Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Front Physiol ; 14: 1081186, 2023.
Article En | MEDLINE | ID: mdl-36891145

Introduction: Cannabis contains cannabidiol (CBD), the main non-psychoactive phytocannabinoid, but also many other phytocannabinoids that have therapeutic potential in the treatment of epilepsy. Indeed, the phytocannabinoids cannabigerolic acid (CBGA), cannabidivarinic acid (CBDVA), cannabichromenic acid (CBCA) and cannabichromene (CBC) have recently been shown to have anti-convulsant effects in a mouse model of Dravet syndrome (DS), an intractable form of epilepsy. Recent studies demonstrate that CBD inhibits voltage-gated sodium channel function, however, whether these other anti-convulsant phytocannabinoids affect these classic epilepsy drug-targets is unknown. Voltage-gated sodium (NaV) channels play a pivotal role in initiation and propagation of the neuronal action potential and NaV1.1, NaV1.2, NaV1.6 and NaV1.7 are associated with the intractable epilepsies and pain conditions. Methods: In this study, using automated-planar patch-clamp technology, we assessed the profile of the phytocannabinoids CBGA, CBDVA, cannabigerol (CBG), CBCA and CBC against these human voltage-gated sodium channels subtypes expressed in mammalian cells and compared the effects to CBD. Results: CBD and CBGA inhibited peak current amplitude in the low micromolar range in a concentration-dependent manner, while CBG, CBCA and CBC revealed only modest inhibition for this subset of sodium channels. CBDVA inhibited NaV1.6 peak currents in the low micromolar range in a concentration-dependent fashion, while only exhibiting modest inhibitory effects on NaV1.1, NaV1.2, and NaV1.7 channels. CBD and CBGA non-selectively inhibited all channel subtypes examined, whereas CBDVA was selective for NaV1.6. In addition, to better understand the mechanism of this inhibition, we examined the biophysical properties of these channels in the presence of each cannabinoid. CBD reduced NaV1.1 and NaV1.7 channel availability by modulating the voltage-dependence of steady-state fast inactivation (SSFI, V0.5 inact), and for NaV1.7 channel conductance was reduced. CBGA also reduced NaV1.1 and NaV1.7 channel availability by shifting the voltage-dependence of activation (V0.5 act) to a more depolarized potential, and for NaV1.7 SSFI was shifted to a more hyperpolarized potential. CBDVA reduced channel availability by modifying conductance, SSFI and recovery from SSFI for all four channels, except for NaV1.2, where V0.5 inact was unaffected. Discussion: Collectively, these data advance our understanding of the molecular actions of lesser studied phytocannabinoids on voltage-gated sodium channel proteins.

2.
J Cannabis Res ; 4(1): 30, 2022 Jun 10.
Article En | MEDLINE | ID: mdl-35689251

BACKGROUND: Purified cannabidiol (CBD), a non-psychoactive phytocannabinoid, has gained regulatory approval to treat intractable childhood epilepsies. Despite this, artisanal and commercial CBD-dominant hemp-based products continue to be used by epilepsy patients. Notably, the CBD doses used in these latter products are much lower than that found to be effective in reducing seizures in clinical trials with purified CBD. This might be because these CBD-dominant hemp products contain other bioactive compounds, including phytocannabinoids and terpenes, which may exert unique effects on epilepsy-relevant drug targets. Voltage-gated sodium (NaV) channels are vital for initiation of neuronal action potential propagation and genetic mutations in these channels result in epilepsy phenotypes. Recent studies suggest that NaV channels are inhibited by purified CBD. However, the effect of cannabis-based products on the function of NaV channels is unknown. METHODS: Using automated-planar patch-clamp technology, we profile a hemp-derived nutraceutical product (NP) against human NaV1.1-NaV1.8 expressed in mammalian cells to examine effects on the biophysical properties of channel conductance, steady-state fast inactivation and recovery from fast inactivation. RESULTS: NP modifies peak current amplitude of the NaV1.1-NaV1.7 subtypes and has variable effects on the biophysical properties for all channel subtypes tested. NP potently inhibits NaV channels revealing half-maximal inhibitory concentration (IC50) values of between 1.6 and 4.2 µg NP/mL. Purified CBD inhibits NaV1.1, NaV1.2, NaV1.6 and NaV1.7 to reveal IC50 values in the micromolar range. The CBD content of the product equates to IC50 values (93-245 nM), which are at least an order of magnitude lower than purified CBD. Unlike NP, hemp seed oil vehicle alone did not inhibit NaV channels, suggesting that the inhibitory effects of NP are independent of hemp seed oil. CONCLUSIONS: This CBD-dominant NP potently inhibits NaV channels. Future study of the individual elements of NP, including phytocannabinoids and terpenes, may reveal a potent individual component or that its components interact to modulate NaV channels.

3.
Methods Mol Biol ; 2188: 133-155, 2021.
Article En | MEDLINE | ID: mdl-33119850

Genetic mutations have long been implicated in epilepsy, particularly in genes that encode ion channels and neurotransmitter receptors. Among some of those identified are voltage-gated sodium, potassium and calcium channels, and ligand-gated gamma-aminobutyric acid (GABA), neuronal nicotinic acetylcholine (CHRN), and glutamate receptors, making them key therapeutic targets. In this chapter we discuss the use of automated electrophysiological technologies to examine the impact of gene defects in two potassium channels associated with different epilepsy syndromes. The hKCNC1 gene encodes the voltage-gated potassium channel hKV3.1, and mutations in this gene cause progressive myoclonus epilepsy (PME) and ataxia due to a potassium channel mutation (MEAK). The hKCNT1 gene encodes the weakly voltage-dependent sodium-activated potassium channel hKCNT1, and mutations in this gene cause a wide spectrum of seizure disorders, including severe autosomal dominant sleep-related hypermotor epilepsy (ADSHE) and epilepsy of infancy with migrating focal seizures (EIMFS), both conditions associated with drug-resistance. Importantly, both of these potassium channels play vital roles in regulating neuronal excitability. Since its discovery in the late nineteen seventies, the patch-clamp technique has been regarded as the bench-mark technology for exploring ion channel characteristics. In more recent times, innovations in automated patch-clamp technologies, of which there are many, are enabling the study of ion channels with much greater productivity that manual systems are capable of. Here we describe aspects of Nanion NPC-16 Patchliner, examining the effects of temperature on stably and transiently transfected mammalian cells, the latter of which for most automated systems on the market is quite challenging. Remarkable breakthroughs in the development of other automated electrophysiological technologies, such as multielectrode arrays that support extracellular signal recordings, provide additional features to examine network activity in the area of ion channel research, particularly epilepsy. Both of these automated technologies enable the acquisition of consistent, robust, and reproducible data. Numerous systems have been developed with very similar capabilities, however, not all the systems on the market are adapted to work with primary cells, particularly neurons that can be problematic. This chapter also showcases methods that demonstrate the versatility of Nanion NPC-16 Patchliner and the Multi Channel Systems (MCS) multielectrode array (MEA) assay for acutely dissociated murine primary cortical neurons, enabling the study of potassium channel mutations implicated in severe refractory epilepsies.


Epilepsy/pathology , Neurons/pathology , Patch-Clamp Techniques/methods , Animals , Cells, Cultured , Electrophysiological Phenomena , Epilepsy/genetics , Epilepsy/metabolism , Equipment Design , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Patch-Clamp Techniques/instrumentation , Potassium Channels, Sodium-Activated/genetics , Potassium Channels, Sodium-Activated/metabolism , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Transfection/instrumentation , Transfection/methods
4.
FASEB J ; 34(3): 3884-3901, 2020 03.
Article En | MEDLINE | ID: mdl-32003498

P2X7 is an ATP-gated membrane ion channel that is expressed by multiple cell types. Brief exposure to ATP induces the opening of a nonselective cation channel; while repeated or prolonged exposure induces formation of a transmembrane pore. This process may be partially regulated by alternative splicing of full-length P2RX7A pre-mRNA, producing isoforms that delete or retain functional domains. Here, we report cloning and expression of a novel P2RX7 splice variant, P2RX7L, that is, characterized by skipping of exons 7 and 8. In HEK 293 cells, expression of P2RX7L produces a protein isoform, P2X7L, that forms a heteromer with P2X7A. A haplotype defined by six single nucleotide polymorphisms (SNPs) (rs208307, rs208306, rs36144485, rs208308, rs208309, and rs373655596) promotes allele-specific alternative splicing, increasing mRNA levels of P2RX7L and another isoform, P2RX7E, which in addition has a truncated C-terminus. Skipping of exons 7 and 8 is predicted to delete critical amino acids in the ATP-binding site. P2X7L-transfected HEK 293 cells have phagocytic but not channel, pore, or membrane-blebbing function, and double-transfected P2X7L and P2X7A cells have reduced pore function. Heteromeric receptor complexes of P2X7A and P2X7L are predicted to have reduced numbers of ATP-binding sites, which potentially alters receptor function compared to homomeric P2X7A complexes.


Exons/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Purinergic P2X7/genetics , Adult , Aged , Binding Sites/genetics , Blotting, Western , Cells, Cultured , Electrophysiology , Female , HEK293 Cells , Haplotypes/genetics , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
5.
Methods Mol Biol ; 2041: 285-300, 2020.
Article En | MEDLINE | ID: mdl-31646497

P2X receptors are a structurally and functionally distinctive family of ligand-gated ion channels that play important roles in mediating extracellular adenosine 5'-triphosphate (ATP) signaling in diverse physiological and pathophysiological processes. For several decades, the "manual" patch-clamp technique was regarded as the gold standard assay for investigating ion channel properties. More recently, breakthroughs in the development of automated patch-clamp technologies are enabling the study of ion channels, with much greater throughput capacities. These automated platforms, of which there are many, generate consistent, reliable, high-fidelity data. This chapter demonstrates the versatility of one of these technologies for ligand-gated ion channels, with a particular emphasis on protocols that address some of the issues of receptor desensitization that are commonly associated with P2X receptor-mediated currents.


