Kv7

Kv7
  • 文章类型: Journal Article
    对Kv7.2/7.3激动剂日益增长的兴趣源于这些通道参与几种大脑过度兴奋障碍。特别是,Kv7.2/7.3突变体与癫痫性脑病(DEE)以及一系列局灶性癫痫疾病明显相关。通常与发育平稳或退化有关。然而,缺乏可用的治疗选择,考虑到瑞替加宾,临床上唯一用作广谱Kv7激动剂的分子,已于2016年底退出市场。这就是为什么学术界和工业界在寻找充当Kv7.2/7.3激动剂的合适的化学型方面都做出了一些努力。在这种情况下,计算机方法发挥了重要作用,因为不同的Kv7同源四聚体的精确结构直到最近才被公开。在本次审查中,在其生物学和结构功能特性的背景下,讨论了用于设计Kv.7.2/7.3小分子激动剂的计算方法和潜在的药物化学。
    The growing interest in Kv7.2/7.3 agonists originates from the involvement of these channels in several brain hyperexcitability disorders. In particular, Kv7.2/7.3 mutants have been clearly associated with epileptic encephalopathies (DEEs) as well as with a spectrum of focal epilepsy disorders, often associated with developmental plateauing or regression. Nevertheless, there is a lack of available therapeutic options, considering that retigabine, the only molecule used in clinic as a broad-spectrum Kv7 agonist, has been withdrawn from the market in late 2016. This is why several efforts have been made both by both academia and industry in the search for suitable chemotypes acting as Kv7.2/7.3 agonists. In this context, in silico methods have played a major role, since the precise structures of different Kv7 homotetramers have been only recently disclosed. In the present review, the computational methods used for the design of Kv.7.2/7.3 small molecule agonists and the underlying medicinal chemistry are discussed in the context of their biological and structure-function properties.
    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
    离子通道是膜蛋白,其也可以具有与其他配体相互作用的细胞内和细胞外结构域。在许多情况下,这些相互作用位点是高度可移动的,并且在与调节信号分子结合时可能会发生构型变化。等温滴定量热法(ITC)是量化溶液中纯化样品的蛋白质-配体相互作用的强大技术。本章介绍了一种基于片段的分析方法,使用ITC来量化电压门控Kv7通道的结构域与钙调节蛋白钙调蛋白之间的相互作用。该实例可用于量化其他离子通道的特定结构域与其调节信号蛋白之间的相互作用。
    Ion channels are membrane proteins that may also have intracellular and extracellular domains that interact with other ligands. In many cases, these interaction sites are highly mobile and may undergo changes in the configuration upon binding with regulatory signaling molecules. Isothermal titration calorimetry (ITC) is a powerful technique to quantify protein-ligand interactions of purified samples in solution. This chapter describes a fragment-based analysis method using ITC to quantify the interactions between a domain of the voltage-gated Kv7 channel and the calcium-regulated protein calmodulin. This example can be used to quantify the interactions between specific domains of other ion channels and their regulatory signaling proteins.
    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
    KCNQ家族由五个基因组成,表达产物形成电压门控钾通道(Kv7.1-7.5),对许多细胞类型的细胞生理学具有重大影响。每个功能性Kv7通道形成为四聚体,通常与KCNE基因家族(KCNE1-5)编码的蛋白质结合,并且严重依赖于磷脂酰肌醇双磷酸(PIP2)和钙调蛋白的结合。其他调节剂,如A激酶锚定蛋白,泛素连接酶和Ca-钙调蛋白激酶II以同工型特异性方式改变Kv7通道功能和运输。现在已经确定,对于Kv7.4,G蛋白βγ亚基(Gβγ)可以添加到关键调节因子列表中,并且对于通道活性至关重要。本文概述了这一新兴的研究领域,突出未来研究的主题和方向。
    The KCNQ family is comprised of five genes and the expression products form voltage-gated potassium channels (Kv7.1-7.5) that have a major impact upon cellular physiology in many cell types. Each functional Kv7 channel forms as a tetramer that often associates with proteins encoded by the KCNE gene family (KCNE1-5) and is critically reliant upon binding of phosphatidylinositol bisphosphate (PIP2) and calmodulin. Other modulators like A-kinase anchoring proteins, ubiquitin ligases and Ca-calmodulin kinase II alter Kv7 channel function and trafficking in an isoform specific manner. It has now been identified that for Kv7.4, G protein βγ subunits (Gβγ) can be added to the list of key regulators and is paramount for channel activity. This article provides an overview of this nascent field of research, highlighting themes and directions for future study.
    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
    癫痫样活动是一组神经元过度兴奋的最惊人的结果,这些神经元可以在不同的大脑区域发生,然后扩散到其他部位。后来发现,这些节律在体外具有细胞相关性,称为阵发性去极化偏移(PDS)。在13-15DIV神经元神经胶质细胞培养中,抑制GABA(A)受体会引起碎屑PDS形式的动作电位爆发和细胞内Ca2浓度([Ca2]i)的振荡。我们证明了表达钙通透性AMPA受体(CP-AMPAR)的GABA能神经元以及Kv7型钾通道在培养的癫痫样活动中调节海马谷氨酸能神经元的兴奋性。
    使用电流钳模式下的全细胞膜片钳和钙成像显微镜的组合同时记录膜电位和[Ca2]i水平。为了鉴定GABA能细胞培养物,在进行重要的[Ca2]i成像后,将其固定并用抗谷氨酸脱羧酶GAD65/67和神经元特异性烯醇化酶(NSE)的抗体染色。
    表明CP-AMPAR参与PDS簇的调节,[Ca2]i脉冲伴随着它们。GABA能神经元的CP-AMPAR的激活被认为会导致GABA的释放,激活其他GABA能中间神经元的GABA(B)受体。假定这些GABA(B)受体的激活导致Gi蛋白的β-γ亚基的释放,激活钾通道,导致这些中间神经元的超极化和抑制。后者导致谷氨酸能神经元的去抑制,这些中间神经元的目标。反过来,CP-AMPAR拮抗剂,NASPM,有相反的效果。在全细胞配置中通过膜片钳方法测量GABA能神经元的膜电位表明,NASPM抑制簇和单个PDS中的超极化。据信Kv7型钾通道参与癫痫样活动期间超极化的控制。Kv7通道的阻断剂,XE991模拟了CP-AMPAR拮抗剂对PDS簇的作用。两种药物都增加了PDS簇的持续时间。反过来,Kv7活化剂,瑞替加宾,减少PDS簇和Ca2+脉冲的持续时间。此外,瑞替加滨在PDS簇的末端导致深度极化。Kv7通道被认为参与了PDS的形成,由于通道阻滞剂降低了PDS中的超极化率几乎三倍。因此,表达CP-AMPAR的GABA能神经元,通过调节其他GABA能神经元的突触后钾通道的活性来调节神经支配的谷氨酸能神经元的膜电位。
    UNASSIGNED: Epileptiform activity is the most striking result of hyperexcitation of a group of neurons that can occur in different brain regions and then spread to other sites. Later it was shown that these rhythms have a cellular correlate in vitro called paroxysmal depolarization shift (PDS). In 13-15 DIV neuron-glial cell culture, inhibition of the GABA(A) receptors induces bursts of action potential in the form of clasters PDS and oscillations of intracellular Ca2+ concentration ([Ca2+]i). We demonstrate that GABAergic neurons expressing calcium-permeable AMPA receptors (CP-AMPARs) as well as Kv7-type potassium channels regulate hippocampal glutamatergic neurons\' excitability during epileptiform activity in culture.
    UNASSIGNED: A combination of whole-cell patch-clamp in current clamp mode and calcium imaging microscopy was used to simultaneously register membrane potential and [Ca2+]i level. To identify GABAergic cell cultures were fixed and stained with antibodies against glutamate decarboxylase GAD 65/67 and neuron-specific enolase (NSE) after vital [Ca2+]i imaging.
    UNASSIGNED: It was shown that CP-AMPARs are involved in the regulation of the PDS clusters and [Ca2+]i pulses accompanied them. Activation of CP-AMPARs of GABAergic neurons is thought to cause the release of GABA, which activates the GABA(B) receptors of other GABAergic interneurons. It is assumed that activation of these GABA(B) receptors leads to the release of beta-gamma subunits of Gi protein, which activate potassium channels, resulting in hyperpolarization and inhibition of these interneurons. The latter causes disinhibition of glutamatergic neurons, the targets of these interneurons. In turn, the CP-AMPAR antagonist, NASPM, has the opposite effect. Measurement of membrane potential in GABAergic neurons by the patch-clamp method in whole-cell configuration demonstrated that NASPM suppresses hyperpolarization in clusters and individual PDSs. It is believed that Kv7-type potassium channels are involved in the control of hyperpolarization during epileptiform activity. The blocker of Kv7 channels, XE 991, mimicked the effect of the CP-AMPARs antagonist on PDS clusters. Both drugs increased the duration of the PDS cluster. In turn, the Kv7 activator, retigabine, decreased the duration of the PDS cluster and Ca2+ pulse. In addition, retigabine led to deep posthyperpolarization at the end of the PDS cluster. The Kv7 channel is believed to be involved in the formation of PDS, as the channel blocker reduced the rate of hyperpolarization in the PDS almost three times. Thus, GABAergic neurons expressing CP-AMPARs, regulate the membrane potential of innervated glutamatergic neurons by modulating the activity of postsynaptic potassium channels of other GABAergic neurons.
    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
    已对梯形体(MNTB)的内侧核作为脑干听觉电路的主要抑制源进行了深入研究。MNTB衍生的抑制在声音位置的计算中起着至关重要的作用,因为声音的时间特征通过Held/MNTB突触的花萼精确地传达。在成年沙鼠中,胆碱能信号通过烟碱乙酰胆碱受体影响MNTB神经元的声音诱发反应(nAChRs;Zhangetal.,2021)建立对该核的胆碱能输入的调节作用。然而,乙酰胆碱(ACh)介导MNTB中这种调节的细胞机制仍然不清楚。为了研究这些机制,我们使用全细胞电流和电压钳记录来检查来自两种性别的蒙古沙鼠(Merionesunguiculatus)的MNTB神经元的胆碱能生理。在脑切片中评估膜兴奋性,在听力前(出生后第9-13天)和听力发作后(P18-20)的MNTB神经元中,使用烟碱(nAChRs)和毒蕈碱受体(mAChRs)的激动剂和拮抗剂。毒蕈碱激活在听力发作之前最明显地引起兴奋性的有效增加,而nAChR调制在较晚的时间点出现。药理学操作进一步证明,电压门控K+通道KCNQ(Kv7)是mAChR激活的下游效应物,其在发育早期影响兴奋性。Kv7的胆碱能调节降低了向外的K电导并使静息膜电位去极化。免疫标记揭示了Kv7通道以及含有M1和M3亚基的mAChR的表达。一起,我们的结果表明,在正在发展的MNTB中,mAChR调制是突出的,但是短暂的,胆碱能调制功能可以塑造听觉电路的发育。
    The medial nucleus of the trapezoid body (MNTB) has been intensively investigated as a primary source of inhibition in brainstem auditory circuitry. MNTB-derived inhibition plays a critical role in the computation of sound location, as temporal features of sounds are precisely conveyed through the calyx of Held/MNTB synapse. In adult gerbils, cholinergic signaling influences sound-evoked responses of MNTB neurons via nicotinic acetylcholine receptors (nAChRs; Zhang et al., 2021) establishing a modulatory role for cholinergic input to this nucleus. However, the cellular mechanisms through which acetylcholine (ACh) mediates this modulation in the MNTB remain obscure. To investigate these mechanisms, we used whole-cell current and voltage-clamp recordings to examine cholinergic physiology in MNTB neurons from Mongolian gerbils (Meriones unguiculatus) of both sexes. Membrane excitability was assessed in brain slices, in pre-hearing (postnatal days 9-13) and post-hearing onset (P18-20) MNTB neurons during bath application of agonists and antagonists of nicotinic (nAChRs) and muscarinic receptors (mAChRs). Muscarinic activation induced a potent increase in excitability most prominently prior to hearing onset with nAChR modulation emerging at later time points. Pharmacological manipulations further demonstrated that the voltage-gated K+ channel KCNQ (Kv7) is the downstream effector of mAChR activation that impacts excitability early in development. Cholinergic modulation of Kv7 reduces outward K+ conductance and depolarizes resting membrane potential. Immunolabeling revealed expression of Kv7 channels as well as mAChRs containing M1 and M3 subunits. Together, our results suggest that mAChR modulation is prominent but transient in the developing MNTB and that cholinergic modulation functions to shape auditory circuit development.
