrations of drugs. In the present study, we used a transgenic mouse model of ALS (G86R mSOD1 mice) to test this hypothesis. Expression and functionality of P-glycoprotein (ABCB1, P-gp) and Breast Cancer Resistance Protein (ABCG2, BCRP), two major efflux pumps, were studied. We observed an increased P-gp expression (1.5-fold) in presymptomatic mSOD1 mice compared to wild-type controls. Consistent with this, P-gp function was also increased by 1.5-fold and riluzole brain disposition was decreased by 1.7-fold in mSOD1 mice. Contrasting with this, BCRP expression and function were unaltered by the pathology. These results demonstrate that BBB transport proteins are modified in G86R mSOD1 mice ALS model. Such findings underline potential problems in extrapolating the results of animal studies to humans and developing clinical trials, especially for drugs transported by P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Amyotrophic Lateral Sclerosis/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Disease Models, Animal , Mice , Mice, Transgenic , Mutation , Neuroprotective Agents/metabolism , Riluzole/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1
18.
Brain Res ; 1289: 106-17, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19615351

ABSTRACT

The mechanisms underlying the disruption of glutamate-glutamine cycle (Glu-Gln cycle) in manganism are still unknown. To approach the concrete mechanisms, the rats were i.p. injected with different doses of MnCl(2) (0, 8, 40, and 200 micromol/kg), and the levels of Mn, Glu, and Gln, the morphological and ultrastructural changes, activities of Na(+)-K(+)-ATPase, GS, and PAG, mRNA and protein expression of GS, GLAST, and GLT-1 in the striatum were investigated. In addition, the effect of 21.35 micromol/kg riluzole (Na(+) channel blocker) was studied at 200 micromol/kg MnCl(2). It was observed that (1) Mn and Glu levels and PAG activity increased; (2) many pathological changes occurred; (3) Gln levels, Na(+)-K(+)-ATPase and GS activities, and GS, GLAST, and GLT-1 mRNA and protein expression inhibited, does dependently. Furthermore, the research indicated that pretreatment of riluzole reversed toxic effects of MnCl(2) significantly. These results suggested that Glu-Gln cycle was disrupted by Mn exposure dose dependently; riluzole might antagonize Mn neurotoxicity.


Subject(s)
Chlorides/toxicity , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Riluzole/pharmacology , Animals , Blotting, Western , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Female , Glutamate-Ammonia Ligase/metabolism , Glutaminase/metabolism , Male , Manganese/metabolism , Manganese Compounds , Manganese Poisoning/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Riluzole/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
19.
Neurosci Lett ; 452(1): 12-6, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19146924

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative fatal disease. Drugs used in this disease need to cross the blood-brain barrier (BBB). Only riluzole is approved for ALS treatment. We have investigated riluzole as a breast cancer resistance protein (BCRP) substrate by studying its brain transport in CF1 mdr1a (-/-) mice and its intracellular uptake on BeWo cells (human placental choriocarcinoma cell line). We have also investigated the effect of riluzole on BCRP expression level and on its activity using the prazocin as a test probe for brain transport and intracellular uptake. Assays on mdr1a (-/-) mice and BeWo cells showed a higher uptake of riluzole when pretreated with a BCRP inhibitor. After repeated doses of riluzole, BCRP activity was increased in CF1 mdr1a (-/-) mice, riluzole uptake was decrease and both BCRP expression and activity were increased in BeWo cells. In conclusion, we report in this study that riluzole is transported by BCRP at the BBB level and can enhance its function. These results taken with our previous studies on riluzole and P-glycoprotein show that drug-drug interactions between riluzole and efflux transporters substrates may occur at the BBB level and should be taken into account in future clinical trial design in ALS.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Neoplasm Proteins/metabolism , Neuroprotective Agents/metabolism , Riluzole/metabolism , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport/drug effects , Biological Transport/genetics , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Choriocarcinoma , Female , Gene Expression/drug effects , Humans , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neuroprotective Agents/pharmacology , Prazosin/metabolism , RNA, Messenger/metabolism , Riluzole/pharmacology , Transfection/methods
20.
Chem Res Toxicol ; 21(3): 711-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18298092

ABSTRACT

Trovafloxacin (Trovan) is a fluoroquinolone antibiotic drug with a long half-life and broad-spectrum activity. Since its entry into the market in 1998, trovafloxacin has been associated with numerous cases of hepatotoxicity, which has limited its clinical usefulness. Trovafloxacin possesses two substructural elements that have the potential to generate reactive intermediates: a cyclopropylamine moiety and a difluoroanilino system. The results presented here describe the in vitro metabolic activation of a synthetic drug model (DM) of trovafloxacin that contains the cyclopropylamine moiety. Cyclopropylamine can be oxidized to reactive ring-opened products-a carbon-centered radical and a subsequently oxidized alpha,beta-unsaturated aldehyde. Experiments with monoamine oxygenases, horseradish peroxidase, flavin monooxygenase 3, and cDNA-expressed P450 isoenzymes revealed that P450 1A2 oxidizes DM to a reactive alpha,beta-unsaturated aldehyde, M 1. Furthermore, myeloperoxidase (MPO) was also demonstrated to oxidize DM in the presence of chloride ion to produce M 1. DM proved to be a suicide inhibitor of MPO while showing no inhibition of P450 1A2. The structure of the reactive metabolite was confirmed by LC-MS/MS analysis by comparison with a synthetic standard. M 1 was further shown to react with glutathione and the related thiol nucleophile, 4-bromobenzyl mercaptan, suggesting the potential of this intermediate to react with protein nucleophiles. In summary, these data provide evidence that trovafloxacin-induced hepatotoxicity may be mediated through the oxidation of the cyclopropylamine substructure to reactive intermediates that may form covalent adducts to hepatic proteins, resulting in damage to liver tissue.


Subject(s)
Anti-Bacterial Agents/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Cross-Linking Reagents/metabolism , Cyclopropanes/metabolism , Fluoroquinolones/toxicity , Naphthyridines/toxicity , Aldehydes/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 Enzyme System/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , In Vitro Techniques , Liver/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Oxidation-Reduction , Peroxidase/metabolism , Procainamide/pharmacology , Riluzole/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...