Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.529
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(20): 5138-5150.e12, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34496225

RESUMEN

Many transient receptor potential (TRP) channels respond to diverse stimuli and conditionally conduct small and large cations. Such functional plasticity is presumably enabled by a uniquely dynamic ion selectivity filter that is regulated by physiological agents. What is currently missing is a "photo series" of intermediate structural states that directly address this hypothesis and reveal specific mechanisms behind such dynamic channel regulation. Here, we exploit cryoelectron microscopy (cryo-EM) to visualize conformational transitions of the capsaicin receptor, TRPV1, as a model to understand how dynamic transitions of the selectivity filter in response to algogenic agents, including protons, vanilloid agonists, and peptide toxins, permit permeation by small and large organic cations. These structures also reveal mechanisms governing ligand binding substates, as well as allosteric coupling between key sites that are proximal to the selectivity filter and cytoplasmic gate. These insights suggest a general framework for understanding how TRP channels function as polymodal signal integrators.


Asunto(s)
Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/metabolismo , Regulación Alostérica , Permeabilidad de la Membrana Celular/efectos de los fármacos , Microscopía por Crioelectrón , Diterpenos/farmacología , Células HEK293 , Humanos , Activación del Canal Iónico , Lípidos/química , Meglumina/farmacología , Modelos Moleculares , Unión Proteica , Conformación Proteica , Protones , Canales Catiónicos TRPV/agonistas
2.
Cell ; 184(4): 957-968.e21, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33567265

RESUMEN

Ligand-gated ion channels mediate signal transduction at chemical synapses and transition between resting, open, and desensitized states in response to neurotransmitter binding. Neurotransmitters that produce maximum open channel probabilities (Po) are full agonists, whereas those that yield lower than maximum Po are partial agonists. Cys-loop receptors are an important class of neurotransmitter receptors, yet a structure-based understanding of the mechanism of partial agonist action has proven elusive. Here, we study the glycine receptor with the full agonist glycine and the partial agonists taurine and γ-amino butyric acid (GABA). We use electrophysiology to show how partial agonists populate agonist-bound, closed channel states and cryo-EM reconstructions to illuminate the structures of intermediate, pre-open states, providing insights into previously unseen conformational states along the receptor reaction pathway. We further correlate agonist-induced conformational changes to Po across members of the receptor family, providing a hypothetical mechanism for partial and full agonist action at Cys-loop receptors.


Asunto(s)
Activación del Canal Iónico , Receptores de Glicina/agonistas , Receptores de Glicina/metabolismo , Animales , Sitios de Unión , Línea Celular , Microscopía por Crioelectrón , Glicina , Células HEK293 , Humanos , Imagenología Tridimensional , Maleatos/química , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Neurotransmisores/metabolismo , Dominios Proteicos , Receptores de Glicina/genética , Receptores de Glicina/ultraestructura , Estireno/química , Pez Cebra , Ácido gamma-Aminobutírico/metabolismo
3.
Cell ; 180(3): 440-453.e18, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32032516

RESUMEN

Recognition of microbe-associated molecular patterns (MAMPs) is crucial for the plant's immune response. How this sophisticated perception system can be usefully deployed in roots, continuously exposed to microbes, remains a mystery. By analyzing MAMP receptor expression and response at cellular resolution in Arabidopsis, we observed that differentiated outer cell layers show low expression of pattern-recognition receptors (PRRs) and lack MAMP responsiveness. Yet, these cells can be gated to become responsive by neighbor cell damage. Laser ablation of small cell clusters strongly upregulates PRR expression in their vicinity, and elevated receptor expression is sufficient to induce responsiveness in non-responsive cells. Finally, localized damage also leads to immune responses to otherwise non-immunogenic, beneficial bacteria. Damage-gating is overridden by receptor overexpression, which antagonizes colonization. Our findings that cellular damage can "switch on" local immune responses helps to conceptualize how MAMP perception can be used despite the presence of microbial patterns in the soil.


Asunto(s)
Arabidopsis/inmunología , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Raíces de Plantas/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Arabidopsis/crecimiento & desarrollo , Arabidopsis/microbiología , Arabidopsis/efectos de la radiación , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/efectos de la radiación , Ascorbato Peroxidasas/metabolismo , Ascorbato Peroxidasas/efectos de la radiación , Flagelina/farmacología , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/efectos de la radiación , Terapia por Láser/métodos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/efectos de la radiación , Microscopía Confocal , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/microbiología , Raíces de Plantas/efectos de la radiación , Proteínas Quinasas/metabolismo , Proteínas Quinasas/efectos de la radiación , Receptores de Reconocimiento de Patrones/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Imagen de Lapso de Tiempo
4.
Cell ; 180(1): 122-134.e10, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31866066

RESUMEN

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.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/ultraestructura , Animales , Línea Celular , Células HEK293 , Corazón/fisiología , Humanos , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp/métodos , Ratas , Sodio/metabolismo , Canales de Sodio/química , Relación Estructura-Actividad , Canales de Sodio Activados por Voltaje/metabolismo , Canales de Sodio Activados por Voltaje/ultraestructura
5.
Cell ; 180(2): 340-347.e9, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31883792

RESUMEN

KCNQ1, also known as Kv7.1, is a voltage-dependent K+ channel that regulates gastric acid secretion, salt and glucose homeostasis, and heart rhythm. Its functional properties are regulated in a tissue-specific manner through co-assembly with beta subunits KCNE1-5. In non-excitable cells, KCNQ1 forms a complex with KCNE3, which suppresses channel closure at negative membrane voltages that otherwise would close it. Pore opening is regulated by the signaling lipid PIP2. Using cryoelectron microscopy (cryo-EM), we show that KCNE3 tucks its single-membrane-spanning helix against KCNQ1, at a location that appears to lock the voltage sensor in its depolarized conformation. Without PIP2, the pore remains closed. Upon addition, PIP2 occupies a site on KCNQ1 within the inner membrane leaflet, which triggers a large conformational change that leads to dilation of the pore's gate. It is likely that this mechanism of PIP2 activation is conserved among Kv7 channels.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Canal de Potasio KCNQ1/ultraestructura , Microscopía por Crioelectrón , Humanos , Activación del Canal Iónico/fisiología , Canal de Potasio KCNQ1/química , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio con Entrada de Voltaje/ultraestructura
6.
Cell ; 176(4): 702-715.e14, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661758

RESUMEN

Voltage-gated sodium (Nav) channels are targets of disease mutations, toxins, and therapeutic drugs. Despite recent advances, the structural basis of voltage sensing, electromechanical coupling, and toxin modulation remains ill-defined. Protoxin-II (ProTx2) from the Peruvian green velvet tarantula is an inhibitor cystine-knot peptide and selective antagonist of the human Nav1.7 channel. Here, we visualize ProTx2 in complex with voltage-sensor domain II (VSD2) from Nav1.7 using X-ray crystallography and cryoelectron microscopy. Membrane partitioning orients ProTx2 for unfettered access to VSD2, where ProTx2 interrogates distinct features of the Nav1.7 receptor site. ProTx2 positions two basic residues into the extracellular vestibule to antagonize S4 gating-charge movement through an electrostatic mechanism. ProTx2 has trapped activated and deactivated states of VSD2, revealing a remarkable ∼10 Å translation of the S4 helix, providing a structural framework for activation gating in voltage-gated ion channels. Finally, our results deliver key templates to design selective Nav channel antagonists.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/ultraestructura , Péptidos/metabolismo , Venenos de Araña/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Cricetulus , Microscopía por Crioelectrón/métodos , Cristalografía por Rayos X/métodos , Células HEK293 , Humanos , Activación del Canal Iónico , Péptidos/toxicidad , Dominios Proteicos , Venenos de Araña/toxicidad , Arañas , Bloqueadores del Canal de Sodio Activado por Voltaje , Canales de Sodio Activados por Voltaje/metabolismo
7.
Cell ; 179(7): 1582-1589.e7, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31787376

RESUMEN

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


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Activación del Canal Iónico , Animales , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Potenciales de la Membrana , Conformación Proteica en Hélice alfa , Células Sf9 , Spodoptera
8.
Cell ; 178(4): 993-1003.e12, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31353218

RESUMEN

Voltage-gated sodium (NaV) channels initiate action potentials in nerve, muscle, and other electrically excitable cells. The structural basis of voltage gating is uncertain because the resting state exists only at deeply negative membrane potentials. To stabilize the resting conformation, we inserted voltage-shifting mutations and introduced a disulfide crosslink in the VS of the ancestral bacterial sodium channel NaVAb. Here, we present a cryo-EM structure of the resting state and a complete voltage-dependent gating mechanism. The S4 segment of the VS is drawn intracellularly, with three gating charges passing through the transmembrane electric field. This movement forms an elbow connecting S4 to the S4-S5 linker, tightens the collar around the S6 activation gate, and prevents its opening. Our structure supports the classical "sliding helix" mechanism of voltage sensing and provides a complete gating mechanism for voltage sensor function, pore opening, and activation-gate closure based on high-resolution structures of a single sodium channel protein.


Asunto(s)
Potenciales de Acción/fisiología , Membrana Externa Bacteriana/metabolismo , Escherichia coli/metabolismo , Activación del Canal Iónico/fisiología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Línea Celular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Mutación , Conformación Proteica en Hélice alfa , Sodio/metabolismo , Spodoptera/citología , Canales de Sodio Activados por Voltaje/química
9.
Cell ; 168(3): 390-399.e11, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28111072

RESUMEN

The stable structural conformations that occur along the complete reaction coordinate for ion channel opening have never been observed. In this study, we describe the equilibrium ensemble of structures of Slo2.2, a neuronal Na+-activated K+ channel, as a function of the Na+ concentration. We find that Slo2.2 exists in multiple closed conformations whose relative occupancies are independent of Na+ concentration. An open conformation emerges from an ensemble of closed conformations in a highly Na+-dependent manner, without evidence of Na+-dependent intermediates. In other words, channel opening is a highly concerted, switch-like process. The midpoint of the structural titration matches that of the functional titration. A maximum open conformation probability approaching 1.0 and maximum functional open probability approaching 0.7 imply that, within the class of open channels, there is a subclass that is not permeable to ions.


Asunto(s)
Proteínas Aviares/química , Pollos/metabolismo , Proteínas del Tejido Nervioso/química , Canales de Potasio/química , Animales , Proteínas Aviares/metabolismo , Microscopía por Crioelectrón , Células HEK293 , Humanos , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio/metabolismo , Conformación Proteica , Sodio/química
10.
Cell ; 169(3): 422-430.e10, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431243

RESUMEN

The human ether-à-go-go-related potassium channel (hERG, Kv11.1) is a voltage-dependent channel known for its role in repolarizing the cardiac action potential. hERG alteration by mutation or pharmacological inhibition produces Long QT syndrome and the lethal cardiac arrhythmia torsade de pointes. We have determined the molecular structure of hERG to 3.8 Å using cryo-electron microscopy. In this structure, the voltage sensors adopt a depolarized conformation, and the pore is open. The central cavity has an atypically small central volume surrounded by four deep hydrophobic pockets, which may explain hERG's unusual sensitivity to many drugs. A subtle structural feature of the hERG selectivity filter might correlate with its fast inactivation rate, which is key to hERG's role in cardiac action potential repolarization.


Asunto(s)
Canal de Potasio ERG1/química , Canal de Potasio ERG1/ultraestructura , Secuencia de Aminoácidos , Microscopía por Crioelectrón , Canal de Potasio ERG1/aislamiento & purificación , Canal de Potasio ERG1/metabolismo , Humanos , Modelos Moleculares , Conformación Proteica , Alineación de Secuencia
11.
Cell ; 170(6): 1234-1246.e14, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28823560

RESUMEN

AMPA receptors mediate fast excitatory neurotransmission in the mammalian brain and transduce the binding of presynaptically released glutamate to the opening of a transmembrane cation channel. Within the postsynaptic density, however, AMPA receptors coassemble with transmembrane AMPA receptor regulatory proteins (TARPs), yielding a receptor complex with altered gating kinetics, pharmacology, and pore properties. Here, we elucidate structures of the GluA2-TARP γ2 complex in the presence of the partial agonist kainate or the full agonist quisqualate together with a positive allosteric modulator or with quisqualate alone. We show how TARPs sculpt the ligand-binding domain gating ring, enhancing kainate potency and diminishing the ensemble of desensitized states. TARPs encircle the receptor ion channel, stabilizing M2 helices and pore loops, illustrating how TARPs alter receptor pore properties. Structural and computational analysis suggests the full agonist and modulator complex harbors an ion-permeable channel gate, providing the first view of an activated AMPA receptor.


Asunto(s)
Canales de Calcio/química , Receptores AMPA/química , Animales , Microscopía por Crioelectrón , Agonistas de Aminoácidos Excitadores/química , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/química , Ácido Kaínico/farmacología , Modelos Moleculares , Ácido Quiscuálico/química , Ácido Quiscuálico/farmacología , Ratas , Receptores AMPA/agonistas
12.
Cell ; 168(1-2): 111-120.e11, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28086084

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels underlie the control of rhythmic activity in cardiac and neuronal pacemaker cells. In HCN, the polarity of voltage dependence is uniquely reversed. Intracellular cyclic adenosine monophosphate (cAMP) levels tune the voltage response, enabling sympathetic nerve stimulation to increase the heart rate. We present cryo-electron microscopy structures of the human HCN channel in the absence and presence of cAMP at 3.5 Å resolution. HCN channels contain a K+ channel selectivity filter-forming sequence from which the amino acids create a unique structure that explains Na+ and K+ permeability. The voltage sensor adopts a depolarized conformation, and the pore is closed. An S4 helix of unprecedented length extends into the cytoplasm, contacts the C-linker, and twists the inner helical gate shut. cAMP binding rotates cytoplasmic domains to favor opening of the inner helical gate. These structures advance understanding of ion selectivity, reversed polarity gating, and cAMP regulation in HCN channels.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Canales de Potasio/química , Secuencia de Aminoácidos , Microscopía por Crioelectrón/métodos , AMP Cíclico/química , AMP Cíclico/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Modelos Moleculares , Canales de Potasio/metabolismo , Alineación de Secuencia
13.
Cell ; 167(1): 145-157.e17, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27662087

RESUMEN

The type-1 ryanodine receptor (RyR1) is an intracellular calcium (Ca(2+)) release channel required for skeletal muscle contraction. Here, we present cryo-EM reconstructions of RyR1 in multiple functional states revealing the structural basis of channel gating and ligand-dependent activation. Binding sites for the channel activators Ca(2+), ATP, and caffeine were identified at interdomain interfaces of the C-terminal domain. Either ATP or Ca(2+) alone induces conformational changes in the cytoplasmic assembly ("priming"), without pore dilation. In contrast, in the presence of all three activating ligands, high-resolution reconstructions of open and closed states of RyR1 were obtained from the same sample, enabling analyses of conformational changes associated with gating. Gating involves global conformational changes in the cytosolic assembly accompanied by local changes in the transmembrane domain, which include bending of the S6 transmembrane segment and consequent pore dilation, displacement, and deformation of the S4-S5 linker and conformational changes in the pseudo-voltage-sensor domain.


Asunto(s)
Agonistas de los Canales de Calcio/química , Activación del Canal Iónico , Contracción Muscular , Canal Liberador de Calcio Receptor de Rianodina/química , Animales , Sitios de Unión , Cafeína/química , Calcio/química , Microscopía por Crioelectrón , Ligandos , Dominios Proteicos , Conejos , Proteínas de Unión a Tacrolimus/química
14.
Mol Cell ; 83(24): 4555-4569.e4, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38035882

RESUMEN

Modulation of large conductance intracellular ligand-activated potassium (BK) channel family (Slo1-3) by auxiliary subunits allows diverse physiological functions in excitable and non-excitable cells. Cryoelectron microscopy (cryo-EM) structures of voltage-gated potassium (Kv) channel complexes have provided insights into how voltage sensitivity is modulated by auxiliary subunits. However, the modulation mechanisms of BK channels, particularly as ligand-activated ion channels, remain unknown. Slo1 is a Ca2+-activated and voltage-gated BK channel and is expressed in neurons, muscle cells, and epithelial cells. Using cryo-EM and electrophysiology, we show that the LRRC26-γ1 subunit modulates not only voltage but also Ca2+ sensitivity of Homo sapiens Slo1. LRRC26 stabilizes the active conformation of voltage-senor domains of Slo1 by an extracellularly S4-locking mechanism. Furthermore, it also stabilizes the active conformation of Ca2+-sensor domains of Slo1 intracellularly, which is functionally equivalent to intracellular Ca2+ in the activation of Slo1. Such a dual allosteric modulatory mechanism may be general in regulating the intracellular ligand-activated BK channel complexes.


Asunto(s)
Calcio , Canales de Potasio de Gran Conductancia Activados por el Calcio , Humanos , Calcio/metabolismo , Microscopía por Crioelectrón , Activación del Canal Iónico/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ligandos , Potasio , Regulación Alostérica
15.
Mol Cell ; 82(13): 2427-2442.e4, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35597238

RESUMEN

The voltage-gated ion channel activity depends on both activation (transition from the resting state to the open state) and inactivation. Inactivation is a self-restraint mechanism to limit ion conduction and is as crucial to membrane excitability as activation. Inactivation can occur when the channel is open or closed. Although open-state inactivation is well understood, the molecular basis of closed-state inactivation has remained elusive. We report cryo-EM structures of human KV4.2 channel complexes in inactivated, open, and closed states. Closed-state inactivation of KV4 involves an unprecedented symmetry breakdown for pore closure by only two of the four S4-S5 linkers, distinct from known mechanisms of open-state inactivation. We further capture KV4 in a putative resting state, revealing how voltage sensor movements control the pore. Moreover, our structures provide insights regarding channel modulation by KChIP2 and DPP6 auxiliary subunits. Our findings elucidate mechanisms of closed-state inactivation and voltage-dependent activation of the KV4 channel.


Asunto(s)
Activación del Canal Iónico , Canales de Potasio Shal , Humanos , Activación del Canal Iónico/fisiología , Cinética , Potenciales de la Membrana/fisiología , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo
16.
Physiol Rev ; 102(3): 1159-1210, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34927454

RESUMEN

Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom, as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive CaV1.2 and CaV1.3 channels to obligatory dimeric assembly and gating of voltage-gated NaV1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine-tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pacemaking activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences, and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.


Asunto(s)
Activación del Canal Iónico , Canal Liberador de Calcio Receptor de Rianodina , Potenciales de Acción , Humanos , Activación del Canal Iónico/fisiología , Neuronas
17.
Mol Cell ; 81(23): 4771-4783.e7, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34678168

RESUMEN

AMPA receptors (AMPARs) mediate the majority of excitatory neurotransmission. Their surface expression, trafficking, gating, and pharmacology are regulated by auxiliary subunits. Of the two types of TARP auxiliary subunits, type I TARPs assume activating roles, while type II TARPs serve suppressive functions. We present cryo-EM structures of GluA2 AMPAR in complex with type II TARP γ5, which reduces steady-state currents, increases single-channel conductance, and slows recovery from desensitization. Regulation of AMPAR function depends on its ligand-binding domain (LBD) interaction with the γ5 head domain. GluA2-γ5 complex shows maximum stoichiometry of two TARPs per AMPAR tetramer, being different from type I TARPs but reminiscent of the auxiliary subunit GSG1L. Desensitization of both GluA2-GSG1L and GluA2-γ5 complexes is accompanied by rupture of LBD dimer interface, while GluA2-γ5 but not GluA2-GSG1L LBD dimers remain two-fold symmetric. Different structural architectures and desensitization mechanisms of complexes with auxiliary subunits endow AMPARs with broad functional capabilities.


Asunto(s)
Canales de Calcio/química , Claudinas/química , Receptores AMPA/química , Secuencias de Aminoácidos , Animales , Microscopía por Crioelectrón , Dimerización , Células HEK293 , Humanos , Procesamiento de Imagen Asistido por Computador , Membrana Dobles de Lípidos/química , Proteínas de la Membrana , Conformación Molecular , Técnicas de Placa-Clamp , Polímeros , Unión Proteica , Conformación Proteica , Dominios Proteicos , Ratas , Transmisión Sináptica
18.
Mol Cell ; 81(14): 2929-2943.e6, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34166608

RESUMEN

The HCN1-4 channel family is responsible for the hyperpolarization-activated cation current If/Ih that controls automaticity in cardiac and neuronal pacemaker cells. We present cryoelectron microscopy (cryo-EM) structures of HCN4 in the presence or absence of bound cAMP, displaying the pore domain in closed and open conformations. Analysis of cAMP-bound and -unbound structures sheds light on how ligand-induced transitions in the channel cytosolic portion mediate the effect of cAMP on channel gating and highlights the regulatory role of a Mg2+ coordination site formed between the C-linker and the S4-S5 linker. Comparison of open/closed pore states shows that the cytosolic gate opens through concerted movements of the S5 and S6 transmembrane helices. Furthermore, in combination with molecular dynamics analyses, the open pore structures provide insights into the mechanisms of K+/Na+ permeation. Our results contribute mechanistic understanding on HCN channel gating, cyclic nucleotide-dependent modulation, and ion permeation.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Activación del Canal Iónico/fisiología , Iones/metabolismo , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Línea Celular , Microscopía por Crioelectrón/métodos , AMP Cíclico/metabolismo , Células HEK293 , Humanos
19.
Mol Cell ; 81(1): 38-48.e4, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33232657

RESUMEN

Voltage-gated sodium channels initiate electrical signals and are frequently targeted by deadly gating-modifier neurotoxins, including tarantula toxins, which trap the voltage sensor in its resting state. The structural basis for tarantula-toxin action remains elusive because of the difficulty of capturing the functionally relevant form of the toxin-channel complex. Here, we engineered the model sodium channel NaVAb with voltage-shifting mutations and the toxin-binding site of human NaV1.7, an attractive pain target. This mutant chimera enabled us to determine the cryoelectron microscopy (cryo-EM) structure of the channel functionally arrested by tarantula toxin. Our structure reveals a high-affinity resting-state-specific toxin-channel interaction between a key lysine residue that serves as a "stinger" and penetrates a triad of carboxyl groups in the S3-S4 linker of the voltage sensor. By unveiling this high-affinity binding mode, our studies establish a high-resolution channel-docking and resting-state locking mechanism for huwentoxin-IV and provide guidance for developing future resting-state-targeted analgesic drugs.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/química , Venenos de Araña/química , Sustitución de Aminoácidos , Animales , Humanos , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.7/genética , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Células Sf9 , Spodoptera
20.
Mol Cell ; 81(22): 4650-4662.e4, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34715014

RESUMEN

Mutations in ATP13A2, also known as PARK9, cause a rare monogenic form of juvenile-onset Parkinson's disease named Kufor-Rakeb syndrome and other neurodegenerative diseases. ATP13A2 encodes a neuroprotective P5B P-type ATPase highly enriched in the brain that mediates selective import of spermine ions from lysosomes into the cytosol via an unknown mechanism. Here we present three structures of human ATP13A2 bound to an ATP analog or to spermine in the presence of phosphomimetics determined by cryoelectron microscopy. ATP13A2 autophosphorylation opens a lysosome luminal gate to reveal a narrow lumen access channel that holds a spermine ion in its entrance. ATP13A2's architecture suggests physical principles underlying selective polyamine transport and anticipates a "pump-channel" intermediate that could function as a counter-cation conduit to facilitate lysosome acidification. Our findings establish a firm foundation to understand ATP13A2 mutations associated with disease and bring us closer to realizing ATP13A2's potential in neuroprotective therapy.


Asunto(s)
Encéfalo/metabolismo , Poliaminas/química , ATPasas de Translocación de Protón/química , ATPasas de Translocación de Protón/genética , Sitio Alostérico , Sitios de Unión , Transporte Biológico , Microscopía por Crioelectrón , Humanos , Iones/química , Lisosomas/química , Mutación , Fosforilación , Dominios Proteicos , Proteínas Recombinantes/química , Espermina/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA