fast inactivation

快速失活
  • 文章类型: Journal Article
    SCN2A基因相关的早期婴儿发育性脑病和癫痫性脑病(EI-DEE)是一种罕见且严重的疾病,表现在婴儿早期。影响快速失活门控机制的SCN2A突变可导致电压依赖性改变和编码的神经元Nav1.2通道的不完全失活,并导致异常的神经元兴奋性。在这项研究中,我们评估了与DEE相关的七个错义Nav1.2变体的临床数据,并进行了分子动力学模拟,膜片钳电生理学,和动态钳夹实时神经元建模,以阐明突变的分子和神经元尺度表型后果。N1662D突变几乎完全阻止了快速失活而不影响活化。野生型和N1662D通道结构的比较表明,残基N1662和Q1494之间的双功能氢键形成对于快速失活至关重要。快速失活也可以防止工程Q1494A或Q1494LNav1.2通道变种,而Q1494E或Q1494K变异导致不完全失活和持续电流。分子动力学模拟显示疏水性IFM-基序相对于野生型具有N1662D和Q1494L变体对其受体位点的亲和力降低。这些结果表明,N1662和Q1494之间的相互作用支持失活门的稳定性和取向,并且对于快速失活的发展至关重要。六个DEE相关的Nav1.2变体,还评估了映射到已知与快速失活有关的通道片段的突变。值得注意的是,L1657P变体还阻止了快速失活,并产生了与N1662D相似的生物物理特征,而M1501V,M1501T,F1651C,P1658S,和A1659V变体产生的生物物理特性与动态动作电位钳制实验中混合神经元的功能获得和增强的动作电位放电一致。矛盾的是,低密度N1662D或L1657P电流增强动作电位激发,而密度增加导致持续的去极化。我们的结果为Nav1.2通道快速失活的分子机制提供了新的结构见解,并为SCN2A相关EI-DEE的治疗策略提供了信息。非失活Nav1.2通道对神经元兴奋性的贡献可能构成SCN2A相关DEE发病机理中的独特细胞机制。
    SCN2A gene-related early-infantile developmental and epileptic encephalopathy (EI-DEE) is a rare and severe disorder that manifests in early infancy. SCN2A mutations affecting the fast inactivation gating mechanism can result in altered voltage dependence and incomplete inactivation of the encoded neuronal Nav1.2 channel and lead to abnormal neuronal excitability. In this study, we evaluated clinical data of seven missense Nav1.2 variants associated with DEE and performed molecular dynamics simulations, patch-clamp electrophysiology, and dynamic clamp real-time neuronal modelling to elucidate the molecular and neuron-scale phenotypic consequences of the mutations. The N1662D mutation almost completely prevented fast inactivation without affecting activation. The comparison of wild-type and N1662D channel structures suggested that the ambifunctional hydrogen bond formation between residues N1662 and Q1494 is essential for fast inactivation. Fast inactivation could also be prevented with engineered Q1494A or Q1494L Nav1.2 channel variants, whereas Q1494E or Q1494 K variants resulted in incomplete inactivation and persistent current. Molecular dynamics simulations revealed a reduced affinity of the hydrophobic IFM-motif to its receptor site with N1662D and Q1494L variants relative to wild-type. These results demonstrate that the interactions between N1662 and Q1494 underpin the stability and the orientation of the inactivation gate and are essential for the development of fast inactivation. Six DEE-associated Nav1.2 variants, with mutations mapped to channel segments known to be implicated in fast inactivation were also evaluated. Remarkably, the L1657P variant also prevented fast inactivation and produced biophysical characteristics that were similar to those of N1662D, whereas the M1501 V, M1501T, F1651C, P1658S, and A1659 V variants resulted in biophysical properties that were consistent with gain-of-function and enhanced action potential firing of hybrid neurons in dynamic action potential clamp experiments. Paradoxically, low densities of N1662D or L1657P currents potentiated action potential firing, whereas increased densities resulted in sustained depolarization. Our results provide novel structural insights into the molecular mechanism of Nav1.2 channel fast inactivation and inform treatment strategies for SCN2A-related EI-DEE. The contribution of non-inactivating Nav1.2 channels to neuronal excitability may constitute a distinct cellular mechanism in the pathogenesis of SCN2A-related DEE.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Nav1.5是心肌中发现的主要电压门控钠通道,它促进Na+离子快速流入细胞膜,导致心脏动作电位的快速去极化阶段0。因此,它在确定心脏冲动的幅度和上冲程速度中起着重要作用。定量地,心脏钠通道在不到一毫秒的时间内激活以触发心脏动作电位,并在2-3ms内失活以促进复极化并返回静息状态,为激发下一个动作电位做准备。编码Nav1.5(SCN5A)的基因中的错义突变,改变这些时间常数,导致广泛的心脏疾病,从长QT综合征3型(LQT3)到心脏猝死。在这篇小型评论中,我将重点关注Nav1.5失活门中导致心律失常的错义突变,试图将错义突变的位置与其特定表型相关联。
    Nav1.5 is the main voltage-gated sodium channel found in cardiac muscle, where it facilitates the fast influx of Na+ ions across the cell membrane, resulting in the fast depolarization phase-phase 0 of the cardiac action potential. As a result, it plays a major role in determining the amplitude and the upstroke velocity of the cardiac impulse. Quantitively, cardiac sodium channel activates in less than a millisecond to trigger the cardiac action potential and inactivates within 2-3 ms to facilitate repolarization and return to the resting state in preparation for firing the next action potential. Missense mutations in the gene that encodes Nav1.5 (SCN5A), change these time constants which leads to a wide spectrum of cardiac diseases ranging from long QT syndrome type 3 (LQT3) to sudden cardiac death. In this mini-review I will focus on the missense mutations in the inactivation gate of Nav1.5 that results in arrhythmia, attempting to correlate the location of the missense mutation to their specific phenotype.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Nav1.7由于其在痛觉中的关键作用而代表了下一代镇痛药的重要目标。在这里,我们报告了与β1和β2亚基复合的野生型(WT)Nav1.7的2.2_分辨率低温EM结构,该结构揭示了几个以前无法识别的胞浆片段。我们报告的Nav1.7(E406K)与各种毒素结合的结构的低温EM数据的再处理鉴定了S6IV的两种不同构象,一个仅由α螺旋圈组成,另一个在中间包含π螺旋圈。无配体的结构Nav1.7(E406K),在3.5-贝达分辨率下确定,与WT通道相同,确认HuwentoxinIV或原毒素II与VSDII的结合变构诱导了S6IV的α→π转变。局部的二级结构转变导致细胞内门收缩,重复I和IV的界面上的开窗闭合,以及快速失活基序的结合位点的重排。
    Nav1.7 represents a preeminent target for next-generation analgesics for its critical role in pain sensation. Here we report a 2.2-Å resolution cryo-EM structure of wild-type (WT) Nav1.7 complexed with the β1 and β2 subunits that reveals several previously indiscernible cytosolic segments. Reprocessing of the cryo-EM data for our reported structures of Nav1.7(E406K) bound to various toxins identifies two distinct conformations of S6IV, one composed of α helical turns only and the other containing a π helical turn in the middle. The structure of ligand-free Nav1.7(E406K), determined at 3.5-Å resolution, is identical to the WT channel, confirming that binding of Huwentoxin IV or Protoxin II to VSDII allosterically induces the α → π transition of S6IV. The local secondary structural shift leads to contraction of the intracellular gate, closure of the fenestration on the interface of repeats I and IV, and rearrangement of the binding site for the fast inactivation motif.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    有害的疼痛信号在周围神经系统中被转换为动作电位,依赖于电压门控钠通道(Navs)的活性。因此,阻断NaV是疼痛治疗的有价值的策略。这里,我们报道了一种新型NaVs拮抗剂的表征,2-(2-(二乙基氨基)乙基)茚并[1,2,3-de]萘嗪-3(2H)-酮(C65780),并调查其作用机制。C65780抑制静止的NaV1.7,NaV1.8和NaV1.9通道,IC50为11.3±0.4μM,2.7±0.3μM和19.2±2.3μM,分别。机制分析显示,C65780迅速结合到其在NaV1.7中的高亲和力受体位点,如快速失活过程所形成的,并使通道稳定在缓慢恢复的状态,它通过在超极化方向上改变它们的失活-电压关系来促进NaV1.7通道失活,增加失活通道的高原比例,削弱了他们依赖时间的恢复。然而,NaV1.7的缓慢失活,不参与C65780的操作。在DRG神经元中,C65780还抑制了NaVs的活性,从而抑制神经元的兴奋性.这些效果表现为口服C65780在各种疼痛模型中的广泛功效,疗效与抗抑郁药/神经性疼痛药物阿米替林相当。令人兴奋的是,与阿米替林相比,C65780对相关的NaV1.4和NaV1.5通道的失活状态抑制较弱,在缓解疼痛剂量的小鼠中,在强迫游泳试验中没有观察到毒性或运动抑制。这些结果表明,C65780通过捕获处于失活和缓慢恢复状态的NaV来产生疼痛缓解,并且可以代表用于开发镇痛药的优异起始化合物。
    Noxious pain signals are transduced in the peripheral nervous system as action potentials, which rely on the activities of voltage-gated sodium channels (NaVs). Blocking NaVs is thus a valuable strategy for pain treatment. Here, we report the characterization of a novel NaVs antagonist, 2-(2-(diethylamino)ethyl)indeno[1,2,3-de]phthalazin-3(2H)-one (C65780), and investigation of its action mechanisms. C65780 inhibited the resting NaV1.7, NaV1.8, and NaV1.9 channels with IC50s of 11.3 ± 0.4 μM, 2.7 ± 0.3 μM and 19.2 ± 2.3 μM, respectively. Mechanistic analysis revealed that C65780 quickly bound to its high-affinity receptor site in NaV1.7 as formed by the fast inactivation process and stabilized the channels in a slowly recovering state, for which it facilitated NaV1.7 channels\' inactivation by shifting their inactivation-voltage relationship in the hyperpolarizing direction, increasing the plateau proportion of inactivated channels, and blunting their time-dependent recovery. The slow inactivation of NaV1.7, however, is not involved in the action of C65780. In DRG neurons, C65780 also inhibited activity of NaVs, thus dampening neuronal excitability. These effects parlayed into a broad efficacy of orally administrated C65780 in various models of pain, with an efficacy comparable to the antidepressant/neuropathic pain drug Amitriptyline. Excitingly, C65780 demonstrated weaker inactivated state inhibition of related NaV1.4 and NaV1.5 channels compared to amitriptyline, and no toxicity or inhibition of locomotion in a forced-swimming test was observed in mice at pain-relieving doses. These results demonstrate that C65780 acts by trapping NaVs in the inactivated and slowly-recovering state to produce pain relief and may represent an excellent starting compound for developing analgesics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Venomous animals have evolved to produce peptide toxins that modulate the activity of voltage-gated sodium (Nav) channels. These specific modulators are powerful probes for investigating the structural and functional features of Nav channels. Here, we report the isolation and characterization of δ-theraphotoxin-Gr4b (Gr4b), a novel peptide toxin from the venom of the spider Grammostola rosea. Gr4b contains 37-amino acid residues with six cysteines forming three disulfide bonds. Patch-clamp analysis confirmed that Gr4b markedly slows the fast inactivation of Nav1.9 and inhibits the currents of Nav1.4 and Nav1.7, but does not affect Nav1.8. It was also found that Gr4b significantly shifts the steady-state activation and inactivation curves of Nav1.9 to the depolarization direction and increases the window current, which is consistent with the change in the ramp current. Furthermore, analysis of Nav1.9/Nav1.8 chimeric channels revealed that Gr4b preferentially binds to the voltage-sensor of domain III (DIII VSD) and has additional interactions with the DIV VSD. The site-directed mutagenesis analysis indicated that N1139 and L1143 in DIII S3-S4 linker participate in toxin binding. In sum, this study reports a novel spider peptide toxin that may slow the fast inactivation of Nav1.9 by binding to the new neurotoxin receptor site-DIII VSD. Taken together, these findings provide insight into the functional role of the Nav channel DIII VSD in fast inactivation and activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    从电压传感器到失活门的运动传输是离子通道一般生理学中的重要问题。在通道hNav1.5的低温EM结构中,胞外环IVP2-S6中的残基N1736和R1739分别接近谷氨酸E1225和E1295,在电压传感域III(VSD-III)中。ClinVar报告的变体E1230K,E1295K,和R1739W/Q和其他变体在环IVP2-S6,IIIS1-S2和IIIS3-S4与心律失常有关,强调IVP2-S6和VSD-III之间的接口是疾病突变的热点。由这些突变引起的通道功能障碍的原子机制是未知的。这里,我们产生了突变体E1295R,R1739E,E1295R/R1739E,和N1736R,在HEK-293T细胞中表达,并探索了生物物理特性。突变E1295R减少了稳态快速失活和增强的稳态缓慢失活。相比之下,突变R1739E轻微增强快速失活和减弱缓慢失活。双突变体E1295R/R1739E的特征与野生型通道的特征相当相似。突变N1736R减弱缓慢失活。分子建模预测了R1739E与激活的电压感应螺旋IIIS4中最外层赖氨酸的盐桥接。相比之下,功能缺失替代E1295R排斥R1739,从而使激活的VSD-III不稳定,这与我们的E1295R引起G-V曲线去极化偏移的数据一致.在约束维持的盐桥E1295-R1739下,VSD-III的计算机模拟失活导致以下变化:1)IIIS4和IVS5之间的接触被切换;2)接头-螺旋IIIS4-S5与IVS5,IVS6的接触和快速失活三肽IFM被修饰;3)IFM三肽与螺旋IVS5和IVS6的接触有助于缓慢的IVS6中的螺旋变化。停用的VSD-III中的盐桥E1295-R1739的可能性得到了泊松-玻尔兹曼计算和循环IVP2-S6的状态相关能量学的支持。一起来看,我们的结果表明,IVP2-S6环路参与了从VSD-III到失活门的运动传递.
    Motion transmission from voltage sensors to inactivation gates is an important problem in the general physiology of ion channels. In a cryo-EM structure of channel hNav1.5, residues N1736 and R1739 in the extracellular loop IVP2-S6 approach glutamates E1225 and E1295, respectively, in the voltage-sensing domain III (VSD-III). ClinVar-reported variants E1230K, E1295K, and R1739W/Q and other variants in loops IVP2-S6, IIIS1-S2, and IIIS3-S4 are associated with cardiac arrhythmias, highlighting the interface between IVP2-S6 and VSD-III as a hot spot of disease mutations. Atomic mechanisms of the channel dysfunction caused by these mutations are unknown. Here, we generated mutants E1295R, R1739E, E1295R/R1739E, and N1736R, expressed them in HEK-293T cells, and explored biophysical properties. Mutation E1295R reduced steady-state fast inactivation and enhanced steady-state slow inactivation. In contrast, mutation R1739E slightly enhanced fast inactivation and attenuated slow inactivation. Characteristics of the double mutant E1295R/R1739E were rather similar to those of the wild-type channel. Mutation N1736R attenuated slow inactivation. Molecular modeling predicted salt bridging of R1739E with the outermost lysine in the activated voltage-sensing helix IIIS4. In contrast, the loss-of-function substitution E1295R repelled R1739, thus destabilizing the activated VSD-III in agreement with our data that E1295R caused a depolarizing shift of the G-V curve. In silico deactivation of VSD-III with constraint-maintained salt bridge E1295-R1739 resulted in the following changes: 1) contacts between IIIS4 and IVS5 were switched; 2) contacts of the linker-helix IIIS4-S5 with IVS5, IVS6, and fast inactivation tripeptide IFM were modified; 3) contacts of the IFM tripeptide with helices IVS5 and IVS6 were altered; 4) mobile loop IVP2-S6 shifted helix IVP2 that contributes to the slow inactivation gate and helix IVS6 that contributes to the fast inactivation gate. The likelihood of salt bridge E1295-R1739 in deactivated VSD-III is supported by Poisson-Boltzmann calculations and state-dependent energetics of loop IVP2-S6. Taken together, our results suggest that loop IVP2-S6 is involved in motion transmission from VSD-III to the inactivation gates.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Cannabidiol (CBD), one of the cannabinoids from the cannabis plant, can relieve the myotonia resulting from sodium channelopathy, which manifests as repetitive discharges of muscle membrane. We investigated the binding kinetics of CBD to Nav1.4 channels on the muscle membrane. The binding affinity of CBD to the channel was evaluated using whole-cell recording. The CDOCKER program was employed to model CBD docking onto the Nav1.4 channel to determine its binding sites. Our results revealed no differential inhibition of sodium current by CBD when the channels were in activation or fast inactivation status. However, differential inhibition was observed with a dose-dependent manner after a prolonged period of depolarization, leaving the channel in a slow-inactivated state. Moreover, CBD binds selectively to the slow-inactivated state with a significantly faster binding kinetics (>64,000 M-1 s-1) and a higher affinity (Kd of fast inactivation vs. slow-inactivation: >117.42 μM vs. 51.48 μM), compared to the fast inactivation state. Five proposed CBD binding sites in a bundle crossing region of the Nav1.4 channels pore was identified as Val793, Leu794, Phe797, and Cys759 in domain I/S6, and Ile1279 in domain II/S6. Our findings imply that CBD favorably binds to the Nav1.4 channel in its slow-inactivated state.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    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.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Among the nine subtypes of human voltage-gated sodium (Nav) channels, the brain and cardiac isoforms, Nav1.1 and Nav1.5, each carry more than 400 missense mutations respectively associated with epilepsy and cardiac disorders. High-resolution structures are required for structure-function relationship dissection of the disease variants. We report the cryo-EM structures of the full-length human Nav1.1-β4 complex at 3.3 Å resolution here and the Nav1.5-E1784K variant in the accompanying paper. Up to 341 and 261 disease-related missense mutations in Nav1.1 and Nav1.5, respectively, are resolved. Comparative structural analysis reveals several clusters of disease mutations that are common to both Nav1.1 and Nav1.5. Among these, the majority of mutations on the extracellular loops above the pore domain and the supporting segments for the selectivity filter may impair structural integrity, while those on the pore domain and the voltage-sensing domains mostly interfere with electromechanical coupling and fast inactivation. Our systematic structural delineation of these mutations provides important insight into their pathogenic mechanism, which will facilitate the development of precise therapeutic interventions against various sodium channelopathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Nav1.5 is the primary voltage-gated Na+ (Nav) channel in the heart. Mutations of Nav1.5 are associated with various cardiac disorders exemplified by the type 3 long QT syndrome (LQT3) and Brugada syndrome (BrS). E1784K is a common mutation that has been found in both LQT3 and BrS patients. Here we present the cryo-EM structure of the human Nav1.5-E1784K variant at an overall resolution of 3.3 Å. The structure is nearly identical to that of the wild-type human Nav1.5 bound to quinidine. Structural mapping of 91- and 178-point mutations that are respectively associated with LQT3 and BrS reveals a unique distribution pattern for LQT3 mutations. Whereas the BrS mutations spread evenly on the structure, LQT3 mutations are clustered mainly to the segments in repeats III and IV that are involved in gating, voltage-sensing, and particularly inactivation. A mutational hotspot involving the fast inactivation segments is identified and can be mechanistically interpreted by our \"door wedge\" model for fast inactivation. The structural analysis presented here, with a focus on the impact of mutations on inactivation and late sodium current, establishes a structure-function relationship for the mechanistic understanding of Nav1.5 channelopathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号