Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Heart Rhythm ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39260661

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) poses a major risk for heart failure, myocardial infarction, and stroke. Several studies have linked SCN5A variants to AF, but their precise mechanistic contribution remains unclear. Human induced pluripotent stem cells (hiPSCs) provide a promising platform for modeling SCN5A-linked AF variants and their functional alterations. OBJECTIVE: The purpose of this study was to assess the electrophysiological impact of three AF-linked SCN5A variants, K1493R, M1875T and N1986K identified in three unrelated individuals. METHODS: CRISPR-Cas9 was used to generate a new hiPSC line in which NaV1.5 was knocked-out. Following differentiation into specific atrial cardiomyocyte by using retinoic acid, the adult WT and SCN5A variants were introduced into the NaV1.5 KO line through transfection. Subsequent analysis including molecular biology, optical mapping, and electrophysiology were performed. RESULTS: The absence of NaV1.5 channels altered the expression of key cardiac genes. NaV1.5 KO hiPSC-aCMs displayed slower conduction velocities, altered action potential (AP) parameters, and impaired calcium transient propagation. The transfection of the WT channel restored sodium current density and AP characteristics. Among the AF variants, one induced a loss-of-function (N1986K) while the other two induced a gain-of-function in NaV1.5 channel activity. Cellular excitability alterations, and early afterdepolarizations were observed in AF variants. CONCLUSION: Our findings suggest that distinct alterations in NaV1.5 channel properties may trigger atrial hyperexcitability and arrhythmogenic activity in AF. Our KO model offers an innovative approach for investigating SCN5A variants in a human cardiac environment.

2.
Stem Cell Res ; 75: 103308, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38232626

ABSTRACT

Dilated cardiomyopathy (DCM) is a prevalent cause of heart failure. We generated induced pluripotent stem cell (iPSC) lines from a DCM patient carrying a mutation in the SCN5A gene, with his healthy father serving as a control. Notably, we employed CRISPR-Cas9 to rectify the mutation in the patient's iPSC line. The resulting iPSC lines expressed pluripotency markers, underwent differentiation into all three embryonic germ layers, maintained a normal karyotype, and lacked reprogramming viral vectors. These iPSC lines serve as a model for delving into the mechanisms of DCM and hold promise for the development of personalized therapeutic approaches.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Humans , Male , Induced Pluripotent Stem Cells/metabolism , Cardiomyopathy, Dilated/genetics , Cell Line , Mutation , Arrhythmias, Cardiac/metabolism , Fathers
3.
Sci Rep ; 12(1): 17182, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229510

ABSTRACT

Epilepsy is a common neurological disorder characterized by recurrent unprovoked seizures. SCN1A encodes NaV1.1, a neuronal voltage-gated Na+ channel that is highly expressed throughout the central nervous system. NaV1.1 is localized within the axon initial segment where it plays a critical role in the initiation and propagation of action potentials and neuronal firing, predominantly in γ-amino-butyric-acid (GABA)ergic neurons of the hippocampus. The objective of this study was to characterize a de novo missense variant of uncertain significance in the SCN1A gene of a proband presented with febrile status epilepticus characterized by generalized tonic clonic movements associated with ictal emesis and an abnormal breathing pattern. Screening a gene panel revealed a heterozygous missense variant of uncertain significance in the SCN1A gene, designated c.4379A>G, p.(Tyr1460Cys). The NaV1.1 wild-type (WT) and mutant channel reproduced in vivo and were transfected in HEK 293 cells. Na+ currents were recorded using the whole-cell configuration of the patch-clamp technique. This NaV1.1 variant (Tyr1460Cys) failed to express functional Na+ currents when expressed in HEK293 cells, most probably due to a pore defect of the channel given that the cell surface expression of the channel was normal. Currents generated after co-transfection with functional WT channels exhibited biophysical properties comparable to those of WT channels, which was mainly due to the functional WT channels at the cell surface. The NaV1.1 variant failed to express functional Na+ currents, most probably due to pore impairment and exhibited a well-established loss of function mechanism. The present study highlights the added-value of functional testing for understanding the pathophysiology and potential treatment decisions for patients with undiagnosed developmental epileptic encephalopathy.


