1
|
Simonson BT, Jiang Z, Ryan JF, Jegla T. Ctenophores and parahoxozoans independently evolved functionally diverse voltage-gated K+ channels. J Gen Physiol 2025; 157:e202413740. [PMID: 40100064 PMCID: PMC11917167 DOI: 10.1085/jgp.202413740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/29/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
The ctenophore species Mnemiopsis leidyi is known to have a large set of voltage-gated K+ channels, but little is known about the functional diversity of these channels or their evolutionary history in other ctenophore species. Here, we searched the genomes of two additional ctenophore species, Beroe ovata and Hormiphora californensis, for voltage-gated K+ channels and functionally expressed a subset of M. leidyi channels. We found that the last common ancestor of these three disparate ctenophore lineages probably had at least 33 voltage-gated K+ channels. Two of these genes belong to the EAG family, and the remaining 31 belong to the Shaker family and form a single clade within the animal/choanoflagellate Shaker phylogeny. We additionally found evidence for 10 of these Shaker channels in a transcriptome of the early branching ctenophore lineage Euplokamis dunlapae, suggesting that the diversification of these channels was already underway early in ctenophore evolution. We functionally expressed 16 Mnemiopsis Shakers and found that they encode a diverse array of voltage-gated K+ conductances with functional orthologs for many classic Shaker family subtypes found in cnidarians and bilaterians. Analysis of Mnemiopsis transcriptome data show these 16 Shaker channels are expressed in a wide variety of cell types, including neurons, muscle, comb cells, and colloblasts. Ctenophores therefore appear to have independently evolved much of the voltage-gated K+ channel diversity that is shared between cnidarians and bilaterians.
Collapse
Affiliation(s)
- Benjamin T Simonson
- Department of Biology and Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - Zhaoyang Jiang
- Department of Biology and Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - Joseph F Ryan
- Whitney Laboratory for Marine Bioscience, University of Florida , St. Augustine, FL, USA
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - Timothy Jegla
- Department of Biology and Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| |
Collapse
|
2
|
Ukachukwu CU, Jimenez-Vazquez EN, Salwi S, Goodrich M, Sanchez-Conde FG, Orland KM, Jain A, Eckhardt LL, Kamp TJ, Jones DK. A PAS-targeting hERG1 activator reduces arrhythmic events in Jervell and Lange-Nielsen syndrome patient-derived hiPSC-CMs. JCI Insight 2025; 10:e183444. [PMID: 39786967 PMCID: PMC11949009 DOI: 10.1172/jci.insight.183444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
The hERG1 potassium channel conducts the cardiac repolarizing current, IKr. hERG1 has emerged as a therapeutic target for cardiac diseases marked by prolonged action potential duration (APD). Unfortunately, many hERG1 activators display off-target and proarrhythmic effects that limit their therapeutic potential. A Per-Arnt-Sim (PAS) domain in the hERG1 N-terminus reduces IKr by slowing channel activation and promoting inactivation. Disrupting PAS activity increases IKr and shortens APD in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). We thus hypothesized that the hERG1 PAS domain could represent a therapeutic target to reduce arrhythmogenic potential in a long QT syndrome (LQTS) background. To test this, we measured the antiarrhythmic capacity of a PAS-disabling single-chain variable fragment antibody, scFv2.10, in a hiPSC-CM line derived from a patient with Jervell and Lange Nielsen (JLN) syndrome. JLN is a severe form of LQTS caused by autosomal recessive mutations in KCNQ1. The patient in this study carried compound heterozygous mutations in KCNQ1. Corresponding JLN hiPSC-CMs displayed prolonged APD and early afterdepolarizations (EADs). Disrupting PAS with scFv2.10 increased IKr, shortened APD, and reduced the incidence of EADs. These data demonstrate that the hERG1 PAS domain could serve as a therapeutic target to treat disorders of cardiac electrical dysfunction.
Collapse
Affiliation(s)
- Chiamaka U. Ukachukwu
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Shreya Salwi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Matthew Goodrich
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Kate M. Orland
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, Madison, Wisconsin, USA
| | - Abhilasha Jain
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lee L. Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Inherited Arrhythmia Clinic, Division of Cardiovascular Medicine, Department of Medicine, Madison, Wisconsin, USA
| | - Timothy J. Kamp
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison Wisconsin, USA
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Abdelaziz R, Tomczak AP, Neef A, Pardo LA. Revealing a hidden conducting state by manipulating the intracellular domains in K V10.1 exposes the coupling between two gating mechanisms. eLife 2024; 12:RP91420. [PMID: 39259196 PMCID: PMC11390113 DOI: 10.7554/elife.91420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
The KCNH family of potassium channels serves relevant physiological functions in both excitable and non-excitable cells, reflected in the massive consequences of mutations or pharmacological manipulation of their function. This group of channels shares structural homology with other voltage-gated K+ channels, but the mechanisms of gating in this family show significant differences with respect to the canonical electromechanical coupling in these molecules. In particular, the large intracellular domains of KCNH channels play a crucial role in gating that is still only partly understood. Using KCNH1(KV10.1) as a model, we have characterized the behavior of a series of modified channels that could not be explained by the current models. With electrophysiological and biochemical methods combined with mathematical modeling, we show that the uncovering of an open state can explain the behavior of the mutants. This open state, which is not detectable in wild-type channels, appears to lack the rapid flicker block of the conventional open state. Because it is accessed from deep closed states, it elucidates intermediate gating events well ahead of channel opening in the wild type. This allowed us to study gating steps prior to opening, which, for example, explain the mechanism of gating inhibition by Ca2+-Calmodulin and generate a model that describes the characteristic features of KCNH channels gating.
Collapse
Affiliation(s)
- Reham Abdelaziz
- Oncophysiology Group. Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Adam P Tomczak
- Oncophysiology Group. Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Andreas Neef
- Neurophysics Laboratory, Göttingen Campus Institute for Dynamics of Biological Networks, Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group. Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| |
Collapse
|
4
|
Codding SJ, Trudeau MC. Photoinhibition of the hERG potassium channel PAS domain by ultraviolet light speeds channel closing. Biophys J 2024; 123:2392-2405. [PMID: 38796698 PMCID: PMC11365103 DOI: 10.1016/j.bpj.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/30/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024] Open
Abstract
hERG potassium channels are critical for cardiac excitability. hERG channels have a Per-Arnt-Sim (PAS) domain at their N-terminus, and here, we examined the mechanism for PAS domain regulation of channel opening and closing (gating). We used TAG codon suppression to incorporate the noncanonical amino acid 4-benzoyl-L-phenylalanine (BZF), which is capable of forming covalent cross-links after photoactivation by ultraviolet (UV) light, at three locations (G47, F48, and E50) in the PAS domain. We found that hERG-G47BZF channels had faster closing (deactivation) when irradiated in the open state (at 0 mV) but showed no measurable changes when irradiated in the closed state (at -100 mV). hERG-F48BZF channels had slower activation, faster deactivation, and a marked rightward shift in the voltage dependence of activation when irradiated in the open (at 0 mV) or closed (at -100 mV) state. hERG-E50BZF channels had no measurable changes when irradiated in the open state (at 0 mV) but had slower activation, faster deactivation, and a rightward shift in the voltage dependence of activation when irradiated in the closed state (at -100mV), indicating that hERG-E50BZF had a state-dependent difference in UV photoactivation, which we interpret to mean that PAS underwent molecular motions between the open and closed states. Moreover, we propose that UV-dependent biophysical changes in hERG-G47BZF, F48BZF, and E50BZF were the direct result of photochemical cross-linking that reduced dynamic motions in the PAS domain and broadly stabilized the closed state relative to the open state of the channel.
Collapse
Affiliation(s)
- Sara J Codding
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
5
|
Miller JM, Carlson AE. Locking hERG channels into place: Using photoreactive unnatural amino acids to study voltage gating. Biophys J 2024; 123:2358-2359. [PMID: 39033327 PMCID: PMC11365217 DOI: 10.1016/j.bpj.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Affiliation(s)
- Jennifer M Miller
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne E Carlson
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
6
|
Trudeau MC. A hydrophobic nexus at the heart of hERG K channel gating. Biophys J 2024; 123:1907-1909. [PMID: 38475996 PMCID: PMC11309969 DOI: 10.1016/j.bpj.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024] Open
Affiliation(s)
- Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
7
|
Stevens-Sostre WA, Flores-Aldama L, Bustos D, Li J, Morais-Cabral JH, Delemotte L, Robertson GA. An intracellular hydrophobic nexus critical for hERG1 channel slow deactivation. Biophys J 2024; 123:2024-2037. [PMID: 38219015 PMCID: PMC11309987 DOI: 10.1016/j.bpj.2024.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/17/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Slow deactivation is a critical property of voltage-gated K+ channels encoded by the human Ether-à-go-go-Related Gene 1 (hERG). hERG1 channel deactivation is modulated by interactions between intracellular N-terminal Per-Arnt-Sim (PAS) and C-terminal cyclic nucleotide-binding homology (CNBh) domains. The PAS domain is multipartite, comprising a globular domain (gPAS; residues 26-135) and an N-terminal PAS-cap that is further subdivided into an initial unstructured "tip" (residues 1-12) and an amphipathic α-helical region (residues 13-25). Although the PAS-cap tip has long been considered the effector of slow deactivation, how its position near the gating machinery is controlled has not been elucidated. Here, we show that a triad of hydrophobic interactions among the gPAS, PAS-cap α helix, and the CNBh domains is required to support slow deactivation in hERG1. The primary sequence of this "hydrophobic nexus" is highly conserved among mammalian ERG channels but shows key differences to fast-deactivating Ether-à-go-go 1 (EAG1) channels. Combining sequence analysis, structure-directed mutagenesis, electrophysiology, and molecular dynamics simulations, we demonstrate that polar serine substitutions uncover an intermediate deactivation mode that is also mimicked by deletion of the PAS-cap α helix. Molecular dynamics simulation analyses of the serine-substituted channels show an increase in distance among the residues of the hydrophobic nexus, a rotation of the intracellular gating ring, and a retraction of the PAS-cap tip from its receptor site near the voltage sensor domain and channel gate. These findings provide compelling evidence that the hydrophobic nexus coordinates the respective components of the intracellular gating ring and positions the PAS-cap tip to control hERG1 deactivation gating.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lisandra Flores-Aldama
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados Del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica Del Maule, Talca, Chile; Laboratorio de Bioinformática y Química Computacional (LBQC), Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica Del Maule, Talca, Chile
| | - Jin Li
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - João H Morais-Cabral
- Instituto de Investigação e Inovação Em Saude da Universidade Do Porto (i3S); Instituto de Biologia Molecular e Celular, Universidade Do Porto, Porto, Portugal
| | - Lucie Delemotte
- KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Gail A Robertson
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
8
|
Peters CH, Singh RK, Langley AA, Nichols WG, Ferris HR, Jeffrey DA, Proenza C, Bankston JR. LRMP inhibits cAMP potentiation of HCN4 channels by disrupting intramolecular signal transduction. eLife 2024; 12:RP92411. [PMID: 38652113 PMCID: PMC11037915 DOI: 10.7554/elife.92411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Lymphoid restricted membrane protein (LRMP) is a specific regulator of the hyperpolarization-activated cyclic nucleotide-sensitive isoform 4 (HCN4) channel. LRMP prevents cAMP-dependent potentiation of HCN4, but the interaction domains, mechanisms of action, and basis for isoform-specificity remain unknown. Here, we identify the domains of LRMP essential for this regulation, show that LRMP acts by disrupting the intramolecular signal transduction between cyclic nucleotide binding and gating, and demonstrate that multiple unique regions in HCN4 are required for LRMP isoform-specificity. Using patch clamp electrophysiology and Förster resonance energy transfer (FRET), we identified the initial 227 residues of LRMP and the N-terminus of HCN4 as necessary for LRMP to associate with HCN4. We found that the HCN4 N-terminus and HCN4-specific residues in the C-linker are necessary for regulation of HCN4 by LRMP. Finally, we demonstrated that LRMP-regulation can be conferred to HCN2 by addition of the HCN4 N-terminus along with mutation of five residues in the S5 region and C-linker to the cognate HCN4 residues. Taken together, these results suggest that LRMP inhibits HCN4 through an isoform-specific interaction involving the N-terminals of both proteins that prevents the transduction of cAMP binding into a change in channel gating, most likely via an HCN4-specific orientation of the N-terminus, C-linker, and S4-S5 linker.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Rohit K Singh
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
- Skaggs School of Pharmacy, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Avery A Langley
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - William G Nichols
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Hannah R Ferris
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Danielle A Jeffrey
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
9
|
Peters CH, Singh RK, Langley AA, Nichols WG, Ferris HR, Jeffrey DA, Proenza C, Bankston JR. LRMP inhibits cAMP potentiation of HCN4 channels by disrupting intramolecular signal transduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.29.555242. [PMID: 37693562 PMCID: PMC10491135 DOI: 10.1101/2023.08.29.555242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Lymphoid restricted membrane protein (LRMP) is a specific regulator of the hyperpolarization-activated cyclic nucleotide-sensitive isoform 4 (HCN4) channel. LRMP prevents cAMP-dependent potentiation of HCN4 but the interaction domains, mechanisms of action, and basis for isoform-specificity remain unknown. Here we identify the domains of LRMP essential for regulation. We show that LRMP acts by disrupting the intramolecular signal transduction between cyclic nucleotide binding and gating. And we demonstrate that multiple unique regions in HCN4 are required for LRMP isoform-specificity. Using patch clamp electrophysiology and Förster resonance energy transfer (FRET), we showed that the initial 227 residues of LRMP and the N-terminus of HCN4 are necessary for LRMP to interact with HCN4. We found that the HCN4 N-terminus and HCN4-specific residues in the C-linker are necessary for regulation of HCN4 by LRMP. And we demonstrate that LRMP-regulation can be conferred to HCN2 by addition of the HCN4 N-terminus along with mutation of 5 residues in the S5 region and C-linker to the cognate HCN4 residues. Taken together, these results suggest that LRMP inhibits HCN4 through an isoform-specific interaction involving the N-terminals of both proteins that prevents the transduction of cAMP binding into a change in channel gating via an HCN4-specific orientation of the N-terminus, C-linker, and S4-S5 linker.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Rohit K Singh
- Skaggs School of Pharmacy, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Boulevard, Aurora, CO 80045
| | - Avery A Langley
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - William G Nichols
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Hannah R Ferris
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Danielle A Jeffrey
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, 12631 E. 17 Avenue, Aurora, CO 80045
| | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, 12800 E. 19 Avenue, Aurora, CO 80045
| |
Collapse
|
10
|
Codding SJ, Trudeau MC. Photo-crosslinking hERG channels causes a U.V.-driven, state-dependent disruption of kinetics and voltage dependence of activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574834. [PMID: 38260338 PMCID: PMC10802430 DOI: 10.1101/2024.01.09.574834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Human ether-à-go-go related gene (hERG) voltage-activated potassium channels are critical for cardiac excitability. Characteristic slow closing (deactivation) in hERG is regulated by direct interaction between the N-terminal Per-Arnt-Sim (PAS) domain and the C-terminal cyclic nucleotide binding homology domain (CNBHD). We aim to understand how the PAS domain that is distal to the pore rearranges during gating to allosterically regulate the channel pore (and ion flux). To achieve this, we utilized the non-canonical amino acid 4-Benzoyl-L-phenylalanine (BZF) which is a photo-activatable cross-linkable probe, that when irradiated with ultraviolet (U.V.) light forms a double radical capable of forming covalent cross-links with C-H bond-containing groups, enabling selective and potent U.V.-driven photoinactivation of ion channel dynamics. Here we incorporate BZF directly into the hERG potassium channel PAS domain at three locations (G47, F48, and E50) using TAG codon suppression technology. hERG channels with BZF incorporated into the PAS domain (hERG-BZF) showed a significant change in the biophysical properties of the channel. hERG-G47BZF activated slowly when irradiated in the closed state (-100mV) but deactivated quickly when irradiated in both the open (0mV) and closed state. hERG-F48BZF channels showed a state independent and U.V. dose-dependent change in channel activation (slowing down) and channel deactivation (speeding up), as well as a marked change (right-shift) in the voltage-dependence of conductance. When irradiated at -100 mV hERG-E50BZF showed a state dependent and U.V. dose-dependent change in a channel activation (slowing down) and deactivation (speeding up) of channel deactivation, as well as a marked change (right-shift) in the voltage-dependence of conductance that occurred only when the channel was irradiated in the closed state (-100mV). This approach demonstrated that direct photo-crosslinking of the PAS domain in hERG channels causes a measurable change in biophysical parameters and more broadly stabilized the closed state of the channel. We propose that altered channel gating is as a direct result of reduced dynamic motions in the PAS domain of hERG due to photo-chemical crosslinking.
Collapse
Affiliation(s)
- Sara J Codding
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
11
|
Ukachukwu CU, Jimenez-Vazquez EN, Jain A, Jones DK. hERG1 channel subunit composition mediates proton inhibition of rapid delayed rectifier potassium current (I Kr) in cardiomyocytes derived from hiPSCs. J Biol Chem 2023; 299:102778. [PMID: 36496073 PMCID: PMC9867984 DOI: 10.1016/j.jbc.2022.102778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/29/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
The voltage-gated channel, hERG1, conducts the rapid delayed rectifier potassium current (IKr) and is critical for human cardiac repolarization. Reduced IKr causes long QT syndrome and increases the risk for cardiac arrhythmia and sudden death. At least two subunits form functional hERG1 channels, hERG1a and hERG1b. Changes in hERG1a/1b abundance modulate IKr kinetics, magnitude, and drug sensitivity. Studies from native cardiac tissue suggest that hERG1 subunit abundance is dynamically regulated, but the impact of altered subunit abundance on IKr and its response to external stressors is not well understood. Here, we used a substrate-driven human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) maturation model to investigate how changes in relative hERG1a/1b subunit abundance impact the response of native IKr to extracellular acidosis, a known component of ischemic heart disease and sudden infant death syndrome. IKr recorded from immatured hiPSC-CMs displays a 2-fold greater inhibition by extracellular acidosis (pH 6.3) compared with matured hiPSC-CMs. Quantitative RT-PCR and immunocytochemistry demonstrated that hERG1a subunit mRNA and protein were upregulated and hERG1b subunit mRNA and protein were downregulated in matured hiPSC-CMs compared with immatured hiPSC-CMs. The shift in subunit abundance in matured hiPSC-CMs was accompanied by increased IKr. Silencing hERG1b's impact on native IKr kinetics by overexpressing a polypeptide identical to the hERG1a N-terminal Per-Arnt-Sim domain reduced the magnitude of IKr proton inhibition in immatured hiPSC-CMs to levels comparable to those observed in matured hiPSC-CMs. These data demonstrate that hERG1 subunit abundance is dynamically regulated and determines IKr proton sensitivity in hiPSC-CMs.
Collapse
Affiliation(s)
- Chiamaka U Ukachukwu
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Eric N Jimenez-Vazquez
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Abhilasha Jain
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David K Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Department of Internal Medicine, University of Michigan Medical School.
| |
Collapse
|
12
|
KCNH2 encodes a nuclear-targeted polypeptide that mediates hERG1 channel gating and expression. Proc Natl Acad Sci U S A 2023; 120:e2214700120. [PMID: 36626562 PMCID: PMC9934303 DOI: 10.1073/pnas.2214700120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
KCNH2 encodes hERG1, the voltage-gated potassium channel that conducts the rapid delayed rectifier potassium current (IKr) in human cardiac tissue. hERG1 is one of the first channels expressed during early cardiac development, and its dysfunction is associated with intrauterine fetal death, sudden infant death syndrome, cardiac arrhythmia, and sudden cardiac death. Here, we identified a hERG1 polypeptide (hERG1NP) that is targeted to the nuclei of immature cardiac cells, including human stem cell-derived cardiomyocytes (hiPSC-CMs) and neonatal rat cardiomyocytes. The nuclear hERG1NP immunofluorescent signal is diminished in matured hiPSC-CMs and absent from adult rat cardiomyocytes. Antibodies targeting distinct hERG1 channel epitopes demonstrated that the hERG1NP signal maps to the hERG1 distal C-terminal domain. KCNH2 deletion using CRISPR simultaneously abolished IKr and the hERG1NP signal in hiPSC-CMs. We then identified a putative nuclear localization sequence (NLS) within the distal hERG1 C-terminus, 883-RQRKRKLSFR-892. Interestingly, the distal C-terminal domain was targeted almost exclusively to the nuclei when overexpressed HEK293 cells. Conversely, deleting the NLS from the distal peptide abolished nuclear targeting. Similarly, blocking α or β1 karyopherin activity diminished nuclear targeting. Finally, overexpressing the putative hERG1NP peptide in the nuclei of HEK cells significantly reduced hERG1a current density, compared to cells expressing the NLS-deficient hERG1NP or GFP. These data identify a developmentally regulated polypeptide encoded by KCNH2, hERG1NP, whose presence in the nucleus indirectly modulates hERG1 current magnitude and kinetics.
Collapse
|
13
|
Zangerl-Plessl EM, Wu W, Sanguinetti MC, Stary-Weinzinger A. Binding of RPR260243 at the intracellular side of the hERG1 channel pore domain slows closure of the helix bundle crossing gate. Front Mol Biosci 2023; 10:1137368. [PMID: 36911523 PMCID: PMC9996038 DOI: 10.3389/fmolb.2023.1137368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
The opening and closing of voltage-dependent potassium channels is dependent on a tight coupling between movement of the voltage sensing S4 segments and the activation gate. A specific interaction between intracellular amino- and carboxyl-termini is required for the characteristically slow rate of channel closure (deactivation) of hERG1 channels. Compounds that increase hERG1 channel currents represent a novel approach for prevention of arrhythmia associated with prolonged ventricular repolarization. RPR260243 (RPR), a quinoline oxo-propyl piperidine derivative, inhibits inactivation and dramatically slows the rate of hERG1 channel deactivation. Here we report that similar to its effect on wild-type channels, RPR greatly slows the deactivation rate of hERG1 channels missing their amino-termini, or of split channels lacking a covalent link between the voltage sensor domain and the pore domain. By contrast, RPR did not slow deactivation of C-terminal truncated hERG1 channels or D540K hERG1 mutant channels activated by hyperpolarization. Together, these findings indicate that ability of RPR to slow deactivation requires an intact C-terminus, does not slow deactivation by stabilizing an interaction involving the amino-terminus or require a covalent link between the voltage sensor and pore domains. All-atom molecular dynamics simulations using the cryo-EM structure of the hERG1 channel revealed that RPR binds to a pocket located at the intracellular ends of helices S5 and S6 of a single subunit. The slowing of channel deactivation by RPR may be mediated by disruption of normal S5-S6 interactions.
Collapse
Affiliation(s)
| | - Wei Wu
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research & Training Institute, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, United States
| | - Michael C Sanguinetti
- 3 Department of Internal Medicine, Division of Cardiovascular Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt LakeCity, UT, United States
| | | |
Collapse
|
14
|
Johnson AA, Crawford TR, Trudeau MC. The N-linker region of hERG1a upregulates hERG1b potassium channels. J Biol Chem 2022; 298:102233. [PMID: 35798139 PMCID: PMC9428852 DOI: 10.1016/j.jbc.2022.102233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 11/29/2022] Open
Abstract
A major physiological role of hERG1 (human Ether-á-go-go-Related Gene 1) potassium channels is to repolarize cardiac action potentials. Two isoforms, hERG1a and hERG1b, associate to form the potassium current IKr in cardiomyocytes. Inherited mutations in hERG1a or hERG1b cause prolonged cardiac repolarization, long QT syndrome, and sudden death arrhythmia. hERG1a subunits assemble with and enhance the number of hERG1b subunits at the plasma membrane, but the mechanism for the increase in hERG1b by hERG1a is not well understood. Here, we report that the hERG1a N-terminal region expressed in trans with hERG1b markedly increased hERG1b currents and increased biotin-labeled hERG1b protein at the membrane surface. hERG1b channels with a deletion of the N-terminal 1b domain did not have a measurable increase in current or biotinylated protein when coexpressed with hERG1a N-terminal regions, indicating that the 1b domain was required for the increase in hERG1b. Using a biochemical pull-down interaction assay and a FRET hybridization experiment, we detected a direct interaction between the hERG1a N-terminal region and the hERG1b N-terminal region. Using engineered deletions and alanine mutagenesis, we identified a short span of amino acids at positions 216 to 220 within the hERG1a "N-linker" region that were necessary for the upregulation of hERG1b. We propose that direct structural interactions between the hERG1a N-linker region and the hERG1b 1b domain increase hERG1b at the plasma membrane. Mechanisms regulating hERG1a and hERG1b are likely critical for cardiac function, may be disrupted by long QT syndrome mutants, and serve as potential targets for therapeutics.
Collapse
Affiliation(s)
- Ashley A Johnson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Taylor R Crawford
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Al Salmani MK, Tavakoli R, Zaman W, Al Harrasi A. Multiple mechanisms underlie reduced potassium conductance in the p.T1019PfsX38 variant of hERG. Physiol Rep 2022; 10:e15341. [PMID: 35854468 PMCID: PMC9296870 DOI: 10.14814/phy2.15341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
Long QT syndrome type II (LQT2) is caused by loss-of-function mutations in the hERG K+ channel, leading to increased incidence of cardiac arrest and sudden death. Many genetic variants have been reported in the hERG gene with various consequences on channel expression, permeation, and gating. Only a small number of LQT2 causing variants has been characterized to define the underlying pathophysiological causes of the disease. We sought to determine the characteristics of the frameshift variant p.Thr1019ProfsX38 (T1019PfsX38) which affects the C-terminus of the protein. This mutation was identified in an extended Omani family of LQT2. It replaces the last 140 amino acids of hERG with 37 unique amino acids. T1019 is positioned at a distinguished region of the C-terminal tail of hERG, as predicted from the deep learning system AlphaFold v2.0. We employed the whole-cell configuration of the patch-clamp technique to study wild-type and mutant channels that were transiently expressed in human embryonic kidney 293 (HEK293) cells. Depolarizing voltages elicited slowly deactivating tail currents that appeared upon repolarization of cells that express either wild-type- or T1019PfsX38-hERG. There were no differences in the voltage and time dependencies of activation between the two variants. However, the rates of hERG channel deactivation at hyperpolarizing potentials were accelerated by T1019PfsX38. In addition, the voltage dependence of inactivation of T1019PfsX38-hERG was shifted by 20 mV in the negative direction when compared with wild-type hERG. The rates of channel inactivation were increased in the mutant channel variant. Next, we employed a step-ramp protocol to mimic membrane repolarization by the cardiac action potential. The amplitudes of outward currents and their integrals were reduced in the mutant variant when compared with the wild-type variant during repolarization. Thus, changes in the gating dynamics of hERG by the T1019PfsX38 variant contribute to the pathology seen in affected LQT2 patients.
Collapse
Affiliation(s)
| | - Rezvan Tavakoli
- Natural and Medical Sciences Research CenterUniversity of NizwaNizwaOman
| | - Wajid Zaman
- Natural and Medical Sciences Research CenterUniversity of NizwaNizwaOman
| | - Ahmed Al Harrasi
- Natural and Medical Sciences Research CenterUniversity of NizwaNizwaOman
| |
Collapse
|
16
|
Jones DK. Hysteretic hERG channel gating current recorded at physiological temperature. Sci Rep 2022; 12:5950. [PMID: 35396394 PMCID: PMC8993916 DOI: 10.1038/s41598-022-10003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/24/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiac hERG channels comprise at least two subunits, hERG 1a and hERG 1b, and drive cardiac action potential repolarization. hERG 1a subunits contain a cytoplasmic PAS domain that is absent in hERG 1b. The hERG 1a PAS domain regulates voltage sensor domain (VSD) movement, but hERG VSD behavior and its regulation by the hERG 1a PAS domain have not been studied at physiological temperatures. We recorded gating charge from homomeric hERG 1a and heteromeric hERG 1a/1b channels at near physiological temperatures (36 ± 1 °C) using pulse durations comparable in length to the human ventricular action potential. The voltage dependence of deactivation was hyperpolarized relative to activation, reflecting VSD relaxation at positive potentials. These data suggest that relaxation (hysteresis) works to delay pore closure during repolarization. Interestingly, hERG 1a VSD deactivation displayed a double Boltzmann distribution, but hERG 1a/1b deactivation displayed a single Boltzmann. Disabling the hERG 1a PAS domain using a PAS-targeting antibody similarly transformed hERG 1a deactivation from a double to a single Boltzmann, highlighting the contribution of the PAS in regulating VSD movement. These data represent, to our knowledge, the first recordings of hERG gating charge at physiological temperature and demonstrate that VSD relaxation (hysteresis) is present in hERG channels at physiological temperature.
Collapse
Affiliation(s)
- David K Jones
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA. .,Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
17
|
Soohoo SM, Tiwari PB, Suzuki YJ, Brelidze TI. Investigation of PAS and CNBH domain interactions in hERG channels and effects of long-QT syndrome-causing mutations with surface plasmon resonance. J Biol Chem 2021; 298:101433. [PMID: 34801551 PMCID: PMC8693265 DOI: 10.1016/j.jbc.2021.101433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Human ether-á-go-go-related gene (hERG) channels are key regulators of cardiac repolarization, neuronal excitability, and tumorigenesis. hERG channels contain N-terminal Per-Arnt-Sim (PAS) and C-terminal cyclic nucleotide-binding homology (CNBH) domains with many long-QT syndrome (LQTS)-causing mutations located at the interface between these domains. Despite the importance of PAS/CNBH domain interactions, little is known about their affinity. Here, we used the surface plasmon resonance (SPR) technique to investigate interactions between isolated PAS and CNBH domains and the effects of LQTS-causing mutations R20G, N33T, and E58D, located at the PAS/CNBH domain interface, on these interactions. We determined that the affinity of the PAS/CNBH domain interactions was ∼1.4 μM. R20G and E58D mutations had little effect on the domain interaction affinity, while N33T abolished the domain interactions. Interestingly, mutations in the intrinsic ligand, a conserved stretch of amino acids occupying the beta-roll cavity in the CNBH domain, had little effect on the affinity of PAS/CNBH domain interactions. Additionally, we determined that the isolated PAS domains formed oligomers with an interaction affinity of ∼1.6 μM. Coexpression of the isolated PAS domains with the full-length hERG channels or addition of the purified PAS protein inhibited hERG currents. These PAS/PAS interactions can have important implications for hERG function in normal and pathological conditions associated with increased surface density of channels or interaction with other PAS-domain-containing proteins. Taken together, our study provides the first account of the binding affinities for wild-type and mutant hERG PAS and CNBH domains and highlights the potential functional significance of PAS/PAS domain interactions.
Collapse
Affiliation(s)
- Stephanie M Soohoo
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Purushottam B Tiwari
- Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Yuichiro J Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA.
| |
Collapse
|
18
|
Conformation-sensitive antibody reveals an altered cytosolic PAS/CNBh assembly during hERG channel gating. Proc Natl Acad Sci U S A 2021; 118:2108796118. [PMID: 34716268 DOI: 10.1073/pnas.2108796118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/22/2021] [Indexed: 11/18/2022] Open
Abstract
The human ERG (hERG) K+ channel has a crucial function in cardiac repolarization, and mutations or channel block can give rise to long QT syndrome and catastrophic ventricular arrhythmias. The cytosolic assembly formed by the Per-Arnt-Sim (PAS) and cyclic nucleotide binding homology (CNBh) domains is the defining structural feature of hERG and related KCNH channels. However, the molecular role of these two domains in channel gating remains unclear. We have previously shown that single-chain variable fragment (scFv) antibodies can modulate hERG function by binding to the PAS domain. Here, we mapped the scFv2.12 epitope to a site overlapping with the PAS/CNBh domain interface using NMR spectroscopy and mutagenesis and show that scFv binding in vitro and in the cell is incompatible with the PAS interaction with CNBh. By generating a fluorescently labeled scFv2.12, we demonstrate that association with the full-length hERG channel is state dependent. We detect Förster resonance energy transfer (FRET) with scFv2.12 when the channel gate is open but not when it is closed. In addition, state dependence of scFv2.12 FRET signal disappears when the R56Q mutation, known to destabilize the PAS-CNBh interaction, is introduced in the channel. Altogether, these data are consistent with an extensive structural alteration of the PAS/CNBh assembly when the cytosolic gate opens, likely favoring PAS domain dissociation from the CNBh domain.
Collapse
|
19
|
Kemp JM, Whittaker DG, Venkateshappa R, Pang Z, Johal R, Sergeev V, Tibbits GF, Mirams GR, Claydon TW. Electrophysiological characterization of the hERG R56Q LQTS variant and targeted rescue by the activator RPR260243. J Gen Physiol 2021; 153:212555. [PMID: 34398210 PMCID: PMC8493834 DOI: 10.1085/jgp.202112923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/11/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022] Open
Abstract
Human Ether-à-go-go (hERG) channels contribute to cardiac repolarization, and inherited variants or drug block are associated with long QT syndrome type 2 (LQTS2) and arrhythmia. Therefore, hERG activator compounds present a therapeutic opportunity for targeted treatment of LQTS. However, a limiting concern is over-activation of hERG resurgent current during the action potential and abbreviated repolarization. Activators that slow deactivation gating (type I), such as RPR260243, may enhance repolarizing hERG current during the refractory period, thus ameliorating arrhythmogenicity with reduced early repolarization risk. Here, we show that, at physiological temperature, RPR260243 enhances hERG channel repolarizing currents conducted in the refractory period in response to premature depolarizations. This occurs with little effect on the resurgent hERG current during the action potential. The effects of RPR260243 were particularly evident in LQTS2-associated R56Q mutant channels, whereby RPR260243 restored WT-like repolarizing drive in the early refractory period and diastolic interval, combating attenuated protective currents. In silico kinetic modeling of channel gating predicted little effect of the R56Q mutation on hERG current conducted during the action potential and a reduced repolarizing protection against afterdepolarizations in the refractory period and diastolic interval, particularly at higher pacing rates. These simulations predicted partial rescue from the arrhythmic effects of R56Q by RPR260243 without risk of early repolarization. Our findings demonstrate that the pathogenicity of some hERG variants may result from reduced repolarizing protection during the refractory period and diastolic interval with limited effect on action potential duration, and that the hERG channel activator RPR260243 may provide targeted antiarrhythmic potential in these cases.
Collapse
Affiliation(s)
- Jacob M Kemp
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Dominic G Whittaker
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | | | - ZhaoKai Pang
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Raj Johal
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Valentine Sergeev
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Glen F Tibbits
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK
| | - Thomas W Claydon
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
20
|
Codding SJ, Johnson AA, Trudeau MC. Gating and regulation of KCNH (ERG, EAG, and ELK) channels by intracellular domains. Channels (Austin) 2021; 14:294-309. [PMID: 32924766 PMCID: PMC7515569 DOI: 10.1080/19336950.2020.1816107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The KCNH family comprises the ERG, EAG, and ELK voltage-activated, potassium-selective channels. Distinct from other K channels, KCNH channels contain unique structural domains, including a PAS (Per-Arnt-Sim) domain in the N-terminal region and a CNBHD (cyclic nucleotide-binding homology domain) in the C-terminal region. The intracellular PAS domains and CNBHDs interact directly and regulate some of the characteristic gating properties of each type of KCNH channel. The PAS-CNBHD interaction regulates slow closing (deactivation) of hERG channels, the kinetics of activation and pre-pulse dependent population of closed states (the Cole-Moore shift) in EAG channels and voltage-dependent potentiation in ELK channels. KCNH channels are all regulated by an intrinsic ligand motif in the C-terminal region which binds to the CNBHD. Here, we focus on some recent advances regarding the PAS-CNBHD interaction and the intrinsic ligand.
Collapse
Affiliation(s)
- Sara J Codding
- Department of Physiology, University of Maryland School of Medicine , Baltimore, MD, USA
| | - Ashley A Johnson
- Department of Physiology, University of Maryland School of Medicine , Baltimore, MD, USA
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine , Baltimore, MD, USA
| |
Collapse
|
21
|
Ben-Bassat A, Giladi M, Haitin Y. Structure of KCNH2 cyclic nucleotide-binding homology domain reveals a functionally vital salt-bridge. J Gen Physiol 2021; 152:151568. [PMID: 32191791 PMCID: PMC7141593 DOI: 10.1085/jgp.201912505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/24/2020] [Accepted: 02/12/2020] [Indexed: 01/04/2023] Open
Abstract
Human KCNH2 channels (hKCNH2, ether-à-go-go [EAG]–related gene, hERG) are best known for their contribution to cardiac action potential repolarization and have key roles in various pathologies. Like other KCNH family members, hKCNH2 channels contain a unique intracellular complex, consisting of an N-terminal eag domain and a C-terminal cyclic nucleotide-binding homology domain (CNBHD), which is crucial for channel function. Previous studies demonstrated that the CNBHD is occupied by an intrinsic ligand motif, in a self-liganded conformation, providing a structural mechanism for the lack of KCNH channel regulation by cyclic nucleotides. While there have been significant advancements in the structural and functional characterization of the CNBHD of KCNH channels, a high-resolution structure of the hKCNH2 intracellular complex has been missing. Here, we report the 1.5 Å resolution structure of the hKCNH2 channel CNBHD. The structure reveals the canonical fold shared by other KCNH family members, where the spatial organization of the intrinsic ligand is preserved within the β-roll region. Moreover, measurements of small-angle x-ray scattering profile in solution, as well as comparison with a recent NMR analysis of hKCNH2, revealed high agreement with the crystallographic structure, indicating an overall low flexibility in solution. Importantly, we identified a novel salt-bridge (E807-R863) which was not previously resolved in the NMR and cryo-EM structures. Electrophysiological analysis of charge-reversal mutations revealed the bridge’s crucial role in hKCNH2 function. Moreover, comparison with other KCNH members revealed the structural conservation of this salt-bridge, consistent with its functional significance. Together with the available structure of the mouse KCNH1 intracellular complex and previous electrophysiological and spectroscopic studies of KCNH family members, we propose that this salt-bridge serves as a strategically positioned linchpin to support both the spatial organization of the intrinsic ligand and the maintenance of the intracellular complex interface.
Collapse
Affiliation(s)
- Ariel Ben-Bassat
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Giladi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yoni Haitin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
22
|
Zequn Z, Jiangfang L. Molecular Insights Into the Gating Kinetics of the Cardiac hERG Channel, Illuminated by Structure and Molecular Dynamics. Front Pharmacol 2021; 12:687007. [PMID: 34168566 PMCID: PMC8217747 DOI: 10.3389/fphar.2021.687007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/20/2021] [Indexed: 11/13/2022] Open
Abstract
The rapidly activating delayed rectifier K+ current generated by the cardiac hERG potassium channel encoded by KCNH2 is the most important reserve current for cardiac repolarization. The unique inward rectification characteristics of the hERG channel depend on the gating regulation, which involves crucial structural domains and key single amino acid residues in the full-length hERG channel. Identifying critical molecules involved in the regulation of gating kinetics for the hERG channel requires high-resolution structures and molecular dynamics simulation models. Based on the latest progress in hERG structure and molecular dynamics simulation research, summarizing the molecules involved in the changes in the channel state helps to elucidate the unique gating characteristics of the channel and the reason for its high affinity to cardiotoxic drugs. In this review, we aim to summarize the significant advances in understanding the voltage gating regulation of the hERG channel based on its structure obtained from cryo-electron microscopy and computer simulations, which reveal the critical roles of several specific structural domains and amino acid residues.
Collapse
Affiliation(s)
- Zheng Zequn
- Department of Cardiovascular, Medical College, Ningbo University, Ningbo, China
| | - Lian Jiangfang
- Department of Cardiovascular, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| |
Collapse
|
23
|
Abstract
Long QT syndrome (LQTS) is a cardiovascular disorder characterized by an abnormality in cardiac repolarization leading to a prolonged QT interval and T-wave irregularities on the surface electrocardiogram. It is commonly associated with syncope, seizures, susceptibility to torsades de pointes, and risk for sudden death. LQTS is a rare genetic disorder and a major preventable cause of sudden cardiac death in the young. The availability of therapy for this lethal disease emphasizes the importance of early and accurate diagnosis. Additionally, understanding of the molecular mechanisms underlying LQTS could help to optimize genotype-specific treatments to prevent deaths in LQTS patients. In this review, we briefly summarize current knowledge regarding molecular underpinning of LQTS, in particular focusing on LQT1, LQT2, and LQT3, and discuss novel strategies to study ion channel dysfunction and drug-specific therapies in LQT1, LQT2, and LQT3 syndromes.
Collapse
Affiliation(s)
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
24
|
Burton MJ, Cresser-Brown J, Thomas M, Portolano N, Basran J, Freeman SL, Kwon H, Bottrill AR, Llansola-Portoles MJ, Pascal AA, Jukes-Jones R, Chernova T, Schmid R, Davies NW, Storey NM, Dorlet P, Moody PCE, Mitcheson JS, Raven EL. Discovery of a heme-binding domain in a neuronal voltage-gated potassium channel. J Biol Chem 2020; 295:13277-13286. [PMID: 32723862 DOI: 10.1074/jbc.ra120.014150] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
The EAG (ether-à-go-go) family of voltage-gated K+ channels are important regulators of neuronal and cardiac action potential firing (excitability) and have major roles in human diseases such as epilepsy, schizophrenia, cancer, and sudden cardiac death. A defining feature of EAG (Kv10-12) channels is a highly conserved domain on the N terminus, known as the eag domain, consisting of a Per-ARNT-Sim (PAS) domain capped by a short sequence containing an amphipathic helix (Cap domain). The PAS and Cap domains are both vital for the normal function of EAG channels. Using heme-affinity pulldown assays and proteomics of lysates from primary cortical neurons, we identified that an EAG channel, hERG3 (Kv11.3), binds to heme. In whole-cell electrophysiology experiments, we identified that heme inhibits hERG3 channel activity. In addition, we expressed the Cap and PAS domain of hERG3 in Escherichia coli and, using spectroscopy and kinetics, identified the PAS domain as the location for heme binding. The results identify heme as a regulator of hERG3 channel activity. These observations are discussed in the context of the emerging role for heme as a regulator of ion channel activity in cells.
Collapse
Affiliation(s)
- Mark J Burton
- Department of Chemistry, University of Leicester, Leicester, United Kingdom; Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | | | - Morgan Thomas
- Department of Chemistry, University of Leicester, Leicester, United Kingdom; Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | - Nicola Portolano
- Department of Chemistry, University of Leicester, Leicester, United Kingdom; Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | - Jaswir Basran
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Samuel L Freeman
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Hanna Kwon
- School of Chemistry, University of Bristol, Bristol, United Kingdom
| | - Andrew R Bottrill
- Protein Nucleic Acid Chemistry Laboratory, University of Leicester, Leicester, United Kingdom
| | - Manuel J Llansola-Portoles
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Andrew A Pascal
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell, Gif-sur-Yvette, France
| | - Rebekah Jukes-Jones
- Medical Research Council Toxicology Unit, University of Cambridge, Leicester, United Kingdom
| | - Tatyana Chernova
- Medical Research Council Toxicology Unit, University of Cambridge, Leicester, United Kingdom
| | - Ralf Schmid
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Noel W Davies
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Nina M Storey
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom; Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Pierre Dorlet
- CNRS, Aix Marseille Université, Laboratoire de Bioenergetique et d'Ingenierie des Protéines, Marseille, France
| | - Peter C E Moody
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - John S Mitcheson
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - Emma L Raven
- School of Chemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
25
|
Wang ZJ, Blanco I, Hayoz S, Brelidze TI. The HCN domain is required for HCN channel cell-surface expression and couples voltage- and cAMP-dependent gating mechanisms. J Biol Chem 2020; 295:8164-8173. [PMID: 32341127 DOI: 10.1074/jbc.ra120.013281] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/23/2020] [Indexed: 11/06/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are major regulators of synaptic plasticity and rhythmic activity in the heart and brain. Opening of HCN channels requires membrane hyperpolarization and is further facilitated by intracellular cyclic nucleotides (cNMPs). In HCN channels, membrane hyperpolarization is sensed by the membrane-spanning voltage sensor domain (VSD), and the cNMP-dependent gating is mediated by the intracellular cyclic nucleotide-binding domain (CNBD) connected to the pore-forming S6 transmembrane segment via the C-linker. Previous functional analysis of HCN channels has suggested a direct or allosteric coupling between the voltage- and cNMP-dependent activation mechanisms. However, the specifics of this coupling remain unclear. The first cryo-EM structure of an HCN1 channel revealed that a novel structural element, dubbed the HCN domain (HCND), forms a direct structural link between the VSD and C-linker-CNBD. In this study, we investigated the functional significance of the HCND. Deletion of the HCND prevented surface expression of HCN2 channels. Based on the HCN1 structure analysis, we identified Arg237 and Gly239 residues on the S2 of the VSD that form direct interactions with Ile135 on the HCND. Disrupting these interactions abolished HCN2 currents. We also identified three residues on the C-linker-CNBD (Glu478, Gln482, and His559) that form direct interactions with residues Arg154 and Ser158 on the HCND. Disrupting these interactions affected both voltage- and cAMP-dependent gating of HCN2 channels. These findings indicate that the HCND is necessary for the cell-surface expression of HCN channels and provides a functional link between voltage- and cAMP-dependent mechanisms of HCN channel gating.
Collapse
Affiliation(s)
- Ze-Jun Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., USA
| | - Ismary Blanco
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D. C., USA
| | - Sebastien Hayoz
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., USA
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., USA .,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D. C., USA
| |
Collapse
|
26
|
Barros F, de la Peña P, Domínguez P, Sierra LM, Pardo LA. The EAG Voltage-Dependent K + Channel Subfamily: Similarities and Differences in Structural Organization and Gating. Front Pharmacol 2020; 11:411. [PMID: 32351384 PMCID: PMC7174612 DOI: 10.3389/fphar.2020.00411] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/18/2020] [Indexed: 12/17/2022] Open
Abstract
EAG (ether-à-go-go or KCNH) are a subfamily of the voltage-gated potassium (Kv) channels. Like for all potassium channels, opening of EAG channels drives the membrane potential toward its equilibrium value for potassium, thus setting the resting potential and repolarizing action potentials. As voltage-dependent channels, they switch between open and closed conformations (gating) when changes in membrane potential are sensed by a voltage sensing domain (VSD) which is functionally coupled to a pore domain (PD) containing the permeation pathway, the potassium selectivity filter, and the channel gate. All Kv channels are tetrameric, with four VSDs formed by the S1-S4 transmembrane segments of each subunit, surrounding a central PD with the four S5-S6 sections arranged in a square-shaped structure. Structural information, mutagenesis, and functional experiments, indicated that in "classical/Shaker-type" Kv channels voltage-triggered VSD reorganizations are transmitted to PD gating via the α-helical S4-S5 sequence that links both modules. Importantly, these Shaker-type channels share a domain-swapped VSD/PD organization, with each VSD contacting the PD of the adjacent subunit. In this case, the S4-S5 linker, acting as a rigid mechanical lever (electromechanical lever coupling), would lead to channel gate opening at the cytoplasmic S6 helices bundle. However, new functional data with EAG channels split between the VSD and PD modules indicate that, in some Kv channels, alternative VSD/PD coupling mechanisms do exist. Noticeably, recent elucidation of the architecture of some EAG channels, and other relatives, showed that their VSDs are non-domain swapped. Despite similarities in primary sequence and predicted structural organization for all EAG channels, they show marked kinetic differences whose molecular basis is not completely understood. Thus, while a common general architecture may establish the gating system used by the EAG channels and the physicochemical coupling of voltage sensing to gating, subtle changes in that common structure, and/or allosteric influences of protein domains relatively distant from the central gating machinery, can crucially influence the gating process. We consider here the latest advances on these issues provided by the elucidation of eag1 and erg1 three-dimensional structures, and by both classical and more recent functional studies with different members of the EAG subfamily.
Collapse
Affiliation(s)
- Francisco Barros
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Edificio Santiago Gascón, Oviedo, Spain
| | - Pilar de la Peña
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Edificio Santiago Gascón, Oviedo, Spain
| | - Pedro Domínguez
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Edificio Santiago Gascón, Oviedo, Spain
| | - Luisa Maria Sierra
- Departamento de Biología Funcional (Area de Genética), Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Oviedo, Spain
| | - Luis A. Pardo
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
27
|
Wang ZJ, Soohoo SM, Tiwari PB, Piszczek G, Brelidze TI. Chlorpromazine binding to the PAS domains uncovers the effect of ligand modulation on EAG channel activity. J Biol Chem 2020; 295:4114-4123. [PMID: 32047112 PMCID: PMC7105296 DOI: 10.1074/jbc.ra119.012377] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
Ether-a-go-go (EAG) potassium selective channels are major regulators of neuronal excitability and cancer progression. EAG channels contain a Per-Arnt-Sim (PAS) domain in their intracellular N-terminal region. The PAS domain is structurally similar to the PAS domains in non-ion channel proteins, where these domains frequently function as ligand-binding domains. Despite the structural similarity, it is not known whether the PAS domain can regulate EAG channel function via ligand binding. Here, using surface plasmon resonance, tryptophan fluorescence, and analysis of EAG currents recorded in Xenopus laevis oocytes, we show that a small molecule chlorpromazine (CH), widely used as an antipsychotic medication, binds to the isolated PAS domain of EAG channels and inhibits currents from these channels. Mutant EAG channels that lack the PAS domain show significantly lower inhibition by CH, suggesting that CH affects currents from EAG channels directly through the binding to the PAS domain. Our study lends support to the hypothesis that there are previously unaccounted steps in EAG channel gating that could be activated by ligand binding to the PAS domain. This has broad implications for understanding gating mechanisms of EAG and related ERG and ELK K+ channels and places the PAS domain as a new target for drug discovery in EAG and related channels. Up-regulation of EAG channel activity is linked to cancer and neurological disorders. Our study raises the possibility of repurposing the antipsychotic drug chlorpromazine for treatment of neurological disorders and cancer.
Collapse
Affiliation(s)
- Ze-Jun Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., 20057
| | - Stephanie M Soohoo
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., 20057
| | - Purushottam B Tiwari
- Department of Oncology, Georgetown University Medical Center, Washington, D. C., 20057
| | - Grzegorz Piszczek
- Biophysics Core, NHLBI, National Institutes of Health, Bethesda, Maryland 20892
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D. C., 20057.
| |
Collapse
|
28
|
Shi YP, Thouta S, Claydon TW. Modulation of hERG K + Channel Deactivation by Voltage Sensor Relaxation. Front Pharmacol 2020; 11:139. [PMID: 32184724 PMCID: PMC7059196 DOI: 10.3389/fphar.2020.00139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022] Open
Abstract
The hERG (human-ether-à-go-go-related gene) channel underlies the rapid delayed rectifier current, Ikr, in the heart, which is essential for normal cardiac electrical activity and rhythm. Slow deactivation is one of the hallmark features of the unusual gating characteristics of hERG channels, and plays a crucial role in providing a robust current that aids repolarization of the cardiac action potential. As such, there is significant interest in elucidating the underlying mechanistic determinants of slow hERG channel deactivation. Recent work has shown that the hERG channel S4 voltage sensor is stabilized following activation in a process termed relaxation. Voltage sensor relaxation results in energetic separation of the activation and deactivation pathways, producing a hysteresis, which modulates the kinetics of deactivation gating. Despite widespread observation of relaxation behaviour in other voltage-gated K+ channels, such as Shaker, Kv1.2 and Kv3.1, as well as the voltage-sensing phosphatase Ci-VSP, the relationship between stabilization of the activated voltage sensor by the open pore and voltage sensor relaxation in the control of deactivation has only recently begun to be explored. In this review, we discuss present knowledge and questions raised related to the voltage sensor relaxation mechanism in hERG channels and compare structure-function aspects of relaxation with those observed in related ion channels. We focus discussion, in particular, on the mechanism of coupling between voltage sensor relaxation and deactivation gating to highlight the insight that these studies provide into the control of hERG channel deactivation gating during their physiological functioning.
Collapse
Affiliation(s)
- Yu Patrick Shi
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Samrat Thouta
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Thomas W Claydon
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
29
|
Whicher JR, MacKinnon R. Regulation of Eag1 gating by its intracellular domains. eLife 2019; 8:49188. [PMID: 31490124 PMCID: PMC6731095 DOI: 10.7554/elife.49188] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/14/2019] [Indexed: 01/01/2023] Open
Abstract
Voltage-gated potassium channels (Kvs) are gated by transmembrane voltage sensors (VS) that move in response to changes in membrane voltage. Kv10.1 or Eag1 also has three intracellular domains: PAS, C-linker, and CNBHD. We demonstrate that the Eag1 intracellular domains are not required for voltage-dependent gating but likely interact with the VS to modulate gating. We identified specific interactions between the PAS, CNBHD, and VS that modulate voltage-dependent gating and provide evidence that VS movement destabilizes these interactions to promote channel opening. Additionally, mutation of these interactions renders Eag1 insensitive to calmodulin inhibition. The structure of the calmodulin insensitive mutant in a pre-open conformation suggests that channel opening may occur through a rotation of the intracellular domains and calmodulin may prevent this rotation by stabilizing interactions between the VS and intracellular domains. Intracellular domains likely play a similar modulatory role in voltage-dependent gating of the related Kv11-12 channels.
Collapse
Affiliation(s)
- Jonathan R Whicher
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, New York, United States
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, The Rockefeller University, Howard Hughes Medical Institute, New York, United States
| |
Collapse
|
30
|
Cowgill J, Chanda B. The contribution of voltage clamp fluorometry to the understanding of channel and transporter mechanisms. J Gen Physiol 2019; 151:1163-1172. [PMID: 31431491 PMCID: PMC6785729 DOI: 10.1085/jgp.201912372] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cowgill and Chanda discuss the importance of voltage clamp fluorometry to the functional interpretation of ion channel and transporter structures. Key advances in single particle cryo-EM methods in the past decade have ushered in a resolution revolution in modern biology. The structures of many ion channels and transporters that were previously recalcitrant to crystallography have now been solved. Yet, despite having atomistic models of many complexes, some in multiple conformations, it has been challenging to glean mechanistic insight from these structures. To some extent this reflects our inability to unambiguously assign a given structure to a particular physiological state. One approach that may allow us to bridge this gap between structure and function is voltage clamp fluorometry (VCF). Using this technique, dynamic conformational changes can be measured while simultaneously monitoring the functional state of the channel or transporter. Many of the important papers that have used VCF to probe the gating mechanisms of channels and transporters have been published in the Journal of General Physiology. In this review, we provide an overview of the development of VCF and discuss some of the key problems that have been addressed using this approach. We end with a brief discussion of the outlook for this technique in the era of high-resolution structures.
Collapse
Affiliation(s)
- John Cowgill
- Graduate Program in Biophysics, University of Wisconsin, Madison, WI.,Department of Neuroscience, University of Wisconsin, Madison, WI
| | - Baron Chanda
- Department of Neuroscience, University of Wisconsin, Madison, WI .,Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI
| |
Collapse
|
31
|
Brelidze TI. N- and C-terminal interactions in KCNH channels: The spotlight on the intrinsic ligand. J Gen Physiol 2019; 151:400-403. [PMID: 30782602 PMCID: PMC6445575 DOI: 10.1085/jgp.201812313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Brelidze examines recent data revealing the new role of the intrinsic ligand in hERG potassium channel gating.
Collapse
Affiliation(s)
- Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
32
|
Codding SJ, Trudeau MC. The hERG potassium channel intrinsic ligand regulates N- and C-terminal interactions and channel closure. J Gen Physiol 2018; 151:478-488. [PMID: 30425124 PMCID: PMC6445578 DOI: 10.1085/jgp.201812129] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023] Open
Abstract
An intersubunit interaction between the N-terminal PAS domain and C-terminal cyclic nucleotide binding homology domain (CNBHD) regulates slow deactivation in hERG potassium channels. By mutating the intrinsic ligand, Codding and Trudeau disrupt slow deactivation and prevent the PAS-CNBHD interaction. Human ether-à-go-go–related gene (hERG, KCNH2) voltage-activated potassium channels are critical for cardiac excitability. hERG channels have characteristic slow closing (deactivation), which is auto-regulated by a direct interaction between the N-terminal Per-Arnt-Sim (PAS) domain and the C-terminal cyclic nucleotide binding homology domain (CNBHD). hERG channels are not activated by the binding of extrinsic cyclic nucleotide ligands, but rather bind an “intrinsic ligand” that is composed of residues 860–862 within the CNBHD and mimics a cyclic nucleotide. The intrinsic ligand is located at the PAS–CNBHD interface, but its mechanism of action in hERG is not well understood. Here we use whole-cell patch-clamp electrophysiology and FRET spectroscopy to examine how the intrinsic ligand regulates gating. To carry out this work, we coexpress PAS (a PAS domain fused to cyan fluorescent protein) in trans with hERG “core” channels (channels with a deletion of the PAS domain fused to citrine fluorescent protein). The PAS domain in trans with hERG core channels has slow (regulated) deactivation, like that of WT hERG channels, as well as robust FRET, which indicates there is a direct functional and structural interaction of the PAS domain with the channel core. In contrast, PAS in trans with hERG F860A core channels has intermediate deactivation and intermediate FRET, indicating perturbation of the PAS domain interaction with the CNBHD. Furthermore, PAS in trans with hERG L862A core channels, or PAS in trans with hERG F860G,L862G core channels, has fast (nonregulated) deactivation and no measurable FRET, indicating abolition of the PAS and CNBHD interaction. These results indicate that the intrinsic ligand is necessary for the functional and structural interaction between the PAS domain and the CNBHD, which regulates the characteristic slow deactivation gating in hERG channels.
Collapse
Affiliation(s)
- Sara J Codding
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
33
|
Kanner SA, Jain A, Colecraft HM. Development of a High-Throughput Flow Cytometry Assay to Monitor Defective Trafficking and Rescue of Long QT2 Mutant hERG Channels. Front Physiol 2018; 9:397. [PMID: 29725305 PMCID: PMC5917007 DOI: 10.3389/fphys.2018.00397] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/04/2018] [Indexed: 11/24/2022] Open
Abstract
Long QT Syndrome (LQTS) is an acquired or inherited disorder characterized by prolonged QT interval, exertion-triggered arrhythmias, and sudden cardiac death. One of the most prevalent hereditary LQTS subtypes, LQT2, results from loss-of-function mutations in the hERG channel, which conducts IKr, the rapid component of the delayed rectifier K+ current, critical for cardiac repolarization. The majority of LQT2 mutations result in Class 2 deficits characterized by impaired maturation and trafficking of hERG channels. Here, we have developed a high-throughput flow cytometric assay to analyze the surface and total expression of wild-type (WT) and mutant hERG channels with single-cell resolution. To test our method, we focused on 16 LQT2 mutations in the hERG Per-Arnt-Sim (PAS) domain that were previously studied via a widely used biochemical approach that compares levels of 135-kDa immature and 155-kDa fully glycosylated hERG protein to infer surface expression. We confirmed that LQT2 mutants expressed in HEK293 cells displayed a decreased surface density compared to WT hERG, and were differentially rescued by low temperature. However, we also uncovered some notable differences from the findings obtained via the biochemical approach. In particular, three mutations (N33T, R56Q, and A57P) with apparent WT-like hERG glycosylation patterns displayed up to 50% decreased surface expression. Furthermore, despite WT-like levels of complex glycosylation, these mutants have impaired forward trafficking, and exhibit varying half-lives at the cell surface. The results highlight utility of the surface labeling/flow cytometry approach to quantitatively assess trafficking deficiencies associated with LQT2 mutations, to discern underlying mechanisms, and to report on interventions that rescue deficits in hERG surface expression.
Collapse
Affiliation(s)
- Scott A Kanner
- Doctoral Program in Neurobiology and Behavior, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Ananya Jain
- Department of Physiology and Cellular Biophysics, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Henry M Colecraft
- Doctoral Program in Neurobiology and Behavior, Columbia University College of Physicians and Surgeons, New York, NY, United States.,Department of Physiology and Cellular Biophysics, Columbia University College of Physicians and Surgeons, New York, NY, United States.,Department of Pharmacology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
34
|
Dai G, James ZM, Zagotta WN. Dynamic rearrangement of the intrinsic ligand regulates KCNH potassium channels. J Gen Physiol 2018; 150:625-635. [PMID: 29567795 PMCID: PMC5881448 DOI: 10.1085/jgp.201711989] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
KCNH potassium channels possess an intrinsic ligand in their cyclic nucleotide-binding homology domain, located at the N- and C-terminal domain interface. Dai et al. show that this intrinsic ligand regulates voltage-dependent potentiation via a rearrangement between the ligand and its binding site. KCNH voltage-gated potassium channels (EAG, ERG, and ELK) play significant roles in neuronal and cardiac excitability. They contain cyclic nucleotide-binding homology domains (CNBHDs) but are not directly regulated by cyclic nucleotides. Instead, the CNBHD ligand-binding cavity is occupied by an intrinsic ligand, which resides at the intersubunit interface between the N-terminal eag domain and the C-terminal CNBHD. We show that, in Danio rerio ELK channels, this intrinsic ligand is critical for voltage-dependent potentiation (VDP), a process in which channel opening is stabilized by prior depolarization. We demonstrate that an exogenous peptide corresponding to the intrinsic ligand can bind to and regulate zebrafish ELK channels. This exogenous intrinsic ligand inhibits the channels before VDP and potentiates the channels after VDP. Furthermore, using transition metal ion fluorescence resonance energy transfer and a fluorescent noncanonical amino acid L-Anap, we show that there is a rearrangement of the intrinsic ligand relative to the CNBHD during VDP. We propose that the intrinsic ligand switches from antagonist to agonist as a result of a rearrangement of the eag domain–CNBHD interaction during VDP.
Collapse
Affiliation(s)
- Gucan Dai
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - Zachary M James
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| |
Collapse
|
35
|
Jones DK, Johnson AC, Roti Roti EC, Liu F, Uelmen R, Ayers RA, Baczko I, Tester DJ, Ackerman MJ, Trudeau MC, Robertson GA. Localization and functional consequences of a direct interaction between TRIOBP-1 and hERG proteins in the heart. J Cell Sci 2018; 131:jcs.206730. [PMID: 29507111 DOI: 10.1242/jcs.206730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
Reduced levels of the cardiac human (h)ERG ion channel protein and the corresponding repolarizing current IKr can cause arrhythmia and sudden cardiac death, but the underlying cellular mechanisms controlling hERG surface expression are not well understood. Here, we identified TRIOBP-1, an F-actin-binding protein previously associated with actin polymerization, as a putative hERG-interacting protein in a yeast-two hybrid screen of a cardiac library. We corroborated this interaction by performing Förster resonance energy transfer (FRET) in HEK293 cells and co-immunoprecipitation in HEK293 cells and native cardiac tissue. TRIOBP-1 overexpression reduced hERG surface expression and current density, whereas reducing TRIOBP-1 expression via shRNA knockdown resulted in increased hERG protein levels. Immunolabeling in rat cardiomyocytes showed that native TRIOBP-1 colocalized predominantly with myosin-binding protein C and secondarily with rat ERG. In human stem cell-derived cardiomyocytes, TRIOBP-1 overexpression caused intracellular co-sequestration of hERG signal, reduced native IKr and disrupted action potential repolarization. Ca2+ currents were also somewhat reduced and cell capacitance was increased. These findings establish that TRIOBP-1 interacts directly with hERG and can affect protein levels, IKr magnitude and cardiac membrane excitability.
Collapse
Affiliation(s)
- David K Jones
- Department of Neuroscience, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison SMPH, 1111 Highland Ave. #5505, Madison, WI 53705, USA
| | - Ashley C Johnson
- Department of Physiology, University of Maryland School of Medicine, 660 W. Redwood St., Baltimore, MD 21201, USA
| | - Elon C Roti Roti
- Department of Neuroscience, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison SMPH, 1111 Highland Ave. #5505, Madison, WI 53705, USA
| | - Fang Liu
- Department of Neuroscience, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison SMPH, 1111 Highland Ave. #5505, Madison, WI 53705, USA
| | - Rebecca Uelmen
- Department of Neuroscience, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison SMPH, 1111 Highland Ave. #5505, Madison, WI 53705, USA
| | - Rebecca A Ayers
- Department of Neuroscience, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison SMPH, 1111 Highland Ave. #5505, Madison, WI 53705, USA
| | - Istvan Baczko
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged 6720, Hungary
| | - David J Tester
- Department of Cardiovascular Diseases, Division of Heart Rhythm Service, Mayo Clinic, Rochester, NY 55905, USA
| | - Michael J Ackerman
- Department of Cardiovascular Diseases, Division of Heart Rhythm Service, Mayo Clinic, Rochester, NY 55905, USA
| | - Matthew C Trudeau
- Department of Physiology, University of Maryland School of Medicine, 660 W. Redwood St., Baltimore, MD 21201, USA
| | - Gail A Robertson
- Department of Neuroscience, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison SMPH, 1111 Highland Ave. #5505, Madison, WI 53705, USA
| |
Collapse
|
36
|
James ZM, Zagotta WN. Structural insights into the mechanisms of CNBD channel function. J Gen Physiol 2017; 150:225-244. [PMID: 29233886 PMCID: PMC5806680 DOI: 10.1085/jgp.201711898] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/28/2017] [Indexed: 12/28/2022] Open
Abstract
James and Zagotta discuss how recent cryoEM structures inform our understanding of cyclic nucleotide–binding domain channels. Cyclic nucleotide-binding domain (CNBD) channels are a family of ion channels in the voltage-gated K+ channel superfamily that play crucial roles in many physiological processes. CNBD channels are structurally similar but functionally very diverse. This family includes three subfamilies: (1) the cyclic nucleotide-gated (CNG) channels, which are cation-nonselective, voltage-independent, and cyclic nucleotide-gated; (2) the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which are weakly K+ selective, hyperpolarization-activated, and cyclic nucleotide-gated; and (3) the ether-à-go-go-type (KCNH) channels, which are strongly K+ selective, depolarization-activated, and cyclic nucleotide-independent. Recently, several high-resolution structures have been reported for intact CNBD channels, providing a structural framework to better understand their diverse function. In this review, we compare and contrast the recent structures and discuss how they inform our understanding of ion selectivity, voltage-dependent gating, and cyclic nucleotide–dependent gating within this channel family.
Collapse
Affiliation(s)
- Zachary M James
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| |
Collapse
|
37
|
McNally BA, Pendon ZD, Trudeau MC. hERG1a and hERG1b potassium channel subunits directly interact and preferentially form heteromeric channels. J Biol Chem 2017; 292:21548-21557. [PMID: 29089383 DOI: 10.1074/jbc.m117.816488] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/24/2017] [Indexed: 01/20/2023] Open
Abstract
Voltage-activated human ether-á-go-go-related gene (hERG) potassium channels are critical for the repolarization of cardiac action potentials and tune-spike frequency adaptation in neurons. Two isoforms of mammalian ERG1 channel subunits, ERG1a and ERG1b, are the principal subunits that conduct the IKr current in the heart and are also broadly expressed in the nervous system. However, there is little direct evidence that ERG1a and ERG1b form heteromeric channels. Here, using electrophysiology, biochemistry, and fluorescence approaches, we systematically tested for direct interactions between hERG1a and hERG1b subunits. We report 1) that hERG1a dominant-negative subunits suppress hERG1b currents (and vice versa), 2) that disulfide bonds form between single cysteine residues experimentally introduced into an extracellular loop of hERG1a and hERG1b subunits and produce hERG1a-hERG1b dimers, and 3) that hERG1a and hERG1b subunits tagged with fluorescent proteins that are FRET pairs exhibit robust energy transfer at the plasma membrane. Thus, multiple lines of evidence indicated a physical interaction between hERG1a and hERG1b, consistent with them forming heteromeric channels. Moreover, co-expression of variable ratios of hERG1a and hERG1b RNA yielded channels with deactivation kinetics that reached a plateau and were different from those of hERG1b channels, consistent with a preference of hERG1b subunits for hERG1a subunits. Cross-linking studies revealed that an equal input of hERG1a and hERG1b yields more hERG1a-hERG1a or hERG1a-hERG1b dimers than hERG1b-hERG1b dimers, also suggesting that hERG1b preferentially interacts with hERG1a. We conclude that hERG1b preferentially forms heteromeric ion channels with hERG1a at the plasma membrane.
Collapse
Affiliation(s)
- Beth A McNally
- From the Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Zeus D Pendon
- From the Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Matthew C Trudeau
- From the Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
38
|
Dai G, Zagotta WN. Molecular mechanism of voltage-dependent potentiation of KCNH potassium channels. eLife 2017; 6. [PMID: 28443815 PMCID: PMC5440166 DOI: 10.7554/elife.26355] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022] Open
Abstract
EAG-like (ELK) voltage-gated potassium channels are abundantly expressed in the brain. These channels exhibit a behavior called voltage-dependent potentiation (VDP), which appears to be a specialization to dampen the hyperexitability of neurons. VDP manifests as a potentiation of current amplitude, hyperpolarizing shift in voltage sensitivity, and slowing of deactivation in response to a depolarizing prepulse. Here we show that VDP of D. rerio ELK channels involves the structural interaction between the intracellular N-terminal eag domain and C-terminal CNBHD. Combining transition metal ion FRET, patch-clamp fluorometry, and incorporation of a fluorescent noncanonical amino acid, we show that there is a rearrangement in the eag domain-CNBHD interaction with the kinetics, voltage-dependence, and ATP-dependence of VDP. We propose that the activation of ELK channels involves a slow open-state dependent rearrangement of the direct interaction between the eag domain and CNBHD, which stabilizes the opening of the channel. DOI:http://dx.doi.org/10.7554/eLife.26355.001 In humans and other animals, electrical signals trigger the heart to beat and carry information around the brain and nervous system. Particular cells can generate these signals by regulating the flow of ions into and out of the cell via proteins called ion channels. These proteins sit in the membrane that surrounds the cell and will open or close in response to specific signals. For example, an ion channel in humans called hERG allows positively-charged potassium ions to flow out of a heart cell to help the cell return to its “resting” state after producing an electrical signal. Defects in hERG can alter the rhythm at which the heart beats, leading to a serious condition called Long QT syndrome. The human hERG channel is part of a family of related channels known as the KCNH channels. These channels are made of four protein subunits that assemble to form a pore that spans the cell membrane. When a cell is resting before producing an electrical signal, KCNH channels are generally closed. However, once an electrical signal starts, the flow of ions through other ion channels in the cell membrane changes an electrical property across the membrane known as the “voltage”. This change in voltage causes KCNH channels to open. Dai and Zagotta studied how a KCNH channel known as ELK from zebrafish responds to changes in membrane voltage. The experiments show that the manner in which ELK channels respond to the voltage is due to changes in how the subunits interact in the part of the channel that lies inside the cell. Further experiments using several new techniques reveal in much more detail how the shape of the channel alters as the voltage changes. These new techniques could also be used to observe how other KCNH channels in the heart and brain change shape in response to changes in voltage. This could lead to the design of new drugs to treat heart and neurological diseases. DOI:http://dx.doi.org/10.7554/eLife.26355.002
Collapse
Affiliation(s)
- Gucan Dai
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| |
Collapse
|
39
|
Phan K, Ng CA, David E, Shishmarev D, Kuchel PW, Vandenberg JI, Perry MD. The S1 helix critically regulates the finely tuned gating of Kv11.1 channels. J Biol Chem 2017; 292:7688-7705. [PMID: 28280240 DOI: 10.1074/jbc.m117.779298] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/26/2017] [Indexed: 11/06/2022] Open
Abstract
Congenital mutations in the cardiac Kv11.1 channel can cause long QT syndrome type 2 (LQTS2), a heart rhythm disorder associated with sudden cardiac death. Mutations act either by reducing protein expression at the membrane and/or by perturbing the intricate gating properties of Kv11.1 channels. A number of clinical LQTS2-associated mutations have been reported in the first transmembrane segment (S1) of Kv11.1 channels, but the role of this region of the channel is largely unexplored. In part, this is due to problems defining the extent of the S1 helix, as a consequence of its low sequence homology with other Kv family members. Here, we used NMR spectroscopy and electrophysiological characterization to show that the S1 of Kv11.1 channels extends seven helical turns, from Pro-405 to Phe-431, and is flanked by unstructured loops. Functional analysis suggests that pre-S1 loop residues His-402 and Tyr-403 play an important role in regulating the kinetics and voltage dependence of channel activation and deactivation. Multiple residues within the S1 helix also play an important role in fine-tuning the voltage dependence of activation, regulating slow deactivation, and modulating C-type inactivation of Kv11.1 channels. Analyses of LQTS2-associated mutations in the pre-S1 loop or S1 helix of Kv11.1 channels demonstrate perturbations to both protein expression and most gating transitions. Thus, S1 region mutations would reduce both the action potential repolarizing current passed by Kv11.1 channels in cardiac myocytes, as well as the current passed in response to premature depolarizations that normally helps protect against the formation of ectopic beats.
Collapse
Affiliation(s)
- Kevin Phan
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010.,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| | - Chai Ann Ng
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010.,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| | - Erikka David
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010
| | - Dmitry Shishmarev
- the School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Philip W Kuchel
- the School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jamie I Vandenberg
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010.,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| | - Matthew D Perry
- From the Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, New South Wales 2010, .,the St. Vincent's Clinical School, University of New South Wales, New South Wales 2052, and
| |
Collapse
|
40
|
Zhao Y, Goldschen-Ohm MP, Morais-Cabral JH, Chanda B, Robertson GA. The intrinsically liganded cyclic nucleotide-binding homology domain promotes KCNH channel activation. J Gen Physiol 2017; 149:249-260. [PMID: 28122815 PMCID: PMC5299623 DOI: 10.1085/jgp.201611701] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/29/2016] [Accepted: 12/21/2016] [Indexed: 12/17/2022] Open
Abstract
hEAG1 is a member of the KCNH family of ion channels, which are characterized by C-terminal regions with homology to cyclic nucleotide–binding domains (CNBhDs). Zhao et al. show that an “intrinsic ligand” occupying the CNBhD binding pocket promotes the activated and open state of the channel. Channels in the ether-à-go-go or KCNH family of potassium channels are characterized by a conserved, C-terminal domain with homology to cyclic nucleotide–binding homology domains (CNBhDs). Instead of cyclic nucleotides, two amino acid residues, Y699 and L701, occupy the binding pocket, forming an “intrinsic ligand.” The role of the CNBhD in KCNH channel gating is still unclear, however, and a detailed characterization of the intrinsic ligand is lacking. In this study, we show that mutating both Y699 and L701 to alanine, serine, aspartate, or glycine impairs human EAG1 channel function. These mutants slow channel activation and shift the conductance–voltage (G–V) relation to more depolarized potentials. The mutations affect activation and the G-V relation progressively, indicating that the gating machinery is sensitive to multiple conformations of the CNBhD. Substitution with glycine at both sites (GG), which eliminates the side chains that interact with the binding pocket, also reduces the ability of voltage prepulses to populate more preactivated states along the activation pathway (i.e., the Cole–Moore effect), as if stabilizing the voltage sensor in deep resting states. Notably, deletion of the entire CNBhD (577–708, ΔCNBhD) phenocopies the GG mutant, suggesting that GG is a loss-of-function mutation and the CNBhD requires an intrinsic ligand to exert its functional effects. We developed a kinetic model for both wild-type and ΔCNBhD mutant channels that describes all our observations on activation kinetics, the Cole–Moore shift, and G-V relations. These findings support a model in which the CNBhD both promotes voltage sensor activation and stabilizes the open pore. The intrinsic ligand is critical for these functional effects.
Collapse
Affiliation(s)
- Yaxian Zhao
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705.,Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Marcel P Goldschen-Ohm
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705.,Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - João H Morais-Cabral
- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4150 Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4150 Porto, Portugal
| | - Baron Chanda
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705.,Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Gail A Robertson
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 .,Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| |
Collapse
|
41
|
Wu W, Gardner A, Sachse FB, Sanguinetti MC. Ginsenoside Rg3, a Gating Modifier of EAG Family K+ Channels. Mol Pharmacol 2016; 90:469-82. [PMID: 27502018 PMCID: PMC5034692 DOI: 10.1124/mol.116.104091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/05/2016] [Indexed: 01/11/2023] Open
Abstract
Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-à-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. However, it is unknown whether this action is unique to hERG1 channels. Here we compare and contrast the mechanisms of actions of Rg3 on hERG1 with three other members of the ether-à-go-go (EAG) K(+) channel gene family, including EAG1 (Kv10.1), ERG3 (Kv11.3), and ELK1 (Kv12.1). All four channel types were heterologously expressed in Xenopus laevis oocytes, and K(+) currents were measured using the two-microelectrode voltage-clamp technique. At a maximally effective concentration, Rg3 shifted the half-point of voltage-dependent activation of currents by -14 mV for ERG1 (EC50 = 414 nM), -20 mV for ERG3 (EC50 = 374 nM), -28 mV for EAG1 (EC50 = 1.18 μM), and more than -100 mV for ELK1 (EC50 = 197 nM) channels. Rg3 also induced slowing of ERG1, ERG3, and ELK1 channel deactivation and accelerated the rate of EAG1 channel activation. A Markov model was developed to simulate gating and the effects of Rg3 on the voltage dependence of activation of hELK1 channels. Understanding the mechanism underlying the action of Rg3 may facilitate the development of more potent and selective EAG family channel activators as therapies for cardiovascular and neural disorders.
Collapse
Affiliation(s)
- Wei Wu
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| | - Alison Gardner
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| | - Michael C Sanguinetti
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| |
Collapse
|
42
|
Toombes GES, Swartz KJ. STRUCTURAL BIOLOGY. Twists and turns in gating ion channels with voltage. Science 2016; 353:646-7. [PMID: 27516583 DOI: 10.1126/science.aah4194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Gilman E S Toombes
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
43
|
Li X, Anishkin A, Liu H, van Rossum DB, Chintapalli SV, Sassic JK, Gallegos D, Pivaroff-Ward K, Jegla T. Bimodal regulation of an Elk subfamily K+ channel by phosphatidylinositol 4,5-bisphosphate. ACTA ACUST UNITED AC 2016; 146:357-74. [PMID: 26503718 PMCID: PMC4621751 DOI: 10.1085/jgp.201511491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PIP2 mediates the bimodal regulation of the EAG family K+ channel ELK1 to produce an overall inhibitory effect. Phosphatidylinositol 4,5-bisphosphate (PIP2) regulates Shaker K+ channels and voltage-gated Ca2+ channels in a bimodal fashion by inhibiting voltage activation while stabilizing open channels. Bimodal regulation is conserved in hyperpolarization-activated cyclic nucleotide–gated (HCN) channels, but voltage activation is enhanced while the open channel state is destabilized. The proposed sites of PIP2 regulation in these channels include the voltage-sensor domain (VSD) and conserved regions of the proximal cytoplasmic C terminus. Relatively little is known about PIP2 regulation of Ether-á-go-go (EAG) channels, a metazoan-specific family of K+ channels that includes three gene subfamilies, Eag (Kv10), Erg (Kv11), and Elk (Kv12). We examined PIP2 regulation of the Elk subfamily potassium channel human Elk1 to determine whether bimodal regulation is conserved within the EAG K+ channel family. Open-state stabilization by PIP2 has been observed in human Erg1, but the proposed site of regulation in the distal C terminus is not conserved among EAG family channels. We show that PIP2 strongly inhibits voltage activation of Elk1 but also stabilizes the open state. This stabilization produces slow deactivation and a mode shift in voltage gating after activation. However, removal of PIP2 has the net effect of enhancing Elk1 activation. R347 in the linker between the VSD and pore (S4–S5 linker) and R479 near the S6 activation gate are required for PIP2 to inhibit voltage activation. The ability of PIP2 to stabilize the open state also requires these residues, suggesting an overlap in sites central to the opposing effects of PIP2 on channel gating. Open-state stabilization in Elk1 requires the N-terminal eag domain (PAS domain + Cap), and PIP2-dependent stabilization is enhanced by a conserved basic residue (K5) in the Cap. Our data shows that PIP2 can bimodally regulate voltage gating in EAG family channels, as has been proposed for Shaker and HCN channels. PIP2 regulation appears fundamentally different for Elk and KCNQ channels, suggesting that, although both channel types can regulate action potential threshold in neurons, they are not functionally redundant.
Collapse
Affiliation(s)
- Xiaofan Li
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802 Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, MD 20742
| | - Hansi Liu
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Damian B van Rossum
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802 Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202 Arkansas Children's Nutrition Center and Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202
| | - Jessica K Sassic
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - David Gallegos
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Kendra Pivaroff-Ward
- Department of Earth and Space Sciences, University of Washington, Seattle, WA 98195
| | - Timothy Jegla
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802 Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
44
|
Lörinczi E, Helliwell M, Finch A, Stansfeld PJ, Davies NW, Mahaut-Smith M, Muskett FW, Mitcheson JS. Calmodulin Regulates Human Ether à Go-Go 1 (hEAG1) Potassium Channels through Interactions of the Eag Domain with the Cyclic Nucleotide Binding Homology Domain. J Biol Chem 2016; 291:17907-18. [PMID: 27325704 PMCID: PMC5016179 DOI: 10.1074/jbc.m116.733576] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Indexed: 11/24/2022] Open
Abstract
The ether à go-go family of voltage-gated potassium channels is structurally distinct. The N terminus contains an eag domain (eagD) that contains a Per-Arnt-Sim (PAS) domain that is preceded by a conserved sequence of 25–27 amino acids known as the PAS-cap. The C terminus contains a region with homology to cyclic nucleotide binding domains (cNBHD), which is directly linked to the channel pore. The human EAG1 (hEAG1) channel is remarkably sensitive to inhibition by intracellular calcium (Ca2+i) through binding of Ca2+-calmodulin to three sites adjacent to the eagD and cNBHD. Here, we show that the eagD and cNBHD interact to modulate Ca2+-calmodulin as well as voltage-dependent gating. Sustained elevation of Ca2+i resulted in an initial profound inhibition of hEAG1 currents, which was followed by a phase when current amplitudes partially recovered, but activation gating was slowed and shifted to depolarized potentials. Deletion of either the eagD or cNBHD abolished the inhibition by Ca2+i. However, deletion of just the PAS-cap resulted in a >15-fold potentiation in response to elevated Ca2+i. Mutations of residues at the interface between the eagD and cNBHD have been linked to human cancer. Glu-600 on the cNBHD, when substituted with residues with a larger volume, resulted in hEAG1 currents that were profoundly potentiated by Ca2+i in a manner similar to the ΔPAS-cap mutant. These findings provide the first evidence that eagD and cNBHD interactions are regulating Ca2+-dependent gating and indicate that the binding of the PAS-cap with the cNBHD is required for the closure of the channels upon CaM binding.
Collapse
Affiliation(s)
- Eva Lörinczi
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN
| | - Matthew Helliwell
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN, the School of Physiology and Pharmacology, University of Bristol, Bristol BS5 1TD, and
| | - Alina Finch
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN
| | - Phillip J Stansfeld
- the Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Noel W Davies
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN
| | - Martyn Mahaut-Smith
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN
| | - Frederick W Muskett
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN
| | - John S Mitcheson
- From the Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN,
| |
Collapse
|
45
|
Perry MD, Ng CA, Phan K, David E, Steer K, Hunter MJ, Mann SA, Imtiaz M, Hill AP, Ke Y, Vandenberg JI. Rescue of protein expression defects may not be enough to abolish the pro-arrhythmic phenotype of long QT type 2 mutations. J Physiol 2016; 594:4031-49. [PMID: 26958806 DOI: 10.1113/jp271805] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/25/2016] [Indexed: 01/28/2023] Open
Abstract
KEY POINTS Most missense long QT syndrome type 2 (LQTS2) mutations result in Kv11.1 channels that show reduced levels of membrane expression. Pharmacological chaperones that rescue mutant channel expression could have therapeutic potential to reduce the risk of LQTS2-associated arrhythmias and sudden cardiac death, but only if the mutant Kv11.1 channels function normally (i.e. like WT channels) after membrane expression is restored. Fewer than half of mutant channels exhibit relatively normal function after rescue by low temperature. The remaining rescued missense mutant Kv11.1 channels have perturbed gating and/or ion selectivity characteristics. Co-expression of WT subunits with gating defective missense mutations ameliorates but does not eliminate the functional abnormalities observed for most mutant channels. For patients with mutations that affect gating in addition to expression, it may be necessary to use a combination therapy to restore both normal function and normal expression of the channel protein. ABSTRACT In the heart, Kv11.1 channels pass the rapid delayed rectifier current (IKr ) which plays critical roles in repolarization of the cardiac action potential and in the suppression of arrhythmias caused by premature stimuli. Over 500 inherited mutations in Kv11.1 are known to cause long QT syndrome type 2 (LQTS2), a cardiac electrical disorder associated with an increased risk of life threatening arrhythmias. Most missense mutations in Kv11.1 reduce the amount of channel protein expressed at the membrane and, as a consequence, there has been considerable interest in developing pharmacological agents to rescue the expression of these channels. However, pharmacological chaperones will only have clinical utility if the mutant Kv11.1 channels function normally after membrane expression is restored. The aim of this study was to characterize the gating phenotype for a subset of LQTS2 mutations to assess what proportion of mutations may be suitable for rescue. As an initial screen we used reduced temperature to rescue expression defects of mutant channels expressed in Xenopus laevis oocytes. Over half (∼56%) of Kv11.1 mutants exhibited functional gating defects that either dramatically reduced the amount of current contributing to cardiac action potential repolarization and/or reduced the amount of protective current elicited in response to premature depolarizations. Our data demonstrate that if pharmacological rescue of protein expression defects is going to have clinical utility in the treatment of LQTS2 then it will be important to assess the gating phenotype of LQTS2 mutations before attempting rescue.
Collapse
Affiliation(s)
- Matthew D Perry
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, 2052, Australia
| | - Chai Ann Ng
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, 2052, Australia
| | - Kevin Phan
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, 2052, Australia
| | - Erikka David
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia
| | - Kieran Steer
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,Faculty of Science, McGill University, Montreal, Quebec, Canada
| | - Mark J Hunter
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia
| | - Stefan A Mann
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, 2052, Australia
| | - Mohammad Imtiaz
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, 2052, Australia
| | - Ying Ke
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, Molecular Cardiology and Biophysics Division, Darlinghurst, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, NSW, 2052, Australia
| |
Collapse
|
46
|
Goodchild SJ, Macdonald LC, Fedida D. Sequence of gating charge movement and pore gating in HERG activation and deactivation pathways. Biophys J 2016; 108:1435-1447. [PMID: 25809256 DOI: 10.1016/j.bpj.2015.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/08/2015] [Accepted: 02/18/2015] [Indexed: 12/26/2022] Open
Abstract
KV11.1 voltage-gated K(+) channels are noted for unusually slow activation, fast inactivation, and slow deactivation kinetics, which tune channel activity to provide vital repolarizing current during later stages of the cardiac action potential. The bulk of charge movement in human ether-a-go-go-related gene (hERG) is slow, as is return of charge upon repolarization, suggesting that the rates of hERG channel opening and, critically, that of deactivation might be determined by slow voltage sensor movement, and also by a mode-shift after activation. To test these ideas, we compared the kinetics and voltage dependence of ionic activation and deactivation with gating charge movement. At 0 mV, gating charge moved ∼threefold faster than ionic current, which suggests the presence of additional slow transitions downstream of charge movement in the physiological activation pathway. A significant voltage sensor mode-shift was apparent by 24 ms at +60 mV in gating currents, and return of charge closely tracked pore closure after pulses of 100 and 300 ms duration. A deletion of the N-terminus PAS domain, mutation R4AR5A or the LQT2-causing mutation R56Q gave faster-deactivating channels that displayed an attenuated mode-shift of charge. This indicates that charge movement is perturbed by N- and C-terminus interactions, and that these domain interactions stabilize the open state and limit the rate of charge return. We conclude that slow on-gating charge movement can only partly account for slow hERG ionic activation, and that the rate of pore closure has a limiting role in the slow return of gating charges.
Collapse
Affiliation(s)
- Samuel J Goodchild
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Logan C Macdonald
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Fedida
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
47
|
Wu W, Sanguinetti MC. Molecular Basis of Cardiac Delayed Rectifier Potassium Channel Function and Pharmacology. Card Electrophysiol Clin 2016; 8:275-84. [PMID: 27261821 DOI: 10.1016/j.ccep.2016.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human cardiomyocytes express 3 distinct types of delayed rectifier potassium channels. Human ether-a-go-go-related gene (hERG) channels conduct the rapidly activating current IKr; KCNQ1/KCNE1 channels conduct the slowly activating current IKs; and Kv1.5 channels conduct an ultrarapid activating current IKur. Here the authors provide a general overview of the mechanistic and structural basis of ion selectivity, gating, and pharmacology of the 3 types of cardiac delayed rectifier potassium ion channels. Most blockers bind to S6 residues that line the central cavity of the channel, whereas activators interact with the channel at 4 symmetric binding sites outside the cavity.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA
| | - Michael C Sanguinetti
- Department of Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
48
|
Calcaterra NE, Hoeppner DJ, Wei H, Jaffe AE, Maher BJ, Barrow JC. Schizophrenia-Associated hERG channel Kv11.1-3.1 Exhibits a Unique Trafficking Deficit that is Rescued Through Proteasome Inhibition for High Throughput Screening. Sci Rep 2016; 6:19976. [PMID: 26879421 PMCID: PMC4754628 DOI: 10.1038/srep19976] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/22/2015] [Indexed: 12/24/2022] Open
Abstract
The primate-specific brain voltage-gated potassium channel isoform Kv11.1-3.1 has been identified as a novel therapeutic target for the treatment of schizophrenia. While this ether-a-go-go related K + channel has shown clinical relevance, drug discovery efforts have been hampered due to low and inconsistent activity in cell-based assays. This poor activity is hypothesized to result from poor trafficking via the lack of an intact channel-stabilizing Per-Ant-Sim (PAS) domain. Here we characterize Kv11.1-3.1 cellular localization and show decreased channel expression and cell surface trafficking relative to the PAS-domain containing major isoform, Kv11.1-1A. Using small molecule inhibition of proteasome degradation, cellular expression and plasma membrane trafficking are rescued. These findings implicate the importance of the unfolded-protein response and endoplasmic reticulum associated degradation pathways in the expression and regulation of this schizophrenia risk factor. Utilizing this identified phenomenon, an electrophysiological and high throughput in-vitro fluorescent assay platform has been developed for drug discovery in order to explore a potentially new class of cognitive therapeutics.
Collapse
Affiliation(s)
| | | | - Huijun Wei
- Lieber Institute for Brain Development, Baltimore, MD 21205
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Baltimore, MD 21205.,Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| | - Brady J Maher
- Lieber Institute for Brain Development, Baltimore, MD 21205.,Departments of Psychiatry and Behavioral Sciences, Baltimore, MD 21205.,Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - James C Barrow
- Departments of Pharmacology and Molecular Sciences, Baltimore, MD 21205.,Lieber Institute for Brain Development, Baltimore, MD 21205
| |
Collapse
|
49
|
Jones DK, Liu F, Dombrowski N, Joshi S, Robertson GA. Dominant negative consequences of a hERG 1b-specific mutation associated with intrauterine fetal death. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:67-76. [PMID: 26772437 DOI: 10.1016/j.pbiomolbio.2016.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 12/16/2022]
Abstract
The human ether-a-go-go related gene (hERG) encodes two subunits, hERG 1a and hERG 1b, that combine in vivo to conduct the rapid delayed rectifier potassium current (IKr). Reduced IKr slows cardiac action potential (AP) repolarization and is an underlying cause of cardiac arrhythmias associated with long QT syndrome (LQTS). Although the physiological importance of hERG 1b has been elucidated, the effects of hERG 1b disease mutations on cardiac IKr and AP behavior have not been described. To explore the disease mechanism of a 1b-specific mutation associated with a case of intrauterine fetal death, we examined the effects of the 1b-R25W mutation on total protein, trafficking and membrane current levels in HEK293 cells at physiological temperatures. By all measures the 1b-R25W mutation conferred diminished expression, and exerted a temperature-sensitive, dominant-negative effect over the WT hERG 1a protein with which it was co-expressed. Membrane currents were reduced by 60% with no apparent effect on voltage dependence or deactivation kinetics. The dominant-negative effects of R25W were demonstrated in iPSC-CMs, where 1b-R25W transfection diminished native IKr compared to controls. R25W also slowed AP repolarization, and increased AP triangulation and variability in iPSC-CMs, reflecting cellular manifestations of pro-arrhythmia. These data demonstrate that R25W is a dominant-negative mutation with significant pathophysiological consequences, and provide the first direct link between hERG 1b mutation and cardiomyocyte dysfunction.
Collapse
Affiliation(s)
- David K Jones
- Dept. of Neuroscience, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison WI 53705, USA
| | - Fang Liu
- Dept. of Neuroscience, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison WI 53705, USA
| | - Natasha Dombrowski
- Dept. of Neuroscience, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison WI 53705, USA
| | - Sunita Joshi
- Dept. of Neuroscience, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison WI 53705, USA
| | - Gail A Robertson
- Dept. of Neuroscience, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Madison WI 53705, USA.
| |
Collapse
|
50
|
Li X, Martinson AS, Layden MJ, Diatta FH, Sberna AP, Simmons DK, Martindale MQ, Jegla TJ. Ether-à-go-go family voltage-gated K+ channels evolved in an ancestral metazoan and functionally diversified in a cnidarian-bilaterian ancestor. ACTA ACUST UNITED AC 2015; 218:526-36. [PMID: 25696816 DOI: 10.1242/jeb.110080] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We examined the evolutionary origins of the ether-à-go-go (EAG) family of voltage-gated K(+) channels, which have a strong influence on the excitability of neurons. The bilaterian EAG family comprises three gene subfamilies (Eag, Erg and Elk) distinguished by sequence conservation and functional properties. Searches of genome sequence indicate that EAG channels are metazoan specific, appearing first in ctenophores. However, phylogenetic analysis including two EAG family channels from the ctenophore Mnemiopsis leidyi indicates that the diversification of the Eag, Erg and Elk gene subfamilies occurred in a cnidarian/bilaterian ancestor after divergence from ctenophores. Erg channel function is highly conserved between cnidarians and mammals. Here we show that Eag and Elk channels from the sea anemone Nematostella vectensis (NvEag and NvElk) also share high functional conservation with mammalian channels. NvEag, like bilaterian Eag channels, has rapid kinetics, whereas NvElk activates at extremely hyperpolarized voltages, which is characteristic of Elk channels. Potent inhibition of voltage activation by extracellular protons is conserved between mammalian and Nematostella EAG channels. However, characteristic inhibition of voltage activation by Mg(2+) in Eag channels and Ca(2+) in Erg channels is reduced in Nematostella because of mutation of a highly conserved aspartate residue in the voltage sensor. This mutation may preserve sub-threshold activation of Nematostella Eag and Erg channels in a high divalent cation environment. mRNA in situ hybridization of EAG channels in Nematostella suggests that they are differentially expressed in distinct cell types. Most notable is the expression of NvEag in cnidocytes, a cnidarian-specific stinging cell thought to be a neuronal subtype.
Collapse
Affiliation(s)
- Xiaofan Li
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Alexandra S Martinson
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Michael J Layden
- Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL 32136, USA
| | - Fortunay H Diatta
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Anna P Sberna
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - David K Simmons
- Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL 32136, USA
| | - Mark Q Martindale
- Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL 32136, USA
| | - Timothy J Jegla
- Department of Biology and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|