Voltage-Gated Sodium Channel Blockers

电压门控钠通道阻滞剂
  • 文章类型: Journal Article
    在传递疼痛信号的感觉神经元中,无论是急性还是慢性,电压门控钠通道(VGSCs)对调节兴奋性至关重要。NaV1.1,NaV1.3,NaV1.6,NaV1.7,NaV1.8和NaV1.9已被证明并根据其生物物理特性和感觉神经元每种亚型中不同的表达模式定义了它们在疼痛信号传导中的功能作用。蝎子和蜘蛛是传统的中药材,属于蜘蛛类。它们的大多数研究物种都进化出了毒液肽,这些毒液肽表现出多种具有亚型选择性和构象特异性的特异性靶向VGSC的结蛋白。这篇综述概述了蝎子和蜘蛛毒液针对疼痛相关的NaV通道的精致结结,描述序列和结构特征以及影响其对特殊亚型和特定构象的选择性的分子决定子,目的是开发NaV通道和镇痛药的新型研究工具,而不良反应最小。
    In sensory neurons that transmit pain signals, whether acute or chronic, voltage-gated sodium channels (VGSCs) are crucial for regulating excitability. NaV1.1, NaV1.3, NaV1.6, NaV1.7, NaV1.8, and NaV1.9 have been demonstrated and defined their functional roles in pain signaling based on their biophysical properties and distinct patterns of expression in each subtype of sensory neurons. Scorpions and spiders are traditional Chinese medicinal materials, belonging to the arachnid class. Most of the studied species of them have evolved venom peptides that exhibit a wide variety of knottins specifically targeting VGSCs with subtype selectivity and conformational specificity. This review provides an overview on the exquisite knottins from scorpion and spider venoms targeting pain-related NaV channels, describing the sequences and the structural features as well as molecular determinants that influence their selectivity on special subtype and at particular conformation, with an aim for the development of novel research tools on NaV channels and analgesics with minimal adverse effects.
    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

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:花椒.(Z.bungeanum),菊科的一员,在亚洲有丰富的传统使用历史来治疗关节炎和牙痛。作为特有的化学成分,从植物中提取了多种生物碱,并报道了其多种生物活性。然而,异喹啉生物碱的研究,一种特定类型的生物碱,在Z.bungeanum是稀缺的。
    目的:本研究旨在从香根草中分离出一种新型异喹啉类生物碱,并探讨其体外药理活性和体内镇痛活性。
    方法:采用色谱和光谱方法从Z.bungeanum中分离和鉴定异喹啉生物碱。全细胞膜片钳技术用于评估其对神经元兴奋性的影响,和急性分离的小鼠小直径背根神经节(DRG)神经元中的内源性电压门控钾(Kv)和钠(Nav)电流。用稳定表达Nav1.7和Nav1.8的HEK293细胞和瞬时表达Kv2.1的中国仓鼠卵巢(CHO)细胞进一步验证了其对通道的抑制作用。福尔马林炎性疼痛模型用于评估体内潜在的镇痛活性。
    结果:首次从Z.bungeanum中分离并鉴定了一种新的异喹啉生物碱,命名为HJ-69(N-13-(3-甲氧基丙-1-基)卢卡地平)。HJ-69可显着抑制DRG神经元中动作电位的激发频率和振幅。始终如一,它状态依赖性地抑制DRG神经元的内源性Nav电流,失活和静息状态的半数最大抑制浓度(IC50)值为13.06±2.06μM和30.19±2.07μM,分别。HJ-69显著抑制DRG神经元的钾电流,显着抑制了延迟整流钾(IK)电流(IC50=6.95±1.29μM),并略微影响了瞬态外向钾(IA)电流(IC50=523.50±39.16μM)。更进一步,HJ-69在异源表达的Nav1.7,Nav1.8和Kv2.1通道上表现出相似的效力,它们相应地代表了神经元中的主要成分。值得注意的是,腹膜内给药30mg/kg和100mg/kgHJ-69可显着减轻福尔马林诱导的小鼠炎性疼痛模型的疼痛行为。
    结论:该研究得出结论,HJ-69是一种新颖的活性异喹啉生物碱,Nav和Kv通道的抑制有助于其镇痛活性。HJ-69可能是基于异喹啉生物碱的未来镇痛药物发现的有希望的原型。
    BACKGROUND: Zanthoxylum bungeanum Maxim. (Z. bungeanum), a member of the Rutaceae family, has a rich history of traditional use in Asia for treating arthritis and toothache conditions. As characteristic chemical components, numerous kinds of alkaloids have been extracted from plants and their diverse biological activities have been reported. However, research on the isoquinoline alkaloid, a specific type of alkaloids, in Z. bungeanum was scarce.
    OBJECTIVE: The study aimed to isolate a novel isoquinoline alkaloid from Z. bungeanum and explore its pharmacological activity in vitro and analgesic activity in vivo.
    METHODS: Isoquinoline alkaloid isolation and identification from Z. bungeanum were conducted using chromatographic and spectroscopic methods. The whole-cell patch-clamp technique was applied to assess its impact on neuronal excitability, and endogenous voltage-gated potassium (Kv) and sodium (Nav) currents in acutely isolated mouse small-diameter dorsal root ganglion (DRG) neurons. Its inhibitory impacts on channels were further validated with HEK293 cells stably expressing Nav1.7 and Nav1.8, and Chinese hamster ovary (CHO) cells transiently expressing Kv2.1. The formalin inflammatory pain model was utilized to evaluate the potential analgesic activity in vivo.
    RESULTS: A novel isoquinoline alkaloid named HJ-69 (N-13-(3-methoxyprop-1-yl)rutaecarpine) was isolated and identified from Z. bungeanum for the first time. HJ-69 significantly suppressed the firing frequency and amplitudes of action potentials in DRG neurons. Consistently, it state-dependently inhibited endogenous Nav currents of DRG neurons, with half maximal inhibitory concentration (IC50) values of 13.06 ± 2.06 μM and 30.19 ± 2.07 μM for the inactivated and resting states, respectively. HJ-69 significantly suppressed potassium currents in DRG neurons, which notably inhibited the delayed rectifier potassium (IK) currents (IC50 = 6.95 ± 1.29 μM) and slightly affected the transient outward potassium (IA) currents (IC50 = 523.50 ± 39.16 μM). Furtherly, HJ-69 exhibited similar potencies on heterologously expressed Nav1.7, Nav1.8, and Kv2.1 channels, which correspondingly represent the main components in neurons. Notably, intraperitoneal administration of 30 mg/kg and 100 mg/kg HJ-69 significantly alleviated pain behaviors in the mouse inflammatory pain model induced by formalin.
    CONCLUSIONS: The study concluded that HJ-69 is a novel and active isoquinoline alkaloid, and the inhibition of Nav and Kv channels contributes to its analgesic activity. HJ-69 may be a promising prototype for future analgesic drug discovery based on the isoquinoline alkaloid.
    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
    艾塞那肽,一种很有前途的心脏保护剂,防止心脏结构重塑和舒张功能障碍。钠和钾通道的联合阻断对于控制心房颤动(AF)很有价值。这里,我们探讨了艾塞那肽是否通过抑制人Kv1.5和Nav1.5通道而发挥抗AF作用.我们使用全细胞膜片钳技术来研究艾塞那肽对人胚肾293细胞中表达的hKv1.5和hNav1.5通道的影响,并研究了艾塞那肽对大鼠心房肌细胞动作电位(AP)和其他心脏离子电流的影响。此外,采用电标测系统研究艾塞那肽对离体大鼠心脏电特性和房颤活性的影响.最后,大鼠房颤模型,使用乙酰胆碱和氯化钙建立,用于评估艾塞那肽在大鼠中的抗AF潜力。艾塞那肽可逆地抑制IKv1.5,IC50为3.08μM,优先阻断HKv1.5通道在其关闭状态,并正向移动电压依赖性激活曲线。艾塞那肽也可逆地抑制INav1.5,IC50为3.30μM,负移动电压依赖性失活曲线,并在5和10Hz时以显着的使用依赖性减慢了其从失活中的恢复。此外,艾塞那肽延长AP持续时间并抑制持续K+电流(Iss)和瞬时外向K+电流(Ito),但不抑制L型Ca2+电流(ICa,L)在年夜鼠心房肌细胞中。艾塞那肽可预防大鼠心脏和大鼠的AF发生率和持续时间。这些发现表明艾塞那肽在体外抑制IKv1.5和INav1.5,并降低离体大鼠心脏和大鼠的AF易感性。
