ABSTRACT
The aim of this study was to examine if the peripheral antinociception of α-bisabolol involves the participation of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) synthesis followed by K+ channel opening in the formalin test. Wistar rats were injected in the dorsal surface of the right hind paw with formalin (1%). Rats received a subcutaneous injection into the dorsal surface of the paw of vehicles or increasing doses of α-bisabolol (100-300 µg/paw). To determine whether the peripheral antinociception induced by α-bisabolol was mediated by either the opioid receptors or the NO-cGMP-K+ channels pathway, the effect of pretreatment (10 min before formalin injection) with the appropriate vehicles, naloxone, naltrexone, NG-nitro-l-arginine methyl ester (L-NAME), 1H-[1,2,4]-oxadiazolo[4,2-a]quinoxalin-1-one (ODQ), glibenclamide, glipizide, apamin, charybdotoxin, tetraethylammonium, or 4-aminopyridine on the antinociceptive effects induced by local peripheral α-bisabolol (300 µg/paw) were assessed. α-Bisabolol produced antinociception during both phases of the formalin test. α-Bisabolol antinociception was blocked by L-NAME, ODQ, and all the K+ channels blockers. The peripheral antinociceptive effect produced by α-bisabolol was not blocked by the opioid receptor inhibitors. α-Bisabolol was able to active the NO-cGMP-K+ channels pathway to produce its antinoceptive effect. The participation of opioid receptors in the peripheral local antinociception induced by α-bisabolol is excluded.
Subject(s)
Analgesics/pharmacology , Cyclic GMP/metabolism , Monocyclic Sesquiterpenes/pharmacology , Nitric Oxide/metabolism , Nociception/drug effects , Potassium Channels/metabolism , Receptors, Opioid/metabolism , Animals , Male , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Potassium Channels/genetics , Rats , Rats, Wistar , Receptors, Opioid/chemistry , Receptors, Opioid/geneticsABSTRACT
Snake venom serine proteases (SVSPs) are complex and multifunctional enzymes, acting primarily on hemostasis. In this work, we report the hitherto unknown inhibitory effect of a SVSP, named collinein-1, isolated from the venom of Crotalus durissus collilineatus, on a cancer-relevant voltage-gated potassium channel (hEAG1). Among 12 voltage-gated ion channels tested, collinein-1 selectively inhibited hEAG1 currents, with a mechanism independent of its enzymatic activity. Corroboratively, we demonstrated that collinein-1 reduced the viability of human breast cancer cell line MCF7 (high expression of hEAG1), but does not affect the liver carcinoma and the non-tumorigenic epithelial breast cell lines (HepG2 and MCF10A, respectively), which present low expression of hEAG1. In order to obtain both functional and structural validation of this unexpected discovery, where an unusually large ligand acts as an inhibitor of an ion channel, a recombinant and catalytically inactive mutant of collinein-1 (His43Arg) was produced and found to preserve its capability to inhibit hEAG1. A molecular docking model was proposed in which Arg79 of the SVSP 99-loop interacts directly with the potassium selectivity filter of the hEAG1 channel.
Subject(s)
Hemostasis , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Serine Proteases/toxicity , Snake Venoms/toxicity , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Catalysis , Cell Line , Drug Design , Electrophysiological Phenomena , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/chemistry , Humans , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Potassium Channel Blockers/chemistry , Potassium Channels/chemistry , Recombinant Proteins , Serine Proteases/chemistry , Snake Venoms/chemistry , Structure-Activity RelationshipABSTRACT
Alkaloids derived from plants have shown great medicinal benefits, and are often reported for their use in cardiovascular disease management. Aristotelia chilensis (Molina) Stuntz (Maqui) has shown important medicinal properties in traditional useage. In this study, we evaluated the effect of the indole-alkaloid aristoteline (ARI), isolated from leaves of Maqui, on vascular reactivity of isolated aortic rings from normotensive rats. ARI induced relaxation (100%) in a concentration-dependent manner in intact or denuded-endothelium aortic rings pre-contracted with phenylephrine (PE; 1 µM). However, a specific soluble guanylyl cyclase inhibitor (ODQ; 1 µM) significantly reduced the relaxation to ARI in aortic rings pre-contracted with PE. In the presence of ARI, the contraction induced by KCl or PE was significantly (p < 0.05) decreased. Interestingly, the potassium channel blockade with 10 µM BaCl2 (Kir), 10 µM glibenclamide (KATP), 1 mM tetraethylammonium (TEA; KCa1.1), or 1 mM 4-aminopyridine (4-AP; Kv) significantly (p < 0.05) reduced the ARI-induced relaxation. ARI significantly (p < 0.05) reduced the contractile response to agonist of CaV1.2 channels (Bay K8644; 10 nM), likely reducing the influx of extracellular calcium through plasma membrane. The mechanisms associated with this process suggest an activation of the potassium channels, a calcium-induced antagonism and endothelium independent vasodilation that possibly involves the nitric oxide-independent soluble guanylate cyclase pathway.
Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Potassium Channels/chemistry , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Chlorates/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Molecular Structure , Nitric Oxide/metabolism , Phenylephrine/pharmacology , Potassium Channels/agonists , Prostaglandins/pharmacology , Rats , Vasodilation/drug effects , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacologyABSTRACT
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Subject(s)
Drug Design , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Potassium Channels/metabolism , Animals , Drug Discovery , Humans , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/metabolismABSTRACT
Traditional antimalarial drugs based on 4-aminoquinolines have exhibited good antiproliferative activities against human tumor cells; however, their low relative efficacy has limited their corresponding clinical uses. In order to identify new potent anticancer agents based on 4-aminoquinoline, we evaluated the antiproliferative activity of a series of dehydroxy isoquines and isotebuquines against five human cancer lines. HeLa and SKBr3 were significantly more sensitive to the action of tested quinolines than the A549, MCF-7, and PC-3 cancer lines. Compound 2h was by far the most potent derivative against four of the tested lines (except to PC3 line), exhibiting low micromolar or nanomolar IC50 values superior to adriamycin reference, low toxicities on dermis human fibroblasts (LD50 > 250 µM), and excellent selectivity indexes against the mentioned cancer cells. A structure-activity relationship analysis put in evidence that a pyrrolidine or morpholine moiety as N-alkyl terminal substitution and the incorporation of the extra phenyl attached to aniline ring are pharmacophore essentials for improvement the anticancer activity of the studied dehydroxy isoquines and isotebuquines. From the results, compound 2h emerged as a promising anticancer candidate for further in vitro assays against resistant-strain and in vivo studies as well as pharmacokinetic and genotoxicity studies. Mechanistic assays suggested that the most active quinoline 2h act as calcium-activated potassium channel activator.
Subject(s)
Aminoquinolines/chemistry , Antineoplastic Agents/chemistry , Potassium Channels/chemistry , Action Potentials/drug effects , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Humans , Potassium Channels/metabolism , Structure-Activity RelationshipABSTRACT
Assuming the selectivity filter of KcsA potassium ion channel may exhibit quantum coherence, we extend a previous model by Vaziri and Plenio (2010 New J. Phys. 12 085001) to take into account Coulomb repulsion between potassium ions. We show that typical ion transit timescales are determined by this interaction, which imposes optimal input/output parameter ranges. Also, as observed in other examples of quantum tunneling in biological systems, the addition of moderate noise helps coherent ion transport.
Subject(s)
Bacterial Proteins/metabolism , Models, Molecular , Potassium Channels/metabolism , Static Electricity , Bacterial Proteins/chemistry , Biological Transport , Kinetics , Potassium Channels/chemistry , Protein Conformation , Quantum TheoryABSTRACT
Tetrameric assembly of channel subunits in the endoplasmic reticulum (ER) is essential for surface expression and function of K+ channels, but the molecular mechanism underlying this process remains unclear. In this study, we found through genetic screening that ER-located J-domain-containing chaperone proteins (J-proteins) are critical for the biogenesis and physiological function of ether-a-go-go-related gene (ERG) K+ channels in both Caenorhabditis elegans and human cells. Human J-proteins DNAJB12 and DNAJB14 promoted tetrameric assembly of ERG (and Kv4.2) K+ channel subunits through a heat shock protein (HSP) 70-independent mechanism, whereas a mutated DNAJB12 that did not undergo oligomerization itself failed to assemble ERG channel subunits into tetramers in vitro and in C. elegans. Overexpressing DNAJB14 significantly rescued the defective function of human ether-a-go-go-related gene (hERG) mutant channels associated with long QT syndrome (LQTS), a condition that predisposes to life-threatening arrhythmia, by stabilizing the mutated proteins. Thus, chaperone proteins are required for subunit stability and assembly of K+ channels.
Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , ERG1 Potassium Channel/metabolism , Endoplasmic Reticulum/metabolism , HSP40 Heat-Shock Proteins/metabolism , HSP47 Heat-Shock Proteins/metabolism , Potassium Channels/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cell Line, Tumor , ERG1 Potassium Channel/chemistry , ERG1 Potassium Channel/genetics , HEK293 Cells , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Membrane Potentials , Molecular Chaperones , Mutation , Myocytes, Cardiac/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , RNA Interference , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Time Factors , TransfectionABSTRACT
TRK transporters, a class of proteins which generally carry out the bulk of K(+) accumulation in plants, fungi, and bacteria, mediate ion currents driven by the large membrane voltages (-150 to -250 mV) common to non-animal cells. Bacterial TRK proteins resemble K(+) channels in their primary sequence, crystallize as membrane dimers having intramolecular K(+)-channel-like folding, and complex with a cytoplasmic collar formed of four RCK domains (Nature 471:336, 2011; Ibid 496:324, 2013). Fungal TRK proteins appear simpler in form than the bacterial members, but do possess two special features: a large built-in regulatory domain, and a highly conserved pair of transmembrane helices (TM7 and TM8, ahead of the C-terminus), which were postulated to facilitate intramembranal oligomerization (Biophys. J. 77:789, 1999; FEMS Yeast Res. 9:278, 2009). A surprising associated functional process in the fungal proteins which have been explored (Saccharomyces, Candida, and Neurospora) is facilitation of channel-like chloride efflux. That process is suppressed by osmoprotective agents, appears to involve hydrophobic gating, and strongly resembles conduction by Cys-loop ligand-gated anion channels. And it leads to a rather general hypothesis: that the thermodynamic tendency for hydrophobic or amphipathic transmembrane helices to self-organize into oligomers can create novel ionic pathways through biological membranes: fundamental hydrophobic nanopores, pathways of low selectivity governed by the chaotropic behavior of individual ionic species and under the strong influence of membrane voltage.
Subject(s)
Chlorides/metabolism , Potassium Channels/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Molecular Sequence Data , Potassium/metabolism , Potassium Channels/chemistry , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Yeasts/genetics , Yeasts/metabolismABSTRACT
Two pore domain potassium (K2P) channels are mostly present in the central nervous system (CNS) where they play important roles in modulating neuronal excitability. K2P channels give rise to background K(+) currents (IKSO) a key component in setting and maintaining the resting membrane potential in excitable cells. Here, we studied the expression and relative abundances of K2P channels in cerebellar granule neurons (CGNs), combining molecular biology, electrophysiology and immunologic techniques. The CGN IKSO was very sensitive to external pH, as previously reported. Quantitative determination of mRNA expression level demonstrated the existence of an accumulation pattern of transcripts in CGN that encode K2P9>K2P1>K2P3>K2P18>K2P2=K2P10>K2P4>K2P5 subunits. The presence of the major K2P subunits expressed was then confirmed by Western blot and immunofluorescence analysis, demonstrating robust expression of K2P1 (TWIK-1), K2P3 (TASK-1), K2P9 (TASK-3) and K2P18 (TRESK) channel protein. Based, on these results, it is concluded that K2P1, -3, -9 and -18 subunits represent the majority component of IKSO current in CGN.
Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Neurons/metabolism , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels/chemistry , Potassium Channels/metabolism , Animals , Cells, Cultured , Gene Expression Regulation/physiology , Ion Channel Gating/physiology , Porosity , Potassium Channels/classification , Protein Structure, Tertiary , Rats , Rats, Sprague-DawleyABSTRACT
Because of their remarkable roles in electrical cell signaling, voltage-gated cation channels (VGCCs) have been the subject of intense investigations and debate for more than 50 years. Ultimately, the prospective implications of such studies have an impact on our understanding of the molecular properties of VGCCs involved in consciousness, anesthesia, and diseases, to mention a few. The following review aims to summarize our current knowledge of activation of VGCCs by highlighting major methodological innovations in the field and the breakthroughs they allowed. Focusing mainly on insights gained through computer simulations, while acknowledging important experimental findings, we hope to inspire experimentalists to benefit from these approaches in the generation of hypotheses and design of experiments. Also, we outline major future challenges for the field, such as channel modulation, lesser-known receptors, and molecular origins of channel dysfunctions.
Subject(s)
Calcium Channels/chemistry , Ion Channel Gating , Potassium Channels/chemistry , Sodium Channels/chemistry , Animals , Calcium Channels/metabolism , Humans , Molecular Dynamics Simulation , Potassium Channels/metabolism , Sodium Channels/metabolismABSTRACT
PURPOSE: The aim of this study was to evaluate the relaxation in vitro of cavernous smooth muscle induced by a new NO donor of the complex nitrosil-ruthenium, named trans-[Ru(NH3)4(caffeine)(NO)]C13 (Rut-Caf) and sodium nitroprusside (SNP). MATERIALS AND METHODS: The tissues, immersed in isolated bath systems, were pre-contracted with phenilephrine (PE) (1 µM) and then concentration-response curves (10 (-12) - 10(-4) M) were obtained. To clarify the mechanism of action involved, it was added to the baths ODQ (10 µM, 30 µM), oxyhemoglobin (10 µM), L-cysteine (100 µM), hydroxicobalamine (100 µM), glibenclamide, iberotoxin and apamine. Tissue samples were frozen in liquid nitrogen to measure the amount of cGMP and cAMP produced. RESULTS: The substances provoked significant relaxation of the cavernous smooth muscle. Both Rut-Caf and SNP determined dose-dependent relaxation with similar potency (pEC50) and maximum effect (E(max)). The substances showed activity through activation of the soluble guanylyl cyclase (sGC), because the relaxations were inhibited by ODQ. Oxyhemoglobin significantly diminished the relaxation effect of the substances. L-cysteine failed to modify the relaxations caused by the agents. Hydroxicobalamine significantly diminished the relaxation effect of Rut-Caf. Glibenclamide significantly increased the efficacy of Rut-Caf (pEC50 4.09 x 7.09). There were no alterations of potency or maximum effect of the substances with the addition of the other ion channel blockers. Rut-Caf induced production of significant amounts of cGMP and cAMP during the relaxation process. CONCLUSIONS: In conclusion, Rut-Caf causes relaxation of smooth muscle of corpus cavernosum by means of activation of sGC with intracellular production of cGMP and cAMP; and also by release of NO in the intracellular environment. Rut-Caf releases the NO free radical and it does not act directly on the potassium ion channels.
Subject(s)
Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Ruthenium Compounds/pharmacology , Animals , Cyclic GMP/biosynthesis , Cyclic GMP/chemistry , Cysteine/pharmacology , Guanosine Monophosphate/biosynthesis , Guanosine Monophosphate/chemistry , Male , Muscle, Smooth/physiology , Nitric Oxide Donors/chemistry , Nitroprusside/chemistry , Potassium Channels/chemistry , Rabbits , Ruthenium Compounds/chemistry , Time FactorsABSTRACT
PURPOSE: The aim of this study was to evaluate the relaxation in vitro of cavernous smooth muscle induced by a new NO donor of the complex nitrosil-ruthenium, named trans-[Ru(NH3)4(caffeine)(NO)]C13 (Rut-Caf) and sodium nitroprusside (SNP). MATERIALS AND METHODS: The tissues, immersed in isolated bath systems, were pre-contracted with phenilephrine (PE) (1 µM) and then concentration-response curves (10-12 - 10-4 M) were obtained. To clarify the mechanism of action involved, it was added to the baths ODQ (10 µM, 30 µM), oxyhemoglobin (10 µM), L-cysteine (100 µM), hydroxicobalamine (100 µM), glibenclamide, iberotoxin and apamine. Tissue samples were frozen in liquid nitrogen to measure the amount of cGMP and cAMP produced. RESULTS: The substances provoked significant relaxation of the cavernous smooth muscle. Both Rut-Caf and SNP determined dose-dependent relaxation with similar potency (pEC50) and maximum effect (Emax). The substances showed activity through activation of the soluble guanylyl cyclase (sGC), because the relaxations were inhibited by ODQ. Oxyhemoglobin significantly diminished the relaxation effect of the substances. L-cysteine failed to modify the relaxations caused by the agents. Hydroxicobalamine significantly diminished the relaxation effect of Rut-Caf. Glibenclamide significantly increased the efficacy of Rut-Caf (pEC50 4.09 x 7.09). There were no alterations of potency or maximum effect of the substances with the addition of the other ion channel blockers. Rut-Caf induced production of significant amounts of cGMP and cAMP during the relaxation process. CONCLUSIONS: In conclusion, Rut-Caf causes relaxation of smooth muscle of corpus cavernosum by means of activation of sGC with intracellular production of cGMP and cAMP; and also by release of NO in the intracellular environment. Rut-Caf releases the NO free radical and it does not act directly on the potassium ion channels.
Subject(s)
Animals , Male , Rabbits , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Ruthenium Compounds/pharmacology , Cyclic GMP/biosynthesis , Cyclic GMP/chemistry , Cysteine/pharmacology , Guanosine Monophosphate/biosynthesis , Guanosine Monophosphate/chemistry , Muscle, Smooth/physiology , Nitric Oxide Donors/chemistry , Nitroprusside/chemistry , Potassium Channels/chemistry , Ruthenium Compounds/chemistry , Time FactorsABSTRACT
The BK channel is one of the most broadly expressed ion channels in mammals. In many tissues, the BK channel pore-forming α-subunit is associated to an auxiliary ß-subunit that modulates the voltage- and Ca(2+)-dependent activation of the channel. Structural components present in ß-subunits that are important for the physical association with the α-subunit are yet unknown. Here, we show through co-immunoprecipitation that the intracellular C-terminus, the second transmembrane domain (TM2) and the extracellular loop of the ß2-subunit are dispensable for association with the α-subunit pointing transmembrane domain 1 (TM1) as responsible for the interaction. Indeed, the TOXCAT assay for transmembrane protein-protein interactions demonstrated for the first time that TM1 of the ß2-subunit physically binds to the transmembrane S1 domain of the α-subunit.
Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Animals , Calcium/chemistry , Dose-Response Relationship, Drug , Epitopes/chemistry , Exons , HEK293 Cells , Humans , Immunoprecipitation , Kinetics , Patch-Clamp Techniques , Potassium/chemistry , Potassium Channels/chemistry , Protein Structure, Tertiary , TransfectionABSTRACT
Potassium channels are particularly important in determining the shape and duration of the action potential, controlling the membrane potential, modulating hormone secretion, epithelial function and, in the case of those K(+) channels activated by Ca(2+), damping excitatory signals. The multiplicity of roles played by K(+) channels is only possible to their mammoth diversity that includes at present 70 K(+) channels encoding genes in mammals. Today, thanks to the use of cloning, mutagenesis, and the more recent structural studies using x-ray crystallography, we are in a unique position to understand the origins of the enormous diversity of this superfamily of ion channels, the roles they play in different cell types, and the relations that exist between structure and function. With the exception of two-pore K(+) channels that are dimers, voltage-dependent K(+) channels are tetrameric assemblies and share an extremely well conserved pore region, in which the ion-selectivity filter resides. In the present overview, we discuss in the function, localization, and the relations between function and structure of the five different subfamilies of K(+) channels: (a) inward rectifiers, Kir; (b) four transmembrane segments-2 pores, K2P; (c) voltage-gated, Kv; (d) the Slo family; and (e) Ca(2+)-activated SK family, SKCa.
Subject(s)
Potassium Channels/metabolism , Amino Acid Sequence , Animals , Humans , Ion Channel Gating , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Potassium Channels/chemistry , Potassium Channels/classification , Potassium Channels/genetics , Protein Structure, TertiaryABSTRACT
A reduction in the risk of coronary heart disease has been associated to moderate red wine consumption. We tested whether a nonalcoholic red wine extract would open mitochondrial K(ATP) channels in guinea pig myocytes. The opening of mitochondrial K(ATP) channels was assessed by endogenous flavoprotein fluorescence. Red wine extract (100 µg·mL(-1)) increased flavoprotein oxidation (10.9% ± 1.2%, n = 20). This effect was prevented by the mitochondrial K(ATP) channel blocker, 5-hydroxydecanoate (500 µmol·L(-1); 0.3% ± 1.1%, n = 13), confirming the hypothesis that red wine extract opens mitochondrial K(ATP) channels.
Subject(s)
Coronary Disease/prevention & control , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels/agonists , Wine/analysis , Animals , Cells, Cultured , Decanoic Acids/pharmacology , Flavoproteins/metabolism , Guinea Pigs , Hydroxy Acids/pharmacology , Kinetics , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidation-Reduction , Plant Extracts/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Wine/adverse effectsABSTRACT
Voltage-gated potassium channel toxins (KTxs) are basic short chain peptides comprising 23-43 amino acid residues that can be cross-linked by 3 or 4 disulfide bridges. KTxs are classified into four large families: α-, ß-, γ- and κ-KTx. These peptides display varying selectivity and affinity for K(v) channel subtypes. In this work, a novel toxin from the Tityus serrulatus venom was isolated, characterized and submitted to a wide electrophysiological screening on 5 different subtypes of Na(V) channels (Na(V)1.4; Na(V)1.5; Na(V)1.6; Na(V)1.8 and DmNa(V)1) and 12 different subtypes of K(V) channels (K(V)1.1 - K(V)1.6; K(V)2.1; K(V)3.1; K(V)4.2; K(V)4.3; Shaker IR and ERG). This novel peptide, named Ts15, has 36 amino acids, is cross-linked by 3 disulfide bridges, has a molecular mass of 3956 Da and pI around 9. Electrophysiological experiments using patch clamp and the two-electrode voltage clamp techniques show that Ts15 preferentially blocks K(V)1.2 and K(V)1.3 channels with an IC50 value of 196 ± 25 and 508 ± 67 nM, respectively. No effect on Na(V) channels was observed, at all tested concentrations. Since Ts15 shows low amino acid identity with other known KTxs, it was considered a bona fide novel type of scorpion toxin. Ts15 is the unique member of the new α-Ktx21 subfamily and therefore was classified as α-Ktx21.1.
Subject(s)
Potassium Channel Blockers/chemistry , Potassium Channels/chemistry , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Molecular Sequence Data , Patch-Clamp Techniques , Potassium Channel Blockers/isolation & purification , Scorpion Venoms/isolation & purification , Scorpions , Sequence Analysis, ProteinABSTRACT
Potassium channel subunits composed of two-pore domains arranged in tandem (K(2P)) are of paramount importance for neural function. A variety of stimuli, such as membrane depolarization and tension, acidification, and anesthetic action, activate K(2P) channels. Most of the channel sensitivity is attributed to its intracellular C-terminal moiety, which works as a sensor domain required for proper integration of the electrical, chemical, and mechanical signals into channel activity. Herein, the structure of K(2P) in a membrane environment has been studied using molecular dynamics (MD). Two distinct fully atomistic models for the most studied K(2P) channel, namely, the TWIK-related (TREK)-1 channel have been built. These constructs were then inserted into a fully hydrated zwitterionic lipid bilayer, and each relaxed by means of MD simulations spanning approximately 0.3 micros. Both simulated TREK-1 structures converged to a final conformation characterized by a closed pore and a C-terminal domain adsorbed onto the lipid bilayer surface. The C-terminus, which is physically linked to the pore and energetically coupled to the bilayer, is poised to gate the channel in response to membrane stimulation. The present study indicates the nature of the direct coupling between the C-terminal domain and the membrane, which is a key structural feature underlying K(2P) channel function.
Subject(s)
Cell Membrane/metabolism , Molecular Dynamics Simulation , Potassium Channels/chemistry , Potassium Channels/metabolism , Arachidonic Acid/pharmacology , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Mechanical Phenomena , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Protein Structure, TertiaryABSTRACT
The mitochondrial permeability transition (PT) involves the opening of a mitochondrial unselective channel (MUC) resulting in membrane depolarization and increased permeability to ions. PT has been observed in many, but not all eukaryotic species. In some species, PT has been linked to cell death, although other functions, such as matrix ion detoxification or regulation of the rate of oxygen consumption have been considered. The identification of the proteins constituting MUC would help understand the biochemistry and physiology of this channel. It has been suggested that the mitochondrial phosphate carrier is a structural component of MUC and we decided to test this in yeast mitochondria. Mersalyl inhibits the phosphate carrier and it has been reported that it also triggers PT. Mersalyl induced opening of the decavanadate-sensitive Yeast Mitochondrial Unselective Channel (YMUC). In isolated yeast mitochondria from a phosphate carrier-null strain the sensitivity to both phosphate and mersalyl was lost, although the permeability transition was still evoked by ATP in a decavanadate-sensitive fashion. Polyethylene glycol (PEG)-induced mitochondrial contraction results indicated that in mitochondria lacking the phosphate carrier the YMUC is smaller: complete contraction for mitochondria from the wild type and the mutant strains was achieved with 1.45 and 1.1 kDa PEGs, respectively. Also, as expected for a smaller channel titration with 1.1 kDa PEG evidenced a higher sensitivity in mitochondria from the mutant strain. The above data suggest that the phosphate carrier is the phosphate sensor in YMUC and contributes to the structure of this channel.
Subject(s)
Phosphate Transport Proteins/metabolism , Potassium Channels/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Mersalyl/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Swelling/drug effects , Permeability/drug effects , Phosphate Transport Proteins/antagonists & inhibitors , Phosphates/metabolism , Polyethylene Glycols/pharmacology , Potassium Channels/chemistry , Potassium Channels/deficiency , Potassium Channels/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion , Vanadates/pharmacology , Voltage-Dependent Anion Channels/metabolismABSTRACT
Connexins are plasma membrane proteins that associate in hexameric complexes to form channels named connexons. Two connexons in neighboring cells may dock to form a "gap junction" channel, i.e. an intercellular conduit that permits the direct exchange of solutes between the cytoplasm of adjacent cells and thus mediate cell-cell ion and metabolic signaling. The lack of high resolution data for connexon structures has hampered so far the study of the structure-function relationships that link molecular effects of disease-causing mutations with their observed phenotypes. Here we present a combination of modeling techniques and molecular dynamics (MD) to infer side chain positions starting from low resolution structures containing only C alpha atoms. We validated this procedure on the structure of the KcsA potassium channel, which is solved at atomic resolution. We then produced a fully atomistic model of a homotypic Cx32 connexon starting from a published model of the C alpha carbons arrangement for the connexin transmembrane helices, to which we added extracellular and cytoplasmic loops. To achieve structural relaxation within a realistic environment, we used MD simulations inserted in an explicit solvent-membrane context and we subsequently checked predictions of putative side chain positions and interactions in the Cx32 connexon against a vast body of experimental reports. Our results provide new mechanistic insights into the effects of numerous spontaneous mutations and their implication in connexin-related pathologies. This model constitutes a step forward towards a structurally detailed description of the gap junction architecture and provides a structural platform to plan new biochemical and biophysical experiments aimed at elucidating the structure of connexin channels and hemichannels.
Subject(s)
Connexins/chemistry , Animals , Bacterial Proteins/chemistry , Carbon/chemistry , Cell Membrane/metabolism , Connexins/metabolism , Gap Junctions , Mice , Microscopy, Electron/methods , Molecular Conformation , Mutation , Phenotype , Potassium Channels/chemistry , Protein Conformation , Protein Structure, Tertiary , Streptomyces/metabolism , Gap Junction beta-1 ProteinABSTRACT
In this study, we present patch-clamp characterization of the background potassium current in human lymphoma (Jurkat cells), generated by voltage-independent 16 pS channels with a high ( approximately 100-fold) K+/Na+ selectivity. Depending on the background K+ channels density, from few per cell up to approximately 1 open channel per microm2, resting membrane potential was in the range of -40 to -83 mV, approaching E (K) = -88 mV. The background K+ channels were insensitive to margotoxin (3 nM), apamine (3 nM), and clotrimazole (1 microM), high-affinity blockers of the lymphocyte Kv1.3, SKCa2, and IKCa1 channels. The current depended weakly on external pH. Arachidonic acid (20 microM) and Hg2+ (0.3-10 microM) suppressed background K+ current in Jurkat cells by 75-90%. Background K+ current was weakly sensitive to TEA+ (IC50 = 14 mM), and was efficiently suppressed by externally applied bupivacaine (IC50 = 5 microM), quinine (IC50 = 16 microM), and Ba2+ (2 mM). Our data, in particular strong inhibition by mercuric ions, suggest that background K+ currents expressed in Jurkat cells are mediated by TWIK-related spinal cord K+ (TRESK) channels belonging to the double-pore domain K+ channel family. The presence of human TRESK in the membrane protein fraction was confirmed by Western blot analysis.