    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
    异常的细胞和回路兴奋性被认为驱动脆性X综合征(FXS)中的许多核心表型。齿状回是执行学习和记忆所必需的关键计算的大脑区域。然而,对FXS中齿状电路缺陷及其机制知之甚少。由于存在两种类型的兴奋性神经元,理解FXS中的齿状回路功能障碍变得复杂,颗粒细胞和苔藓细胞。在这里,我们报告FMRP的丢失显着降低齿状苔藓细胞的兴奋性,与FXS中兴奋性神经元的所有其他已知兴奋性缺陷相反的变化。这种苔藓细胞低兴奋性是由Fmr1敲除(KO)小鼠中Kv7功能增加引起的。通过减少对局部肺门中间神经元的兴奋驱动,苔藓细胞的低兴奋性导致颗粒细胞的兴奋/抑制率增加,因此矛盾地导致齿状输出过多。Fmr1KO小鼠中Kv7通道的全电路抑制增加了对颗粒细胞的抑制驱动,并使齿状输出正常化,以响应生理相关的θ-γ偶联刺激。我们的研究表明,基于回路的干预措施可能为这种疾病提供有希望的策略,以绕过不同细胞类型中不可调和的兴奋性缺陷,并在回路水平上恢复其病理生理后果。
    Abnormal cellular and circuit excitability is believed to drive many core phenotypes in fragile X syndrome (FXS). The dentate gyrus is a brain area performing critical computations essential for learning and memory. However, little is known about dentate circuit defects and their mechanisms in FXS. Understanding dentate circuit dysfunction in FXS has been complicated by the presence of two types of excitatory neurons, the granule cells and mossy cells. Here we report that loss of FMRP markedly decreased excitability of dentate mossy cells, a change opposite to all other known excitability defects in excitatory neurons in FXS. This mossy cell hypo-excitability is caused by increased Kv7 function in Fmr1 knockout (KO) mice. By reducing the excitatory drive onto local hilar interneurons, hypo-excitability of mossy cells results in increased excitation/inhibition ratio in granule cells and thus paradoxically leads to excessive dentate output. Circuit-wide inhibition of Kv7 channels in Fmr1 KO mice increases inhibitory drive onto granule cells and normalizes the dentate output in response to physiologically relevant theta-gamma coupling stimulation. Our study suggests that circuit-based interventions may provide a promising strategy in this disorder to bypass irreconcilable excitability defects in different cell types and restore their pathophysiological consequences at the circuit level.
    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
    神经元Kv7电压门控钾通道产生M电流并调节神经元兴奋性。这里,我们报道硫酸脱氢表雄酮(DHEAS)是一种内源性Kv7通道调节剂,可减弱Gq偶联受体诱导的M电流抑制.DHEAS降低了毒蕈碱激动剂诱导的Kv7电流对大鼠交感神经节神经元中Kv7.1,Kv7.2,Kv7.4或Kv7.5同源电流和内源性M电流的抑制。然而,DHEAS本身没有改变这些Kv7同源通道的电压依赖性或m1受体诱导的磷脂酶C或蛋白激酶C的激活。DHEAS处理的Kv7.2同源电流对磷脂酰肌醇4,5-二磷酸(PIP2)的消耗具有抗性由电压激活的磷酸酶,Ci-VSP或eVSP。我们的计算模型预测了Kv7亚基的细胞质结构域中DHEAS的新结合位点。预测的关键组氨酸在大鼠Kv7.2亚基中单点突变为半胱氨酸,rKv7.2(H558C),导致DHEAS对毒蕈碱Kv7电流抑制的作用丧失。此外,在福尔马林爪试验中,在两性小鼠体内施用DHEAS可减少晚期疼痛反应。然而,它对福尔马林爪试验的早期反应或热板试验的反应没有影响。联合使用选择性Kv7抑制剂,XE991和DHEAS在福尔马林爪试验中消除了DHEAS在晚期反应中的镇痛作用。总的来说,这些结果表明,DHEAS通过稳定PIP2-Kv7亚基相互作用减弱M-电流抑制,并可减轻炎性疼痛.意义StatementM-通过刺激Gq偶联受体诱导的电流抑制是Kv7电流调制的一种形式,可以可逆地增加神经元兴奋性。这项研究表明,DHEAS,一种内源性类固醇激素,是一种新型的Kv7通道调制器,可以在不影响基础Kv7通道动力学的情况下衰减M电流抑制。体内施用DHEAS减轻了啮齿动物的炎性疼痛。这些结果表明,小分子可以动态调节M电流抑制的程度。因此,这种新形式的Kv7通道调节具有作为致敏神经活动如炎性疼痛的治疗靶点的潜力.
    Neuronal Kv7 voltage-gated potassium channels generate the M-current and regulate neuronal excitability. Here, we report that dehydroepiandrosterone sulfate (DHEAS) is an endogenous Kv7 channel modulator that attenuates Gq-coupled receptor-induced M-current suppression. DHEAS reduced muscarinic agonist-induced Kv7-current suppression of Kv7.1, Kv7.2, Kv7.4, or Kv7.5 homomeric currents and endogenous M-currents in rat sympathetic ganglion neurons. However, DHEAS per se did not alter the voltage dependence of these Kv7 homomeric channels or the m1 receptor-induced activation of phospholipase C or protein kinase C. DHEAS-treated Kv7.2 homomeric currents became resistant to depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) induced by voltage-activated phosphatase, Ci-VSP or eVSP. Our computational models predicted a novel binding site for DHEAS in the cytoplasmic domain of Kv7 subunits. A single-point mutation of the predicted key histidine into cysteine in the rat Kv7.2 subunit, rKv7.2(H558C), resulted in a loss of effects of DHEAS on muscarinic Kv7 current suppression. Furthermore, in vivo administration of DHEAS in mice of both sexes reduced late phase pain responses in the formalin paw test. However, it did not have effects on early phase responses in the formalin paw test or responses in the hot plate test. Coadministration of a selective Kv7 inhibitor, XE991, and DHEAS eliminated analgesic effects of DHEAS in late phase responses in the formalin paw test. Collectively, these results suggest that DHEAS attenuates M-current suppression by stabilizing PIP2-Kv7 subunit interaction and can mitigate inflammatory pain.SIGNIFICANCE STATEMENT M-current suppression induced by stimulation of Gq-coupled receptors is a form of Kv7 current modulation that can reversibly increase neuronal excitability. This study demonstrates that DHEAS, an endogenous steroid hormone, is a novel Kv7 channel modulator that can attenuate M-current suppression without affecting basal Kv7 channel kinetics. Administration of DHEAS in vivo alleviated inflammatory pain in rodents. These results suggest that the degree of M-current suppression can be dynamically regulated by small molecules. Therefore, this novel form of Kv7 channel regulation holds promising potential as a therapeutic target for sensitized nervous activities, such as inflammatory pain.
    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
    K+通道通过影响动作电位波形,调节神经元兴奋性和释放概率,在神经递质释放过程中起着重要作用。K+通道激活的这些不同作用是由各种各样的K+通道基因及其在不同细胞类型中的差异表达所确保的。因此,多种K+通道参与调节神经递质的释放,包括Ca2+和电压门控K+通道Slo1(也称为BK通道),Kv3(肖型)的电压门控K+通道,Kv1(振动筛型),和Kv7(KCNQ)家族,G蛋白门控向内整流K(GIRK)通道,和SLO-2(aCa2+-。Cl-,和电压门控K+通道在C.elegans)。这些通道的表达方式各不相同,亚细胞定位,和生物物理特性。它们在神经递质释放中的作用也可能因突触和生理或实验条件而异。本章总结了有关K通道在调节神经递质释放中的作用的关键发现。
    K+ channels play potent roles in the process of neurotransmitter release by influencing the action potential waveform and modulating neuronal excitability and release probability. These diverse effects of K+ channel activation are ensured by the wide variety of K+ channel genes and their differential expression in different cell types. Accordingly, a variety of K+ channels have been implicated in regulating neurotransmitter release, including the Ca2+- and voltage-gated K+ channel Slo1 (also known as BK channel), voltage-gated K+ channels of the Kv3 (Shaw-type), Kv1 (Shaker-type), and Kv7 (KCNQ) families, G-protein-gated inwardly rectifying K+ (GIRK) channels, and SLO-2 (a Ca2+-. Cl-, and voltage-gated K+ channel in C. elegans). These channels vary in their expression patterns, subcellular localization, and biophysical properties. Their roles in neurotransmitter release may also vary depending on the synapse and physiological or experimental conditions. This chapter summarizes key findings about the roles of K+ channels in regulating neurotransmitter release.
    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
    背景:患有心脏信道病长QT综合征(LQTS)的基因型阳性患者可能表现出一系列临床表型,通常原因不明。因此,有必要确定影响疾病严重程度的因素,以实现LQTS的个体化临床管理.影响疾病表型的一个可能因素是内源性大麻素系统,已经成为心血管功能的调节剂。在这项研究中,我们旨在阐明内源性大麻素是否靶向心脏电压门控钾通道KV7.1/KCNE1,后者是LQTS中最常突变的离子通道.
    方法:我们使用了双电极电压钳,分子动力学模拟和E4031药物诱导的离体豚鼠心脏LQT2模型。
    结果:我们发现了一组促进通道激活的内源性大麻素,被视为通道开口的电压依赖性偏移,总电流幅度和电导增加。我们建议带负电荷的内源性大麻素与通道上带正电荷的氨基酸处的已知脂质结合位点相互作用,提供为什么只有特异性内源性大麻素调节KV7.1/KCNE1的结构见解。使用内源性大麻素ARA-S作为原型,我们表明,这种作用不依赖于KCNE1亚基或通道的磷酸化状态。在豚鼠的心脏里,发现ARA-S逆转E4031延长的动作电位持续时间和QT间期。
    结论:我们认为内源性大麻素是一类有趣的hKV7.1/KCNE1通道调节剂,在LQTS环境中具有推定的保护作用。
    背景:ERC(编号:850622),加拿大卫生研究院,加拿大研究主席和计算加拿大,瑞典国家计算基础设施。
    BACKGROUND: Genotype-positive patients who suffer from the cardiac channelopathy Long QT Syndrome (LQTS) may display a spectrum of clinical phenotypes, with often unknown causes. Therefore, there is a need to identify factors influencing disease severity to move towards an individualized clinical management of LQTS. One possible factor influencing the disease phenotype is the endocannabinoid system, which has emerged as a modulator of cardiovascular function. In this study, we aim to elucidate whether endocannabinoids target the cardiac voltage-gated potassium channel KV7.1/KCNE1, which is the most frequently mutated ion channel in LQTS.
    METHODS: We used two-electrode voltage clamp, molecular dynamics simulations and the E4031 drug-induced LQT2 model of ex-vivo guinea pig hearts.
    RESULTS: We found a set of endocannabinoids that facilitate channel activation, seen as a shifted voltage-dependence of channel opening and increased overall current amplitude and conductance. We propose that negatively charged endocannabinoids interact with known lipid binding sites at positively charged amino acids on the channel, providing structural insights into why only specific endocannabinoids modulate KV7.1/KCNE1. Using the endocannabinoid ARA-S as a prototype, we show that the effect is not dependent on the KCNE1 subunit or the phosphorylation state of the channel. In guinea pig hearts, ARA-S was found to reverse the E4031-prolonged action potential duration and QT interval.
    CONCLUSIONS: We consider the endocannabinoids as an interesting class of hKV7.1/KCNE1 channel modulators with putative protective effects in LQTS contexts.
    BACKGROUND: ERC (No. 850622), Canadian Institutes of Health Research, Canada Research Chairs and Compute Canada, Swedish National Infrastructure for Computing.
    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
    KV7通道开放剂已证明其在治疗疼痛和癫痫方面的治疗价值,此外,它们有可能扩展到医疗需求未得到满足的其他适应症。然而,经临床验证但同时停止的KV7通道开放剂氟吡汀和瑞替加滨具有氧化敏感的三氨基芳基支架,怀疑通过形成醌类氧化产物引起药物不良反应。这里,我们报道了烟酰胺类似物和相关化合物的设计和合成,这些化合物修复了氟吡汀和瑞替加滨化学结构中的缺陷。烟酰胺铅结构的优化产生具有优异的KV7.2/3开放活性的类似物,正如EC50值接近个位数纳摩尔范围所证明的那样。另一方面,加权KV7.2/3开放活性数据,包括允许建立结构-活性关系的非活性化合物以及通过对接和分子动力学模拟验证的合理结合模式假设。
    KV 7 channel openers have proven their therapeutic value in the treatment of pain as well as epilepsy and, moreover, they hold the potential to expand into additional indications with unmet medical needs. However, the clinically validated but meanwhile discontinued KV 7 channel openers flupirtine and retigabine bear an oxidation-sensitive triaminoraryl scaffold, which is suspected of causing adverse drug reactions via the formation of quinoid oxidation products. Here, we report the design and synthesis of nicotinamide analogs and related compounds that remediate the liability in the chemical structure of flupirtine and retigabine. Optimization of a nicotinamide lead structure yielded analogs with excellent KV 7.2/3 opening activity, as evidenced by EC50 values approaching the single-digit nanomolar range. On the other hand, weighted KV 7.2/3 opening activity data including inactive compounds allowed for the establishment of structure-activity relationships and a plausible binding mode hypothesis verified by docking and molecular dynamics simulations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号