NaV1.6

Nav1.6
  • 文章类型: Journal Article
    目的:电压门控钠通道(NaVs)抑制剂是重要的抗癫痫药物,但特定通道亚型的贡献是未知的,因为可用的抑制剂是非选择性的。我们的目标是创作小说,Nav通道的同工型选择性抑制剂作为一种手段,告知改进的抗癫痫药物的开发。
    方法:我们创建了一系列具有不同选择性的化合物,可以单独或与NaV1.2一起阻断NaV1.6。评估了这些新型NaV抑制剂在Scn8a(编码NaV1.6)和野生型小鼠中具有杂合功能获得突变(N1768D/)的小鼠中抑制电诱发癫痫发作的能力。
    结果:在Scn8aN1768D/+小鼠中对NaV1.6的药物抑制可防止由6-Hz电击引起的癫痫发作。抑制剂在野生型小鼠的直流电最大电击癫痫发作测定中也是有效的。NaV1.6抑制作用与两种模型的疗效相关,即使没有抑制其他CNSNaV亚型。
    结论:我们的数据表明,抑制NaV1.6是NaV抑制剂抗癫痫药物疗效的驱动因素。保留抑制性中间神经元的NaV1.1通道不会损害功效。选择性NaV1.6抑制剂可以为人类Scn8a发育性和癫痫性脑病提供靶向治疗,并改善特发性癫痫的治疗。
    OBJECTIVE: Inhibitors of voltage-gated sodium channels (NaVs) are important anti-epileptic drugs, but the contribution of specific channel isoforms is unknown since available inhibitors are non-selective. We aimed to create novel, isoform selective inhibitors of Nav channels as a means of informing the development of improved antiseizure drugs.
    METHODS: We created a series of compounds with diverse selectivity profiles enabling block of NaV1.6 alone or together with NaV1.2. These novel NaV inhibitors were evaluated for their ability to inhibit electrically evoked seizures in mice with a heterozygous gain-of-function mutation (N1768D/+) in Scn8a (encoding NaV1.6) and in wild-type mice.
    RESULTS: Pharmacologic inhibition of NaV1.6 in Scn8aN1768D/+ mice prevented seizures evoked by a 6-Hz shock. Inhibitors were also effective in a direct current maximal electroshock seizure assay in wild-type mice. NaV1.6 inhibition correlated with efficacy in both models, even without inhibition of other CNS NaV isoforms.
    CONCLUSIONS: Our data suggest NaV1.6 inhibition is a driver of efficacy for NaV inhibitor anti-seizure medicines. Sparing the NaV1.1 channels of inhibitory interneurons did not compromise efficacy. Selective NaV1.6 inhibitors may provide targeted therapies for human Scn8a developmental and epileptic encephalopathies and improved treatments for idiopathic epilepsies.
    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
    奎尼丁已被用作抗惊厥药,通过靶向功能获得KCNT1致病性突变变体来治疗KCNT1相关癫痫患者。然而,奎尼丁阻断KCNT1(Slack)的详细机制仍然难以捉摸。这里,我们报道了电压门控钠通道NaV1.6和Slack的功能和物理耦合。NaV1.6结合并高度敏感Slack对奎尼丁阻断。NaV1.6的纯合敲除降低了天然钠激活的钾电流对奎尼丁阻断的敏感性。NaV1.6介导的致敏需要NaV1.6的N-和C-末端与Slack的C-末端结合的参与,并且通过NaV1.6的瞬时钠流入增强。此外,通过Slack的C端病毒表达破坏Slack-NaV1.6相互作用可以防止SlackG269S诱导的小鼠癫痫发作。这些关于Slack-NaV1.6复合物的见解挑战了传统的观点,即“Slack作为抗癫痫药物发现工作的孤立目标”,并可以指导KCNT1相关癫痫的创新治疗策略的开发。
    Quinidine has been used as an anticonvulsant to treat patients with KCNT1-related epilepsy by targeting gain-of-function KCNT1 pathogenic mutant variants. However, the detailed mechanism underlying quinidine\'s blockade against KCNT1 (Slack) remains elusive. Here, we report a functional and physical coupling of the voltage-gated sodium channel NaV1.6 and Slack. NaV1.6 binds to and highly sensitizes Slack to quinidine blockade. Homozygous knockout of NaV1.6 reduces the sensitivity of native sodium-activated potassium currents to quinidine blockade. NaV1.6-mediated sensitization requires the involvement of NaV1.6\'s N- and C-termini binding to Slack\'s C-terminus and is enhanced by transient sodium influx through NaV1.6. Moreover, disrupting the Slack-NaV1.6 interaction by viral expression of Slack\'s C-terminus can protect against SlackG269S-induced seizures in mice. These insights about a Slack-NaV1.6 complex challenge the traditional view of \'Slack as an isolated target\' for anti-epileptic drug discovery efforts and can guide the development of innovative therapeutic strategies for KCNT1-related epilepsy.
    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
    目的:尽管程序性细胞死亡蛋白1(PD-1)通常作为免疫疗法的靶标,最近的一些研究发现PD-1在神经系统中表达,神经元PD-1可能在调节神经元兴奋性中起关键作用。然而,脑局部PD-1是否参与癫痫发作和癫痫发生尚不清楚,值得深入探索。
    方法:通过免疫组织化学证实人神经元中PD-1的存在,通过实时定量PCR(RT-qPCR)和蛋白质印迹法测量PD-1的表达水平。化疗惊厥药,戊四氮(PTZ)和环噻嗪(CTZ),用于建立体内(啮齿动物)和体外(原代海马神经元)癫痫发作模型,分别。SHR-1210(一种PD-1单克隆抗体)和葡萄糖酸钠(SSG,一种经过验证的含SH2的蛋白酪氨酸磷酸酶-1[SHP-1]抑制剂),用于研究PD-1通路阻断对啮齿动物癫痫行为和神经元癫痫样放电的影响。应用miRNA策略来确定PD-1敲低对神经元兴奋性的影响。通过全细胞膜片钳记录确定神经元的电活动和钠通道功能。通过进行共免疫染色和共免疫沉淀(co-IP)实验,验证了PD-1和人电压门控钠通道(Nav1.6)的α-6亚基之间的相互作用。
    结果:我们的结果表明,与难治性癫痫患者手术切除标本的病灶周围组织相比,PD-1蛋白和mRNA水平在病灶核心中上调。此外,我们表明抗PD-1治疗在体内和体外均具有抗癫痫发作作用。然后,我们发现PD-1阻断可以改变钠通道的电生理特性。此外,我们发现PD-1与下游SHP-1共同作用调节钠通道功能,从而调节神经元兴奋性。进一步的研究表明,神经元PD-1和Nav1.6之间存在直接相互作用。
    结论:我们的研究表明,神经元PD-1在癫痫中起重要作用,抗PD-1治疗通过抑制钠通道功能来预防癫痫发作,确定抗PD-1治疗作为癫痫的新治疗策略。
    Although programmed cell death protein 1 (PD-1) typically serves as a target for immunotherapies, a few recent studies have found that PD-1 is expressed in the nervous system and that neuronal PD-1 might play a crucial role in regulating neuronal excitability. However, whether brain-localized PD-1 is involved in seizures and epileptogenesis is still unknown and worthy of in-depth exploration.
    The existence of PD-1 in human neurons was confirmed by immunohistochemistry, and PD-1 expression levels were measured by real-time quantitative PCR (RT-qPCR) and western blotting. Chemoconvulsants, pentylenetetrazol (PTZ) and cyclothiazide (CTZ), were applied for the establishment of in vivo (rodents) and in vitro (primary hippocampal neurons) models of seizure, respectively. SHR-1210 (a PD-1 monoclonal antibody) and sodium stibogluconate (SSG, a validated inhibitor of SH2-containing protein tyrosine phosphatase-1 [SHP-1]) were administrated to investigate the impact of PD-1 pathway blockade on epileptic behaviors of rodents and epileptiform discharges of neurons. A miRNA strategy was applied to determine the impact of PD-1 knockdown on neuronal excitability. The electrical activities and sodium channel function of neurons were determined by whole-cell patch-clamp recordings. The interaction between PD-1 and α-6 subunit of human voltage-gated sodium channel (Nav1.6) was validated by performing co-immunostaining and co-immunoprecipitation (co-IP) experiments.
    Our results reveal that PD-1 protein and mRNA levels were upregulated in lesion cores compared with perifocal tissues of surgically resected specimens from patients with intractable epilepsy. Furthermore, we show that anti-PD-1 treatment has anti-seizure effects both in vivo and in vitro. Then, we reveal that PD-1 blockade can alter the electrophysiological properties of sodium channels. Moreover, we reveal that PD-1 acts together with downstream SHP-1 to regulate sodium channel function and hence neuronal excitability. Further investigation suggests that there is a direct interaction between neuronal PD-1 and Nav1.6.
    Our study reveals that neuronal PD-1 plays an important role in epilepsy and that anti-PD-1 treatment protects against seizures by suppressing sodium channel function, identifying anti-PD-1 treatment as a novel therapeutic strategy for epilepsy.
    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
    脊柱运动网络中的持续钠电流(INaP)促进了两种不同的非线性放电模式:由双稳态运动神经元的短暂激发触发的自持尖峰和中央模式发生器(CPG)的中间神经元的爆发振荡。这里,我们确定了负责INaP的NaV通道及其在运动行为中的作用。我们报告轴突Nav1.6是腰运动神经元INaP的主要分子参与者。在运动神经元中抑制Nav1.6而不是Nav1.1会损害INaP,双稳态,姿势语气,和运动性能。在节律性CPG区域的中间神经元中,Nav1.6和Nav1.1同样介导INaP。需要抑制两个通道以消除振荡爆发活动和运动节律。总的来说,Nav1.6通过控制运动神经元中INaP依赖性双稳态并与Nav1.1协同工作以提供CPG的INaP依赖性节律特性,在姿势和运动中都起着重要作用。
    Persistent sodium current (INaP) in the spinal locomotor network promotes two distinct nonlinear firing patterns: a self-sustained spiking triggered by a brief excitation in bistable motoneurons and bursting oscillations in interneurons of the central pattern generator (CPG). Here, we identify the NaV channels responsible for INaP and their role in motor behaviors. We report the axonal Nav1.6 as the main molecular player for INaP in lumbar motoneurons. The inhibition of Nav1.6, but not of Nav1.1, in motoneurons impairs INaP, bistability, postural tone, and locomotor performance. In interneurons of the rhythmogenic CPG region, both Nav1.6 and Nav1.1 equally mediate INaP. Inhibition of both channels is required to abolish oscillatory bursting activities and the locomotor rhythm. Overall, Nav1.6 plays a significant role both in posture and locomotion by governing INaP-dependent bistability in motoneurons and working in tandem with Nav1.1 to provide INaP-dependent rhythmogenic properties of the CPG.
    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
    帕金森病(Parkinson’sdisease,PD)是老年人常见的神经退行性疾病,其特征是PD患者的运动障碍。Nav1.6是成年哺乳动物和啮齿动物脑中电压门控钠通道(VGSC)最丰富的亚型。在这里,我们研究了Nav1.6在单侧6-OHDA(6-羟基多巴胺)损伤大鼠运动缺陷发病机理中涉及的外部苍白球(GP)中的作用。结果表明,在中期阶段,同侧GP的反应性星形胶质细胞中Nav1.6急剧增加,但在6-OHDA损伤大鼠病理过程的后期与对照大鼠没有区别。此外,同侧GP中Nav1.6表达的下调可以显着改善6-OHDA损伤大鼠在病理过程中期的运动缺陷。电生理实验表明,同侧GP中Nav1.6表达的下调显着降低了6-OHDA损伤大鼠同侧M1(初级运动皮层)和GP之间的异常高同步性。Ca2成像显示,Nav1.6表达的下调降低了原代培养的星形胶质细胞中Ca2([Ca2]i)的细胞内浓度。这些发现表明,GP中反应性星形胶质细胞Nav1.6表达的增加在6-OHDA损伤大鼠中期运动功能障碍的发病机理中起重要作用。可能通过调节星形胶质细胞的[Ca2]i参与星形胶质细胞-神经元的通讯,从而有助于6-OHDA病变大鼠基底神经节(BG)异常电信号的形成。
    Parkinson\'s disease (PD) is a common neurodegenerative disease in elderly people, which is characterized by motor disabilities in PD patients. Nav1.6 is the most abundant subtype of voltage-gated sodium channels (VGSCs) in the brain of adult mammals and rodents. Here we investigated the role of Nav1.6 in the external globus pallidus (GP) involved in the pathogenesis of motor deficits in unilateral 6-OHDA(6-hydroxydopamine)lesioned rats. The results show that Nav1.6 is dramatically increased in reactive astrocytes of the ipsilateral GP in the middle stage, but not different from the control rats in the later stage of the pathological process in 6-OHDA lesioned rats. Furthermore, the down-regulation of Nav1.6 expression in the ipsilateral GP can significantly improve motor deficits in 6-OHDA lesioned rats in the middle stage of the pathological process. The electrophysiological experiments show that the down-regulation of Nav1.6 expression in the ipsilateral GP significantly decreases the abnormal high synchronization between the ipsilateral M1 (the primary motor cortex) and GP in 6-OHDA lesioned rats. Ca2+ imaging reveals that the down-regulation of Nav1.6 expression reduces the intracellular concentration of Ca2+ ([Ca2+ ]i) in primary cultured astrocytes. These findings suggest that the increased Nav1.6 expression of reactive astrocytes in the GP play an important role in the pathogenesis of motor dysfunction in the middle stage in 6-OHDA lesioned rats, which may participate in astrocyte-neuron communication by regulating [Ca2+ ]i of astrocytes, thereby contributing to the formation of abnormal electrical signals of the basal ganglia (BG) in 6-OHDA lesioned rats.
    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
    胶质瘤仍然是一个临床挑战,常见的和致命的。胶质母细胞瘤的治疗仍然难以捉摸,研究人员专注于发现新的机制和药物。已经确定,电压门控钠通道(VGSC)的表达在许多恶性肿瘤中异常增加,总的来说,在相应的正常组织中很少表达。这表明离子通道活性似乎与肿瘤的恶性进展有关。VGSC对于它们的活性如何导致癌细胞活性或侵袭性的增加仍然是未知的。某些钠离子通道亚型(例如,Nav1.5和Nav1.7)与包括乳腺癌和结直肠癌在内的癌症的转移和侵袭有关。作者先前的一项研究探索了某些离子通道在神经胶质瘤中的表达,但是很少有与Nav1.6相关的研究。本研究旨在阐明Nav1.6在脑胶质瘤中的表达和作用,并通过虚拟筛选和药物敏感性分析筛选潜在的脑胶质瘤治疗药物。通过逆转录定量PCR和蛋白质印迹分析确定Nav1.6mRNA和蛋白质的相对表达。细胞增殖通过细胞计数试剂盒-8测定法测定。通过细胞伤口愈合测定评估细胞迁移。通过Transwell细胞侵袭实验和流式细胞术检测细胞侵袭和凋亡。最后但并非最不重要的,FDA批准的药物使用虚拟筛选进行筛选,基于Nav1.6的表达和结构的分子对接和NCI‑60药物敏感性分析。在神经胶质瘤细胞中,Nav1.6表达明显上调,多见于细胞质和细胞膜,其表达与病理分级呈正相关。A172和U251细胞增殖减少,当Nav1.6表达被击倒时迁移和入侵,细胞凋亡增加。发现作用于神经胶质瘤细胞的TNF-α(100pg/ml)上调Nav1.6的表达水平,TNF-α参与Nav1.6促进神经胶质瘤恶性进展的过程。最后,某些FDA批准的药物是通过虚拟筛选和药物敏感性分析确定的。总之,本研究证明了Nav1.6在神经胶质瘤中的表达和作用,并确定了几种FDA批准的与Nav1.6高度相关的药物,这些药物可能是神经胶质瘤患者的候选药物.
    Gliomas remain a clinical challenge, common and fatal. Treatment of glioblastoma remains elusive, and researchers have focused on discovering new mechanisms and drugs. It has been well established that the expression of voltage‑gated sodium channels (VGSCs) is abnormally increased in numerous malignancies and, in general, is rarely expressed in the corresponding normal tissues. This suggests that ion channel activity appears to be associated with malignant progression of tumors. VGSCs remain largely unknown as to how their activity leads to an increase in cancer cell activity or invasiveness. Certain sodium ion channel subtypes (for instance, Nav1.5 and Nav1.7) are associated with metastasis and invasion in cancers including breast and colorectal cancers. A previous study by the authors explored the expression of certain ion channels in glioma, but there are few studies related to Nav1.6. The current study aimed to elucidate the expression and role of Nav1.6 in glioma and to screen potential drugs for the treatment of glioma by virtual screening and drug sensitivity analysis. Nav1.6 relative expression of mRNA and protein was determined by reverse transcription‑quantitative PCR and western blot analysis. Cell proliferation was determined by Cell Counting Kit‑8 assay. Cell migration was assessed by cellular wound healing assay. Cell invasion and apoptosis were detected by Transwell cell invasion assay and flow cytometry. Last but not least, FDA‑approved drugs were screened using virtual screening, molecular docking and NCI‑60 drug sensitivity analyses based on the expression and structure of Nav1.6. In glioma cells, Nav1.6 was significantly upregulated and expressed mostly in the cytoplasm and cell membrane; its expression was positively correlated with pathological grade. A172 and U251 cells exhibited reduced proliferation, migration and invasion when Nav1.6 expression was knocked down, and apoptosis was increased. TNF‑α (100 pg/ml) acting on glioma cells was found to upregulate the expression level of Nav1.6, and TNF‑α was involved in the process of Nav1.6 promoting malignant progression of glioma. Finally, certain FDA‑approved drugs were identified by virtual screening and drug sensitivity analysis. In conclusion, the present study demonstrated the expression and role of Nav1.6 in glioma and identified several FDA‑approved drugs that are highly correlated with Nav1.6 and could be candidate drugs for patients with glioma.
    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Nav1.6在中枢神经系统(CNS)的神经元放电中起着至关重要的作用。Nav1.6的异常功能可能导致癫痫和其他神经系统疾病。因此,Nav1.6的特异性抑制剂具有治疗潜力。在这里,我们展示了在存在辅助亚基β1和成纤维细胞生长因子同源因子2B(FHF2B)的情况下人类Nav1.6的冷冻EM结构,总分辨率为3.1µ。总体结构表示具有闭合孔域(PD)和所有“向上”电压感应域的失活状态。涉及Trp302和Asn326以及β1亚基的保守的碳水化合物-芳香族相互作用,稳定重复I中的胞外环。除了在EM图中解析的常规脂质之外,属于未知分子的前所未有的Y形密度与PD结合,揭示了开发Nav1.6特异性阻断剂的潜在位点。疾病相关Nav1.6突变的结构图谱提供了对其致病机制的见解。
    Voltage-gated sodium channel Nav1.6 plays a crucial role in neuronal firing in the central nervous system (CNS). Aberrant function of Nav1.6 may lead to epilepsy and other neurological disorders. Specific inhibitors of Nav1.6 thus have therapeutic potentials. Here we present the cryo-EM structure of human Nav1.6 in the presence of auxiliary subunits β1 and fibroblast growth factor homologous factor 2B (FHF2B) at an overall resolution of 3.1 Å. The overall structure represents an inactivated state with closed pore domain (PD) and all \"up\" voltage-sensing domains. A conserved carbohydrate-aromatic interaction involving Trp302 and Asn326, together with the β1 subunit, stabilizes the extracellular loop in repeat I. Apart from regular lipids that are resolved in the EM map, an unprecedented Y-shaped density that belongs to an unidentified molecule binds to the PD, revealing a potential site for developing Nav1.6-specific blockers. Structural mapping of disease-related Nav1.6 mutations provides insights into their pathogenic mechanism.
    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
    癌症引起的骨痛(CIBP)经常发生在晚期癌症患者中。电压门控钠通道(VGSCs)与慢性疼痛有关,但对VGSCs在CIBP中的作用知之甚少。这里,我们旨在研究VGSCs在背根神经节(DRGs)在CIBP中的具体作用。通过皮下接种MRMT-1乳腺癌细胞产生ACIBP大鼠模型。进行转录组测序以评估基因表达谱。通过Westernblotting和qPCR检测与激活途径相关的关键基因和分化基因的表达水平。我们在鞘内植入了导管,用于慢病毒和药物的给药。然后,测量机械退缩阈值(MWT)的变化。我们在CIBP模型大鼠的DRG中鉴定了149个差异表达的mRNA(DEmRNAs)。Nav1.6的表达,在这些DMRNAs中,显着上调。DemRNA的京都基因和基因组百科全书(KEGG)途径分析表明,它们主要富集在丝裂原活化蛋白激酶(MAPK)途径中。Nav1.6敲低减弱了骨癌诱导的MWT降低。蛋白质印迹分析显示p38抑制剂降低Nav1.6的表达并减轻疼痛行为。我们的研究表明,p38MAPK在大鼠DRGs中Nav1.6表达上调有助于CIBP。
    Cancer-induced bone pain (CIBP) occurs frequently among advanced cancer patients. Voltage-gated sodium channels (VGSCs) have been associated with chronic pain, but how VGSCs function in CIBP is poorly understood. Here, we aimed to investigate the specific role of VGSCs in the dorsal root ganglia (DRGs) in CIBP. A CIBP rat model was generated by the intratibial inoculation of MRMT-1 breast carcinoma cells. Transcriptome sequencing was conducted to assess the gene expression profiles. The expression levels of key genes and differentiated genes related to activated pathways were measured by Western blotting and qPCR. We implanted a catheter intrathecally for the administration of lentivirus and drugs. Then, the changes in the mechanical withdrawal threshold (MWT) were measured. We identified 149 differentially expressed mRNAs (DEmRNAs) in the DRGs of CIBP model rats. The expression of Nav1.6, which was among these DEmRNAs, was significantly upregulated. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the DEmRNAs showed that they were mainly enriched in the mitogen-activated protein kinase (MAPK) pathway. The decrease in MWT induced by bone cancer was attenuated by Nav1.6 knockdown. Western blot analysis revealed that a p38 inhibitor decreased the expression of Nav1.6 and attenuated pain behavior. Our study shows that the upregulation of Nav1.6 expression by p38 MAPK in the DRGs of rats contributes to CIBP.
    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
    目标:SCN8A的变体,NaV1.6通道的编码基因,有多种症状,包括顽固性癫痫发作,精神运动延迟,进行性认知能力下降,自闭症特征,共济失调或肌张力障碍。标准抗惊厥治疗对病程的影响有限。
    方法:我们研究了艾司利卡西平(S-licarbazepine;S-lic)的治疗潜力,电压门控钠通道缓慢失活的增强剂,在神经母细胞瘤细胞和鼠原代海马神经元培养物中,有两个具有生物物理和神经元功能获得的变体(G1475R和M1760I)和一个具有生物物理功能获得但神经元功能丧失的变体(A1622D)。这三种变异涵盖了NaV1.6相关疾病的广谱,并与轻度至中度癫痫(G1475R)的代表性表型有关。发育性和癫痫性脑病(M1760I)和无癫痫的智力障碍(A1622D)。
    结果:与已知对NaV1.6野生型通道的影响类似,S-lic主要增强所有测试变体的缓慢失活,无论其特定的生物物理机制。除此之外,S-lic表现出变体特异性效应,包括部分逆转病理性减慢的快速失活动力学(A1622D和M1760I),以及减少A1622D变异通道增强的持续Na电流的趋势。此外,我们在原代转染神经元中的数据表明,S-lic不仅可以调节与变异相关的兴奋过度(M1760I和G1475R),而且还可以调节兴奋过度(A1622D)。
    结论:S-lic不仅具有物质特异性作用,而且具有变体特异性作用。优化以实现这种变体特异性药理学调节的个性化治疗方案可有助于减少不良副作用并改善SCN8A相关疾病的总体治疗结果。
    Variants in SCN8A, the NaV 1.6 channel\'s coding gene, are characterized by a variety of symptoms, including intractable epileptic seizures, psychomotor delay, progressive cognitive decline, autistic features, ataxia or dystonia. Standard anticonvulsant treatment has a limited impact on the course of disease.
    We investigated the therapeutic potential of eslicarbazepine (S-licarbazepine; S-lic), an enhancer of slow inactivation of voltage gated sodium channels, on two variants with biophysical and neuronal gain-of-function (G1475R and M1760I) and one variant with biophysical gain-of-function but neuronal loss-of-function (A1622D) in neuroblastoma cells and in murine primary hippocampal neuron cultures. These three variants cover the broad spectrum of NaV 1.6-associated disease and are linked to representative phenotypes of mild to moderate epilepsy (G1475R), developmental and epileptic encephalopathy (M1760I) and intellectual disability without epilepsy (A1622D).
    Similar to known effects on NaV 1.6 wildtype channels, S-lic predominantly enhances slow inactivation on all tested variants, irrespective of their particular biophysical mechanisms. Beyond that, S-lic exhibits variant-specific effects including a partial reversal of pathologically slowed fast inactivation dynamics (A1622D and M1760I) and a trend to reduce enhanced persistent Na+ current by A1622D variant channels. Furthermore, our data in primary transfected neurons reveal that not only variant-associated hyperexcitability (M1760I and G1475R) but also hypoexcitability (A1622D) can be modulated by S-lic.
    S-lic has not only substance-specific effects but also variant-specific effects. Personalized treatment regimens optimized to achieve such variant-specific pharmacological modulation may help to reduce adverse side effects and improve the overall therapeutic outcome of SCN8A-related disease.
    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
    Nav1.6活动异常可诱发与癫痫相关的兴奋过度。编码Nav1.6的SCN8A基因中的功能增益突变与癫痫的发展有关;然而,介导这些变化的分子机制具有显著异质性,可能涉及Nav1.6的翻译后调控。因为钙/钙调蛋白依赖性蛋白激酶II(CaMKII)是Nav1.6通道的强大调节剂,我们调查了CaMKII是否调节与疾病相关的Nav1.6突变体。ND7/23细胞中的全细胞电压钳记录表明,CaMKII对癫痫相关突变R850Q的抑制在很大程度上概括了先前对WTNav1.6观察到的影响。我们还描述了一个罕见的错义变体,R639C,位于Nav1.6的CaMKII调制的监管热点内。预测软件算法和电生理记录揭示了R639C突变体通道活性的功能增益效应,包括与WTNav1.6相比增加的钠电流和超极化激活。重要的是,R639C突变在关键调控位点消除CaMKII磷酸化,T642,和,与WT和R850Q通道相比,显示对CaMKII抑制的不同反应。计算模拟表明,带有R639C或R850Q突变的建模神经元是可过度激活的,并模拟CaMKII抑制对建模神经元中Nav1.6活性的影响,从而降低了过度兴奋性。急性CaMKII抑制可能是减弱Nav1.6突变产生的功能获得效应的有希望的机制。
    Aberrant Nav1.6 activity can induce hyperexcitability associated with epilepsy. Gain-of-function mutations in the SCN8A gene encoding Nav1.6 are linked to epilepsy development; however, the molecular mechanisms mediating these changes are remarkably heterogeneous and may involve post-translational regulation of Nav1.6. Because calcium/calmodulin-dependent protein kinase II (CaMKII) is a powerful modulator of Nav1.6 channels, we investigated whether CaMKII modulates disease-linked Nav1.6 mutants. Whole-cell voltage clamp recordings in ND7/23 cells show that CaMKII inhibition of the epilepsy-related mutation R850Q largely recapitulates the effects previously observed for WT Nav1.6. We also characterized a rare missense variant, R639C, located within a regulatory hotspot for CaMKII modulation of Nav1.6. Prediction software algorithms and electrophysiological recordings revealed gain-of-function effects for R639C mutant channel activity, including increased sodium currents and hyperpolarized activation compared to WT Nav1.6. Importantly, the R639C mutation ablates CaMKII phosphorylation at a key regulatory site, T642, and, in contrast to WT and R850Q channels, displays a distinct response to CaMKII inhibition. Computational simulations demonstrate that modeled neurons harboring the R639C or R850Q mutations are hyperexcitable, and simulating the effects of CaMKII inhibition on Nav1.6 activity in modeled neurons differentially reduced hyperexcitability. Acute CaMKII inhibition may represent a promising mechanism to attenuate gain-of-function effects produced by Nav1.6 mutations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号