Adenosine Triphosphate/metabolism , Ion Channel Gating/physiology , Ion Channels/physiology , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Receptors, Purinergic P2X/metabolism , Automation , HEK293 Cells , Humans , Membrane Potentials , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 115(34): E8077-E8085, 2018 08 21.
Article En | MEDLINE | ID: mdl-30076230

Dravet syndrome is a catastrophic, pharmacoresistant epileptic encephalopathy. Disease onset occurs in the first year of life, followed by developmental delay with cognitive and behavioral dysfunction and substantially elevated risk of premature death. The majority of affected individuals harbor a loss-of-function mutation in one allele of SCN1A, which encodes the voltage-gated sodium channel NaV1.1. Brain NaV1.1 is primarily localized to fast-spiking inhibitory interneurons; thus the mechanism of epileptogenesis in Dravet syndrome is hypothesized to be reduced inhibitory neurotransmission leading to brain hyperexcitability. We show that selective activation of NaV1.1 by venom peptide Hm1a restores the function of inhibitory interneurons from Dravet syndrome mice without affecting the firing of excitatory neurons. Intracerebroventricular infusion of Hm1a rescues Dravet syndrome mice from seizures and premature death. This precision medicine approach, which specifically targets the molecular deficit in Dravet syndrome, presents an opportunity for treatment of this intractable epilepsy.


Epilepsies, Myoclonic/drug therapy , Interneurons/metabolism , Mutation , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Spider Venoms/pharmacology , Synaptic Transmission/drug effects , Animals , CHO Cells , Cricetulus , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/pathology , HEK293 Cells , Humans , Interneurons/pathology , Mice , Mice, Mutant Strains , NAV1.1 Voltage-Gated Sodium Channel/genetics
7.
Pharmacol Res Perspect ; 5(4): e00319, 2017 08.
Article En | MEDLINE | ID: mdl-28713569

The molecular composition and functional diversity of native GABAB receptors (GABABR) are still poorly understood, thus hindering development of selective GABABR ligands. Potassium channel tetramerization domain-containing protein (KCTD) 12 is a GABABR auxiliary subunit and mouse KCTD12 can alter GABABR function. In this study, we sought to characterize the effects of human KCTD12 on GABABR kinetics and pharmacology, using an automated electrophysiological assay. Seizure susceptibility and ethanol consumption were also investigated in a KCTD12 knockout mouse model. Human KCTD12 co-expression altered the kinetics of GABABR-mediated GIRK channels, speeding rates of both activation and desensitization. Analysis of concentration-response curves showed that KCTD12 coexpression did not alter effects of the agonists GABA or baclofen on GABABR. KCTD12 coexpression enhanced the potentiating effects of the positive allosteric modulator CGP7930, and its effects on GABABR activation and desensitization. The function of KCTD12 in vivo was examined, using the KCTD12 knockout mouse model. The knockout mice were more resistant to a pentylenetetrazole proconvulsant challenge suggesting reduced seizure susceptibility. In the two bottle preference test, KCTD12 knockout mice demonstrated a reduced consumption at high ethanol concentrations. In summary, human KCTD12 accelerated the kinetics of GABABR in vitro, in a manner possibly sensitive to allosteric pharmacological modulation. This study also provides novel in vivo evidence that the interaction between KCTD12 and GABABR is of physiological significance, and may be a mechanism to more selectively modulate GABABR.

