Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Sci Rep ; 14(1): 8541, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609508

ABSTRACT

Ultrasonic imaging, using ultrasonic phased arrays, has an enormous impact in science, medicine and society and is a widely used modality in many application fields. The maximum amount of information which can be captured by an array is provided by the data acquisition method capturing the complete data set of signals from all possible combinations of ultrasonic generation and detection elements of a dense array. However, capturing this complete data set requires long data acquisition time, large number of array elements and transmit channels and produces a large volume of data. All these reasons make such data acquisition unfeasible due to the existing phased array technology or non-applicable to cases requiring fast measurement time. This paper introduces the concept of an adaptive data acquisition process, the Selective Matrix Capture (SMC), which can adapt, dynamically, to specific imaging requirements for efficient ultrasonic imaging. SMC is realised experimentally using Laser Induced Phased Arrays (LIPAs), that use lasers to generate and detect ultrasound. The flexibility and reconfigurability of LIPAs enable the evolution of the array configuration, on-the-fly. The SMC methodology consists of two stages: a stage for detecting and localising regions of interest, by means of iteratively synthesising a sparse array, and a second stage for array optimisation to the region of interest. The delay-and-sum is used as the imaging algorithm and the experimental results are compared to images produced using the complete generation-detection data set. It is shown that SMC, without a priori knowledge of the test sample, is able to achieve comparable results, while preforming ∼ 10 times faster data acquisition and achieving ∼ 10 times reduction in data size.

2.
Sci Rep ; 13(1): 21757, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38066004

ABSTRACT

Agriculturally important crop plants emit a multitude of volatile organic compounds (VOCs), which are excellent indicators of their health status and their interactions with pathogens and pests. In this study, we have developed a novel cellular olfactory panel for detecting fungal pathogen-related VOCs we had identified in the field, as well as during controlled inoculations of several crop plants. The olfactory panel consists of seven stable HEK293 cell lines each expressing a functional Drosophila olfactory receptor as a biosensing element along with GCaMP6, a fluorescent calcium indicator protein. An automated 384-well microplate reader was used to characterize the olfactory receptor cell lines for their sensitivity to reference VOCs. Subsequently, we profiled a set of 66 VOCs on all cell lines, covering a concentration range from 1 to 100 µM. Results showed that 49 VOCs (74.2%) elicited a response in at least one olfactory receptor cell line. Some VOCs activated the cell lines even at nanomolar (ppb) concentrations. The interaction profiles obtained here will support the development of biosensors for agricultural applications. Additionally, the olfactory receptor proteins can be purified from these cell lines with sufficient yields for further processing, such as structure determination or integration with sensor devices.


Subject(s)
Olfactory Receptor Neurons , Receptors, Odorant , Volatile Organic Compounds , Humans , Animals , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/chemistry , Ligands , HEK293 Cells , Insecta/metabolism , Drosophila/metabolism , Volatile Organic Compounds/metabolism , Biomarkers
3.
Front Pharmacol ; 13: 809802, 2022.
Article in English | MEDLINE | ID: mdl-35586063

ABSTRACT

Background and purpose: Ivabradine is clinically administered to lower the heart rate, proposedly by inhibiting hyperpolarization-activated cyclic nucleotide-gated cation channels in the sinoatrial node. Recent evidence suggests that voltage-gated sodium channels (VGSC) are inhibited within the same concentration range. VGSCs are expressed within the sinoatrial node and throughout the conduction system of the heart. A block of these channels thus likely contributes to the established and newly raised clinical indications of ivabradine. We, therefore, investigated the pharmacological action of ivabradine on VGSCs in sufficient detail in order to gain a better understanding of the pro- and anti-arrhythmic effects associated with the administration of this drug. Experimental Approach: Ivabradine was tested on VGSCs in native cardiomyocytes isolated from mouse ventricles and the His-Purkinje system and on human Nav1.5 in a heterologous expression system. We investigated the mechanism of channel inhibition by determining its voltage-, frequency-, state-, and temperature-dependence, complemented by a molecular drug docking to the recent Nav1.5 cryoEM structure. Automated patch-clamp experiments were used to investigate ivabradine-mediated changes in Nav1.5 inactivation parameters and inhibition of different VGSC isoforms. Key results: Ivabradine inhibited VGSCs in a voltage- and frequency-dependent manner, but did not alter voltage-dependence of activation and fast inactivation, nor recovery from fast inactivation. Cardiac (Nav1.5), neuronal (Nav1.2), and skeletal muscle (Nav1.4) VGSC isoforms were inhibited by ivabradine within the same concentration range, as were sodium currents in native cardiomyocytes isolated from the ventricles and the His-Purkinje system. Molecular drug docking suggested an interaction of ivabradine with the classical local anesthetic binding site. Conclusion and Implications: Ivabradine acts as an atypical inhibitor of VGSCs. Inhibition of VGSCs likely contributes to the heart rate lowering effect of ivabradine, in particular at higher stimulation frequencies and depolarized membrane potentials, and to the observed slowing of intra-cardiac conduction. Inhibition of VGSCs in native cardiomyocytes and across channel isoforms may provide a potential basis for the anti-arrhythmic potential as observed upon administration of ivabradine.

4.
Polymers (Basel) ; 14(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35160496

ABSTRACT

The design, simulation, realization, and measurement of an ultra-wideband (UWB) antenna on a polymeric substrate have been realized. The UWB antenna was prepared using conventional technology, such as copper etching; inkjet printing, which is regarded as a modern and progressive nano-technology; and polymer thick-film technology in the context of screen-printing technology. The thick-film technology-based UWB antenna has a bandwidth of 3.8 GHz, with a central frequency of 9 GHz, and a frequency range of 6.6 to 10.4 GHz. In addition to a comparison of the technologies described, the results show that the mesh of the screens has a significant impact on the quality of the UWB antenna when utilizing polymeric screen-printing pastes. Last but not least, the eco-friendly combination of polyimide substrate and graphene-based screen-printing paste is thoroughly detailed. From 5 to 9.42 GHz, the graphene-based UWB antenna achieved a bandwidth of 4.42 GHz. The designed and realized UWB antenna well exceeds the Federal Communications Commission's (FCC) standards for UWB antenna definition. The modification of the energy surface of the polyimide substrate by plasma treatment is also explained in this paper, in addition to the many types of screen-printing pastes and technologies. According to the findings, plasma treatment improved the bandwidth of UWB antennas to 5.45 GHz, and the combination of plasma treatment with graphene provides a suitable replacement for traditional etching technologies. The characteristics of graphene-based pastes can also be altered by plasma treatment in terms of their usability on flexible substrates.

5.
Front Pharmacol ; 12: 738260, 2021.
Article in English | MEDLINE | ID: mdl-34658875

ABSTRACT

Standard high throughput screening projects using automated patch-clamp instruments often fail to grasp essential details of the mechanism of action, such as binding/unbinding dynamics and modulation of gating. In this study, we aim to demonstrate that depth of analysis can be combined with acceptable throughput on such instruments. Using the microfluidics-based automated patch clamp, IonFlux Mercury, we developed a method for a rapid assessment of the mechanism of action of sodium channel inhibitors, including their state-dependent association and dissociation kinetics. The method is based on a complex voltage protocol, which is repeated at 1 Hz. Using this time resolution we could monitor the onset and offset of both channel block and modulation of gating upon drug perfusion and washout. Our results show that the onset and the offset of drug effects are complex processes, involving several steps, which may occur on different time scales. We could identify distinct sub-processes on the millisecond time scale, as well as on the second time scale. Automated analysis of the results allows collection of detailed information regarding the mechanism of action of individual compounds, which may help the assessment of therapeutic potential for hyperexcitability-related disorders, such as epilepsies, pain syndromes, neuromuscular disorders, or neurodegenerative diseases.

6.
Front Pharmacol ; 12: 738460, 2021.
Article in English | MEDLINE | ID: mdl-34497526

ABSTRACT

We have developed an automated patch-clamp protocol that allows high information content screening of sodium channel inhibitor compounds. We have observed that individual compounds had their specific signature patterns of inhibition, which were manifested irrespective of the concentration. Our aim in this study was to quantify these properties. Primary biophysical data, such as onset rate, the shift of the half inactivation voltage, or the delay of recovery from inactivation, are concentration-dependent. We wanted to derive compound-specific properties, therefore, we had to neutralize the effect of concentration. This study describes how this is done, and shows how compound-specific properties reflect the mechanism of action, including binding dynamics, cooperativity, and interaction with the membrane phase. We illustrate the method using four well-known sodium channel inhibitor compounds, riluzole, lidocaine, benzocaine, and bupivacaine. Compound-specific biophysical properties may also serve as a basis for deriving parameters for kinetic modeling of drug action. We discuss how knowledge about the mechanism of action may help to predict the frequency-dependence of individual compounds, as well as their potential persistent current component selectivity. The analysis method described in this study, together with the experimental protocol described in the accompanying paper, allows screening for inhibitor compounds with specific kinetic properties, or with specific mechanisms of inhibition.

7.
Br J Pharmacol ; 178(5): 1200-1217, 2021 03.
Article in English | MEDLINE | ID: mdl-33450052

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.


Subject(s)
Sodium Channel Blockers , Sodium Channels , Binding Sites , HEK293 Cells , Humans , Membrane Potentials , Riluzole/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism
8.
PeerJ ; 7: e7542, 2019.
Article in English | MEDLINE | ID: mdl-31534841

ABSTRACT

Cognitive impairment often involves the decreased expression or hypofunction of alpha 7-type nicotinic acetylcholine receptors (α7 nAChRs). Agonists or positive allosteric modulators (PAMs) of α7 nAChRs are known to be potential treatments for dementias, different neurodegenerative disorders, pain syndromes and conditions involving inflammation. In some of these conditions, it is desirable to maintain the temporal precision of fast cholinergic events, while in others, this temporal precision is unnecessary. For this reason, the optimal therapeutic effect for distinct indications may require PAMs with different mechanisms of action. The two major mechanisms are called "type I", which are compounds that augment α7 nAChR-mediated currents but maintain their characteristic fast kinetics; and "type II", which are compounds that produce augmented and prolonged currents. In this study, we performed a kinetic analysis of two type II PAMs of the α7 nAChR: PNU-120596 and A-867744, using a fast perfusion method that allowed high temporal resolution. We characterized the type of modulation produced by the two compounds, the state-dependence of the modulatory action, and the interaction between the two compounds. We found fundamental differences between the modulation mechanisms by PNU-120596 and A-867744. Most importantly, during brief agonist pulses, A-867744 caused a strikingly type I-like modulation, while PNU-120596 caused a type II-like prolonged activation. Our results demonstrate that specific compounds, even though all labeled as type II PAMs, can behave in completely different ways, including their onset and offset kinetics, state preference, and single channel open time. Our results emphasize that subtle details of the mechanism of action may be significant in assessing the therapeutic applicability of α7 nAChR PAM compounds.

9.
Sci Rep ; 8(1): 8110, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802266

ABSTRACT

Sodium channel inhibitor drugs decrease pathological hyperactivity in various diseases including pain syndromes, myotonia, arrhythmias, nerve injuries and epilepsies. Inhibiting pathological but not physiological activity, however, is a major challenge in drug development. Sodium channel inhibitors exert their effects by a dual action: they obstruct ion flow ("block"), and they alter the energetics of channel opening and closing ("modulation"). Ideal drugs would be modulators without blocking effect, because modulation is inherently activity-dependent, therefore selective for pathological hyperactivity. Can block and modulation be separated? It has been difficult to tell, because the effect of modulation is obscured by conformation-dependent association/dissociation of the drug. To eliminate dynamic association/dissociation, we used a photoreactive riluzole analog which could be covalently bound to the channel; and found, unexpectedly, that drug-bound channels could still conduct ions, although with modulated gating. The finding that non-blocking modulation is possible, may open a novel avenue for drug development because non-blocking modulators could be more specific in treating hyperactivity-linked diseases.


Subject(s)
Muscle Proteins/antagonists & inhibitors , Riluzole/analogs & derivatives , Riluzole/pharmacology , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Ultraviolet Rays , Animals , Azides/chemistry , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Ion Channel Gating/drug effects , Muscle Proteins/metabolism , Rats , Sodium Channels/metabolism
10.
Sci Rep ; 8(1): 631, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29330525

ABSTRACT

Inactivation of voltage-gated Na+ channels (VGSC) is essential for the regulation of cellular excitability. The molecular rearrangement underlying inactivation is thought to involve the intracellular linker between domains III and IV serving as inactivation lid, the receptor for the lid (domain III S4-S5 linker) and the pore-lining S6 segements. To better understand the role of the domain IV S6 segment in inactivation we performed a cysteine scanning mutagenesis of this region in rNav 1.4 channels and screened the constructs for perturbations in the voltage-dependence of steady state inactivation. This screen was performed in the background of wild-type channels and in channels carrying the mutation K1237E, which profoundly alters both permeation and gating-properties. Of all tested constructs the mutation I1581C was unique in that the mutation-induced gating changes were strongly influenced by the mutational background. This suggests that I1581 is involved in specific short-range interactions during inactivation. In recently published crystal structures VGSCs the respective amino acids homologous to I1581 appear to control a bend of the S6 segment which is critical to the gating process. Furthermore, I1581 may be involved in the transmission of the movement of the DIII voltage-sensor to the domain IV S6 segment.


Subject(s)
Cysteine/genetics , Muscle Proteins/genetics , Mutation , Sodium Channels/genetics , Xenopus laevis/genetics , Animals , Enzyme Activation , Models, Molecular , Molecular Dynamics Simulation , Muscle Proteins/chemistry , Protein Structure, Tertiary , Rats , Sodium Channels/chemistry
11.
Mol Pharmacol ; 88(5): 866-79, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26358763

ABSTRACT

The clinically important suppression of high-frequency discharges of excitable cells by local anesthetics (LA) is largely determined by drug-induced prolongation of the time course of repriming (recovery from inactivation) of voltage-gated Na(+) channels. This prolongation may result from periodic drug-binding to a high-affinity binding site during the action potentials and subsequent slow dissociation from the site between action potentials ("dissociation hypothesis"). For many drugs it has been suggested that the fast inactivated state represents the high-affinity binding state. Alternatively, LAs may bind with high affinity to a native slow-inactivated state, thereby accelerating the development of this state during action potentials ("stabilization hypothesis"). In this case, slow recovery between action potentials occurs from enhanced native slow inactivation. To test these two hypotheses we produced serial cysteine mutations of domain IV segment 6 in rNav1.4 that resulted in constructs with varying propensities to enter fast- and slow-inactivated states. We tested the effect of the LA lidocaine on the time course of recovery from short and long depolarizing prepulses, which, under drug-free conditions, recruited mainly fast- and slow-inactivated states, respectively. Among the tested constructs the mutation-induced changes in native slow recovery induced by long depolarizations were not correlated with the respective lidocaine-induced slow recovery after short depolarizations. On the other hand, for long depolarizations the mutation-induced alterations in native slow recovery were significantly correlated with the kinetics of lidocaine-induced slow recovery. These results favor the "dissociation hypothesis" for short depolarizations but the "stabilization hypothesis" for long depolarizations.


Subject(s)
Anesthetics, Local/pharmacology , Lidocaine/pharmacology , Muscle Proteins/antagonists & inhibitors , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Muscle Proteins/physiology , Mutagenesis , Rats , Sodium Channels/physiology , Structure-Activity Relationship
12.
J Biol Chem ; 289(31): 21770-81, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24947510

ABSTRACT

Despite the availability of several crystal structures of bacterial voltage-gated Na(+) channels, the structure of eukaryotic Na(+) channels is still undefined. We used predictions from available homology models and crystal structures to modulate an external access pathway for the membrane-impermeant local anesthetic derivative QX-222 into the internal vestibule of the mammalian rNaV1.4 channel. Potassium channel-based homology models predict amino acid Ile-1575 in domain IV segment 6 to be in close proximity to Lys-1237 of the domain III pore-loop selectivity filter. The mutation K1237E has been shown previously to increase the diameter of the selectivity filter. We found that an access pathway for external QX-222 created by mutations of Ile-1575 was abolished by the additional mutation K1237E, supporting the notion of a close spatial relationship between sites 1237 and 1575. Crystal structures of bacterial voltage-gated Na(+) channels predict that the side chain of rNaV1.4 Trp-1531 of the domain IV pore-loop projects into the space between domain IV segment 6 and domain III pore-loop and, therefore, should obstruct the putative external access pathway. Indeed, mutations W1531A and W1531G allowed for exceptionally rapid access of QX-222. In addition, W1531G created a second non-selective ion-conducting pore, bypassing the outer vestibule but probably merging into the internal vestibule, allowing for control by the activation gate. These data suggest a strong structural similarity between bacterial and eukaryotic voltage-gated Na(+) channels.


Subject(s)
Anesthetics, Local/pharmacology , Ion Channel Gating , Sodium Channels/drug effects , Animals , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Protein Conformation , Sodium Channels/chemistry , Sodium Channels/genetics , Xenopus laevis
13.
Am J Physiol Heart Circ Physiol ; 306(4): H564-H573, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24337461

ABSTRACT

Duchenne muscular dystrophy (DMD), induced by mutations in the gene encoding for the cytoskeletal protein dystrophin, is an inherited disease characterized by progressive muscle weakness. Besides the relatively well characterized skeletal muscle degenerative processes, DMD is also associated with cardiac complications. These include cardiomyopathy development and cardiac arrhythmias. The current understanding of the pathomechanisms in the heart is very limited, but recent research indicates that dysfunctional ion channels in dystrophic cardiomyocytes play a role. The aim of the present study was to characterize abnormalities in L-type calcium channel function in adult dystrophic ventricular cardiomyocytes. By using the whole cell patch-clamp technique, the properties of currents through calcium channels in ventricular cardiomyocytes isolated from the hearts of normal and dystrophic adult mice were compared. Besides the commonly used dystrophin-deficient mdx mouse model for human DMD, we also used mdx-utr mice, which are both dystrophin- and utrophin-deficient. We found that calcium channel currents were significantly increased, and channel inactivation was reduced in dystrophic cardiomyocytes. Both effects enhance the calcium influx during an action potential (AP). Whereas the AP in dystrophic mouse cardiomyocytes was nearly normal, implementation of the enhanced dystrophic calcium conductance in a computer model of a human ventricular cardiomyocyte considerably prolonged the AP. Finally, the described dystrophic calcium channel abnormalities entailed alterations in the electrocardiograms of dystrophic mice. We conclude that gain of function in cardiac L-type calcium channels may disturb the electrophysiology of the dystrophic heart and thereby cause arrhythmias.


Subject(s)
Calcium Channels, L-Type/metabolism , Heart/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Myocardium/metabolism , Myocytes, Cardiac/physiology , Action Potentials/physiology , Animals , Cardiomyopathies/complications , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Computer Simulation , Disease Models, Animal , Humans , Mice , Mice, Inbred mdx , Models, Cardiovascular , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/metabolism
14.
Toxicol Appl Pharmacol ; 273(2): 259-68, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23707769

ABSTRACT

The plant alkaloid ibogaine has promising anti-addictive properties. Albeit not licensed as a therapeutic drug, and despite hints that ibogaine may perturb the heart rhythm, this alkaloid is used to treat drug addicts. We have recently reported that ibogaine inhibits human ERG (hERG) potassium channels at concentrations similar to the drugs affinity for several of its known brain targets. Thereby the drug may disturb the heart's electrophysiology. Here, to assess the drug's cardiac ion channel profile in more detail, we studied the effects of ibogaine and its congener 18-Methoxycoronaridine (18-MC) on various cardiac voltage-gated ion channels. We confirmed that heterologously expressed hERG currents are reduced by ibogaine in low micromolar concentrations. Moreover, at higher concentrations, the drug also reduced human Nav1.5 sodium and Cav1.2 calcium currents. Ion currents were as well reduced by 18-MC, yet with diminished potency. Unexpectedly, although blocking hERG channels, ibogaine did not prolong the action potential (AP) in guinea pig cardiomyocytes at low micromolar concentrations. Higher concentrations (≥ 10 µM) even shortened the AP. These findings can be explained by the drug's calcium channel inhibition, which counteracts the AP-prolonging effect generated by hERG blockade. Implementation of ibogaine's inhibitory effects on human ion channels in a computer model of a ventricular cardiomyocyte, on the other hand, suggested that ibogaine does prolong the AP in the human heart. We conclude that therapeutic concentrations of ibogaine have the propensity to prolong the QT interval of the electrocardiogram in humans. In some cases this may lead to cardiac arrhythmias.


Subject(s)
Behavior, Addictive , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ibogaine/pharmacology , Myocytes, Cardiac/drug effects , Potassium Channel Blockers/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Behavior, Addictive/drug therapy , Behavior, Addictive/metabolism , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/physiology , Female , Guinea Pigs , Humans , Ibogaine/chemistry , Ibogaine/therapeutic use , Ion Channels/antagonists & inhibitors , Ion Channels/physiology , Myocytes, Cardiac/physiology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/therapeutic use
15.
PLoS One ; 6(5): e20300, 2011.
Article in English | MEDLINE | ID: mdl-21677768

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is associated with severe cardiac complications including cardiomyopathy and cardiac arrhythmias. Recent research suggests that impaired voltage-gated ion channels in dystrophic cardiomyocytes accompany cardiac pathology. It is, however, unknown if the ion channel defects are primary effects of dystrophic gene mutations, or secondary effects of the developing cardiac pathology. METHODOLOGY/PRINCIPAL FINDINGS: To address this question, we first investigated sodium channel impairments in cardiomyocytes derived from dystrophic neonatal mice prior to cardiomyopahty development, by using the whole cell patch clamp technique. Besides the most common model for DMD, the dystrophin-deficient mdx mouse, we also used mice additionally carrying an utrophin mutation. In neonatal cardiomyocytes, dystrophin-deficiency generated a 25% reduction in sodium current density. In addition, extra utrophin-deficiency significantly altered sodium channel gating parameters. Moreover, also calcium channel inactivation was considerably reduced in dystrophic neonatal cardiomyocytes, suggesting that ion channel abnormalities are universal primary effects of dystrophic gene mutations. To assess developmental changes, we also studied sodium channel impairments in cardiomyocytes derived from dystrophic adult mice, and compared them with the respective abnormalities in dystrophic neonatal cells. Here, we found a much stronger sodium current reduction in adult cardiomyocytes. The described sodium channel impairments slowed the upstroke of the action potential in adult cardiomyocytes, and only in dystrophic adult mice, the QRS interval of the electrocardiogram was prolonged. CONCLUSIONS/SIGNIFICANCE: Ion channel impairments precede pathology development in the dystrophic heart, and may thus be considered potential cardiomyopathy triggers.


Subject(s)
Calcium Channels, L-Type/metabolism , Cardiomyopathies/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Sodium Channels/metabolism , Action Potentials/physiology , Animals , Animals, Newborn , Barium/metabolism , Cardiomyopathies/pathology , Cells, Cultured , Dystrophin/genetics , Electrocardiography , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Mutation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Sodium/metabolism , Utrophin/deficiency
16.
PLoS One ; 5(12): e15568, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21187965

ABSTRACT

BACKGROUND: There is only one established drug binding site on sodium channels. However, drug binding of sodium channels shows extreme promiscuity: ∼25% of investigated drugs have been found to potently inhibit sodium channels. The structural diversity of these molecules suggests that they may not share the binding site, and/or the mode of action. Our goal was to attempt classification of sodium channel inhibitors by measuring multiple properties of inhibition in electrophysiology experiments. We also aimed to investigate if different properties of inhibition correlate with specific chemical properties of the compounds. METHODOLOGY/PRINCIPAL FINDINGS: A comparative electrophysiological study of 35 compounds, including classic sodium channel inhibitors (anticonvulsants, antiarrhythmics and local anesthetics), as well as antidepressants, antipsychotics and neuroprotective agents, was carried out using rNav1.2 expressing HEK-293 cells and the QPatch automatic patch-clamp instrument. In the multi-dimensional space defined by the eight properties of inhibition (resting and inactivated affinity, potency, reversibility, time constants of onset and offset, use-dependence and state-dependence), at least three distinct types of inhibition could be identified; these probably reflect distinct modes of action. The compounds were clustered similarly in the multi-dimensional space defined by relevant chemical properties, including measures of lipophilicity, aromaticity, molecular size, polarity and electric charge. Drugs of the same therapeutic indication typically belonged to the same type. We identified chemical properties, which were important in determining specific properties of inhibition. State-dependence correlated with lipophilicity, the ratio of the neutral form of molecules, and aromaticity: We noticed that the highly state dependent inhibitors had at least two aromatic rings, logP>4.0, and pKa<8.0. CONCLUSIONS/SIGNIFICANCE: The correlations of inhibition properties both with chemical properties and therapeutic profiles would not have been evident through the sole determination of IC(50); therefore, recording multiple properties of inhibition may allow improved prediction of therapeutic usefulness.


Subject(s)
Sodium Channel Blockers/classification , Sodium Channel Blockers/pharmacology , Anesthetics/pharmacology , Anticonvulsants/pharmacology , Automation , Chemistry, Pharmaceutical/methods , Dose-Response Relationship, Drug , Electrophysiology/methods , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Kinetics , Patch-Clamp Techniques , Sodium/chemistry , Sodium Channels/chemistry
17.
J Biol Chem ; 285(50): 39458-70, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20926383

ABSTRACT

Voltage-gated ion channels are transmembrane proteins that undergo complex conformational changes during their gating transitions. Both functional and structural data from K(+) channels suggest that extracellular and intracellular parts of the pore communicate with each other via a trajectory of interacting amino acids. No crystal structures are available for voltage-gated Na(+) channels, but functional data suggest a similar intramolecular communication involving the inner and outer vestibules. However, the mechanism of such communication is unknown. Here, we report that amino acid Ile-1575 in the middle of transmembrane segment 6 of domain IV (DIV-S6) in the adult rat skeletal muscle isoform of the voltage-gated sodium channel (rNa(V)1.4) may act as molecular switch allowing for interaction between outer and inner vestibules. Cysteine scanning mutagenesis of the internal part of DIV-S6 revealed that only mutations at site 1575 rescued the channel from a unique kinetic state ("ultra-slow inactivation," I(US)) produced by the mutation K1237E in the selectivity filter. A similar effect was seen with I1575A. Previously, we reported that conformational changes of both the internal and the external vestibule are involved in the generation of I(US). The fact that mutations at site 1575 modulate I(US) produced by K1237E strongly suggests an interaction between these sites. Our data confirm a previously published molecular model in which Ile-1575 of DIV-S6 is in close proximity to Lys-1237 of the selectivity filter. Furthermore, these functional data define the position of the selectivity filter relative to the adjacent DIV-S6 segment within the ionic permeation pathway.


Subject(s)
Muscle Proteins/metabolism , Potassium Channels/chemistry , Sodium Channels/chemistry , Animals , Cysteine/chemistry , Electrophysiology/methods , Female , Ion Channel Gating , Isoleucine/chemistry , Kinetics , Muscle, Skeletal/metabolism , Mutation , Protein Conformation , Protein Structure, Tertiary , Rats , Sodium Channels/metabolism , Xenopus laevis
18.
Curr Mol Pharmacol ; 3(3): 129-44, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20565383

ABSTRACT

Local anesthetics have been in clinical use since 1884, and different aspects of the local anesthetic binding site have been studied in enormous detail. In spite of all these efforts, some of the most fundamental questions--such as which exact residues constitute the binding site, how many binding sites exist, do local anesthetics share their binding site(s) with other sodium channel inhibitors, and what are the mechanisms of inhibition--are still largely unanswered. We review accumulated data on the "local anesthetic receptor"and discuss controversial points, such as possible mechanisms of inhibition, the possibility of additional binding sites, the orientation of S6 helices, and the internal vs. external position of the anticonvulsant binding site. We describe the four following specific groups of functionally important residues: i) conserved asparagines six residues below the hinge residues; we propose that they are oriented toward the external surface of S6 helices, and have a critical role in the coupling of voltage sensors to gating, ii) residues lining the inner vestibule and constructing the "orthodox" binding site, iii) residues around the outer vestibule, which have been proposed to constitute an alternative external binding site, and iv) residues determining external access for quaternary amine inhibitors, such as QX314. We conclude that sodium channel inhibitors must be heterogenous in terms of binding sites and inhibition mechanisms, and propose that this heterogeneity should be taken into consideration during drug development.


Subject(s)
Sodium Channel Blockers/chemistry , Sodium Channels/chemistry , Anesthetics, Local/chemistry , Anesthetics, Local/therapeutic use , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Binding Sites , Lidocaine/analogs & derivatives , Lidocaine/chemistry , Lidocaine/pharmacology , Mutagenesis , Protein Structure, Secondary , Protein Structure, Tertiary , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics , Sodium Channels/metabolism
19.
Mar Drugs ; 8(4): 1373-93, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20479982

ABSTRACT

The outer vestibule of voltage-gated Na(+) channels is formed by extracellular loops connecting the S5 and S6 segments of all four domains ("P-loops"), which fold back into the membrane. Classically, this structure has been implicated in the control of ion permeation and in toxin blockage. However, conformational changes of the outer vestibule may also result in alterations in gating, as suggested by several P-loop mutations that gave rise to gating changes. Moreover, partial pore block by mutated toxins may reverse gating changes induced by mutations. Therefore, toxins that bind to the outer vestibule can be used to modulate channel gating.


Subject(s)
Saxitoxin/metabolism , Sodium Channels/metabolism , Tetrodotoxin/metabolism , Animals , Binding Sites , Humans , Mutation , Permeability , Protein Binding , Sodium Channels/genetics
20.
Fogorv Sz ; 100(6): 289-93, 2007 Dec.
Article in Hungarian | MEDLINE | ID: mdl-18361201

ABSTRACT

The number of old people shows increasing tendency worldwide. The prevalence of oral diseases has been increased with age. In the older adult population tooth loss, dental caries and periodontal diseases frequently can be observed as characteristic features of their oral health condition. Additionally people in elderly are frequently suffered from other oral diseases such as xerostomia, orofacial pain, oral cancer. Results of the latest epidemiological studies show that the level of oral health of Hungarian old population is very low. In many cases oral healthcare can't show an optimal situation due to low economic and social circumstances. The present situation need more changes in oral care. It is necessary to recognize the risk factors, to treat the oral diseases properly and to organize an effective oral/dental care system for the old population.


Subject(s)
Dental Care for Aged , Geriatric Dentistry/trends , Periodontal Diseases/prevention & control , Preventive Dentistry/trends , Primary Prevention/trends , Tooth Diseases/prevention & control , Aged , Aged, 80 and over , DMF Index , Dental Health Surveys , Female , Humans , Hungary/epidemiology , Male , Middle Aged , Oral Health , Oral Hygiene , Oral Medicine/trends , Risk Factors , Socioeconomic Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...