Voltage-Gated Sodium Channel Blockers

电压门控钠通道阻滞剂
  • 文章类型: Editorial
    暂无摘要。
    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
    我们研究了雷诺嗪和伊伐布雷定预处理后对灌注的离体兔肺进行肺动脉血栓栓塞建模时,肺微血流动力学的变化。肺动脉压升高,肺血管阻力,毛细血管前和毛细血管后的阻力不如对照动物明显,但在用电压门控Na+通道阻断剂利多卡因和罗哌卡因预处理后肺血栓栓塞的情况下接近。毛细管过滤系数的增加与毛细管静水压力值呈负相关。因此,雷诺嗪和伊伐布雷定主要在肺动脉平滑肌中表现出电压门控Na通道阻断剂的特性,并促进内皮通透性的降低。
    We studied changes of pulmonary microhemodynamics when modeling pulmonary artery thromboembolism on perfused isolated rabbit lungs after pretreatment with ranolazine and ivabradine. The increase in pulmonary artery pressure, pulmonary vascular resistance, and pre- and postcapillary resistance was less pronounced than in control animals, but was close to that in case of pulmonary thromboembolism after pretreatment with voltage-gated Na+ channel blockers lidocaine and ropivacaine. The increase of capillary filtration coefficient inversely correlated with values of capillary hydrostatic pressure. Thus, ranolazine and ivabradine exhibit the properties of voltage-gated Na+ channel blockers mainly in smooth muscles of pulmonary arterial vessels and promote the decrease in endothelial permeability.
    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
    ω-Grammotoxin-SIA(GrTX-SIA)最初是从智利玫瑰狼草的毒液中分离出来的,并被证明是电压门控Ca2(CaV)通道的门控改性剂。后来的实验表明,GrTX-SIA还可以通过类似的作用机制来抑制电压门控K(KV)通道电流,该机制涉及与电压感应域(VSD)中保守的S3-S4区域的结合。由于电压门控Na(NaV)通道含有同源结构基序,我们假设GrTX-SIA也可以抑制该离子通道家族的成员。这里,我们表明,GrTX-SIA确实可以阻止多个NaV通道亚型的门控过程,而NaV1.6是最易受影响的靶标。此外,GrTX-SIA在NaV1.6上的分子对接,由p.E1607K突变支持,揭示了IV域中的电压传感器(VSDIV)是主要的作用部位。电流抑制似乎发生的双相方式表明,可能是敏感性较低的结合位点,通过使用KV2.1/NaV1.6嵌合电压传感器构建体鉴定为VSDII。随后,NaV1.6p.E782K/p。E838K(VSDII),NaV1.6p.E1607K(VSDIV),特别是组合的VSDII/VSDIV突变体几乎失去了对GrTX-SIA的所有易感性。结合现有文献,我们的数据表明,GrTX-SIA识别NaV通道VSD中离子通道家族之间保守的模块,从而使其充当全面的离子通道门控修饰肽。
    ω-Grammotoxin-SIA (GrTX-SIA) was originally isolated from the venom of the Chilean rose tarantula and demonstrated to function as a gating modifier of voltage-gated Ca2+ (CaV) channels. Later experiments revealed that GrTX-SIA could also inhibit voltage-gated K+ (KV) channel currents via a similar mechanism of action that involved binding to a conserved S3-S4 region in the voltage-sensing domains (VSDs). Since voltage-gated Na+ (NaV) channels contain homologous structural motifs, we hypothesized that GrTX-SIA could inhibit members of this ion channel family as well. Here, we show that GrTX-SIA can indeed impede the gating process of multiple NaV channel subtypes with NaV1.6 being the most susceptible target. Moreover, molecular docking of GrTX-SIA onto NaV1.6, supported by a p.E1607K mutation, revealed the voltage sensor in domain IV (VSDIV) as being a primary site of action. The biphasic manner in which current inhibition appeared to occur suggested a second, possibly lower-sensitivity binding locus, which was identified as VSDII by using KV2.1/NaV1.6 chimeric voltage-sensor constructs. Subsequently, the NaV1.6p.E782K/p.E838K (VSDII), NaV1.6p.E1607K (VSDIV), and particularly the combined VSDII/VSDIV mutant lost virtually all susceptibility to GrTX-SIA. Together with existing literature, our data suggest that GrTX-SIA recognizes modules in NaV channel VSDs that are conserved among ion channel families, thereby allowing it to act as a comprehensive ion channel gating modifier peptide.