8.
Ann Neurol ; 81(5): 677-689, 2017 May.
Article En | MEDLINE | ID: mdl-28380698

OBJECTIVE: To comprehensively describe the new syndrome of myoclonus epilepsy and ataxia due to potassium channel mutation (MEAK), including cellular electrophysiological characterization of observed clinical improvement with fever. METHODS: We analyzed clinical, electroclinical, and neuroimaging data for 20 patients with MEAK due to recurrent KCNC1 p.R320H mutation. In vitro electrophysiological studies were conducted using whole cell patch-clamp to explore biophysical properties of wild-type and mutant KV 3.1 channels. RESULTS: Symptoms began at between 3 and 15 years of age (median = 9.5), with progressively severe myoclonus and rare tonic-clonic seizures. Ataxia was present early, but quickly became overshadowed by myoclonus; 10 patients were wheelchair-bound by their late teenage years. Mild cognitive decline occurred in half. Early death was not observed. Electroencephalogram (EEG) showed generalized spike and polyspike wave discharges, with documented photosensitivity in most. Polygraphic EEG-electromyographic studies demonstrated a cortical origin for myoclonus and striking coactivation of agonist and antagonist muscles. Magnetic resonance imaging revealed symmetrical cerebellar atrophy, which appeared progressive, and a prominent corpus callosum. Unexpectedly, transient clinical improvement with fever was noted in 6 patients. To explore this, we performed high-temperature in vitro recordings. At elevated temperatures, there was a robust leftward shift in activation of wild-type KV 3.1, increasing channel availability. INTERPRETATION: MEAK has a relatively homogeneous presentation, resembling Unverricht-Lundborg disease, despite the genetic and biological basis being quite different. A remarkable improvement with fever may be explained by the temperature-dependent leftward shift in activation of wild-type KV 3.1 subunit-containing channels, which would counter the loss of function observed for mutant channels, highlighting KCNC1 as a potential target for precision therapeutics. Ann Neurol 2017;81:677-689.


Ataxia , Cognitive Dysfunction/etiology , Epilepsies, Myoclonic , Hot Temperature , Shaw Potassium Channels/metabolism , Adolescent , Adult , Age of Onset , Ataxia/complications , Ataxia/diagnostic imaging , Ataxia/genetics , Ataxia/physiopathology , Electroencephalography , Epilepsies, Myoclonic/complications , Epilepsies, Myoclonic/diagnostic imaging , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/physiopathology , Female , HEK293 Cells , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Mutation , Pedigree , Shaw Potassium Channels/genetics , Syndrome , Young Adult
9.
Hum Mutat ; 38(6): 736-744, 2017 06.
Article En | MEDLINE | ID: mdl-28326637

Genetic variants in the purinergic receptors P2RX4 and P2RX7 have been shown to affect susceptibility to multiple sclerosis (MS). In this study, we set out to evaluate whether rare coding variants of major effect could also be identified in these purinergic receptors. Sequencing analysis of P2RX4 and P2RX7 in 193 MS patients and 100 controls led to the identification of a rare three variant haplotype (P2RX7 rs140915863:C>T [p.T205M], P2RX7 rs201921967:A>G [p.N361S], and P2RX4 rs765866317:G>A [p.G135S]) segregating with disease in a multi-incident family with six family members diagnosed with MS (logarithm of odds = 3.07). Functional analysis of this haplotype in HEK293 cells revealed impaired P2X7 surface expression (P < 0.01), resulting in over 95% inhibition of adenosine triphosphate (ATP)-induced pore function (P < 0.001) and a marked reduction in phagocytic ability (P < 0.05). In addition, transfected cells showed 40% increased peak ATP-induced inward current (P < 0.01), and a greater Ca2+ response to the P2X4 135S variant compared with wild type (P < 0.0001). Our study nominates rare genetic variants in P2RX4 and P2RX7 as major genetic contributors to disease, further supporting a role for these purinergic receptors in MS and the disruption of transmembrane cation channels leading to impairment of phagocytosis as the pathological mechanisms of disease.


Genetic Predisposition to Disease , Multiple Sclerosis/genetics , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X7/genetics , Female , HEK293 Cells , Haplotypes , Humans , Male , Multiple Sclerosis/pathology , Polymorphism, Single Nucleotide
10.
J Neurochem ; 139(5): 806-822, 2016 12.
Article En | MEDLINE | ID: mdl-27696399

Toluene is a commonly abused inhalant that is easily accessible to adolescents. Despite the increasing incidence of use, our understanding of its long-term impact remains limited. Here, we used a range of techniques to examine the acute and chronic effects of toluene exposure on glutameteric and GABAergic function, and on indices of psychological function in adult rats after adolescent exposure. Metabolomics conducted on cortical tissue established that acute exposure to toluene produces alterations in cellular metabolism indicative of a glutamatergic and GABAergic profile. Similarly, in vitro electrophysiology in Xenopus oocytes found that acute toluene exposure reduced NMDA receptor signalling. Finally, in an adolescent rodent model of chronic intermittent exposure to toluene (10 000 ppm), we found that, while toluene exposure did not affect initial learning, it induced a deficit in updating that learning when response-outcome relationships were reversed or degraded in an instrumental conditioning paradigm. There were also group differences when more effort was required to obtain the reward; toluene-exposed animals were less sensitive to progressive ratio schedules and to delayed discounting. These behavioural deficits were accompanied by changes in subunit expression of both NMDA and GABA receptors in adulthood, up to 10 weeks after the final exposure to toluene in the hippocampus, prefrontal cortex and ventromedial striatum; regions with recognized roles in behavioural flexibility and decision-making. Collectively, our data suggest that exposure to toluene is sufficient to induce adaptive changes in glutamatergic and GABAergic systems and in adaptive behaviour that may underlie the deficits observed following adolescent inhalant abuse, including susceptibility to further drug-use.


Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Toluene/administration & dosage , Toluene/toxicity , Administration, Inhalation , Age Factors , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Female , Guinea Pigs , Learning/drug effects , Learning/physiology , Male , Organ Culture Techniques , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Solvents/administration & dosage , Solvents/toxicity , Xenopus laevis
11.
Epilepsia ; 57(9): 1398-405, 2016 09.
Article En | MEDLINE | ID: mdl-27440235

OBJECTIVE: Fracture risk is a serious comorbidity in epilepsy and may relate to the use of antiepileptic drugs (AEDs). Many AEDs inhibit ion channel function, and the expression of these channels in osteoblasts raises the question of whether altered bone signaling increases bone fragility. We aimed to confirm the expression of voltage-gated sodium (NaV ) channels in mouse osteoblasts, and to investigate the action of carbamazepine and phenytoin on NaV channels. METHODS: Immunocytochemistry was performed on primary calvarial osteoblasts extracted from neonatal C57BL/6J mice and additional RNA sequencing (RNASeq) was included to confirm expression of NaV . Whole-cell patch-clamp recordings were made to identify the native currents expressed and to assess the actions of carbamazepine (50 µm) or phenytoin (50 µm). RESULTS: NaV expression was demonstrated with immunocytochemistry, RNA sequencing, and functionally, with demonstration of robust tetrodotoxin-sensitive and voltage-activated inward currents. Application of carbamazepine or phenytoin resulted in significant inhibition of current amplitude for carbamazepine (31.6 ± 5.9%, n = 9; p < 0.001), and for phenytoin (35.5 ± 6.9%, n = 7; p < 0.001). SIGNIFICANCE: Mouse osteoblasts express NaV , and native NaV currents are blocked by carbamazepine and phenytoin, supporting our hypothesis that AEDs can directly influence osteoblast function and potentially affect bone strength.


Anticonvulsants/pharmacology , Carbamazepine/pharmacology , Osteoblasts/drug effects , Phenytoin/pharmacology , Sodium Channels/physiology , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Electric Stimulation , Gene Expression Regulation/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Patch-Clamp Techniques , RNA, Messenger , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
12.
Neurology ; 86(17): 1605-12, 2016 Apr 26.
Article En | MEDLINE | ID: mdl-27029629

OBJECTIVES: We report development of a targeted resequencing gene panel for focal epilepsy, the most prevalent phenotypic group of the epilepsies. METHODS: The targeted resequencing gene panel was designed using molecular inversion probe (MIP) capture technology and sequenced using massively parallel Illumina sequencing. RESULTS: We demonstrated proof of principle that mutations can be detected in 4 previously genotyped focal epilepsy cases. We searched for both germline and somatic mutations in 251 patients with unsolved sporadic or familial focal epilepsy and identified 11 novel or very rare missense variants in 5 different genes: CHRNA4, GRIN2B, KCNT1, PCDH19, and SCN1A. Of these, 2 were predicted to be pathogenic or likely pathogenic, explaining ∼0.8% of the cohort, and 8 were of uncertain significance based on available data. CONCLUSIONS: We have developed and validated a targeted resequencing panel for focal epilepsies, the most important clinical class of epilepsies, accounting for about 60% of all cases. Our application of MIP technology is an innovative approach that will be advantageous in the clinical setting because it is highly sensitive, efficient, and cost-effective for screening large patient cohorts. Our findings indicate that mutations in known genes likely explain only a small proportion of focal epilepsy cases. This is not surprising given the established clinical and genetic heterogeneity of these disorders and underscores the importance of further gene discovery studies in this complex syndrome.


Epilepsies, Partial/genetics , Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , Mutation , Sequence Analysis, DNA/methods , Cadherins/genetics , Cohort Studies , Female , Genetic Predisposition to Disease , Humans , Male , NAV1.1 Voltage-Gated Sodium Channel/genetics , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , Protocadherins , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, Nicotinic/genetics
13.
Ann Neurol ; 78(6): 995-9, 2015 Dec.
Article En | MEDLINE | ID: mdl-26369628

We report 2 patients with drug-resistant epilepsy caused by KCNT1 mutations who were treated with quinidine. Both mutations manifested gain of function in vitro, showing increased current that was reduced by quinidine. One, who had epilepsy of infancy with migrating focal seizures, had 80% reduction in seizure frequency as recorded in seizure diaries, and partially validated by objective seizure evaluation on EEG. The other, who had a novel phenotype, with severe nocturnal focal and secondary generalized seizures starting in early childhood with developmental regression, did not improve. Although quinidine represents an encouraging opportunity for therapeutic benefits, our experience suggests caution in its application and supports the need to identify more targeted drugs for KCNT1 epilepsies.