    Exenatide, a promising cardioprotective agent, protects against cardiac structural remodeling and diastolic dysfunction. Combined blockade of sodium and potassium channels is valuable for managing atrial fibrillation (AF). Here, we explored whether exenatide displayed anti-AF effects by inhibiting human Kv1.5 and Nav1.5 channels. We used the whole-cell patch-clamp technique to investigate the effects of exenatide on hKv1.5 and hNav1.5 channels expressed in human embryonic kidney 293 cells and studied the effects of exenatide on action potential (AP) and other cardiac ionic currents in rat atrial myocytes. Additionally, an electrical mapping system was used to explore the effects of exenatide on electrical properties and AF activity in isolated rat hearts. Finally, a rat AF model, established using acetylcholine and calcium chloride, was employed to evaluate the anti-AF potential of exenatide in rats. Exenatide reversibly suppressed IKv1.5 with IC50 of 3.08 μM, preferentially blocked the hKv1.5 channel in its closed state, and positively shifted the voltage-dependent activation curve. Exenatide also reversibly inhibited INav1.5 with IC50 of 3.30 μM, negatively shifted the voltage-dependent inactivation curve, and slowed its recovery from inactivation with significant use-dependency at 5 and 10 Hz. Furthermore, exenatide prolonged AP duration and suppressed the sustained K+ current (Iss) and transient outward K+ current (Ito), but without inhibition of L-type Ca2+ current (ICa,L) in rat atrial myocytes. Exenatide prevented AF incidence and duration in rat hearts and rats. These findings demonstrate that exenatide inhibits IKv1.5 and INav1.5in vitro and reduces AF susceptibility in isolated rat hearts and rats.
    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
    背景:慢性口面部疼痛(COP)治疗具有挑战性,因为目前的医学治疗非常缺乏。牡丹皮(MC)是一种广泛用于慢性炎症性疾病的中药。然而,COP治疗中MC背后的机制尚未建立.这项研究的目的是确定MC的活性成分及其在COP治疗中的特定潜在机制。
    方法:在本研究中,通过网络药理学和生物信息学分析,确定了MC在COP治疗中的主要活性成分和复合靶点网络。成年雄性Sprague-Dawley大鼠接受口腔粘膜脂多糖(LPS)注射诱导COP。神经节内注射后,通过口面部机械伤害性评估来评估疼痛行为。通过实时定量聚合酶链反应(RT-qPCR)定量LPS预处理的人牙周膜干细胞(hPDLSCs)和大鼠原代培养三叉神经节(TG)神经元中的体外炎性细胞因子。使用Schrödinger软件验证槲皮素与关键靶标的分子对接。全细胞记录电生理用于评估槲皮素对大鼠TG神经元电压门控钠(Nav)通道的影响。
    结果:组装的化合物-靶标网络由4个化合物和46个靶标组成。由于MC的1个活性成分与最相关的目标相关,槲皮素减轻LPS诱导的COP大鼠模型的机械异常性疼痛(给药后0.5小时的机械异常性疼痛阈值中位数[四分位距(IQR):载体1.3[0.6-2.0]g与槲皮素7.0[6.0-8.5]g,P=.002)。基因本体论(GO)富集和京都基因和基因组百科全书(KEGG)途径分析显示,免疫反应和膜功能在MC-COP治疗中起着至关重要的作用。通过蛋白质-蛋白质相互作用分析将5个相关靶标鉴定为核心靶标。槲皮素有镇痛作用,可能通过阻断TG感觉神经元的Nav通道(峰值电流密度中位数[IQR]:LPS-850.2[-983.6至-660.7]mV与LPS槲皮素-589.6[-711.0至-147.8]mV,P=.006),同时下调促炎细胞因子-FOS的表达水平(标准化信使RNA[mRNA]水平平均值±平均值标准误差[SEM]:LPS[2.22±0.33]与LPS+槲皮素[1.33±0.14],P=.034)和TNF-α(标准化mRNA水平平均值±SEM:LPS[8。93±0.78]与LPS+槲皮素[3.77±0.49],P<.0001)。
    结论:确定Nav作为槲皮素的分子靶标阐明了MC的镇痛机制,为新型选择性、高效的慢性止痛药的开发提供了思路。
    BACKGROUND: Chronic orofacial pain (COP) therapy is challenging, as current medical treatments are extremely lacking. Moutan Cortex (MC) is a traditional Chinese medicine herb widely used for chronic inflammatory diseases. However, the mechanism behind MC in COP therapy has not been well-established. The purpose of this study was to identify the active ingredients of MC and their specific underlying mechanisms in COP treatment.
    METHODS: In this study, the main active ingredients and compound-target network of MC in COP therapy were identified through network pharmacology and bioinformatics analysis. Adult male Sprague-Dawley rats received oral mucosa lipopolysaccharide (LPS) injection to induce COP. Pain behaviors were evaluated by orofacial mechanical nociceptive assessment after intraganglionar injection. In vitro inflammatory cytokines in LPS-pretreated human periodontal ligament stem cells (hPDLSCs) and rat primary cultural trigeminal ganglion (TG) neurons were quantified by real-time quantitative polymerase chain reaction (RT-qPCR). Schrödinger software was used to verify the molecular docking of quercetin and critical targets. Whole-cell recording electrophysiology was used to evaluate the effect of quercetin on voltage-gated sodium (Na v ) channel in rat TG neurons.
    RESULTS: The assembled compound-target network consisted of 4 compounds and 46 targets. As 1 of the active components of MC correlated with most related targets, quercetin alleviated mechanical allodynia in LPS-induced rat model of COP (mechanical allodynia threshold median [interquartile range (IQR) 0.5 hours after drug administration: vehicle 1.3 [0.6-2.0] g vs quercetin 7.0 [6.0-8.5] g, P = .002). Gene ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that immune response and membrane functions play essential roles in MC-COP therapy. Five of the related targets were identified as core targets by protein-protein interaction analysis. Quercetin exerted an analgesic effect, possibly through blocking Na v channel in TG sensory neurons (peak current density median [IQR]: LPS -850.2 [-983.6 to -660.7] mV vs LPS + quercetin -589.6 [-711.0 to -147.8] mV, P = .006) while downregulating the expression level of proinflammatory cytokines-FOS (normalized messenger RNA [mRNA] level mean ± standard error of mean [SEM]: LPS [2. 22 ± 0.33] vs LPS + quercetin [1. 33 ± 0.14], P = .034) and TNF-α (normalized mRNA level mean ± SEM: LPS [8. 93 ± 0.78] vs LPS + quercetin [3. 77 ± 0.49], P < .0001).
    CONCLUSIONS: Identifying Na v as the molecular target of quercetin clarifies the analgesic mechanism of MC, and provides ideas for the development of novel selective and efficient chronic pain relievers.
    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
    电压门控钠(Nav)通道是许多广泛使用和研究的癫痫治疗药物的目标,心律失常,疼痛,和其他疾病。尽管最近在导航通道的结构阐明方面取得了进展,大多数Nav靶向药物的结合模式仍然未知.在这里,我们报告了用药物和铅化合物处理的人Nav1.7的高分辨率低温EM结构,其代表性化学骨架的分辨率为2.6-3.2µ。细胞内门下方的结合位点(位点BIG)容纳卡马西平,布比卡因,还有Lacosamide.出乎意料的是,拉科酰胺的第二个分子从中央腔插入选择性过滤器。Fenestation是各种状态依赖性药物的流行场所。我们证明长春西汀,长春花生物碱的合成衍生物,和硬维甲酸,一种具有抗伤害作用的天然产品,结合III-IV开窗,而维索三嗪,镇痛药候选人,穿透孔域的IV-I开窗。我们的结果允许从现有和以前的结构中总结出Nav通道上已知药物结合位点的3D结构图。
    Voltage-gated sodium (Nav) channels are targeted by a number of widely used and investigational drugs for the treatment of epilepsy, arrhythmia, pain, and other disorders. Despite recent advances in structural elucidation of Nav channels, the binding mode of most Nav-targeting drugs remains unknown. Here we report high-resolution cryo-EM structures of human Nav1.7 treated with drugs and lead compounds with representative chemical backbones at resolutions of 2.6-3.2 Å. A binding site beneath the intracellular gate (site BIG) accommodates carbamazepine, bupivacaine, and lacosamide. Unexpectedly, a second molecule of lacosamide plugs into the selectivity filter from the central cavity. Fenestrations are popular sites for various state-dependent drugs. We show that vinpocetine, a synthetic derivative of a vinca alkaloid, and hardwickiic acid, a natural product with antinociceptive effect, bind to the III-IV fenestration, while vixotrigine, an analgesic candidate, penetrates the IV-I fenestration of the pore domain. Our results permit building a 3D structural map for known drug-binding sites on Nav channels summarized from the present and previous structures.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号