Subject(s)
Epilepsy, Generalized , Epilepsy , Action Potentials/physiology , Epilepsy/genetics , HEK293 Cells , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/genetics , NAV1.6 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Seizures , gamma-Aminobutyric Acid
4.
J Neurophysiol ; 127(5): 1388-1397, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35417276

ABSTRACT

SCN2A encodes a voltage-gated sodium channel (NaV1.2) expressed throughout the central nervous system in predominantly excitatory neurons. Pathogenic variants in SCN2A are associated with epilepsy and neurodevelopmental disorders. Genotype-phenotype correlations have been described, with loss-of-function variants typically being associated with neurodevelopmental delay and later-onset seizures, whereas gain-of-function variants more often result in early infantile-onset epilepsy. However, the true electrophysiological effects of most disease-causing SCN2A variants have yet to be characterized. We report an infant who presented with migrating focal seizures in the neonatal period. She was found to have a mosaic c.2635G>A, p.Gly879Arg variant in SCN2A. Voltage-clamp studies of the variant expressed on adult and neonatal NaV1.2 isoforms demonstrated a mixed gain and loss of function, with predominantly a loss-of-function effect with reduced cell surface expression and current density. Additional small electrophysiological alterations included a decrease in the voltage dependence of activation and an increase in the voltage dependence of inactivation. This finding of a predominantly loss-of-function effect was unexpected, as the infant's early epilepsy onset would have suggested a predominantly gain-of-function effect. This case illustrates that our understanding of genotype-phenotype correlations is still limited and highlights the complexity of the underlying electrophysiological effects of SCN2A variants.NEW & NOTEWORTHY Voltage-gated sodium channels play an important role in the central nervous system, mutations in which have been reported to be responsible for epilepsy. We report here an infant presenting with epilepsy of infancy with migrating focal seizures (EIMFS) in the neonatal period with a mosaic c.2635G>A, resulting in a p.Gly879Arg missense mutation on the SCN2A gene encoding NaV1.2 sodium channels. Biophysical characterization of this variant revealed a mixture of gain- and loss-of-function effects.


Subject(s)
Epilepsy , NAV1.2 Voltage-Gated Sodium Channel , Epilepsy/genetics , Female , Humans , Infant , Mutation , NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Phenotype , Seizures/genetics
5.
Sci Rep ; 11(1): 17168, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433864

ABSTRACT

Cardiomyocytes derived from patient-specific induced pluripotent stem cells (iPSC-CMs) successfully reproduce the mechanisms of several channelopathies. However, this approach involve cell reprogramming from somatic tissue biopsies or genomic editing in healthy iPSCs for every mutation found and to be investigated. We aim to knockout (KO) NaV1.5, the cardiac sodium channel, in a healthy human iPSC line, characterize the model and then, use it to express variants of NaV1.5. We develop a homozygous NaV1.5 KO iPSC line able to differentiate into cardiomyocytes with CRISPR/Cas9 tool. The NaV1.5 KO iPSC-CMs exhibited an organized contractile apparatus, spontaneous contractile activity, and electrophysiological recordings confirmed the major reduction in total Na+ currents. The action potentials (APs) exhibited a reduction in their amplitude and in their maximal rate of rise. Voltage optical mapping recordings revealed that the conduction velocity Ca2+ transient waves propagation velocities were slow. A wild-type (WT) NaV1.5 channel expressed by transient transfection in the KO iPSC-CMs restored Na+ channel expression and AP properties. The expression of NaV1.5/delQKP, a long QT type 3 (LQT3) variant, in the NaV1.5 KO iPSC-CMs showed that dysfunctional Na+ channels exhibited a persistent Na+ current and caused prolonged AP duration that led to arrhythmic events, characteristics of LQT3.


Subject(s)
Action Potentials , Induced Pluripotent Stem Cells/metabolism , Long QT Syndrome/genetics , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Calcium Signaling , Cell Differentiation , Cell Line , Gene Knockout Techniques/methods , Homozygote , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques/methods
6.
J Physiol ; 599(5): 1651-1664, 2021 03.
Article in English | MEDLINE | ID: mdl-33442870

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.