Drug Resistant Epilepsy/drug therapy , Enzyme Inhibitors/pharmacology , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Quinidine/pharmacology , Child , Child, Preschool , Drug Resistant Epilepsy/genetics , Enzyme Inhibitors/administration & dosage , Female , Humans , Male , Mutation , Potassium Channels, Sodium-Activated , Quinidine/administration & dosage
14.
PLoS One ; 10(3): e0118946, 2015.
Article En | MEDLINE | ID: mdl-25794116

Nucleotide alterations in the gene encoding proline-rich transmembrane protein 2 (PRRT2) have been identified in most patients with benign partial epilepsies in infancy (BPEI)/benign familial infantile epilepsy (BFIE). However, not all patients harbor these PRRT2 mutations, indicating the involvement of genes other than PRRT2. In this study, we performed whole exome sequencing analysis for a large family affected with PRRT2-unrelated BPEI. We identified a non-synonymous single nucleotide variation (SNV) in the voltage-sensitive chloride channel 6 gene (CLCN6). A cohort study of 48 BPEI patients without PRRT2 mutations revealed a different CLCN6 SNV in a patient, his sibling and his father who had a history of febrile seizures (FS) but not BPEI. Another study of 48 patients with FS identified an additional SNV in CLCN6. Chloride channels (CLCs) are involved in a multitude of physiologic processes and some members of the CLC family have been linked to inherited diseases. However, a phenotypic correlation has not been confirmed for CLCN6. Although we could not detect significant biological effects linked to the identified CLCN6 SNVs, further studies should investigate potential CLCN6 variants that may underlie the genetic susceptibility to convulsive disorders.


Chloride Channels/genetics , Epilepsy, Benign Neonatal/complications , Epilepsy, Benign Neonatal/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Seizures, Febrile/complications , Seizures, Febrile/genetics , Amino Acid Sequence , Base Sequence , Chloride Channels/chemistry , DNA Mutational Analysis , Exons/genetics , Female , Genetic Association Studies , Humans , Infant, Newborn , Male , Molecular Sequence Data , Mutagenesis , Pedigree , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Ann Neurol ; 75(4): 581-90, 2014 Apr.
Article En | MEDLINE | ID: mdl-24591078

OBJECTIVE: Mutations in KCNT1 have been implicated in autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE) and epilepsy of infancy with migrating focal seizures (EIMFS). More recently, a whole exome sequencing study of epileptic encephalopathies identified an additional de novo mutation in 1 proband with EIMFS. We aim to investigate the electrophysiological and pharmacological characteristics of hKCNT1 mutations and examine developmental expression levels. METHODS: Here we use a Xenopus laevis oocyte-based automated 2-electrode voltage clamp assay. The effects of quinidine (100 and 300 µM) are also tested. Using quantitative reverse transcriptase polymerase chain reaction, the relative levels of mouse brain mKcnt1 mRNA expression are determined. RESULTS: We demonstrate that KCNT1 mutations implicated in epilepsy cause a marked increase in function. Importantly, there is a significant group difference in gain of function between mutations associated with ADNFLE and EIMFS. Finally, exposure to quinidine significantly reduces this gain of function for all mutations studied. INTERPRETATION: These results establish direction for a targeted therapy and potentially exemplify a translational paradigm for in vitro studies informing novel therapies in a neuropsychiatric disease.


Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Quinidine/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Animals , Brain/growth & development , Brain/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , Humans , Male , Mice , Mice, Inbred C57BL , Microinjections , Oocytes , Patch-Clamp Techniques , Potassium Channels, Sodium-Activated , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Xenopus laevis
16.
Methods Mol Biol ; 998: 171-87, 2013.
Article En | MEDLINE | ID: mdl-23529429

Ion channels are integral membrane proteins that regulate the flow of ions across the plasma membrane and the membranes of intracellular organelles of both excitable and non-excitable cells. Ion channels are vital to a wide variety of biological processes and are prominent components of the nervous system and cardiovascular system, as well as controlling many metabolic functions. Furthermore, ion channels are known to be involved in many disease states and as such have become popular therapeutic targets. For many years now manual patch-clamping has been regarded as one of the best approaches for assaying ion channel function, through direct measurement of ion flow across these membrane proteins. Over the last decade there have been many remarkable breakthroughs in the development of technologies enabling the study of ion channels. One of these breakthroughs is the development of automated planar patch-clamp technology. Automated platforms have demonstrated the ability to generate high-quality data with high throughput capabilities, at great efficiency and reliability. Additional features such as simultaneous intracellular and extracellular perfusion of the cell membrane, current clamp operation, fast compound application, an increasing rate of parallelization, and more recently temperature control have been introduced. Furthermore, in addition to the well-established studies of over-expressed ion channel proteins in cell lines, new generations of planar patch-clamp systems have enabled successful studies of native and primary mammalian cells. This technology is becoming increasingly popular and extensively used both within areas of drug discovery as well as academic research. Many platforms have been developed including NPC-16 Patchliner(®) and SyncroPatch(®) 96 (Nanion Technologies GmbH, Munich), CytoPatch™ (Cytocentrics AG, Rostock), PatchXpress(®) 7000A, IonWorks(®) Quattro and IonWorks Barracuda™, (Molecular Devices, LLC); Dynaflow(®) HT (Cellectricon AB, Mölndal), QPatch HT (Sophion A/S, Copenhagen), IonFlux HT (Fluxion Bioscience Inc, USA), which have demonstrated the capability to generate recordings similar in quality to that of conventional patch clamping. Here we describe features of Nanion's NPC-16 Patchliner(®) and processes and protocols suited for this particularly flexible and successful high-throughput automated platform, which is based on planar patch-clamp technology. However, many of the protocols and notes given in this chapter can be applied to other automated patch-clamp platforms, similarly.


