EIMFS

EIMFS
  • 文章类型: Journal Article
    KCNT1基因的功能增益突变,编码钠激活钾通道SLACK,与罕见但破坏性的发育性脑病和癫痫性脑病有关,称为婴儿期癫痫伴迁移局灶性癫痫(EIMFS)。SLACK通道的小分子抑制剂的设计代表了治疗EIMFS的潜在治疗方法,其他儿童癫痫症,和发育障碍。在这里,我们描述了以通过高通量筛选发现的黄嘌呤SLACK抑制剂(8)为中心的命中优化努力。在化学型的三个不同区域,我们合成了58种新的类似物,并在全细胞自动化膜片钳试验中对每种类似物进行了测试,以建立抑制SLACK通道的构效关系.我们进一步评估了所选择的类似物对各种其他离子通道的选择性以及对临床相关SLACK突变体的活性。系列中的选择性相当不错,包括对hERG。类似物80(VU0948578)是WT的有效抑制剂,A934T,和G288S松弛,这些变体的IC50值在0.59和0.71µM之间。VU0948578代表来自化学型的有用的体外工具化合物,其不同于先前报道的SLACK通道的小分子抑制剂。
    Gain-of-function mutations in the KCNT1 gene, which encodes the sodium-activated potassium channel known as SLACK, are associated with the rare but devastating developmental and epileptic encephalopathy known as epilepsy of infancy with migrating focal seizures (EIMFS). The design of small molecule inhibitors of SLACK channels represents a potential therapeutic approach to the treatment of EIMFS, other childhood epilepsies, and developmental disorders. Herein, we describe a hit optimization effort centered on a xanthine SLACK inhibitor (8) discovered via a high-throughput screen. Across three distinct regions of the chemotype, we synthesized 58 new analogs and tested each one in a whole-cell automated patch-clamp assay to develop structure-activity relationships for inhibition of SLACK channels. We further evaluated selected analogs for their selectivity versus a variety of other ion channels and for their activity versus clinically relevant SLACK mutants. Selectivity within the series was quite good, including versus hERG. Analog 80 (VU0948578) was a potent inhibitor of WT, A934T, and G288S SLACK, with IC50 values between 0.59 and 0.71 µM across these variants. VU0948578 represents a useful in vitro tool compound from a chemotype that is distinct from previously reported small molecule inhibitors of SLACK channels.
    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
    KCNT1基因编码钠激活钾通道Slack(KCNT1,KNa1.1),神经元兴奋性的调节器。人类的功能增益突变导致皮层网络过度兴奋,癫痫发作,和严重的智力残疾。使用表达Slack-R455H突变的小鼠模型,我们发现,在兴奋性和抑制性皮质神经元中,Na依赖性K(KNa)和电压依赖性钠(NaV)电流均增加。这些增加的电流,然而,增强兴奋性神经元的放电,但抑制抑制性神经元的放电。我们进一步表明,NaV通道亚基的表达,特别是NaV1.6的水平上调,并且两种神经元类型的轴突初始节段和轴突NaV免疫染色的长度都增加。我们对KNa电流和NaV通道表达的协调调节的研究可能为理解和治疗癫痫和其他神经系统疾病提供了途径。
    The KCNT1 gene encodes the sodium-activated potassium channel Slack (KCNT1, KNa1.1), a regulator of neuronal excitability. Gain-of-function mutations in humans cause cortical network hyperexcitability, seizures, and severe intellectual disability. Using a mouse model expressing the Slack-R455H mutation, we find that Na+-dependent K+ (KNa) and voltage-dependent sodium (NaV) currents are increased in both excitatory and inhibitory cortical neurons. These increased currents, however, enhance the firing of excitability neurons but suppress that of inhibitory neurons. We further show that the expression of NaV channel subunits, particularly that of NaV1.6, is upregulated and that the length of the axon initial segment and of axonal NaV immunostaining is increased in both neuron types. Our study on the coordinate regulation of KNa currents and the expression of NaV channels may provide an avenue for understanding and treating epilepsies and other neurological disorders.
    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
    婴儿期恶性移行性部分癫痫(MMPSI)是一种破坏性和药物抗性的婴儿癫痫。MMPSI已与KCNT1基因中的多个功能获得(GOF)突变有关,编码钾通道,通常称为SLACK。SLACK通道是分布在整个中枢神经系统(CNS)和外周的钠激活钾通道。本文描述的研究旨在发现SLACK通道抑制剂工具化合物并概述其药代动力学和药效学性质。通过高通量筛选(HTS)活动鉴定SLACK通道抑制剂VU0531245(VU245)。在击中的VU245的五个不同区域进行了结构-活性关系(SAR)研究。在稳定表达野生型(WT)人SLACK的HEK-293细胞中使用铊通量测定评价VU245类似物影响SLACK通道活性的能力。测试所选择的类似物在小鼠肝微粒体中的代谢稳定性和在小鼠血浆中的血浆-蛋白结合。通过铊通量测试同一组类似物与人A934TSLACK和其他结构相关的钾通道的活性,包括SLICK和Maxi-K.此外,通过自动膜片钳系统,在稳定表达WT人SLACK的CHO细胞中使用全细胞电生理学(EP)测定获得了所选VU245类似物的效力.结果表明,这种支架可以容忍某些区域的结构变化,一些类似物表现出改善的SLACK抑制活性,对测试的其他通道具有良好的选择性,代谢清除率的适度改善。模拟VU0935685代表一个新的,SLACK通道的结构独特的小分子抑制剂,可作为研究该靶标的体外工具。
    Malignant migrating partial seizure of infancy (MMPSI) is a devastating and pharmacoresistant form of infantile epilepsy. MMPSI has been linked to multiple gain-of-function (GOF) mutations in the KCNT1 gene, which encodes for a potassium channel often referred to as SLACK. SLACK channels are sodium-activated potassium channels distributed throughout the central nervous system (CNS) and the periphery. The investigation described here aims to discover SLACK channel inhibitor tool compounds and profile their pharmacokinetic and pharmacodynamic properties. A SLACK channel inhibitor VU0531245 (VU245) was identified via a high-throughput screen (HTS) campaign. Structure-activity relationship (SAR) studies were conducted in five distinct regions of the hit VU245. VU245 analogs were evaluated for their ability to affect SLACK channel activity using a thallium flux assay in HEK-293 cells stably expressing wild-type (WT) human SLACK. Selected analogs were tested for metabolic stability in mouse liver microsomes and plasma-protein binding in mouse plasma. The same set of analogs was tested via thallium flux for activity versus human A934T SLACK and other structurally related potassium channels, including SLICK and Maxi-K. In addition, potencies for selected VU245 analogs were obtained using whole-cell electrophysiology (EP) assays in CHO cells stably expressing WT human SLACK through an automated patch clamp system. Results revealed that this scaffold tolerates structural changes in some regions, with some analogs demonstrating improved SLACK inhibitory activity, good selectivity against the other channels tested, and modest improvements in metabolic clearance. Analog VU0935685 represents a new, structurally distinct small-molecule inhibitor of SLACK channels that can serve as an in vitro tool for studying this target.
    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
    已在编码GABAA受体β3亚基的GABRB3基因中鉴定了与发育和癫痫性脑病相关的遗传变体。通常,变体改变受体对GABA的敏感性,导致功能的增益或丧失,与患者表型相关。然而,目前还不清楚另一个重要的受体特性,脱敏,导致功能获得变异的临床严重程度更高。使用双电极电压钳电生理学评估了20种功能获得GABRB3变体受体的脱敏特性。测量的参数包括电流衰减率和稳态电流。还在转染的哺乳动物细胞系中使用全细胞电生理学评估了具有增加或减少的脱敏作用的选定变体。在评估的20种功能增益变体中,发现13改变受体脱敏特性。七个变体在平衡时减少了脱敏,这会恶化功能获得特征。六个变体加速电流衰减动力学,这限制了功能获得特征。所有受影响的患者均表现出严重的临床表型,伴有智力障碍和难以治疗的癫痫。然而,在平衡状态下降低脱敏的变异与更严重的临床结局相关.这包括首次发作的年龄较小(中位数为0.5个月),运动障碍(肌张力障碍和运动障碍),婴儿期癫痫伴转移性局灶性癫痫(EIMFS)和早期死亡风险。加速电流衰减动力学的变体与癫痫发作后期(中位数4个月)稍微温和的表型相关。无法分类的发育性和癫痫性脑病或Lennox-Gastaut综合征,没有运动障碍。我们的研究表明,功能获得GABRB3变体可以增加或减少受体脱敏特性,并且与疾病严重程度相关。在平衡状态下降低脱敏的变体聚集在构成通道孔的跨膜区域中,并与更大的疾病严重程度相关。而加速电流衰减的变体聚集在负责受体激活的耦合环中,并与较低的严重程度相关。
    Genetic variants associated with developmental and epileptic encephalopathies have been identified in the GABRB3 gene that encodes the β3 subunit of GABAA receptors. Typically, variants alter receptor sensitivity to GABA resulting in either gain- or loss-of-function, which correlates with patient phenotypes. However, it is unclear how another important receptor property, desensitization, contributes to the greater clinical severity of gain-of-function variants. Desensitization properties of 20 gain-of-function GABRB3 variant receptors were evaluated using two-electrode voltage-clamp electrophysiology. The parameters measured included current decay rates and steady-state currents. Selected variants with increased or reduced desensitization were also evaluated using whole-cell electrophysiology in transfected mammalian cell lines. Of the 20 gain-of-function variants assessed, 13 were found to alter receptor desensitization properties. Seven variants reduced desensitization at equilibrium, which acts to worsen gain-of-function traits. Six variants accelerated current decay kinetics, which limits gain-of-function traits. All affected patients displayed severe clinical phenotypes with intellectual disability and difficult-to-treat epilepsy. Nevertheless, variants that reduced desensitization at equilibrium were associated with more severe clinical outcomes. This included younger age of first seizure onset (median 0.5 months), movement disorders (dystonia and dyskinesia), epilepsy of infancy with migrating focal seizures (EIMFS) and risk of early mortality. Variants that accelerated current decay kinetics were associated with slightly milder phenotypes with later seizure onset (median 4 months), unclassifiable developmental and epileptic encephalopathies or Lennox-Gastaut syndrome and no movement disorders. Our study reveals that gain-of-function GABRB3 variants can increase or decrease receptor desensitization properties and that there is a correlation with the degree of disease severity. Variants that reduced the desensitization at equilibrium were clustered in the transmembrane regions that constitute the channel pore and correlated with greater disease severity, while variants that accelerated current decay were clustered in the coupling loops responsible for receptor activation and correlated with lesser severity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    婴儿期癫痫伴转移性局灶性癫痫发作(EIMFS)是一种罕见的破坏性婴儿癫痫性脑病,其特征在于多灶性同时癫痫发作的独特脑电图(EEG)特征。虽然EIMFS没有明确的病因,某些离子通道中的突变,有牵连。同样,苯丙氨酰-tRNA合成酶2(FARS2)缺乏症是一种罕见的,功能失调的线粒体翻译引起难治性癫痫发作的常染色体隐性遗传疾病,乳酸性酸中毒,和发育回归与各种脑电图的发现。然而,FARS2缺乏症的EEG类似EIMFS模式最近才报道过一次。在这里,我们描述了一名7周大的男性癫痫发作,其中全外显子组测序(WES)显示致病性FARS2变异和EEG的EIMFS模式.这个案例为EIMFS提供了一种新的遗传机制。当检测到EIMFS以评估FARS2缺陷时,我们鼓励早期考虑WES,尤其是在严重的乳酸性酸中毒中。
    Epilepsy of infancy with migrating focal seizures (EIMFS) is a rare devastating infantile epileptic encephalopathy that is characterized by a unique electroencephalopgraphy (EEG) signature of multifocal simultaneous seizures. Although no definite etiology is understood for EIMFS, mutations in certain ion channels, are implicated. Similarly, phenylalanyl-tRNA synthetase 2 (FARS2) deficiency is a rare, autosomal recessive disorder of dysfunctional mitochondrial translation causing refractory seizures, lactic acidosis, and developmental regression with a variety of EEG findings. However, an EIMFS-like pattern on EEG in FARS2 deficiency has only recently been reported once. Herein, we describe a seven-week-old male with seizures where whole exome sequencing (WES) revealed pathogenic FARS2 variants and an EIMFS pattern on EEG. This case provides an insight on a novel genetic mechanism for EIMFS. We encourage early consideration of WES when EIMFS is detected to evaluate for FARS2 deficiency, especially in the setting of profound lactic acidosis.