Subject(s)
Epilepsy , NAV1.6 Voltage-Gated Sodium Channel , Action Potentials , Animals , Epilepsy/drug therapy , Epilepsy/genetics , Humans , Mutation , NAV1.6 Voltage-Gated Sodium Channel/genetics , Neurons , Rats
7.
Sci Rep ; 11(1): 2500, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510259

ABSTRACT

Cardiac complications such as electrical abnormalities including conduction delays and arrhythmias are the main cause of death in individuals with Myotonic Dystrophy type 1 (DM1). We developed a disease model using iPSC-derived cardiomyocytes (iPSC-CMs) from a healthy individual and two DM1 patients with different CTG repeats lengths and clinical history (DM1-1300 and DM1-300). We confirmed the presence of toxic RNA foci and mis-spliced MBNL1/2 transcripts in DM1 iPSC-CMs. In DM1-1300, we identified a switch in the cardiac sodium channel SCN5A from the adult to the neonatal isoform. The down-regulation of adult SCN5A isoforms is consistent with a shift in the sodium current activation to depolarized potentials observed in DM1-1300. L-type calcium current density was higher in iPSC-CMs from DM1-1300, which is correlated with the overexpression of the CaV1.2 transcript and proteins. Importantly, INa and ICaL dysfunctions resulted in prolonged action potentials duration, slower velocities, and decreased overshoots. Optical mapping analysis revealed a slower conduction velocity in DM1-1300 iPSC-CM monolayers. In conclusion, our data revealed two distinct ions channels perturbations in DM1 iPSC-CM from the patient with cardiac dysfunction, one affecting Na+ channels and one affecting Ca2+ channels. Both have an impact on cardiac APs and ultimately on heart conduction.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Ion Channel Gating , Ion Channels/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/physiopathology , Action Potentials , Adult , Biopsy , Calcium/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Disease Susceptibility , Fluorescent Antibody Technique , Humans , Male
8.
Biochem Biophys Res Commun ; 516(1): 222-228, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31208718

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are a valuable tool for investigating complex cellular and molecular events that occur in several human diseases. Importantly, the ability to differentiate hiPSCs into any human cell type provides a unique way for investigating disease mechanisms such as complex mental health diseases. The in vitro transformation of human lymphocytes into lymphoblasts (LCLs) using the Epstein-Barr virus (EBV) has been the main method for generating immortalized human cell lines for half a century. However, the derivation of iPSCs from LCLs has emerged as an alternative source from which these cell lines can be generated. We show that iPSCs derived from LCLs using the Sendai virus procedure can be successfully differentiated into cardiomyocytes, neurons, and myotubes that express neuron- and myocyte-specific markers. We further show that these cardiac and neuronal cells are functional and generate action potentials that are required for cell excitability. We conclude that the ability to differentiate LCLs into neurons and myocytes will increase the use of LCLs in the future as a potential source of cells for modelling a number of diseases.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Myoblasts/cytology , Myocytes, Cardiac/cytology , Neurons/cytology , Cell Differentiation , Cell Line , Cellular Reprogramming Techniques , Humans
9.
Sci Rep ; 8(1): 2041, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391559

ABSTRACT

Mutations in NaV1.4, the skeletal muscle voltage-gated Na+ channel, underlie several skeletal muscle channelopathies. We report here the functional characterization of two substitutions targeting the R1451 residue and resulting in 3 distinct clinical phenotypes. The R1451L is a novel pathogenic substitution found in two unrelated individuals. The first individual was diagnosed with non-dystrophic myotonia, whereas the second suffered from an unusual phenotype combining hyperkalemic and hypokalemic episodes of periodic paralysis (PP). The R1451C substitution was found in one individual with a single attack of hypoPP induced by glucocorticoids. To elucidate the biophysical mechanism underlying the phenotypes, we used the patch-clamp technique to study tsA201 cells expressing WT or R1451C/L channels. Our results showed that both substitutions shifted the inactivation to hyperpolarized potentials, slowed the kinetics of inactivation, slowed the recovery from slow inactivation and reduced the current density. Cooling further enhanced these abnormalities. Homology modeling revealed a disruption of hydrogen bonds in the voltage sensor domain caused by R1451C/L. We concluded that the altered biophysical properties of R1451C/L well account for the PMC-hyperPP cluster and that additional factors likely play a critical role in the inter-individual differences of clinical expression resulting from R1451C/L.


Subject(s)
Mutation, Missense , Myotonic Disorders/genetics , NAV1.4 Voltage-Gated Sodium Channel/metabolism , HEK293 Cells , Humans , Ion Channel Gating , NAV1.4 Voltage-Gated Sodium Channel/chemistry , NAV1.4 Voltage-Gated Sodium Channel/genetics
10.
Neurology ; 86(2): 161-9, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26659129

ABSTRACT

OBJECTIVE: To determine the molecular basis of a complex phenotype of congenital muscle weakness observed in an isolated but consanguineous patient. METHODS: The proband was evaluated clinically and neurophysiologically over a period of 15 years. Genetic testing of candidate genes was performed. Functional characterization of the candidate mutation was done in mammalian cell background using whole cell patch clamp technique. RESULTS: The proband had fatigable muscle weakness characteristic of congenital myasthenic syndrome with acute and reversible attacks of most severe muscle weakness as observed in periodic paralysis. We identified a novel homozygous SCN4A mutation (p.R1454W) linked to this recessively inherited phenotype. The p.R1454W substitution induced an important enhancement of fast and slow inactivation, a slower recovery for these inactivated states, and a frequency-dependent regulation of Nav1.4 channels in the heterologous expression system. CONCLUSION: We identified a novel loss-of-function mutation of Nav1.4 that leads to a recessive phenotype combining clinical symptoms and signs of congenital myasthenic syndrome and periodic paralysis, probably by decreasing channel availability for muscle action potential genesis at the neuromuscular junction and propagation along the sarcolemma.


Subject(s)
Genetic Predisposition to Disease , Mutation/genetics , Myasthenic Syndromes, Congenital/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Paralyses, Familial Periodic/genetics , Adult , Female , Humans , Muscle Weakness/genetics , Myasthenic Syndromes, Congenital/diagnosis , Neuromuscular Junction/genetics , Paralyses, Familial Periodic/diagnosis , Patch-Clamp Techniques/methods
11.
Eur J Pharmacol ; 764: 395-403, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26187311

ABSTRACT

Antidepressant drugs of the SSRI family are used as a third-line treatment for neuropathic pain. In contrast MAOi antidepressants, that also increase extracellular serotonin bioavailability have little or no effects on this condition. In addition to their action of the serotonin transporter, some SSRI have been shown to inhibit voltage gated sodium channels. Here we investigated the potential inhibition of SSRIs and MAOi antidepressants on Nav1.7 or Nav1.8, which are expressed in sensory neurons and play an important role in pain sensation. We used the whole-cell patch-clamp technique on HEK293 cells expressing either Nav1.7 or Nav1.8, and evaluated the effects of the SSRIs fluoxetine, paroxetine, and citalopram as well as one MAOi antidepressants on the electrophysiological properties of the Na(+) channels. Paroxetine exhibited the greatest affinity for Na(+) channels. In ascending order of affinity for Nav1.7 were paroxetine (IC50=10 µM), followed by fluoxetine (IC50=66 µM), then citalopram (IC50=174 µM). In ascending order of affinity for Nav1.8 were paroxetine (IC50=9 µM), followed by fluoxetine (IC50=49 µM), then citalopram (IC50=100 µM). Paroxetine and fluoxetine accelerated the onset of slow-inactivation and delayed the time-course of recovery from inactivation for both channels. Paroxetine and fluoxetine also had a prominent effect on the frequency-dependent inhibition, with a greater effect on Nav1.7. In contrast to SSRIs, MAOi did not affect Na(+) channels currents. These results suggest that, in certain conditions, the analgesic effect of SSRIs may in part be due to their interactions with Na(+) channels.


Subject(s)
Analgesics/pharmacology , Antidepressive Agents/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Sodium Channel Blockers/pharmacology , Citalopram/pharmacology , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , HEK293 Cells , Humans , Membrane Potentials , Moclobemide/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Paroxetine/pharmacology , Patch-Clamp Techniques , Time Factors , Transfection
12.
Mol Pharmacol ; 86(4): 378-89, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25028482

ABSTRACT

The voltage-gated Nav1.5 channel is essential for the propagation of action potentials in the heart. Malfunctions of this channel are known to cause hereditary diseases. It is a prime target for class 1 antiarrhythmic drugs and a number of antidepressants. Our study investigated the Nav1.5 blocking properties of fluoxetine, a selective serotonin reuptake inhibitor. Nav1.5 channels were expressed in HEK-293 cells, and Na(+) currents were recorded using the patch-clamp technique. Dose-response curves of racemic fluoxetine (IC50 = 39 µM) and its optical isomers had a similar IC50 [40 and 47 µM for the (+) and (-) isomers, respectively]. Norfluoxetine, a fluoxetine metabolite, had a higher affinity than fluoxetine, with an IC50 of 29 µM. Fluoxetine inhibited currents in a frequency-dependent manner, shifted steady-state inactivation to more hyperpolarized potentials, and slowed the recovery of Nav1.5 from inactivation. Mutating a phenylalanine (F1760) and a tyrosine (Y1767) in the S6 segment of domain (D) IV (DIVS6) significantly reduced the affinity of fluoxetine and its frequency-dependent inhibition. We used a noninactivating Nav1.5 mutant to show that fluoxetine displays open-channel block behavior. The molecular model of fluoxetine in Nav1.5 was in agreement with mutational experiments in which F1760 and Y1767 were found to be the key residues in binding fluoxetine. We concluded that fluoxetine blocks Nav1.5 by binding to the class 1 antiarrhythmic site. The blocking of cardiac Na(+) channels should be taken into consideration when prescribing fluoxetine alone or in association with other drugs that may be cardiotoxic or for patients with conduction disorders.


Subject(s)
Fluoxetine/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Sodium Channel Blockers/pharmacology , Amino Acid Sequence , Anti-Arrhythmia Agents/pharmacology , Binding Sites , Fluoxetine/adverse effects , Fluoxetine/pharmacokinetics , HEK293 Cells , Humans , Inhibitory Concentration 50 , Ion Channel Gating , Molecular Sequence Data , Mutation , NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/genetics , Protein Binding , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Sodium Channel Blockers/pharmacokinetics
13.
Can J Physiol Pharmacol ; 92(3): 189-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24593783

ABSTRACT

P2X receptors are cation-permeable ligand-gated ion channels that open in response to the binding of ATP. These receptors are present in many excitable cells, including neurons, striated muscle cells, epithelial cells, and leukocytes. They mediate fast excitatory neurotransmission in the central and peripheral nervous systems and are thought to be involved in neuropathic pain, inflammation, and cell damage following ischemia-reperfusion injuries. P2X receptors are thus a target for the development of new therapeutics to treat chronic pain and inflammation. In this study, we characterized the inhibition caused by pyridoxal-5'-phosphate, a natural metabolite of vitamin B6 (MC-1), of P2X2, P2X4, P2X7, and P2X2/3 receptors stably expressed in HEK293 cells using the patch-clamp technique in the whole-cell configuration. We also tested a new approach using VC6.1, a modified cameleon calcium-sensitive fluorescent protein, to characterize the inhibition of P2X2 and P2X2/3. MC-1 blocked these two P2X receptors, with an IC50 of 7 and 13 µmol/L, respectively. P2X2 exhibited the highest affinity for VC6.1, and the chimeric receptor P2X2/3, the lowest. The patch-clamp and imaging approaches gave similar results and indicated that VC6.1 may be useful for high throughput drug screening. Pyridoxal-5'-phosphate is an efficient P2X blocker and can be classified as a P2X antagonist.


Subject(s)
Purinergic P2X Receptor Antagonists/pharmacology , Pyridoxal Phosphate/pharmacology , Receptors, Purinergic P2X/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Ion Channel Gating , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
Eur J Pharmacol ; 727: 158-66, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24486399

ABSTRACT

n-butyl-p-aminobenzoate (BAB), a local anesthetic, is administered epidurally in cancer patients to treat pain that is poorly controlled by other drugs that have a number of adverse effects. The purpose of the study was to unravel the mechanisms underlying the apparent selective pain suppressant effect of BAB. We used the whole-cell patch-clamp technique to record Na(+) currents and action potentials (APs) in dissociated, nociceptive dorsal root ganglion (DRG) cells from rats, two types of peripheral sensory neuron Na(+) channels (Nav1.7 and Nav1.8), and the motor neuron-specific Na(+) channel (Nav1.6) expressed in HEK293 cells. BAB (1-100µM) inhibited, in a concentration-dependent manner, the depolarization evoked repetitive firing in DRG cells, the three types of Na(+) current expressed in HEK293 cells, and the TTXr Na(+) current of the DRG neurons. BAB induced a use-dependent block that caused a shift of the inactivation curve in the hyperpolarizing direction. BAB enhanced the onset of slow inactivation of Nav1.7 and Nav1.8 currents but not of Nav1.6 currents. At clinically relevant concentrations (1-100µM), BAB is thus a more potent inhibitor of peripheral TTX-sensitive TTXs, Nav1.7 and TTX-resistant NaV1.8 Na(+) channels than of motor neuron axonal Nav1.6 Na(+) channels. BAB had similar effects on the TTXr Na(+) channels of rat DRG neurons and Nav1.8 channels expressed in HEK293 cells. The observed selectivity of BAB in treating cancer pain may be due to an enhanced and selective responsiveness of Na(+) channels in nociceptive neurons to this local anesthetic.


Subject(s)
Anesthetics, Local/pharmacology , Benzocaine/analogs & derivatives , Ganglia, Spinal/drug effects , Neurons/drug effects , Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/drug effects , Action Potentials , Animals , Benzocaine/pharmacology , Dose-Response Relationship, Drug , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , NAV1.6 Voltage-Gated Sodium Channel/drug effects , NAV1.6 Voltage-Gated Sodium Channel/metabolism , NAV1.7 Voltage-Gated Sodium Channel/drug effects , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neurons/metabolism , Rats, Sprague-Dawley , Time Factors , Transfection , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
15.
Gene ; 536(2): 348-56, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24334129

ABSTRACT

BACKGROUND: A variant of the ether-à-go-go related channel (hERG), p.Arg148Trp (R148W) was found at heterozygous state in two infants who died from sudden infant death syndrome (SIDS), one with documented prolonged QTc and Torsade de Pointes (TdP), and in an adult woman with QTc >500 ms, atrioventricular block and TdP. This variant was previously reported in cases of severe ventricular arrhythmia but very rarely in control subjects. Its classification as mutation or polymorphism awaited electrophysiological characterization. METHODS: The properties of this N-terminal, proximal domain, hERG variant were explored in Xenopus oocytes injected with the same amount of RNA encoding for either hERG/WT or hERG/R148W or their equimolar mixture. The human ventricular cell (TNNP) model was used to test the effects of changes in hERG current. RESULTS: R148W alone produced a current similar to the WT (369 ± 76 nA (mean ± SEM), n=13 versus 342 ± 55 nA in WT, n=13), while the co-expression of 1/2 WT+1/2 R148W lowered the current by 29% versus WT (243 ± 35 nA, n=13, p<0.05). The voltage dependencies of steady-state activation and inactivation were not changed in the variant alone or in co-expression with the WT. The time constants of fast recovery from inactivation and of fast and slow deactivation analyzed between -120 and +20 mV were not changed. The voltage-dependent distribution of the current amplitudes among fast-, slow- and non-deactivating fractions was unaltered. A 6.6% increase in APD90 from 323.5 ms to 345 ms was observed using the human cardiac ventricular myocyte model. CONCLUSIONS: Such a decrease in hERG current as evidenced here when co-expressing the hERG/R148W variant with the WT may have predisposed to the observed long QT syndrome and associated TdP. Therefore, the heterozygous carriers of hERG/R148W may be at risk of cardiac sudden death.


Subject(s)
Arrhythmias, Cardiac/genetics , Heart Conduction System/abnormalities , Long QT Syndrome/genetics , Mutation/genetics , Trans-Activators/genetics , Adult , Animals , Arrhythmias, Cardiac/metabolism , Brugada Syndrome , Cardiac Conduction System Disease , Cell Line , Death, Sudden, Cardiac , Female , HEK293 Cells , Heart/physiopathology , Heart Conduction System/metabolism , Heterozygote , Humans , Infant , Long QT Syndrome/metabolism , Male , Myocytes, Cardiac/metabolism , Oocysts/metabolism , Trans-Activators/metabolism , Transcriptional Regulator ERG , Xenopus/genetics , Xenopus/metabolism
16.
Biochem Biophys Res Commun ; 346(1): 306-13, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16756948

ABSTRACT

The NR4A3 nuclear receptor (also known as NOR1) is involved in tumorigenesis by the t(9;22) chromosome translocation encoding the EWS/NOR1 fusion protein found in approximately 75% of all cases of extraskeletal myxoid chondrosarcomas (EMC). Several observations suggest that one role of EWS/NOR1 in tumorigenesis may be to deregulate the expression of specific target genes. We have shown previously that constitutive expression of EWS/NOR1 in CFK2 fetal rat chondrogenic cells induces their transformation as measured by growth beyond confluency and growth in soft agar. To identify genes regulated by the fusion protein in this model, we have generated a CFK2 cell line in which the expression of EWS/NOR1 is controlled by tetracycline. Using the differential display technique, we have identified the serum- and glucocorticoid-regulated kinase 1 (SGK1) mRNA as being up-regulated in the presence of EWS/NOR1. Co-immunocytochemistry confirmed over-expression of the SGK1 protein in cells expressing EWS/NOR1. Significantly, immunohistochemistry of 10 EMC tumors positive for EWS/NOR1 showed that all of them over-express the SGK1 protein in contrast to non-neoplastic cells in the same biopsies and various other sarcoma types. These results strongly suggest that SGK1 may be a genuine in vivo target of EWS/NOR1 in EMC.


Subject(s)
DNA-Binding Proteins/physiology , Immediate-Early Proteins/biosynthesis , Nerve Tissue Proteins/physiology , Protein Serine-Threonine Kinases/biosynthesis , RNA-Binding Protein EWS/physiology , Animals , Bone Neoplasms/physiopathology , Cell Line , Chondrosarcoma/physiopathology , Enzyme Induction , Promoter Regions, Genetic/drug effects , Rats , Recombinant Fusion Proteins/pharmacology , Up-Regulation
17.
Cancer Lett ; 227(2): 185-91, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16112421

ABSTRACT

In approximately 70% of human extraskeletal myxoid chondrosarcoma (EMC) tumors, a t(9;22) chromosome translocation gives rise to a fusion protein, named EWS/NOR1, containing the amino-terminal domain of EWS fused to the complete amino acid sequence of the nuclear receptor NOR1. Several observations suggest that one role of EWS/NOR1 in EMC may be to deregulate the expression of specific genes involved in the tumoral process. In order to identify these genes, we have used a CFK2 chondrogenic cell line over-expressing EWS/NOR1. A differential display analysis has identified the PLAGL1 gene as being down-regulated in the CFK2(EWS/NOR1) cell line compared to native CFK2 cells. RT-PCR analyses show that whereas the PLAGL1 mRNAs encoding the two isoforms of the protein are highly expressed in four human chondrocyte immortalized cell lines and two human chondrocyte primary cultures, they are strongly down-regulated in six EMC tumors. We conclude that down-regulation of PLAGL1 may be a significant contributing factor in the development of EMC tumors.


Subject(s)
Cell Cycle Proteins/metabolism , Chondrosarcoma/metabolism , Genes, Tumor Suppressor/physiology , RNA-Binding Protein EWS/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Chondrosarcoma/genetics , Down-Regulation , Gene Expression Profiling , Humans , Membrane Transport Proteins , Nasopharyngeal Neoplasms , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein EWS/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL