Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 179(7): 1582-1589.e7, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31787376

ABSTRACT

The hyperpolarization-activated cyclic nucleotide-gated (HCN) channel is a voltage-gated cation channel that mediates neuronal and cardiac pacemaker activity. The HCN channel exhibits reversed voltage dependence, meaning it closes with depolarization and opens with hyperpolarization. Different from Na+, Ca2+, and Kv1-Kv7 channels, the HCN channel does not have domain-swapped voltage sensors. We introduced a reversible, metal-mediated cross bridge into the voltage sensors to create the chemical equivalent of a hyperpolarized conformation and determined the structure using cryoelectron microscopy (cryo-EM). Unlike the depolarized HCN channel, the S4 helix is displaced toward the cytoplasm by two helical turns. Near the cytoplasm, the S4 helix breaks into two helices, one running parallel to the membrane surface, analogous to the S4-S5 linker of domain-swapped voltage-gated channels. These findings suggest a basis for allosteric communication between voltage sensors and the gate in this kind of channel. They also imply that voltage sensor movements are not the same in all voltage-gated channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Ion Channel Gating , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Membrane Potentials , Protein Conformation, alpha-Helical , Sf9 Cells , Spodoptera
2.
Cell ; 169(6): 1042-1050.e9, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575668

ABSTRACT

KCNQ1 is the pore-forming subunit of cardiac slow-delayed rectifier potassium (IKs) channels. Mutations in the kcnq1 gene are the leading cause of congenital long QT syndrome (LQTS). Here, we present the cryoelectron microscopy (cryo-EM) structure of a KCNQ1/calmodulin (CaM) complex. The conformation corresponds to an "uncoupled," PIP2-free state of KCNQ1, with activated voltage sensors and a closed pore. Unique structural features within the S4-S5 linker permit uncoupling of the voltage sensor from the pore in the absence of PIP2. CaM contacts the KCNQ1 voltage sensor through a specific interface involving a residue on CaM that is mutated in a form of inherited LQTS. Using an electrophysiological assay, we find that this mutation on CaM shifts the KCNQ1 voltage-activation curve. This study describes one physiological form of KCNQ1, depolarized voltage sensors with a closed pore in the absence of PIP2, and reveals a regulatory interaction between CaM and KCNQ1 that may explain CaM-mediated LQTS.


Subject(s)
Calmodulin/chemistry , KCNQ1 Potassium Channel/chemistry , Long QT Syndrome/metabolism , Amino Acid Sequence , Animals , Calmodulin/metabolism , Cryoelectron Microscopy , Humans , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Models, Molecular , Mutation , Sequence Alignment , Xenopus laevis
3.
Biopolymers ; 114(7): e23540, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37254962

ABSTRACT

In the CryoEM-structure of the hSkMNaV1.4 ion channel (PDB:6AGF), the 59-residue DIS5-S6 linker peptide was omitted due to absence of electron density. This peptide is intriguing - comprised of unique sequence and found only in mammalian skeletal muscle sodium ion channels. To probe potential physiological and evolutionary significance, we constructed an homology model of the complete hSkMNaV1.4 channel. Rather than a flexible random coil potentiating drift across the channel, the linker folds into a compact configuration through self-assembling secondary structural elements. Analogous sequences from 48 mammalian organisms show hypervariability with between 40% and 100% sequence similarity. To investigate structural implications, sequences from 14 representative organisms were additionally modelled. All showed highly conserved N-and C-terminal residues closely superimposed, suggesting a critical functional role. An optimally located asparagine residue within the conserved region was investigated for N-linked glycosylation and MD simulations carried out. Results suggest a complex glycan added at this site in the linker may form electrostatic interactions with the DIV voltage sensing domain and be mechanistically involved in channel gating. The relationship of unique sequence, compact configuration, potential glycosylation and MD simulations are discussed relative to SkMNaV1.4 structure and function.


Subject(s)
Molecular Dynamics Simulation , Sodium Channels , Animals , Glycosylation , Sodium Channels/chemistry , Sodium Channels/metabolism , Mammals/metabolism
4.
Int J Mol Sci ; 24(11)2023 May 25.
Article in English | MEDLINE | ID: mdl-37298178

ABSTRACT

Ion channels play important roles in fundamental biological processes, such as electric signaling in cells, muscle contraction, hormone secretion, and regulation of the immune response. Targeting ion channels with drugs represents a treatment option for neurological and cardiovascular diseases, muscular degradation disorders, and pathologies related to disturbed pain sensation. While there are more than 300 different ion channels in the human organism, drugs have been developed only for some of them and currently available drugs lack selectivity. Computational approaches are an indispensable tool for drug discovery and can speed up, especially, the early development stages of lead identification and optimization. The number of molecular structures of ion channels has considerably increased over the last ten years, providing new opportunities for structure-based drug development. This review summarizes important knowledge about ion channel classification, structure, mechanisms, and pathology with the main focus on recent developments in the field of computer-aided, structure-based drug design on ion channels. We highlight studies that link structural data with modeling and chemoinformatic approaches for the identification and characterization of new molecules targeting ion channels. These approaches hold great potential to advance research on ion channel drugs in the future.


Subject(s)
Ion Channels , Muscular Diseases , Humans , Ion Channels/metabolism , Drug Design , Drug Discovery , Molecular Structure , Computers , Computer-Aided Design
5.
Pflugers Arch ; 473(9): 1423-1435, 2021 09.
Article in English | MEDLINE | ID: mdl-34357442

ABSTRACT

Cyclic nucleotide-gated (CNG) channels are key to the signal transduction machinery of certain sensory modalities both in vertebrate and invertebrate organisms. They translate a chemical change in cyclic nucleotide concentration into an electrical signal that can spread through sensory cells. Despite CNG and voltage-gated potassium channels sharing a remarkable amino acid sequence homology and basic architectural plan, their functional properties are dramatically different. While voltage-gated potassium channels are highly selective and require membrane depolarization to open, CNG channels have low ion selectivity and are not very sensitive to voltage. In the last few years, many high-resolution structures of intact CNG channels have been released. This wealth of new structural information has provided enormous progress toward the understanding of the molecular mechanisms and driving forces underpinning CNG channel activation. In this review, we report on the current understanding and controversies surrounding the gating mechanism in CNG channels, as well as the deep intertwining existing between gating, the ion permeation process, and its modulation by membrane voltage. While the existence of this powerful coupling was recognized many decades ago, its direct structural demonstration, and ties to the CNG channel inherent pore flexibility, is a recent achievement.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/physiology , Ion Channel Gating/physiology , Animals , Humans , Protein Conformation , Protein Structure, Secondary , Retinal Rod Photoreceptor Cells/chemistry , Retinal Rod Photoreceptor Cells/physiology , Signal Transduction/physiology
6.
Int J Mol Sci ; 22(2)2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33430308

ABSTRACT

Cell survival and normal cell function require a highly coordinated and precise regulation of basal cytosolic Ca2+ concentrations. The primary source of Ca2+ entry into the cell is mediated by the Ca2+ release-activated Ca2+ (CRAC) channel. Its action is stimulated in response to internal Ca2+ store depletion. The fundamental constituents of CRAC channels are the Ca2+ sensor, stromal interaction molecule 1 (STIM1) anchored in the endoplasmic reticulum, and a highly Ca2+-selective pore-forming subunit Orai1 in the plasma membrane. The precise nature of the Orai1 pore opening is currently a topic of intensive research. This review describes how Orai1 gating checkpoints in the middle and cytosolic extended transmembrane regions act together in a concerted manner to ensure an opening-permissive Orai1 channel conformation. In this context, we highlight the effects of the currently known multitude of Orai1 mutations, which led to the identification of a series of gating checkpoints and the determination of their role in diverse steps of the Orai1 activation cascade. The synergistic action of these gating checkpoints maintains an intact pore geometry, settles STIM1 coupling, and governs pore opening. We describe the current knowledge on Orai1 channel gating mechanisms and summarize still open questions of the STIM1-Orai1 machinery.


Subject(s)
Calcium Signaling/genetics , Calcium/metabolism , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , Calcium Release Activated Calcium Channels/metabolism , Cell Survival/genetics , Cytosol/metabolism , Endoplasmic Reticulum/genetics , Humans
7.
J Cell Sci ; 132(4)2018 12 14.
Article in English | MEDLINE | ID: mdl-30552138

ABSTRACT

Inositol 1,4,5-trisphosphate receptors (IP3Rs) are widely expressed intracellular channels that release Ca2+ from the endoplasmic reticulum (ER). We review how studies of IP3Rs removed from their intracellular environment ('ex cellula'), alongside similar analyses of ryanodine receptors, have contributed to understanding IP3R behaviour. Analyses of permeabilized cells have demonstrated that the ER is the major intracellular Ca2+ store, and that IP3 stimulates Ca2+ release from this store. Radioligand binding confirmed that the 4,5-phosphates of IP3 are essential for activating IP3Rs, and facilitated IP3R purification and cloning, which paved the way for structural analyses. Reconstitution of IP3Rs into lipid bilayers and patch-clamp recording from the nuclear envelope have established that IP3Rs have a large conductance and select weakly between Ca2+ and other cations. Structural analyses are now revealing how IP3 binding to the N-terminus of the tetrameric IP3R opens the pore ∼7 nm away from the IP3-binding core (IBC). Communication between the IBC and pore passes through a nexus of interleaved domains contributed by structures associated with the pore and cytosolic domains, which together contribute to a Ca2+-binding site. These structural analyses provide evidence to support the suggestion that IP3 gates IP3Rs by first stimulating Ca2+ binding, which leads to pore opening and Ca2+ release.


Subject(s)
Calcium Signaling/physiology , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Animals , Calcium/metabolism , Humans , Ryanodine Receptor Calcium Release Channel/metabolism
8.
Neurochem Res ; 42(9): 2495-2504, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28589518

ABSTRACT

Voltage-gated sodium channels initiate action potentials in brain neurons. In the 1970s, much was known about the function of sodium channels from measurements of ionic currents using the voltage clamp method, but there was no information about the sodium channel molecules themselves. As a postdoctoral fellow and staff scientist at the National Institutes of Health, I developed neurotoxins as molecular probes of sodium channels in cultured neuroblastoma cells. During those years, Bruce Ransom and I crossed paths as members of the laboratories of Marshall Nirenberg and Philip Nelson and shared insights about sodium channels in neuroblastoma cells from my work and electrical excitability and synaptic transmission in cultured spinal cord neurons from Bruce's pioneering electrophysiological studies. When I established my laboratory at the University of Washington in 1977, my colleagues and I used those neurotoxins to identify the protein subunits of sodium channels, purify them, and reconstitute their ion conductance activity in pure form. Subsequent studies identified the molecular basis for the main functions of sodium channels-voltage-dependent activation, rapid and selective ion conductance, and fast inactivation. Bruce Ransom and I re-connected in the 1990s, as ski buddies at the Winter Conference on Brain Research and as faculty colleagues at the University of Washington when Bruce became our founding Chair of Neurology and provided visionary leadership of that department. In the past decade my work on sodium channels has evolved into structural biology. Molecular modeling and X-ray crystallographic studies have given new views of sodium channel function at atomic resolution. Sodium channels are also the molecular targets for genetic diseases, including Dravet Syndrome, an intractable pediatric epilepsy disorder with major co-morbidities of cognitive deficit, autistic-like behaviors, and premature death that is caused by loss-of-function mutations in the brain sodium channel NaV1.1. Our work on a mouse genetic model of this disease has shown that its multi-faceted pathophysiology and co-morbidities derive from selective loss of electrical excitability and action potential firing in GABAergic inhibitory neurons, which disinhibits neural circuits throughout the brain and leads directly to the epilepsy, premature death and complex co-morbidities of this disease. It has been rewarding for me to use our developing knowledge of sodium channels to help understand the pathophysiology and to suggest potential therapeutic approaches for this devastating childhood disease.


Subject(s)
Epilepsy/metabolism , Sodium Channels/chemistry , Sodium Channels/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Neurotoxins/metabolism , Neurotoxins/pharmacology , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/physiology
9.
J Biol Chem ; 288(25): 17968-75, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23667254

ABSTRACT

Oxidation is an important biochemical defense mechanism, but it also elicits toxicity; therefore, oxidation must be under strict control. In phagocytotic events in neutrophils, the voltage-gated H(+) (Hv) channel is a key regulator of the production of reactive oxygen species against invading bacteria. The cytoplasmic domain of the Hv channel forms a dimeric coiled coil underpinning a dimerized functional unit. Importantly, in the alignment of the coiled-coil core, a conserved cysteine residue forms a potential intersubunit disulfide bond. In this study, we solved the crystal structures of the coiled-coil domain in reduced, oxidized, and mutated (Cys → Ser) states. The crystal structures indicate that a pair of Cys residues forms an intersubunit disulfide bond dependent on the redox conditions. CD spectroscopy revealed that the disulfide bond increases the thermal stability of the coiled-coil protein. We also reveal that two thiol modifier molecules are able to bind to Cys in a redox-dependent manner without disruption of the dimeric coiled-coil assembly. Thus, the biochemical properties of the cytoplasmic coiled-coil domain in the Hv channel depend on the redox condition, which may play a role in redox sensing in the phagosome.


Subject(s)
Ion Channels/chemistry , Ion Channels/physiology , Protein Structure, Quaternary , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Blotting, Western , Circular Dichroism , Crystallography, X-Ray , Disulfides/chemistry , Disulfides/metabolism , HEK293 Cells , Humans , Ion Channels/genetics , Macrophages/metabolism , Membrane Potentials , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Oxidation-Reduction , Patch-Clamp Techniques , Phagosomes/metabolism , Protein Multimerization , Sequence Homology, Amino Acid , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism
10.
Cell Calcium ; 123: 102927, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38959762

ABSTRACT

In order to understand protein function, the field of structural biology makes extensive use of cryogenic electron microscopy (cryo-EM), a technique that enables structure determination at atomic resolution following embedding of protein particles in vitreous ice. Considering the profound effects of temperature on macromolecule function, an important-but often neglected-question is how the frozen particles relate to the actual protein conformations at physiological temperatures. In a recent study, Hu et al. compare structures of the cation channel TRPM4 "frozen" at 4 °C versus 37 °C, revealing how temperature critically affects the binding of activating Ca2+ ions and other channel modulators.


Subject(s)
Cryoelectron Microscopy , TRPM Cation Channels , Animals , Humans , Calcium/metabolism , Cryoelectron Microscopy/methods , Temperature , TRPM Cation Channels/metabolism , TRPM Cation Channels/chemistry
11.
Neuron ; 109(18): 2902-2913.e4, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34390650

ABSTRACT

TRAAK is a mechanosensitive two-pore domain K+ (K2P) channel localized to nodes of Ranvier in myelinated neurons. TRAAK deletion in mice results in mechanical and thermal allodynia, and gain-of-function mutations cause the human neurodevelopmental disorder FHEIG. TRAAK displays basal and stimulus-gated activities typical of K2Ps, but the mechanistic and structural differences between these modes are unknown. Here, we demonstrate that basal and mechanically gated openings are distinguished by their conductance, kinetics, and structure. Basal openings are low conductance, short duration, and due to a conductive channel conformation with the interior cavity exposed to the surrounding membrane. Mechanically gated openings are high conductance, long duration, and due to a channel conformation in which the interior cavity is sealed to the surrounding membrane. Our results explain how dual modes of activity are produced by a single ion channel and provide a basis for the development of state-selective pharmacology with the potential to treat disease.


Subject(s)
Ion Channel Gating/physiology , Mechanotransduction, Cellular/physiology , Neurons/physiology , Potassium Channels/chemistry , Potassium Channels/physiology , Animals , Female , Humans , Physical Stimulation/methods , Protein Structure, Secondary , Protein Structure, Tertiary , Saccharomycetales , Xenopus laevis
12.
Methods Enzymol ; 653: 189-206, 2021.
Article in English | MEDLINE | ID: mdl-34099171

ABSTRACT

Nicotinic acetylcholine receptors are members of the Cys-loop superfamily of pentameric ligand-gated ion channels. The electric organ of the Torpedo ray is extraordinarily rich in an acetylcholine receptor that is homologous to the human nicotinic receptor found at the neuromuscular junction. Due to this abundant natural source in the fish and the relatively accessible preparation of the neuromuscular junction (compared to a central synapse), this muscle-type receptor and specifically the fish receptors have long been used as the prototype for study of nicotinic receptors. However, an absence of structural detail at high resolution has limited the chemical interpretation of this archetypal nicotinic receptor. One of the main concerns in preparing receptor for high resolution structural analysis was its documented sensitivity to particular detergents and requirements for specific lipids in order to maintain function after reconstitution in a membrane. Here, we present methods for purifying native nicotinic receptor from Torpedo electric tissue that maintains functionality after reconstitution and that is amenable to high resolution structural analysis. The specific developments we describe include detergent exchange during purification, inclusion of specific lipids during purification and for nanodisc reconstitution, and synthesis of a new affinity reagent for rapid isolation of receptors.


Subject(s)
Ligand-Gated Ion Channels , Receptors, Nicotinic , Torpedo , Animals , Fish Proteins/isolation & purification , Ligand-Gated Ion Channels/isolation & purification , Receptors, Nicotinic/isolation & purification
13.
Neuron ; 106(6): 952-962.e5, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32275860

ABSTRACT

The nicotinic acetylcholine receptor, a pentameric ligand-gated ion channel, converts the free energy of binding of the neurotransmitter acetylcholine into opening of its central pore. Here we present the first high-resolution structure of the receptor type found in muscle-endplate membrane and in the muscle-derived electric tissues of fish. The native receptor was purified from Torpedo electric tissue and functionally reconstituted in lipids optimal for cryo-electron microscopy. The receptor was stabilized in a closed state by the binding of α-bungarotoxin. The structure reveals the binding of a toxin molecule at each of two subunit interfaces in a manner that would block the binding of acetylcholine. It also reveals a closed gate in the ion-conducting pore, formed by hydrophobic amino acid side chains, located ∼60 Å from the toxin binding sites. The structure provides a framework for understanding gating in ligand-gated channels and how mutations in the acetylcholine receptor cause congenital myasthenic syndromes.


Subject(s)
Bungarotoxins/metabolism , Electric Organ/metabolism , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/ultrastructure , Animals , Binding Sites , Bungarotoxins/pharmacology , Carbachol/pharmacology , Cryoelectron Microscopy , Molecular Conformation , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Protein Conformation , Receptors, Nicotinic/drug effects , Torpedo
14.
Toxins (Basel) ; 11(9)2019 09 19.
Article in English | MEDLINE | ID: mdl-31546848

ABSTRACT

Venoms are comprised of diverse mixtures of proteins, peptides, and small molecules. Identifying individual venom components and their target(s) with mechanism of action is now attainable to understand comprehensively the effectiveness of venom cocktails and how they collectively function in the defense and predation of an organism. Here, structure-based computational methods were used with bioinformatics tools to screen and identify potential biological targets of tertiapin (TPN), a venom peptide from Apis mellifera (European honey bee). The small hive beetle (Aethina tumida (A. tumida)) is a natural predator of the honey bee colony and was found to possess multiple inwardly rectifying K+ (Kir) channel subunit genes from a genomic BLAST search analysis. Structure-based virtual screening of homology modelled A. tumida Kir (atKir) channels found TPN to interact with a docking profile and interface "footprint" equivalent to known TPN-sensitive mammalian Kir channels. The results support the hypothesis that atKir channels, and perhaps other insect Kir channels, are natural biological targets of TPN that help defend the bee colony from infestations by blocking K+ transport via atKir channels. From these in silico findings, this hypothesis can now be subsequently tested in vitro by validating atKir channel block as well as in vivo TPN toxicity towards A. tumida. This study highlights the utility and potential benefits of screening in virtual space for venom peptide interactions and their biological targets, which otherwise would not be feasible.


Subject(s)
Bee Venoms/pharmacology , Insect Proteins/physiology , Peptides/pharmacology , Potassium Channels, Inwardly Rectifying/physiology , Animals , Bee Venoms/chemistry , Coleoptera , Female , Molecular Dynamics Simulation , Oocytes/physiology , Peptides/chemistry , Structure-Activity Relationship , Xenopus laevis
15.
Cell Calcium ; 76: 132-134, 2018 12.
Article in English | MEDLINE | ID: mdl-30420160

ABSTRACT

Transient receptor potential cation channel, subtype melastatin 2 (TRPM2), is important for several physiological functions, such as immune response or temperature regulation. Recently, the structure of full-length TRPM2 from zebrafish was published (Huang et al., 2018) proposing a new activation mechanism - is it really a paradigm shift or just reflects evolution of the channel?.


Subject(s)
Adenosine Diphosphate Ribose , TRPM Cation Channels , Animals , Calcium , Calcium Signaling , Ion Channel Gating
16.
Elife ; 72018 08 10.
Article in English | MEDLINE | ID: mdl-30095067

ABSTRACT

SWELL1 (LRRC8A) is the only essential subunit of the Volume Regulated Anion Channel (VRAC), which regulates cellular volume homeostasis and is activated by hypotonic solutions. SWELL1, together with four other LRRC8 family members, potentially forms a vastly heterogeneous cohort of VRAC channels with different properties; however, SWELL1 alone is also functional. Here, we report a high-resolution cryo-electron microscopy structure of full-length human homo-hexameric SWELL1. The structure reveals a trimer of dimers assembly with symmetry mismatch between the pore-forming domain and the cytosolic leucine-rich repeat (LRR) domains. Importantly, mutational analysis demonstrates that a charged residue at the narrowest constriction of the homomeric channel is an important pore determinant of heteromeric VRAC. Additionally, a mutation in the flexible N-terminal portion of SWELL1 affects pore properties, suggesting a putative link between intracellular structures and channel regulation. This structure provides a scaffold for further dissecting the heterogeneity and mechanism of activation of VRAC.


Subject(s)
Membrane Proteins/chemistry , Protein Multimerization/genetics , Structure-Activity Relationship , Voltage-Dependent Anion Channels/chemistry , Amino Acids/chemistry , Amino Acids/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Multigene Family , Mutation , Protein Structure, Quaternary , Voltage-Dependent Anion Channels/genetics
17.
Neuron ; 99(5): 956-968.e4, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30122377

ABSTRACT

AMPA receptors mediate fast excitatory neurotransmission and are critical for CNS development and function. Calcium-permeable subsets of AMPA receptors are strongly implicated in acute and chronic neurological disorders. However, despite the clinical importance, the therapeutic landscape for specifically targeting them, and not the calcium-impermeable AMPA receptors, remains largely undeveloped. To address this problem, we used cryo-electron microscopy and electrophysiology to investigate the mechanisms by which small-molecule blockers selectively inhibit ion channel conductance in calcium-permeable AMPA receptors. We determined the structures of calcium-permeable GluA2 AMPA receptor complexes with the auxiliary subunit stargazin bound to channel blockers, including the orb weaver spider toxin AgTx-636, the spider toxin analog NASPM, and the adamantane derivative IEM-1460. Our structures provide insights into the architecture of the blocker binding site and the mechanism of trapping, which are critical for development of small molecules that specifically target calcium-permeable AMPA receptors.


Subject(s)
Calcium/metabolism , Excitatory Amino Acid Antagonists/chemistry , Excitatory Amino Acid Antagonists/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Animals , Excitatory Amino Acid Antagonists/pharmacology , HEK293 Cells , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Receptors, AMPA/antagonists & inhibitors , Spider Venoms/chemistry , Spider Venoms/metabolism , Spider Venoms/pharmacology
18.
Neuropharmacology ; 127: 161-172, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28716449

ABSTRACT

Inwardly rectifying K+ (Kir) channels play a significant role in vertebrate and invertebrate biology by regulating the movement of K+ ions involved in membrane transport and excitability. Yet unlike other ion channels including their ancestral K+-selective homologs, there are very few venom toxins known to target and inhibit Kir channels with the potency and selectivity found for the Ca2+-activated and voltage-gated K+ channel families. It is unclear whether this is simply due to a lack of discovery, or instead a consequence of the evolutionary processes that drive the development of venom components towards their targets based on a collective efficacy to 1) elicit pain for defensive purposes, 2) promote paralysis for prey capture, or 3) facilitate delivery of venom components into the circulation. The past two decades of venom screening has yielded three venom peptides with inhibitory activity towards mammalian Kir channels, including the discovery of tertiapin, a high-affinity pore blocker from the venom of the European honey bee Apis mellifera. Venomics and structure-based computational approaches represent exciting new frontiers for venom peptide development, where re-engineering peptide 'scaffolds' such as tertiapin may aid in the quest to expand the palette of potent and selective Kir channel blockers for future research and potentially new therapeutics. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'


Subject(s)
Peptides , Potassium Channel Blockers , Potassium Channels, Inwardly Rectifying/metabolism , Venoms , Animals , History, 20th Century , History, 21st Century , Humans , Models, Molecular , Peptides/chemistry , Peptides/history , Peptides/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/history , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/history , Venoms/chemistry , Venoms/history , Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL