NaV1.3

  • 文章类型: Journal Article
    心肌细胞中活性氧(ROS)的积累可以通过去除电压门控Na通道的失活来诱导致心律失常的晚期Na电流,其中包括抗河豚毒素(TTX)的心脏α亚基Nav1.5以及TTX敏感的α亚基,例如Nav1.2和Nav1.3。这里,我们探索了小鼠心肌细胞和人诱导的多能干细胞衍生的心肌细胞(hiPSC-CM)以及表达Nav1.2,Nav1.3或Nav1.5的HEK293细胞中氧化剂诱导的晚期Na电流。用氧化剂氯胺T(ChT)处理的小鼠心肌细胞和hiPSC-CM中的Na电流发生了峰值电流幅度的适度降低,并伴有大的晚期Na电流。虽然ChT引起峰值电流幅度的强烈降低,但在Nav1.5上只有很小的持续电流,但Nav1.2和Nav1.3在氧化后都产生了增加的峰值电流幅度和大的持续电流。与小鼠心肌细胞和hiPSC-CM的峰值Na+电流相比,TTX(300nM)阻断了ChT诱导的晚期Na+电流明显更强。当UVA光(380nm)或半胱氨酸选择性氧化剂硝酰基(HNO)诱导氧化时,Nav1.2,Nav1.3和Nav1.5之间在ROS敏感性方面的相似差异也很明显。最后,我们在心肌细胞中表达的TTX敏感性Na+通道的数据可能与氧化应激后晚期Na+电流的产生有关.
    An accumulation of reactive oxygen species (ROS) in cardiomyocytes can induce pro-arrhythmogenic late Na+ currents by removing the inactivation of voltage-gated Na+ channels including the tetrodotoxin (TTX)-resistant cardiac α-subunit Nav1.5 as well as TTX-sensitive α-subunits like Nav1.2 and Nav1.3. Here, we explored oxidant-induced late Na+ currents in mouse cardiomyocytes and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as well as in HEK 293 cells expressing Nav1.2, Nav1.3, or Nav1.5. Na+ currents in mouse cardiomyocytes and hiPSC-CMs treated with the oxidant chloramine T (ChT) developed a moderate reduction in peak current amplitudes accompanied by large late Na+ currents. While ChT induced a strong reduction in peak current amplitudes but only small persistent currents on Nav1.5, both Nav1.2 and Nav1.3 produced increased peak current amplitudes and large persistent currents following oxidation. TTX (300 nM) blocked ChT-induced late Na+ currents significantly stronger as compared to peak Na+ currents in both mouse cardiomyocytes and hiPSC-CMs. Similar differences between Nav1.2, Nav1.3, and Nav1.5 regarding ROS sensitivity were also evident when oxidation was induced with UVA-light (380 nm) or the cysteine-selective oxidant nitroxyl (HNO). To conclude, our data on TTX-sensitive Na+ channels expressed in cardiomyocytes may be relevant for the generation of late Na+ currents following oxidative stress.
    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:神经性疼痛是一种常见的慢性疼痛障碍,这主要归因于脊髓中枢致敏。脊髓背角(SDH)中的钙/钙调蛋白依赖性蛋白激酶IIα(CaMKIIα)激活是脊髓致敏的主要原因。然而,SDH中CaMKIIα阳性(CaMKIIα)神经元诱导神经性疼痛的确切方式尚不清楚。本研究旨在探讨脊髓CaMKIIα+神经元在慢性压迫性损伤(CCI)引起的神经病理性疼痛中的作用,并探讨CaMKIIα+神经元激活的潜在表观遗传机制。
    方法:CCI诱导的神经病理性疼痛小鼠模型,Sirt1loxP/loxP小鼠,和化学遗传病毒用于研究脊髓CaMKIIα神经元的激活是否参与神经性疼痛及其参与机制。转录组序列,西方印迹,qRT-PCR,和免疫荧光分析检测相关分子的表达和神经元的激活。免疫共沉淀用于观察蛋白质的结合关系。染色质免疫沉淀(ChIP)-PCR用于分析Scn3a启动子区域组蛋白H3的乙酰化。
    结果:CCI小鼠脊髓CaMKIIα+神经元钠通道Nav1.3表达增加,SIRT1表达降低。CCI后CaMKIIα神经元变得过度活跃,抑制其激活缓解了CCI引起的疼痛。SIRT1的过表达逆转了Nav1.3的增加并减轻了疼痛,而SIRT1的敲除或Nav1.3的过表达促进了CaMKIIα神经元的激活并诱导了疼痛。通过敲除脊髓SIRT1,Scn3a(编码Nav1.3)启动子区组蛋白H3的乙酰化增加,导致Nav1.3的表达增加。
    结论:研究结果表明,神经损伤后脊髓SIRT1的异常减少会在表观遗传学上增加Nav1.3,随后激活CaMKIIα神经元并引起神经性疼痛。
    OBJECTIVE: Neuropathic pain is a common chronic pain disorder, which is largely attributed to spinal central sensitization. Calcium/calmodulin-dependent protein kinase II alpha (CaMKIIα) activation in the spinal dorsal horn (SDH) is a major contributor to spinal sensitization. However, the exact way that CaMKIIα-positive (CaMKIIα+) neurons in the SDH induce neuropathic pain is still unclear. This study aimed to explore the role of spinal CaMKIIα+ neurons in neuropathic pain caused by chronic constriction injury (CCI) and investigate the potential epigenetic mechanisms involved in CaMKIIα+ neuron activation.
    METHODS: CCI-induced neuropathic pain mice model, Sirt1loxP/loxP mice, and chemogenetic virus were used to investigate whether the activation of spinal CaMKIIα+ neurons is involved in neuropathic pain and its involved mechanism. Transcriptome sequence, western blotting, qRT-PCR, and immunofluorescence analysis were performed to assay the expression of related molecules and activation of neurons. Co-immunoprecipitation was used to observe the binding relationship of protein. Chromatin immunoprecipitation (ChIP)-PCR was applied to analyze the acetylation of histone H3 in the Scn3a promoter region.
    RESULTS: The expression of sodium channel Nav1.3 was increased and the expression of SIRT1 was decreased in the spinal CaMKIIα+ neurons of CCI mice. CaMKIIα neurons became overactive after CCI, and inhibiting their activation relieved CCI-induced pain. Overexpression of SIRT1 reversed the increase of Nav1.3 and alleviated pain, while knockdown of SIRT1 or overexpression of Nav1.3 promoted CaMKIIα+ neuron activation and induced pain. By knocking down spinal SIRT1, the acetylation of histone H3 in the Scn3a (encoding Nav1.3) promoter region was increased, leading to an increased expression of Nav1.3.
    CONCLUSIONS: The findings suggest that an aberrant reduction of spinal SIRT1 after nerve injury epigenetically increases Nav1.3, subsequently activating CaMKIIα+ neurons and causing neuropathic pain.
    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:三叉神经痛(TN)是临床实践中常见且难以治疗的神经性疼痛障碍。先前的研究表明,Toll样受体4(TLR4)调节NF-κB通路的激活以影响大鼠的神经性疼痛。已知电压门控钠通道(VGSC)在神经性疼痛电活动中起重要作用。
    目的:探讨眶下神经慢性压迫性损伤(ION-CCI)后TLR4是否通过TRAF6/NF-κBp65通路调节Nav1.3。
    方法:对SD(SpragueDawley)大鼠进行ION-CCI建模。为了验证建模的成功,我们需要检测机械痛阈值和ATF3。然后,检测大鼠TG中TLR4、TRAF6、NF-κBp65、p-p65和Nav1.3的表达。随后,鞘内注射LPS-rs(TLR4拮抗剂),探讨TLR4/TRAF6/NF-κB通路在ION-CCI模型中的作用,C25-140(TRAF6抑制剂),和PDTC(NF-κBp65抑制剂)。
    结果:ION-CCI手术可降低大鼠机械痛阈,增加ATF3、TLR4、TRAF6、NF-κBp-p65和Nav1.3的表达,但NF-κBp65表达无差异。注射TLR4/TRAF6/NF-κB通路拮抗剂或抑制剂后,Nav1.3表达降低,机械性痛阈升高。
    结论:在ION-CCI大鼠模型中,大鼠三叉神经节TLR4通过TRAF6/NF-κBp65通路调节Nav1.3,TLR4拮抗剂可减轻ION-CCI大鼠的神经病理性疼痛。
    BACKGROUND: Trigeminal neuralgia (TN) is a common and difficult-to-treat neuropathic pain disorder in clinical practice. Previous studies have shown that Toll-like receptor 4 (TLR4) modulates the activation of the NF-κB pathway to affect neuropathic pain in rats. Voltage-gated sodium channels (VGSCs) are known to play an important role in neuropathic pain electrical activity.
    OBJECTIVE: To investigate whether TLR4 can regulate Nav1.3 through the TRAF6/NF-κB p65 pathway after infraorbital nerve chronic constriction injury (ION-CCI).
    METHODS: ION-CCI modeling was performed on SD (Sprague Dawley) rats. To verify the success of the modeling, we need to detect the mechanical pain threshold and ATF3. Then, detecting the expression of TLR4, TRAF6, NF-κB p65, p-p65, and Nav1.3 in rat TG. Subsequently, investigate the role of TLR4/TRAF6/NF-κB pathway in ION-CCI model by intrathecal injections of LPS-rs (TLR4 antagonist), C25-140 (TRAF6 inhibitor), and PDTC (NF-κB p65 inhibitor).
    RESULTS: ION-CCI surgery decreased the mechanical pain threshold of rats and increased the expression of ATF3, TLR4, TRAF6, NF-κB p-p65 and Nav1.3, but there was no difference in NF-κB p65 expression. After inject antagonist or inhibitor of the TLR4/TRAF6/NF-κB pathway, the expression of Nav1.3 was decreased and mechanical pain threshold was increased.
    CONCLUSIONS: In the rat model of ION-CCI, TLR4 in the rat trigeminal ganglion regulates Nav1.3 through the TRAF6/NF-κB p65 pathway, and TLR4 antagonist alleviates neuropathic pain in ION-CCI 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
    SCN3A中编码电压门控钠(Na)通道α亚基Nav1.3的错义变体与SCN3A相关的神经发育障碍(SCN3A-NDD)有关,包括癫痫和皮质发育畸形的一系列疾病。SCN3A中的遗传变异如何导致病理仍不清楚,因为先前关于疾病相关变异的电生理学工作仅在异源细胞系统中进行。进一步探讨SCN3A-NDD发病机制,我们使用CRISPR/Cas9基因编辑来修饰对照人类诱导多能干细胞(iPSC)系,以表达反复的从头错义变体SCN3Ac.2624T>C(pIle875Thr)。随着Ngn2快速诱导方案的建立,我们产生谷氨酸能的前脑样神经元(iNeurons),我们显示其表达SCN3AmRNA和Nav1.3介导的Na电流。我们进行详细的全细胞膜片钳记录以确定SCN3A-p的效果。Ile875Thr变体对内源性Na电流的影响,和内在的兴奋性,人类神经元。与对照神经元相比,表达变体的iNeuros表现出显著增加的缓慢失活/持续的Na+电流,异常放电模式伴有阵发性爆裂和高原样电位伴有动作电位失效,以及用于产生动作电位的超极化电压阈值。然后,我们使用从携带SCN3A-p的患者产生的单独的iPSC线验证这些发现。Ile875Thr变体与相应的CRISPR校正的同基因对照品系相比。最后,我们发现Nav1.3选择性阻断剂ICA-121431的应用使SCN3A-p中的动作电位阈值和异常放电模式正常化。Ile1875ThriNeuroons;相比之下,与作为Na+通道阻滞剂的作用一致,ICA-121431降低对照神经元的兴奋性。我们的发现表明,iNeuons可以模拟SCN3A中遗传变异的影响,但揭示了离子通道水平的功能获得与对神经元兴奋性的影响之间的复杂关系。鉴于SCN3A在发育中的人类神经系统中的瞬时表达,选择性阻断或抑制含Nav1.3的Na+通道可以代表对SCN3A-NDD的治疗方法。
    Missense variants in SCN3A encoding the voltage-gated sodium (Na+) channel α subunit Nav1.3 are associated with SCN3A-related neurodevelopmental disorder (SCN3A-NDD), a spectrum of disease that includes epilepsy and malformation of cortical development. How genetic variation in SCN3A leads to pathology remains unclear, as prior electrophysiological work on disease-associated variants has been performed exclusively in heterologous cell systems. To further investigate the mechanisms of SCN3A-NDD pathogenesis, we used CRISPR/Cas9 gene editing to modify a control human induced pluripotent stem cell (iPSC) line to express the recurrent de novo missense variant SCN3A c.2624T>C (p.Ile875Thr). With the established Ngn2 rapid induction protocol, we generated glutamatergic forebrain-like neurons (iNeurons), which we showed to express SCN3A mRNA and Nav1.3-mediated Na+ currents. We performed detailed whole-cell patch clamp recordings to determine the effect of the SCN3A-p.Ile875Thr variant on endogenous Na+ currents in, and intrinsic excitability of, human neurons. Compared to control iNeurons, variant-expressing iNeurons exhibit markedly increased slowly-inactivating/persistent Na+ current, abnormal firing patterns with paroxysmal bursting and plateau-like potentials with action potential failure, and a hyperpolarized voltage threshold for action potential generation. We then validated these findings using a separate iPSC line generated from a patient harbouring the SCN3A-p.Ile875Thr variant compared to a corresponding CRISPR-corrected isogenic control line. Finally, we found that application of the Nav1.3-selective blocker ICA-121431 normalizes action potential threshold and aberrant firing patterns in SCN3A-p.Ile1875Thr iNeurons; in contrast, consistent with action as a Na+ channel blocker, ICA-121431 decreases excitability of control iNeurons. Our findings demonstrate that iNeurons can model the effects of genetic variation in SCN3A yet reveal a complex relationship between gain-of-function at the level of the ion channel versus impact on neuronal excitability. Given the transient expression of SCN3A in the developing human nervous system, selective blockade or suppression of Nav1.3-containing Na+ channels could represent a therapeutic approach towards SCN3A-NDD.
    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
    Nav1.3,由SCN3A基因编码,是细胞膜上的电压门控钠通道。它在胎儿大脑中大量表达,但在正常成人大脑中很少表达。它参与可兴奋细胞中动作电位的产生和传导。Nav1.3在许多神经系统疾病中起着重要作用。这篇综述的目的是总结有关Nav1.3在神经病学领域的新发现。SCN3A的许多突变可导致神经元过度兴奋,然后引起癫痫。Nav1.3从失活中的快速恢复和缓慢的关闭状态失活动力学导致通道的激活阈值降低和神经元的高频率放电。Nav1.3的过度活跃也会诱导感觉神经元的兴奋性增加,较低的伤害性阈值,和神经性疼痛。本文就Nav1.3的结构和功能作一综述,并重点探讨其与癫痫和神经性疼痛的关系。
    Nav1.3, encoded by the SCN3A gene, is a voltage-gated sodium channel on the cell membrane. It is expressed abundantly in the fetal brain but little in the normal adult brain. It is involved in the generation and conduction of action potentials in excitable cells. Nav1.3 plays an important role in many neurological diseases. The aim of this review is to summarize new findings about Nav1.3 in the field of neurology. Many mutations of SCN3A can lead to neuronal hyperexcitability and then cause epilepsy. The rapid recovery from inactivation and slow closed-state inactivation kinetics of Nav1.3 leads to a reduced activation threshold of the channel and a high frequency of firing of neurons. Hyperactivity of Nav1.3 also induces increased excitability of sensory neurons, a lower nociceptive threshold, and neuropathic pain. This review summarizes the structure and the function of Nav1.3 and focuses on its relationship with epilepsy and 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
    在损伤的外周神经元中持续增加的兴奋性和自发活动对于许多形式的神经性疼痛的发展和持续是必要的。这种异常活性涉及电压门控Na和Ca2通道和超极化激活的环核苷酸门控(HCN)通道的活性和/或表达增加,以及K通道的功能降低。因为它们显示有限的中央副作用,外周限制的Na和Ca2通道阻滞剂和K通道激活剂为疼痛管理提供了潜在的治疗方法。这篇综述概述了外周作用通道调节剂的现状和未来的治疗前景。Nav1.3,Nav1.7,Nav1.8,Cav3.2和HCN2的选择性阻断剂和Kv7.2的激活剂在动物模型中消除了神经性疼痛的体征。不幸的是,它们在临床上的表现令人失望;一些物质未能达到治疗终点,而另一些则产生剂量限制性副作用.尽管如此,外周电压门控阳离子通道保留其作为治疗靶标的希望。前进的道路可能包括(i)进一步改善K通道激活剂如瑞替加滨和ASP0819的结构,以提高选择性和限制毒性;使用或修饰Na通道阻滞剂如维索三嗪,PF-05089771、A803467、PF-01247324、VX-150或蛛形纲毒素如Tap1a;使用Ca2+通道阻滞剂如TTA-P2、TTA-A2、Z944、ACT709478和CNCB-2;(ii)改进评估“疼痛”的方法,而不是使用其他通过NGF基因表达的抗疼痛模型的个性化治疗性治疗性的vor用于治疗T细胞淋巴瘤的组蛋白甲基转移酶抑制剂,或用于治疗类风湿性关节炎的MNK1/2抑制剂的cercosporamide;(vi)使用对不同通道类型或调节过程具有选择性的药物的联合治疗;(vii)指导临床前验证工作以使用人类或人类来源的组织样品;(viii)分子生物学方法的应用,例如成簇规则间隔的短回文重复(CRISPR)技术。
    The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing \"pain\" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
    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
    由周围神经损伤引起的慢性疼痛代表了重要的临床挑战,因为即使是最有效的抗惊厥药物治疗也受到其副作用的限制。我们调查了疼痛行为,轴突信号传导和三叉神经元兴奋性的变化,以及眶下神经卡压(IoNE)后眶下神经和三叉神经节(TG)中电压门控钠通道(Navs)的表达。与Sham相比,IoNE大鼠从神经支配的纤维中增加了A-和C-纤维复合动作电位(CAPs)和A-CAP区域的Aδ成分。在Ione之后,A-和C-纤维CAPs对河豚毒素(TTX)的阻断更敏感,抗TTX的纤维对NaV1.8选择性阻断剂的阻断更敏感,A-803467.虽然NaV1.7阻断剂,单独使用ICA-121431,不影响Aδ-光纤信号的传播,A-803467和4,9-脱水-TTX的累积应用显着降低了IoNE大鼠的Aδ纤维CAP。在中小型TG神经元的膜片钳记录中,IoNE导致动作电位(AP)去极化电流阈值降低,超极化AP电压阈值,AP持续时间增加,和更加去极化的膜电位。虽然IoNE后同侧TG中大多数Navs的转录物减少,NaV1.3、NaV1.7和NaV1.8mRNA,和NaV1.8蛋白,在神经中显著增加。总之,我们的数据表明,NaV1.8的轴突再分布以及较小程度的NaV1.3和NaV1.7有助于IoNE后周围神经中伤害性信号的传播增强.
    Chronic pain arising from peripheral nerve injuries represents a significant clinical challenge because even the most efficacious anticonvulsant drug treatments are limited by their side effects profile. We investigated pain behavior, changes in axonal signal conduction and excitability of trigeminal neurons, and expression of voltage-gated sodium channels (NaVs) in the infraorbital nerve and trigeminal ganglion (TG) after infraorbital nerve entrapment (IoNE). Compared to Sham, IoNE rats had increased A- and C-fiber compound action potentials (CAPs) and Aδ component of A-CAP area from fibers innervating the vibrissal pad. After IoNE, A- and C-fiber CAPs were more sensitive to blockade by tetrodotoxin (TTX), and those fibers that were TTX-resistant were more sensitive to blockade by the NaV1.8 selective blocker, A-803467. Although NaV1.7 blocker, ICA-121431 alone, did not affect Aδ-fiber signal propagation, cumulative application with A-803467 and 4,9-anhydro-TTX significantly reduced the Aδ-fiber CAP in IoNE rats. In patch clamp recordings from small- and medium-sized TG neurons, IoNE resulted in reduced action potential (AP) depolarizing current threshold, hyperpolarized AP voltage threshold, increased AP duration, and a more depolarized membrane potential. While the transcripts of most NaVs were reduced in the ipsilateral TG after IoNE, NaV1.3, NaV1.7, and NaV1.8 mRNAs, and NaV1.8 protein, were significantly increased in the nerve. Altogether, our data suggest that axonal redistribution of NaV1.8, and to a lesser extent NaV1.3, and NaV1.7 contributes to enhanced nociceptive signal propagation in peripheral nerve after IoNE.
    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
    Proparacaine(PPC)是一种先前发现的局部麻醉药,可通过阻断中心Nav1.3进行眼科验光和手术。在这项研究中,我们发现盐酸丙对卡因(PPC-HCl)在毛果芸香碱诱导的癫痫小鼠中具有急性稳健的抗癫痫作用.更重要的是,PPC-HCl的慢性治疗完全终止了自发性复发性癫痫发作,无明显毒性。PPC-HCl的慢性治疗没有引起明显的细胞毒性,神经精神不良反应,肝毒性,心脏毒性,甚至通过全基因组尺度转录组学分析评估的遗传毒性。仅在高剂量(50mg/kg)时,心电图测量的QRS间期略有延长,这与左乙拉西坦的影响相似。然而,为了克服这个潜在的问题,我们采用脂质体包封策略,可以减轻心脏毒性,并制备了一种含有PPC-HCl的水凝胶用于持续释放。将含有脂质体PPC-HCl的热敏壳聚糖基水凝胶植入皮下组织,可在不影响QRS间期的情况下立即缓解癫痫小鼠的自发性复发性癫痫发作。因此,这种新的脂质体水凝胶制剂的普美卡因可以开发作为治疗癫痫的透皮贴剂,避免了目前抗癫痫药物在临床上长期治疗后的严重毒性。
    Proparacaine (PPC) is a previously discovered topical anesthetic for ophthalmic optometry and surgery by blocking the central Nav1.3. In this study, we found that proparacaine hydrochloride (PPC-HCl) exerted an acute robust antiepileptic effect in pilocarpine-induced epilepsy mice. More importantly, chronic treatment with PPC-HCl totally terminated spontaneous recurrent seizure occurrence without significant toxicity. Chronic treatment with PPC-HCl did not cause obvious cytotoxicity, neuropsychiatric adverse effects, hepatotoxicity, cardiotoxicity, and even genotoxicity that evaluated by whole genome-scale transcriptomic analyses. Only when in a high dose (50 mg/kg), the QRS interval measured by electrocardiography was slightly prolonged, which was similar to the impact of levetiracetam. Nevertheless, to overcome this potential issue, we adopt a liposome encapsulation strategy that could alleviate cardiotoxicity and prepared a type of hydrogel containing PPC-HCl for sustained release. Implantation of thermosensitive chitosan-based hydrogel containing liposomal PPC-HCl into the subcutaneous tissue exerted immediate and long-lasting remission from spontaneous recurrent seizure in epileptic mice without affecting QRS interval. Therefore, this new liposomal hydrogel formulation of proparacaine could be developed as a transdermal patch for treating epilepsy, avoiding the severe toxicity after chronic treatment with current antiepileptic drugs in clinic.
    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
    NaV1.3 is a subtype of the voltage-gated sodium channel family. It has been implicated in the pathogenesis of neuropathic pain, although the contribution of this channel to neuronal excitability is not well understood. Tf2, a β-scorpion toxin previously identified from the venom of Tityus fasciolatus, has been reported to selectively activate NaV1.3. Here, we describe the activity of synthetic Tf2 and assess its suitability as a pharmacological probe for NaV1.3. As described for the native toxin, synthetic Tf2 (1 µM) caused early channel opening, decreased the peak current, and shifted the voltage dependence of NaV1.3 activation in the hyperpolarizing direction by -11.3 mV, with no activity at NaV1.1, NaV1.2, and NaV1.4-NaV1.8. Additional activity was found at NaV1.9, tested using the hNav1.9_C4 chimera, where Tf2 (1 µM) shifted the voltage dependence of activation by -6.3 mV. In an attempt to convert Tf2 into an NaV1.3 inhibitor, we synthetized the analogue Tf2[S14R], a mutation previously described to remove the excitatory activity of related β-scorpion toxins. Indeed, Tf2[S14R](10 µM) had reduced excitatory activity at NaV1.3, although it still caused a small -5.8 mV shift in the voltage dependence of activation. Intraplantar injection of Tf2 (1 µM) in mice caused spontaneous flinching and swelling, which was not reduced by the NaV1.1/1.3 inhibitor ICA-121431 nor in NaV1.9-/- mice, suggesting off-target activity. In addition, despite a loss of excitatory activity, intraplantar injection of Tf2[S14R](10 µM) still caused swelling, providing strong evidence that Tf2 has additional off-target activity at one or more non-neuronal targets. Therefore, due to activity at NaV1.9 and other yet to be identified target(s), the use of Tf2 as a selective pharmacological probe may be limited.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Background: To verify the hypothesis that the nature of trigeminal neuralgia (TN) is an ectopic impulse induced by sodium channel modulated by cytokines, we conducted an animal study using the infraorbital nerve chronic constriction injury (CCI) model in rats.Method: The expression of Nav1.3 or IL-6 in the infraorbital nerve (ION) and trigeminal ganglion (TG) were detected by western blot and immunocytochemistry after administration of antisense oligodeoxynucleotide sequence (AS), IL-6 or Anti-IL-6.Results: With intrathecal administration of AS or mismatch oligodeoxynucleotide sequence (MM) in the CCI rats, the Nav1.3-IR in ION and TG accounted for 2.2 ± 0.51% and 8.5 ± 3.1% in AS+CCI group vs. 6.9 ± 1.3% and 38.7 ± 4.8% in MM+CCI group (p < 0.05), respectively. While with local administration of IL-6 in those with sham operation, it accounted for 7.4 ± 2.1% and 45.5 ± 3.4% in IL-6+ sham group vs. 1.9 ± 0.67% and 8.1 ± 1.3% in vehicle+sham group (p < 0.05); with local administration of anti-IL-6 in CCI rats, 4.5 ± 0.78% and 32.1 ± 9.6% in Anti-IL-6+ CCI group vs 8.9 ± 2.1% and 61.4 ± 11.2% in vehicle+CCI group (p < 0.05).Discussion: We believe that the emergence of Nav1.3 from the compressed trigeminal nerve might be an important structural basis for the development of the ectopic excitability on the axon and IL-6 may play a role of necessary precondition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号