Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 436
Filter
1.
Methods Mol Biol ; 2796: 157-184, 2024.
Article in English | MEDLINE | ID: mdl-38856901

ABSTRACT

Kir channels are potassium (K+) channels responsible for the mechanism of inward rectification, which plays a fundamental role in maintaining the resting membrane potential. There are seven Kir subfamilies, and their opening and closing mechanism is regulated by different regulatory factors. Genetically inherited defects in Kir channels are responsible for several rare human diseases, and for most of them, there are currently no effective therapeutic treatments. High-resolution structural information is not available for several members within the Kir subfamilies. Recently, our group achieved a significant breakthrough by utilizing cryo-EM single-particle analysis to elucidate the first structure of the human Kir2.1 channel. We present here the data processing protocol of the cryo-EM data of the human Kir2.1 channel, which is applicable to the structural determination of other ion channels by cryo-EM single-particle analysis. We also introduce a protocol designed to assess the structural heterogeneity within the cryo-EM data, allowing for the identification of other possible protein structure conformations present in the collected data. Moreover, we present a protocol for conducting all-atom molecular dynamics (MD) simulations for K+ channels, which can be incorporated into various membrane models to simulate different environments. We also propose some methods for analyzing the MD simulations, with a particular emphasis on assessing the local mobility of protein residues.


Subject(s)
Cryoelectron Microscopy , Molecular Dynamics Simulation , Potassium Channels, Inwardly Rectifying , Cryoelectron Microscopy/methods , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Humans , Protein Conformation
2.
Molecules ; 29(8)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38675722

ABSTRACT

Diabetes mellitus (DM) represents a problem for the healthcare system worldwide. DM has very serious complications such as blindness, kidney failure, and cardiovascular disease. In addition to the very bad socioeconomic impacts, it influences patients and their families and communities. The global costs of DM and its complications are huge and expected to rise by the year 2030. DM is caused by genetic and environmental risk factors. Genetic testing will aid in early diagnosis and identification of susceptible individuals or populations using ATP-sensitive potassium (KATP) channels present in different tissues such as the pancreas, myocardium, myocytes, and nervous tissues. The channels respond to different concentrations of blood sugar, stimulation by hormones, or ischemic conditions. In pancreatic cells, they regulate the secretion of insulin and glucagon. Mutations in the KCNJ11 gene that encodes the Kir6.2 protein (a major constituent of KATP channels) were reported to be associated with Type 2 DM, neonatal diabetes mellitus (NDM), and maturity-onset diabetes of the young (MODY). Kir6.2 harbors binding sites for ATP and phosphatidylinositol 4,5-diphosphate (PIP2). The ATP inhibits the KATP channel, while the (PIP2) activates it. A Kir6.2 mutation at tyrosine330 (Y330) was demonstrated to reduce ATP inhibition and predisposes to NDM. In this study, we examined the effect of mutations on the Kir6.2 structure using bioinformatics tools and molecular dynamic simulations (SIFT, PolyPhen, SNAP2, PANTHER, PhD&SNP, SNP&Go, I-Mutant, MuPro, MutPred, ConSurf, HOPE, and GROMACS). Our results indicated that M199R, R201H, R206H, and Y330H mutations influence Kir6.2 structure and function and therefore may cause DM. We conclude that MD simulations are useful techniques to predict the effects of mutations on protein structure. In addition, the M199R, R201H, R206H, and Y330H variant in the Kir6.2 protein may be associated with DM. These results require further verification in protein-protein interactions, Kir6.2 function, and case-control studies.


Subject(s)
Diabetes Mellitus , Molecular Dynamics Simulation , Potassium Channels, Inwardly Rectifying , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Humans , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Mutation , Genetic Predisposition to Disease , Binding Sites , Protein Binding
3.
J Struct Biol ; 216(2): 108091, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641256

ABSTRACT

Cholesterol is a negative regulator of a variety of ion channels. We have previously shown that cholesterol suppresses Kir2.2 channels via residue-residue uncoupling on the inter-subunit interfaces within the close state of the channels (3JYC). In this study, we extend this analysis to the other known structure of Kir2.2 that is closer to the open state of Kir2.2 channels (3SPI) and provide additional analysis of the residue distances between the uncoupled residues and cholesterol binding domains in the two conformation states of the channels. We found that the general phenomenon of cholesterol binding leading to uncoupling between specific residues is conserved in both channel states but the specific pattern of the uncoupling residues is distinct between the two states and implies different mechanisms. Specifically, we found that cholesterol binding in the 3SPI state results in an uncoupling of residues in three distinct regions; the transmembrane domain, membrane-cytosolic interface, and the cytosolic domain, with the first two regions forming an envelope around PI(4,5)P2 and cholesterol binding sites and the distal region overlapping with the subunit-subunit interface characterized in our previous study of the disengaged state. We also found that this uncoupling is dependent upon the number of cholesterol molecules bound to the channel. We further generated a mutant channel Kir2.2P187V with a single point mutation in a residue proximal to the PI(4,5)P2 binding site, which is predicted to be uncoupled from other residues in its vicinity upon cholesterol binding and found that this mutation abrogates the sensitivity of Kir2.2 to cholesterol changes in the membrane. These findings suggest that cholesterol binding to this conformation state of Kir2.2 channels may destabilize the PI(4,5)P2 interactions with the channels while in the disengaged state the destabilization occurs where the subunits interact. These findings give insight into the structural mechanistic basis for the functional effects of cholesterol binding to the Kir2.2 channel.


Subject(s)
Cholesterol , Potassium Channels, Inwardly Rectifying , Protein Binding , Cholesterol/metabolism , Cholesterol/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Binding Sites , Animals , Humans , Protein Conformation
4.
J Am Chem Soc ; 146(7): 4421-4432, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38334076

ABSTRACT

Lipids adhere to membrane proteins to stimulate or suppress molecular and ionic transport and signal transduction. Yet, the molecular details of lipid-protein interaction and their functional impact are poorly characterized. Here we combine NMR, coarse-grained molecular dynamics (CGMD), and functional assays to reveal classic cooperativity in the binding and subsequent activation of a bacterial inward rectifier potassium (Kir) channel by phosphatidylglycerol (PG), a common component of many membranes. Past studies of lipid activation of Kir channels focused primarily on phosphatidylinositol bisphosphate, a relatively rare signaling lipid that is tightly regulated in space and time. We use solid-state NMR to quantify the binding of unmodified 13C-PG to the K+ channel KirBac1.1 in liposomes. This specific lipid-protein interaction has a dissociation constant (Kd) of ∼7 mol percentage PG (ΧPG) with positive cooperativity (n = 3.8) and approaches saturation near 20% ΧPG. Liposomal flux assays show that K+ flux also increases with PG in a cooperative manner with an EC50 of ∼20% ΧPG, within the physiological range. Further quantitative fitting of these data reveals that PG acts as a partial (80%) agonist with fivefold K+ flux amplification. Comparisons of NMR chemical shift perturbation and CGMD simulations at different ΧPG confirm the direct interaction of PG with key residues, several of which would not be accessible to lipid headgroups in the closed state of the channel. Allosteric regulation by a common lipid is directly relevant to the activation mechanisms of several human ion channels. This study highlights the role of concentration-dependent lipid-protein interactions and tightly controlled protein allostery in the activation and regulation of ion channels.


Subject(s)
Potassium Channels, Inwardly Rectifying , Humans , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Liposomes , Membrane Proteins/metabolism , Lipids , Magnetic Resonance Spectroscopy
5.
Bioessays ; 46(3): e2300151, 2024 03.
Article in English | MEDLINE | ID: mdl-38227376

ABSTRACT

SUR2, similar to SUR1, is a regulatory subunit of the ATP-sensitive potassium channel (KATP), which plays a key role in numerous important physiological processes and is implicated in various diseases. Recent structural studies have revealed that, like SUR1, SUR2 can undergo ligand-dependent dynamic conformational changes, transitioning between an inhibitory inward-facing conformation and an activating occluded conformation. In addition, SUR2 possesses a unique inhibitory Regulatory helix (R helix) that is absent in SUR1. The binding of the activating Mg-ADP to NBD2 of SUR2 competes with the inhibitory Mg-ATP, thereby promoting the release of the R helix and initiating the activation process. Moreover, the signal generated by Mg-ADP binding to NBD2 might be directly transmitted to the TMD of SUR2, prior to NBD dimerization. Furthermore, the C-terminal 42 residues (C42) of SUR2 might allosterically regulate the kinetics of Mg-nucleotide binding on NBD2. These distinctive properties render SUR2 intricate sensors for intracellular Mg-nucleotides.


Subject(s)
Nucleotides , Potassium Channels, Inwardly Rectifying , Nucleotides/metabolism , Sulfonylurea Receptors/chemistry , Sulfonylurea Receptors/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/metabolism
6.
Structure ; 32(2): 168-176.e2, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38101402

ABSTRACT

ATP-sensitive potassium channels (KATP) are inhibited by ATP but activated by Mg-ADP, coupling the intracellular ATP/ADP ratio to the potassium conductance of the plasma membrane. Although there has been progress in determining the structure of KATP, the functional significance of the domain-domain interface in the gating properties of KATP channels remains incompletely understood. In this study, we define the structure of KATP as two modules: KATPcore and SURABC. Based on this model, we identified two functionally important interfaces between these two modules, namely interface I and interface II. Further structure-guided mutagenesis experiments indicate that destabilizing interface II by deleting ECL3 on the SUR1 subunit impairs KNtp-independent Mg-ADP activation, demonstrating the essential role of intramolecular interactions between KATPcore and SURABC in Mg-ADP activation. Additionally, interface II is functionally conserved between SUR1 and SUR2, and the hydrophobic residue F351 on ECL3 of SUR1 is crucial for maintaining the stability of this interface.


Subject(s)
KATP Channels , Potassium Channels, Inwardly Rectifying , KATP Channels/genetics , KATP Channels/metabolism , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/metabolism , Cell Membrane/metabolism
7.
Mol Pharmacol ; 102(5): 234-239, 2022 11.
Article in English | MEDLINE | ID: mdl-36253099

ABSTRACT

ATP-sensitive potassium channels (KATP) are energy sensors that participate in a range of physiologic processes. These channels are also clinically validated drug targets. For decades, KATP inhibitors have been prescribed for diabetes and KATP activators have been used for the treatment of hypoglycemia, hypertension, and hair loss. In this Emerging Concepts article, we highlight our current knowledge about the drug binding modes observed using cryogenic electron microscopy techniques. The inhibitors and activators bind to two distinct sites in the transmembrane domain of the sulfonylurea receptor (SUR) subunit. We also discuss the possible mechanism of how these drugs allosterically modulate the dimerization of SUR nucleotide-binding domains (NBDs) and thus KATP channel activity. SIGNIFICANCE STATEMENT: ATP-sensitive potassium channels (KATP) are fundamental to energy homeostasis, and they participate in many vital physiological processes. KATP channels are important drug targets. Both KATP inhibitors (insulin secretagogues) and KATP activators are broadly used clinically for the treatment of related diseases. Recent cryogenic electron microscopy studies allow us to understand the emerging concept of KATP structural pharmacology.


Subject(s)
Insulins , Potassium Channels, Inwardly Rectifying , Adenosine Triphosphate/metabolism , Insulins/metabolism , KATP Channels/metabolism , Nucleotides/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Receptors, Drug/chemistry , Receptors, Drug/metabolism , Secretagogues , Sulfonylurea Receptors/metabolism
8.
Sci Adv ; 8(38): eabq8489, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36149965

ABSTRACT

We present the first structure of the human Kir2.1 channel containing both transmembrane domain (TMD) and cytoplasmic domain (CTD). Kir2.1 channels are strongly inward-rectifying potassium channels that play a key role in maintaining resting membrane potential. Their gating is modulated by phosphatidylinositol 4,5-bisphosphate (PIP2). Genetically inherited defects in Kir2.1 channels are responsible for several rare human diseases, including Andersen's syndrome. The structural analysis (cryo-electron microscopy), surface plasmon resonance, and electrophysiological experiments revealed a well-connected network of interactions between the PIP2-binding site and the G-loop through residues R312 and H221. In addition, molecular dynamics simulations and normal mode analysis showed the intrinsic tendency of the CTD to tether to the TMD and a movement of the secondary anionic binding site to the membrane even without PIP2. Our results revealed structural features unique to human Kir2.1 and provided insights into the connection between G-loop and gating and the pathological mechanisms associated with this channel.


Subject(s)
Molecular Dynamics Simulation , Phosphatidylinositols , Potassium Channels, Inwardly Rectifying , Cryoelectron Microscopy , Humans , Membrane Potentials , Potassium Channels, Inwardly Rectifying/chemistry
9.
Angew Chem Int Ed Engl ; 61(13): e202112232, 2022 03 21.
Article in English | MEDLINE | ID: mdl-34985791

ABSTRACT

Cholesterol oligomers reside in multiple membrane protein X-ray crystal structures. Yet, there is no direct link between these oligomers and a biological function. Here we present the structural and functional details of a cholesterol dimer that stabilizes the inactivated state of an inward-rectifier potassium channel KirBac1.1. K+ efflux assays confirm that high cholesterol concentration reduces K+ conductance. We then determine the structure of the cholesterol-KirBac1.1 complex using Xplor-NIH simulated annealing calculations driven by solid-state NMR distance measurements. These calculations identified an α-α cholesterol dimer docked to a cleft formed by adjacent subunits of the homotetrameric protein. We compare these results to coarse grain molecular dynamics simulations. This is one of the first examples of a cholesterol oligomer performing a distinct biological function and structural characterization of a conserved promiscuous lipid binding region.


Subject(s)
Potassium Channels, Inwardly Rectifying , Cholesterol , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism
10.
Nat Commun ; 13(1): 490, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35079013

ABSTRACT

Ion currents through potassium channels are gated. Constriction of the ion conduction pathway at the inner helix bundle, the textbook gate of Kir potassium channels, has been shown to be an ineffective permeation control, creating a rift in our understanding of how these channels are gated. Here we present evidence that anionic lipids act as interactive response elements sufficient to gate potassium conduction. We demonstrate the limiting barrier to K+ permeation lies within the ion conduction pathway and show that this gate is operated by the fatty acyl tails of lipids that infiltrate the conduction pathway via fenestrations in the walls of the pore. Acyl tails occupying a surface groove extending from the cytosolic interface to the conduction pathway provide a potential means of relaying cellular signals, mediated by anionic lipid head groups bound at the canonical lipid binding site, to the internal gate.


Subject(s)
Ion Channel Gating , Membrane Lipids/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , Anions/chemistry , Anions/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Ion Transport , Liposomes/chemistry , Liposomes/metabolism , Membrane Lipids/chemistry , Molecular Dynamics Simulation , Mutation , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics
11.
Hum Genet ; 141(2): 217-227, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34821995

ABSTRACT

Cooks syndrome (CS) is an ultrarare limb malformation due to in tandem microduplications involving KCNJ2 and extending to the 5' regulatory element of SOX9. To date, six CS families were resolved at the molecular level. Subsequent studies explored the evolutionary and pathological complexities of the SOX9-KCNJ2/Sox9-Kcnj2 locus, and suggested a key role for the formation of novel topologically associating domain (TAD) by inter-TAD duplications in causing CS. Here, we report a unique case of CS associated with a de novo 1;17 translocation affecting the KCNJ2 locus. On chromosome 17, the breakpoint mapped between KCNJ16 and KCNJ2, and combined with a ~ 5 kb deletion in the 5' of KCNJ2. Based on available capture Hi-C data, the breakpoint on chromosome 17 separated KCNJ2 from a putative enhancer. Gene expression analysis demonstrated downregulation of KCNJ2 in both patient's blood cells and cultured skin fibroblasts. Our findings suggest that a complex rearrangement falling in the 5' of KCNJ2 may mimic the developmental consequences of in tandem duplications affecting the SOX9-KCNJ2/Sox9-Kcnj2 locus. This finding adds weight to the notion of an intricate role of gene regulatory regions and, presumably, the related three-dimensional chromatin structure in normal and abnormal human morphology.


Subject(s)
Fingers/abnormalities , Foot Deformities, Congenital/genetics , Gene Rearrangement , Hand Deformities, Congenital/genetics , Potassium Channels, Inwardly Rectifying/genetics , Regulatory Sequences, Nucleic Acid , Adolescent , Adult , Chromosome Breakpoints , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 17/genetics , Facies , Female , Humans , In Situ Hybridization, Fluorescence , Male , Potassium Channels, Inwardly Rectifying/chemistry , Sequence Deletion , Translocation, Genetic , Young Adult
12.
Nat Commun ; 12(1): 7114, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880224

ABSTRACT

Protein domains are the basic units of protein structure and function. Comparative analysis of genomes and proteomes showed that domain recombination is a main driver of multidomain protein functional diversification and some of the constraining genomic mechanisms are known. Much less is known about biophysical mechanisms that determine whether protein domains can be combined into viable protein folds. Here, we use massively parallel insertional mutagenesis to determine compatibility of over 300,000 domain recombination variants of the Inward Rectifier K+ channel Kir2.1 with channel surface expression. Our data suggest that genomic and biophysical mechanisms acted in concert to favor gain of large, structured domain at protein termini during ion channel evolution. We use machine learning to build a quantitative biophysical model of domain compatibility in Kir2.1 that allows us to derive rudimentary rules for designing domain insertion variants that fold and traffic to the cell surface. Positional Kir2.1 responses to motif insertion clusters into distinct groups that correspond to contiguous structural regions of the channel with distinct biophysical properties tuned towards providing either folding stability or gating transitions. This suggests that insertional profiling is a high-throughput method to annotate function of ion channel structural regions.


Subject(s)
Biophysics , Potassium Channels/chemistry , Potassium Channels/genetics , Recombination, Genetic , Cell Line , Cell Membrane , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Gene Expression Profiling , HEK293 Cells , Humans , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Machine Learning , Mutagenesis, Insertional , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Protein Domains/genetics , Transcriptome
13.
Biochemistry ; 60(44): 3292-3301, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34676745

ABSTRACT

Membrane proteins represent a large family of proteins that perform vital physiological roles and represent key drug targets. Despite their importance, bioanalytical methods aiming to comprehensively characterize the post-translational modification (PTM) of membrane proteins remain challenging compared to other classes of proteins in part because of their inherent low expression and hydrophobicity. The inward rectifier potassium channel (Kir) 2.1, an integral membrane protein, is critical for the maintenance of the resting membrane potential and phase-3 repolarization of the cardiac action potential in the heart. The importance of this channel to cardiac physiology is highlighted by the recognition of several sudden arrhythmic death syndromes, Andersen-Tawil and short QT syndromes, which are associated with loss or gain of function mutations in Kir2.1, often triggered by changes in the ß-adrenergic tone. Therefore, understanding the PTMs of this channel (particularly ß-adrenergic tone-driven phosphorylation) is important for arrhythmia prevention. Here, we developed a proteomic method, integrating both top-down (intact protein) and bottom-up (after enzymatic digestion) proteomic analyses, to characterize the PTMs of recombinant wild-type and mutant Kir2.1, successfully mapping five novel sites of phosphorylation and confirming a sixth site. Our study provides a framework for future work to assess the role of PTMs in regulating Kir2.1 functions.


Subject(s)
Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Proteomics/methods , Action Potentials , HEK293 Cells , Heart , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Potentials , Membrane Proteins/metabolism , Phosphorylation , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Protein Processing, Post-Translational , Research Design
14.
Sci Rep ; 11(1): 6668, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758250

ABSTRACT

Developmental delay, epilepsy, and neonatal diabetes (DEND) syndrome, the most severe end of neonatal diabetes mellitus, is caused by mutation in the ATP-sensitive potassium (KATP) channel. In addition to diabetes, DEND patients present muscle weakness as one of the symptoms, and although the muscle weakness is considered to originate in the brain, the pathological effects of mutated KATP channels in skeletal muscle remain elusive. Here, we describe the local effects of the KATP channel on muscle by expressing the mutation present in the KATP channels of the DEND syndrome in the murine skeletal muscle cell line C2C12 in combination with computer simulation. The present study revealed that the DEND mutation can lead to a hyperpolarized state of the muscle cell membrane, and molecular dynamics simulations based on a recently reported high-resolution structure provide an explanation as to why the mutation reduces ATP sensitivity and reveal the changes in the local interactions between ATP molecules and the channel.


Subject(s)
Diabetes Mellitus/genetics , Epilepsy/genetics , Infant, Newborn, Diseases/genetics , KATP Channels/chemistry , KATP Channels/genetics , Muscle, Skeletal/metabolism , Mutation , Psychomotor Disorders/genetics , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Calcium/metabolism , Gene Expression , Glucose/metabolism , KATP Channels/metabolism , Membrane Potentials , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Muscle Development , Muscle Fibers, Skeletal , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship
15.
Biochim Biophys Acta Biomembr ; 1863(4): 183555, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33444624

ABSTRACT

Expression, purification, and functional reconstitution of mammalian ion channels are often challenging. Heterologous expression of mammalian channels in bacteria can be advantageous due to unrelated protein environment and the lack of risk of copurification of endogenous proteins, e.g., accessory channel subunits that can influence the channel activity. Also, direct recording of channel activity could be challenging due to their intracellular localization like in the case of mitochondrial channels. The activity of purified channels can be characterized at the single-molecule level by electrophysiological techniques, such as planar lipid bilayers (PLB). In this work, we describe a simple approach to accomplish PLB recording of the activity of single renal outer medullary potassium channels ROMK expressed in E. coli. We focused on the ROMK2 isoform that is present at low levels in the mitochondria and can be responsible for mitoKATP activity. We screened for the best construct to express the codon-optimized ROMK proteins with a 6xHis tag for protein purification. The strategy involved the use of optimal styrene-maleic acid (SMA) copolymer, which forms so-called polymer nanodiscs, to solubilize and purify ROMK-containing SMA lipid particles (SMALPs), which were amenable for fusion with PLB. Reconstituted ROMK channels exhibited ion selectivity, rectification, and pharmacological properties, which are in agreement with previous work on ROMK channels.


Subject(s)
Maleates/chemistry , Nanostructures/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Styrene/chemistry , Humans , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
16.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008764

ABSTRACT

ATP-sensitive potassium (K-ATP) channels are ubiquitously expressed on the plasma membrane of cells in several organs, including the heart, pancreas, and brain, and they govern a wide range of physiological processes. In pancreatic ß-cells, K-ATP channels composed of Kir6.2 and SUR1 play a key role in coupling blood glucose and insulin secretion. A tryptophan residue located at the cytosolic end of the transmembrane helix is highly conserved in eukaryote and prokaryote Kir channels. Any mutation on this amino acid causes a gain of function and neonatal diabetes mellitus. In this study, we have investigated the effect of mutation on this highly conserved residue on a KirBac channel (prokaryotic homolog of mammalian Kir6.2). We provide the crystal structure of the mutant KirBac3.1 W46R (equivalent to W68R in Kir6.2) and its conformational flexibility properties using HDX-MS. In addition, the detailed dynamical view of the mutant during the gating was investigated using the in silico method. Finally, functional assays have been performed. A comparison of important structural determinants for the gating mechanism between the wild type KirBac and the mutant W46R suggests interesting structural and dynamical clues and a mechanism of action of the mutation that leads to the gain of function.


Subject(s)
Conserved Sequence , Mutation/genetics , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Tryptophan/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Hydrogen Deuterium Exchange-Mass Spectrometry , Ion Channel Gating , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Domains , Protein Interaction Maps , Protein Structure, Secondary
18.
Elife ; 92020 08 26.
Article in English | MEDLINE | ID: mdl-32844743

ABSTRACT

G-protein-gated inward rectifier potassium (GIRK) channels are regulated by G proteins and PIP2. Here, using cryo-EM single particle analysis we describe the equilibrium ensemble of structures of neuronal GIRK2 as a function of the C8-PIP2 concentration. We find that PIP2 shifts the equilibrium between two distinguishable structures of neuronal GIRK (GIRK2), extended and docked, towards the docked form. In the docked form the cytoplasmic domain, to which Gßγ binds, becomes accessible to the cytoplasmic membrane surface where Gßγ resides. Furthermore, PIP2 binding reshapes the Gßγ binding surface on the cytoplasmic domain, preparing it to receive Gßγ. We find that cardiac GIRK (GIRK1/4) can also exist in both extended and docked conformations. These findings lead us to conclude that PIP2 influences GIRK channels in a structurally similar manner to Kir2.2 channels. In Kir2.2 channels, the PIP2-induced conformational changes open the pore. In GIRK channels, they prepare the channel for activation by Gßγ.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels , Phosphatidylinositol 4,5-Diphosphate , Animals , Cryoelectron Microscopy , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Mice , Neurons/chemistry , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Protein Conformation
19.
Int J Mol Sci ; 21(16)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796537

ABSTRACT

Inward rectifier potassium ion channels (IK1-channels) of the Kir2.x family are responsible for maintaining a stable negative resting membrane potential in excitable cells, but also play a role in processes of non-excitable tissues, such as bone development. IK1-channel loss-of-function, either congenital or acquired, has been associated with cardiac disease. Currently, basic research and specific treatment are hindered by the absence of specific and efficient Kir2.x channel activators. However, twelve different compounds, including approved drugs, show off-target IK1 activation. Therefore, these compounds contain valuable information towards the development of agonists of Kir channels, AgoKirs. We reviewed the mechanism of IK1 channel activation of these compounds, which can be classified as direct or indirect activators. Subsequently, we examined the most viable starting points for rationalized drug development and possible safety concerns with emphasis on cardiac and skeletal muscle adverse effects of AgoKirs. Finally, the potential value of AgoKirs is discussed in view of the current clinical applications of potentiators and activators in cystic fibrosis therapy.


Subject(s)
Heart Diseases/drug therapy , Potassium Channels, Inwardly Rectifying/agonists , Animals , Humans , Ion Channel Gating , Potassium Channels, Inwardly Rectifying/chemistry
20.
Proc Natl Acad Sci U S A ; 117(28): 16626-16637, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601236

ABSTRACT

Neuronal activity leads to an increase in local cerebral blood flow (CBF) to allow adequate supply of oxygen and nutrients to active neurons, a process termed neurovascular coupling (NVC). We have previously shown that capillary endothelial cell (cEC) inwardly rectifying K+ (Kir) channels can sense neuronally evoked increases in interstitial K+ and induce rapid and robust dilations of upstream parenchymal arterioles, suggesting a key role of cECs in NVC. The requirements of this signal conduction remain elusive. Here, we utilize mathematical modeling to investigate how small outward currents in stimulated cECs can elicit physiologically relevant spread of vasodilatory signals within the highly interconnected brain microvascular network to increase local CBF. Our model shows that the Kir channel can act as an "on-off" switch in cECs to hyperpolarize the cell membrane as extracellular K+ increases. A local hyperpolarization can be amplified by the voltage-dependent activation of Kir in neighboring cECs. Sufficient Kir density enables robust amplification of the hyperpolarizing stimulus and produces responses that resemble action potentials in excitable cells. This Kir-mediated excitability can remain localized in the stimulated region or regeneratively propagate over significant distances in the microvascular network, thus dramatically increasing the efficacy of K+ for eliciting local hyperemia. Modeling results show how changes in cEC transmembrane current densities and gap junctional resistances can affect K+-mediated NVC and suggest a key role for Kir as a sensor of neuronal activity and an amplifier of retrograde electrical signaling in the cerebral vasculature.


Subject(s)
Neurons/metabolism , Neurovascular Coupling , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , Animals , Brain/blood supply , Brain/metabolism , Cerebrovascular Circulation , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/chemistry , Potassium/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Signal Transduction , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...