1,069 results on '"Sodium Channel"'
Search Results
2. SCN9A variant in a family of mixed breed dogs with congenital insensitivity to pain
- Author
-
Rodrigo Gutierrez‐Quintana, Matthias Christen, Kiterie M. E. Faller, Julien Guevar, Vidhya Jagannathan, and Tosso Leeb
- Subjects
animal model ,Canis lupus familiaris ,genetics ,neurology ,precision medicine ,sodium channel ,Veterinary medicine ,SF600-1100 - Abstract
Abstract Background Congenital insensitivity to pain (CIP) and hereditary sensory and autonomic neuropathies (HSANs) are a rare group of genetic disorders causing inability to feel pain. Three different associated variants have been identified in dogs: 1 in Border Collies, 1 in mixed breed dogs, and 1 in Spaniels and Pointers. Objectives To clinically and genetically characterize CIP in a family of mixed breed dogs. Animals Two mixed breed dogs from the same litter were independently presented: 1 for evaluation of painless fractures, and the other for chronic thermal skin injuries. Methods Physical, neurological, and histopathological evaluations were performed. Whole genome sequencing of 1 affected dog was used to identify homozygous protein‐changing variants that were not present in 926 control genomes from diverse dog breeds. Results Physical and neurological examinations showed the absence of superficial and deep pain perception in the entire body. Histopathological evaluations of the brain, spinal cord and sensory ganglia were normal. Whole genome sequencing identified a homozygous missense variant in SCN9A, XM_038584713.1:c.2761C>T or XP_038440641.1:(p.Arg921Cys). Both affected dogs were homozygous for the mutant allele, which was not detected in 926 dogs of different breeds. Conclusions and Clinical Importance We confirmed the diagnosis of CIP in a family of mixed breed dogs and identified a likely pathogenic variant in the SCN9A gene. The clinical signs observed in these dogs mimic those reported in humans with pathogenic SCN9A variants causing CIP. This report is the first of a spontaneous pathogenic SCN9A variant in domestic animals.
- Published
- 2023
- Full Text
- View/download PDF
3. Editorial to "An extremely wide QRS complex tachycardia induced by anamorelin".
- Author
-
Inoue S
- Abstract
Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2024
- Full Text
- View/download PDF
4. Transcriptional profiles of genes related to electrophysiological function in Scn5a+/− murine hearts
- Author
-
Michael Takla, Charlotte E. Edling, Kevin Zhang, Khalil Saadeh, Gary Tse, Samantha C. Salvage, Christopher L.‐H. Huang, and Kamalan Jeevaratnam
- Subjects
arrhythmia ,Brugada syndrome ,mechanisms ,sodium channel ,transcription ,Physiology ,QP1-981 - Abstract
Abstract The Scn5a gene encodes the major pore‐forming Nav1.5 (α) subunit, of the voltage‐gated Na+ channel in cardiomyocytes. The key role of Nav1.5 in action potential initiation and propagation in both atria and ventricles predisposes organisms lacking Scn5a or carrying Scn5a mutations to cardiac arrhythmogenesis. Loss‐of‐function Nav1.5 genetic abnormalities account for many cases of the human arrhythmic disorder Brugada syndrome (BrS) and related conduction disorders. A murine model with a heterozygous Scn5a deletion recapitulates many electrophysiological phenotypes of BrS. This study examines the relationships between its Scn5a+/− genotype, resulting transcriptional changes, and the consequent phenotypic presentations of BrS. Of 62 selected protein‐coding genes related to cardiomyocyte electrophysiological or homeostatic function, concentrations of mRNA transcribed from 15 differed significantly from wild type (WT). Despite halving apparent ventricular Scn5a transcription heterozygous deletion did not significantly downregulate its atrial expression, raising possibilities of atria‐specific feedback mechanisms. Most of the remaining 14 genes whose expression differed significantly between WT and Scn5a+/− animals involved Ca2+ homeostasis specifically in atrial tissue, with no overlap with any ventricular changes. All statistically significant changes in expression were upregulations in the atria and downregulations in the ventricles. This investigation demonstrates the value of future experiments exploring for and clarifying links between transcriptional control of Scn5a and of genes whose protein products coordinate Ca2+ regulation and examining their possible roles in BrS.
- Published
- 2021
- Full Text
- View/download PDF
5. Possible role of SCN4A skeletal muscle mutation in apnea during seizure
- Author
-
Dilşad Türkdoğan, Emma Matthews, Sunay Usluer, Aslı Gündoğdu, Kayıhan Uluç, Roope Mannikko, Michael G. Hanna, Sanjay M. Sisodiya, and Hande S. Çağlayan
- Subjects
laryngospasm ,SUDEP ,myotonia ,sodium channel ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract SCN4A gene mutations cause a number of neuromuscular phenotypes including myotonia. A subset of infants with myotonia‐causing mutations experience severe life‐threatening episodic laryngospasm with apnea. We have recently identified similar SCN4A mutations in association with sudden infant death syndrome. Laryngospasm has also been proposed as a contributory mechanism to some cases of sudden unexpected death in epilepsy (SUDEP). We report an infant with EEG‐confirmed seizures and recurrent apneas. Whole‐exome sequencing identified a known pathogenic mutation in the SCN4A gene that has been reported in several unrelated families with myotonic disorder. We propose that the SCN4A mutation contributed to the apneas in our case, irrespective of the underlying cause of the epilepsy. We suggest this supports the notion that laryngospasm may contribute to some cases of SUDEP, and implicates a possible shared mechanism between a proportion of sudden infant deaths and sudden unexpected deaths in epilepsy.
- Published
- 2019
- Full Text
- View/download PDF
6. Identification of rare heterozygous linkage R965C‐R1309H mutations in the pore‐forming region of SCN5A gene associated with complex arrhythmia
- Author
-
Yubi Lin, Jiading Qin, Yuhui Shen, Jiana Huang, Zuoquan Zhang, ZhiLing Zhu, Huifang Lu, Yin Huang, Yuelan Yin, Ani Wang, Lizi Jin, Zhenyu Hu, Xiufang Lin, and Bin Jiang
- Subjects
atrial fibrillation ,hereditary arrhythmia ,SCN5A ,sodium channel ,Genetics ,QH426-470 - Abstract
Abstract Background We examined the genetic background of a Chinese Han family in which some members presented with complex arrhythmias including sick sinus syndrome, progressive conduction block, atrial fibrillation, atrial standstill and Brugada syndrome. The possible underlying mechanism associated with the genetic mutation was explored. Methods Targeted capture sequencing was conducted in the probands in the coding and splicing regions of genes implicated in inherited arrhythmias. Stable cell lines overexpressing wild type (WT) or mutant SCN5A were generated in HEK293T cells. Whole‐cell recording was performed to evaluate the functional changes in sodium channels. Results The rare heterozygous linkage mutations, SCN5A R965C and R1309H, were found in these patients with complex familial arrhythmias. Compared to WT, R965C or R1309H, the peak current of sodium channel was dramatically reduced in HEK293T cell with linkage R965C‐R1309H mutation when testing potentials ranging from −45 to 15 mV. Notably, the maximum peak current of sodium channels with R1309H and linkage R965C‐R1309H displayed significant decreases of 31.5% and 73.34%, respectively, compared to WT. Additionally, compared to R965C or R1309H alone, the linkage mutation R965C‐R1309H demonstrated not only a more obvious depolarisation‐shifted activation and hyperpolarisation‐shifted inactivation, but also a more significant alteration in the time constant, V1/2 and the slope factor of activation and inactivation. Conclusions The linkage mutation SCN5A R965C‐R1309H led to a more dramatically reduced current density, as well as more significant depolarisation‐shifted activation and hyperpolarisation‐shifted inactivation in sodium channels than R965C or R1309H alone, which potentially explain this complex familial arrhythmia syndrome.
- Published
- 2021
- Full Text
- View/download PDF
7. The role of sodium channels in sudden unexpected death in pediatrics
- Author
-
Anne M. Rochtus, Richard D. Goldstein, Ingrid A. Holm, Catherine A. Brownstein, Eduardo Pérez‐Palma, Robin Haynes, Dennis Lal, and Annapurna H. Poduri
- Subjects
arrhythmia ,epilepsy ,sodium channel ,sudden unexpected death ,Genetics ,QH426-470 - Abstract
Abstract Background Sudden Unexpected Death in Pediatrics (SUDP) is a tragic event, likely caused by the complex interaction of multiple factors. The presence of hippocampal abnormalities in many children with SUDP suggests that epilepsy‐related mechanisms may contribute to death, similar to Sudden Unexplained Death in Epilepsy. Because of known associations between the genes SCN1A and SCN5A and sudden death, and shared mechanisms and patterns of expression in genes encoding many voltage‐gated sodium channels (VGSCs), we hypothesized that individuals dying from SUDP have pathogenic variants across the entire family of cardiac arrhythmia‐ and epilepsy‐associated VGSC genes. Methods To address this hypothesis, we evaluated whole‐exome sequencing data from infants and children with SUDP for variants in VGSC genes, reviewed the literature for all SUDP‐associated variants in VGSCs, applied a novel paralog analysis to all variants, and evaluated all variants according to American College of Medical Genetics and Genomics (ACMG) guidelines. Results In our cohort of 73 cases of SUDP, we assessed 11 variants as pathogenic in SCN1A, SCN1B, and SCN10A, genes with long‐standing disease associations, and in SCN3A, SCN4A, and SCN9A, VGSC gene paralogs with more recent disease associations. From the literature, we identified 82 VGSC variants in SUDP cases. Pathogenic variants clustered at conserved amino acid sites intolerant to variation across the VGSC genes, which is unlikely to occur in the general population (p
- Published
- 2020
- Full Text
- View/download PDF
8. SCN5A gene mutations and the risk of ventricular fibrillation and syncope in Brugada syndrome patients: A meta‐analysis
- Author
-
Sunu Budhi Raharjo, Rido Maulana, Irma Maghfirah, Fatimah Alzahra, Agnes Dinar Putrinarita, Dicky A. Hanafy, and Yoga Yuniadi
- Subjects
Brugada syndrome ,SCN5A mutations ,sodium channel ,syncope ,ventricular fibrillation ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Abstract Mutations in the gene encoding the main cardiac sodium channel (SCN5A) are the commonest genetic cause of Brugada syndrome (BrS). However, the effect of SCN5A mutations on the outcomes of ventricular fibrillation (VF) and syncope remains uncertain. To clarify this relationship, a meta‐analysis was performed. A comprehensive search was conducted to identify all eligible studies from PubMed, MEDLINE, EBSCO, ProQuest, Science Direct, Clinical Key, and Cochrane database for cohort studies of BrS populations that had been systematically tested for SCN5A mutations. We did meta‐analysis to see the relationship between SCN5A mutations and the occurrence of VF and/or syncope using RevMan 5.3. Five clinical studies met our criteria and included a total of 665 BrS patients. These studies included 45 patients with VF and 178 patients with syncope. We found that in BrS patients with SCN5A mutations the rate of VF event was 30.7% while in patients without mutations was 28.5% (Risk Ratio [RR] = 1.11, [95% CI: 0.61, 2.00], P = 0.73, I2 = 0%). The occurrence of syncope events was 35.9% in patients with SCN5A mutations and 34.5% in patients without mutations (RR = 1.12, [95% CI: 0.87, 1.45], P = 0.37, I2 = 39%). Furthermore, the occurrence of combined VF and syncope events were similar between the 2 groups (RR = 1.12, [95% CI: 0.89, 1.42], P = 0.34, I2 = 11%). BrS patients with SCN5A mutations exhibit a similar risk of future occurence of VF and/or syncope as compared to those without SCN5A mutations.
- Published
- 2018
- Full Text
- View/download PDF
9. Functional diversity of sodium channel variants in common eastern bumblebee, Bombus impatiens.
- Author
-
Chen L, Wang Y, Zhang K, and Wu S
- Subjects
- Bees genetics, Animals, Insecta metabolism, Alternative Splicing, Sodium metabolism, Amino Acids metabolism, Insecticides pharmacology, Voltage-Gated Sodium Channels genetics, Voltage-Gated Sodium Channels chemistry, Voltage-Gated Sodium Channels metabolism, Pyrethrins pharmacology
- Abstract
For the past decade, Colony Collapse Disorder has been reported worldwide. Insecticides containing pyrethroids may be responsible for a decline in bees, which are more sensitive to pyrethroids compared with other insects. Voltage-gated sodium channels (Na
v ) are the major target sites of pyrethroids, and the sodium channel diversity is generated through extensive alternative splicing and RNA editing. In this study, we cloned and analyzed the function of variants of the Nav channel, BiNav , from Bombus impatiens. BiNav covers a 46 kb genome region including 30 exons. Sequence analysis of 56 clones showed that the clones can be grouped into 22 splice types with 11 optional exons (exons j, w, p, q, r, b, e, t, l/k, and z). Here, a special alternative exon w is identified, encoding a stretch of 31 amino acid resides in domain I between S3 and S4. RNA editing generates 18 amino acid changes in different positions in individual variants. Among 56 variants examined, only six variants generated sufficient sodium currents for functional characterization in Xenopus oocytes. In the presence of B. impatiens TipE and TEH1, the sodium current amplitude of BiNav 1-1 increased by fourfold, while TipE of other insect species had no effect on the expression. Abundant alternative splicing and RNA editing of BiNav suggests the molecular and functional pharmacology diversity of the Nav channel for bumblebees. This study provides a theoretical basis for the design of insecticides that specifically target pests without affecting beneficial insects., (© 2023 Wiley Periodicals LLC.)- Published
- 2023
- Full Text
- View/download PDF
10. Fast voltage‐dependent sodium (Na V ) currents are functionally expressed in mouse corpus cavernosum smooth muscle cells
- Author
-
Xin Rui Lim, Eamonn Bradley, Gerard P. Sergeant, Keith D. Thornbury, Caoimhin S. Griffin, and Mark A. Hollywood
- Subjects
Pharmacology ,Sodium channel ,Stimulation ,Cell biology ,Contractility ,chemistry.chemical_compound ,Electrophysiology ,chemistry ,Nifedipine ,medicine ,Patch clamp ,Veratridine ,Ion channel ,medicine.drug - Abstract
Background and purpose Corpus cavernosum smooth muscle (CCSM) exhibits phasic contractions that are coordinated by ion channels. Mouse models are commonly used to study erectile dysfunction, but there are few published electrophysiological studies of mouse CCSM. We describe, for the first time, voltage-dependent sodium (NaV ) currents in mouse CCSM and investigate their function. Experimental approach Electrophysiological, pharmacological, and immunocytochemical studies on isolated CCSM cells. Tension measurements in whole tissue. Key results A fast, voltage-dependent sodium current was induced by depolarising steps. Steady-state activation and inactivation curves revealed a window current between -60 and -30 mV. Two populations of NaV currents, ('TTX-sensitive') and ('TTX-insensitive'), were distinguished. TTX-sensitive current showed 48% block with the NaV -subtype-specific blockers ICA-121431 (NaV 1.1-1.3), PF-05089771 (NaV 1.7), and 4,9-anhydro-TTX (NaV 1.6). TTX-insensitive current was insensitive to A803467, a NaV 1.8 blocker. Immunocytochemistry confirmed the expression of NaV 1.5 and NaV 1.4 in freshly dispersed CCSM cells. Veratridine, a NaV activator, reduced time-dependent inactivation of the current and increased the duration of evoked action potentials. Veratridine induced phasic contractions in CCSM strips. This effect was reversible with TTX and nifedipine but not by KB-R7943. Conclusion and implications We report, for the first time, a fast voltage-dependent sodium current in mouse CCSM. Stimulation of this current increases the contractility of corpus cavernosum in vitro, suggesting that it may contribute to the mechanisms of detumescence, and potentially serve as a clinically relevant target for pharmaceutical intervention in erectile dysfunction. Further work will be necessary to define its role.
- Published
- 2021
11. Neonatal Na V 1.5 channels: pharmacological distinctiveness of a cancer‐related voltage‐gated sodium channel splice variant
- Author
-
Frank Bosmans, Margaux Theys, Rustem Onkal, Scott P. Fraser, and Mustafa B.A. Djamgoz
- Subjects
EXPRESSION ,INVASION ,spider toxin ,Xenopus ,Nav1.5 ,Pharmacology ,ACTIVATION ,NA+ CHANNELS ,antibody ,Mexiletine ,voltage-gated sodium channel ,Medicine and Health Sciences ,medicine ,cancer ,metastasis ,Patch clamp ,biology ,Chemistry ,Sodium channel ,IN-VITRO ,biology.organism_classification ,Spider toxin ,CONCISE GUIDE ,PROSTATE-CANCER ,Riluzole ,Electrophysiology ,NAV1.5 ,FAST INACTIVATION ,biology.protein ,LIDOCAINE BLOCK ,medicine.drug - Abstract
Background and Purpose Voltage-gated sodium (Na-V) channels are expressed de novo in carcinomas where their activity promotes invasiveness. Breast and colon cancer cells express the neonatal splice variant of Na(V)1.5 (nNa(V)1.5), which has several amino acid substitutions in the domain I voltage-sensor compared with its adult counterpart (aNa(V)1.5). This study aimed to determine whether nNa(V)1.5 channels could be distinguished pharmacologically from aNa(V)1.5 channels. Experimental Approach Cells expressing either nNa(V)1.5 or aNa(V)1.5 channels were exposed to low MW inhibitors, an antibody or natural toxins, and changes in electrophysiological parameters were measured. Stable expression in EBNA cells and transient expression in Xenopus laevis oocytes were used. Currents were recorded by whole-cell patch clamp and two-electrode voltage-clamp, respectively. Key Results Several clinically used blockers of Na-V channels (lidocaine, procaine, phenytoin, mexiletine, ranolazine, and riluzole) could not distinguish between nNa(V)1.5 or aNa(V)1.5 channels. However, two tarantula toxins (HaTx and ProTx-II) and a polyclonal antibody (NESOpAb) preferentially inhibited currents elicited by either nNa(V)1.5 or aNa(V)1.5 channels by binding to the spliced region of the channel. Furthermore, the amino acid residue at position 211 (aspartate in aNa(V)1.5/lysine in nNa(V)1.5), that is, the charge reversal in the spliced region of the channel, played a key role in the selectivity, especially in antibody binding. Conclusion and Implications We conclude that the cancer-related nNa(V)1.5 channel can be distinguished pharmacologically from its nearest neighbour, aNa(V)1.5 channels. Thus, it may be possible to design low MW compounds as antimetastatic drugs for non-toxic therapy of nNa(V)1.5-expressing carcinomas.
- Published
- 2021
12. Role of Na V 1.7 in action potential conduction along human bronchial vagal afferent C‐fibres
- Author
-
Nikoleta Pavelkova, Marian Kollarik, Bradley J. Undem, John C. Hunter, John Mulcahy, and Fei Ru
- Subjects
Pharmacology ,Contraction (grammar) ,Chemistry ,Sodium channel ,Vagus nerve ,Compound muscle action potential ,chemistry.chemical_compound ,Tetrodotoxin ,Reflex ,medicine ,Cholinergic ,Bronchoconstriction ,medicine.symptom - Abstract
BACKGROUND AND PURPOSE The purpose of this study was to determine the role of NaV 1.7 in action potential conduction in C-fibres in the bronchial branches of the human vagus nerve. EXPERIMENTAL APPROACH Bronchial branches of the vagus nerve were dissected from human donor tissue. The C-wave of the electrically evoked compound action potential was quantified in the absence and presence of increasing concentrations of the selective NaV 1.7 blocking drugs, PF-05089771 and ST-2262, as well as the NaV 1.1, 1.2, and 1.3 blocking drug ICA121-431. The efficacy and potency of these inhibitors were compared to the standard NaV 1 blocker, tetrodotoxin. We then compared the relative potencies of the NaV 1 blockers in inhibiting the C-wave of the compound action potential, with their ability to inhibit parasympathetic cholinergic contraction of human isolated bronchi, a response previously shown to be strictly dependent on NaV 1.7 channels. KEY RESULTS The selective NaV 1.7 blockers inhibited the C-wave of the compound action potential with potencies similar to that observed in the NaV 1.7 bronchial contractions assay. Using rt-PCR, we noted that NaV 1.7 mRNA was strongly expressed and transported down the vagus nerve bundles. CONCLUSIONS AND IMPLICATIONS NaV 1.7 blockers can prevent action potential conduction in the majority of vagal C-fibres arising from human bronchi. Blockers of NaV 1.7 channels may therefore have value in inhibiting the responses to excessive airway C-fibre activation in inflammatory airway disease, responses that include coughing as well as reflex bronchoconstriction and secretions.
- Published
- 2021
13. The effect of sodium channels on neurological/neuronal disorders: A systematic review
- Author
-
Alireza Komaki, Shokufeh Bagheri, Rasool Haddadi, Sahar Saki, and Masoumeh Kourosh-Arami
- Subjects
Neurons ,Nervous system ,Epilepsy ,Multiple Sclerosis ,business.industry ,Multiple sclerosis ,Sodium channel ,Brain ,Voltage-Gated Sodium Channels ,Disease ,medicine.disease ,Electrophysiology ,medicine.anatomical_structure ,Sodium channel blocker ,Developmental Neuroscience ,medicine ,Humans ,business ,Neuroscience ,Ion channel ,Developmental Biology - Abstract
Neurological and neuronal disorders are associated with structural, biochemical, or electrical abnormalities in the nervous system. Many neurological diseases have not yet been discovered. Interventions used for the treatment of these disorders include avoidance measures, lifestyle changes, physiotherapy, neurorehabilitation, pain management, medication, and surgery. In the sodium channelopathies, alterations in the structure, expression, and function of voltage-gated sodium channels (VGSCs) are considered as the causes of neurological and neuronal diseases. Online databases, including Scopus, Science Direct, Google Scholar, and PubMed were assessed for studies published between 1977 and 2020 using the keywords of review, sodium channels blocker, neurological diseases, and neuronal diseases. VGSCs consist of one α subunit and two β subunits. These subunits are known to regulate the gating kinetics, functional characteristics, and localization of the ion channel. These channels are involved in cell migration, cellular connections, neuronal pathfinding, and neurite outgrowth. Through the VGSC, the action potential is triggered and propagated in the neurons. Action potentials are physiological functions and passage of impermeable ions. The electrophysiological properties of these channels and their relationship with neurological and neuronal disorders have been identified. Subunit mutations are involved in the development of diseases, such as epilepsy, multiple sclerosis, autism, and Alzheimer's disease. Accordingly, we conducted a review of the link between VGSCs and neurological and neuronal diseases. Also, novel therapeutic targets were introduced for future drug discoveries.
- Published
- 2021
14. Bioisosteric Modification of To042: Synthesis and Evaluation of Promising Use‐Dependent Inhibitors of Voltage‐Gated Sodium Channels
- Author
-
Antonio Carrieri, Alessia Carocci, Elisabetta Casalino, Marilena Muraglia, Carlo Franchini, Filomena Corbo, Maria Maddalena Cavalluzzi, Mariagrazia Perrone, Jean-François Desaphy, Nicola Antonio Colabufo, Antonella Santoro, Gualtiero Milani, Giovanni Lentini, Concetta Altamura, and Isabella Pisano
- Subjects
Pharmacology ,Molecular model ,Stereochemistry ,Sodium channel ,Organic Chemistry ,HEK 293 cells ,Ligand (biochemistry) ,Biochemistry ,In vitro ,chemistry.chemical_compound ,Sodium channel blocker ,Membrane ,chemistry ,Drug Discovery ,Molecular Medicine ,General Pharmacology, Toxicology and Pharmaceutics ,Lead compound - Abstract
Three analogues of To042, a tocainide-related lead compound recently reported for the treatment of myotonia, were synthesized and evaluated in vitro as skeletal muscle sodium channel blockers possibly endowed with enhanced use-dependent behavior. Patch-clamp experiments on hNav1.4 expressed in HEK293 cells showed that N -[(naphthalen-1-yl)methyl]-4-[(2,6-dimethyl)phenoxy]butan-2-amine, the aryloxyalkyl bioisoster of To042, exerted a higher use-dependent block than To042 thus being able to preferentially block the channels in over-excited membranes while preserving healthy tissue function. It also showed the lowest active transport across BBB according to the results of P-glycoprotein (P-gp) interacting activity evaluation and the highest cytoprotective effect on HeLa cells. Quantum mechanical calculations and dockings gave insights on the most probable conformation of the aryloxyalkyl bioisoster of To042 in solution and the target residues involved in the binding, respectively. Both approaches indicated the conformations that might be both adopted in both the unbound and bound state of the ligand. Overall, N -[(naphthalen-1-yl)methyl]-4-[(2,6-dimethyl)phenoxy]butan-2-amine exhibits an interesting toxico-pharmacological profile and deserves further investigation.
- Published
- 2021
15. Scorpion venom heat‐resistant synthesized peptide ameliorates 6‐OHDA‐induced neurotoxicity and neuroinflammation: likely role of Na v 1.6 inhibition in microglia
- Author
-
Ao-Ran Sui, Donglai Li, Jie Zhao, Xue-Fei Wu, Xiujie Li, Khizar Khan, Na Li, Shao Li, Sheng Li, and Bi-Ying Ge
- Subjects
0301 basic medicine ,Pharmacology ,Parkinson's disease ,Microglia ,Chemistry ,Sodium channel ,Neurotoxicity ,Inflammation ,medicine.disease ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,nervous system ,Dopamine ,medicine ,Neuron ,medicine.symptom ,030217 neurology & neurosurgery ,Neuroinflammation ,medicine.drug - Abstract
BACKGROUND AND PURPOSE Microglia-related inflammation is associated with the pathology of Parkinson's disease. Functional voltage-gated sodium channels (VGSCs) are involved in regulating microglial function. Here, we aim to investigate the effects of scorpion venom heat-resistant synthesized peptide (SVHRSP) on 6-hydroxydopamine (6-OHDA)-induced Parkinson's disease-like mouse model and reveal its underlying mechanism. EXPERIMENTAL APPROACH Unilateral brain injection of 6-OHDA was performed to establish Parkinson's disease mouse model. After behaviour test, brain tissues were collected for morphological analysis and protein/gene expression examination. Primary microglia culture was used to investigate the role of sodium channel Nav 1.6 in the regulation of microglia inflammation by SVHRSP. KEY RESULTS SVHRSP treatment attenuated motor deficits, dopamine neuron degeneration, activation of glial cells and expression of pro-inflammatory cytokines induced by 6-OHDA lesion. Primary microglia activation and the production of pro-inflammatory cytokines induced by lipopolysaccharide (LPS) were also suppressed by SVHRSP treatment. In addition, SVHRSP could inhibit mitogen-activated protein kinases (MAPKs) pathway, which plays pivotal roles in the pro-inflammatory response. Notably, SVHRSP treatment suppressed the overexpression of microglial Nav 1.6 induced by 6-OHDA and LPS. Finally, it was shown that the anti-inflammatory effect of SVHRSP in microglia was Nav 1.6 dependent and was related to suppression of sodium current and probably the consequent Na+ /Ca2+ exchange. CONCLUSIONS AND IMPLICATIONS SVHRSP might inhibit neuroinflammation and protect dopamine neurons via down-regulating microglial Nav 1.6 and subsequently suppressing intracellular Ca2+ accumulation to attenuate the activation of MAPKs signalling pathway in microglia.
- Published
- 2021
16. Cardiac ion channel expression in the equine model ‐ In‐silico prediction utilising RNA sequencing data from mixed tissue samples
- Author
-
Premont, Antoine, Saadeh, Khalil, Edling, Charlotte, Lewis, Rebecca, Marr, Celia M, Jeevaratnam, Kamalan, Jeevaratnam, Kamalan [0000-0002-6232-388X], and Apollo - University of Cambridge Repository
- Subjects
Sequence Analysis, RNA ,Physiology ,calcium-handling proteins ,cardiomyocyte ,Arrhythmias, Cardiac ,equine cardiac electrophysiology ,Ion Channels ,in‐silico prediction ,cardiac arrhythmia ,transcriptomics ,in-silico prediction ,Physiology (medical) ,calcium‐handling proteins ,Animals ,Humans ,RNA ,Calcium ,Myocytes, Cardiac ,Horses ,ORIGINAL ARTICLES ,ORIGINAL ARTICLE ,sodium channel - Abstract
Funder: University of Surrey; Id: http://dx.doi.org/10.13039/501100003513, Understanding cardiomyocyte ion channel expression is crucial to understanding normal cardiac electrophysiology and underlying mechanisms of cardiac pathologies particularly arrhythmias. Hitherto, equine cardiac ion channel expression has rarely been investigated. Therefore, we aim to predict equine cardiac ion channel gene expression. Raw RNAseq data from normal horses from 9 datasets was retrieved from ArrayExpress and European Nucleotide Archive and reanalysed. The normalised (FPKM) read counts for a gene in a mix of tissue were hypothesised to be the average of the expected expression in each tissue weighted by the proportion of the tissue in the mix. The cardiac-specific expression was predicted by estimating the mean expression in each other tissues. To evaluate the performance of the model, predicted gene expression values were compared to the human cardiac gene expression. Cardiac-specific expression could be predicted for 91 ion channels including most expressed Na+ channels, K+ channels and Ca2+ -handling proteins. These revealed interesting differences from what would be expected based on human studies. These differences included predominance of NaV 1.4 rather than NaV 1.5 channel, and RYR1, SERCA1 and CASQ1 rather than RYR2, SERCA2, CASQ2 Ca2+ -handling proteins. Differences in channel expression not only implicate potentially different regulatory mechanisms but also pathological mechanisms of arrhythmogenesis.
- Published
- 2022
17. A Low‐Frequency Pulsed Magnetic Field Reduces Neuropathic Pain by Regulating NaV 1.8 and NaV 1.9 Sodium Channels at the Transcriptional Level in Diabetic Rats
- Author
-
Cagil Coskun, Ismail Gunay, and Isil Ocal
- Subjects
Control level ,Physiology ,business.industry ,Sodium channel ,Biophysics ,020206 networking & telecommunications ,02 engineering and technology ,General Medicine ,Low frequency ,Pain management ,Pharmacology ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,SCN3A ,0302 clinical medicine ,Downregulation and upregulation ,Gene expression ,Neuropathic pain ,0202 electrical engineering, electronic engineering, information engineering ,Medicine ,Radiology, Nuclear Medicine and imaging ,business - Abstract
Low-frequency pulsed magnetic field (LF-PMF) application is a non-invasive, easy, and inexpensive treatment method in pain management. However, the molecular mechanism underlying the effect of LF-PMF on pain is not fully understood. Considering the obvious dysregulations of gene expression observed in certain types of voltage-gated sodium channels (VGSCs) in pain conditions, the present study tested the hypothesis that LF-PMF shows its pain-relieving effect by regulating genes that code VGSCs proteins. Five experimental rat groups (Control, Streptozotocin-induced experimental painful diabetic neuropathy (PDN), PDN Sham, PDN 10 Hz PMF, and PDN 30 Hz PMF) were established. After the pain formation in PDN groups, the magnetic field groups were exposed to 10/30 Hz, 1.5 mT PMF for 4 weeks, an hour daily. Progression of pain was evaluated using behavioral pain tests during the entire experimental processes. After the end of PMF treatment, SCN9A (NaV1.7 ), SCN10A (NaV1.8 ), SCN11A (NaV1.9 ), and SCN3A (NaV1.3 ) gene expression level changes were determined by analyzing real-time polymerase chain reaction results. We found that 10 Hz PMF application was more effective than 30 Hz on pain management. In addition, NaV1.7 and NaV1.3 transcriptions were upregulated while NaV1.8 and NaV1.9 were downregulated in painful conditions. Notably, the downregulated expression of the genes encoding NaV1.8 and NaV1.9 were re-regulated and increased to control level by 10 Hz PMF application. Consequently, it may be deduced that 10 Hz PMF application reduces pain by modulating certain VGSCs at the transcriptional level. © 2021 Bioelectromagnetics Society.
- Published
- 2021
18. Fluorescent‐ and tagged‐protoxin II peptides: potent markers of the Na v 1.7 channel pain target
- Author
-
Claude Zoukimian, Jérôme Montnach, Patrick Delmas, Nancy Osorio, Michel De Waard, Céline Marionneau, Sophie Burel, Massimo Mantegazza, Rachid Boukaiba, Sébastien Nicolas, Rémy Béroud, Michel Partiseti, Stephan De Waard, Unité de recherche de l'institut du thorax (ITX-lab), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Laboratoire de Neurosciences Cognitives [Marseille] (LNC), Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Smartox Biotechnology, Institut de pharmacologie moléculaire et cellulaire (IPMC), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), Sanofi-Aventis R&D, SANOFI Recherche, ANR-16-CE92-0013,Progress DHF,Mécanismes de la progression de la dysfonction diastolique vers l'insuffisance cardiaque(2016), unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)
- Subjects
0301 basic medicine ,Gene isoform ,pain target ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Protoxin II ,Peptide ,Gating ,03 medical and health sciences ,0302 clinical medicine ,Automated patch clamp ,voltage-gated sodium channel ,automated patchclamp ,Receptor ,Nav1.7 ,cellular distribution ,Pharmacology ,chemistry.chemical_classification ,pull-down ,Sodium channel ,cell line ,030104 developmental biology ,chemistry ,Cell culture ,Biotinylation ,biotinylated analogue ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Biophysics ,fluorescent analogue ,030217 neurology & neurosurgery - Abstract
Background and purpose Protoxin II (ProTx II) is a high affinity gating modifier that is thought to selectively block the Nav 1.7 voltage-dependent Na+ channel, a major therapeutic target for the control of pain. We aimed at producing ProTx II analogues entitled with novel functionalities for cell distribution studies and biochemical characterization of its Nav channel targets. Experimental approach We took advantage of the high affinity properties of the peptide, combined to its slow off rate, to design a number of new tagged analogues useful for imaging and biochemistry purposes. We used high-throughput automated patch-clamp to identify the analogues best matching the native properties of ProTx II and validated them on various Nav -expressing cells in pull-down and cell distribution studies. Key results Two of the produced ProTx II analogues, Biot-ProTx II and ATTO488-ProTx II, best recapitulate the pharmacological properties of unlabelled ProTx II, while other analogues remain high affinity blockers of Nav 1.7. The biotinylated version of ProTx II efficiently works for the pull-down of several Nav isoforms tested in a concentration-dependent manner, while the fluorescent ATTO488-ProTx II specifically labels the Nav 1.7 channel over other Nav isoforms tested in various experimental conditions. Conclusions and implications The properties of these ProTx II analogues as tools for Nav channel purification and cell distribution studies pave the way for a better understanding of ProTx II channel receptors in pain and their pathophysiological implications in sensory neuronal processing. The new fluorescent ProTx II should also reveal itself useful for the design of new drug screening strategies.
- Published
- 2021
19. Early onset epilepsy and sudden unexpected death in epilepsy with cardiac arrhythmia in mice carrying the early infantile epileptic encephalopathy 47 gain‐of‐function FHF1(FGF12) missense mutation
- Author
-
Glenn I. Fishman, David S. Park, Mitchell Goldfarb, Ying Xie, Yue Liu, Akshay Shekhar, Vasilisa Iatckova, Christopher Marra, Libor Velíšek, and Jana Velíšková
- Subjects
0301 basic medicine ,Bradycardia ,medicine.medical_specialty ,Genotype ,Mutation, Missense ,Oligonucleotides ,Mice, Transgenic ,Voltage-Gated Sodium Channels ,Electroencephalography ,Article ,Electrocardiography ,Mice ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Seizures ,Internal medicine ,Heart rate ,medicine ,Animals ,Humans ,Missense mutation ,Age of Onset ,Sudden Unexpected Death in Epilepsy ,medicine.diagnostic_test ,business.industry ,Sodium channel ,Cardiac arrhythmia ,Arrhythmias, Cardiac ,medicine.disease ,Penetrance ,Fibroblast Growth Factors ,030104 developmental biology ,Endocrinology ,Animals, Newborn ,Neurology ,Epilepsy, Tonic-Clonic ,Neurology (clinical) ,CRISPR-Cas Systems ,medicine.symptom ,business ,Spasms, Infantile ,030217 neurology & neurosurgery - Abstract
Objective Fibroblast growth factor homologous factors (FHFs) are brain and cardiac sodium channel-binding proteins that modulate channel density and inactivation gating. A recurrent de novo gain-of-function missense mutation in the FHF1(FGF12) gene (p.Arg52His) is associated with early infantile epileptic encephalopathy 47 (EIEE47; Online Mendelian Inheritance in Man database 617166). To determine whether the FHF1 missense mutation is sufficient to cause EIEE and to establish an animal model for EIEE47, we sought to engineer this mutation into mice. Methods The Arg52His mutation was introduced into fertilized eggs by CRISPR (clustered regularly interspaced short palindromic repeats) editing to generate Fhf1R52H /F+ mice. Spontaneous epileptiform events in Fhf1R52H /+ mice were assessed by cortical electroencephalography (EEG) and video monitoring. Basal heart rhythm and seizure-induced arrhythmia were recorded by electrocardiography. Modulation of cardiac sodium channel inactivation by FHF1BR52H protein was assayed by voltage-clamp recordings of FHF-deficient mouse cardiomyocytes infected with adenoviruses expressing wild-type FHF1B or FHF1BR52H protein. Results All Fhf1R52H /+ mice experienced seizure or seizurelike episodes with lethal ending between 12 and 26 days of age. EEG recordings in 19-20-day-old mice confirmed sudden unexpected death in epilepsy (SUDEP) as severe tonic seizures immediately preceding loss of brain activity and death. Within 2-53 s after lethal seizure onset, heart rate abruptly declined from 572 ± 16 bpm to 108 ± 15 bpm, suggesting a parasympathetic surge accompanying seizures that may have contributed to SUDEP. Although ectopic overexpression of FHF1BR52H in cardiomyocytes induced a 15-mV depolarizing shift in voltage of steady-state sodium channel inactivation and slowed the rate of channel inactivation, heart rhythm was normal in Fhf1R52H /+ mice prior to seizure. Significance The Fhf1 missense mutation p.Arg52His induces epileptic encephalopathy with full penetrance in mice. Both Fhf1 (p.Arg52His) and Scn8a (p.Asn1768Asp) missense mutations enhance sodium channel Nav 1.6 currents and induce SUDEP with bradycardia in mice, suggesting an FHF1/Nav 1.6 functional axis underlying altered brain sodium channel gating in epileptic encephalopathy.
- Published
- 2021
20. Structural Basis for Pore Blockade of the Human Cardiac Sodium Channel Na v 1.5 by the Antiarrhythmic Drug Quinidine**
- Author
-
Nieng Yan, Jianlin Lei, Zhangqiang Li, Gaoxingyu Huang, Xiaojing Pan, Tong Wu, Kun Wu, and Xueqin Jin
- Subjects
Quinidine ,Drug ,biology ,010405 organic chemistry ,Chemistry ,media_common.quotation_subject ,Sodium channel ,General Chemistry ,General Medicine ,Nav1.5 ,Pharmacology ,010402 general chemistry ,01 natural sciences ,Catalysis ,0104 chemical sciences ,Blockade ,Mechanism of action ,Biophysics ,biology.protein ,medicine ,medicine.symptom ,Flecainide ,Intracellular ,medicine.drug ,media_common - Abstract
Na v 1.5, the primary voltage-gated Na + (Na v ) channel in heart, is a major target for class I antiarrhythmic agents. Here we present the cryo-EM structure of full-length human Na v 1.5 bound to quinidine, a class Ia antiarrhythmic drug, at 3.3 A resolution. Quinidine is positioned right beneath the selectivity filter in the pore domain and coordinated by residues from repeats I, III, and IV. Pore blockade by quinidine is achieved through both direct obstruction of the ion permeation path and induced rotation of an invariant Tyr residue that tightens the intracellular gate. Structural comparison with a truncated rat Na v 1.5 in the presence of flecainide, a class Ic agent, reveals distinct binding poses for the two antiarrhythmics within the pore domain. Our work reported here, along with previous studies, reveals the molecular basis for the mechanism of action of class I antiarrhythmic drugs.
- Published
- 2021
21. A first, naturally occurring substitution at the second pyrethroid receptor of voltage‐gated sodium channel of <scp> Aedes aegypti </scp>
- Author
-
Yoshihide Maekawa, Kyoko Sawabe, Kentaro Itokawa, José Luiz de Lima Filho, Osamu Komagata, Aki Takaoka, Shogo Furutani, Shinji Kasai, Luiz Carlos Alves, and Takashi Tomita
- Subjects
0106 biological sciences ,Insecticides ,knockdown resistance ,Voltage-Gated Sodium Channels ,Aedes aegypti ,Biology ,01 natural sciences ,Dengue fever ,Insecticide Resistance ,chemistry.chemical_compound ,pyrethroids ,Aedes ,Pyrethrins ,parasitic diseases ,medicine ,Animals ,Research Articles ,Genetics ,Pyrethroid ,Zika Virus Infection ,Haplotype ,Yellow fever ,Knockdown resistance ,Zika Virus ,General Medicine ,medicine.disease ,biology.organism_classification ,010602 entomology ,Mosquito control ,chemistry ,Insect Science ,Vector (epidemiology) ,Mutation ,Agronomy and Crop Science ,Brazil ,Research Article ,sodium channel ,010606 plant biology & botany - Abstract
BACKGROUND Aedes aegypti is a remarkably effective mosquito vector of epidemiologically important arboviral diseases including dengue fever, yellow fever and Zika. The present spread of resistance against pyrethroids, the primary insecticides used for mosquito control, in global populations of this species is of great concern. The voltage‐gated sodium channel (VGSC) in the nervous system is the known target site of pyrethroids in insects. Past studies have revealed several amino‐acid substitutions in this channel that confer pyrethroid resistance, which are known as knockdown resistance (kdr) mutations. RESULTS This study investigated a laboratory colony of Ae. aegypti, MCNaeg, established from larvae collected in Rio de Janeiro, Brazil in 2016. The MCNaeg colony showed strong resistance against pyrethroids without laboratory selection. Of the two VGSC gene haplotypes present within this colony, one harbored three known kdr mutations, V410L, V1016I, and F1534C, and the other harbored only the known F1534C mutation. In latter haplotype, we also found novel amino‐acid substations including V253F. Previous molecular modeling and electrophysiological studies suggest that this residue serves a pyrethroid‐sensing site in the second receptor, PyR2. Our genetical analysis showed that the haplotype harboring V253F and F1534C is associated with equal or slightly stronger resistance than the other triple kdr haplotype to both Type I and Type II pyrethroids. CONCLUSION The novel substitution V253F is potentially involved in pyrethroid resistance in Ae. aegypti. Further studies are needed to elucidate the role of this substitution in the pyrethroid susceptibility of VGSC. © 2021 The Authors. Pest Management Science published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry., We found a novel amino‐acid substitution V253F in the voltage‐gated sodium channel in Aedes aegypti which is potentially involved in pyrethroid resistance of this vector mosquito
- Published
- 2021
22. Voltage‐gated sodium channel‐dependent retroaxonal modulation of photoreceptor function during post‐natal development in mice
- Author
-
Patrice D. Côté, François Tremblay, and Benjamin J. Smith
- Subjects
Retinal Ganglion Cells ,0301 basic medicine ,genetic structures ,Voltage-Gated Sodium Channels ,Tropomyosin receptor kinase B ,Biology ,Retina ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,Neurotrophic factors ,Electroretinography ,medicine ,Animals ,Mice, Knockout ,Sodium channel ,Wild type ,eye diseases ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Retinal ganglion cell ,NAV1.6 Voltage-Gated Sodium Channel ,Axoplasmic transport ,Optic nerve ,sense organs ,030217 neurology & neurosurgery - Abstract
Juvenile (postnatal day 16) mice lacking Nav 1.6 channels (null-mutant Scn8admu ) have reduced photoreceptor function, which is unexpected given that Nav channels have not been detected in mouse photoreceptors and do not contribute appreciably to photoreceptor function in adults. We demonstrate that acute block of Nav channels with intravitreal TTX in juvenile (P16) wild-type mice has no effect on photoreceptor function. However, reduced light activity by prolonged dark adaptation from P8 caused significant reduction in photoreceptor function at P16. Injecting TTX into the retrobulbar space at P16 to specifically block Nav channels in the optic nerve also caused a reduction in photoreceptor function comparable to that seen at P16 in null-mutant Scn8a mice. In both P16 null-mutant Scn8admu and retrobulbar TTX-injected wild type mice, photoreceptor function was restored following intravitreal injection of the TrkB receptor agonist 7,8 dihydroxyflavone, linking Nav -dependent retrograde transport to TrkB-dependent neurotrophic factor production pathways as a modulatory influence of photoreceptor function at P16. We also found that in Scn8admu mice, photoreceptor function recovers by P22-25 despite more precarious general health of the animal. Retrobulbar injection of TTX in the wild type still reduced the photoreceptor response at this age but to a lesser extent, suggesting that Nav -dependent modulation of photoreceptor function is largely transient, peaking soon after eye opening. Together, these results suggest that the general photosensitivity of the retina is modulated following eye opening by retrograde transport through activity-dependent retinal ganglion cell axonal signaling targeting TrkB receptors.
- Published
- 2021
23. The mechanism of non‐blocking inhibition of sodium channels revealed by conformation‐selective photolabeling
- Author
-
Tamás Hegedűs, Katalin Zboray, Peter Lukacs, Arpad Mike, Krisztina Pesti, András Málnási-Csizmadia, Ádám Tóth, and Mátyás C. Földi
- Subjects
0301 basic medicine ,Drug ,media_common.quotation_subject ,Sodium Channels ,Membrane Potentials ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Binding site ,media_common ,Pharmacology ,Binding Sites ,Riluzole ,Mechanism (biology) ,Chemistry ,Sodium channel ,Resting potential ,Small molecule ,Electrophysiology ,HEK293 Cells ,030104 developmental biology ,Biophysics ,030217 neurology & neurosurgery ,Sodium Channel Blockers ,medicine.drug - Abstract
Background and purpose Sodium channel inhibitors can be used to treat hyperexcitability-related diseases, including epilepsies, pain syndromes, neuromuscular disorders and cardiac arrhythmias. The applicability of these drugs is limited by their nonspecific effect on physiological function. They act mainly by sodium channel block and in addition by modulation of channel kinetics. While channel block inhibits healthy and pathological tissue equally, modulation can preferentially inhibit pathological activity. An ideal drug designed to target the sodium channels of pathological tissue would act predominantly by modulation. Thus far, no such drug has been described. Experimental approach Patch-clamp experiments with ultra-fast solution exchange and photolabeling-coupled electrophysiology were applied to describe the unique mechanism of riluzole on Nav1.4 sodium channels. In silico docking experiments were used to study the molecular details of binding. Key results We present evidence that riluzole acts predominantly by non-blocking modulation. We propose that, being a relatively small molecule, riluzole is able to stay bound to the binding site, but nonetheless stay off the conduction pathway, by residing in one of the fenestrations. We demonstrate how this mechanism can be recognized. Conclusions and implications Our results identify riluzole as the prototype of this new class of sodium channel inhibitors. Drugs of this class are expected to selectively prevent hyperexcitability, while having minimal effect on cells firing at a normal rate from a normal resting potential.
- Published
- 2021
24. R1617Q epilepsy mutation slows Na V 1.6 sodium channel inactivation and increases the persistent current and neuronal firing
- Author
-
Mohamed Chahine and Hugo Poulin
- Subjects
0301 basic medicine ,Mutation ,Arginine ,Physiology ,Sodium channel ,Stimulation ,Hippocampal formation ,medicine.disease ,medicine.disease_cause ,Riluzole ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,Epilepsy ,030104 developmental biology ,0302 clinical medicine ,chemistry ,medicine ,Tetrodotoxin ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Key points A human NaV 1.6 construct was established to study the biophysical consequences of the R1617Q mutation on NaV 1.6 identified in patients with unclassified epileptic encephalopathy and severe intellectual disability. The R1617Q mutation disrupts the inactivation process of the channel, and more specifically, slows the current decay, increases the persistent sodium current that was blocked by tetrodotoxin and riluzole, and disrupts the inactivation voltage-dependence and increases the kinetics of recovery. In native hippocampal neurons, the R1617Q mutation exhibited a significant increase in action potentials triggered in response to stimulation and a significant increase in the number of neurons that exhibited spontaneous activity compared to neurons expressing WT channels that were inhibited by riluzole. The abnormally persistent current activity caused by the disruption of the channel inactivation process in NaV 1.6/R1617Q may result in epileptic encephalopathy in patients. Abstract The voltage-gated sodium channel NaV 1.6 is the most abundantly expressed sodium channel isoform in the central nervous system. It plays a critical role in saltatory and continuous conduction. Although over 40 NaV 1.6 mutations have been linked to epileptic encephalopathy, only a few have been functionally analysed. In the present study, we characterized a NaV 1.6 mutation (R1617Q) identified in patients with epileptic encephalopathy and intellectual disability. R1617Q substitutes an arginine for a glutamine in the S4 segment of domain IV, which plays a major role in coupling the activation and inactivation of sodium channels. We used patch-clamp to show that R1617Q is a gain-of-function mutation. It is typified by slower inactivation kinetics and a loss of inactivation of voltage-dependence, which result in a 2.5-fold increase in the window current. In addition, sodium currents exhibited an enhanced rate of recovery from inactivation, most likely due to the destabilization of the inactivation state. The alterations in the fast inactivation caused a significant increase in the persistent sodium current. Overexpression of R1617Q in rat hippocampal neurons resulted in an increase in action potential firing activity that was inhibited by riluzole, consistent with the gain-of-function observed. We conclude that the R1617Q mutation causes neuronal hyperexcitability and may result in epileptic encephalopathy.
- Published
- 2021
25. Complex effects of eslicarbazepine on inhibitory micro networks in chronic experimental epilepsy
- Author
-
Thoralf Opitz, Heinz Beck, Leonie Pothmann, Sarah Schmidt, Daniel Müller-Komorowska, and Patrício Soares da Silva
- Subjects
0301 basic medicine ,Long-Term Potentiation ,Adamantane ,AMPA receptor ,Muscarinic Agonists ,Hippocampal formation ,Inhibitory postsynaptic potential ,Hippocampus ,03 medical and health sciences ,0302 clinical medicine ,Dibenzazepines ,Interneurons ,medicine ,Animals ,Receptors, AMPA ,Receptor ,CA1 Region, Hippocampal ,Feedback, Physiological ,Neurons ,Epilepsy ,Neuronal Plasticity ,Dose-Response Relationship, Drug ,Chemistry ,Pyramidal Cells ,Sodium channel ,Pilocarpine ,Neural Inhibition ,Long-term potentiation ,Carbamazepine ,Rats ,Disease Models, Animal ,030104 developmental biology ,Inhibitory Postsynaptic Potentials ,Neurology ,Excitatory postsynaptic potential ,Anticonvulsants ,Calcium ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Objective Many antiseizure drugs (ASDs) act on voltage-dependent sodium channels, and the molecular basis of these effects is well established. In contrast, how ASDs act on the level of neuronal networks is much less understood. Methods In the present study, we determined the effects of eslicarbazepine (S-Lic) on different types of inhibitory neurons, as well as inhibitory motifs. Experiments were performed in hippocampal slices from both sham-control and chronically epileptic pilocarpine-treated rats. Results We found that S-Lic causes an unexpected reduction of feed-forward inhibition in the CA1 region at high concentrations (300 µM), but not at lower concentrations (100 µM). Concurrently, 300 but not 100 μM S-Lic significantly reduced maximal firing rates in putative feed-forward interneurons located in the CA1 stratum radiatum of sham-control and epileptic animals. In contrast, feedback inhibition was not inhibited by S-Lic. Instead, application of S-Lic, in contrast to previous data for other drugs like carbamazepine (CBZ), resulted in a lasting potentiation of feedback inhibitory post-synaptic currents (IPSCs) only in epileptic and not in sham-control animals, which persisted after washout of S-Lic. We hypothesized that this plasticity of inhibition might rely on anti-Hebbian potentiation of excitatory feedback inputs onto oriens-lacunosum moleculare (OLM) interneurons, which is dependent on Ca2+ -permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Indeed, we show that blocking Ca2+ -permeable AMPA receptors completely prevents upmodulation of feedback inhibition. Significance These results suggest that S-Lic affects inhibitory circuits in the CA1 hippocampal region in unexpected ways. In addition, ASD actions may not be sufficiently explained by acute effects on their target channels, rather, it may be necessary to take plasticity of inhibitory circuits into account.
- Published
- 2021
26. First report of voltage‐gated sodium channel M918V and molecular diagnostics of nicotinic acetylcholine receptor R81T in the cotton aphid
- Author
-
Xinghui Qiu, Mei Li, Otgonzaya Munkhbayar, and Nian Liu
- Subjects
Nicotinic acetylcholine receptor ,Aphid ,biology ,Insect Science ,Sodium channel ,Biophysics ,biology.organism_classification ,Molecular diagnostics ,Agronomy and Crop Science - Published
- 2020
27. Excitatory and inhibitory neuron defects in a mouse model of Scn1b‐linked EIEE52
- Author
-
Chunling Chen, Charles Anumonwo, Heather A. O'Malley, Alexandra A. Bouza, Yukun Yuan, Jacob M. Hull, Luis F. Lopez-Santiago, Nicholas Denomme, and Lori L. Isom
- Subjects
0301 basic medicine ,Interneuron ,EMX1 ,Cell Count ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Inhibitory postsynaptic potential ,03 medical and health sciences ,SCN3A ,Mice ,Mice, Congenic ,0302 clinical medicine ,SCN1B ,Interneurons ,CAMK2A ,Medicine ,Animals ,Humans ,RC346-429 ,Research Articles ,Cerebral Cortex ,business.industry ,General Neuroscience ,Sodium channel ,Pyramidal Cells ,Infant, Newborn ,Voltage-Gated Sodium Channel beta-1 Subunit ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Parvalbumins ,nervous system ,Excitatory postsynaptic potential ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,business ,Neuroscience ,Spasms, Infantile ,030217 neurology & neurosurgery ,Research Article ,RC321-571 - Abstract
Objective Human variants in voltage‐gated sodium channel (VGSC) α and β subunit genes are linked to developmental and epileptic encephalopathies (DEEs). Inherited, biallelic, loss‐of‐function variants in SCN1B, encoding the β1/β1B subunits, are linked to early infantile DEE (EIEE52). De novo, monoallelic variants in SCN1A (Nav1.1), SCN2A (Nav1.2), SCN3A (Nav1.3), and SCN8A (Nav1.6) are also linked to DEEs. While these VGSC‐linked DEEs have similar presentations, they have diverse mechanisms of altered neuronal excitability. Mouse models have suggested that Scn2a‐, Scn3a‐, and Scn8a‐linked DEE variants are, in general, gain of function, resulting in increased persistent or resurgent sodium current (INa) and pyramidal neuron hyperexcitability. In contrast, Scn1a‐linked DEE variants, in general, are loss‐of‐function, resulting in decreased INa and hypoexcitability of fast‐spiking interneurons. VGSC β1 subunits associate with Nav1.1, Nav1.2, Nav1.3, and Nav1.6 and are expressed throughout the brain, raising the possibility that insults to both pyramidal and interneuron excitability may drive EIEE52 pathophysiology. Methods We investigated excitability defects in pyramidal and parvalbumin‐positive (PV +) interneurons in the Scn1b −/− model of EIEE52. We also used Scn1bFL/FL mice to delete Scn1b in specific neuronal populations. Results Scn1b −/− cortical PV + interneurons were hypoexcitable, with reduced INa density. Scn1b −/− cortical pyramidal neurons had population‐specific changes in excitability and impaired INa density. Scn1b deletion in PV + neurons resulted in 100% lethality, whereas deletion in Emx1 + or Camk2a + neurons did not affect survival. Interpretation This work suggests that SCN1B‐linked DEE variants impact both excitatory and inhibitory neurons, leading to the increased severity of EIEE52 relative to other DEEs.
- Published
- 2020
28. Three‐dimensional neuron–astrocyte construction on matrigel enhances establishment of functional voltage‐gated sodium channels
- Author
-
Mohammad Mohammadi Aria, Emine Sekerdag, Yasemin Gursoy-Ozdemir, Ihsan Solaroglu, Sedat Nizamoglu, Sercin Karahuseyinoglu, and Yagmur Cetin Tas
- Subjects
0301 basic medicine ,Patch-Clamp Techniques ,Neurite ,Primary Cell Culture ,Voltage-Gated Sodium Channels ,Biochemistry ,law.invention ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Pregnancy ,Confocal microscopy ,law ,Neurites ,medicine ,Animals ,Patch clamp ,Cerebral Cortex ,Neurons ,Mice, Inbred BALB C ,Matrigel ,NAV1.2 Voltage-Gated Sodium Channel ,Chemistry ,Sodium channel ,Nervous tissue ,Embryo, Mammalian ,Electrophysiological Phenomena ,Cell biology ,Drug Combinations ,030104 developmental biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,nervous system ,NAV1.6 Voltage-Gated Sodium Channel ,Astrocytes ,Female ,Proteoglycans ,Collagen ,Laminin ,Neuron ,030217 neurology & neurosurgery ,Astrocyte - Abstract
This study aimed to investigate and compare cell growth manners and functional differences of primary cortical neurons cultured on either poly-d-lysine (PDL) and or Matrigel, to delineate the role of extracellular matrix on providing resemblance to in vivo cellular interactions in nervous tissue. Primary cortical neurons, obtained from embryonic day 15 mice pups, seeded either on PDL- or Matrigel-coated culture ware were investigated by DIC/bright field and fluorescence/confocal microscopy for their morphology, 2D and 3D structure, and distribution patterns. Patch clamp, western blot, and RT-PCR studies were performed to investigate neuronal firing thresholds and sodium channel subtypes Nav1.2 and Nav1.6 expression. Cortical neurons cultured on PDL coating possessed a 2D structure composed of a few numbers of branched and tortuous neurites that contacted with each other in one to one manner, however, neurons on Matrigel coating showed a more complicated dimensional network that depicted tight, linear axonal bundles forming a 3D interacted neuron-astrocyte construction. This difference in growth patterns also showed a significant alteration in neuronal firing threshold which was recorded between 80 Iinj 120 pA on PDL and 2 Iinj 160 pA on Matrigel. Neurons grown up on Matrigel showed increased levels of sodium channel protein expression of Nav1.2 and Nav1.6 compared to neurons on PDL. These results have demonstrated that a 3D interacted neuron-astrocyte construction on Matrigel enhances the development of Nav1.2 and Nav1.6 in vitro and decreases neuronal firing threshold by 40 times compared to conventional PDL, resembling in vivo neuronal networks and hence would be a better in vitro model of adult neurons.
- Published
- 2020
29. Action Potentials: Generation and Propagation
- Author
-
Mohamed A. Fouda and Peter C. Ruben
- Subjects
Membrane potential ,Voltage-gated ion channel ,Chemistry ,Sodium channel ,Biophysics ,Analytical chemistry ,Voltage-gated potassium channel ,Hyperpolarization (biology) ,Resting potential ,Ion channel ,Ventricular action potential - Abstract
All cells maintain a voltage across their plasma membranes. Only excitable cells, however, can generate action potentials, the rapid, transient changes in membrane potential that spread along the surface of these unique cells. Action potential generation and propagation occurs through, and is regulated by, the function of voltage-gated ion channels – proteins with ion-selective pores that span the cell membrane. Ion channels undergo changes in their structural conformation in response to changes in the electrical field across the membrane. These structural changes cause the opening of pores – channels – through which ions can flow down their electrochemical gradient. The charge carried by ions creates an electrical current and rapidly alters the membrane potential with time- and voltage-dependent properties. This rapid, transient membrane potential change is called the action potential. Action potentials transmit information within neurons, trigger contractions within muscle cells, and lead to exocytosis in secretory cells. Key Concepts: All cells maintain a voltage difference across their plasma membranes. Action potentials are all-or-nothing, transient changes in membrane potentials of electrically excitable cells that carry important cellular information. Influx of sodium ions through voltage-gated sodium channels is responsible for the upstroke of the action potential, whereas efflux of potassium ions through voltage-gated potassium channels is responsible for the falling phase. Propagation of action potentials depends on gating kinetics of ion channels and intracellular and membrane resistances. Keywords: membrane potential; ionic current; threshold; refractoriness; length constant
- Published
- 2020
30. Telethonin variants found in Brugada syndrome, J‐wave pattern ECG, and ARVC reduce peak Na v 1.5 currents in HEK‐293 cells
- Author
-
Minoru Horie, Masaru Yamakawa, Takeshi Ueyama, Peng Sheng Chen, Takeru Makiyama, Isik Turker, Matteo Vatta, Tomohiko Ai, and Akihiko Shimizu
- Subjects
medicine.medical_specialty ,030204 cardiovascular system & hematology ,Telethonin ,sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,ARVC ,Medicine ,030212 general & internal medicine ,telethonin ,J wave ,Brugada syndrome ,business.industry ,Brugada ,Sodium channel ,HEK 293 cells ,General Medicine ,Transfection ,medicine.disease ,Phenotype ,Electrophysiology ,Endocrinology ,J-wave syndromes ,Cardiology and Cardiovascular Medicine ,business ,arrhythmias - Abstract
Background:Telethonin (TCAP) is a Z-disk protein that maintains cytoskeletal integrity and various signaling pathways in cardiomyocytes. TCAP is shown to modulate α-subunit of the human cardiac sodium channel (hNav 1.5) by direct interactions. Several TCAP variants are found in cardiomyopathies. We sought to investigate whether TCAP variants are associated with arrhythmia syndromes., Methods:Mutational analyses for TCAP were performed in 303 Japanese patients with Brugada syndrome, arrhythmogenic right ventricular cardiomyopathy, and J-wave pattern ECG. Using patch-clamp techniques, electrophysiological characteristics of hNav 1.5 were studied in HEK-293 cells stably expressing hNav 1.5 and transiently transfected with wild-type (WT) or variant TCAP., Results:We identified two TCAP variants, c.145G>A:p.E49K and c.458G>A:p.R153H, in four individuals. p.E49K was found in two patients with ARVC or BrS. p.R153H was found in two patients with BrS or J-wave pattern ECG. No patient had variant hNav 1.5. Patch-clamp experiments demonstrated that peak sodium currents were significantly reduced in cells expressing p.R153H and p.E49K compared with WT-TCAP (66%, p.R153H; 72%, p.E49K). Voltage dependency of peak IV curve was rightward-shifted by 5 mV in cells expressing p.E49K compared with WT-TCAP. Voltage dependency of activation was not leftward-shifted by p.R153H, while voltage dependency of steady-state inactivation was leftward-shifted by p.E49K., Conclusions:We found two TCAP variants in the patients with BrS, J-wave pattern ECG, and ARVC that can cause loss-of-function of the hNav 1.5 in heterologous expression systems. Our observation suggests that these variants might impair INa and be associated with the patients' electrophysiological phenotypes. Further studies linking our experimental data to clinical phenotypes are warranted.
- Published
- 2020
31. Sodium channel epilepsies and neurodevelopmental disorders: from disease mechanisms to clinical application
- Author
-
Andreas Brunklaus and Dennis Lal
- Subjects
030506 rehabilitation ,Brain development ,Gene Expression ,Biology ,03 medical and health sciences ,SCN3A ,symbols.namesake ,Epilepsy ,0302 clinical medicine ,Developmental Neuroscience ,medicine ,Humans ,Genetic heterogeneity ,Sodium channel ,Disease mechanisms ,Precision medicine ,medicine.disease ,Electrophysiological Phenomena ,NAV1.1 Voltage-Gated Sodium Channel ,Neurodevelopmental Disorders ,Pediatrics, Perinatology and Child Health ,Mendelian inheritance ,symbols ,Neurology (clinical) ,0305 other medical science ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Genetic variants in brain-expressed voltage-gated sodium channels (SCNs) have emerged as one of the most frequent causes of Mendelian forms of epilepsy and neurodevelopmental disorders (NDDs). This review explores the biological concepts that underlie sodium channel NDDs, explains their phenotypic heterogeneity, and appraises how this knowledge may inform clinical practice. We observe that excitatory/inhibitory neuronal expression ratios of sodium channels are important regulatory mechanisms underlying brain development, homeostasis, and neurological diseases. We hypothesize that a detailed understanding of gene expression, variant tolerance, location, and function, as well as timing of seizure onset can aid the understanding of how variants in SCN1A, SCN2A, SCN3A, and SCN8A contribute to seizure aetiology and inform treatment choice. We propose a model in which variant type, development-specific gene expression, and functions of SCNs explain the heterogeneity of sodium channel associated NDDs. Understanding of basic disease mechanisms and detailed knowledge of variant characteristics have increasing influence on clinical decision making, enabling us to stratify treatment and move closer towards precision medicine in sodium channel epilepsy and NDDs. WHAT THIS PAPER ADDS: Sodium-channel disorder heterogeneity is explained by variant-specific gene expression timing and function. Gene tolerance and location analyses aid sodium channel variant interpretation. Sodium-channel variant characteristics can contribute to clinical decision making.
- Published
- 2020
32. Identification of transmembrane protein 168 mutation in familial Brugada syndrome
- Author
-
Satoru Yamazaki, Akio Shimizu, Yoshihiro Asano, Naomasa Makita, Toshinori Makita, Taichi Noda, Masahito Ikawa, Minoru Horie, Hiroshi Matsuura, Hiroshi Morita, Taisuke Ishikawa, Hisakazu Ogita, Seiji Takashima, Dimitar P. Zankov, Yohei Miyashita, Akira Sato, Masahiro Komeno, Futoshi Toyoda, Seiko Ohno, and Masahito Hitosugi
- Subjects
Adult ,Male ,0301 basic medicine ,Mutant ,Biology ,ubiquitination ,medicine.disease_cause ,Biochemistry ,NAV1.5 Voltage-Gated Sodium Channel ,Mice ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Channelopathy ,Genetics ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Myocytes, Cardiac ,Molecular Biology ,Gene ,Exome sequencing ,Brugada Syndrome ,Brugada syndrome ,Mutation ,Sodium channel ,Membrane Proteins ,medicine.disease ,Molecular biology ,Transmembrane protein ,Pedigree ,030104 developmental biology ,fatal ventricular arrhythmia ,Female ,030217 neurology & neurosurgery ,sodium channel ,Biotechnology - Abstract
Brugada syndrome (BrS) is an inherited channelopathy responsible for almost 20% of sudden cardiac deaths in patients with nonstructural cardiac diseases. Approximately 70% of BrS patients, the causative gene mutation(s) remains unknown. In this study, we used whole exome sequencing to investigate candidate mutations in a family clinically diagnosed with BrS. A heterozygous 1616G>A substitution (R539Q mutation) was identified in the transmembrane protein 168 (TMEM168) gene of symptomatic individuals. Similar to endogenous TMEM168, both TMEM168 wild-type (WT) and mutant proteins that were ectopically induced in HL-1 cells showed nuclear membrane localization. A significant decrease in Na+ current and Nav 1.5 protein expression was observed in HL-1 cardiomyocytes expressing mutant TMEM168. Ventricular tachyarrhythmias and conduction disorders were induced in the heterozygous Tmem168 1616G>A knock-in mice by pharmacological stimulation, but not in WT mice. Na+ current was reduced in ventricular cardiomyocytes isolated from the Tmem168 knock-in heart, and Nav 1.5 expression was also impaired. This impairment was dependent on increased Nedd4-2 binding to Nav 1.5 and subsequent ubiquitination. Collectively, our results show an association between the TMEM168 1616G>A mutation and arrhythmogenesis in a family with BrS.
- Published
- 2020
33. New insights into the early mechanisms of epileptogenesis in a zebrafish model of Dravet syndrome
- Author
-
Suresh Poovathingal, Kinga Gawel, Ju Xu, Maria Lorena Cordero-Maldonado, Kamil Grzyb, Wietske van der Ent, Ettore Tiraboschi, Silvia Martina, Jarle Brattespe, Somisetty V. Satheesh, Maximiliano L. Suster, Sarah Heintz, Camila V. Esguerra, and Alexander Skupin
- Subjects
0301 basic medicine ,Epilepsies, Myoclonic ,Epileptogenesis ,0302 clinical medicine ,Gliosis ,RNA-Seq ,GABAergic Neurons ,NEURONS ,Zebrafish ,education.field_of_study ,Neuronal Plasticity ,SEVERE MYOCLONIC EPILEPSY ,Brain ,Electroencephalography ,MOUSE MODEL ,Astrogliosis ,Neurology ,NEUROPEPTIDE-Y ,GABAergic ,Anticonvulsants ,Single-Cell Analysis ,Life Sciences & Biomedicine ,Locomotion ,sodium channel ,medicine.medical_specialty ,Population ,Mutation, Missense ,Clinical Neurology ,Biology ,Real-Time Polymerase Chain Reaction ,Inhibitory postsynaptic potential ,03 medical and health sciences ,Glutamatergic ,Dravet syndrome ,GABAERGIC INTERNEURONS ,Internal medicine ,Fenfluramine ,OSCILLATIONS ,medicine ,Animals ,education ,Cell Proliferation ,Diazepam ,Science & Technology ,Gene Expression Profiling ,SOMATOSTATIN ,Zebrafish Proteins ,zebrafish ,medicine.disease ,biology.organism_classification ,NAV1.1 Voltage-Gated Sodium Channel ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,DENDRITIC ARBORIZATION ,HIPPOCAMPUS ,epileptogenesis ,fenfluramine ,Neurosciences & Neurology ,Neurology (clinical) ,CRISPR-Cas Systems ,Serotonin 5-HT2 Receptor Agonists ,030217 neurology & neurosurgery - Abstract
Objective: To pinpoint the earliest cellular defects underlying seizure onset (epileptogenic period) during perinatal brain development in a new zebrafish model of Dravet syndrome (DS) and to investigate potential disease-modifying activity of the 5HT2 receptor agonist fenfluramine. Methods: We used CRISPR/Cas9 mutagenesis to introduce a missense mutation, designed to perturb ion transport function in all channel isoforms, into scn1lab, the zebrafish orthologue of SCN1A (encoding voltage-gated sodium channel alpha subunit 1). We performed behavioral analysis and electroencephalographic recordings to measure convulsions and epileptiform discharges, followed by single-cell RNA-Seq, morphometric analysis of transgenic reporter-labeled γ-aminobutyric acidergic (GABAergic) neurons, and pharmacological profiling of mutant larvae. Results: Homozygous mutant (scn1labmut/mut ) larvae displayed spontaneous seizures with interictal, preictal, and ictal discharges (mean = 7.5 per 20-minute recording; P < .0001; one-way analysis of variance). Drop-Seq analysis revealed a 2:1 shift in the ratio of glutamatergic to GABAergic neurons in scn1labmut/mut larval brains versus wild type (WT), with dynamic changes in neuronal, glial, and progenitor cell populations. To explore disease pathophysiology further, we quantified dendritic arborization in GABAergic neurons and observed a 40% reduction in arbor number compared to WT (P < .001; n = 15 mutant, n = 16 WT). We postulate that the significant reduction in inhibitory arbors causes an inhibitory to excitatory neurotransmitter imbalance that contributes to seizures and enhanced electrical brain activity in scn1labmut/mut larvae (high-frequency range), with subsequent GABAergic neuronal loss and astrogliosis. Chronic fenfluramine administration completely restored dendritic arbor numbers to normal in scn1labmut/mut larvae, whereas similar treatment with the benzodiazepine diazepam attenuated seizures, but was ineffective in restoring neuronal cytoarchitecture. BrdU labeling revealed cell overproliferation in scn1labmut/mut larval brains that were rescued by fenfluramine but not diazepam. Significance: Our findings provide novel insights into early mechanisms of DS pathogenesis, describe dynamic cell population changes in the scn1labmut/mut brain, and present first-time evidence for potential disease modification by fenfluramine. Keywords: Dravet syndrome; epileptogenesis; fenfluramine; sodium channel; zebrafish. publishedVersion
- Published
- 2020
34. Molecular characterization and functional expression of voltage‐gated sodium channel variants inApolygus lucorum(<scp>Meyer‐Dür</scp>)
- Author
-
Wenbo Duan, Qiang Wang, Mengli Chen, Elizabeth Bandason, Shaoying Wu, Denghui Deng, Hao Wang, Kun Zhang, and Fen Li
- Subjects
0106 biological sciences ,China ,Population ,Voltage-Gated Sodium Channels ,01 natural sciences ,Heteroptera ,Exon ,Pyrethrins ,Animals ,Coding region ,education ,Gene ,Apolygus lucorum ,Genetics ,education.field_of_study ,biology ,Sodium channel ,Sodium ,Alternative splicing ,General Medicine ,biology.organism_classification ,010602 entomology ,RNA editing ,Insect Science ,Agronomy and Crop Science ,010606 plant biology & botany - Abstract
BACKGROUND Apolygus lucorum (Meyer-Dur) is a serious worldwide agricultural pest, especially for Bt cotton in China. Pyrethroids, neonicotinoids and organophosphates are the most effective insecticides to control piercing and sucking insects, including A. lucorum. The voltage-gated sodium channel (Nav ) is major target site of pyrethroids. Extensive alternative splicing and RNA editing, two major post-transcriptional mechanisms, contribute to generate different functional sodium channel variants. In our research, we characterized the sodium channel variants of A. lucorum. RESULTS In this study, we isolated numerous sodium channel variants that cover the entire coding region of the VGSC gene from A. lucorum. All clones could be grouped into 47 splice types based on the presence of nine alternative exons (exons j, n, o, a, p, b, s, q and t). Exons j, b and t were located independently, while exons n, o, a and p were located adjacently, as were exons s and q. We also found 35 nucleotide changes in different positions in individual variants, of which 18 nucleotide changes were A-to-I RNA editing, 11 nucleotide changes were likely due to U-to-C or C-to-U editing, and the others were likely natural sequence polymorphisms in the population. Furthermore, we expressed all of the variants in Xenopus oocytes. Eighteen of them were expressed in oocytes and sensitive to tetrodotoxin. CONCLUSION Our results provide a functional basis for understanding how A. lucorum sodium channel variants work in regulating channel expression, pharmacology and gating properties for agricultural insects. Apolygus lucorum is widely distributed in cotton production. Our results suggest how AlNav (the sodium channel of A.lucorum) variants work in regulating channel expression, pharmacology and sodium channel gating for agricultural insects in the future. © 2020 Society of Chemical Industry.
- Published
- 2020
35. The sodium channel Na X : Possible player in excitation–contraction coupling
- Author
-
J. A. Wasserstrom, Robert D. Galiano, Hari Iyer, Kai P. Leung, Alfred L. George, Thomas A. Mustoe, Elena Bogdanovic, William Marszalec, Seok Jong Hong, and Franck Potet
- Subjects
0301 basic medicine ,Sarcolemma ,biology ,Voltage-gated ion channel ,Chemistry ,Sodium channel ,Clinical Biochemistry ,Cardiac muscle ,Skeletal muscle ,Depolarization ,Cell Biology ,Nav1.5 ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Genetics ,Biophysics ,medicine ,biology.protein ,Myocyte ,Molecular Biology - Abstract
The sodium channel NaX (encoded by the SCN7A gene) was originally identified in the heart and skeletal muscle and is structurally similar to the other voltage-gated sodium channels but does not appear to be voltage gated. Although NaX is expressed at high levels in cardiac and skeletal muscle, little information exists on the function of NaX in these tissues. Transcriptional profiling of ion channels in the heart in a subset of patients with Brugada syndrome revealed an inverse relationship between the expression of NaX and NaV 1.5 suggesting that, in cardiac myocytes, the expression of these channels may be linked. We propose that NaX plays a role in excitation-contraction coupling based on our experimental observations. Here we show that in cardiac myocytes, NaX is expressed in a striated pattern on the sarcolemma in regions corresponding to the sarcomeric M-line. Knocking down NaX expression decreased NaV 1.5 mRNA and protein and reduced the inward sodium current (INa+ ) following cell depolarization. When the expression of NaV 1.5 was knocked down, ~85% of the INa+ was reduced consistent with the observations that NaV 1.5 is the main voltage-gated sodium channel in cardiac muscle and that NaX likely does not directly participate in mediating the INa+ following depolarization. Silencing NaV 1.5 expression led to significant upregulation of NaX mRNA. Similar to NaV 1.5, NaX protein levels were rapidly downregulated when the intracellular [Ca2+ ] was increased either by CaCl2 or caffeine. These data suggest that a relationship exists between NaX and NaV 1.5 and that NaX may play a role in excitation-contraction coupling.
- Published
- 2020
36. Scn8a Antisense Oligonucleotide Is Protective in Mouse Models of SCN8A Encephalopathy and Dravet Syndrome
- Author
-
Corrine E. Smolen, Jacy L. Wagnon, Wenxi Yu, Sophie F. Hill, Paymaan Jafar-Nejad, Roman J. Giger, Frank Rigo, Guy M. Lenk, Miriam H. Meisler, Julie Ziobro, Kritika Bhatia, Lucas D. Huffman, Jack M. Parent, and Hayley Petit
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Encephalopathy ,Epilepsies, Myoclonic ,Mice, Transgenic ,Open field ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Dravet syndrome ,Seizures ,Internal medicine ,medicine ,Animals ,Brain Diseases ,Dose-Response Relationship, Drug ,business.industry ,Sodium channel ,Oligonucleotides, Antisense ,medicine.disease ,Penetrance ,3. Good health ,Infusions, Intraventricular ,030104 developmental biology ,Endocrinology ,Neurology ,NAV1.6 Voltage-Gated Sodium Channel ,Mutation ,Stereotactic injection ,Female ,Neurology (clinical) ,medicine.symptom ,business ,Haploinsufficiency ,Weight gain ,030217 neurology & neurosurgery - Abstract
Objective SCN8A encephalopathy is a developmental and epileptic encephalopathy (DEE) caused by de novo gain-of-function mutations of sodium channel Nav 1.6 that result in neuronal hyperactivity. Affected individuals exhibit early onset drug-resistant seizures, developmental delay, and cognitive impairment. This study was carried out to determine whether reducing the abundance of the Scn8a transcript with an antisense oligonucleotide (ASO) would delay seizure onset and prolong survival in a mouse model of SCN8A encephalopathy. Methods ASO treatment was tested in a conditional mouse model with Cre-dependent expression of the pathogenic patient SCN8A mutation p.Arg1872Trp (R1872W). This model exhibits early onset of seizures, rapid progression, and 100% penetrance. An Scn1a +/- haploinsufficient mouse model of Dravet syndrome was also treated. ASO was administered by intracerebroventricular injection at postnatal day 2, followed in some cases by stereotactic injection at postnatal day 30. Results We observed a dose-dependent increase in length of survival from 15 to 65 days in the Scn8a-R1872W/+ mice treated with ASO. Electroencephalographic recordings were normal prior to seizure onset. Weight gain and activity in an open field were unaffected, but treated mice were less active in a wheel running assay. A single treatment with Scn8a ASO extended survival of Dravet syndrome mice from 3 weeks to >5 months. Interpretation Reduction of Scn8a transcript by 25 to 50% delayed seizure onset and lethality in mouse models of SCN8A encephalopathy and Dravet syndrome. Reduction of SCN8A transcript is a promising approach to treatment of intractable childhood epilepsies. Ann Neurol 2020;87:339-346.
- Published
- 2020
37. The clustering of voltage‐gated sodium channels in various excitable membranes
- Author
-
Yael Eshed-Eisenbach and Elior Peles
- Subjects
0301 basic medicine ,Node of Ranvier ,Sodium channel ,Action Potentials ,Voltage-Gated Sodium Channels ,Biology ,Axon initial segment ,Axons ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,Membrane ,medicine.anatomical_structure ,nervous system ,Developmental Neuroscience ,Postsynaptic potential ,Biophysics ,medicine ,Cluster Analysis ,Cytoskeleton ,Cluster analysis ,030217 neurology & neurosurgery ,Actin - Abstract
In excitable membranes, the clustering of voltage-gated sodium channels (VGSC) serves to enhance excitability at critical sites. The two most profoundly studied sites of channel clustering are the axon initial segment, where action potentials are generated and the node of Ranvier, where action potentials propagate along myelinated axons. The clustering of VGSC is found, however, in other highly excitable sites such as axonal terminals, postsynaptic membranes of dendrites and muscle fibers, and pre-myelinated axons. In this review, different examples of axonal as well as non-axonal clustering of VGSC are discussed and the underlying mechanisms are compared. Whether the clustering of channels is intrinsically or extrinsically induced, it depends on the submembranous actin-based cytoskeleton that organizes these highly specialized membrane microdomains through specific adaptor proteins.
- Published
- 2020
38. Astrocytes control the spiking of mouse visual cortex layer 5 pyramidal neurons
- Author
-
Pretty Garg, Sivaraj Mohana Sundaram, Heiko M. Leßlich, and Sakthikumar Mathivanan
- Subjects
0303 health sciences ,Patch-Clamp Techniques ,Physiology ,Chemistry ,Pyramidal Cells ,Sodium channel ,Neuronal firing ,Photostimulation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Visual cortex ,medicine.anatomical_structure ,Astrocytes ,medicine ,Animals ,Layer (electronics) ,Neuroscience ,030217 neurology & neurosurgery ,Visual Cortex ,030304 developmental biology - Published
- 2021
39. Transcriptional profiles of genes related to electrophysiological function in Scn5a+/- murine hearts
- Author
-
Samantha C. Salvage, Kamalan Jeevaratnam, Charlotte E. Edling, Michael Takla, Gary Tse, Khalil Saadeh, Christopher L.-H. Huang, Kevin Zhang, Salvage, Samantha [0000-0002-5793-2349], Huang, Christopher [0000-0001-9553-6112], Apollo - University of Cambridge Repository, Huang, Christopher L.‐H. [0000-0001-9553-6112], and Jeevaratnam, Kamalan [0000-0002-6232-388X]
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,mechanisms ,Physiology ,Sodium channel ,Wild type ,Biology ,arrhythmia ,Phenotype ,Cell biology ,Downregulation and upregulation ,Physiology (medical) ,Transcriptional regulation ,cardiovascular system ,QP1-981 ,Brugada syndrome ,cardiovascular diseases ,ORIGINAL ARTICLES ,Cardiology and Cardiovascular Medicine ,transcription ,Gene ,Homeostasis ,ORIGINAL ARTICLE ,Action potential initiation ,sodium channel - Abstract
Funder: University of Surrey; Id: http://dx.doi.org/10.13039/501100003513, The Scn5a gene encodes the major pore‐forming Nav1.5 (α) subunit, of the voltage‐gated Na+ channel in cardiomyocytes. The key role of Nav1.5 in action potential initiation and propagation in both atria and ventricles predisposes organisms lacking Scn5a or carrying Scn5a mutations to cardiac arrhythmogenesis. Loss‐of‐function Nav1.5 genetic abnormalities account for many cases of the human arrhythmic disorder Brugada syndrome (BrS) and related conduction disorders. A murine model with a heterozygous Scn5a deletion recapitulates many electrophysiological phenotypes of BrS. This study examines the relationships between its Scn5a+/− genotype, resulting transcriptional changes, and the consequent phenotypic presentations of BrS. Of 62 selected protein‐coding genes related to cardiomyocyte electrophysiological or homeostatic function, concentrations of mRNA transcribed from 15 differed significantly from wild type (WT). Despite halving apparent ventricular Scn5a transcription heterozygous deletion did not significantly downregulate its atrial expression, raising possibilities of atria‐specific feedback mechanisms. Most of the remaining 14 genes whose expression differed significantly between WT and Scn5a+/− animals involved Ca2+ homeostasis specifically in atrial tissue, with no overlap with any ventricular changes. All statistically significant changes in expression were upregulations in the atria and downregulations in the ventricles. This investigation demonstrates the value of future experiments exploring for and clarifying links between transcriptional control of Scn5a and of genes whose protein products coordinate Ca2+ regulation and examining their possible roles in BrS.
- Published
- 2021
- Full Text
- View/download PDF
40. Human‐induced pluripotent stem cell‐derived cardiomyocytes: Cardiovascular properties and metabolism and pharmacokinetics of deuterated mexiletine analogs
- Author
-
Karl J. Okolotowicz, Jorge Gomez-Galeno, John R. Cashman, Wesley L. McKeithan, Mark R. Johnson, and Mark Mercola
- Subjects
Male ,Induced Pluripotent Stem Cells ,QT prolongation ,human‐induced pluripotent stem cells‐derived cardiomyocytes ,RM1-950 ,Pharmacology ,Rats, Sprague-Dawley ,Seizures ,Mexiletine ,phenyl mexiletine ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,General Pharmacology, Toxicology and Pharmaceutics ,Induced pluripotent stem cell ,Cells, Cultured ,Mice, Inbred BALB C ,Behavior, Animal ,Chemistry ,Drug discovery ,Sodium channel ,Cardiac action potential ,Original Articles ,Hit to lead ,Potassium channel ,Long QT Syndrome ,Liver ,Neurology ,Original Article ,Female ,deuterated phenyl mexiletines ,Therapeutics. Pharmacology ,mexiletine ,arrhythmias ,metabolism ,Drug metabolism ,medicine.drug - Abstract
Prolongation of the cardiac action potential (AP) and early after depolarizations (EADs) are electrical anomalies of cardiomyocytes that can lead to lethal arrhythmias and are potential liabilities for existing drugs and drug candidates in development. For example, long QT syndrome‐3 (LQTS3) is caused by mutations in the Nav1.5 sodium channel that debilitate channel inactivation and cause arrhythmias. We tested the hypothesis that a useful drug (i.e., mexiletine) with potential liabilities (i.e., potassium channel inhibition and adverse reactions) could be re‐engineered by dynamic medicinal chemistry to afford a new drug candidate with greater efficacy and less toxicity. Human cardiomyocytes were generated from LQTS3 patient‐derived induced pluripotent stem cells (hIPSCs) and normal hIPSCs to determine beneficial (on‐target) and detrimental effects (off‐target) of mexiletine and synthetic analogs, respectively. The approach combined “drug discovery” and "hit to lead” refinement and showed that iterations of medicinal chemistry and physiological testing afforded optimized compound 22. Compared to mexiletine, compound 22 showed a 1.85‐fold greater AUC and no detectable CNS toxicity at 100 mg/kg. In vitro hepatic metabolism studies showed that 22 was metabolized via cytochrome P‐450, as previously shown, and by the flavin‐containing monooxygenase (FMO). Deuterated‐22 showed decreased metabolism and showed acceptable cardiovascular and physicochemical properties., Scheme for identification of Lead compounds.
- Published
- 2021
41. Voltage‐dependent activation of Rac1 by Na v 1.5 channels promotes cell migration
- Author
-
Peter O'Toole, William J. Brackenbury, Michaela Nelson, Rakesh Suman, Ming Yang, Richard Kasprowicz, and Andrew D. James
- Subjects
0301 basic medicine ,Membrane potential ,biology ,Physiology ,Chemistry ,Sodium channel ,Clinical Biochemistry ,Motility ,Depolarization ,Cell migration ,RAC1 ,Cell Biology ,Nav1.5 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,biology.protein ,Biophysics ,Ion channel - Abstract
Ion channels can regulate the plasma membrane potential (Vm ) and cell migration as a result of altered ion flux. However, the mechanism by which Vm regulates motility remains unclear. Here, we show that the Nav 1.5 sodium channel carries persistent inward Na+ current which depolarizes the resting Vm at the timescale of minutes. This Nav 1.5-dependent Vm depolarization increases Rac1 colocalization with phosphatidylserine, to which it is anchored at the leading edge of migrating cells, promoting Rac1 activation. A genetically encoded FRET biosensor of Rac1 activation shows that depolarization-induced Rac1 activation results in acquisition of a motile phenotype. By identifying Nav 1.5-mediated Vm depolarization as a regulator of Rac1 activation, we link ionic and electrical signaling at the plasma membrane to small GTPase-dependent cytoskeletal reorganization and cellular migration. We uncover a novel and unexpected mechanism for Rac1 activation, which fine tunes cell migration in response to ionic and/or electric field changes in the local microenvironment.
- Published
- 2019
42. Subicular pyramidal neurons gate drug resistance in temporal lobe epilepsy
- Author
-
Xiaoying Ying, Yao Liu, Shuang Wang, Cenglin Xu, Shuo Zhang, Zhong Chen, Yu-Dong Zhou, Shumin Duan, Kai Zhong, Liying Chen, Ying Wang, Fang Ding, Jiazhen Nao, and Yi Wang
- Subjects
Male ,0301 basic medicine ,Drug Resistant Epilepsy ,Hippocampal formation ,Optogenetics ,Hippocampus ,Sodium Channels ,Temporal lobe ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,otorhinolaryngologic diseases ,medicine ,Animals ,Humans ,Clozapine ,Chemistry ,Pyramidal Cells ,Sodium channel ,digestive, oral, and skin physiology ,Subiculum ,food and beverages ,Neural Inhibition ,Chemogenetics ,medicine.disease ,Electric Stimulation ,Rats ,nervous system diseases ,Electrophysiology ,030104 developmental biology ,Epilepsy, Temporal Lobe ,nervous system ,Neurology ,Phenytoin ,Female ,Neurology (clinical) ,Atrophy ,Neuroscience ,030217 neurology & neurosurgery ,Sodium Channel Blockers - Abstract
Objective Drug-resistant epilepsy causes great clinical danger and still lacks effective treatments. Methods Here, we used multifaceted approaches combining electrophysiology, optogenetics, and chemogenetics in a classic phenytoin-resistant epilepsy model to reveal the key target of subicular pyramidal neurons in phenytoin resistance. Results In vivo neural recording showed that the firing rate of pyramidal neurons in the subiculum, but not other hippocampal subregions, could not be inhibited by phenytoin in phenytoin-resistant rats. Selective inhibition of subicular pyramidal neurons by optogenetics or chemogenetics reversed phenytoin resistance, whereas selective activation of subicular pyramidal neurons induced phenytoin resistance. Moreover, long-term low-frequency stimulation at the subiculum, which is clinically feasible, significantly inhibited the subicular pyramidal neurons and reversed phenytoin resistance. Furthermore, in vitro electrophysiology revealed that off-target use of phenytoin on sodium channels of subicular pyramidal neurons was involved in the phenytoin resistance, and clinical neuroimaging data suggested the volume of the subiculum in drug-resistant patients was related to the usage of sodium channel inhibitors. Interpretation These results highlight that the subicular pyramidal neurons may be a key switch control of drug-resistant epilepsy and represent a new potential target for precise treatments. ANN NEUROL 2019;86:626-640.
- Published
- 2019
43. Acquired cardiac channelopathies in epilepsy: Evidence, mechanisms, and clinical significance
- Author
-
Terence J. O'Brien, Michael C H Li, Marian Todaro, and Kim L. Powell
- Subjects
0301 basic medicine ,Long QT syndrome ,Sudden death ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,medicine ,Humans ,Sudden Unexpected Death in Epilepsy ,Ion channel ,Cardiac channelopathy ,Cardiac electrophysiology ,business.industry ,Sodium channel ,Arrhythmias, Cardiac ,Heart ,medicine.disease ,Potassium channel ,030104 developmental biology ,Neurology ,cardiovascular system ,Channelopathies ,Neurology (clinical) ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
There is growing evidence that cardiac dysfunction in patients with chronic epilepsy could play a pathogenic role in sudden unexpected death in epilepsy (SUDEP). Recent animal studies have revealed that epilepsy secondarily alters the expression of cardiac ion channels alongside abnormal cardiac electrophysiology and remodeling. These molecular findings represent novel evidence for an acquired cardiac channelopathy in epilepsy, distinct from inherited ion channels mutations associated with cardiocerebral phenotypes. Specifically, seizure activity has been shown to alter the messenger RNA (mRNA) and protein expression of voltage-gated sodium channels (Nav 1.1, Nav 1.5), voltage-gated potassium channels (Kv 4.2, Kv 4.3), sodium-calcium exchangers (NCX1), and nonspecific cation-conducting channels (HCN2, HCN4). The pathophysiology may involve autonomic dysfunction and structural cardiac disease, as both are independently associated with epilepsy and ion channel dysregulation. Indeed, in vivo and in vitro studies of cardiac pathology reveal a complex network of signaling pathways and transcription factors regulating ion channel expression in the setting of sympathetic overactivity, cardiac failure, and hypertrophy. Other mechanisms such as circulating inflammatory mediators or exogenous effects of antiepileptic medications lack evidence. Moreover, an acquired cardiac channelopathy may underlie the electrophysiologic cardiac abnormalities seen in chronic epilepsy, potentially contributing to the increased risk of malignant arrhythmias and sudden death. Therefore, further investigation is necessary to establish whether cardiac ion channel dysregulation similarly occurs in patients with epilepsy, and to characterize any pathogenic relationship with SUDEP.
- Published
- 2019
44. Conservation of the voltage‐sensitive sodium channel protein within the Insecta
- Author
-
Jeffrey G. Scott and Juan J. Silva
- Subjects
0106 biological sciences ,0301 basic medicine ,Insecta ,Population ,Voltage-Gated Sodium Channels ,01 natural sciences ,Insecticide Resistance ,03 medical and health sciences ,Exon ,Aedes ,Pyrethrins ,Genetics ,Melanogaster ,Animals ,splice ,Amino Acid Sequence ,education ,Molecular Biology ,education.field_of_study ,biology ,Sodium channel ,Knockdown resistance ,Exons ,Sequence Analysis, DNA ,biology.organism_classification ,010602 entomology ,Drosophila melanogaster ,030104 developmental biology ,Insect Science ,Codon usage bias ,Mutation ,Insect Proteins - Abstract
The voltage-sensitive sodium channel (VSSC) is essential for the generation and propagation of action potentials. VSSC kinetics can be modified by producing different splice variants. The functionality of VSSC depends on features such as the voltage sensors, the selectivity filter and the inactivation loop. Mutations in Vssc conferring resistance to pyrethroid insecticides are known as knockdown resistance (kdr). We analysed the conservation of VSSC in both a broad scope and a narrow scope by three approaches: (1) we compared conservation of sequences and of differential exon use across orders of the Insecta; (2) we determined which kdr mutations were possible with a single nucleotide mutation in nine populations of Aedes aegypti; and (3) we examined the individual VSSC variation that exists within a population of Drosophila melanogaster. There is an increasing amount of transcript diversity possible from Diplura towards Diptera. The residues of the voltage sensors, selectivity filter and inactivation loop are highly conserved. The majority of exon sequences were >88.6% similar. Strain-specific differences in codon constraints exist for kdr mutations in nine strains of A. aegypti. Three Vssc mutations were found in one population of D. melanogaster. This study shows that, overall, Vssc is highly conserved across Insecta and within a population of an insect, but that important differences do exist.
- Published
- 2019
45. Structural adaption of axons during de‐ and remyelination in the Cuprizone mouse model
- Author
-
Petra Fallier-Becker, Gabriele Frommer-Kaestle, and Friederike Pfeiffer
- Subjects
0301 basic medicine ,Genetically modified mouse ,Multiple Sclerosis ,Mice, Transgenic ,Biology ,Neuroprotection ,Sodium Channels ,Pathology and Forensic Medicine ,Cuprizone ,Mice ,03 medical and health sciences ,Myelin ,0302 clinical medicine ,Ranvier's Nodes ,medicine ,Animals ,Premovement neuronal activity ,Remyelination ,Axon ,Myelin Sheath ,Research Articles ,Neurons ,General Neuroscience ,Sodium channel ,Neurodegeneration ,medicine.disease ,Axons ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,NAV1.6 Voltage-Gated Sodium Channel ,Neurology (clinical) ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
Multiple Sclerosis is an autoimmune disorder causing neurodegeneration mostly in young adults. Thereby, myelin is lost in the inflammatory lesions leaving unmyelinated axons at a high risk to degenerate. Oligodendrocyte precursor cells maintain their regenerative capacity into adulthood and are able to remyelinate axons if they are properly activated and differentiate. Neuronal activity influences the success of myelination indicating a close interplay between neurons and oligodendroglia. The myelination profile determines the distribution of voltage‐gated ion channels along the axon. Here, we analyze the distribution of the sodium channel subunit Nav1.6 and the ultrastructure of axons after cuprizone‐induced demyelination in transgenic mice expressing GFP in oligodendroglial cells. Using this mouse model, we found an increased number of GFP‐expressing oligodendroglial cells compared to untreated mice. Analyzing the axons, we found an increase in the number of nodes of Ranvier in mice that had received cuprizone. Furthermore, we found an enhanced portion of unmyelinated axons showing vesicles in the cytoplasm. These vesicles were labeled with VGlut1, indicating that they are involved in axonal signaling. Our results highlight the flexibility of axons towards changes in the glial compartment and depict the structural changes they undergo upon myelin removal. These findings might be considered if searching for new neuroprotective therapies that aim at blocking neuronal activity in order to avoid interfering with the process of remyelination.
- Published
- 2019
46. Possible role of SCN4A skeletal muscle mutation in apnea during seizure
- Author
-
Michael G. Hanna, Kayihan Uluc, Roope Männikkö, Sanjay M. Sisodiya, Asli Gundogdu, Dilsad Turkdogan, Hande Caglayan, Sunay Usluer, Emma Matthews, Turkdogan, Dilsad, Matthews, Emma, Usluer, Sunay, Gundogdu, Asli, Uluc, Kayihan, Mannikko, Roope, Hanna, Michael G., Sisodiya, Sanjay M., and Caglayan, Hande S.
- Subjects
SUDEP ,Myotonic Disorder ,Gene mutation ,medicine.disease_cause ,Bioinformatics ,lcsh:RC346-429 ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,030225 pediatrics ,medicine ,Short Research Article ,Laryngospasm ,lcsh:Neurology. Diseases of the nervous system ,Mutation ,laryngospasm ,business.industry ,Apnea ,Sudden infant death syndrome ,medicine.disease ,Myotonia ,myotonia ,Neurology ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,sodium channel - Abstract
SCN4A gene mutations cause a number of neuromuscular phenotypes including myotonia. A subset of infants with myotonia‐causing mutations experience severe life‐threatening episodic laryngospasm with apnea. We have recently identified similar SCN4A mutations in association with sudden infant death syndrome. Laryngospasm has also been proposed as a contributory mechanism to some cases of sudden unexpected death in epilepsy (SUDEP). We report an infant with EEG‐confirmed seizures and recurrent apneas. Whole‐exome sequencing identified a known pathogenic mutation in the SCN4A gene that has been reported in several unrelated families with myotonic disorder. We propose that the SCN4A mutation contributed to the apneas in our case, irrespective of the underlying cause of the epilepsy. We suggest this supports the notion that laryngospasm may contribute to some cases of SUDEP, and implicates a possible shared mechanism between a proportion of sudden infant deaths and sudden unexpected deaths in epilepsy.
- Published
- 2019
47. Propranolol inhibits neonatal Nav1.5 activity and invasiveness of MDA‐MB‐231 breast cancer cells: Effects of combination with ranolazine
- Author
-
Mustafa B.A. Djamgoz, Scott P. Fraser, and Alice Lee
- Subjects
0301 basic medicine ,Physiology ,Clinical Biochemistry ,Motility ,Ranolazine ,Triple Negative Breast Neoplasms ,Propranolol ,Pharmacology ,Nav1.5 ,NAV1.5 Voltage-Gated Sodium Channel ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Receptors, Adrenergic, beta ,medicine ,Humans ,Neoplasm Invasiveness ,Receptor ,Cell Proliferation ,biology ,Chemistry ,Sodium channel ,Antagonist ,Cell Biology ,Gene Expression Regulation, Neoplastic ,Drug Combinations ,030104 developmental biology ,Potassium Channels, Voltage-Gated ,030220 oncology & carcinogenesis ,biology.protein ,Tumor Hypoxia ,Female ,Proteoglycans ,Collagen ,Laminin ,Fetal bovine serum ,medicine.drug - Abstract
The MDA-MB-231 cell line was used as a model of triple negative breast cancer to investigate the interaction of β-adrenergic receptor (β-AR) and voltage-gated sodium channel (VGSC). There was significant (86%) overlap in their expression. Short-term (acute) application of the β-AR antagonist propranolol (25 μM) led to reduction of peak current and quickening of current inactivation (the latter occurred only in 5% fetal bovine serum). Long-term (48 hr) incubation with propranolol (25 μM) resulted in several changes in VGSC characteristics: shifts in (a) current-voltage relationship and (b) steady-state inactivation, both to more negative potentials and (c) the slowing of recovery from inactivation. We then investigated the effects of propranolol and ranolazine, a blocker of VGSC activity, alone and in combination, on lateral motility and Matrigel invasion. These assays were carried out under hypoxic conditions more representative of tumor progression. Propranolol (2.5 and 25 μM) and ranolazine (5 μM), and their combination inhibited lateral motility. Also, propranolol (25 μM) and ranolazine (5 μM), and their combination inhibited invasion. However, no synergy was observed in the pharmacological combinations for both assays. Propranolol also significantly decreased total neonatal Nav1.5 protein expression, the predominant VGSC subtype expressed in these cells. We conclude (a) that β-AR and VGSC are functionally coupled in MDA-MB-231 cells; (b) that propranolol has direct blocking action on the VGSC; (c) that the action of propranolol is modulated by serum; and (d) that the antimetastatic cellular effects of propranolol and ranolazine are not additive.
- Published
- 2019
48. Na V 1.6 regulates excitability of mechanosensitive sensory neurons
- Author
-
Peng Zhao, Thomas Durek, Sulayman D. Dib-Hajj, Brian S. Tanaka, Mathilde R. Israel, David J. Craik, Irina Vetter, Samuel D. Robinson, Stephen G. Waxman, Panumart Thongyoo, Stuart M. Brierley, Jennifer R. Deuis, and Joel Castro
- Subjects
0301 basic medicine ,Physiology ,Chemistry ,Sodium channel ,Sensory system ,Stimulus (physiology) ,Sensory neuron ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Nociception ,Dorsal root ganglion ,medicine ,Mechanosensitive channels ,Transduction (physiology) ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Key points: Voltage-gated sodium channels are critical for peripheral sensory neuron transduction and have been implicated in a number of painful and painless disorders. The β-scorpion toxin, Cn2, is selective for Na1.6 in dorsal root ganglion neurons. Na1.6 plays an essential role in peripheral sensory neurons, specifically at the distal terminals of mechanosensing fibres innervating the skin and colon. Na1.6 activation also leads to enhanced response to mechanical stimulus in vivo. This works highlights the use of toxins in elucidating pain pathways moreover the importance of non-peripherally restricted Na isoforms in pain generation. Abstract: Peripheral sensory neurons express multiple voltage-gated sodium channels (Na) critical for the initiation and propagation of action potentials and transmission of sensory input. Three pore-forming sodium channel isoforms are primarily expressed in the peripheral nervous system (PNS): Na1.7, Na1.8 and Na1.9. These sodium channels have been implicated in painful and painless channelopathies and there has been intense interest in them as potential therapeutic targets in human pain. Emerging evidence suggests Na1.6 channels are an important isoform in pain sensing. This study aimed to assess, using pharmacological approaches, the function of Na1.6 channels in peripheral sensory neurons. The potent and Na1.6 selective β-scorpion toxin Cn2 was used to assess the effect of Na1.6 channel activation in the PNS. The multidisciplinary approach included Ca imaging, whole-cell patch-clamp recordings, skin–nerve and gut–nerve preparations and in vivo behavioural assessment of pain. Cn2 facilitates Na1.6 early channel opening, and increased persistent and resurgent currents in large-diameter dorsal root ganglion (DRG) neurons. This promotes enhanced excitatory drive and tonic action potential firing in these neurons. In addition, Na1.6 channel activation in the skin and gut leads to increased response to mechanical stimuli. Finally, intra-plantar injection of Cn2 causes mechanical but not thermal allodynia. This study confirms selectivity of Cn2 on Na1.6 channels in sensory neurons. Activation of Na1.6 channels, in terminals of the skin and viscera, leads to profound changes in neuronal responses to mechanical stimuli. In conclusion, sensory neurons expressing Na1.6 are important for the transduction of mechanical information in sensory afferents innervating the skin and viscera.
- Published
- 2019
49. Natural Voltage‐Gated Sodium Channel Ligands: Biosynthesis and Biology
- Author
-
April L. Lukowski and Alison R. H. Narayan
- Subjects
chemistry.chemical_classification ,Natural product ,Molecular Structure ,Sodium channel ,Neurotoxins ,Organic Chemistry ,Voltage-Gated Sodium Channels ,Plants ,Biology ,Ligands ,Biochemistry ,Small molecule ,Article ,chemistry.chemical_compound ,Enzyme ,chemistry ,Biosynthesis ,Biocatalysis ,Animals ,Molecular Medicine ,Neurotoxin ,Receptor ,Molecular Biology ,Sodium Channel Blockers - Abstract
Natural product biosynthetic pathways are composed of enzymes that use powerful chemistry to assemble complex molecules. Small molecule neurotoxins are examples of natural products with intricate scaffolds which often have high affinities for their biological targets. The focus of this Minireview is small molecule neurotoxins targeting voltage-gated sodium channels (VGSCs) and the state of knowledge on their associated biosynthetic pathways. There are three small molecule neurotoxin receptor sites on VGSCs associated with three different classes of molecules: guanidinium toxins, alkaloid toxins, and ladder polyethers. Each of these types of toxins have unique structural features which are assembled by biosynthetic enzymes and the extent of information known about these enzymes varies among each class. The biosynthetic enzymes involved in the formation of these toxins have the potential to become useful tools in the efficient synthesis of VGSC probes.
- Published
- 2019
50. Magi‐1 scaffolds Na v 1‐8 and Slack K Na channels in dorsal root ganglion neurons regulating excitability and pain
- Author
-
Danielle L. Tomasello, Garrett D. Sheehan, Arin Bhattacharjee, Rasheen Powell, Katherine M. Evely, Kerri D. Pryce, Allan Nip, Sushmitha Gururaj, and Dalia Agwa
- Subjects
Nociception ,0301 basic medicine ,Scaffold protein ,Sensory Receptor Cells ,Guanylate kinase ,PDZ Domains ,Nerve Tissue Proteins ,Potassium Channels, Sodium-Activated ,Biochemistry ,Injections ,NAV1.8 Voltage-Gated Sodium Channel ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Dorsal root ganglion ,Postsynaptic potential ,Ganglia, Spinal ,Protein Interaction Mapping ,Ranvier's Nodes ,Genetics ,medicine ,Animals ,Amino Acid Sequence ,RNA, Small Interfering ,Molecular Biology ,Cells, Cultured ,Ion transporter ,Sequence Homology, Amino Acid ,Chemistry ,Research ,Sodium channel ,Membrane Proteins ,Axons ,Potassium channel ,Rats ,Cell biology ,Spinal Nerves ,030104 developmental biology ,medicine.anatomical_structure ,NAV1 ,Female ,Guanylate Kinases ,Sequence Alignment ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Voltage-dependent sodium (Na(V)) 1.8 channels regulate action potential generation in nociceptive neurons, identifying them as putative analgesic targets. Here, we show that Na(V)1.8 channel plasma membrane localization, retention, and stability occur through a direct interaction with the postsynaptic density-95/discs large/zonula occludens-1–and WW domain–containing scaffold protein called membrane-associated guanylate kinase with inverted orientation (Magi)-1. The neurophysiological roles of Magi-1 are largely unknown, but we found that dorsal root ganglion (DRG)–specific knockdown of Magi-1 attenuated thermal nociception and acute inflammatory pain and produced deficits in Na(V)1.8 protein expression. A competing cell-penetrating peptide mimetic derived from the Na(V)1.8 WW binding motif decreased sodium currents, reduced Na(V)1.8 protein expression, and produced hypoexcitability. Remarkably, a phosphorylated variant of the very same peptide caused an opposing increase in Na(V)1.8 surface expression and repetitive firing. Likewise, in vivo, the peptides produced diverging effects on nocifensive behavior. Additionally, we found that Magi-1 bound to sequence like a calcium-activated potassium channel sodium-activated (Slack) potassium channels, demonstrating macrocomplexing with Na(V)1.8 channels. Taken together, these findings emphasize Magi-1 as an essential scaffold for ion transport in DRG neurons and a central player in pain.—Pryce, K. D., Powell, R., Agwa, D., Evely, K. M., Sheehan, G. D., Nip, A., Tomasello, D. L., Gururaj, S., Bhattacharjee, A. Magi-1 scaffolds Na(V)1.8 and Slack K(Na) channels in dorsal root ganglion neurons regulating excitability and pain.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.