Patch-Clamp Techniques/methods , Animals , Automation , Embryonic Stem Cells/cytology , Mice , Myocytes, Cardiac/cytology , Patch-Clamp Techniques/instrumentation , Rats , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Temperature , alpha7 Nicotinic Acetylcholine Receptor
17.
Am J Respir Crit Care Med ; 186(7): 648-56, 2012 Oct 01.
Article En | MEDLINE | ID: mdl-22822026

RATIONALE: Clinical reports describe life-threatening cardiac arrhythmias after environmental exposure to carbon monoxide (CO) or accidental CO poisoning. Numerous case studies describe disruption of repolarization and prolongation of the QT interval, yet the mechanisms underlying CO-induced arrhythmias are unknown. OBJECTIVES: To understand the cellular basis of CO-induced arrhythmias and to identify an effective therapeutic approach. METHODS: Patch-clamp electrophysiology and confocal Ca(2+) and nitric oxide (NO) imaging in isolated ventricular myocytes was performed together with protein S-nitrosylation to investigate the effects of CO at the cellular and molecular levels, whereas telemetry was used to investigate effects of CO on electrocardiogram recordings in vivo. MEASUREMENTS AND MAIN RESULTS: CO increased the sustained (late) component of the inward Na(+) current, resulting in prolongation of the action potential and the associated intracellular Ca(2+) transient. In more than 50% of myocytes these changes progressed to early after-depolarization-like arrhythmias. CO elevated NO levels in myocytes and caused S-nitrosylation of the Na(+) channel, Na(v)1.5. All proarrhythmic effects of CO were abolished by the NO synthase inhibitor l-NAME, and reversed by ranolazine, an inhibitor of the late Na(+) current. Ranolazine also corrected QT variability and arrhythmias induced by CO in vivo, as monitored by telemetry. CONCLUSIONS: Our data indicate that the proarrhythmic effects of CO arise from activation of NO synthase, leading to NO-mediated nitrosylation of Na(V)1.5 and to induction of the late Na(+) current. We also show that the antianginal drug ranolazine can abolish CO-induced early after-depolarizations, highlighting a novel approach to the treatment of CO-induced arrhythmias.


Arrhythmias, Cardiac/etiology , Carbon Monoxide Poisoning/complications , Carbon Monoxide/pharmacology , Myocytes, Cardiac/drug effects , Voltage-Gated Sodium Channels/drug effects , Acetanilides/therapeutic use , Action Potentials/drug effects , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/physiopathology , Calcium Signaling/drug effects , Carbon Monoxide/adverse effects , Carbon Monoxide Poisoning/physiopathology , Cell Culture Techniques , Disease Models, Animal , Environmental Exposure/adverse effects , Enzyme Inhibitors/therapeutic use , Male , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Piperazines/therapeutic use , Ranolazine , Rats , Rats, Wistar , Voltage-Gated Sodium Channels/physiology
18.
Mol Pharmacol ; 79(6): 1023-30, 2011 Jun.
Article En | MEDLINE | ID: mdl-21406603

The aim of this study was to generate new insight into chemical regulation of transient receptor potential (TRP) channels with relevance to glucose homeostasis and the metabolic syndrome. Human TRP melastatin 2 (TRPM2), TRPM3, and TRP canonical 5 (TRPC5) were conditionally overexpressed in human embryonic kidney 293 cells and studied by using calcium-measurement and patch-clamp techniques. Rosiglitazone and other peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists were investigated. TRPM2 was unaffected by rosiglitazone at concentrations up to 10 µM but was inhibited completely at higher concentrations (IC(50), ∼22.5 µM). TRPM3 was more potently inhibited, with effects occurring in a biphasic concentration-dependent manner such that there was approximately 20% inhibition at low concentrations (0.1-1 µM) and full inhibition at higher concentrations (IC(50), 5-10 µM). PPAR-γ antagonism by 2-chloro-5-nitrobenzanilide (GW9662) did not prevent inhibition of TRPM3 by rosiglitazone. TRPC5 was strongly stimulated by rosiglitazone at concentrations of ≥10 µM (EC(50), ∼30 µM). Effects on TRPM3 and TRPC5 occurred rapidly and reversibly. Troglitazone and pioglitazone inhibited TRPM3 (IC(50), 12 µM) but lacked effect on TRPC5, suggesting no relevance of PPAR-γ or the thiazolidinedione moiety to rosiglitazone stimulation of TRPC5. A rosiglitazone-related but nonthiazolidinedione PPAR-γ agonist, N-(2-benzoylphenyl)-O-[2-(methyl-2-pyridinylamino)ethyl]-l-tyrosine (GW1929), was a weak stimulator of TRPM3 and TRPC5. The natural PPAR-γ agonist 15-deoxy prostaglandin J(2), had no effect on TRPM3 or TRPC5. The data suggest that rosiglitazone contains chemical moieties that rapidly, strongly, and differentially modulate TRP channels independently of PPAR-γ, potentially contributing to biological consequences of the agent and providing the basis for novel TRP channel pharmacology.


