Nav1.2

NaV1.2
  • 文章类型: Journal Article
    本征塑性,一个使神经元能够修改其内在属性的基本过程,在塑造神经元输入输出功能中起着至关重要的作用,并且与各种神经和精神疾病有关。尽管它很重要,内在可塑性的潜在分子机制仍然知之甚少。在这项研究中,一种新的泛素连接酶适配器,蛋白酪氨酸磷酸酶受体N型(PTPRN),在颞叶癫痫的背景下,被确定为内在神经元兴奋性的调节剂。PTPRN将NEDD4样E3泛素蛋白连接酶(NEDD4L)招募到NaV1.2钠通道,促进NEDD4L介导的泛素化,NaV1.2的内吞作用在海马颗粒细胞中敲除PTPRN导致NaV1.2介导的钠电流增强和更高的内在兴奋性,导致转基因小鼠癫痫发作易感性增加。相反,腺相关病毒介导的PTPRN在齿状回区域的递送降低了内在兴奋性并降低了癫痫发作易感性。此外,本研究结果表明PTPRN对电压门控钠通道具有选择性调节作用。总的来说,PTPRN在调节内在兴奋性和癫痫发作易感性中起重要作用,提出了精确调制NaV1.2通道功能的潜在策略。
    Intrinsic plasticity, a fundamental process enabling neurons to modify their intrinsic properties, plays a crucial role in shaping neuronal input-output function and is implicated in various neurological and psychiatric disorders. Despite its importance, the underlying molecular mechanisms of intrinsic plasticity remain poorly understood. In this study, a new ubiquitin ligase adaptor, protein tyrosine phosphatase receptor type N (PTPRN), is identified as a regulator of intrinsic neuronal excitability in the context of temporal lobe epilepsy. PTPRN recruits the NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L) to NaV1.2 sodium channels, facilitating NEDD4L-mediated ubiquitination, and endocytosis of NaV1.2. Knockout of PTPRN in hippocampal granule cells leads to augmented NaV1.2-mediated sodium currents and higher intrinsic excitability, resulting in increased seizure susceptibility in transgenic mice. Conversely, adeno-associated virus-mediated delivery of PTPRN in the dentate gyrus region decreases intrinsic excitability and reduces seizure susceptibility. Moreover, the present findings indicate that PTPRN exerts a selective modulation effect on voltage-gated sodium channels. Collectively, PTPRN plays a significant role in regulating intrinsic excitability and seizure susceptibility, suggesting a potential strategy for precise modulation of NaV1.2 channels\' function.
    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
    头颈部鳞状细胞癌(HNSCC)是头颈部肿瘤(HNTs)最常见的组织学形式,起源于嘴唇和口腔的上皮,咽部,喉部,唾液腺,鼻腔,和鼻窦。主要风险因素包括消费各种形式的烟草和酒精,以及高危型人乳头瘤病毒或EB病毒感染。不管病因是什么,男性患不同类型HNTs的风险比女性高2至6倍以上。造成这种差异的原因可能在于生物和社会心理因素的结合。因此,假设暴露于女性性激素,主要是雌激素,为女性提供保护,防止HNTs的形成和转移。在这次审查中,我们合成了有关雌激素和雌激素受体(ERs)在HNTs发育和进展中的作用的现有知识,特别强调膜ER,这些研究要少得多。我们可以总结一下,除了流行病学研究明确指出雌激素对女性的保护作用之外,两个核ER的表达增加,ERα,和ERβ,和膜ER,ERα36、GPER1和NaV1.2存在于不同类型的HNSCC中,抗雌激素可作为一种有效的治疗方法。
    Head and neck squamous cell carcinoma (HNSCC) is the most common histological form of head and neck tumors (HNTs), which originate from the epithelium of the lips and oral cavity, pharynx, larynx, salivary glands, nasal cavity, and sinuses. The main risk factors include consumption of tobacco in all forms and alcohol, as well as infections with high-risk human papillomaviruses or the Epstein-Barr virus. Regardless of the etiological agent, the risk of developing different types of HNTs is from two to more than six times higher in males than in females. The reason for such disparities probably lies in a combination of both biological and psychosocial factors. Therefore, it is hypothesized that exposure to female sex hormones, primarily estrogen, provides women with protection against the formation and metastasis of HNTs. In this review, we synthesized available knowledge on the role of estrogen and estrogen receptors (ERs) in the development and progression of HNTs, with special emphasis on membrane ERs, which are much less studied. We can summarize that in addition to epidemiologic studies unequivocally pointing to the protective effect of estrogen in women, an increased expression of both nuclear ERs, ERα, and ERβ, and membrane ERs, ERα36, GPER1, and NaV1.2, was present in different types of HNSCC, for which anti-estrogens could be used as an effective therapeutic approach.
    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
    继发于电压门控钠通道NaV1.2功能改变的SCN2A相关疾病很少有临床异质性表达,包括癫痫,自闭症,以及多种严重到严重的损伤和其他疾病。为了进一步了解临床表型及其与通道功能的关系,81例患者(36,44%为女性,中位年龄5.4岁),对69种独特的SCN2A变体进行了系统表型分析,并对其NaV1.2通道功能进行了系统评估。参与者是通过FamileSCN2A基金会招募的。原发性表型(新生儿发作的癫痫,N=27;婴儿发病,N=18;发病后期N=24;自闭症无癫痫发作,(N=12)与非癫痫发作严重程度指数(p=0.002)密切相关,这是基于粗大运动的严重损伤,精细电机,沟通能力,胃造瘘管依赖,以及皮质视觉障碍和脊柱侧凸的诊断。非癫痫发作严重程度在新生儿发作组中最大,在自闭症组中最小(p=0.002)。严重程度指数最低的儿童仍然严重受损,Vineland自适应行为综合得分平均为49.5分(比测试的标准参考平均值低>3SD)。癫痫痉挛在婴儿发作(67%)比新生儿(22%)或晚期(29%)癫痫中明显更常见(p=0.007)。原发性表型也与变异功能密切相关(p<0.0001);在新生儿发作性癫痫中,功能获得和混合功能变异占主导地位,婴儿发作性癫痫患者的功能逐渐丧失,和严重和完全的功能丧失在后期发作的癫痫和自闭症组。探索性聚类分析确定了五组,代表(1)主要是晚期发作的癫痫,具有中度功能变异和低严重程度指数,(2)大多数婴儿发作性癫痫,功能变异中度丧失,但严重程度指数较高,(3)迟发性和自闭症,仅具有最低的严重程度指数(大多数为0)和功能变异的严重/完全丧失。两个唯一的新生儿群在非癫痫发作严重程度评分上彼此区分,其次在变异功能上彼此区分。主要表型和变异功能之间的关系强调了发育因素在SCN2A变异体对NaV1.2通道功能的影响的差异临床表达中的作用。SCN2A疾病的非癫痫发作严重程度取决于癫痫发作时的年龄(主要表型)和变异功能的组合。随着SCN2A相关疾病的精准治疗迈向临床试验,了解变异功能与临床疾病表达之间的关系对于确定这些试验的合适患者和选择有效的临床结局很有价值.
