Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(25): e2322475121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857404

ABSTRACT

Low temperatures and cooling agents like menthol induce cold sensation by activating the peripheral cold receptors TRPM8 and TRPA1, cation channels belonging to the TRP channel family, while the reduction of potassium currents provides an additional and/or synergistic mechanism of cold sensation. Despite extensive studies over the past decades to identify the molecular receptors that mediate thermosensation, cold sensation is still not fully understood and many cold-sensitive peripheral neurons do not express the well-established cold sensor TRPM8. We found that the voltage-gated potassium channel KCNQ1 (Kv7.1), which is defective in cardiac LQT1 syndrome, is, in addition to its known function in the heart, a highly relevant and sex-specific sensor of moderately cold temperatures. We found that KCNQ1 is expressed in skin and dorsal root ganglion neurons, is sensitive to menthol and cooling agents, and is highly sensitive to moderately cold temperatures, in a temperature range at which TRPM8 is not thermosensitive. C-fiber recordings from KCNQ1-/- mice displayed altered action potential firing properties. Strikingly, only male KCNQ1-/- mice showed substantial deficits in cold avoidance at moderately cold temperatures, with a strength of the phenotype similar to that observed in TRPM8-/- animals. While sex-dependent differences in thermal sensitivity have been well documented in humans and mice, KCNQ1 is the first gene reported to play a role in sex-specific temperature sensation. Moreover, we propose that KCNQ1, together with TRPM8, is a key instrumentalist that orchestrates the range and intensity of cold sensation.


Subject(s)
Cold Temperature , KCNQ1 Potassium Channel , Animals , Male , Female , Mice , KCNQ1 Potassium Channel/metabolism , KCNQ1 Potassium Channel/genetics , Mice, Knockout , Ganglia, Spinal/metabolism , Thermosensing/physiology , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Mice, Inbred C57BL , Action Potentials/physiology , Sex Characteristics , Menthol/pharmacology
2.
Commun Biol ; 5(1): 301, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365746

ABSTRACT

Loss-of-function mutations in Kv7.1 often lead to long QT syndrome (LQTS), a cardiac repolarization disorder associated with arrhythmia and subsequent sudden cardiac death. The discovery of agonistic IKs modulators may offer a new potential strategy in pharmacological treatment of this disorder. The benzodiazepine derivative (R)-L3 potently activates Kv7.1 channels and shortens action potential duration, thus may represent a starting point for drug development. However, the molecular mechanisms underlying modulation by (R)-L3 are still unknown. By combining alanine scanning mutagenesis, non-canonical amino acid incorporation, voltage-clamp electrophysiology and fluorometry, and in silico protein modelling, we show that (R)-L3 not only stimulates currents by allosteric modulation of the pore domain but also alters the kinetics independently from the pore domain effects. We identify novel (R)-L3-interacting key residues in the lower S4-segment of Kv7.1 and observed an uncoupling of the outer S4 segment with the inner S5, S6 and selectivity filter segments.


Subject(s)
Benzodiazepines , Ion Channel Gating , Benzodiazepines/pharmacology , Mutation
3.
Cell Physiol Biochem ; 49(3): 1197-1207, 2018.
Article in English | MEDLINE | ID: mdl-30196304

ABSTRACT

BACKGROUND/AIMS: The hyperpolarization-activated cyclic nucleotide-gated cation channel HCN4 contributes significantly to the generation of basic cardiac electrical activity in the sinus node and is a mediator of modulation by ß-adrenergic stimulation. Heterologous expression of sick sinus syndrome (SSS) and bradycardia associated mutations within the human HCN4 gene results in altered channel function. The main aim was to describe the functional characterization of three (two novel and one known) missense mutations of HCN4 identified in families with SSS. METHODS: Here, the two-electrode voltage clamp technique on Xenopus laevis oocytes and confocal imaging on transfected COS7 cells respectively, were used to analyze the functional effects of three HCN4 mutations; R378C, R550H, and E1193Q. Membrane surface expressions of wild type and the mutant channels were assessed by confocal microscopy, chemiluminescence assay, and Western blot in COS7 and HeLa cells. RESULTS: The homomeric mutant channels R550H and E1193Q showed loss of function through increased rates of deactivation and distinctly reduced surface expression in all three homomeric mutant channels. HCN4 channels containing R550H and E1193Q mutant subunits only showed minor effects on the voltage dependence and rates of activation/deactivation. In contrast, homomeric R378C exerted a left-shifted activation curve and slowed activation kinetics. These effects were reduced in heteromeric co-expression of R378C with wild-type (WT) channels. CONCLUSION: Dysfunction of homomeric/heteromeric mutant HCN4-R378C, R550H, and E1193Q channels in the present study was primarily caused by loss of function due to decreased channel surface expression.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Sick Sinus Syndrome/genetics , Action Potentials/physiology , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Microscopy, Confocal , Molecular Dynamics Simulation , Muscle Proteins/chemistry , Mutagenesis, Site-Directed , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels/chemistry , Protein Structure, Tertiary , Xenopus laevis
4.
Cell Physiol Biochem ; 40(6): 1549-1558, 2016.
Article in English | MEDLINE | ID: mdl-27997884

ABSTRACT

BACKGROUND/AIMS: Acquired as well as inherited channelopathies are disorders that are caused by altered ion channel function. A family of channels whose malfunction is associated with different channelopathies is the Kv7 K+ channel family; and restoration of normal Kv7 channel function by small molecule modulators is a promising approach for treatment of these often fatal diseases. METHODS: Here, we show the modulation of Kv7 channels by the natural compound Rottlerin heterologously expressed in Xenopus laevis oocytes and on iPSC cardiomyocytes overexpressing Kv7.1 channels. RESULTS: We show that currents carried by Kv7.1 (EC50 = 1.48 µM), Kv7.1/KCNE1 (EC50 = 4.9 µM), and Kv7.4 (EC50 = 0.148 µM) are strongly enhanced by the compound, whereas Kv7.2, Kv7.2/Kv7.3, and Kv7.5 are not sensitive to Rottlerin. Studies on Kv7.1/KCNE1 mutants and in silico modelling indicate that Rottlerin binds to the R-L3-activator site. Rottlerin mediated activation of Kv7.1/KCNE1 channels might be a promising approach in long QT syndrome. As a proof of concept, we show that Rottlerin shortens cardiac repolarisation in iPSC-derived cardiomyocytes expressing Kv7.1. CONCLUSION: Rottlerin or an optimized derivative holds a potential as QT interval correcting drug.


Subject(s)
Acetophenones/pharmacology , Benzopyrans/pharmacology , Biological Products/pharmacology , Ion Channel Gating/drug effects , KCNQ1 Potassium Channel/metabolism , Acetophenones/chemistry , Animals , Benzopyrans/chemistry , Biological Products/chemistry , Computer Simulation , Humans , Induced Pluripotent Stem Cells/cytology , KCNQ1 Potassium Channel/chemistry , Membrane Potentials/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Domains , Protein Multimerization/drug effects , Xenopus laevis
5.
Nat Commun ; 7: 12795, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27731317

ABSTRACT

Most small-molecule inhibitors of voltage-gated ion channels display poor subtype specificity because they bind to highly conserved residues located in the channel's central cavity. Using a combined approach of scanning mutagenesis, electrophysiology, chemical ligand modification, chemical cross-linking, MS/MS-analyses and molecular modelling, we provide evidence for the binding site for adamantane derivatives and their putative access pathway in Kv7.1/KCNE1 channels. The adamantane compounds, exemplified by JNJ303, are highly potent gating modifiers that bind to fenestrations that become available when KCNE1 accessory subunits are bound to Kv7.1 channels. This mode of regulation by auxiliary subunits may facilitate the future development of potent and highly subtype-specific Kv channel inhibitors.


Subject(s)
Adamantane/analogs & derivatives , Adamantane/pharmacology , Ion Channel Gating/drug effects , KCNQ1 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Adamantane/chemistry , Allosteric Regulation/drug effects , Animals , Binding Sites , Cross-Linking Reagents/chemistry , Humans , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Models, Molecular , Mutagenesis , Mutation , Oocytes , Potassium Channel Blockers/chemistry , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Tandem Mass Spectrometry , Xenopus laevis
7.
Proc Natl Acad Sci U S A ; 111(50): E5383-92, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25453094

ABSTRACT

Jervell and Lange-Nielsen syndrome (JLNS) is one of the most severe life-threatening cardiac arrhythmias. Patients display delayed cardiac repolarization, associated high risk of sudden death due to ventricular tachycardia, and congenital bilateral deafness. In contrast to the autosomal dominant forms of long QT syndrome, JLNS is a recessive trait, resulting from homozygous (or compound heterozygous) mutations in KCNQ1 or KCNE1. These genes encode the α and ß subunits, respectively, of the ion channel conducting the slow component of the delayed rectifier K(+) current, IKs. We used complementary approaches, reprogramming patient cells and genetic engineering, to generate human induced pluripotent stem cell (hiPSC) models of JLNS, covering splice site (c.478-2A>T) and missense (c.1781G>A) mutations, the two major classes of JLNS-causing defects in KCNQ1. Electrophysiological comparison of hiPSC-derived cardiomyocytes (CMs) from homozygous JLNS, heterozygous, and wild-type lines recapitulated the typical and severe features of JLNS, including pronounced action and field potential prolongation and severe reduction or absence of IKs. We show that this phenotype had distinct underlying molecular mechanisms in the two sets of cell lines: the previously unidentified c.478-2A>T mutation was amorphic and gave rise to a strictly recessive phenotype in JLNS-CMs, whereas the missense c.1781G>A lesion caused a gene dosage-dependent channel reduction at the cell membrane. Moreover, adrenergic stimulation caused action potential prolongation specifically in JLNS-CMs. Furthermore, sensitivity to proarrhythmic drugs was strongly enhanced in JLNS-CMs but could be pharmacologically corrected. Our data provide mechanistic insight into distinct classes of JLNS-causing mutations and demonstrate the potential of hiPSC-CMs in drug evaluation.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Jervell-Lange Nielsen Syndrome/drug therapy , Jervell-Lange Nielsen Syndrome/genetics , Jervell-Lange Nielsen Syndrome/physiopathology , KCNQ1 Potassium Channel/genetics , Models, Biological , Phenotype , Action Potentials/physiology , Analysis of Variance , Base Sequence , Cell Line , Genes, Recessive/genetics , Genetic Engineering , Humans , In Vitro Techniques , KCNQ1 Potassium Channel/chemistry , Models, Molecular , Molecular Sequence Data , Mutation, Missense/genetics , Myocytes, Cardiac/physiology , Sequence Analysis, DNA
9.
J Biol Chem ; 289(33): 22749-22758, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24947509

ABSTRACT

Kv7.1 to Kv7.5 α-subunits belong to the family of voltage-gated potassium channels (Kv). Assembled with the ß-subunit KCNE1, Kv7.1 conducts the slowly activating potassium current IKs, which is one of the major currents underlying repolarization of the cardiac action potential. A known regulator of Kv7 channels is the lipid phosphatidylinositol 4,5-bisphosphate (PIP2). PIP2 increases the macroscopic current amplitude by stabilizing the open conformation of 7.1/KCNE1 channels. However, knowledge about the exact nature of the interaction is incomplete. The aim of this study was the identification of the amino acids responsible for the interaction between Kv7.1 and PIP2. We generated 13 charge neutralizing point mutations at the intracellular membrane border and characterized them electrophysiologically in complex with KCNE1 under the influence of diC8-PIP2. Electrophysiological analysis of corresponding long QT syndrome mutants suggested impaired PIP2 regulation as the cause for channel dysfunction. To clarify the underlying structural mechanism of PIP2 binding, molecular dynamics simulations of Kv7.1/KCNE1 complexes containing two PIP2 molecules in each subunit at specific sites were performed. Here, we identified a subset of nine residues participating in the interaction of PIP2 and Kv7.1/KCNE1. These residues may form at least two binding pockets per subunit, leading to the stabilization of channel conformations upon PIP2 binding.


Subject(s)
KCNQ1 Potassium Channel/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Action Potentials/physiology , Amino Acid Substitution , Animals , Binding Sites , Humans , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/genetics , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/genetics , Point Mutation , Protein Binding , Xenopus laevis
10.
Pflugers Arch ; 466(10): 1885-97, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24389605

ABSTRACT

Ionotropic glutamate receptors are the most important excitatory receptors in the central nervous system, and their impairment can lead to multiple neuronal diseases. Here, we show that glutamate-induced currents in oocytes expressing GluA1 are increased by coexpression of the schizophrenia-associated phosphoinositide kinase PIP5K2A. This effect was due to enhanced membrane abundance and was blunted by a point mutation (N251S) in PIP5K2A. An increase in GluA1 currents was also observed upon acute injection of PI(4,5)P2, the main product of PIP5K2A. By expression of wild-type and mutant PIP5K2A in human embryonic kidney cells, we were able to provide evidence of impaired kinase activity of the mutant PIP5K2A. We defined the region K813-K823 of GluA1 as critical for the PI(4,5)P2 effect by performing an alanine scan that suggested PI(4,5)P2 binding to this area. A PIP strip assay revealed PI(4,5)P2 binding to the C-terminal GluA1 peptide. The present observations disclose a novel mechanism in the regulation of GluA1.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/chemistry , Receptors, AMPA/chemistry , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Animals , Binding Sites , HEK293 Cells , Humans , Molecular Sequence Data , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Xenopus
11.
Front Pharmacol ; 3: 142, 2012.
Article in English | MEDLINE | ID: mdl-22876232

ABSTRACT

The classical tango is a dance characterized by a 2/4 or 4/4 rhythm in which the partners dance in a coordinated way, allowing dynamic contact. There is a surprising similarity between the tango and how KCNE ß-subunits "dance" to the fast rhythm of the cell with their partners from the Kv channel family. The five KCNE ß-subunits interact with several members of the Kv channels, thereby modifying channel gating via the interaction of their single transmembrane-spanning segment, the extracellular amino terminus, and/or the intracellular carboxy terminus with the Kv α-subunit. Best studied is the molecular basis of interactions between KCNE1 and Kv7.1, which, together, supposedly form the native cardiac I(Ks) channel. Here we review the current knowledge about functional and molecular interactions of KCNE1 with Kv7.1 and try to summarize and interpret the tango of the KCNEs.

SELECTION OF CITATIONS
SEARCH DETAIL
...