    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
    在这封信中,我们描述了在新的2-氨基-N-苯基乙酰胺系列Slack钾通道抑制剂的五个不同区域进行的结构-活性关系(SAR)研究,例如最近公开的高通量筛选(HTS)命中VU0606170(4)。在稳定表达野生型人(WT)Slack的HEK-293细胞中,在铊(Tl+)通量测定中筛选新的类似物。在Tl+通量相对于A934TSlack和其他Slo家族成员Slick和Maxi-K中筛选所选择的类似物,并使用自动膜片钳系统在全细胞电生理学(EP)测定中进行评估。结果表明,该系列具有平坦的SAR,具有明显的结构修饰,导致Slack活性丧失。更微小的改变导致具有类似于HTS命中的Slack活性和Slo家族选择性的化合物。
    In this Letter we describe structure-activity relationship (SAR) studies conducted in five distinct regions of a new 2-amino-N-phenylacetamides series of Slack potassium channel inhibitors exemplified by recently disclosed high-throughput screening (HTS) hit VU0606170 (4). New analogs were screened in a thallium (Tl+) flux assay in HEK-293 cells stably expressing wild-type human (WT) Slack. Selected analogs were screened in Tl+ flux versus A934T Slack and other Slo family members Slick and Maxi-K and evaluated in whole-cell electrophysiology (EP) assays using an automated patch clamp system. Results revealed the series to have flat SAR with significant structural modifications resulting in a loss of Slack activity. More minor changes led to compounds with Slack activity and Slo family selectivity similar to the HTS hit.
    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
    松弛通道是由KCNT1基因编码的钠激活钾通道。几种KCNT1功能获得突变已与婴儿期恶性迁移部分癫痫发作有关。奎尼丁是一种抗心律失常药物,可作为Slack通道的中等有效抑制剂;然而,奎尼丁的使用受到选择性差的限制,安全性和药代动力学特征。松弛通道代表了开发用于治疗婴儿期和其他儿童癫痫的恶性迁移部分性癫痫发作的新型疗法的有趣目标;因此,正在进行的努力是针对抑制Slack电流的小分子的发现。这篇综述总结了2020-2021年发表的专利申请,描述了新型小分子Slack抑制剂的发现。
    Slack channels are sodium-activated potassium channels that are encoded by the KCNT1 gene. Several KCNT1 gain of function mutations have been linked to malignant migrating partial seizures of infancy. Quinidine is an anti-arrhythmic drug that functions as a moderately potent inhibitor of Slack channels; however, quinidine use is limited by its poor selectivity, safety and pharmacokinetic profile. Slack channels represent an interesting target for developing novel therapeutics for the treatment of malignant migrating partial seizures of infancy and other childhood epilepsies; thus, ongoing efforts are directed toward the discovery of small-molecules that inhibit Slack currents. This review summarizes patent applications published in 2020-2021 that describe the discovery of novel small-molecule Slack inhibitors.
    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
    KCNT1的功能增益(GOF)致病变体,该基因编码已知的最大钾通道亚基,KNa1.1与发育性和癫痫性脑病相关,伴有严重的精神运动性和智力障碍。用奎尼丁阻断可过度激活的KNa1.1通道,一类抗心律失常药,在患者中显示出不同的成功,部分原因是剂量限制的脱靶效应,血脑屏障(BBB)穿透性差,和低效力。近年来,已经确定了不同激活状态下的鸡KNa1.1通道的高分辨率低温电子显微镜(cryo-EM)结构,并且已经产生了疾病的动物模型。随着关于GOF致病变体对KNa1.1通道行为的功能影响以及它们如何导致过度兴奋的信息的增加,这些工具将有助于制定更有效的治疗策略。我们回顾了KCNT1变体的范围及其功能效应,当前治疗策略带来的挑战,以及在寻找更有效和选择性治疗KCNT1相关癫痫的干预措施方面的最新进展。
    Gain-of-function (GOF) pathogenic variants of KCNT1, the gene encoding the largest known potassium channel subunit, KNa1.1, are associated with developmental and epileptic encephalopathies accompanied by severe psychomotor and intellectual disabilities. Blocking hyperexcitable KNa1.1 channels with quinidine, a class I antiarrhythmic drug, has shown variable success in patients in part because of dose-limiting off-target effects, poor blood-brain barrier (BBB) penetration, and low potency. In recent years, high-resolution cryogenic electron microscopy (cryo-EM) structures of the chicken KNa1.1 channel in different activation states have been determined, and animal models of the diseases have been generated. Alongside increasing information about the functional effects of GOF pathogenic variants on KNa1.1 channel behaviour and how they lead to hyperexcitability, these tools will facilitate the development of more effective treatment strategies. We review the range of KCNT1 variants and their functional effects, the challenges posed by current treatment strategies, and recent advances in finding more potent and selective therapeutic interventions for KCNT1-related epilepsies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Epilepsy of infancy with migrating focal seizures (EIMFS) is a rare epilepsy syndrome, characterized by an onset of multifocal seizures before the age of six months and a rather typical ictal EEG pattern. The ketogenic diet (KD) has been shown to be a treatment option in these patients with variable results. The KD is generally given by enteral formula or solid food, however, patients on the KD often have coexisting medical disorders that may impair the gastrointestinal tract and, in these cases, parenteral nutrition support may be needed. We present our experience with three patients who had been on the KD because of EIMFS, who were acutely unable to absorb nutrients through the intestinal tract. For these patients, we were unable to reach ketogenic ratios higher than 1.5:1 because of the limited fat intake via the parenteral route. This ratio, nevertheless, was adequate for maintenance of seizure control while allowing short-term bowel rest. Even though our report is limited as it provides no controlled evidence, ketogenic parenteral nutrition should be considered in children on the KD when enteral nutrition is not feasible. Special care should be taken to maintain ketosis and avoid undesired carbohydrates. Patients may respond well to ketogenic parenteral nutrition in spite of a lower ketogenic ratio.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    KCNT1中的突变(Slack,KNa1.1)钠激活钾通道可产生严重的癫痫性脑病。在异源系统中的表达已经表明,致病突变产生具有增加的电流幅度的通道。不知道,然而,这种功能的获得是否发生在人类神经元中,这种增加的KNa电流是否会抑制或增加皮质神经元的兴奋性。使用基因工程人类诱导多能干细胞(iPSC)衍生的神经元,我们现在发现,在带有纯合P924L突变的神经元中,钠依赖性钾电流增加了数倍.在电流钳记录中,具有P924L突变的神经元中增加的KNa电流可以缩短动作电位的持续时间,并增加每个动作电位之后的超极化后的幅度。引人注目的是,在表达突变通道的神经元中,由去极化电流引起的动作电位数量以及最大放电率增加。在自发活跃的神经元网络中,平均射击率,动作电位快速爆发的发生,在P924LSlack突变的神经元中,爆发期间的放电强度均增加。通过数值模拟验证了增加KNa电流以独立于任何补偿性变化来增加点火速率的可行性。我们的发现表明,SlackKNa通道的功能获得会导致孤立的神经元和神经网络的过度兴奋,并通过不需要网络相互作用的细胞自主机制发生。显著性声明KCNT1突变导致严重的癫痫性脑病,没有有效的治疗方法。这项研究首次证明了KCNT1突变会增加神经元中的Slack电流。它还提供了第一个解释这种增加的钾电流如何诱导过度兴奋,这可能是导致癫痫发作的重要因素。
    Mutations in the KCNT1 (Slack, KNa1.1) sodium-activated potassium channel produce severe epileptic encephalopathies. Expression in heterologous systems has shown that the disease-causing mutations give rise to channels that have increased current amplitude. It is not known, however, whether such gain of function occurs in human neurons, nor whether such increased KNa current is expected to suppress or increase the excitability of cortical neurons. Using genetically engineered human induced pluripotent stem cell (iPSC)-derived neurons, we have now found that sodium-dependent potassium currents are increased several-fold in neurons bearing a homozygous P924L mutation. In current-clamp recordings, the increased KNa current in neurons with the P924L mutation acts to shorten the duration of action potentials and to increase the amplitude of the afterhyperpolarization that follows each action potential. Strikingly, the number of action potentials that were evoked by depolarizing currents as well as maximal firing rates were increased in neurons expressing the mutant channel. In networks of spontaneously active neurons, the mean firing rate, the occurrence of rapid bursts of action potentials, and the intensity of firing during the burst were all increased in neurons with the P924L Slack mutation. The feasibility of an increased KNa current to increase firing rates independent of any compensatory changes was validated by numerical simulations. Our findings indicate that gain-of-function in Slack KNa channels causes hyperexcitability in both isolated neurons and in neural networks and occurs by a cell-autonomous mechanism that does not require network interactions.SIGNIFICANCE STATEMENT KCNT1 mutations lead to severe epileptic encephalopathies for which there are no effective treatments. This study is the first demonstration that a KCNT1 mutation increases the Slack current in neurons. It also provides the first explanation for how this increased potassium current induces hyperexcitability, which could be the underlining factor causing seizures.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号