Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3691, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693179

ABSTRACT

Voltage-gated sodium (NaV) channels mediate a plethora of electrical activities. NaV channels govern cellular excitability in response to depolarizing stimuli. Inactivation is an intrinsic property of NaV channels that regulates cellular excitability by controlling the channel availability. The fast inactivation, mediated by the Ile-Phe-Met (IFM) motif and the N-terminal helix (N-helix), has been well-characterized. However, the molecular mechanism underlying NaV channel slow inactivation remains elusive. Here, we demonstrate that the removal of the N-helix of NaVEh (NaVEhΔN) results in a slow-inactivated channel, and present cryo-EM structure of NaVEhΔN in a potential slow-inactivated state. The structure features a closed activation gate and a dilated selectivity filter (SF), indicating that the upper SF and the inner gate could serve as a gate for slow inactivation. In comparison to the NaVEh structure, NaVEhΔN undergoes marked conformational shifts on the intracellular side. Together, our results provide important mechanistic insights into NaV channel slow inactivation.


Subject(s)
Cryoelectron Microscopy , Ion Channel Gating , Voltage-Gated Sodium Channels , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/chemistry , Humans , Animals , HEK293 Cells , Models, Molecular
2.
Nat Struct Mol Biol ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664565

ABSTRACT

RNA uptake by cells is critical for RNA-mediated gene interference (RNAi) and RNA-based therapeutics. In Caenorhabditis elegans, RNAi is systemic as a result of SID-1-mediated double-stranded RNA (dsRNA) across cells. Despite the functional importance, the underlying mechanisms of dsRNA internalization by SID-1 remain elusive. Here we describe cryogenic electron microscopy structures of SID-1, SID-1-dsRNA complex and human SID-1 homologs SIDT1 and SIDT2, elucidating the structural basis of dsRNA recognition and import by SID-1. The homodimeric SID-1 homologs share conserved architecture, but only SID-1 possesses the molecular determinants within its extracellular domains for distinguishing dsRNA from single-stranded RNA and DNA. We show that the removal of the long intracellular loop between transmembrane helix 1 and 2 attenuates dsRNA uptake and systemic RNAi in vivo, suggesting a possible endocytic mechanism of SID-1-mediated dsRNA internalization. Our study provides mechanistic insights into dsRNA internalization by SID-1, which may facilitate the development of dsRNA applications based on SID-1.

3.
Nature ; 626(7998): 427-434, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38081299

ABSTRACT

Vesicular monoamine transporter 2 (VMAT2) accumulates monoamines in presynaptic vesicles for storage and exocytotic release, and has a vital role in monoaminergic neurotransmission1-3. Dysfunction of monoaminergic systems causes many neurological and psychiatric disorders, including Parkinson's disease, hyperkinetic movement disorders and depression4-6. Suppressing VMAT2 with reserpine and tetrabenazine alleviates symptoms of hypertension and Huntington's disease7,8, respectively. Here we describe cryo-electron microscopy structures of human VMAT2 complexed with serotonin and three clinical drugs at 3.5-2.8 Å, demonstrating the structural basis for transport and inhibition. Reserpine and ketanserin occupy the substrate-binding pocket and lock VMAT2 in cytoplasm-facing and lumen-facing states, respectively, whereas tetrabenazine binds in a VMAT2-specific pocket and traps VMAT2 in an occluded state. The structures in three distinct states also reveal the structural basis of the VMAT2 transport cycle. Our study establishes a structural foundation for the mechanistic understanding of substrate recognition, transport, drug inhibition and pharmacology of VMAT2 while shedding light on the rational design of potential therapeutic agents.


Subject(s)
Cryoelectron Microscopy , Vesicular Monoamine Transport Proteins , Humans , Binding Sites , Cytoplasm/drug effects , Cytoplasm/metabolism , Ketanserin/chemistry , Ketanserin/metabolism , Ketanserin/pharmacology , Reserpine/chemistry , Reserpine/metabolism , Reserpine/pharmacology , Serotonin/chemistry , Serotonin/metabolism , Substrate Specificity , Tetrabenazine/chemistry , Tetrabenazine/metabolism , Tetrabenazine/pharmacology , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Vesicular Monoamine Transport Proteins/chemistry , Vesicular Monoamine Transport Proteins/metabolism , Vesicular Monoamine Transport Proteins/ultrastructure
4.
Cell Rep ; 42(8): 112858, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37494189

ABSTRACT

The sodium-activated Slo2.2 channel is abundantly expressed in the brain, playing a critical role in regulating neuronal excitability. The Na+-binding site and the underlying mechanisms of Na+-dependent activation remain unclear. Here, we present cryoelectron microscopy (cryo-EM) structures of human Slo2.2 in closed, open, and inhibitor-bound form at resolutions of 2.6-3.2 Å, revealing gating mechanisms of Slo2.2 regulation by cations and a potent inhibitor. The cytoplasmic gating ring domain of the closed Slo2.2 harbors multiple K+ and Zn2+ sites, which stabilize the channel in the closed conformation. The open Slo2.2 structure reveals at least two Na+-sensitive sites where Na+ binding induces expansion and rotation of the gating ring that opens the inner gate. Furthermore, a potent inhibitor wedges into a pocket formed by pore helix and S6 helix and blocks the pore. Together, our results provide a comprehensive structural framework for the investigation of Slo2.2 channel gating, Na+ sensation, and inhibition.


Subject(s)
Potassium Channels , Sodium , Humans , Potassium Channels/metabolism , Cryoelectron Microscopy , Potassium Channels, Sodium-Activated , Sodium/metabolism
5.
Nat Commun ; 14(1): 2442, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37117223

ABSTRACT

Voltage-gated sodium (NaV) channels are critical regulators of neuronal excitability and are targeted by many toxins that directly interact with the pore-forming α subunit, typically via extracellular loops of the voltage-sensing domains, or residues forming part of the pore domain. Excelsatoxin A (ExTxA), a pain-causing knottin peptide from the Australian stinging tree Dendrocnide excelsa, is the first reported plant-derived NaV channel modulating peptide toxin. Here we show that TMEM233, a member of the dispanin family of transmembrane proteins expressed in sensory neurons, is essential for pharmacological activity of ExTxA at NaV channels, and that co-expression of TMEM233 modulates the gating properties of NaV1.7. These findings identify TMEM233 as a previously unknown NaV1.7-interacting protein, position TMEM233 and the dispanins as accessory proteins that are indispensable for toxin-mediated effects on NaV channel gating, and provide important insights into the function of NaV channels in sensory neurons.


Subject(s)
Toxins, Biological , Urtica dioica , Australia , Pain , Peptides , NAV1.7 Voltage-Gated Sodium Channel/metabolism
6.
Nat Commun ; 14(1): 1030, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36823201

ABSTRACT

The sodium channel NaV1.6 is widely expressed in neurons of the central and peripheral nervous systems, which plays a critical role in regulating neuronal excitability. Dysfunction of NaV1.6 has been linked to epileptic encephalopathy, intellectual disability and movement disorders. Here we present cryo-EM structures of human NaV1.6/ß1/ß2 alone and complexed with a guanidinium neurotoxin 4,9-anhydro-tetrodotoxin (4,9-ah-TTX), revealing molecular mechanism of NaV1.6 inhibition by the blocker. The apo-form structure reveals two potential Na+ binding sites within the selectivity filter, suggesting a possible mechanism for Na+ selectivity and conductance. In the 4,9-ah-TTX bound structure, 4,9-ah-TTX binds to a pocket similar to the tetrodotoxin (TTX) binding site, which occupies the Na+ binding sites and completely blocks the channel. Molecular dynamics simulation results show that subtle conformational differences in the selectivity filter affect the affinity of TTX analogues. Taken together, our results provide important insights into NaV1.6 structure, ion conductance, and inhibition.


Subject(s)
NAV1.6 Voltage-Gated Sodium Channel , Sodium Channel Blockers , Tetrodotoxin , Humans , Molecular Dynamics Simulation , Neurons/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/analogs & derivatives , Tetrodotoxin/pharmacology , NAV1.6 Voltage-Gated Sodium Channel/chemistry
7.
Nat Chem Biol ; 19(1): 81-90, 2023 01.
Article in English | MEDLINE | ID: mdl-36302896

ABSTRACT

The TRPV3 channel plays vital roles in skin physiology. Dysfunction of TRPV3 causes skin diseases, including Olmsted syndrome. However, the lack of potent and selective inhibitors impedes the validation of TRPV3 as a therapeutic target. In this study, we identified Trpvicin as a potent and subtype-selective inhibitor of TRPV3. Trpvicin exhibits pharmacological potential in the inhibition of itch and hair loss in mouse models. Cryogenic electron microscopy structures of TRPV3 and the pathogenic G573S mutant complexed with Trpvicin reveal detailed ligand-binding sites, suggesting that Trpvicin inhibits the TRPV3 channel by stabilizing it in a closed state. Our G573S mutant structures demonstrate that the mutation causes a dilated pore, generating constitutive opening activity. Trpvicin accesses additional binding sites inside the central cavity of the G573S mutant to remodel the channel symmetry and block the channel. Together, our results provide mechanistic insights into the inhibition of TRPV3 by Trpvicin and support TRPV3-related drug development.


Subject(s)
TRPV Cation Channels , Mice , Animals , TRPV Cation Channels/genetics , TRPV Cation Channels/chemistry , Mutation , Binding Sites
8.
Nat Struct Mol Biol ; 29(12): 1208-1216, 2022 12.
Article in English | MEDLINE | ID: mdl-36424527

ABSTRACT

Voltage-gated sodium channel NaV1.7 plays essential roles in pain and odor perception. NaV1.7 variants cause pain disorders. Accordingly, NaV1.7 has elicited extensive attention in developing new analgesics. Here we present cryo-EM structures of human NaV1.7/ß1/ß2 complexed with inhibitors XEN907, TC-N1752 and NaV1.7-IN2, explaining specific binding sites and modulation mechanism for the pore blockers. These inhibitors bind in the central cavity blocking ion permeation, but engage different parts of the cavity wall. XEN907 directly causes α- to π-helix transition of DIV-S6 helix, which tightens the fast inactivation gate. TC-N1752 induces π-helix transition of DII-S6 helix mediated by a conserved asparagine on DIII-S6, which closes the activation gate. NaV1.7-IN2 serves as a pore blocker without causing conformational change. Electrophysiological results demonstrate that XEN907 and TC-N1752 stabilize NaV1.7 in inactivated state and delay the recovery from inactivation. Our results provide structural framework for NaV1.7 modulation by pore blockers, and important implications for developing subtype-selective analgesics.


Subject(s)
Pain , Humans , Binding Sites
9.
Front Pharmacol ; 13: 908867, 2022.
Article in English | MEDLINE | ID: mdl-35721169

ABSTRACT

Voltage-gated sodium (NaV) channels are responsible for the rapid rising-phase of action potentials in excitable cells. Over 1,000 mutations in NaV channels are associated with human diseases including epilepsy, periodic paralysis, arrhythmias and pain disorders. Natural toxins and clinically-used small-molecule drugs bind to NaV channels and modulate their functions. Recent advances from cryo-electron microscopy (cryo-EM) structures of NaV channels reveal invaluable insights into the architecture, activation, fast inactivation, electromechanical coupling, ligand modulation and pharmacology of eukaryotic NaV channels. These structural analyses not only demonstrate molecular mechanisms for NaV channel structure and function, but also provide atomic level templates for rational development of potential subtype-selective therapeutics. In this review, we summarize recent structural advances of eukaryotic NaV channels, highlighting the structural features of eukaryotic NaV channels as well as distinct modulation mechanisms by a wide range of modulators from natural toxins to synthetic small-molecules.

10.
Nat Commun ; 13(1): 2713, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35581266

ABSTRACT

Voltage-gated sodium (NaV) channels initiate action potentials. Fast inactivation of NaV channels, mediated by an Ile-Phe-Met motif, is crucial for preventing hyperexcitability and regulating firing frequency. Here we present cryo-electron microscopy structure of NaVEh from the coccolithophore Emiliania huxleyi, which reveals an unexpected molecular gating mechanism for NaV channel fast inactivation independent of the Ile-Phe-Met motif. An N-terminal helix of NaVEh plugs into the open activation gate and blocks it. The binding pose of the helix is stabilized by multiple electrostatic interactions. Deletion of the helix or mutations blocking the electrostatic interactions completely abolished the fast inactivation. These strong interactions enable rapid inactivation, but also delay recovery from fast inactivation, which is ~160-fold slower than human NaV channels. Together, our results provide mechanistic insights into fast inactivation of NaVEh that fundamentally differs from the conventional local allosteric inhibition, revealing both surprising structural diversity and functional conservation of ion channel inactivation.


Subject(s)
Eukaryota , Voltage-Gated Sodium Channels , Action Potentials , Cryoelectron Microscopy , Eukaryota/metabolism , Humans , Sodium/metabolism , Voltage-Gated Sodium Channels/genetics
11.
Nat Commun ; 13(1): 1286, 2022 03 11.
Article in English | MEDLINE | ID: mdl-35277491

ABSTRACT

Voltage-gated sodium (NaV) channels play fundamental roles in initiating and propagating action potentials. NaV1.3 is involved in numerous physiological processes including neuronal development, hormone secretion and pain perception. Here we report structures of human NaV1.3/ß1/ß2 in complex with clinically-used drug bulleyaconitine A and selective antagonist ICA121431. Bulleyaconitine A is located around domain I-II fenestration, providing the detailed view of the site-2 neurotoxin binding site. It partially blocks ion path and expands the pore-lining helices, elucidating how the bulleyaconitine A reduces peak amplitude but improves channel open probability. In contrast, ICA121431 preferentially binds to activated domain IV voltage-sensor, consequently strengthens the Ile-Phe-Met motif binding to its receptor site, stabilizes the channel in inactivated state, revealing an allosterically inhibitory mechanism of NaV channels. Our results provide structural details of distinct small-molecular modulators binding sites, elucidate molecular mechanisms of their action on NaV channels and pave a way for subtype-selective therapeutic development.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Voltage-Gated Sodium Channel Blockers , Binding Sites , Humans , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Protein Structure, Secondary , Sodium/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacology
12.
Cell Rep ; 37(5): 109931, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731621

ABSTRACT

N-type voltage-gated calcium (CaV) channels mediate Ca2+ influx at presynaptic terminals in response to action potentials and play vital roles in synaptogenesis, release of neurotransmitters, and nociceptive transmission. Here, we elucidate a cryo-electron microscopy (cryo-EM) structure of the human CaV2.2 complex in apo, ziconotide-bound, and two CaV2.2-specific pore blockers-bound states. The second voltage-sensing domain (VSD) is captured in a resting-state conformation, trapped by a phosphatidylinositol 4,5-bisphosphate (PIP2) molecule, which is distinct from the other three VSDs of CaV2.2, as well as activated VSDs observed in previous structures of CaV channels. This structure reveals the molecular basis for the unique inactivation process of CaV2.2 channels, in which the intracellular gate formed by S6 helices is closed and a W-helix from the domain II-III linker stabilizes closed-state inactivation. The structures of this inactivated, drug-bound complex lay a solid foundation for developing new state-dependent blockers for treatment of chronic pain.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Dipeptides/pharmacology , Ion Channel Gating/drug effects , omega-Conotoxins/pharmacology , Action Potentials , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/ultrastructure , Calcium Signaling , Cryoelectron Microscopy , HEK293 Cells , Humans , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Conformation, alpha-Helical , Structure-Activity Relationship
13.
Cell ; 184(20): 5151-5162.e11, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34520724

ABSTRACT

The heartbeat is initiated by voltage-gated sodium channel NaV1.5, which opens rapidly and triggers the cardiac action potential; however, the structural basis for pore opening remains unknown. Here, we blocked fast inactivation with a mutation and captured the elusive open-state structure. The fast inactivation gate moves away from its receptor, allowing asymmetric opening of pore-lining S6 segments, which bend and rotate at their intracellular ends to dilate the activation gate to ∼10 Å diameter. Molecular dynamics analyses predict physiological rates of Na+ conductance. The open-state pore blocker propafenone binds in a high-affinity pose, and drug-access pathways are revealed through the open activation gate and fenestrations. Comparison with mutagenesis results provides a structural map of arrhythmia mutations that target the activation and fast inactivation gates. These results give atomic-level insights into molecular events that underlie generation of the action potential, open-state drug block, and fast inactivation of cardiac sodium channels, which initiate the heartbeat.


Subject(s)
NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Animals , Arrhythmias, Cardiac/genetics , Cryoelectron Microscopy , HEK293 Cells , Heart Rate/drug effects , Humans , Ion Channel Gating , Models, Molecular , Molecular Dynamics Simulation , Mutation/genetics , Myocardium , NAV1.5 Voltage-Gated Sodium Channel/isolation & purification , NAV1.5 Voltage-Gated Sodium Channel/ultrastructure , Propafenone/pharmacology , Protein Conformation , Rats , Sodium/metabolism , Time Factors , Water/chemistry
14.
Methods Enzymol ; 653: 89-101, 2021.
Article in English | MEDLINE | ID: mdl-34099183

ABSTRACT

Voltage-gated sodium channel NaV1.5 is responsible for initiating and propagating cardiac action potentials by selectively conducting Na+ into cardiomyocytes. Class-I antiarrhythmic drugs target NaV1.5 for treatment of arrhythmias. During the last few years, cryogenic electron microscopy (cryo-EM) has become a powerful technique to determine the structures of ion channels at atomic level. In order to reveal the structural features of NaV1.5 and the structural basis for its interaction with antiarrhythmic drugs by cryo-EM, NaV1.5 protein must be expressed at high levels and purified to homogeneity. In this chapter, we discuss the expression and purification of NaV1.5 in a mammalian expression system. We optimized the construct by deleting unstructured intracellular loops of rat NaV1.5 while retaining core functional regions. The resulting rNaV1.5C is fully functional and is blocked by Class-I antiarrhythmic drugs in a state-dependent manner. Protocols are presented for expressing and purifying sufficient sample of NaV1.5 for preparing cryo-EM grids. The resulting cryo-EM structure is briefly described.


Subject(s)
Myocytes, Cardiac , Voltage-Gated Sodium Channels , Action Potentials , Animals , Cryoelectron Microscopy , Myocytes, Cardiac/metabolism , Rats , Sodium/metabolism
15.
Nat Commun ; 12(1): 128, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397917

ABSTRACT

Voltage-gated sodium (NaV) channels initiate action potentials in excitable cells, and their function is altered by potent gating-modifier toxins. The α-toxin LqhIII from the deathstalker scorpion inhibits fast inactivation of cardiac NaV1.5 channels with IC50 = 11.4 nM. Here we reveal the structure of LqhIII bound to NaV1.5 at 3.3 Å resolution by cryo-EM. LqhIII anchors on top of voltage-sensing domain IV, wedged between the S1-S2 and S3-S4 linkers, which traps the gating charges of the S4 segment in a unique intermediate-activated state stabilized by four ion-pairs. This conformational change is propagated inward to weaken binding of the fast inactivation gate and favor opening the activation gate. However, these changes do not permit Na+ permeation, revealing why LqhIII slows inactivation of NaV channels but does not open them. Our results provide important insights into the structural basis for gating-modifier toxin binding, voltage-sensor trapping, and fast inactivation of NaV channels.


Subject(s)
Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Scorpion Venoms/toxicity , Animals , Binding Sites , Cryoelectron Microscopy , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Molecular Dynamics Simulation , NAV1.5 Voltage-Gated Sodium Channel/ultrastructure , Protein Conformation , Rats , Scorpion Venoms/chemistry , Sodium/metabolism
16.
Nature ; 585(7823): 129-134, 2020 09.
Article in English | MEDLINE | ID: mdl-32848250

ABSTRACT

Transmembrane channels and pores have key roles in fundamental biological processes1 and in biotechnological applications such as DNA nanopore sequencing2-4, resulting in considerable interest in the design of pore-containing proteins. Synthetic amphiphilic peptides have been found to form ion channels5,6, and there have been recent advances in de novo membrane protein design7,8 and in redesigning naturally occurring channel-containing proteins9,10. However, the de novo design of stable, well-defined transmembrane protein pores that are capable of conducting ions selectively or are large enough to enable the passage of small-molecule fluorophores remains an outstanding challenge11,12. Here we report the computational design of protein pores formed by two concentric rings of α-helices that are stable and monodisperse in both their water-soluble and their transmembrane forms. Crystal structures of the water-soluble forms of a 12-helical pore and a 16-helical pore closely match the computational design models. Patch-clamp electrophysiology experiments show that, when expressed in insect cells, the transmembrane form of the 12-helix pore enables the passage of ions across the membrane with high selectivity for potassium over sodium; ion passage is blocked by specific chemical modification at the pore entrance. When incorporated into liposomes using in vitro protein synthesis, the transmembrane form of the 16-helix pore-but not the 12-helix pore-enables the passage of biotinylated Alexa Fluor 488. A cryo-electron microscopy structure of the 16-helix transmembrane pore closely matches the design model. The ability to produce structurally and functionally well-defined transmembrane pores opens the door to the creation of designer channels and pores for a wide variety of applications.


Subject(s)
Computer Simulation , Genes, Synthetic/genetics , Ion Channels/chemistry , Ion Channels/genetics , Models, Molecular , Synthetic Biology , Cell Line , Cryoelectron Microscopy , Crystallography, X-Ray , Electric Conductivity , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrazines , Ion Channels/metabolism , Ion Transport , Liposomes/metabolism , Patch-Clamp Techniques , Porins/chemistry , Porins/genetics , Porins/metabolism , Protein Engineering , Protein Structure, Secondary , Solubility , Water/chemistry
17.
Cell ; 180(1): 122-134.e10, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31866066

ABSTRACT

Voltage-gated sodium channel Nav1.5 generates cardiac action potentials and initiates the heartbeat. Here, we report structures of NaV1.5 at 3.2-3.5 Å resolution. NaV1.5 is distinguished from other sodium channels by a unique glycosyl moiety and loss of disulfide-bonding capability at the NaVß subunit-interaction sites. The antiarrhythmic drug flecainide specifically targets the central cavity of the pore. The voltage sensors are partially activated, and the fast-inactivation gate is partially closed. Activation of the voltage sensor of Domain III allows binding of the isoleucine-phenylalanine-methionine (IFM) motif to the inactivation-gate receptor. Asp and Ala, in the selectivity motif DEKA, line the walls of the ion-selectivity filter, whereas Glu and Lys are in positions to accept and release Na+ ions via a charge-delocalization network. Arrhythmia mutation sites undergo large translocations during gating, providing a potential mechanism for pathogenic effects. Our results provide detailed insights into Nav1.5 structure, pharmacology, activation, inactivation, ion selectivity, and arrhythmias.


Subject(s)
NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , NAV1.5 Voltage-Gated Sodium Channel/ultrastructure , Animals , Cell Line , HEK293 Cells , Heart/physiology , Humans , Ion Channel Gating/physiology , Membrane Potentials/physiology , Patch-Clamp Techniques/methods , Rats , Sodium/metabolism , Sodium Channels/chemistry , Structure-Activity Relationship , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/ultrastructure
18.
Nature ; 557(7706): 590-594, 2018 05.
Article in English | MEDLINE | ID: mdl-29769724

ABSTRACT

Potassium-sensitive hypokalaemic and normokalaemic periodic paralysis are inherited skeletal muscle diseases characterized by episodes of flaccid muscle weakness1,2. They are caused by single mutations in positively charged residues ('gating charges') in the S4 transmembrane segment of the voltage sensor of the voltage-gated sodium channel Nav1.4 or the calcium channel Cav1.11,2. Mutations of the outermost gating charges (R1 and R2) cause hypokalaemic periodic paralysis1,2 by creating a pathogenic gating pore in the voltage sensor through which cations leak in the resting state3,4. Mutations of the third gating charge (R3) cause normokalaemic periodic paralysis 5 owing to cation leak in both activated and inactivated states 6 . Here we present high-resolution structures of the model bacterial sodium channel NavAb with the analogous gating-charge mutations7,8, which have similar functional effects as in the human channels. The R2G and R3G mutations have no effect on the backbone structures of the voltage sensor, but they create an aqueous cavity near the hydrophobic constriction site that controls gating charge movement through the voltage sensor. The R3G mutation extends the extracellular aqueous cleft through the entire length of the activated voltage sensor, creating an aqueous path through the membrane. Conversely, molecular modelling shows that the R2G mutation creates a continuous aqueous path through the membrane only in the resting state. Crystal structures of NavAb(R2G) in complex with guanidinium define a potential drug target site. Molecular dynamics simulations illustrate the mechanism of Na+ permeation through the mutant gating pore in concert with conformational fluctuations of the gating charge R4. Our results reveal pathogenic mechanisms of periodic paralysis at the atomic level and suggest designs of drugs that may prevent ionic leak and provide symptomatic relief from hypokalaemic and normokalaemic periodic paralysis.


Subject(s)
Ion Channel Gating , NAV1.4 Voltage-Gated Sodium Channel/chemistry , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Paralyses, Familial Periodic/metabolism , Binding Sites , Electric Conductivity , Guanidine/metabolism , Humans , Hypokalemic Periodic Paralysis/genetics , Hypokalemic Periodic Paralysis/metabolism , Ion Channel Gating/genetics , Molecular Dynamics Simulation , Mutation , NAV1.4 Voltage-Gated Sodium Channel/genetics , Paralyses, Familial Periodic/genetics , Sodium/metabolism , Thermodynamics
19.
Protein Sci ; 24(10): 1560-79, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26234418

ABSTRACT

Major facilitator superfamily (MFS) is a large class of secondary active transporters widely expressed across all life kingdoms. Although a common 12-transmembrane helix-bundle architecture is found in most MFS crystal structures available, a common mechanism of energy coupling remains to be elucidated. Here, we discuss several models for energy-coupling in the transport process of the transporters, largely based on currently available structures and the results of their biochemical analyses. Special attention is paid to the interaction between protonation and the negative-inside membrane potential. Also, functional roles of the conserved sequence motifs are discussed in the context of the 3D structures. We anticipate that in the near future, a unified picture of the functions of MFS transporters will emerge from the insights gained from studies of the common architectures and conserved motifs.


Subject(s)
Energy Metabolism/physiology , Membrane Transport Proteins/metabolism , Models, Biological , Binding Sites , Biological Transport , Crystallography, X-Ray , Membrane Transport Proteins/chemistry , Protein Conformation
20.
Biochem Biophys Res Commun ; 450(2): 929-35, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24952155

ABSTRACT

YajR is an Escherichia coli transporter that belongs to the major facilitator superfamily. Unlike most MFS transporters, YajR contains a carboxyl terminal, cytosolic domain of 67 amino acid residues termed YAM domain. Although it is speculated that the function of this small soluble domain is to regulate the conformational change of the 12-helix transmembrane domain, its precise regulatory role remains unclear. Here, we report the crystal structure of the YAM domain at 1.07-Å resolution, along with its structure determined using nuclear magnetic resonance. Detailed analysis of the high resolution structure revealed a symmetrical dimer in which a belt of well-ordered poly-pentagonal water molecules is embedded. A mutagenesis experiment and a thermal stability assay were used to analyze the putative role of this dimerization in response to changes in halogen concentration.


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Multimerization , Protein Stability , Protein Structure, Tertiary , Sodium Chloride/chemistry , Water/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...