    SCN2A-related disorders secondary to altered function in the voltage-gated sodium channel NaV1.2 are rare with clinically heterogeneous expressions that include epilepsy, autism, and multiple severe to profound impairments and other conditions. To advance understanding of the clinical phenotypes and their relation to channel function, 81 patients (36, 44% female, median age 5.4 years) with 69 unique SCN2A variants were systematically phenotyped and their NaV1.2 channel function systematically assessed. Participants were recruited through the FamileSCN2A Foundation. Primary phenotype (epilepsy of neonatal-onset, N=27; infant onset, N=18; and later onset N=24; and autism without seizures, (N=12) was strongly correlated with a non-seizure severity index (p=0.002), which was based on presence of severe impairments in gross motor, fine motor, communication abilities, gastrostomy tube dependence, and diagnosis of cortical visual impairment and scoliosis. Non-seizure severity was greatest in the neonatal-onset group and least in the autism group (p=0.002). Children with the lowest severity indices were still severely impaired, as reflected by an average Vineland adaptive behavior composite score of 49.5 (>3 SD below the test\'s norm-referenced mean). Epileptic spasms were significantly more common in infant onset (67%) than in neonatal (22%) or later-onset (29%) epilepsy (p=0.007). Primary phenotype also strongly correlated with variant function (p<0.0001); gain of function and mixed function variants predominated in neonatal-onset epilepsy, shifting to moderate loss of function in infant-onset epilepsy, and severe and complete loss of function in later-onset epilepsy and autism groups. Exploratory cluster analysis identified five groups representing (1) primarily later-onset epilepsy with moderate loss of function variants and low severity indices, (2) mostly infant-onset epilepsy with moderate loss of function variants but higher severity indices, (3) late-onset and autism only with the lowest severity indices (mostly 0) and severe/complete loss of function variants. Two exclusively neonatal clusters were distinguished from each other largely on non-seizure severity scores and secondarily on variant function. The relation between primary phenotype and variant function emphasizes the role of developmental factors in the differential clinical expression of SCN2A variants based on their effects on NaV1.2 channel function. The non-seizure severity of SCN2A disorders depends on a combination of the age at seizure onset (primary phenotype) and variant function. As precision therapies for SCN2A-related disorders advance toward clinical trials, knowledge of the relationship between variant function and clinical disease expression will be valuable for identifying appropriate patients for these trials and in selecting efficient clinical outcomes.
    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
    SCN2A基因的许多致病变异,编码电压门控钠通道α2亚基Nav1.2蛋白,已经在包括精神分裂症在内的广泛的神经精神疾病中被发现。然而,由这些变异引起的精神分裂症相关行为异常的病理机制仍然知之甚少.在这项研究中,我们对精神分裂症相关脑区有选择性Scn2a缺失的小鼠系进行了表征,内侧前额叶皮质(mPFC)或腹侧被盖区(VTA),通过将表达Cre重组酶的腺相关病毒(AAV)注射到纯合Scn2a-floxed(Scn2afl/fl)小鼠中获得,其中Scn2a的表达在Cre重组酶存在下局部缺失。mPFC中缺乏Scn2a的小鼠表现出声学惊吓反应中的前脉冲抑制(PPI)降低的趋势。相反,VTA中缺乏Scn2a的小鼠显示PPI显著增加。我们还发现,mPFC中缺乏Scn2a的小鼠表现出增强的社交能力,运动活动减少,增加了焦虑样的行为,而在VTA中缺乏Scn2a的小鼠除了作为运动活动之一的垂直活动外,这些参数没有显示任何其他异常。这些结果表明,mPFC和VTA中的Scn2a缺陷与SCN2A变体患者的精神分裂症表型成反比。
    Numerous pathogenic variants of SCN2A gene, encoding voltage-gated sodium channel α2 subunit Nav1.2 protein, have been identified in a wide spectrum of neuropsychiatric disorders including schizophrenia. However, pathological mechanisms for the schizophrenia-relevant behavioral abnormalities caused by the variants remain poorly understood. Here in this study, we characterized mouse lines with selective Scn2a deletion at schizophrenia-related brain regions, medial prefrontal cortex (mPFC) or ventral tegmental area (VTA), obtained by injecting adeno-associated viruses (AAV) expressing Cre recombinase into homozygous Scn2a-floxed (Scn2afl/fl) mice, in which expression of the Scn2a was locally deleted in the presence of Cre recombinase. The mice lacking Scn2a in the mPFC exhibited a tendency for a reduction in prepulse inhibition (PPI) in acoustic startle response. Conversely, the mice lacking Scn2a in the VTA showed a significant increase in PPI. We also found that the mice lacking Scn2a in the mPFC displayed increased sociability, decreased locomotor activity, and increased anxiety-like behavior, while the mice lacking Scn2a in the VTA did not show any other abnormalities in these parameters except for vertical activity which is one of locomotor activities. These results suggest that Scn2a-deficiencies in mPFC and VTA are inversely relevant for the schizophrenic phenotypes in patients with SCN2A variants.
