Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 3608, 2023 06 17.
Article in English | MEDLINE | ID: mdl-37330603

ABSTRACT

KATP channels are metabolic sensors for intracellular ATP/ADP ratios, play essential roles in many physiological processes, and are implicated in a spectrum of pathological conditions. SUR2A-containing KATP channels differ from other subtypes in their sensitivity to Mg-ADP activation. However, the underlying structural mechanism remains poorly understood. Here we present a series of cryo-EM structures of SUR2A in the presence of different combinations of Mg-nucleotides and the allosteric inhibitor repaglinide. These structures uncover regulatory helix (R helix) on the NBD1-TMD2 linker, which wedges between NBD1 and NBD2. R helix stabilizes SUR2A in the NBD-separated conformation to inhibit channel activation. The competitive binding of Mg-ADP with Mg-ATP to NBD2 mobilizes the R helix to relieve such inhibition, allowing channel activation. The structures of SUR2B in similar conditions suggest that the C-terminal 42 residues of SUR2B enhance the structural dynamics of NBD2 and facilitate the dissociation of the R helix and the binding of Mg-ADP to NBD2, promoting NBD dimerization and subsequent channel activation.


Subject(s)
Potassium Channels, Inwardly Rectifying , Sulfonylurea Receptors/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/metabolism , Adenosine Diphosphate/metabolism , Dimerization , KATP Channels/metabolism
2.
Elife ; 112022 11 22.
Article in English | MEDLINE | ID: mdl-36413210

ABSTRACT

Phagocyte oxidase plays an essential role in the first line of host defense against pathogens. It oxidizes intracellular NADPH to reduce extracellular oxygen to produce superoxide anions that participate in pathogen killing. The resting phagocyte oxidase is a heterodimeric complex formed by two transmembrane proteins NOX2 and p22. Despite the physiological importance of this complex, its structure remains elusive. Here, we reported the cryo-EM structure of the functional human NOX2-p22 complex in nanodisc in the resting state. NOX2 shows a canonical 6-TM architecture of NOX and p22 has four transmembrane helices. M3, M4, and M5 of NOX2, and M1 and M4 helices of p22 are involved in the heterodimer formation. Dehydrogenase (DH) domain of NOX2 in the resting state is not optimally docked onto the transmembrane domain, leading to inefficient electron transfer and NADPH binding. Structural analysis suggests that the cytosolic factors might activate the NOX2-p22 complex by stabilizing the DH in a productive docked conformation.


Subject(s)
NADPH Oxidases , Phagocytes , Humans , NADP , Superoxides , Membrane Proteins
3.
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
4.
Front Pharmacol ; 13: 929684, 2022.
Article in English | MEDLINE | ID: mdl-35847046

ABSTRACT

Mitiglinide is a highly selective fast-acting anti-diabetic drug that induces insulin secretion by inhibiting pancreatic KATP channels. However, how mitiglinide binds KATP channels remains unknown. Here, we show the cryo-EM structure of the SUR1 subunit complexed with mitiglinide. The structure reveals that mitiglinide binds inside the common insulin secretagogue-binding site of SUR1, which is surrounded by TM7, TM8, TM16, and TM17. Mitiglinide locks SUR1 in the NBD-separated inward-facing conformation. The detailed structural analysis of the mitiglinide-binding site uncovers the molecular basis of its high selectivity.

5.
Nat Commun ; 13(1): 2770, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589716

ABSTRACT

ATP-sensitive potassium channels (KATP) are metabolic sensors that convert the intracellular ATP/ADP ratio to the excitability of cells. They are involved in many physiological processes and implicated in several human diseases. Here we present the cryo-EM structures of the pancreatic KATP channel in both the closed state and the pre-open state, resolved in the same sample. We observe the binding of nucleotides at the inhibitory sites of the Kir6.2 channel in the closed but not in the pre-open state. Structural comparisons reveal the mechanism for ATP inhibition and Mg-ADP activation, two fundamental properties of KATP channels. Moreover, the structures also uncover the activation mechanism of diazoxide-type KATP openers.


Subject(s)
Adenosine Triphosphate , KATP Channels , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Humans , KATP Channels/metabolism , Pancreas/metabolism , Sulfonylurea Receptors/metabolism
6.
Nat Commun ; 13(1): 2675, 2022 05 13.
Article in English | MEDLINE | ID: mdl-35562524

ABSTRACT

ATP-sensitive potassium channels (KATP), composed of Kir6 and SUR subunits, convert the metabolic status of the cell into electrical signals. Pharmacological activation of SUR2- containing KATP channels by class of small molecule drugs known as KATP openers leads to hyperpolarization of excitable cells and to vasodilation. Thus, KATP openers could be used to treat cardiovascular diseases. However, where these vasodilators bind to KATP and how they activate the channel remains elusive. Here, we present cryo-EM structures of SUR2A and SUR2B subunits in complex with Mg-nucleotides and P1075 or levcromakalim, two chemically distinct KATP openers that are specific to SUR2. Both P1075 and levcromakalim bind to a common site in the transmembrane domain (TMD) of the SUR2 subunit, which is between TMD1 and TMD2 and is embraced by TM10, TM11, TM12, TM14, and TM17. These KATP openers synergize with Mg-nucleotides to stabilize SUR2 in the NBD-dimerized occluded state to activate the channel.


Subject(s)
Potassium Channels, Inwardly Rectifying , Vasodilator Agents , Adenosine Triphosphate/metabolism , Binding Sites , Cromakalim , KATP Channels/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
7.
Neuron ; 110(6): 1023-1035.e5, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35051376

ABSTRACT

TRPC3 and TRPC6 channels are calcium-permeable non-selective cation channels that are involved in many physiological processes. The gain-of-function (GOF) mutations of TRPC6 lead to familial focal segmental glomerulosclerosis (FSGS) in humans, but their pathogenic mechanism remains elusive. Here, we report the cryo-EM structures of human TRPC3 in both high-calcium and low-calcium conditions. Based on these structures and accompanying electrophysiological studies, we identified both inhibitory and activating calcium-binding sites in TRPC3 that couple intracellular calcium concentrations to the basal channel activity. These calcium sensors are also structurally and functionally conserved in TRPC6. We uncovered that the GOF mutations of TRPC6 activate the channel by allosterically abolishing the inhibitory effects of intracellular calcium. Furthermore, structures of human TRPC6 in complex with two chemically distinct inhibitors bound at different ligand-binding pockets reveal different conformations of the transmembrane domain, providing templates for further structure-based drug design targeting TRPC6-related diseases such as FSGS.


Subject(s)
Calcium , Glomerulosclerosis, Focal Segmental , TRPC Cation Channels , TRPC6 Cation Channel , Binding Sites , Calcium/metabolism , Calcium Channels/metabolism , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism
8.
Elife ; 102021 03 08.
Article in English | MEDLINE | ID: mdl-33683200

ABSTRACT

TRPC5 channel is a nonselective cation channel that participates in diverse physiological processes. TRPC5 inhibitors show promise in the treatment of anxiety disorder, depression, and kidney disease. However, the binding sites and inhibitory mechanism of TRPC5 inhibitors remain elusive. Here, we present the cryo-EM structures of human TRPC5 in complex with two distinct inhibitors, namely clemizole and HC-070, to the resolution of 2.7 Å. The structures reveal that clemizole binds inside the voltage sensor-like domain of each subunit. In contrast, HC-070 is wedged between adjacent subunits and replaces the glycerol group of a putative diacylglycerol molecule near the extracellular side. Moreover, we found mutations in the inhibitor binding pockets altered the potency of inhibitors. These structures suggest that both clemizole and HC-070 exert the inhibitory functions by stabilizing the ion channel in a nonconductive closed state. These results pave the way for further design and optimization of inhibitors targeting human TRPC5.


Subject(s)
Benzimidazoles/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/chemistry , Benzimidazoles/metabolism , Binding Sites , Heterocyclic Compounds, 4 or More Rings/metabolism , Humans , Models, Molecular , TRPC Cation Channels/metabolism
9.
Nat Commun ; 12(1): 155, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420071

ABSTRACT

Dual oxidases (DUOXs) produce hydrogen peroxide by transferring electrons from intracellular NADPH to extracellular oxygen. They are involved in many crucial biological processes and human diseases, especially in thyroid diseases. DUOXs are protein complexes co-assembled from the catalytic DUOX subunits and the auxiliary DUOXA subunits and their activities are regulated by intracellular calcium concentrations. Here, we report the cryo-EM structures of human DUOX1-DUOXA1 complex in both high-calcium and low-calcium states. These structures reveal the DUOX1 complex is a symmetric 2:2 hetero-tetramer stabilized by extensive inter-subunit interactions. Substrate NADPH and cofactor FAD are sandwiched between transmembrane domain and the cytosolic dehydrogenase domain of DUOX. In the presence of calcium ions, intracellular EF-hand modules might enhance the catalytic activity of DUOX by stabilizing the dehydrogenase domain in a conformation that allows electron transfer.


Subject(s)
Calcium/metabolism , Dual Oxidases/chemistry , Membrane Proteins/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cryoelectron Microscopy , Dual Oxidases/genetics , Enzyme Activation , Enzyme Assays , Flavin-Adenine Dinucleotide/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Models, Molecular , NADP/metabolism , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
10.
Nat Commun ; 11(1): 6199, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273469

ABSTRACT

Resting membrane potential determines the excitability of the cell and is essential for the cellular electrical activities. The NALCN channel mediates sodium leak currents, which positively adjust resting membrane potential towards depolarization. The NALCN channel is involved in several neurological processes and has been implicated in a spectrum of neurodevelopmental diseases. Here, we report the cryo-EM structure of rat NALCN and mouse FAM155A complex to 2.7 Å resolution. The structure reveals detailed interactions between NALCN and the extracellular cysteine-rich domain of FAM155A. We find that the non-canonical architecture of NALCN selectivity filter dictates its sodium selectivity and calcium block, and that the asymmetric arrangement of two functional voltage sensors confers the modulation by membrane potential. Moreover, mutations associated with human diseases map to the domain-domain interfaces or the pore domain of NALCN, intuitively suggesting their pathological mechanisms.


Subject(s)
Calcium Channels/chemistry , Ion Channels/chemistry , Membrane Proteins/chemistry , Sodium/metabolism , Amino Acid Sequence , Animals , Calcium Channels/metabolism , HEK293 Cells , Humans , Ion Channels/metabolism , Membrane Potentials , Membrane Proteins/metabolism , Mice , Models, Molecular , Protein Domains , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats
11.
Nat Commun ; 11(1): 2478, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424158

ABSTRACT

Sterol O-acyltransferase 1 (SOAT1) is an endoplasmic reticulum (ER) resident, multi-transmembrane enzyme that belongs to the membrane-bound O-acyltransferase (MBOAT) family. It catalyzes the esterification of cholesterol to generate cholesteryl esters for cholesterol storage. SOAT1 is a target to treat several human diseases. However, its structure and mechanism remain elusive since its discovery. Here, we report the structure of human SOAT1 (hSOAT1) determined by cryo-EM. hSOAT1 is a tetramer consisted of a dimer of dimer. The structure of hSOAT1 dimer at 3.5 Å resolution reveals that a small molecule inhibitor CI-976 binds inside the catalytic chamber and blocks the accessibility of the active site residues H460, N421 and W420. Our results pave the way for future mechanistic study and rational drug design targeting hSOAT1 and other mammalian MBOAT family members.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Sterol O-Acyltransferase/chemistry , Binding Sites , Biocatalysis , HEK293 Cells , Humans , Ligands , Protein Multimerization , Sterol O-Acyltransferase/ultrastructure , Structure-Activity Relationship , Substrate Specificity/drug effects
12.
Biochemistry ; 59(1): 18-25, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31566370

ABSTRACT

Sulfonylureas and glinides are commonly used oral insulin secretagogues (ISs) that act on the pancreatic ATP-sensitive potassium (KATP) channel to promote insulin secretion in order to lower the blood glucose level. Physiologically, KATP channels are inhibited by intracellular ATP and activated by Mg-ADP. Therefore, they sense the cellular energy status to regulate the permeability of potassium ions across the plasma membrane. The pancreatic KATP channel is composed of the pore-forming Kir6.2 subunits and the regulatory SUR1 subunits. Previous electrophysiological studies have established that ISs bind to the SUR1 subunit and inhibit the channel activity primarily by two mechanisms. First, ISs prevent Mg-ADP activation. Second, ISs inhibit the channel activity of Kir6.2 directly. Several cryo-EM structures of the pancreatic KATP channel determined recently have provided remarkable structural insights into these two mechanisms.


Subject(s)
Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Secretagogues/metabolism , Sulfonylurea Receptors/metabolism , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites , Humans , Protein Conformation , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Sequence Alignment
13.
Nature ; 574(7777): 206-210, 2019 10.
Article in English | MEDLINE | ID: mdl-31514202

ABSTRACT

Soluble guanylate cyclase (sGC) is the primary sensor of nitric oxide. It has a central role in nitric oxide signalling and has been implicated in many essential physiological processes and disease conditions. The binding of nitric oxide boosts the enzymatic activity of sGC. However, the mechanism by which nitric oxide activates the enzyme is unclear. Here we report the cryo-electron microscopy structures of the human sGCα1ß1 heterodimer in different functional states. These structures revealed that the transducer module bridges the nitric oxide sensor module and the catalytic module. Binding of nitric oxide to the ß1 haem-nitric oxide and oxygen binding (H-NOX) domain triggers the structural rearrangement of the sensor module and a conformational switch of the transducer module from bending to straightening. The resulting movement of the N termini of the catalytic domains drives structural changes within the catalytic module, which in turn boost the enzymatic activity of sGC.


Subject(s)
Cryoelectron Microscopy , Soluble Guanylyl Cyclase/metabolism , Soluble Guanylyl Cyclase/ultrastructure , Animals , Disulfides/chemistry , Disulfides/metabolism , Drosophila melanogaster , Enzyme Activation , HEK293 Cells , Heme/metabolism , Humans , Hydrazines/pharmacology , Mice , Models, Molecular , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Oxygen/metabolism , Protein Domains , Protein Multimerization , Soluble Guanylyl Cyclase/chemistry , Soluble Guanylyl Cyclase/genetics
14.
Cell Rep ; 27(6): 1848-1857.e4, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31067468

ABSTRACT

Repaglinide (RPG) is a short-acting insulin secretagogue widely prescribed for the treatment of type 2 diabetes. It boosts insulin secretion by inhibiting the pancreatic ATP-sensitive potassium channel (KATP). However, the mechanisms by which RPG binds to the KATP channel are poorly understood. Here, we describe two cryo-EM structures: the pancreatic KATP channel in complex with inhibitory RPG and adenosine-5'-(γ-thio)-triphosphate (ATPγS) at 3.3 Å and a medium-resolution structure of a RPG-bound mini SUR1 protein in which the N terminus of the inward-rectifying potassium channel 6.1 (Kir6.1) is fused to the ABC transporter module of the sulfonylurea receptor 1 (SUR1). These structures reveal the binding site of RPG in the SUR1 subunit. Furthermore, the high-resolution structure reveals the complex architecture of the ATP binding site, which is formed by both Kir6.2 and SUR1 subunits, and the domain-domain interaction interfaces.


Subject(s)
Carbamates/chemistry , Carbamates/metabolism , Pancreas/metabolism , Piperidines/chemistry , Piperidines/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Binding , Protein Interaction Mapping , Protein Subunits/chemistry , Protein Subunits/metabolism , Sulfonylurea Receptors/chemistry , Sulfonylurea Receptors/metabolism , Sulfonylurea Receptors/ultrastructure
15.
BMC Anesthesiol ; 18(1): 195, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30577757

ABSTRACT

BACKGROUND: Laparoscopic surgery typically requires deep neuromuscular blockade (NMB), but whether deep or moderate NMB is superior for thoracoscopic surgery remains controversial. METHODS: Patients scheduled for thoracoscopic lobectomy under intravenous anesthesia were randomly assigned to receive moderate [train of four (TOF) 1-2] or deep NMB [TOF 0, post-tetanic count (PTC) 1-5]. Depth of anesthesia was controlled at a Narcotrend rating of 30 ± 5 in both groups. The primary outcome was the need to use an additional muscle relaxant (cisatracurium) during surgery. Secondary outcomes included surgeon satisfaction, recovery time of each stage after drug withdrawal [time from withdrawal until TOF recovery to 20% (antagonists administration), 25, 75, 90, 100%], blood gas data, VAS pain grade after extubation, the time it takes for patients to begin walking after surgery, postoperative complications and hospitalization time. Results were analyzed on an intention-to-treat basis. RESULTS: Thirty patients were enrolled per arm, and all but one patient in each arm was included in the final analysis. Among patients undergoing moderate NMB, surgeons applied additional cisatracurium in 8 patients because of body movement and 5 because of coughing (13/29, 44.8%). Additional cisatracurium was not applied to any of the patients undergoing deep NMB (p < 0.001). Surgeons reported significantly higher satisfaction for patients undergoing deep NMB (p < 0.001, Wilcoxon rank sum test). The mean difference between the two groups in the time from withdrawal until TOF recovery of 25% or 90% was 10 min (p < 0.001). The two groups were similar in other recovery data, blood gas analysis, VAS pain grade, days for beginning to walk and mean hospitalization time. CONCLUSIONS: Deep NMB can reduce the use of additional muscle relaxant and increase surgeon satisfaction during thoracoscopic lobectomy. TRIAL REGISTRATION: Chinese Clinical Trial Registry, ChiCTR-IOR-15007117 , 22 September 2015.


Subject(s)
Atracurium/analogs & derivatives , Laparoscopy/methods , Neuromuscular Blockade/methods , Thoracoscopy/methods , Aged , Anesthesia, Intravenous/methods , Atracurium/administration & dosage , Blood Gas Analysis , Double-Blind Method , Female , Hospitalization/statistics & numerical data , Humans , Length of Stay , Lung/surgery , Male , Middle Aged , Neuromuscular Blocking Agents/administration & dosage , Neuromuscular Monitoring/methods , Postoperative Complications/epidemiology , Time Factors
16.
Nat Struct Mol Biol ; 25(9): 850-858, 2018 09.
Article in English | MEDLINE | ID: mdl-30190597

ABSTRACT

Mechanosensitive ion channels convert mechanical stimuli into a flow of ions. These channels are widely distributed from bacteria to higher plants and humans, and are involved in many crucial physiological processes. Here we show that two members of the OSCA protein family in Arabidopsis thaliana, namely AtOSCA1.1 and AtOSCA3.1, belong to a new class of mechanosensitive ion channels. We solve the structure of the AtOSCA1.1 channel at 3.5-Å resolution and AtOSCA3.1 at 4.8-Å resolution by cryo-electron microscopy. OSCA channels are symmetric dimers that are mediated by cytosolic inter-subunit interactions. Strikingly, they have structural similarity to the mammalian TMEM16 family proteins. Our structural analysis accompanied with electrophysiological studies identifies the ion permeation pathway within each subunit and suggests a conformational change model for activation.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/physiology , Ion Channels/chemistry , Ion Channels/physiology , Mechanotransduction, Cellular , Animals , Cryoelectron Microscopy , Cytoplasm/chemistry , Dimerization , Humans
18.
Cell Res ; 28(7): 746-755, 2018 07.
Article in English | MEDLINE | ID: mdl-29700422

ABSTRACT

TRPC6 and TRPC3 are receptor-activated nonselective cation channels that belong to the family of canonical transient receptor potential (TRPC) channels. They are activated by diacylglycerol, a lipid second messenger. TRPC6 and TRPC3 are involved in many physiological processes and implicated in human genetic diseases. Here we present the structure of human TRPC6 homotetramer in complex with a newly identified high-affinity inhibitor BTDM solved by single-particle cryo-electron microscopy to 3.8 Å resolution. We also present the structure of human TRPC3 at 4.4 Å resolution. These structures show two-layer architectures in which the bell-shaped cytosolic layer holds the transmembrane layer. Extensive inter-subunit interactions of cytosolic domains, including the N-terminal ankyrin repeats and the C-terminal coiled-coil, contribute to the tetramer assembly. The high-affinity inhibitor BTDM wedges between the S5-S6 pore domain and voltage sensor-like domain to inhibit channel opening. Our structures uncover the molecular architecture of TRPC channels and provide a structural basis for understanding the mechanism of these channels.


Subject(s)
TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/chemistry , TRPC6 Cation Channel/antagonists & inhibitors , TRPC6 Cation Channel/chemistry , Animals , Cryoelectron Microscopy/methods , HEK293 Cells , Humans , Protein Domains , Protein Multimerization , Protein Structure, Quaternary , Sf9 Cells , Spodoptera
19.
Protein Cell ; 9(6): 553-567, 2018 06.
Article in English | MEDLINE | ID: mdl-29594720

ABSTRACT

ATP-sensitive potassium channels (KATP) are energy sensors on the plasma membrane. By sensing the intracellular ADP/ATP ratio of ß-cells, pancreatic KATP channels control insulin release and regulate metabolism at the whole body level. They are implicated in many metabolic disorders and diseases and are therefore important drug targets. Here, we present three structures of pancreatic KATP channels solved by cryo-electron microscopy (cryo-EM), at resolutions ranging from 4.1 to 4.5 Å. These structures depict the binding site of the antidiabetic drug glibenclamide, indicate how Kir6.2 (inward-rectifying potassium channel 6.2) N-terminus participates in the coupling between the peripheral SUR1 (sulfonylurea receptor 1) subunit and the central Kir6.2 channel, reveal the binding mode of activating nucleotides, and suggest the mechanism of how Mg-ADP binding on nucleotide binding domains (NBDs) drives a conformational change of the SUR1 subunit.


Subject(s)
Pancreas/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Sulfonylurea Receptors/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , Ligands , Mesocricetus , Mice , Models, Molecular , Nucleotides/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Sf9 Cells , Spodoptera , Sulfonylurea Receptors/metabolism
20.
Cell ; 168(1-2): 101-110.e10, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28086082

ABSTRACT

ATP-sensitive potassium channels (KATP) couple intracellular ATP levels with membrane excitability. These channels play crucial roles in many essential physiological processes and have been implicated extensively in a spectrum of metabolic diseases and disorders. To gain insight into the mechanism of KATP, we elucidated the structure of a hetero-octameric pancreatic KATP channel in complex with a non-competitive inhibitor glibenclamide by single-particle cryoelectron microscopy to 5.6-Å resolution. The structure shows that four SUR1 regulatory subunits locate peripherally and dock onto the central Kir6.2 channel tetramer through the SUR1 TMD0-L0 fragment. Glibenclamide-bound SUR1 uses TMD0-L0 fragment to stabilize Kir6.2 channel in a closed conformation. In another structural population, a putative co-purified phosphatidylinositol 4,5-bisphosphate (PIP2) molecule uncouples Kir6.2 from glibenclamide-bound SUR1. These structural observations suggest a molecular mechanism for KATP regulation by anti-diabetic sulfonylurea drugs, intracellular adenosine nucleotide concentrations, and PIP2 lipid.


Subject(s)
KATP Channels/chemistry , KATP Channels/metabolism , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Cryoelectron Microscopy , Humans , Hydrolases/chemistry , Hydrolases/metabolism , Mammals/metabolism , Mesocricetus , Mice , Models, Molecular , Phosphoinositide Phospholipase C/chemistry , Phosphoinositide Phospholipase C/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Receptors/chemistry , Sulfonylurea Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL