NAV1.9 Voltage-Gated Sodium Channel

NAV1.9 电压门控钠通道
  • 文章类型: Journal Article
    家族性发作性疼痛综合征(FEPS)是一种儿童早期发作的严重发作性肢体疼痛疾病,主要由SCN11A的致病变体引起,SCN10A,SCN9A,它们编码三个电压门控钠通道(VGSCs),在初级感觉神经元中表达为伤害感受器兴奋性的关键决定因素。可能仍有许多未确诊的FEPS患者。更好地理解相关的发病机制,流行病学,和临床特征需要提供适当的诊断和护理。对于这项研究,在全国范围内招募日本患者是使用临时临床诊断标准进行的,其次是SCN11A基因检测,SCN10A,SCN9A在招募的212名患者中,基因检测显示,64名患者(30.2%)携带这些基因的致病性或可能的致病性变异,由42(19.8%)组成,14(6.60%),和8例(3.77%)患有SCN11A变异的患者,SCN10A,SCN9A,分别。同时,符合暂定临床标准的患者比例为89.1%,52.0%,在具有三个基因中的每一个的致病性或可能致病性变异的患者中,有54.5%,表明这些临床标准的有效性,特别是SCN11A变异的患者。FEPS的这些临床诊断标准将加速在日本意外流行的具有潜在致病变异的患者的招募。
    Familial episodic pain syndrome (FEPS) is an early childhood onset disorder of severe episodic limb pain caused mainly by pathogenic variants of SCN11A, SCN10A, and SCN9A, which encode three voltage-gated sodium channels (VGSCs) expressed as key determinants of nociceptor excitability in primary sensory neurons. There may still be many undiagnosed patients with FEPS. A better understanding of the associated pathogenesis, epidemiology, and clinical characteristics is needed to provide appropriate diagnosis and care. For this study, nationwide recruitment of Japanese patients was conducted using provisional clinical diagnostic criteria, followed by genetic testing for SCN11A, SCN10A, and SCN9A. In the cohort of 212 recruited patients, genetic testing revealed that 64 patients (30.2%) harbored pathogenic or likely pathogenic variants of these genes, consisting of 42 (19.8%), 14 (6.60%), and 8 (3.77%) patients with variants of SCN11A, SCN10A, and SCN9A, respectively. Meanwhile, the proportions of patients meeting the tentative clinical criteria were 89.1%, 52.0%, and 54.5% among patients with pathogenic or likely pathogenic variants of each of the three genes, suggesting the validity of these clinical criteria, especially for patients with SCN11A variants. These clinical diagnostic criteria of FEPS will accelerate the recruitment of patients with underlying pathogenic variants who are unexpectedly prevalent in Japan.
    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
    电压门控钠通道亚型,Nav1.7、Nav1.8和Nav1.9主要在外周感觉神经元中表达。最近的遗传研究表明,它们参与病理性疼痛的处理,并且Nav1.7,Nav1.8或Nav1.9的阻断将成为有前途的药物疗法,尤其是对于神经性疼痛。越来越多的药物发现计划已针对任一亚型获得选择性抑制剂,该抑制剂可在不影响心血管和中枢神经系统的情况下缓解疼痛。尽管它们都没有被批准。在这里,我们描述了ANP-230的体外特征,这是一种正在临床开发中的新型钠通道阻滞剂。令人惊讶的是,显示ANP-230可阻断三种疼痛相关亚型,具有相似效力的人Nav1.7、Nav1.8和Nav1.9,但对人心脏Nav1.5通道和大鼠中枢Nav通道的抑制活性较低。使用不同阶跃脉冲协议的电压钳实验表明,ANP-230具有“强直阻滞”作用模式,没有状态和使用依赖性。此外,ANP-230在人Nav1.7稳定表达细胞中引起激活曲线的去极化偏移并减慢门控动力学。在人Nav1.8稳定表达细胞以及大鼠背根神经节神经元中通常观察到激活曲线的去极化位移。这些数据表明ANP-230抑制Nav通道的非常独特的机制。最后,ANP-230以浓度依赖的方式降低大鼠背根神经节神经元的兴奋性。总的来说,这些有希望的结果表明ANP-230可能是治疗神经性疼痛的有效药物.
    Voltage-gated sodium channel subtypes, Nav1.7, Nav1.8, and Nav1.9 are predominantly expressed in peripheral sensory neurons. Recent genetic studies have revealed that they are involved in pathological pain processing and that the blockade of Nav1.7, Nav1.8, or Nav1.9 will become a promising pharmacotherapy especially for neuropathic pain. A growing number of drug discovery programs have targeted either of the subtypes to obtain a selective inhibitor which can provide pain relief without affecting the cardiovascular and central nervous systems, though none of them has been approved yet. Here we describe the in vitro characteristics of ANP-230, a novel sodium channel blocker under clinical development. Surprisingly, ANP-230 was shown to block three pain-related subtypes, human Nav1.7, Nav1.8, and Nav1.9 with similar potency, but had only low inhibitory activity to human cardiac Nav1.5 channel and rat central Nav channels. The voltage clamp experiments using different step pulse protocols revealed that ANP-230 had a \"tonic block\" mode of action without state- and use-dependency. In addition, ANP-230 caused a depolarizing shift of the activation curve and decelerated gating kinetics in human Nav1.7-stably expressing cells. The depolarizing shift of activation curve was commonly observed in human Nav1.8-stably expressing cells as well as rat dorsal root ganglion neurons. These data suggested a quite unique mechanism of Nav channel inhibition by ANP-230. Finally, ANP-230 reduced excitability of rat dorsal root ganglion neurons in a concentration dependent manner. Collectively, these promising results indicate that ANP-230 could be a potent drug for neuropathic 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
    抑郁症(MDD)的发病机制涉及脂质代谢。我们早期的研究还表明,MDD患者的总胆固醇(TC)浓度远低于健康对照(HCs)。然而,目前尚不清楚为什么MDD中TC降低。这里,基于独创性知识库的独创性途径分析,我们发现钠电压门控通道α亚基11A(SCN11A)可能是低脂水平与MDD之间的联系.我们分析了TC水平,并使用ELISA试剂盒测量了139例MDD患者血清中SCN11A的水平,和65个HC来证实这一理论,并探索SCN11A在MDD中的潜在参与。结果显示,MDD患者的TC水平明显低于HCs,SCN11A水平显着升高,而药物治疗的MDD患者比药物初治的MDD患者明显逆转。在使用单一或多种抗抑郁药的MDD患者中,SCN11A水平没有显着差异,和选择性5-羟色胺再摄取抑制剂或其他抗抑郁药。Pearson相关分析显示,TC和SCN11A水平与汉密尔顿抑郁量表评分相关。在TC和SCN11A之间也发现了实质性的关联。此外,发现了由SCN11A组成的判别模型,这在训练集中的曲线下产生了0.9571的面积,在测试集中产生了0.9357的面积。一起来看,我们的研究结果表明,SCN11A可能是低脂水平与MDD之间的联系,并有望成为MDD的候选生物标志物。
    The pathogenesis of major depressive disorder (MDD) involves lipid metabolism. Our earlier research also revealed that MDD patients had much lower total cholesterol (TC) concentrations than healthy controls (HCs). However, it is still unclear why TC decreased in MDD. Here, based on the Ingenuity Knowledge Base\'s ingenuity pathway analysis, we found that sodium voltage-gated channel alpha subunit 11A (SCN11A) might serve as a link between low lipid levels and MDD. We analyzed the TC levels and used ELISA kits to measure the levels of SCN11A in the serum from 139 MDD patients, and 65 HCs to confirm this theory and explore the potential involvement of SCN11A in MDD. The findings revealed that TC levels were considerably lower and SCN11A levels were remarkably increased in MDD patients than those in HCs, while they were significantly reversed in drug-treatment MDD patients than in drug-naïve MDD patients. There was no significant difference in SCN11A levels among MDD patients who used single or multiple antidepressants, and selective serotonin reuptake inhibitors or other antidepressants. Pearson correlation analysis showed that the levels of TC and SCN11A were linked with the Hamilton Depression Rating Scales score. A substantial association was also found between TC and SCN11A. Moreover, a discriminative model made up of SCN11A was discovered, which produced an area under a curve of 0.9571 in the training set and 0.9357 in the testing set. Taken together, our findings indicated that SCN11A may serve as a link between low lipid levels and MDD, and showed promise as a candidate biomarker for MDD.
    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
    Nav1.9通道是电压门控钠通道。它在炎症后疼痛的产生和神经元过度兴奋的形成中起着至关重要的作用。它在肠神经系统的背根神经节的小直径神经元和DogielII神经元中高度表达。背根神经节中的小直径神经元是疼痛传导的主要感觉神经元。Nav1.9通道也参与调节肠运动。Nav1.9通道的功能增强在一定程度上导致小直径背根神经节神经元的兴奋过度。神经元的过度兴奋可引起内脏痛觉过敏。肠神经系统中的肠传入神经元和内在初级传入神经元属于DogielII型神经元。它们的兴奋性也可以通过Nav1.9通道调节。肠fugal传入神经元的过度兴奋异常激活肠-肠抑制反射。内在初级传入神经元的过度兴奋通过异常激活蠕动反射来干扰蠕动波。这篇综述讨论了Nav1.9通道在肠道肥大和运动障碍中的作用。
    The Nav1.9 channel is a voltage-gated sodium channel. It plays a vital role in the generation of pain and the formation of neuronal hyperexcitability after inflammation. It is highly expressed in small diameter neurons of dorsal root ganglions and Dogiel II neurons in enteric nervous system. The small diameter neurons in dorsal root ganglions are the primary sensory neurons of pain conduction. Nav1.9 channels also participate in regulating intestinal motility. Functional enhancements of Nav1.9 channels to a certain extent lead to hyperexcitability of small diameter dorsal root ganglion neurons. The hyperexcitability of the neurons can cause visceral hyperalgesia. Intestinofugal afferent neurons and intrinsic primary afferent neurons in enteric nervous system belong to Dogiel type II neurons. Their excitability can also be regulated by Nav1.9 channels. The hyperexcitability of intestinofugal afferent neurons abnormally activate entero-enteric inhibitory reflexes. The hyperexcitability of intrinsic primary afferent neurons disturb peristaltic waves by abnormally activating peristaltic reflexes. This review discusses the role of Nav1.9 channels in intestinal hyperpathia and dysmotility.
    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
    钠通道Nav1.9在传递疼痛信号的小直径背根神经节的感觉神经元中表达,和功能获得Nav1.9突变与疼痛和无痛性疾病有关。我们最初确定一些Nav1.9突变是在日本人群中观察到的家族性发作性疼痛综合征的原因。因此,我们产生了带有一种更痛苦的日本突变的模型小鼠,R222S,并确定背根神经节过度兴奋是相关疼痛的原因。ANP-230是一种新型非阿片类药物,对Nav1.7、1.8和1.9具有很强的抑制作用,目前正在接受家族性发作性疼痛综合征患者的临床试验。然而,对其作用机制和对疼痛敏感性的影响知之甚少。在这项研究中,因此,我们研究了ANP-230对Nav1.9p.R222S突变模型小鼠对疼痛的超敏反应的抑制作用。在行为测试中,ANP-230降低了小鼠的疼痛反应,特别是热或机械刺激,以浓度和时间依赖的方式。此外,ANP-230抑制了这些突变小鼠的背根神经节神经元的重复放电。我们的结果清楚地表明,ANP-230是由DRG神经元兴奋过度引起的家族性发作性疼痛综合征的有效镇痛剂,这种镇痛作用可能具有临床意义。
    The sodium channel Nav1.9 is expressed in the sensory neurons of small diameter dorsal root ganglia that transmit pain signals, and gain-of-function Nav1.9 mutations have been associated with both painful and painless disorders. We initially determined that some Nav1.9 mutations are responsible for familial episodic pain syndrome observed in the Japanese population. We therefore generated model mice harboring one of the more painful Japanese mutations, R222S, and determined that dorsal root ganglia hyperexcitability was the cause of the associated pain. ANP-230 is a novel non-opioid drug with strong inhibitory effects on Nav1.7, 1.8 and 1.9, and is currently under clinical trials for patients suffering from familial episodic pain syndrome. However, little is known about its mechanism of action and effects on pain sensitivity. In this study, we therefore investigated the inhibitory effects of ANP-230 on the hypersensitivity of Nav1.9 p.R222S mutant model mouse to pain. In behavioral tests, ANP-230 reduced the pain response of the mice, particularly to heat or mechanical stimuli, in a concentration- and time-dependent manner. Furthermore, ANP-230 suppressed the repetitive firing of dorsal root ganglion neurons of these mutant mice. Our results clearly demonstrate that ANP-230 is an effective analgesic for familial episodic pain syndrome resulting from DRG neuron hyperexcitability, and that such analgesic effects are likely to be of clinical significance.
    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
    通过比较野生型与野生型的反应来评估NaV1.9对炎症介质诱导的气道迷走神经滴注C纤维活化的影响NaV1.9-/-小鼠。单细胞rt-PCR分析表明,几乎所有的nodoseC纤维神经元都表达NaV1.9(SCN11A)mRNA。在分离的迷走神经支配的小鼠气管-肺制剂中使用细胞外电生理记录,注意到介体通过G蛋白偶联受体(PAR2)起作用,在没有NaV1.9的情况下,或亲离子受体(P2×3)在激发动作电位放电方面的效率降低了70-85%。然而,当刺激是快速点状机械刺激时,野生型和NaV1.9-/-之间的动作电位放电没有差异。对分离的无剂量神经元的被动和主动特性的分析显示,野生型和NaV1.9-/-小鼠的神经元之间没有差异,除了后超极化(AHP)的持续时间差异不大。引起动作电位(流变碱)或动作电位电压阈值所需的电流量也没有差异。由P2×3激动剂引起的化学介质引起的内向电流在野生型与野生型中相同。NaV1.9-/-神经元。然而,电流仅在野生型神经元中足以引起动作电位。数据支持以下推测:通过选择性抑制炎症介质相关的迷走神经C纤维激活,NaV1.9可能是炎症性气道疾病的有吸引力的治疗靶标。要点:在缺乏NaV1.9的小鼠中,炎症介质在激活迷走神经气道C纤维末端方面的作用要小得多。野生型神经元和NaV1.9-/-神经元的主动和被动特性相同。缺乏NaV1.9的神经有反应,通常,具有动作电位放电以对末端进行快速点状机械刺激或对细胞体进行向内电流注入的快速刺激。NaV1.9通道可能是选择性抑制由炎症介质引起的迷走神经伤害性C纤维激活而不阻断神经对与误吸相关的潜在危险刺激的反应的有吸引力的靶标。摘要图例:我们检验了以下假设:NaV1.9在气道迷走神经滴注C纤维的兴奋性中起重要作用。数据显示,NaV1.9在通过GPCRs或离子型受体起作用的炎症介质诱发的无剂量C纤维末端的激活中很重要,但在通过快速点状机械刺激的激活中不那么重要。NaV1.9的存在不会影响神经元的膜特性,通过快速注入去极化电流来评估它们的兴奋性。NaV1.9为治疗慢性咳嗽和过度反射性支气管痉挛以及与炎性气道疾病相关的分泌物提供了有趣的靶标。本文受版权保护。保留所有权利。
    The influence of NaV 1.9 on inflammatory mediator-induced activation of airway vagal nodose C-fibres was evaluated by comparing responses in wild-type versus NaV 1.9-/- mice. A single-cell RT-PCR analysis indicated that virtually all nodose C-fibre neurons expressed NaV 1.9 (SCN11A) mRNA. Using extracellular electrophysiological recordings in an isolated vagally innervated mouse trachea-lung preparation, it was noted that mediators acting via G protein-coupled receptors (PAR2), or ionotropic receptors (P2×3) were 70-85% less effective in evoking action potential discharge in the absence of NaV 1.9. However, there was no difference in action potential discharge between wild-type and NaV 1.9-/- when the stimulus was a rapid punctate mechanical stimulus. An analysis of the passive and active properties of isolated nodose neurons revealed no difference between neurons from wild-type and NaV 1.9-/- mice, with the exception of a modest difference in the duration of the afterhyperpolarization. There was also no difference in the amount of current required to evoke action potentials (rheobase) or the action potential voltage threshold. The inward current evoked by the chemical mediator by a P2×3 agonist was the same in wild-type versus NaV 1.9-/- neurons. However, the current was sufficient to evoke action potential only in the wild-type neurons. The data support the speculation that NaV 1.9 could be an attractive therapeutic target for inflammatory airway disease by selectively inhibiting inflammatory mediator-associated vagal C-fibre activation. KEY POINTS: Inflammatory mediators were much less effective in activating the terminals of vagal airway C-fibres in mice lacking NaV 1.9. The active and passive properties of nodose neurons were the same between wild-type neurons and NaV 1.9-/- neurons. Nerves lacking NaV 1.9 responded, normally, with action potential discharge to rapid punctate mechanical stimulation of the terminals or the rapid stimulation of the cell bodies with inward current injections. NaV 1.9 channels could be an attractive target to selectively inhibit vagal nociceptive C-fibre activation evoked by inflammatory mediators without blocking the nerves\' responses to the potentially hazardous stimuli associated with aspiration.
    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
    骨骼肌收缩会触发运动压力反射(EPR),以调节心血管系统对运动的反应。肌肉收缩时,释放的物质在介导EPR的III和IV组传入中产生动作电位活性。这些物质中的一些通过G蛋白偶联受体(GPCR)激活增加传入活性,但是机制还不完全清楚。我们有兴趣确定是否涉及抗河豚毒素(TTX-R)电压依赖性钠通道(NaV),并研究了此类化合物混合物(缓激肽,前列腺素,去甲肾上腺素,ATP,称为肌肉代谢物)。使用全细胞膜片钳电生理学,我们表明,肌肉代谢物显着增加TTX-RNaV电流。这种增强的上升时间平均为2分钟,这表明参与了可扩散的第二信使途径。肌肉代谢物对电流-电压关系的影响,通道激活和失活动力学支持NaV1.9通道作为这种增强的目标。以混合物中使用的浓度单独施用时,只有前列腺素和缓激肽显着增强NaV电流,但是当肌肉代谢物一起应用时,这些增强的总和<1/3。这表明活化的GPCR之间的协同作用以增强NaV1.9电流。当在较高浓度下使用时,所有四种物质都可以增强电流,这表明由每种代谢物激活的GPCRs可以增强通道活性。NaV1.9通道活性的增强是GPCR激活增加产生EPR的传入体中的动作电位活性的可能机制。NEW和NOTEWORTHYG蛋白偶联受体(GPCR)激活增加了肌肉传入的动作电位活性,从而产生运动压迫反射(EPR),但是机制还不完全清楚。我们提供证据表明,通过应用在肌肉收缩过程中可能释放的代谢物混合物,NaV1.9电流可协同增强。NaV1.9电流的增强可能是心血管疾病患者中GPCR激活产生EPR和EPR不适当激活的一种机制。
    Skeletal muscle contraction triggers the exercise pressor reflex (EPR) to regulate the cardiovascular system response to exercise. During muscle contraction, substances are released that generate action potential activity in group III and IV afferents that mediate the EPR. Some of these substances increase afferent activity via G-protein-coupled receptor (GPCR) activation, but the mechanisms are incompletely understood. We were interested in determining if tetrodotoxin-resistant (TTX-R) voltage-dependent sodium channels (NaV) were involved and investigated the effect of a mixture of such compounds (bradykinin, prostaglandin, norepinephrine, and ATP, called muscle metabolites). Using whole cell patch-clamp electrophysiology, we show that the muscle metabolites significantly increased TTX-R NaV currents. The rise time of this enhancement averaged ∼2 min, which suggests the involvement of a diffusible second messenger pathway. The effect of muscle metabolites on the current-voltage relationship, channel activation and inactivation kinetics support NaV1.9 channels as the target for this enhancement. When applied individually at the concentration used in the mixture, only prostaglandin and bradykinin significantly enhanced NaV current, but the sum of these enhancements was <1/3 that observed when the muscle metabolites were applied together. This suggests synergism between the activated GPCRs to enhance NaV1.9 current. When applied at a higher concentration, all four substances could enhance the current, which demonstrates that the GPCRs activated by each metabolite can enhance channel activity. The enhancement of NaV1.9 channel activity is a likely mechanism by which GPCR activation increases action potential activity in afferents generating the EPR.NEW & NOTEWORTHY G-protein-coupled receptor (GPCR) activation increases action potential activity in muscle afferents to produce the exercise pressor reflex (EPR), but the mechanisms are incompletely understood. We provide evidence that NaV1.9 current is synergistically enhanced by application of a mixture of metabolites potentially released during muscle contraction. The enhancement of NaV1.9 current is likely one mechanism by which GPCR activation generates the EPR and the inappropriate activation of the EPR in patients with cardiovascular disease.
    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
    间充质同源异型盒蛋白2(MEOX2)是参与中胚层分化的转录因子,包括骨骼的发育,肌肉,脉管系统和皮体。我们以前已经确定了MEOX2在成纤维细胞中的失调,来自先天性疼痛患者的不敏感,并确认了BTN,MEOX2的果蝇同源物在对有害热刺激的有害反应中起作用。为了确定MEOX2在哺乳动物周围神经系统中的重要性,我们使用Meox2杂合(Meox2+/-)小鼠模型来表征其在感觉神经系统中的功能,更具体地说,在伤害性感受中。MEOX2在小鼠背根神经节(DRG)和脊髓中表达,并定位在感觉神经元子集的核中。小鼠模型的功能研究,包括行为,细胞和电生理分析,显示出改变的伤害感受,包括去极化时动作电位启动受损。机械上,我们注意到分别编码Nav1.7和Nav1.9电压门控钠通道的Scn9a和Scn11a基因的表达降低,这对痛觉感受器的亚阈值扩增和动作电位启动至关重要。对Meox2+/-DRG的进一步转录组学分析揭示了特定基因子集的下调,包括先前与疼痛感知相关的基因。如PENK和NPY。基于这些观察,我们提出了MEOX2在初级传入伤害性感受器神经元中的新作用,以维持正确感知急性和炎性有害刺激所需的转录程序。
    Mesenchyme homeobox protein 2 (MEOX2) is a transcription factor involved in mesoderm differentiation, including development of bones, muscles, vasculature and dermatomes. We have previously identified dysregulation of MEOX2 in fibroblasts from Congenital Insensitivity to Pain patients, and confirmed that btn, the Drosophila homologue of MEOX2, plays a role in nocifensive responses to noxious heat stimuli. To determine the importance of MEOX2 in the mammalian peripheral nervous system, we used a Meox2 heterozygous (Meox2+/- ) mouse model to characterise its function in the sensory nervous system, and more specifically, in nociception. MEOX2 is expressed in the mouse dorsal root ganglia (DRG) and spinal cord, and localises in the nuclei of a subset of sensory neurons. Functional studies of the mouse model, including behavioural, cellular and electrophysiological analyses, showed altered nociception encompassing impaired action potential initiation upon depolarisation. Mechanistically, we noted decreased expression of Scn9a and Scn11a genes encoding Nav 1.7 and Nav 1.9 voltage-gated sodium channels respectively, that are crucial in subthreshold amplification and action potential initiation in nociceptors. Further transcriptomic analyses of Meox2+/- DRG revealed downregulation of a specific subset of genes including those previously associated with pain perception, such as PENK and NPY. Based on these observations, we propose a novel role of MEOX2 in primary afferent nociceptor neurons for the maintenance of a transcriptional programme required for proper perception of acute and inflammatory noxious stimuli.
    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 inhibition of voltage-gated sodium (NaV) channels in somatosensory neurons presents a promising novel modality for the treatment of pain. However, the precise contribution of these channels to neuronal excitability, the cellular correlate of pain, is unknown; previous studies using genetic knockout models or pharmacologic block of NaV channels have identified general roles for distinct sodium channel isoforms, but have never quantified their exact contributions to these processes. To address this deficit, we have utilized dynamic clamp electrophysiology to precisely tune in varying levels of NaV1.8 and NaV1.9 currents into induced pluripotent stem cell-derived sensory neurons (iPSC-SNs), allowing us to quantify how graded changes in these currents affect different parameters of neuronal excitability and electrogenesis. We quantify and report direct relationships between NaV1.8 current density and action potential half-width, overshoot, and repetitive firing. We additionally quantify the effect varying NaV1.9 current densities have on neuronal membrane potential and rheobase. Furthermore, we examined the simultaneous interplay between NaV1.8 and NaV1.9 on neuronal excitability. Finally, we show that minor biophysical changes in the gating of NaV1.8 can render human iPSC-SNs hyperexcitable, in a first-of-its-kind investigation of a gain-of-function NaV1.8 mutation in a human neuronal background.
    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
    Pancreatic cancer (PC) is a malignant tumor with poor prognosis. The poor effect of surgery and chemotherapy makes the research of immunotherapy target molecules significant. Therefore, identifying the new molecular targets of PC is important for patients. In our study, we systematically analyzed molecular correlates of pancreatic cancer by bioinformatic analysis. We characterized differentially expressed analysis based on the TCGA pancreatic cancer dataset. Then, univariate Cox regression was employed to screen out overall survival- (OS-) related DEGs. Based on these genes, we established a risk signature by the multivariate Cox regression model. The ICGC cohort and GSE62452 cohort were used to validate the reliability of the risk signature. The impact of T lymphocyte-related genes from risk signature was confirmed in PC. Here, we observed the correlation between the T lymphocyte-related genes and the expression level of targeted therapy. We established a five-mRNA (LY6D, ANLN, ZNF488, MYEOV, and SCN11A) prognostic risk signature. Next, we identified ANLN and MYEOV that were associated with T lymphocyte infiltrations (P < 0.05). High ANLN and MYEOV expression levels had a poorer prognosis in decreased T lymphocyte subgroup in PC. Correlation analysis between ANLN and MYEOV and immunomodulators showed that ANLN and MYEOV may have potential value in pancreatic cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号