1
|
Jegla T, Simonson BT, Spafford JD. A broad survey of choanoflagellates revises the evolutionary history of the Shaker family of voltage-gated K + channels in animals. Proc Natl Acad Sci U S A 2024; 121:e2407461121. [PMID: 39018191 PMCID: PMC11287247 DOI: 10.1073/pnas.2407461121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/21/2024] [Indexed: 07/19/2024] Open
Abstract
The Shaker family of voltage-gated K+ channels has been thought of as an animal-specific ion channel family that diversified in concert with nervous systems. It comprises four functionally independent gene subfamilies (Kv1-4) that encode diverse neuronal K+ currents. Comparison of animal genomes predicts that only the Kv1 subfamily was present in the animal common ancestor. Here, we show that some choanoflagellates, the closest protozoan sister lineage to animals, also have Shaker family K+ channels. Choanoflagellate Shaker family channels are surprisingly most closely related to the animal Kv2-4 subfamilies which were believed to have evolved only after the divergence of ctenophores and sponges from cnidarians and bilaterians. Structural modeling predicts that the choanoflagellate channels share a T1 Zn2+ binding site with Kv2-4 channels that is absent in Kv1 channels. We functionally expressed three Shakers from Salpingoeca helianthica (SheliKvT1.1-3) in Xenopus oocytes. SheliKvT1.1-3 function only in two heteromultimeric combinations (SheliKvT1.1/1.2 and SheliKvT1.1/1.3) and encode fast N-type inactivating K+ channels with distinct voltage dependence that are most similar to the widespread animal Kv1-encoded A-type Shakers. Structural modeling of the T1 assembly domain supports a preference for heteromeric assembly in a 2:2 stoichiometry. These results push the origin of the Shaker family back into a common ancestor of metazoans and choanoflagellates. They also suggest that the animal common ancestor had at least two distinct molecular lineages of Shaker channels, a Kv1 subfamily lineage predicted from comparison of animal genomes and a Kv2-4 lineage predicted from comparison of animals and choanoflagellates.
Collapse
Affiliation(s)
- Timothy Jegla
- Department of Biology, Eberly College of Sciences and Huck Institutes of the Life Sciences, Penn State University, University Park, PA16802
| | - Benjamin T. Simonson
- Department of Biology, Eberly College of Sciences and Huck Institutes of the Life Sciences, Penn State University, University Park, PA16802
| | - J. David Spafford
- Department of Biology, University of Waterloo, Waterloo, ONN2L 3G1, Canada
| |
Collapse
|
2
|
Burtscher V, Mount J, Huang J, Cowgill J, Chang Y, Bickel K, Chen J, Yuan P, Chanda B. Structural basis for hyperpolarization-dependent opening of human HCN1 channel. Nat Commun 2024; 15:5216. [PMID: 38890331 PMCID: PMC11189445 DOI: 10.1038/s41467-024-49599-x] [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: 08/14/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Hyperpolarization and cyclic nucleotide (HCN) activated ion channels are critical for the automaticity of action potentials in pacemaking and rhythmic electrical circuits in the human body. Unlike most voltage-gated ion channels, the HCN and related plant ion channels activate upon membrane hyperpolarization. Although functional studies have identified residues in the interface between the voltage-sensing and pore domain as crucial for inverted electromechanical coupling, the structural mechanisms for this unusual voltage-dependence remain unclear. Here, we present cryo-electron microscopy structures of human HCN1 corresponding to Closed, Open, and a putative Intermediate state. Our structures reveal that the downward motion of the gating charges past the charge transfer center is accompanied by concomitant unwinding of the inner end of the S4 and S5 helices, disrupting the tight gating interface observed in the Closed state structure. This helix-coil transition at the intracellular gating interface accompanies a concerted iris-like dilation of the pore helices and underlies the reversed voltage dependence of HCN channels.
Collapse
Affiliation(s)
- Verena Burtscher
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jonathan Mount
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - John Cowgill
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yongchang Chang
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kathleen Bickel
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - Peng Yuan
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Baron Chanda
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA.
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
3
|
Ghorbani M, Wang ZJ, Chen X, Tiwari PB, Klauda JB, Brelidze TI. Chlorpromazine inhibits EAG1 channels by altering the coupling between the PAS, CNBH and pore domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581826. [PMID: 38464246 PMCID: PMC10925124 DOI: 10.1101/2024.02.23.581826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
EAG1 depolarization-activated potassium selective channels are important targets for treatment of cancer and neurological disorders. EAG1 channels are formed by a tetrameric subunit assembly with each subunit containing an N-terminal Per-Arnt-Sim (PAS) domain and C-terminal cyclic nucleotide-binding homology (CNBH) domain. The PAS and CNBH domains from adjacent subunits interact and form an intracellular tetrameric ring that regulates the EAG1 channel gating, including the movement of the voltage sensor domain (VSD) from closed to open states. Small molecule ligands can inhibit EAG1 channels by binding to their PAS domains. However, the allosteric pathways of this inhibition are not known. Here we show that chlorpromazine, a PAS domain small molecule binder, alters interactions between the PAS and CNBH domains and decreases the coupling between the intracellular tetrameric ring and the pore of the channel, while having little effect on the coupling between the PAS and VSD domains. In addition, chlorpromazine binding to the PAS domain did not alter Cole-Moore shift characteristic of EAG1 channels, further indicating that chlorpromazine has no effect on VSD movement from the deep closed to opened states. Our study provides a framework for understanding global pathways of EAG1 channel regulation by small molecule PAS domain binders.
Collapse
|
4
|
Tiwari PB, Kamgar-Dayhoff P, Tiwari P, McKillop MI, Brelidze TI. Use of Surface Plasmon Resonance Technique for Studies of Inter-domain Interactions in Ion Channels. Methods Mol Biol 2024; 2796:105-118. [PMID: 38856898 PMCID: PMC11225882 DOI: 10.1007/978-1-0716-3818-7_7] [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: 06/11/2024]
Abstract
Ion channels are transmembrane proteins essential for cellular functions and are important drug targets. Surface plasmon resonance (SPR) is a powerful technique for investigating protein-protein and protein-small molecule ligand interactions. SPR has been underutilized for studies of ion channels, even though it could provide a wealth of information on the mechanisms of ion channel regulation and aid in ion channel drug discovery. Here we provide a detailed description of the use of SPR technology for investigating inter-domain interactions in KCNH potassium-selective and voltage-gated ion channels.
Collapse
Affiliation(s)
- Purushottam B Tiwari
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
| | - Pareesa Kamgar-Dayhoff
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Prakriti Tiwari
- Department of Oncology, Georgetown University Medical Center, Washington, DC, USA
- Department of Biology, College of Arts & Sciences, Georgetown University, Washington, DC, USA
| | - Maria I McKillop
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
5
|
Wang ZJ, Ghorbani M, Chen X, Tiwari PB, Klauda JB, Brelidze TI. Molecular mechanism of EAG1 channel inhibition by imipramine binding to the PAS domain. J Biol Chem 2023; 299:105391. [PMID: 37898402 PMCID: PMC10687071 DOI: 10.1016/j.jbc.2023.105391] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023] Open
Abstract
Ether-a-go-go (EAG) channels are key regulators of neuronal excitability and tumorigenesis. EAG channels contain an N-terminal Per-Arnt-Sim (PAS) domain that can regulate currents from EAG channels by binding small molecules. The molecular mechanism of this regulation is not clear. Using surface plasmon resonance and electrophysiology we show that a small molecule ligand imipramine can bind to the PAS domain of EAG1 channels and inhibit EAG1 currents via this binding. We further used a combination of molecular dynamics (MD) simulations, electrophysiology, and mutagenesis to investigate the molecular mechanism of EAG1 current inhibition by imipramine binding to the PAS domain. We found that Tyr71, located at the entrance to the PAS domain cavity, serves as a "gatekeeper" limiting access of imipramine to the cavity. MD simulations indicate that the hydrophobic electrostatic profile of the cavity facilitates imipramine binding and in silico mutations of hydrophobic cavity-lining residues to negatively charged glutamates decreased imipramine binding. Probing the PAS domain cavity-lining residues with site-directed mutagenesis, guided by MD simulations, identified D39 and R84 as residues essential for the EAG1 channel inhibition by imipramine binding to the PAS domain. Taken together, our study identified specific residues in the PAS domain that could increase or decrease EAG1 current inhibition by imipramine binding to the PAS domain. These findings should further the understanding of molecular mechanisms of EAG1 channel regulation by ligands and facilitate the development of therapeutic agents targeting these channels.
Collapse
Affiliation(s)
- Ze-Jun Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Mahdi Ghorbani
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland, USA
| | - Xi Chen
- 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
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, Maryland, USA; Institute for Physical Science and Technology and Biophysics Graduate Program, University of Maryland, College Park, Maryland, USA.
| | - Tinatin I Brelidze
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA.
| |
Collapse
|
6
|
Burtscher V, Mount J, Cowgill J, Chang Y, Bickel K, Yuan P, Chanda B. Structural Basis for Hyperpolarization-dependent Opening of the Human HCN1 Channel. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553623. [PMID: 37645882 PMCID: PMC10462129 DOI: 10.1101/2023.08.17.553623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Hyperpolarization and cyclic-nucleotide (HCN) activated ion channels play a critical role in generating self-propagating action potentials in pacemaking and rhythmic electrical circuits in the human body. Unlike most voltage-gated ion channels, the HCN channels activate upon membrane hyperpolarization, but the structural mechanisms underlying this gating behavior remain unclear. Here, we present cryo-electron microscopy structures of human HCN1 in Closed, Intermediate, and Open states. Our structures reveal that the inward motion of two gating charges past the charge transfer center (CTC) and concomitant tilting of the S5 helix drives the opening of the central pore. In the intermediate state structure, a single gating charge is positioned below the CTC and the pore appears closed, whereas in the open state structure, both charges move past CTC and the pore is fully open. Remarkably, the downward motion of the voltage sensor is accompanied by progressive unwinding of the inner end of S4 and S5 helices disrupting the tight gating interface that stabilizes the Closed state structure. This "melting" transition at the intracellular gating interface leads to a concerted iris-like displacement of S5 and S6 helices, resulting in pore opening. These findings reveal key structural features that are likely to underlie reversed voltage-dependence of HCN channels.
Collapse
Affiliation(s)
- Verena Burtscher
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Mount
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Cowgill
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yongchang Chang
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kathleen Bickel
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Peng Yuan
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Baron Chanda
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
7
|
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: 2.0] [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
|
8
|
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: 8] [Impact Index Per Article: 2.0] [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
|
9
|
The environmental toxicant ziram enhances neurotransmitter release and increases neuronal excitability via the EAG family of potassium channels. Neurobiol Dis 2020; 143:104977. [PMID: 32553709 DOI: 10.1016/j.nbd.2020.104977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 12/21/2022] Open
Abstract
Environmental toxicants have the potential to contribute to the pathophysiology of multiple complex diseases, but the underlying mechanisms remain obscure. One such toxicant is the widely used fungicide ziram, a dithiocarbamate known to have neurotoxic effects and to increase the risk of Parkinson's disease. We have used Drosophila melanogaster as an unbiased discovery tool to identify novel molecular pathways by which ziram may disrupt neuronal function. Consistent with previous results in mammalian cells, we find that ziram increases the probability of synaptic vesicle release by dysregulation of the ubiquitin signaling system. In addition, we find that ziram increases neuronal excitability. Using a combination of live imaging and electrophysiology, we find that ziram increases excitability in both aminergic and glutamatergic neurons. This increased excitability is phenocopied and occluded by null mutant animals of the ether a-go-go (eag) potassium channel. A pharmacological inhibitor of the temperature sensitive hERG (human ether-a-go-go related gene) phenocopies the excitability effects of ziram but only at elevated temperatures. seizure (sei), a fly ortholog of hERG, is thus another candidate target of ziram. Taken together, the eag family of potassium channels emerges as a candidate for mediating some of the toxic effects of ziram. We propose that ziram may contribute to the risk of complex human diseases by blockade of human eag and sei orthologs, such as hERG.
Collapse
|
10
|
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: 5.0] [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
|
11
|
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: 14] [Impact Index Per Article: 3.5] [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
|
12
|
He S, Moutaoufik MT, Islam S, Persad A, Wu A, Aly KA, Fonge H, Babu M, Cayabyab FS. HERG channel and cancer: A mechanistic review of carcinogenic processes and therapeutic potential. Biochim Biophys Acta Rev Cancer 2020; 1873:188355. [PMID: 32135169 DOI: 10.1016/j.bbcan.2020.188355] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/21/2022]
Abstract
The human ether-à-go-go related gene (HERG) encodes the alpha subunit of Kv11.1, which is a voltage-gated K+ channel protein mainly expressed in heart and brain tissue. HERG plays critical role in cardiac repolarization, and mutations in HERG can cause long QT syndrome. More recently, evidence has emerged that HERG channels are aberrantly expressed in many kinds of cancer cells and play important roles in cancer progression. HERG could therefore be a potential biomarker for cancer and a possible molecular target for anticancer drug design. HERG affects a number of cellular processes, including cell proliferation, apoptosis, angiogenesis and migration, any of which could be affected by dysregulation of HERG. This review provides an overview of available information on HERG channel as it relates to cancer, with focus on the mechanism by which HERG influences cancer progression. Molecular docking attempts suggest two possible protein-protein interactions of HERG with the ß1-integrin receptor and the transcription factor STAT-1 as novel HERG-directed therapeutic targeting which avoids possible cardiotoxicity. The role of epigenetics in regulating HERG channel expression and activity in cancer will also be discussed. Finally, given its inherent extracellular accessibility as an ion channel, we discuss regulatory roles of this molecule in cancer physiology and therapeutic potential. Future research should be directed to explore the possibilities of therapeutic interventions targeting HERG channels while minding possible complications.
Collapse
Affiliation(s)
- Siyi He
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | | | - Saadul Islam
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Amit Persad
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Adam Wu
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Khaled A Aly
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W8, Canada; Department of Medical Imaging, Royal University Hospital, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Francisco S Cayabyab
- Department of Surgery, Neuroscience Research Group, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
13
|
Meléndez TA, Huanosta-Gutiérrez A, Barriga-Montoya C, González-Andrade M, Gómez-Lagunas F. Dronedarone blockage of the tumor-related Kv10.1 channel: a comparison with amiodarone. Pflugers Arch 2020; 472:75-87. [DOI: 10.1007/s00424-019-02342-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022]
|
14
|
Bouagnon AD, Lin L, Srivastava S, Liu CC, Panda O, Schroeder FC, Srinivasan S, Ashrafi K. Intestinal peroxisomal fatty acid β-oxidation regulates neural serotonin signaling through a feedback mechanism. PLoS Biol 2019; 17:e3000242. [PMID: 31805041 PMCID: PMC6917301 DOI: 10.1371/journal.pbio.3000242] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 12/17/2019] [Accepted: 11/15/2019] [Indexed: 02/02/2023] Open
Abstract
The ability to coordinate behavioral responses with metabolic status is fundamental to the maintenance of energy homeostasis. In numerous species including Caenorhabditis elegans and mammals, neural serotonin signaling regulates a range of food-related behaviors. However, the mechanisms that integrate metabolic information with serotonergic circuits are poorly characterized. Here, we identify metabolic, molecular, and cellular components of a circuit that links peripheral metabolic state to serotonin-regulated behaviors in C. elegans. We find that blocking the entry of fatty acyl coenzyme As (CoAs) into peroxisomal β-oxidation in the intestine blunts the effects of neural serotonin signaling on feeding and egg-laying behaviors. Comparative genomics and metabolomics revealed that interfering with intestinal peroxisomal β-oxidation results in a modest global transcriptional change but significant changes to the metabolome, including a large number of changes in ascaroside and phospholipid species, some of which affect feeding behavior. We also identify body cavity neurons and an ether-a-go-go (EAG)-related potassium channel that functions in these neurons as key cellular components of the circuitry linking peripheral metabolic signals to regulation of neural serotonin signaling. These data raise the possibility that the effects of serotonin on satiety may have their origins in feedback, homeostatic metabolic responses from the periphery.
Collapse
Affiliation(s)
- Aude D. Bouagnon
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Lin Lin
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Shubhi Srivastava
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Chung-Chih Liu
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Oishika Panda
- Boyce Thompson Institute, Cornell University, Ithaca, New York, United States of America
| | - Frank C. Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, New York, United States of America
| | - Supriya Srinivasan
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, United States of America
| | - Kaveh Ashrafi
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
15
|
Wang X, Chen Y, Li J, Guo S, Lin X, Zhang H, Zhan Y, An H. Tetrandrine, a novel inhibitor of ether-à-go-go-1 (Eag1), targeted to cervical cancer development. J Cell Physiol 2019; 234:7161-7173. [PMID: 30362536 DOI: 10.1002/jcp.27470] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022]
Abstract
Mortality-to-incidence ratios in patients with cancer are extremely high, positioning cancer as a major cause of death worldwide. Ether-à-go-go-1 (Eag1) is an ion channel that plays important roles in tumour proliferation, malignant transformation, invasion, metastasis, recurrence, and prognosis. Therefore, identifying potent and specific Eag1 channel inhibitors is crucial. In this study, we identified the first natural inhibitor of Eag1, the traditional Chinese medicine agent tetrandrine, and explored the underlying mechanism. Tetrandrine directly interacted with Eag1 and inhibited the currents in a concentration-dependent manner (IC50 of 69.97 ± 5.2 μM), and the amino acids Ile 550 , Thr 552 , and Gln 557 in the Eag1 C-linker domain were critical for tetrandrine's inhibitory effect. Moreover, tetrandrine reduced the proliferation of HeLa cells and Chinese hamster ovary (CHO) cells stably expressing Eag1 in a concentration-dependent manner. Finally, tetrandrine (30 mg/kg/day) inhibited tumor growth in mice, demonstrating a 64.21% inhibitory rate of HeLa cell-transplanted tumors. These results suggest that tetrandrine is a potent and selective Eag1 channel inhibitor, and could act as a leading compound in the development of therapies for Eag1 ion channel dysfunction-induced diseases.
Collapse
Affiliation(s)
- Xuzhao Wang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, China
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| | - Junwei Li
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| | - Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| | - Xiaoe Lin
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| | - Hailin Zhang
- Department of Pharmacology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yong Zhan
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, China
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| | - Hailong An
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, China
- Key Laboratory of Molecular Biophysics, Hebei Province, School of Science, Hebei University of Technology, Tianjin, China
| |
Collapse
|
16
|
The Interaction between the Drosophila EAG Potassium Channel and the Protein Kinase CaMKII Involves an Extensive Interface at the Active Site of the Kinase. J Mol Biol 2018; 430:5029-5049. [PMID: 30381148 DOI: 10.1016/j.jmb.2018.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
The Drosophila EAG (dEAG) potassium channel is the founding member of the superfamily of KNCH channels, which are involved in cardiac repolarization, neuronal excitability and cellular proliferation. In flies, dEAG is involved in regulation of neuron firing and assembles with CaMKII to form a complex implicated in memory formation. We have characterized the interaction between the kinase domain of CaMKII and a 53-residue fragment of the dEAG channel that includes a canonical CaMKII recognition sequence. Crystal structures together with biochemical/biophysical analysis show a substrate-kinase complex with an unusually tight and extensive interface that appears to be strengthened by phosphorylation of the channel fragment. Electrophysiological recordings show that catalytically active CaMKII is required to observe active dEAG channels. A previously identified phosphorylation site in the recognition sequence is not the substrate for this crucial kinase activity, but rather contributes importantly to the tight interaction of the kinase with the channel. The available data suggest that the dEAG channel is a docking platform for the kinase and that phosphorylation of the channel's kinase recognition sequence modulates the strength of the interaction between the channel and the kinase.
Collapse
|
17
|
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: 4.0] [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
|
18
|
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: 64] [Impact Index Per Article: 9.1] [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
|
19
|
Ruan H, Ueda A, Xing X, Wan X, Strub B, Mukai S, Certel K, Green D, Belozerov K, Yao WD, Johnson W, Jung-Ching Lin J, Hilliker AJ, Wu CF. Generation and characterization of new alleles of quiver (qvr) that encodes an extracellular modulator of the Shaker potassium channel. J Neurogenet 2017; 31:325-336. [PMID: 29117754 DOI: 10.1080/01677063.2017.1393076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Our earlier genetic screen uncovered a paraquat-sensitive leg-shaking mutant quiver1 (qvr1), whose gene product interacts with the Shaker (Sh) K+ channel. We also mapped the qvr locus to EY04063 and noticed altered day-night activity patterns in these mutants. Such circadian behavioral defects were independently reported by another group, who employed the qvr1 allele we supplied them, and attributed the extreme restless phenotype of EY04063 to the qvr gene. However, their report adopted a new noncanonical gene name sleepless (sss) for qvr. In addition to qvr1 and qvrEY, our continuous effort since the early 2000s generated a number of novel recessive qvr alleles, including ethyl methanesulfonate (EMS)-induced mutations qvr2 and qvr3, and P-element excision lines qvrip6 (imprecise jumpout), qvrrv7, and qvrrv9 (revertants) derived from qvrEY. Distinct from the original intron-located qvr1 allele that generates abnormal-sized mRNAs, qvr2, and qvr3 had their lesion sites in exons 6 and 7, respectively, producing nearly normal-sized mRNA products. A set of RNA-editing sites are nearby the lesion sites of qvr3 and qvrEY on exon 7. Except for the revertants, all qvr alleles display a clear ether-induced leg-shaking phenotype just like Sh, and weakened climbing abilities to varying degrees. Unlike Sh, all shaking qvr alleles (except for qvrf01257) displayed a unique activity-dependent enhancement in excitatory junction potentials (EJPs) at larval neuromuscular junctions (NMJs) at very low stimulus frequencies, with qvrEY displaying the largest EJP and more significant NMJ overgrowth than other alleles. Our detailed characterization of a collection of qvr alleles helps to establish links between novel molecular lesions and different behavioral and physiological consequences, revealing how modifications of the qvr gene lead to a wide spectrum of phenotypes, including neuromuscular hyperexcitability, defective motor ability and activity-rest cycles.
Collapse
Affiliation(s)
- Hongyu Ruan
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Atsushi Ueda
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Xiaomin Xing
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Xuxuan Wan
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Benjamin Strub
- b Department of Biology , York University , Toronto , ON , Canada
| | - Spencer Mukai
- b Department of Biology , York University , Toronto , ON , Canada
| | - Kaan Certel
- c Department of Molecular Physiology and Biophysics , University of Iowa , Iowa City , IA , USA
| | - David Green
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Kyle Belozerov
- b Department of Biology , York University , Toronto , ON , Canada
| | - Wei-Dong Yao
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Wayne Johnson
- c Department of Molecular Physiology and Biophysics , University of Iowa , Iowa City , IA , USA
| | | | | | - Chun-Fang Wu
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| |
Collapse
|
20
|
Campos-Lira E, Carrillo E, Aguilar MB, Gajewiak J, Gómez-Lagunas F, López-Vera E. Conorfamide-Sr3, a structurally novel specific inhibitor of the Shaker K + channel. Toxicon 2017; 138:53-58. [DOI: 10.1016/j.toxicon.2017.07.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022]
|
21
|
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: 40] [Impact Index Per Article: 5.7] [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
|
22
|
Sun X, Xu B, Xue Y, Li H, Zhang H, Zhang Y, Kang L, Zhang X, Zhang J, Jia Z, Zhang X. Characterization and structure-activity relationship of natural flavonoids as hERG K + channel modulators. Int Immunopharmacol 2017; 45:187-193. [DOI: 10.1016/j.intimp.2017.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/05/2017] [Accepted: 02/11/2017] [Indexed: 01/21/2023]
|
23
|
Eag1 Voltage-Dependent Potassium Channels: Structure, Electrophysiological Characteristics, and Function in Cancer. J Membr Biol 2017; 250:123-132. [DOI: 10.1007/s00232-016-9944-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/19/2016] [Indexed: 01/07/2023]
|
24
|
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: 3.0] [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
|
25
|
Gómez-Lagunas F, Carrillo E, Pardo LA, Stühmer W. Gating Modulation of the Tumor-Related Kv10.1 Channel by Mibefradil. J Cell Physiol 2017; 232:2019-2032. [DOI: 10.1002/jcp.25448] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 06/02/2016] [Indexed: 01/24/2023]
Affiliation(s)
- Froylán Gómez-Lagunas
- Department of Physiology, School of Medicine. National Autonomous University of Mexico, UNAM; Ciudad Universitaria; México City México DF
| | - Elisa Carrillo
- Department of Physiology, School of Medicine. National Autonomous University of Mexico, UNAM; Ciudad Universitaria; México City México DF
| | - Luis A. Pardo
- Max-Planck Institute for Experimental Medicine; Göttingen Germany
| | - Walter Stühmer
- Max-Planck Institute for Experimental Medicine; Göttingen Germany
| |
Collapse
|
26
|
Cid Uribe JI, Jiménez Vargas JM, Ferreira Batista CV, Zamudio Zuñiga F, Possani LD. Comparative proteomic analysis of female and male venoms from the Mexican scorpion Centruroides limpidus: Novel components found. Toxicon 2017; 125:91-98. [DOI: 10.1016/j.toxicon.2016.11.256] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/23/2016] [Indexed: 10/20/2022]
|
27
|
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: 1.9] [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
|
28
|
Aman TK, Gordon SE, Zagotta WN. Regulation of CNGA1 Channel Gating by Interactions with the Membrane. J Biol Chem 2016; 291:9939-47. [PMID: 26969165 DOI: 10.1074/jbc.m116.723932] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Indexed: 11/06/2022] Open
Abstract
Cyclic nucleotide-gated (CNG) channels are expressed in rod photoreceptors and open in response to direct binding of cyclic nucleotides. We have previously shown that potentiation of CNGA1 channels by transition metals requires a histidine in the A' helix following the S6 transmembrane segment. Here, we used transition metal ion FRET and patch clamp fluorometry with a fluorescent, noncanonical amino acid (3-(6-acetylnaphthalen-2-ylamino)-2-aminopropanoic acid (Anap)) to show that the potentiating transition metal Co(2+) binds in or near the A' helix. Adding high-affinity metal-binding sites to the membrane (stearoyl-nitrilotriacetic acid (C18-NTA)) increased potentiation for low Co(2+) concentrations, indicating that the membrane can coordinate metal ions with the A' helix. These results suggest that restraining the A' helix to the plasma membrane potentiates CNGA1 channel opening. Similar interactions between the A' helix and the plasma membrane may underlie regulation of structurally related hyperpolarization-activated cyclic nucleotide-gated (HCN) and voltage-gated potassium subfamily H (KCNH) channels by plasma membrane components.
Collapse
Affiliation(s)
- Teresa K Aman
- From the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| | - Sharona E Gordon
- From the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| | - William N Zagotta
- From the Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| |
Collapse
|
29
|
Kv10.1 K+ channel: from physiology to cancer. Pflugers Arch 2016; 468:751-62. [DOI: 10.1007/s00424-015-1784-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/11/2015] [Accepted: 12/27/2015] [Indexed: 12/18/2022]
|
30
|
Zhu W, Varga Z, Silva JR. Molecular motions that shape the cardiac action potential: Insights from voltage clamp fluorometry. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 120:3-17. [PMID: 26724572 DOI: 10.1016/j.pbiomolbio.2015.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/11/2015] [Accepted: 12/16/2015] [Indexed: 01/04/2023]
Abstract
Very recently, voltage-clamp fluorometry (VCF) protocols have been developed to observe the membrane proteins responsible for carrying the ventricular ionic currents that form the action potential (AP), including those carried by the cardiac Na(+) channel, NaV1.5, the L-type Ca(2+) channel, CaV1.2, the Na(+)/K(+) ATPase, and the rapid and slow components of the delayed rectifier, KV11.1 and KV7.1. This development is significant, because VCF enables simultaneous observation of ionic current kinetics with conformational changes occurring within specific channel domains. The ability gained from VCF, to connect nanoscale molecular movement to ion channel function has revealed how the voltage-sensing domains (VSDs) control ion flux through channel pores, mechanisms of post-translational regulation and the molecular pathology of inherited mutations. In the future, we expect that this data will be of great use for the creation of multi-scale computational AP models that explicitly represent ion channel conformations, connecting molecular, cell and tissue electrophysiology. Here, we review the VCF protocol, recent results, and discuss potential future developments, including potential use of these experimental findings to create novel computational models.
Collapse
Affiliation(s)
- Wandi Zhu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Zoltan Varga
- MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, Debrecen, Hungary
| | - Jonathan R Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
31
|
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: 2.7] [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
|
32
|
CRYPTOCHROME-mediated phototransduction by modulation of the potassium ion channel β-subunit redox sensor. Proc Natl Acad Sci U S A 2015; 112:2245-50. [PMID: 25646452 DOI: 10.1073/pnas.1416586112] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Blue light activation of the photoreceptor CRYPTOCHROME (CRY) evokes rapid depolarization and increased action potential firing in a subset of circadian and arousal neurons in Drosophila melanogaster. Here we show that acute arousal behavioral responses to blue light significantly differ in mutants lacking CRY, as well as mutants with disrupted opsin-based phototransduction. Light-activated CRY couples to membrane depolarization via a well conserved redox sensor of the voltage-gated potassium (K(+)) channel β-subunit (Kvβ) Hyperkinetic (Hk). The neuronal light response is almost completely absent in hk(-/-) mutants, but is functionally rescued by genetically targeted neuronal expression of WT Hk, but not by Hk point mutations that disable Hk redox sensor function. Multiple K(+) channel α-subunits that coassemble with Hk, including Shaker, Ether-a-go-go, and Ether-a-go-go-related gene, are ion conducting channels for CRY/Hk-coupled light response. Light activation of CRY is transduced to membrane depolarization, increased firing rate, and acute behavioral responses by the Kvβ subunit redox sensor.
Collapse
|
33
|
Shimizu N, Sato N, Kikuchi T, Ishizaki T, Kobayashi K, Kita K, Takimoto K. A sustained increase in the intracellular Ca²⁺ concentration induces proteolytic cleavage of EAG2 channel. Int J Biochem Cell Biol 2014; 59:126-34. [PMID: 25542181 DOI: 10.1016/j.biocel.2014.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 11/19/2014] [Accepted: 12/15/2014] [Indexed: 11/28/2022]
Abstract
Voltage-gated EAG2 channel is abundant in the brain and enhances cancer cell growth by controlling cell volume. The channel contains a cyclic nucleotide-binding homology (CNBH) domain and multiple calmodulin-binding motifs. Here we show that a raised intracellular Ca(2+) concentration causes proteolytic digestion of heterologously expressed and native EAG2 channels. A treatment of EAG2-expressing cells with the Ca(2+) ionophore A23187 for 1h reduces the full-length protein by ∼80% with a concomitant appearance of 30-35-kDa peptides. Similarly, a treatment with the Ca(2+)-ATPase inhibitor thapsigargin for 3h removes 30-35-kDa peptides from ∼1/3 of the channel protein. Moreover, an incubation of the isolated rat brain membrane with CaCl2 leads to the generation of fragments with similar sizes. This Ca(2+)-induced digestion is not seen with EAG1. Mutations in a C-terminal calmodulin-binding motif alter the degrees and positions of the cleavage. Truncated channels that mimic the digested proteins exhibit a reduced current density and altered channel gating. In particular, these shorter channels lack a rapid activation typical in EAG channels with more than 20-mV positive shifts in voltage dependence of activation. The truncation also eliminates the ability of EAG2 channel to reduce cell volume. These results suggest that a sustained increase in the intracellular Ca(2+) concentration leads to proteolytic cleavage at the C-terminal cytosolic region following the CNBH domain by altering its interaction with calmodulin. The observed Ca(2+)-induced proteolytic cleavage of EAG2 channel may act as an adaptive response under physiological and/or pathological conditions.
Collapse
Affiliation(s)
- Nobuhiro Shimizu
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Natsumi Sato
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Teppei Kikuchi
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Takuro Ishizaki
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Kazuto Kobayashi
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | - Kaori Kita
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Koichi Takimoto
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan; Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
34
|
Wei H, Yasar H, Funk NW, Giese M, Baz ES, Stengl M. Signaling of pigment-dispersing factor (PDF) in the Madeira cockroach Rhyparobia maderae. PLoS One 2014; 9:e108757. [PMID: 25269074 PMCID: PMC4182629 DOI: 10.1371/journal.pone.0108757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 08/05/2014] [Indexed: 11/19/2022] Open
Abstract
The insect neuropeptide pigment-dispersing factor (PDF) is a functional ortholog of vasoactive intestinal polypeptide, the coupling factor of the mammalian circadian pacemaker. Despite of PDF's importance for synchronized circadian locomotor activity rhythms its signaling is not well understood. We studied PDF signaling in primary cell cultures of the accessory medulla, the circadian pacemaker of the Madeira cockroach. In Ca²⁺ imaging studies four types of PDF-responses were distinguished. In regularly bursting type 1 pacemakers PDF application resulted in dose-dependent long-lasting increases in Ca²⁺ baseline concentration and frequency of oscillating Ca²⁺ transients. Adenylyl cyclase antagonists prevented PDF-responses in type 1 cells, indicating that PDF signaled via elevation of intracellular cAMP levels. In contrast, in type 2 pacemakers PDF transiently raised intracellular Ca²⁺ levels even after blocking adenylyl cyclase activity. In patch clamp experiments the previously characterized types 1-4 could not be identified. Instead, PDF-responses were categorized according to ion channels affected. Application of PDF inhibited outward potassium or inward sodium currents, sometimes in the same neuron. In a comparison of Ca²⁺ imaging and patch clamp experiments we hypothesized that in type 1 cells PDF-dependent rises in cAMP concentrations block primarily outward K⁺ currents. Possibly, this PDF-dependent depolarization underlies PDF-dependent phase advances of pacemakers. Finally, we propose that PDF-dependent concomitant modulation of K⁺ and Na⁺ channels in coupled pacemakers causes ultradian membrane potential oscillations as prerequisite to efficient synchronization via resonance.
Collapse
Affiliation(s)
- Hongying Wei
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Hanzey Yasar
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Nico W. Funk
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Maria Giese
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - El-Sayed Baz
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Monika Stengl
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
- * E-mail:
| |
Collapse
|
35
|
Li Q, Ng HQ, Yoon HS, Kang C. Solution structure of the cyclic-nucleotide binding homology domain of a KCNH channel. J Struct Biol 2014; 186:68-74. [PMID: 24632450 DOI: 10.1016/j.jsb.2014.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/03/2014] [Accepted: 03/10/2014] [Indexed: 12/28/2022]
Abstract
The carboxy-terminal region of the KCNH family of potassium channels contains a cyclic-nucleotide binding homology domain (CNBHD) that is important for channel gating and trafficking. The solution structure of the CNBHD of the KCNH potassium of zebrafish was determined using solution NMR spectroscopy. This domain exists as a monomer under solution conditions and adopts a similar fold to that determined by X-ray crystallography. The CNBHD does not bind cAMP because residue Y740 blocks the entry of cyclic-nucleotide to the binding pocket. Relaxation results show that the CNBHD is rigid except that some residues in the loop between β6 and β7 are flexible. Our results will be useful to understand the gating mechanism of KCNH family members through the CNBHD.
Collapse
Affiliation(s)
- Qingxin Li
- Institute of Chemical & Engineering Sciences, Agency for Science, Technology and Research (A∗STAR), Singapore 627833, Singapore
| | - Hui Qi Ng
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A∗STAR), Singapore 138669, Singapore
| | - Ho Sup Yoon
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637511, Singapore
| | - Congbao Kang
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A∗STAR), Singapore 138669, Singapore.
| |
Collapse
|
36
|
Multistate structural modeling and voltage-clamp analysis of epilepsy/autism mutation Kv10.2-R327H demonstrate the role of this residue in stabilizing the channel closed state. J Neurosci 2013; 33:16586-93. [PMID: 24133262 DOI: 10.1523/jneurosci.2307-13.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Voltage-gated potassium channel Kv10.2 (KCNH5) is expressed in the nervous system, but its functions and involvement in human disease are poorly understood. We studied a human Kv10.2 channel mutation (R327H) recently identified in a child with epileptic encephalopathy and autistic features. Using multistate structural modeling, we demonstrate that the Arg327 residue in the S4 helix of voltage-sensing domain has strong ionic interactions with negatively charged residues within the S1-S3 helices in the resting (closed) and early-activation state but not in the late-activation and fully-activated (open) state. The R327H mutation weakens ionic interactions between residue 327 and these negatively charged residues, thus favoring channel opening. Voltage-clamp analysis showed a strong hyperpolarizing (∼70 mV) shift of voltage dependence of activation and an acceleration of activation. Our results demonstrate the critical role of the Arg327 residue in stabilizing the channel closed state and explicate for the first time the structural and functional change of a Kv10.2 channel mutation associated with neurological disease.
Collapse
|
37
|
Haitin Y, Carlson AE, Zagotta WN. The structural mechanism of KCNH-channel regulation by the eag domain. Nature 2013; 501:444-8. [PMID: 23975098 PMCID: PMC3910112 DOI: 10.1038/nature12487] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/18/2013] [Indexed: 12/22/2022]
Abstract
The KCNH voltage-dependent potassium channels (ether-à-go-go, EAG; EAG-related gene, ERG; EAG-like channels, ELK) are important regulators of cellular excitability and have key roles in diseases such as cardiac long QT syndrome type 2 (LQT2), epilepsy, schizophrenia and cancer. The intracellular domains of KCNH channels are structurally distinct from other voltage-gated channels. The amino-terminal region contains an eag domain, which is composed of a Per-Arnt-Sim (PAS) domain and a PAS-cap domain, whereas the carboxy-terminal region contains a cyclic nucleotide-binding homology domain (CNBHD), which is connected to the pore through a C-linker domain. Many disease-causing mutations localize to these specialized intracellular domains, which underlie the unique gating and regulation of KCNH channels. It has been suggested that the eag domain may regulate the channel by interacting with either the S4-S5 linker or the CNBHD. Here we present a 2 Å resolution crystal structure of the eag domain-CNBHD complex of the mouse EAG1 (also known as KCNH1) channel. It displays extensive interactions between the eag domain and the CNBHD, indicating that the regulatory mechanism of the eag domain primarily involves the CNBHD. Notably, the structure reveals that a number of LQT2 mutations at homologous positions in human ERG, in addition to cancer-associated mutations in EAG channels, localize to the eag domain-CNBHD interface. Furthermore, mutations at the interface produced marked effects on channel gating, demonstrating the important physiological role of the eag domain-CNBHD interaction. Our structure of the eag domain-CNBHD complex of mouse EAG1 provides unique insights into the physiological and pathophysiological mechanisms of KCNH channels.
Collapse
Affiliation(s)
- Yoni Haitin
- Department of Physiology and Biophysics, University of Washington
School of Medicine, Seattle, WA 98195
| | - Anne E. Carlson
- Department of Physiology and Biophysics, University of Washington
School of Medicine, Seattle, WA 98195
| | - William N. Zagotta
- Department of Physiology and Biophysics, University of Washington
School of Medicine, Seattle, WA 98195
| |
Collapse
|
38
|
Structure of the C-terminal region of an ERG channel and functional implications. Proc Natl Acad Sci U S A 2013; 110:11648-53. [PMID: 23801759 DOI: 10.1073/pnas.1306887110] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human ether-à-go-go-related gene (hERG) encodes a K(+) channel crucial for repolarization of the cardiac action potential. EAG-related gene (ERG) channels contain a C-terminal cyclic nucleotide-binding homology domain coupled to the pore of the channel by a C-linker. Here, we report the structure of the C-linker/cyclic nucleotide-binding homology domain of a mosquito ERG channel at 2.5-Å resolution. The structure reveals that the region expected to form the cyclic nucleotide-binding pocket is negatively charged and is occupied by a short β-strand, referred to as the intrinsic ligand, explaining the lack of direct regulation of ERG channels by cyclic nucleotides. In hERG channels, the intrinsic ligand harbors hereditary mutations associated with long-QT syndrome (LQTS), a potentially lethal cardiac arrhythmia. Mutations in the intrinsic ligand affected hERG channel gating and LQTS mutations abolished hERG currents and altered trafficking of hERG channels, which explains the LQT phenotype. The structure also reveals a dramatically different conformation of the C-linker compared with the structures of the related ether-à-go-go-like K(+) and hyperpolarization-activated cyclic nucleotide-modulated channels, suggesting that the C-linker region may be highly dynamic in the KCNH, hyperpolarization-activated cyclic nucleotide-modulated, and cyclic nucleotide-gated channels.
Collapse
|
39
|
Ryan MY, Maloney R, Fineberg JD, Reenan RA, Horn R. RNA editing in eag potassium channels: biophysical consequences of editing a conserved S6 residue. Channels (Austin) 2012; 6:443-52. [PMID: 23064203 DOI: 10.4161/chan.22314] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
RNA editing at four sites in eag, a Drosophila voltage-gated potassium channel, results in the substitution of amino acids into the final protein product that are not encoded by the genome. These sites and the editing alterations introduced are K467R (Site 1, top of the S6 segment), Y548C, N567D and K699R (sites 2-4, within the cytoplasmic C-terminal domain). We mutated these residues individually and expressed the channels in Xenopus oocytes. A fully edited construct (all four sites) has the slowest activation kinetics and a paucity of inactivation, whereas the fully unedited channel exhibits the fastest activation and most dramatic inactivation. Editing Site 1 inhibits steady-state inactivation. Mutating Site 1 to the neutral residues resulted in intermediate inactivation phenotypes and a leftward shift of the peak current-voltage relationship. Activation kinetics display a Cole-Moore shift that is enhanced by RNA editing. Normalized open probability relationships for 467Q, 467R and 467K are superimposable, indicating little effect of the mutations on steady-state activation. 467Q and 467R enhance instantaneous inward rectification, indicating a role of this residue in ion permeation. Intracellular tetrabutylammonium blocks 467K significantly better than 467R. Block by intracellular, but not extracellular, tetraethylammonium interferes with inactivation. The fraction of inactivated current is reduced at higher extracellular Mg(+2) concentrations, and channels edited at Site 1 are more sensitive to changes in extracellular Mg(+2) than unedited channels. These results show that even a minor change in amino acid side-chain chemistry and size can have a dramatic impact on channel biophysics, and that RNA editing is important for fine-tuning the channel's function.
Collapse
Affiliation(s)
- Mary Y Ryan
- Department of Molecular Physiology & Biophysics, Institute of Hyperexcitability, Jefferson Medical College, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
40
|
Huang X, Dubuc AM, Hashizume R, Berg J, He Y, Wang J, Chiang C, Cooper MK, Northcott PA, Taylor MD, Barnes MJ, Tihan T, Chen J, Hackett CS, Weiss WA, James CD, Rowitch DH, Shuman MA, Jan YN, Jan LY. Voltage-gated potassium channel EAG2 controls mitotic entry and tumor growth in medulloblastoma via regulating cell volume dynamics. Genes Dev 2012; 26:1780-96. [PMID: 22855790 DOI: 10.1101/gad.193789.112] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.