    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: Preprint
    SCN2A中的致病变体与一系列神经发育障碍(NDD)相关。尽管很大程度上是单基因的,与SCN2A相关的NDD显示出相当大的表型变异和复杂的基因型-表型相关性。遗传修饰可导致与罕见驱动突变相关的疾病表型的变异性。因此,不同的遗传背景在近交啮齿动物品系已被证明影响疾病相关的表型,包括与SCN2A相关的NDD。最近,我们开发了变异体SCN2A-p的小鼠模型。K1422E在C57BL/6J(B6)菌株上保持为等基因系。我们对杂合Scn2aK1422E小鼠中NDD表型的初步表征揭示了焦虑相关行为和癫痫发作易感性的改变。为了确定背景菌株是否影响Scn2aK1422E小鼠模型中的表型严重性,比较了B6和[DBA/2JxB6]F1杂种(F1D2)菌株的小鼠表型。来自神经行为测定的收敛证据表明,与野生型相比,Scn2aK1422E小鼠中的焦虑样行为更低,并且进一步表明,与F1D2背景相比,这种效应在B6背景上更显著。尽管罕见的自发性癫痫发作的发生没有应变依赖性差异,对化学惊厥海人酸的反应揭示了癫痫发作的普遍性和致死风险的差异,基于应变和性别的变化。在Scn2aK1422E小鼠模型中继续检查菌株依赖性效应可以揭示具有独特易感性谱的遗传背景,这将与未来对特定性状的研究相关,并能够鉴定高度渗透的表型和修饰基因,这些基因可以提供有关K1422E变体的主要致病机制的线索。
    Pathogenic variants in SCN2A are associated with a range of neurodevelopmental disorders (NDD). Despite being largely monogenic, SCN2A-related NDD show considerable phenotypic variation and complex genotype-phenotype correlations. Genetic modifiers can contribute to variability in disease phenotypes associated with rare driver mutations. Accordingly, different genetic backgrounds across inbred rodent strains have been shown to influence disease-related phenotypes, including those associated with SCN2A-related NDD. Recently, we developed a mouse model of the variant SCN2A-p.K1422E that was maintained as an isogenic line on the C57BL/6J (B6) strain. Our initial characterization of NDD phenotypes in heterozygous Scn2aK1422E mice revealed alterations in anxiety-related behavior and seizure susceptibility. To determine if background strain affects phenotype severity in the Scn2aK1422E mouse model, phenotypes of mice on B6 and [DBA/2J×B6]F1 hybrid (F1D2) strains were compared. Convergent evidence from neurobehavioral assays demonstrated lower anxiety-like behavior in Scn2aK1422E mice compared to wild-type and further suggested that this effect is more pronounced on the B6 background compared to the F1D2 background. Although there were no strain-dependent differences in occurrence of rare spontaneous seizures, response to the chemoconvulsant kainic acid revealed differences in seizure generalization and lethality risk, with variation based on strain and sex. Continued examination of strain-dependent effects in the Scn2aK1422E mouse model could reveal genetic backgrounds with unique susceptibility profiles that would be relevant for future studies on specific traits and enable the identification of highly penetrant phenotypes and modifier genes that could provide clues about the primary pathogenic mechanism of the K1422E variant.
    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
    将传统的突变扫描应用于电压门控钠通道(Navs)并在功能上注释这些基因中的大量编码变体是具有挑战性的。使用胞嘧啶碱基编辑器和合并的活力测定,我们筛选了368个指导RNA(gRNA)的文库,以鉴定超过100个改变NaV1.2功能的gRNA。我们对由这些gRNA的子集进行的碱基编辑进行测序,以确认驱动通道功能变化的特定变体。这些通道变体的电生理表征验证了筛选结果并提供了通道扰动的功能机制。由这些gRNA引起的大多数变化可归类为功能丧失以及导致NaV1.2通道功能获得的两个错义突变。这种在功能上大规模表征离子通道蛋白变体的两级策略识别了NaV1.2中的大量功能丧失突变。
    It is challenging to apply traditional mutational scanning to voltage-gated sodium channels (NaVs) and functionally annotate the large number of coding variants in these genes. Using a cytosine base editor and a pooled viability assay, we screen a library of 368 guide RNAs (gRNAs) tiling NaV1.2 to identify more than 100 gRNAs that change NaV1.2 function. We sequence base edits made by a subset of these gRNAs to confirm specific variants that drive changes in channel function. Electrophysiological characterization of these channel variants validates the screen results and provides functional mechanisms of channel perturbation. Most of the changes caused by these gRNAs are classifiable as loss of function along with two missense mutations that lead to gain of function in NaV1.2 channels. This two-tiered strategy to functionally characterize ion channel protein variants at scale identifies a large set of loss-of-function mutations in NaV1.2.
    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
    视网膜神经节细胞(RGC)是眼睛的尖峰投射神经元,可编码视觉环境的不同特征。对向不同RGC类型提供突触输入以驱动特征选择性的电路进行了广泛研究,但是,旨在了解内在属性以及它们如何影响特征选择性的研究较少。我们在鼠标中引入RGC类型,突发对比压制(bSbC)RGC,并将其与OFF持续阿尔法(OFFsA)进行比较。它们的对比响应函数的差异不是由突触输入的差异引起的,而是由其固有特性引起的。尖峰生成是这种功能差异背后的关键内在属性;bSbCRGC经历去极化阻滞,而OFFsARGC保持较高的尖峰率。我们的结果表明,内在特性的差异使这两种RGC类型能够检测并将相同视觉刺激的不同特征传递到大脑。
    Retinal ganglion cells (RGCs) are the spiking projection neurons of the eye that encode different features of the visual environment. The circuits providing synaptic input to different RGC types to drive feature selectivity have been studied extensively, but there has been less research aimed at understanding the intrinsic properties and how they impact feature selectivity. We introduce an RGC type in the mouse, the Bursty Suppressed-by-Contrast (bSbC) RGC, and compared it to the OFF sustained alpha (OFFsA). Differences in their contrast response functions arose from differences not in synaptic inputs but in their intrinsic properties. Spike generation was the key intrinsic property behind this functional difference; the bSbC RGC undergoes depolarization block while the OFFsA RGC maintains a high spike rate. Our results demonstrate that differences in intrinsic properties allow these two RGC types to detect and relay distinct features of an identical visual stimulus to the brain.
    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
    SCN2A编码在整个中枢神经系统中主要在兴奋性神经元中表达的电压门控钠通道(NaV1.2)。SCN2A的致病变异与癫痫和神经发育障碍有关。已经描述了基因型-表型相关性,功能丧失变异通常与神经发育迟缓和迟发性癫痫发作有关,而功能获得变异更常导致早期婴儿发作癫痫。然而,大多数致病SCN2A变异体的真正电生理效应尚未被表征.我们报告了一名在新生儿期出现局灶性癫痫发作的婴儿。她被发现有一个马赛克c.2635G>A,SCN2A中的p.Gly879Arg变体。在成人和新生儿NaV1.2同种型上表达的变体的电压钳研究表明,功能的增加和丧失是混合的,主要具有功能丧失效应,细胞表面表达和电流密度降低。其他小的电生理改变包括激活的电压依赖性降低和失活的电压依赖性增加。这种主要的功能丧失效应的发现是出乎意料的,因为婴儿的早期癫痫发作表明主要是功能获得效应。这种情况说明我们对基因型-表型相关性的理解仍然有限,并且突出了SCN2A变体的潜在电生理效应的复杂性。电压门控钠通道在中枢神经系统中起着重要作用,据报道,这些突变是导致癫痫的原因。我们在这里报告了一名婴儿,在新生儿期出现癫痫伴转移性局灶性癫痫发作(EIMFS),马赛克c.2635G>A,在编码NaV1.2钠通道的SCN2A基因上产生p.Gly879Arg错义突变。该变体的生物物理表征揭示了功能增益和功能丧失效应的混合。
    SCN2A encodes a voltage-gated sodium channel (NaV1.2) expressed throughout the central nervous system in predominantly excitatory neurons. Pathogenic variants in SCN2A are associated with epilepsy and neurodevelopmental disorders. Genotype-phenotype correlations have been described, with loss-of-function variants typically being associated with neurodevelopmental delay and later-onset seizures, whereas gain-of-function variants more often result in early infantile-onset epilepsy. However, the true electrophysiological effects of most disease-causing SCN2A variants have yet to be characterized. We report an infant who presented with migrating focal seizures in the neonatal period. She was found to have a mosaic c.2635G>A, p.Gly879Arg variant in SCN2A. Voltage-clamp studies of the variant expressed on adult and neonatal NaV1.2 isoforms demonstrated a mixed gain and loss of function, with predominantly a loss-of-function effect with reduced cell surface expression and current density. Additional small electrophysiological alterations included a decrease in the voltage dependence of activation and an increase in the voltage dependence of inactivation. This finding of a predominantly loss-of-function effect was unexpected, as the infant\'s early epilepsy onset would have suggested a predominantly gain-of-function effect. This case illustrates that our understanding of genotype-phenotype correlations is still limited and highlights the complexity of the underlying electrophysiological effects of SCN2A variants.NEW & NOTEWORTHY Voltage-gated sodium channels play an important role in the central nervous system, mutations in which have been reported to be responsible for epilepsy. We report here an infant presenting with epilepsy of infancy with migrating focal seizures (EIMFS) in the neonatal period with a mosaic c.2635G>A, resulting in a p.Gly879Arg missense mutation on the SCN2A gene encoding NaV1.2 sodium channels. Biophysical characterization of this variant revealed a mixture of gain- and loss-of-function effects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Editorial
    癫痫是最常见的神经系统疾病之一。癫痫患者面临癫痫发作,这主要是由增强的神经元兴奋性和/或降低的神经元抑制活性引起的。SCN2A编码神经元电压门控钠通道,NaV1.2主要存在于整个大脑的兴奋性神经元中。NaV1.2最集中在皮质纹状体回路的主要神经元内,其中包括内侧前额叶皮质的锥体神经元和纹状体的中刺神经元。在成人发育的早期阶段,NaV1.2通道在这些神经元中动作电位的产生和传播中起关键作用。SCN2A的功能变异导致无源性癫痫发作和癫痫,而SCN2A的功能丧失变异体是自闭症谱系障碍和智力障碍的主要原因。先前的研究表明,小鼠中Scn2a基因的完全缺失是致命的,而Scn2a基因的部分破坏(小于50%)会导致神经元兴奋性的抑制。Yang博士实验室最近的一项研究揭示了严重NaV1.2缺乏症小鼠的意外结果,他们证明了NaV1.2中Scn2a基因的严重缺失(约68%的基因破坏)会触发成年小鼠的神经元过度兴奋。他们的发现可以解释令人困惑的临床观察,即某些患有NaV1.2缺乏症的人仍然会出现无缘无故的癫痫发作。知道使用钠通道阻滞剂只会加剧癫痫发作,需要了解NaV1.2通道的内在性质提供了未来的重要研究课题。
    Epilepsy is one of the most common neurological diseases. Epileptic individuals are faced with seizures, which are largely caused by enhanced neuronal excitability and/or decreased neuronal inhibitory activity. SCN2A encodes a neuronal voltage-gated sodium channel, NaV1.2 that is primarily found in excitatory neurons throughout the brain. NaV1.2 is most concentrated within the principal neurons of the corticostriatal circuit, which includes pyramidal neurons in the medial prefrontal cortex and medium spiny neurons in the striatum. In the early stage of adult development, the NaV1.2 channel plays critical roles in generation and propagation of action potentials in these neurons. Gain of Function variants of SCN2A results in unprovoked seizures and epilepsy, while loss-of-function variants of SCN2A is a leading cause for autism spectrum disorder as well as intellectual disability. Previous studies have shown that full deletion of Scn2a gene in mice is lethal and partial disruption of Scn2a gene (less than 50%) leads to inhibition of neuronal excitability. A recent study from Dr. Yang\'s laboratory revealed an unexpected result from mice with severe NaV1.2 deficiency and they demonstrated that severe deletion of Scn2a gene (around 68% gene disruption) in NaV1.2 triggers neuronal hyperexcitability in adult mice. Their findings may explain the puzzling clinical observation that certain individuals with NaV1.2 deficiency still develop unprovoked seizure. With the knowledge that using sodium-channel blockers simply exacerbates the seizure, the need for understanding the intrinsic nature of the NaV1.2 channel provides an important research topic in the future.
    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
    自闭症谱系障碍(ASD)影响美国约2%的人口,单基因形式的ASD通常导致该疾病最严重的表现。最近,SCN2A已成为与ASD相关的主要基因,其中异常睡眠模式是一种常见的合并症。SCN2A编码电压门控钠通道NaV1.2。主要在大脑中表达,NaV1.2介导神经元的动作电位放电。临床研究发现,很大一部分SCN2A缺乏症儿童存在睡眠障碍,严重影响受影响的个人及其照顾者的生活质量。与NaV1.2缺乏相关的睡眠障碍的潜在机制,然而,不知道。使用基因陷阱Scn2a缺陷小鼠模型(Scn2atrap),我们发现Scn2a缺乏导致觉醒增加和非快速眼动(NREM)睡眠减少.视交叉上核(SCN)包含区域的大脑区域特异性Scn2a缺陷,这与昼夜节律有关,部分概括了睡眠障碍表型。在细胞层面,我们发现Scn2a缺乏破坏了SCN区域自发放电神经元的放电模式。在分子水平上,RNA测序分析揭示了昼夜节律夹带途径中差异表达的基因,包括核心时钟基因Per1和Per2。进行基于转录组的化合物发现,我们鉴定了右他比诺(HU-211),一种推定的谷氨酸受体调节剂,可以部分逆转小鼠的睡眠障碍。总的来说,我们的研究揭示了Scn2a缺乏相关睡眠障碍的潜在分子和细胞机制,这可能会为受影响的个体提供潜在的药物遗传学干预措施。
    Autism spectrum disorder (ASD) affects ~2% of the population in the US, and monogenic forms of ASD often result in the most severe manifestation of the disorder. Recently, SCN2A has emerged as a leading gene associated with ASD, of which abnormal sleep pattern is a common comorbidity. SCN2A encodes the voltage-gated sodium channel NaV1.2. Predominantly expressed in the brain, NaV1.2 mediates the action potential firing of neurons. Clinical studies found that a large portion of children with SCN2A deficiency have sleep disorders, which severely impact the quality of life of affected individuals and their caregivers. The underlying mechanism of sleep disturbances related to NaV1.2 deficiency, however, is not known. Using a gene-trap Scn2a-deficient mouse model (Scn2atrap), we found that Scn2a deficiency results in increased wakefulness and reduced non-rapid-eye-movement (NREM) sleep. Brain region-specific Scn2a deficiency in the suprachiasmatic nucleus (SCN) containing region, which is involved in circadian rhythms, partially recapitulates the sleep disturbance phenotypes. At the cellular level, we found that Scn2a deficiency disrupted the firing pattern of spontaneously firing neurons in the SCN region. At the molecular level, RNA-sequencing analysis revealed differentially expressed genes in the circadian entrainment pathway including core clock genes Per1 and Per2. Performing a transcriptome-based compound discovery, we identified dexanabinol (HU-211), a putative glutamate receptor modulator, that can partially reverse the sleep disturbance in mice. Overall, our study reveals possible molecular and cellular mechanisms underlying Scn2a deficiency-related sleep disturbances, which may inform the development of potential pharmacogenetic interventions for the affected individuals.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号