    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
    电压门控钠通道Nav1.2的功能障碍导致各种癫痫性疾病,和抑制通道已成为一种有吸引力的治疗策略。然而,目前可用的Nav1.2抑制剂表现出低效力和有限的结构多样性。在这项研究中,设计了一系列具有Nav1.2抑制活性的嘧啶基衍生物,合成,并进行了评估。化合物14和35对Nav1.2表现出有效的活性,IC50值为120和65nM,分别。化合物14在腹膜内注射后显示出有利的药代动力学(F=43%)和优异的脑渗透效力(B/P=3.6)。化合物14和35在最大电击测试中表现出强大的抗癫痫活性,ED50值为3.2和11.1mg/kg,分别。化合物35在6Hz(32mA)模型中也表现出有效的抗癫痫活性。ED50值为18.5mg/kg。总的来说,化合物14和35是开发新的癫痫小分子疗法的有希望的线索。
    Dysfunction of voltage-gated sodium channel Nav1.2 causes various epileptic disorders, and inhibition of the channel has emerged as an attractive therapeutic strategy. However, currently available Nav1.2 inhibitors exhibit low potency and limited structural diversity. In this study, a novel series of pyrimidine-based derivatives with Nav1.2 inhibitory activity were designed, synthesized, and evaluated. Compounds 14 and 35 exhibited potent activity against Nav1.2, boasting IC50 values of 120 and 65 nM, respectively. Compound 14 displayed favorable pharmacokinetics (F = 43%) following intraperitoneal injection and excellent brain penetration potency (B/P = 3.6). Compounds 14 and 35 exhibited robust antiepileptic activities in the maximal electroshock test, with ED50 values of 3.2 and 11.1 mg/kg, respectively. Compound 35 also demonstrated potent antiepileptic activity in a 6 Hz (32 mA) model, with an ED50 value of 18.5 mg/kg. Overall, compounds 14 and 35 are promising leads for the development of new small-molecule therapeutics for epilepsy.
    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
    在本期《细胞化学生物学》中,Elleman等人介绍了一种具有高时空分辨率的转化化学方法来控制神经元活动。作者介绍了STX-BPC,一种有效的神经毒素,天然抑制电压门控钠通道(Navs),补充可用的光遗传学方法来操纵神经元活动,蜂窝通信,和行为。
    In this issue of Cell Chemical Biology, Elleman et al.1 introduce a transformative chemical approach to control neuronal activity with high spatial and temporal resolution. The authors present STX-bpc, a potent neurotoxin that naturally inhibits voltage-gated sodium channels (NaVs), complementing available optogenetic methods for manipulating neuronal activity, cellular communication, and behavior.
    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
    背根神经节(DRG)是负责将外周疼痛信号传递到中枢神经系统的初级神经元,在疼痛转导中起着至关重要的作用。调节DRG兴奋性被认为是疼痛管理的可行方法。神经元兴奋性与神经元上的离子通道错综复杂地联系在一起。中小型DRG神经元主要参与疼痛传导,并具有高水平的TTX-S钠通道,Nav1.7约占目前的80%。电压门控钠通道(VGSC或Nav)阻滞剂是治疗中枢神经系统疾病的重要靶点,尤其是慢性疼痛。VGSCs在控制细胞兴奋性中起关键作用。临床研究表明,Nav1.7在疼痛感觉中起着至关重要的作用,并且有强有力的遗传证据将Nav1.7及其编码基因SCN9A基因与人类疼痛疾病联系起来。许多研究表明,Nav1.7在疼痛管理中起着重要作用。Nav1.7在疼痛信号通路中的作用使其成为潜在开发新型疼痛药物的有吸引力的靶标。同时,了解Nav1.7的结构可能有助于开发下一代止痛药。这篇综述提供了最近报道的针对Nav1.7途径的分子抑制剂的最新信息。总结了它们的结构-活性关系(SARs),并讨论了它们对疼痛疾病的治疗效果。药物化学家正在努力提高Nav1.7抑制剂的治疗指数,达到更好的镇痛效果,减少副作用。我们希望这篇综述将有助于开发新型Nav1.7抑制剂作为潜在药物。
    The dorsal root ganglion (DRG) is the primary neuron responsible for transmitting peripheral pain signals to the central nervous system and plays a crucial role in pain transduction. Modulation of DRG excitability is considered a viable approach for pain management. Neuronal excitability is intricately linked to the ion channels on the neurons. The small and medium-sized DRG neurons are chiefly engaged in pain conduction and have high levels of TTX-S sodium channels, with Nav1.7 accounting for approximately 80% of the current. Voltage-gated sodium channel (VGSC or Nav) blockers are vital targets for the management of central nervous system diseases, particularly chronic pain. VGSCs play a key role in controlling cellular excitability. Clinical research has shown that Nav1.7 plays a crucial role in pain sensation, and there is strong genetic evidence linking Nav1.7 and its encoding gene SCN9A gene to painful disorders in humans. Many studies have shown that Nav1.7 plays an important role in pain management. The role of Nav1.7 in pain signaling pathways makes it an attractive target for the potential development of new pain drugs. Meanwhile, understanding the architecture of Nav1.7 may help to develop the next generation of painkillers. This review provides updates on the recently reported molecular inhibitors targeting the Nav1.7 pathway, summarizes their structure-activity relationships (SARs), and discusses their therapeutic effects on painful diseases. Pharmaceutical chemists are working to improve the therapeutic index of Nav1.7 inhibitors, achieve better analgesic effects, and reduce side effects. We hope that this review will contribute to the development of novel Nav1.7 inhibitors as potential drugs.
    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
    慢性疼痛是一个常见且具有挑战性的临床问题,严重影响患者的生活质量。钠通道Nav1.8在慢性疼痛的发生发展中起着至关重要的作用,使其成为治疗慢性疼痛的关键目标之一。在这篇文章中,我们将虚拟筛选与细胞膜层析技术相结合,建立了一种快速高通量筛选Nav1.8选择性抑制剂的新方法。使用这种方法,我们鉴定了一种小分子化合物6,其不仅表现出对Nav1.8的高亲和力和抑制活性,而且还表现出对CFA诱导的慢性炎性疼痛的显著抑制作用。与阳性药物VX-150相比,化合物6显示出更长时间的镇痛作用,使其成为具有潜在临床应用的Nav1.8抑制剂的有希望的候选物。这一发现为慢性疼痛的治疗提供了新的治疗选择。
    Chronic pain is a common and challenging clinical problem that significantly impacts patients\' quality of life. The sodium channel Nav1.8 plays a crucial role in the occurrence and development of chronic pain, making it one of the key targets for treating chronic pain. In this article, we combined virtual screening with cell membrane chromatography techniques to establish a novel method for rapid high-throughput screening of selective Nav1.8 inhibitors. Using this approach, we identified a small molecule compound 6, which not only demonstrated high affinity and inhibitory activity against Nav1.8 but also exhibited significant inhibitory effects on CFA-induced chronic inflammatory pain. Compared to the positive drug VX-150, compound 6 showed a more prolonged analgesic effect, making it a promising candidate as a Nav1.8 inhibitor with potential clinical applications. This discovery provides a new therapeutic option for the treatment of chronic pain.
    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
    电压门控钠(NaV)通道与动作电位的产生和传递密切相关,这些通道的功能障碍可能会导致神经系统疾病,如癫痫,神经性疼痛,精神病,自闭症和心律失常.许多毒液肽选择性地作用于NaV通道。这些包括香菇毒素,它们是由蜗牛分泌的神经毒素,用于捕获猎物或自卫,但是它们也是用于识别和/或治疗人类疾病的有价值的药理学工具。通常,conotoxins含有两个或三个二硫键,这些内部交叉括号有助于conotoxins具有紧密的,明确的结构和高稳定性。在含有三个二硫键的芋螺毒素中,一些选择性靶向哺乳动物NaV通道,刺激,或调节这些频道。这样的芋螺毒素具有作为用于研究NaV通道的功能和特征的药理学工具或作为与NaV通道相关的神经疾病的药物线索的巨大潜力。因此,发现或设计具有高效力和选择性的靶向NaV通道的螺毒素是重要的。氨基酸序列,二硫键连接,三维结构是影响芋螺毒素生物活性的关键因素,和有针对性的合成改性的芋螺毒素可以大大提高其活性和选择性。这篇综述研究了NaV通道靶向的螺毒素,专注于它们的结构,活动和设计的修改,以扩大其应用范围。意义声明NaV通道在各种神经系统疾病中至关重要。一些芋螺毒素选择性地靶向NaV通道,导致封锁或激活,从而使它们能够用作药理学工具来研究通道的特性和功能。二恶英毒素也具有被开发为药物先导物的有希望的潜力。这些肽中的二硫键对于稳定它们的结构很重要,从而导致增强的特异性和效力。一起,以NaV通道为靶点的芋螺毒素既有研究价值,也有很好的应用前景。
    Voltage-gated sodium (NaV) channels are intimately involved in the generation and transmission of action potentials, and dysfunction of these channels may contribute to nervous system diseases, such as epilepsy, neuropathic pain, psychosis, autism, and cardiac arrhythmia. Many venom peptides selectively act on NaV channels. These include conotoxins, which are neurotoxins secreted by cone snails for prey capture or self-defense but which are also valuable pharmacological tools for the identification and/or treatment of human diseases. Typically, conotoxins contain two or three disulfide bonds, and these internal crossbraces contribute to conotoxins having compact, well defined structures and high stability. Of the conotoxins containing three disulfide bonds, some selectively target mammalian NaV channels and can block, stimulate, or modulate these channels. Such conotoxins have great potential to serve as pharmacological tools for studying the functions and characteristics of NaV channels or as drug leads for neurologic diseases related to NaV channels. Accordingly, discovering or designing conotoxins targeting NaV channels with high potency and selectivity is important. The amino acid sequences, disulfide bond connectivity, and three-dimensional structures are key factors that affect the biological activity of conotoxins, and targeted synthetic modifications of conotoxins can greatly improve their activity and selectivity. This review examines NaV channel-targeted conotoxins, focusing on their structures, activities, and designed modifications, with a view toward expanding their applications. SIGNIFICANCE STATEMENT: NaV channels are crucial in various neurologic diseases. Some conotoxins selectively target NaV channels, causing either blockade or activation, thus enabling their use as pharmacological tools for studying the channels\' characteristics and functions. Conotoxins also have promising potential to be developed as drug leads. The disulfide bonds in these peptides are important for stabilizing their structures, thus leading to enhanced specificity and potency. Together, conotoxins targeting NaV channels have both immediate research value and promising future application prospects.
    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
    暂无摘要。
    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
    疼痛,影响全球数百万人的复杂和令人衰弱的状况,是一个重要的问题,尤其是在术后恢复的背景下。这篇全面的综述探讨了疼痛的复杂性及其全球影响,强调电压门控钠通道(VGSC或NaV通道)的调节是疼痛管理的有希望的途径,目的是减少对阿片类药物的依赖。本文深入研究了特定NaV同工型的作用,特别是疼痛过程中的NaV1.7,NaV1.8和NaV1.9,并讨论了钠通道阻滞剂的发展以精确地靶向这些同工型。传统局部麻醉药和选择性NaV亚型抑制剂,尽管在疼痛管理中表现出不同的功效,在系统分布和潜在副作用方面面临挑战。该综述强调了纳米医学在改善局部麻醉药递送方面的潜力,毒素和选择性NaV亚型抑制剂,用于在疼痛部位的靶向和持续释放。这一创新战略旨在提高药物的生物利用度,尽量减少全身暴露,优化治疗结果,为从外科手术中恢复或患有慢性疼痛的患者提供安全的疼痛管理和提高生活质量的重要希望。
    Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic pain.
    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