Collapse
Affiliation(s)
- Xi Huang
- Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Structure of the carboxy-terminal region of a KCNH channel. Nature 2012; 481:530-3. [PMID: 22230959 PMCID: PMC3267858 DOI: 10.1038/nature10735] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 11/23/2011] [Indexed: 11/15/2022]
Abstract
The KCNH family of ion channels, comprising ether-à-go-go (EAG), EAG-related gene (ERG), and EAG-like (ELK) K+ channel subfamilies, is crucial for repolarization of the cardiac action potential1, regulation of neuronal excitability2, and proliferation of tumor cells3. The C-terminal region of KCNH channels contains a cyclic nucleotide-binding homology domain (CNBHD) and C-linker that couples the CNBHD to the pore4. The C-linker/CNBHD is essential for proper function and trafficking of ion channels in the KCNH family5–9. However, despite the importance of the C-linker/CNBHD for the function of KCNH channels, the structural basis of ion channel regulation by the C-linker/CNBHD is unknown. Here we report the crystal structure of the C-linker/CNBHD of zebrafish ELK channels at 2.2 Å resolution. While the overall structure of the C-linker/CNBHD of zELK channels is similar to the cyclic nucleotide-binding domain (CNBD) structure of the related HCN channels10, there are dramatic differences. Unlike the CNBD of HCN, the CNBHD of zELK displays a negatively charged electrostatic profile that explains the lack of binding and regulation of KCNH channels by cyclic nucleotides4,11. Instead of cyclic nucleotide, the binding pocket is occupied by a short β-strand. Mutations of the β-strand shift the voltage dependence of activation to more depolarized voltages, implicating the β-strand as an intrinsic ligand for the CNBHD of zELK channels. In both zELK and HCN channels the C-linker is the site of virtually all of the intersubunit interactions in the C-terminal region. However, in the zELK structure there is a reorientation in the C-linker so the subunits form dimers instead of tetramers as observed in HCN channels. These results provide a structural framework for understanding the regulation of ion channels in the KCNH family by the C-linker/CNBHD and may guide the design of specific drugs.
Collapse
|
42
|
Asher V, Warren A, Shaw R, Sowter H, Bali A, Khan R. The role of Eag and HERG channels in cell proliferation and apoptotic cell death in SK-OV-3 ovarian cancer cell line. Cancer Cell Int 2011; 11:6. [PMID: 21392380 PMCID: PMC3063814 DOI: 10.1186/1475-2867-11-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 03/10/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The voltage gated potassium (K+) channels Eag and HERG have been implicated in the pathogenesis of various cancers, through association with cell cycle changes and programmed cell death. The role of these channels in the onset and progression of ovarian cancer is unknown. An understanding of mechanism by which Eag and HERG channels affect cell proliferation in ovarian cancer cells is required and therefore we investigated their role in cell proliferation and their effect on the cell cycle and apoptosis of ovarian cancer cells. METHODS The presence of Eag and HERG was determined in SK-OV-3 cells using immunofluorescence and western blotting. The effect of the Eag blockers (imipramine and clofilium) and HERG blockers (E-4031 and ergtoxin) on cell proliferation was assessed using the MTS assay with further investigation of their role in the cell cycle and apoptosis determined by flow cytometry. RESULTS Eag and HERG channels were present in the cytoplasm and nuclei of SK-OV-3 cells. There was significant inhibition of proliferation of SK-OV-3 cells by imipramine (P < 0.001) and ergtoxin (P < 0.05) at 72 hours of culture. Incubation of cells with ergtoxin led to the accumulation of cells in the S and G2/M phase, while cells accumulated in S phase after incubation with E-4031, with no effect on apoptosis. Imipramine did not affect the cell cycle but increased the proportion of SK-OV-3 cells undergoing early apoptosis. CONCLUSION Both Eag and HERG channels are expressed in SK-OV-3 ovarian cancer cells and have a role in cell proliferation. HERG channels affect the cell cycle while Eag channels are implicated in the inhibition of apoptosis of ovarian cancer cells. The family of Eag channels may represent a new therapeutic target for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Viren Asher
- School of Graduate Entry Medicine and Health Royal Derby Hospital, Uttoxeter Road, Derby DE22 3DT, UK.
| | | | | | | | | | | |
Collapse
|
43
|
LeBoeuf B, Guo X, García LR. The effects of transient starvation persist through direct interactions between CaMKII and ether-a-go-go K+ channels in C. elegans males. Neuroscience 2011; 175:1-17. [PMID: 21145946 PMCID: PMC3059131 DOI: 10.1016/j.neuroscience.2010.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 11/30/2010] [Accepted: 12/01/2010] [Indexed: 12/29/2022]
Abstract
Prolonged nutrient limitation has been extensively studied due to its positive effects on life span. However, less is understood of how brief periods of starvation can have lasting consequences. In this study, we used genetics, biochemistry, pharmacology and behavioral analysis to show that after a limited period of starvation, the synthesis of egl-2-encoded ether-a-go-go (EAG) K+ channels and its C-terminal modifications by unc-43-encoded CaMKII have a perduring effect on C. elegans male sexual behavior. EGL-2 and UNC-43 interactions, induced after food deprivation, maintain reduced excitability in muscles involved in sex. In young adult males, spastic contractions occur in cholinergic-activated sex muscles that lack functional unc-103-encoded ERG-like K+ channels. Promoting EGL-2 and UNC-43 interactions in unc-103 mutant adult males by starving them for a few hours reduce spastic muscle contractions over multiple days. Although transient starvation during early adulthood has a hormetic effect of suppressing mutation-induced muscle contractions, the treatment reduces the ability of young wild-type (WT) males to compete with well-fed cohorts in siring progeny.
Collapse
Affiliation(s)
- Brigitte LeBoeuf
- Howard Hughes Medical Institute, Texas A&M University, 3258 TAMU, College Station, TX 77843-3258
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX 77843-3258
| | - Xiaoyan Guo
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX 77843-3258
| | - L. René García
- Howard Hughes Medical Institute, Texas A&M University, 3258 TAMU, College Station, TX 77843-3258
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX 77843-3258
| |
Collapse
|
44
|
Asher V, Sowter H, Shaw R, Bali A, Khan R. Eag and HERG potassium channels as novel therapeutic targets in cancer. World J Surg Oncol 2010; 8:113. [PMID: 21190577 PMCID: PMC3022597 DOI: 10.1186/1477-7819-8-113] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/29/2010] [Indexed: 12/03/2022] Open
Abstract
Voltage gated potassium channels have been extensively studied in relation to cancer. In this review, we will focus on the role of two potassium channels, Ether à-go-go (Eag), Human ether à-go-go related gene (HERG), in cancer and their potential therapeutic utility in the treatment of cancer. Eag and HERG are expressed in cancers of various organs and have been implicated in cell cycle progression and proliferation of cancer cells. Inhibition of these channels has been shown to reduce proliferation both in vitro and vivo studies identifying potassium channel modulators as putative inhibitors of tumour progression. Eag channels in view of their restricted expression in normal tissue may emerge as novel tumour biomarkers.
