Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.633
1.
Elife ; 122024 Apr 23.
Article En | MEDLINE | ID: mdl-38652113

Lymphoid restricted membrane protein (LRMP) is a specific regulator of the hyperpolarization-activated cyclic nucleotide-sensitive isoform 4 (HCN4) channel. LRMP prevents cAMP-dependent potentiation of HCN4, but the interaction domains, mechanisms of action, and basis for isoform-specificity remain unknown. Here, we identify the domains of LRMP essential for this regulation, show that LRMP acts by disrupting the intramolecular signal transduction between cyclic nucleotide binding and gating, and demonstrate that multiple unique regions in HCN4 are required for LRMP isoform-specificity. Using patch clamp electrophysiology and Förster resonance energy transfer (FRET), we identified the initial 227 residues of LRMP and the N-terminus of HCN4 as necessary for LRMP to associate with HCN4. We found that the HCN4 N-terminus and HCN4-specific residues in the C-linker are necessary for regulation of HCN4 by LRMP. Finally, we demonstrated that LRMP-regulation can be conferred to HCN2 by addition of the HCN4 N-terminus along with mutation of five residues in the S5 region and C-linker to the cognate HCN4 residues. Taken together, these results suggest that LRMP inhibits HCN4 through an isoform-specific interaction involving the N-terminals of both proteins that prevents the transduction of cAMP binding into a change in channel gating, most likely via an HCN4-specific orientation of the N-terminus, C-linker, and S4-S5 linker.


Cyclic AMP , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Proteins , Muscle Proteins , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Cyclic AMP/metabolism , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Animals , Protein Binding , HEK293 Cells , Potassium Channels/metabolism , Potassium Channels/genetics , Potassium Channels/chemistry , Patch-Clamp Techniques , Fluorescence Resonance Energy Transfer , Protein Isoforms/metabolism , Protein Isoforms/genetics
2.
PLoS Comput Biol ; 20(3): e1011559, 2024 Mar.
Article En | MEDLINE | ID: mdl-38517941

Cyclic AMP controls neuronal ion channel activity. For example hyperpolarization-activated cyclic nucleotide-gated (HCN) and M-type K+ channels are activated by cAMP. These effects have been suggested to be involved in astrocyte control of neuronal activity, for example, by controlling the action potential firing frequency. In cortical neurons, cAMP can induce mixed-mode oscillations (MMOs) consisting of small-amplitude, subthreshold oscillations separating complete action potentials, which lowers the firing frequency greatly. We extend a model of neuronal activity by including HCN and M channels, and show that it can reproduce a series of experimental results under various conditions involving and inferring with cAMP-induced activation of HCN and M channels. In particular, we find that the model can exhibit MMOs as found experimentally, and argue that both HCN and M channels are crucial for reproducing these patterns. To understand how M and HCN channels contribute to produce MMOs, we exploit the fact that the model is a three-time scale dynamical system with one fast, two slow, and two super-slow variables. We show that the MMO mechanism does not rely on the super-slow dynamics of HCN and M channel gating variables, since the model is able to produce MMOs even when HCN and M channel activity is kept constant. In other words, the cAMP-induced increase in the average activity of HCN and M channels allows MMOs to be produced by the slow-fast subsystem alone. We show that the slow-fast subsystem MMOs are due to a folded node singularity, a geometrical structure well known to be involved in the generation of MMOs in slow-fast systems. Besides raising new mathematical questions for multiple-timescale systems, our work is a starting point for future research on how cAMP signalling, for example resulting from interactions between neurons and glial cells, affects neuronal activity via HCN and M channels.


Nucleotides, Cyclic , Potassium Channels , Potassium Channels/chemistry , Nucleotides, Cyclic/pharmacology , Neurons , Cyclic AMP , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Cyclic Nucleotide-Gated Cation Channels
3.
Methods ; 225: 89-99, 2024 May.
Article En | MEDLINE | ID: mdl-38508347

A variety of equilibrium and non-equilibrium methods have been used in a multidisciplinary approach to study the conformational landscape associated with the binding of different cations to the pore of potassium channels. These binding processes, and the conformational changes resulting therefrom, modulate the functional properties of such integral membrane properties, revealing these permeant and blocking cations as true effectors of such integral membrane proteins. KcsA, a prototypic K+ channel from Streptomyces lividans, has been extensively characterized in this regard. Here, we revise several fluorescence-based approaches to monitor cation binding under different experimental conditions in diluted samples, analyzing the advantages and disadvantages of each approach. These studies have contributed to explain the selectivity, conduction, and inactivation properties of K+ channels at the molecular level, together with the allosteric communication between the two gates that control the ion channel flux, and how they are modulated by lipids.


Potassium Channels , Protein Conformation , Potassium Channels/chemistry , Potassium Channels/metabolism , Streptomyces lividans/metabolism , Streptomyces lividans/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Spectrometry, Fluorescence/methods , Protein Binding , Fluorescent Dyes/chemistry , Ion Channel Gating
4.
J Biol Chem ; 300(4): 107155, 2024 Apr.
Article En | MEDLINE | ID: mdl-38479597

Despite significant advances in the development of therapeutic interventions targeting autoimmune diseases and chronic inflammatory conditions, lack of effective treatment still poses a high unmet need. Modulating chronically activated T cells through the blockade of the Kv1.3 potassium channel is a promising therapeutic approach; however, developing selective Kv1.3 inhibitors is still an arduous task. Phage display-based high throughput peptide library screening is a rapid and robust approach to develop promising drug candidates; however, it requires solid-phase immobilization of target proteins with their binding site preserved. Historically, the KcsA bacterial channel chimera harboring only the turret region of the human Kv1.3 channel was used for screening campaigns. Nevertheless, literature data suggest that binding to this type of chimera does not correlate well with blocking potency on the native Kv1.3 channels. Therefore, we designed and successfully produced advanced KcsA-Kv1.3, KcsA-Kv1.1, and KcsA-Kv1.2 chimeric proteins in which both the turret and part of the filter regions of the human Kv1.x channels were transferred. These T+F (turret-filter) chimeras showed superior peptide ligand-binding predictivity compared to their T-only versions in novel phage ELISA assays. Phage ELISA binding and competition results supported with electrophysiological data confirmed that the filter region of KcsA-Kv1.x is essential for establishing adequate relative affinity order among selected peptide toxins (Vm24 toxin, Hongotoxin-1, Kaliotoxin-1, Maurotoxin, Stichodactyla toxin) and consequently obtaining more reliable selectivity data. These new findings provide a better screening tool for future drug development efforts and offer insight into the target-ligand interactions of these therapeutically relevant ion channels.


Kv1.3 Potassium Channel , Potassium Channel Blockers , Recombinant Fusion Proteins , Humans , Kv1.3 Potassium Channel/metabolism , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/chemistry , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Binding Sites , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/antagonists & inhibitors , Ligands , Potassium Channels/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Peptide Library
5.
Int J Mol Sci ; 25(3)2024 Feb 04.
Article En | MEDLINE | ID: mdl-38339159

KCTD ((K)potassium Channel Tetramerization Domain-containing) proteins constitute an emerging class of proteins involved in fundamental physio-pathological processes. In these proteins, the BTB domain, which represents the defining element of the family, may have the dual role of promoting oligomerization and favoring functionally important partnerships with different interactors. Here, by exploiting the potential of recently developed methodologies for protein structure prediction, we report a comprehensive analysis of the interactions of all KCTD proteins with their most common partner Cullin 3 (Cul3). The data here presented demonstrate the impressive ability of this approach to discriminate between KCTDs that interact with Cul3 and those that do not. Indeed, reliable and stable models of the complexes were only obtained for the 15 members of the family that are known to interact with Cul3. The generation of three-dimensional models for all KCTD-Cul3 complexes provides interesting clues on the determinants of the structural basis of this partnership as clear structural differences emerged between KCTDs that bind or do not bind Cul3. Finally, the availability of accurate three-dimensional models for KCTD-Cul3 interactions may be valuable for the ad hoc design and development of compounds targeting specific KCTDs that are involved in several common diseases.


Cullin Proteins , Potassium Channels , Humans , Amino Acid Sequence , Cullin Proteins/chemistry , Potassium Channels/chemistry , Protein Binding , Protein Multimerization
6.
Chem Commun (Camb) ; 60(5): 482-500, 2024 Jan 11.
Article En | MEDLINE | ID: mdl-38111319

Potassium channels represent the most prevalent class of ion channels, exerting regulatory control over numerous vital biological processes, including muscle contraction, neurotransmitter release, cell proliferation, and apoptosis. The seamless integration of astonishing functions into a sophisticated structure, as seen in these protein channels, inspires the chemical community to develop artificial versions, gearing toward simplifying their structure while replicating their key functions. In particular, over the past ten years or so, a number of elegant artificial potassium transporters have emerged, demonstrating high selectivity, high transport efficiency or unprecedented transport mechanisms. In this review, we will provide a detailed exposition of these artificial potassium transporters that are derived from a single molecular backbone or self-assembled from multiple components, with their respective structural designs, channel functions, transport mechanisms and biomedical applications thoroughly reviewed.


Ion Channels , Potassium , Potassium/metabolism , Ion Channels/metabolism , Potassium Channels/chemistry , Potassium Channels/metabolism , Biological Transport
7.
J Am Chem Soc ; 145(33): 18529-18537, 2023 08 23.
Article En | MEDLINE | ID: mdl-37578394

The potassium ion (K+) configurations of the selectivity filter of the KcsA ion channel protein are investigated with two-dimensional infrared (2D IR) spectroscopy of amide I vibrations. Single 13C-18O isotope labels are used, for the first time, to selectively probe the S1/S2 or S2/S3 binding sites in the selectivity filter. These binding sites have the largest differences in ion occupancy in two competing K+ transport mechanisms: soft-knock and hard-knock. According to the former, water molecules alternate between K+ ions in the selectivity filter while the latter assumes that K+ ions occupy the adjacent sites. Molecular dynamics simulations and computational spectroscopy are employed to interpret experimental 2D IR spectra. We find that in the closed conductive state of the KcsA channel, K+ ions do not occupy adjacent binding sites. The experimental data is consistent with simulated 2D IR spectra of soft-knock ion configurations. In contrast, the simulated spectra for the hard-knock ion configurations do not reproduce the experimental results. 2D IR spectra of the hard-knock mechanism have lower frequencies, homogeneous 2D lineshapes, and multiple peaks. In contrast, ion configurations of the soft-knock model produce 2D IR spectra with a single peak at a higher frequency and inhomogeneous lineshape. We conclude that under equilibrium conditions, in the absence of transmembrane voltage, both water and K+ ions occupy the selectivity filter of the KcsA channel in the closed conductive state. The ion configuration is central to the mechanism of ion transport through potassium channels.


Potassium Channels , Potassium , Potassium Channels/chemistry , Potassium/chemistry , Spectrophotometry, Infrared , Isotopes , Ions/chemistry , Water/metabolism , Bacterial Proteins/chemistry , Protein Conformation
8.
Neurobiol Dis ; 185: 106244, 2023 09.
Article En | MEDLINE | ID: mdl-37524211

Lysosomes are acidic intracellular organelles with autophagic functions that are critical for protein degradation and mitochondrial homeostasis, while abnormalities in lysosomal physiological functions are closely associated with neurological disorders. Transmembrane protein 175 (TMEM175), an ion channel in the lysosomal membrane that is essential for maintaining lysosomal acidity, has been proven to coordinate with V-ATPase to modulate the luminal pH of the lysosome to assist the digestion of abnormal proteins and organelles. However, there is considerable controversy about the characteristics of TMEM175. In this review, we introduce the research progress on the structural, modulatory, and functional properties of TMEM175, followed by evidence of its relevance for neurological disorders. Finally, we discuss the potential value of TMEM175 as a therapeutic target in the hope of providing new directions for the treatment of neurodegenerative diseases.


Ion Channels , Neurodegenerative Diseases , Humans , Ion Channels/analysis , Ion Channels/metabolism , Neurodegenerative Diseases/metabolism , Lysosomes/metabolism , Autophagy , Potassium Channels/chemistry
9.
Commun Biol ; 6(1): 745, 2023 07 18.
Article En | MEDLINE | ID: mdl-37464013

The TWIK-related spinal cord K+ channel (TRESK, K2P18.1) is a K2P channel contributing to the maintenance of membrane potentials in various cells. Recently, physiological TRESK function was identified as a key player in T-cell differentiation rendering the channel a new pharmacological target for treatment of autoimmune diseases. The channel activator cloxyquin represents a promising lead compound for the development of a new class of immunomodulators. Identification of cloxyquin binding site and characterization of the molecular activation mechanism can foster the future drug development. Here, we identify the cloxyquin binding site at the M2/M4 interface by mutational scan and analyze the molecular mechanism of action by protein modeling as well as in silico and in vitro electrophysiology using different permeating ion species (K+ / Rb+). In combination with kinetic analyses of channel inactivation, our results suggest that cloxyquin allosterically stabilizes the inner selectivity filter facilitating the conduction process subsequently activating hTRESK.


Chloroquinolinols , Potassium Channels , Potassium Channels/chemistry , Binding Sites , Chloroquinolinols/chemistry , Chloroquinolinols/pharmacology , Membrane Potentials
10.
J Chem Theory Comput ; 19(9): 2574-2589, 2023 May 09.
Article En | MEDLINE | ID: mdl-37040262

Potassium channels are responsible for the selective yet efficient permeation of potassium ions across cell membranes. Despite many available high-resolution structures of potassium channels, those conformations inform only on static information on the ion permeation processes. Here, we use molecular dynamics simulations and Markov state models to obtain dynamical details of ion permeation. The permeation cycles, expressed in terms of selectivity filter occupancy and representing ion permeation events, are illustrated. We show that the direct knock-on permeation represents the dominant permeation mechanism over a wide range of potassium concentrations, temperatures, and membrane voltages for the pore of MthK. Direct knock-on is also observed in other potassium channels with a highly conserved selectivity filter, demonstrating the robustness of the permeation mechanism. Lastly, we investigate the charge strength dependence of permeation cycles. Our results shed light on the underlying permeation details, which are valuable in studying conduction mechanisms in potassium channels.


Molecular Dynamics Simulation , Potassium Channels , Potassium Channels/chemistry , Cell Membrane/metabolism , Potassium/chemistry
11.
J Phys Chem Lett ; 14(11): 2886-2890, 2023 Mar 23.
Article En | MEDLINE | ID: mdl-36924459

Silver and silver ions have a long history of antimicrobial activity and medical applications. Nevertheless, the activity of Ag+ against bacteria, how it enters a cell, has not yet been established. The K+ channel, a membrane protein, is a possible route. The addition of a channel inhibitor (4-aminopyridine) to modulate the Ag+ uptake could support this view. However, the inhibitor enhances the uptake of Ag+, the opposite result. We have applied cold ion trap infrared laser spectroscopy to complexes of Ag+ and Ac-Tyr-NHMe (a model for GYG) which is a portion of the selectivity filter in the K+ channel to consider the question of permeation. With support from quantum chemical calculations, we have determined the stable conformations of the complex. The conformations strongly suggest that Ag+ would not readily permeate the K+ channel. The mechanism of the unexpected enhancement by the inhibitor is discussed.


Potassium Channels , Silver , Potassium Channels/chemistry , Spectrophotometry, Infrared , Peptides/metabolism , Ions
12.
J Mol Biol ; 435(6): 167970, 2023 03 15.
Article En | MEDLINE | ID: mdl-36682679

Ionotropic glutamate receptors are ligand-gated cation channels that play essential roles in the excitatory synaptic transmission throughout the central nervous system. A number of open-pore structures of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic-acid (AMPA)-type glutamate receptors became available recently by cryo-electron microscopy (cryo-EM). These structures provide valuable insights into the conformation of the selectivity filter (SF), the part of the ion channel that determines the ion selectivity. Nonetheless, due to the moderate resolution of the cryo-EM structures, detailed information such as ion occupancy of monovalent and divalent cations as well as positioning of the side-chains in the SF is still missing. Here, in an attempt to obtain high-resolution information about glutamate receptor SFs, we incorporated partial SF sequences of the AMPA and kainate receptors into the bacterial tetrameric cation channel NaK, which served as a structural scaffold. We determined a series of X-ray structures of NaK-CDI, NaK-SDI and NaK-SELM mutants at 1.42-2.10 Å resolution, showing distinct ion occupation of different monovalent cations. Molecular dynamics (MD) simulations of NaK-CDI indicated the channel to be conductive to monovalent cations, which agrees well with our electrophysiology recordings. Moreover, previously unobserved structural asymmetry of the SF was revealed by the X-ray structures and MD simulations, implying its importance in ion non-selectivity of tetrameric cation channels.


Bacterial Proteins , Potassium Channels , Receptors, AMPA , Receptors, Kainic Acid , Cryoelectron Microscopy , Molecular Dynamics Simulation , Receptors, AMPA/chemistry , Receptors, AMPA/genetics , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/genetics , Protein Multimerization , Potassium Channels/chemistry , Potassium Channels/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics
13.
Annu Rev Biophys ; 52: 91-111, 2023 05 09.
Article En | MEDLINE | ID: mdl-36626766

Carefully orchestrated opening and closing of ion channels control the diffusion of ions across cell membranes, generating the electrical signals required for fast transmission of information throughout the nervous system. Inactivation is a parsimonious means for channels to restrict ion conduction without the need to remove the activating stimulus. Voltage-gated channel inactivation plays crucial physiological roles, such as controlling action potential duration and firing frequency in neurons. The ball-and-chain moniker applies to a type of inactivation proposed first for sodium channels and later shown to be a universal mechanism. Still, structural evidence for this mechanism remained elusive until recently. We review the ball-and-chain inactivation research starting from its introduction as a crucial component of sodium conductance during electrical signaling in the classical Hodgkin and Huxley studies, through the discovery of its simple intuitive mechanism in potassium channels during the molecular cloning era, to the eventual elucidation of a potassium channel structure in a ball-and-chain inactivated state.


Potassium Channels , Signal Transduction , Potassium Channels/chemistry , Cell Membrane
14.
Funct Plant Biol ; 50(1): 17-28, 2023 01.
Article En | MEDLINE | ID: mdl-36220140

Plant vacuoles are the main cellular reservoirs to store K+ . The vacuolar K+ channels play a pivotal role in K+ exchange between cytosol and vacuolar sap. Among vacuolar K+ transporters, the Two Pore Potassium Channels (TPKs) are highly selective K+ channels present in most or all plant vacuoles and could be involved in various plant stress responses and developmental processes. Although the majority of TPK members have a vacuolar specialisation, some TPKs display different membrane localisation including the plasma membrane, tonoplast of protein storage vacuoles and probably chloroplast membranes. The functional properties as well as physiological roles of TPKs remains largely unexplored. In this review, we have collected recent data about the physiology, structure, functionality and evolution of TPK/KCO3 channels. We also critically evaluate the latest findings on the biological role, physiological functions, and regulation of TPK/KCO3 channels in relation to their structure and phylogenetic position. The possible role of TPK/KCO3 channels in plant tolerance to various abiotic stresses is summarised, and the future priority directions for TPK/KCO3 studies are addressed.


Plants , Potassium Channels , Phylogeny , Plants/metabolism , Potassium Channels/genetics , Potassium Channels/chemistry , Potassium Channels/metabolism , Vacuoles/metabolism , Cell Membrane/metabolism
15.
J Biomol Struct Dyn ; 41(13): 6272-6281, 2023.
Article En | MEDLINE | ID: mdl-35916276

Discrepin is a 38-residue α-toxin extracted from the venom of the Venezuelan scorpion Tityus discrepans, which inhibits ionic transit in the voltage-dependent potassium channels (Kv) of A-type current. The effect of specific residues on the IC50 between Discrepine and Kv4.3, the main component of A-type currents, is known; however, the molecular details of the toxin-channel interaction are not known. In this work, we present interaction models between Discrepin (wt) and two peptide variants (V6K/D20K and K13A) on the pore-forming domain of the Kv4.3 channel obtained from homology, docking, and molecular dynamics modeling techniques. The free energy calculations in these models correspond to the order of the experimentally determined IC50 values. Our studies shed light on the role of the K13 residue as responsible for occluding the Kv4.3 selectivity filter and the importance of the V6K mutation in the approach and stabilization of toxin-channel complex interactions.Communicated by Ramaswamy H. Sarma.


Molecular Dynamics Simulation , Scorpion Venoms , Amino Acid Sequence , Scorpion Venoms/pharmacology , Scorpion Venoms/chemistry , Potassium Channels/chemistry , Peptides/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry
16.
Function (Oxf) ; 3(6): zqac052, 2022.
Article En | MEDLINE | ID: mdl-36325512

Ever since they were first observed in Purkinje fibers of the heart, funny channels have had close connections to potassium channels. Indeed, funny channels were initially thought to produce a potassium current in the heart called I K2. However, funny channels are completely unlike potassium channels in ways that make their contributions to the physiology of cells unique. An important difference is the greater ability for sodium to permeate funny channels. Although it does not flow through the funny channel as easily as does potassium, sodium does permeate well enough to allow for depolarization of cells following a strong hyperpolarization. This is critical for the function of funny channels in places like the heart and brain. Computational analyses using recent structures of the funny channels have provided a possible mechanism for their unusual permeation properties.


Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Potassium Channels , Potassium Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/physiology , Potassium , Sodium
17.
Molecules ; 27(19)2022 Oct 07.
Article En | MEDLINE | ID: mdl-36235195

The nanostructuration of solid matrices with lipid nanoparticles containing membrane proteins is a promising tool for the development of high-throughput screening devices. Here, sol-gel silica-derived nanocomposites loaded with liposome-reconstituted KcsA, a prokaryotic potassium channel, have been synthesized. The conformational and functional stability of these lipid nanoparticles before and after sol-gel immobilization have been characterized by using dynamic light scattering, and steady-state and time-resolved fluorescence spectroscopy methods. The lipid-reconstituted KcsA channel entrapped in the sol-gel matrix retained the conformational and stability changes induced by the presence of blocking or permeant cations in the buffer (associated with the conformation of the selectivity filter) or by a drop in the pH (associated with the opening of the activation gate of the protein). Hence, these results indicate that this novel device has the potential to be used as a screening platform to test new modulating drugs of potassium channels.


Liposomes , Nanocomposites , Bacterial Proteins/metabolism , Cations , Ion Channels/metabolism , Lipids , Nanoparticles , Potassium Channels/chemistry , Protein Conformation , Silicon Dioxide/metabolism
18.
Function (Oxf) ; 3(3): zqac019, 2022.
Article En | MEDLINE | ID: mdl-36156894

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels generate the pacemaker current which plays an important role in the timing of various biological processes like the heart beat. We used umbrella sampling to explore the potential of mean force for the conduction of potassium and sodium through the open HCN4 pore. Our data explain distinct functional features like low unitary conductance and weak selectivity as a result of high energetic barriers inside the selectivity filter of this channel. They exceed the 3-5 kJ/mol threshold which is presumed as maximal barrier for diffusion-limited conductance. Furthermore, simulations provide a thermodynamic explanation for the weak cation selectivity of HCN channels that contain only two ion binding sites in the selectivity filter (SF). We find that sodium ions bind more strongly to the SF than potassium and are easier released by binding of potassium than of another sodium. Hence ion transport and selectivity in HCN channels is not determined by the same mechanism as in potassium-selective channels; it rather relies on sodium as a weak blocker that can only be released by potassium.


Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Potassium Channels , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Potassium Channels/chemistry , Cations/metabolism , Binding Sites , Sodium/metabolism , Potassium/chemistry
19.
Nat Commun ; 13(1): 5682, 2022 09 27.
Article En | MEDLINE | ID: mdl-36167696

The voltage-gated potassium channel AKT1 is responsible for primary K+ uptake in Arabidopsis roots. AKT1 is functionally activated through phosphorylation and negatively regulated by a potassium channel α-subunit AtKC1. However, the molecular basis for the modulation mechanism remains unclear. Here we report the structures of AKT1, phosphorylated-AKT1, a constitutively-active variant, and AKT1-AtKC1 complex. AKT1 is assembled in 2-fold symmetry at the cytoplasmic domain. Such organization appears to sterically hinder the reorientation of C-linkers during ion permeation. Phosphorylated-AKT1 adopts an alternate 4-fold symmetric conformation at cytoplasmic domain, which indicates conformational changes associated with symmetry switch during channel activation. To corroborate this finding, we perform structure-guided mutagenesis to disrupt the dimeric interface and identify a constitutively-active variant Asp379Ala mediates K+ permeation independently of phosphorylation. This variant predominantly adopts a 4-fold symmetric conformation. Furthermore, the AKT1-AtKC1 complex assembles in 2-fold symmetry. Together, our work reveals structural insight into the regulatory mechanism for AKT1.


Arabidopsis Proteins , Arabidopsis , Potassium Channels, Voltage-Gated , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Potassium/metabolism , Potassium Channels/chemistry , Potassium Channels, Voltage-Gated/chemistry
20.
Virus Res ; 320: 198903, 2022 Oct 15.
Article En | MEDLINE | ID: mdl-36037849

Ion channels are membrane proteins essential for a plethora of cellular functions including maintaining cell shape, ion homeostasis, cardiac rhythm and action potential in neurons. The complexity and often extensive structure of eukaryotic membrane proteins makes it difficult to understand their basic biological regulation. Therefore, this article suggests, viroporins - the miniature versions of eukaryotic protein homologs from viruses - might serve as model systems to provide insights into behaviour of eukaryotic ion channels in general. The structural requirements for correct assembly of the channel along with the basic functional properties of a K+ channel exist in the minimal design of the viral K+ channels from two viruses, Chlorella virus (Kcv) and Ectocarpus siliculosus virus (Kesv). These small viral proteins readily assemble into tetramers and they sort in cells to distinct target membranes. When these viruses-encoded channels are expressed into the mammalian cells, they utilise their protein machinery and hence can serve as excellent tools to study the cells protein sorting machinery. This combination of small size and robust function makes viral K+ channels a valuable model system for detection of basic structure-function correlations. It is believed that molecular and physiochemical analyses of these viroporins may serve as basis for the development of inhibitors or modulators to ion channel activity for targeting ion channel diseases - so called channelopathies. Therefore, it may provide a potential different scope for molecular pharmacology studies aiming at novel and innovative therapeutics associated with channel related diseases. This article reviews the structural and functional properties of Kcv and Kesv upon expression in mammalian cells and Xenopus oocytes. The mechanisms behind differential protein sorting in Kcv and Kesv are also thoroughly discussed.


Chlorella , Phycodnaviridae , Viruses , Animals , Chlorella/metabolism , Eukaryota , Ion Channels/metabolism , Mammals/metabolism , Phycodnaviridae/genetics , Phycodnaviridae/metabolism , Potassium/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels/metabolism , Viroporin Proteins , Viruses/metabolism
...