TRPC Cation Channels/drug effects , TRPM Cation Channels/drug effects , Thiazolidinediones/pharmacology , Calcium/metabolism , Cell Line , Humans , Patch-Clamp Techniques , Rosiglitazone
19.
FASEB J ; 25(5): 1519-30, 2011 May.
Article En | MEDLINE | ID: mdl-21248240

Oxidative stress induces neuronal apoptosis and is implicated in cerebral ischemia, head trauma, and age-related neurodegenerative diseases. An early step in this process is the loss of intracellular K(+) via K(+) channels, and evidence indicates that K(v)2.1 is of particular importance in this regard, being rapidly inserted into the plasma membrane in response to apoptotic stimuli. An additional feature of neuronal oxidative stress is the up-regulation of the inducible enzyme heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). CO provides neuronal protection against stresses such as stroke and excitotoxicity, although the underlying mechanisms are not yet elucidated. Here, we demonstrate that CO reversibly inhibits K(v)2.1. Channel inhibition by CO involves reactive oxygen species and protein kinase G activity. Overexpression of K(v)2.1 in HEK293 cells increases their vulnerability to oxidant-induced apoptosis, and this is reversed by CO. In hippocampal neurons, CO selectively inhibits K(v)2.1, reverses the dramatic oxidant-induced increase in K(+) current density, and provides marked protection against oxidant-induced apoptosis. Our results provide a novel mechanism to account for the neuroprotective effects of CO against oxidative apoptosis, which has potential for therapeutic exploitation to provide neuronal protection in situations of oxidative stress.


2,2'-Dipyridyl/analogs & derivatives , Apoptosis/drug effects , Carbon Monoxide/pharmacology , Disulfides/pharmacology , Oxidants/pharmacology , Shab Potassium Channels/metabolism , 2,2'-Dipyridyl/pharmacology , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line , Electrophysiology , HEK293 Cells , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Rats , Rats, Wistar
20.
Arterioscler Thromb Vasc Biol ; 30(7): 1453-9, 2010 Jul.
Article En | MEDLINE | ID: mdl-20378846

OBJECTIVE: To determine whether calcium-permeable channels are targets for the oxidized phospholipids: 1-palmitoyl-2-glutaroyl-phosphatidylcholine (PGPC) and 1-palmitoyl-2-oxovaleroyl-phosphatidylcholine (POVPC). METHODS AND RESULTS: Oxidized phospholipids are key factors in inflammation and associated diseases, including atherosclerosis; however, the initial reception mechanisms for cellular responses to the factors are poorly understood. Low micromolar concentrations of PGPC and POVPC evoked increases in intracellular calcium in human embryonic kidney 293 cells that overexpressed human transient receptor potential canonical 5 (TRPC5) but not human TRP melastatin (TRPM) 2 or 3. The results of electrophysiological experiments confirmed stimulation of TRPC5. To investigate relevance to endogenous channels, we studied proliferating vascular smooth muscle cells from patients undergoing coronary artery bypass surgery. PGPC and POVPC elicited calcium entry that was inhibited by anti-TRPC5 or anti-TRPC1 antibodies or dominant-negative mutant TRPC5. Calcium release did not occur. The effect was functionally relevant because it enhanced cell migration. The actions of PGPC and POVPC depended on G(i/o) proteins but not on previously identified G protein-coupled receptors for oxidized phospholipids. CONCLUSIONS: Stimulation of calcium-permeable TRPC5-containing channels may be an early event in cellular responses to oxidized phospholipids that couples to cell migration and requires an unidentified G protein-coupled receptor.


Calcium Signaling , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phospholipid Ethers/metabolism , TRPC Cation Channels/metabolism , Cell Line , Cell Movement , Cell Proliferation , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Membrane Potentials , Mutation , Oxidation-Reduction , TRPC Cation Channels/genetics , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Time Factors , Transfection
...