Collapse
Affiliation(s)
- Viren Asher
- Department of Obstetrics and Gynaecology, School of Graduate Medicine and Health, Royal Derby Hospital, Uttoxeter road, Derby DE22 3DT, UK.
| | | | | | | | | |
Collapse
|
45
|
Xu X, Vysotskaya ZV, Liu Q, Zhou L. Structural basis for the cAMP-dependent gating in the human HCN4 channel. J Biol Chem 2010; 285:37082-91. [PMID: 20829353 PMCID: PMC2978636 DOI: 10.1074/jbc.m110.152033] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Revised: 09/08/2010] [Indexed: 11/06/2022] Open
Abstract
Hyperpolarization-activated cAMP-regulated (HCN) channels play important physiological roles in both cardiovascular and central nervous systems. Among the four HCN isoforms, HCN2 and HCN4 show high expression levels in the human heart, with HCN4 being the major cardiac isoform. The previously published crystal structure of the mouse HCN2 (mHCN2) C-terminal fragment, including the C-linker and the cyclic-nucleotide binding domain (CNBD), has provided many insights into cAMP-dependent gating in HCN channels. However, structures of other mammalian HCN channel isoforms have been lacking. Here we used a combination of approaches including structural biology, biochemistry, and electrophysiology to study cAMP-dependent gating in HCN4 channel. First we solved the crystal structure of the C-terminal fragment of human HCN4 (hHCN4) channel at 2.4 Å. Overall we observed a high similarity between mHCN2 and hHCN4 crystal structures. Functional comparison between two isoforms revealed that compared with mHCN2, the hHCN4 protein exhibited marked different contributions to channel function, such as a ∼3-fold reduction in the response to cAMP. Guided by structural differences in the loop region between β4 and β5 strands, we identified residues that could partially account for the differences in response to cAMP between mHCN2 and hHCN4 proteins. Moreover, upon cAMP binding, the hHCN4 C-terminal protein exerts a much prolonged effect in channel deactivation that could have significant physiological contributions.
Collapse
Affiliation(s)
- Xinping Xu
- From the Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Zhanna V. Vysotskaya
- From the Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Qinglian Liu
- From the Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Lei Zhou
- From the Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298
| |
Collapse
|
46
|
Li Y, Liu X, Wu Y, Xu Z, Li H, Griffith LC, Zhou Y. Intracellular regions of the Eag potassium channel play a critical role in generation of voltage-dependent currents. J Biol Chem 2010; 286:1389-99. [PMID: 21059657 DOI: 10.1074/jbc.m110.184077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Folding, assembly, and trafficking of ion channels are tightly controlled processes and are important for biological functions relevant to health and disease. Here, we report that functional expression of the Eag channel is temperature-sensitive by a mechanism that is independent of trafficking or surface targeting of the channel protein. Eag channels in cells grown at 37 °C exhibit voltage-evoked gating charge movements but fail to conduct K(+) ions. By mutagenesis and chimeric channel studies, we show that the N- and C-terminal regions are involved in controlling a step after movement of the voltage sensor, as well as in regulating biophysical properties of the Eag channel. Synthesis and assembly of Eag at high temperature disrupt the ability of these domains to carry out their function. These results suggest an important role of the intracellular regions in the generation of Eag currents.
Collapse
Affiliation(s)
- Yong Li
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida 32306, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Brelidze TI, Carlson AE, Davies DR, Stewart LJ, Zagotta WN. Identifying regulators for EAG1 channels with a novel electrophysiology and tryptophan fluorescence based screen. PLoS One 2010; 5. [PMID: 20824064 PMCID: PMC2932742 DOI: 10.1371/journal.pone.0012523] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 07/02/2010] [Indexed: 11/19/2022] Open
Abstract
Background Ether-à-go-go (EAG) channels are expressed throughout the central nervous system and are also crucial regulators of cell cycle and tumor progression. The large intracellular amino- and carboxy- terminal domains of EAG1 each share similarity with known ligand binding motifs in other proteins, yet EAG1 channels have no known regulatory ligands. Methodology/Principal Findings Here we screened a library of small biologically relevant molecules against EAG1 channels with a novel two-pronged screen to identify channel regulators. In one arm of the screen we used electrophysiology to assess the functional effects of the library compounds on full-length EAG1 channels. In an orthogonal arm, we used tryptophan fluorescence to screen for binding of the library compounds to the isolated C-terminal region. Conclusions/Significance Several compounds from the flavonoid, indole and benzofuran chemical families emerged as binding partners and/or regulators of EAG1 channels. The two-prong screen can aid ligand and drug discovery for ligand-binding domains of other ion channels.
Collapse
Affiliation(s)
- Tinatin I. Brelidze
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Anne E. Carlson
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Douglas R. Davies
- Emerald BioStructures, Inc., Bainbridge Island, Washington, United States of America
| | - Lance J. Stewart
- Emerald BioStructures, Inc., Bainbridge Island, Washington, United States of America
| | - William N. Zagotta
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
48
|
Liu X, Wu Y, Zhou Y. Intracellular linkers are involved in Mg2+-dependent modulation of the Eag potassium channel. Channels (Austin) 2010; 4:311-8. [PMID: 20855938 DOI: 10.4161/chan.4.4.12329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Modulation of activation kinetics by divalent ions is one of the characteristic features of Eag channels. Here, we report that Mg(2+)-dependent deceleration of Eag channel activation is significantly attenuated by a G297E mutation, which exhibits a gain-of-function phenotype in Drosophila by suppressing the effect of shaker mutation on behavior and neuronal excitability. The G297 residue is located in the intracellular linker of transmembrane segments S2 and S3, and is thus not involved in direct binding of Mg(2+) ions. Moreover, mutation of the only positively charged residue in the other intracellular linker between S4 and S5 also results in a dramatic reduction of Mg(2+)-dependent modulation of Eag activation kinetics. Collectively, the two mutations in eag eliminate or even paradoxically reverse the effect of Mg(2+) on channel activation and inactivation kinetics. Together, these results suggest an important role of the intracellular linker regions in gating processes of Eag channels.
Collapse
Affiliation(s)
- Xinqiu Liu
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| | | | | |
Collapse
|
49
|
Noma K, Kimura K, Minatohara K, Nakashima H, Nagao Y, Mizoguchi A, Fujiyoshi Y. Triple N-glycosylation in the long S5-P loop regulates the activation and trafficking of the Kv12.2 potassium channel. J Biol Chem 2009; 284:33139-50. [PMID: 19808681 DOI: 10.1074/jbc.m109.021519] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mammalian voltage-dependent potassium (Kv) channels regulate the excitability of nerve and muscle cells. Kv12.2 features the longest S5-P loop among all known mammalian Kv channels with the most N-linked glycosylation sites (three sites). Despite its unique structural features, Kv12.2 is not well characterized. Because glycosylation plays important roles in the folding, trafficking, and function of various Kv channels, we focused on the N-glycosylation of Kv12.2. We show that Kv12.2 is N-glycosylated in Chinese hamster ovary (CHO) cells and in cultured neurons as well as in the mouse brain. As an effect of N-glycosylation on the function of Kv12.2, we demonstrate that removal of sugar chains causes a depolarizing shift in the steady-state activation without a significant reduction in current amplitude. Unlike the previously reported shift for Shaker-type Kv channels, this shift does not appear to be due to negatively charged sialic acid residues in the sugar chains. We next examined the trafficking in CHO cells to address whether the unglycosylated Kv12.2 channels are utilized in vivo. Although double mutants, retaining only one glycosylation site, are trafficked to the surface of CHO cells irrespective of the position of the glycosylated site, unglycosylated channels are not trafficked to the cell surface. Furthermore, we could not detect unglycosylated channels in the mouse brain. Our data suggest that only glycosylated Kv12.2 channels show proper voltage dependence and are utilized in vivo.
Collapse
Affiliation(s)
- Kentaro Noma
- Department of Biophysics, Graduate School of Science, Kyoto University, Oiwake, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | | | | | | | | | |
Collapse
|
50
|
Erg K+ currents modulate excitability in mouse mitral/tufted neurons. Pflugers Arch 2009; 459:55-70. [DOI: 10.1007/s00424-009-0709-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 06/13/2009] [Accepted: 07/30/2009] [Indexed: 10/20/2022]
|