KCNK4

KCNK4
  • 文章类型: Journal Article
    背景:钾离子通道在维持细胞电稳定性中起着至关重要的作用,并且与各种癫痫有关。KCNK4中的杂合致病变异导致可识别的神经发育综合征,伴有面部畸形,多毛症,癫痫,智力残疾(ID),和牙龈过度生长(FHEIG)。迄今为止,全球范围内对FHEIG患者的描述不超过9例,但对KCNK4相关疾病的癫痫表型知之甚少.
    方法:我们在患有耐药性夜间癫痫发作的患者中,在KCNK4中发现了一种新的从头p。(Gly139Arg)变体,轻微的ID,和畸形特征。对该变体的计算机模拟分析强烈表明存在功能增益效应。我们对以前发表的病例进行了回顾性审查,专注于癫痫的特征和对各种治疗的反应。
    结果:迄今为止,据报道,有8/10的KCNK4相关疾病患者出现癫痫。癫痫发作的平均年龄为1.8岁,最常见的发作类型是双侧强直阵挛性发作(5/8)。钠通道阻滞剂和丙戊酸盐对大多数患者有效,但在3/8的癫痫是耐药的。在使用碳酸酐酶抑制剂sulthiame治疗后,我们的患者癫痫发作控制得到了改善。有趣的是,患者表现出周围神经兴奋过度综合征的特征,钾电导增加引起的钾通道病中以前没有描述过的现象。
    结论:KCNK4的功能增益变异导致一系列癫痫,从良性孤立性癫痫到癫痫性脑病,最常观察到的是局灶性至双侧强直阵挛性癫痫发作。重要的是,表现为轻度神经外表型而无特征性面部畸形或全身性多毛症的患者亚组。该报告扩展了KNCK4相关疾病的表型谱,并为这种罕见的神经发育综合征的临床异质性提供了新的见解。
    BACKGROUND: Potassium ion channels play a crucial role in maintaining cellular electrical stability and are implicated in various epilepsies. Heterozygous pathogenic variants in KCNK4 cause a recognizable neurodevelopmental syndrome with facial dysmorphism, hypertrichosis, epilepsy, intellectual disability (ID), and gingival overgrowth (FHEIG). To date, no more than nine patients with FHEIG have been described worldwide and still little is known about epileptic phenotype in KCNK4-related disease.
    METHODS: We identified a novel de novo p.(Gly139Arg) variant in KCNK4 in a patient with drug-resistant nocturnal seizures, mild ID, and dysmorphic features. In silico analyses of the variant strongly suggest a gain-of-function effect. We conducted a retrospective review of previously published cases, focusing on the epileptic features and response to various treatments.
    RESULTS: To date, epilepsy has been reported in 8/10 patients with KCNK4-related disease. The mean age of seizure onset was 1.8 years, and the most common seizure type was focal to bilateral tonic-clonic (5/8). Sodium channel blockers and valproate were effective in the majority of patients, but in 3/8 the epilepsy was drug-resistant. Our patient showed improved seizure control after treatment with the carbonic anhydrase inhibitor sulthiame. Interestingly, the patient showed features of peripheral nerve hyperexcitability syndrome, a phenomenon not previously described in potassium channelopathies caused by increased K+ conductance.
    CONCLUSIONS: Gain-of-function variants in KCNK4 cause a spectrum of epilepsies, ranging from benign isolated epilepsy to epileptic encephalopathy, with focal to bilateral tonic-clonic seizures being the most commonly observed. Importantly, a subgroup of patients present with a mild extra-neurological phenotype without characteristic facial dysmorphism or generalized hypertrichosis. This report expands the phenotypic spectrum of KNCK4-associated disease and provides new insights into the clinical heterogeneity of this rare neurodevelopmental syndrome.
    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
    轴突初始节段(AIS)是神经元中的关键区室。它将突触后输入转换为动作电位,然后触发信息传递到目标神经元。该过程依赖于多个电压门控钠(NaV)和钾(KV)通道的存在,这些通道在AIS上以高密度积累。TRAAK是一种机械敏感性泄漏钾通道,最近位于Ranvier的节点。这里,我们发现TRAAK也存在于成年大鼠大脑的海马和皮质神经元的AISs以及培养的大鼠海马神经元的AISs中。我们表明,AIS定位是由C端锚蛋白G结合序列驱动的,该序列以190nm间隔的周期性模式组织TRAAK,该模式与周期性组织的锚蛋白G共同分布。我们还发现,虽然已识别的锚蛋白G结合基序类似于NaV1和KV7.2/KV7.3通道中已知的锚蛋白G结合基序,它是通过趋同进化获得的。我们的发现将TRAAK确定为AIS离子通道,该通道可融合地获得锚蛋白G结合基序,并扩展了锚蛋白G的作用,以包括AIS上离子通道的纳米级组织。
    The axon initial segment (AIS) is a critical compartment in neurons. It converts postsynaptic input into action potentials that subsequently trigger information transfer to target neurons. This process relies on the presence of several voltage-gated sodium (NaV) and potassium (KV) channels that accumulate in high densities at the AIS. TRAAK is a mechanosensitive leak potassium channel that was recently localized to the nodes of Ranvier. Here, we uncover that TRAAK is also present in AISs of hippocampal and cortical neurons in the adult rat brain as well as in AISs of cultured rat hippocampal neurons. We show that the AIS localization is driven by a C-terminal ankyrin G-binding sequence that organizes TRAAK in a 190 nm spaced periodic pattern that codistributes with periodically organized ankyrin G. We furthermore uncover that while the identified ankyrin G-binding motif is analogous to known ankyrin G-binding motifs in NaV1 and KV7.2/KV7.3 channels, it was acquired by convergent evolution. Our findings identify TRAAK as an AIS ion channel that convergently acquired an ankyrin G-binding motif and expand the role of ankyrin G to include the nanoscale organization of ion channels at the AIS.
    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
    KCNK4基因的异常超极化,在神经系统中表达,大脑,和牙周膜成纤维细胞,导致神经递质敏感性受损,心律失常,和内分泌功能障碍,还有,进行性细胞增殖。据报道,KCNK4基因的功能变异从头获得可引起以面部畸形为特征的可识别综合征,多毛症,癫痫,智力/发育迟缓,和牙龈过度生长(FHEIG,OMIM#618381)。FHEIG极为罕见,文献中仅报道了三例。在这里,我们描述了第一个继承的KCNK4变体(c.730G>C,p.Ala244Pro)在一个埃及男孩和他的母亲中。由于患者表现出综合征的全面图像,而母亲仅表现为多毛症和牙龈过度生长,而没有任何神经系统表现,因此注意到了表型表达。c.730G>C(p。Ala244Pro)变体之前在单个患者中进行了描述,并且在将表型与我们的患者进行比较时,表型-基因型相关性似乎很可能.心房颤动和关节松弛是我们患者发现的新的相关发现,扩展了该综合征的临床表型。为受影响的男孩提供了牙科管理,并且随着患者恢复微笑,出现了显着改善,恢复咀嚼功能,恢复了他的心理稳定.
    Abnormal hyperpolarization of the KCNK4 gene, expressed in the nervous system, brain, and periodontal ligament fibroblasts, leads to impaired neurotransmitter sensitivity, cardiac arrhythmias, and endocrine dysfunction, as well as, progressive cell proliferation. De novo gain of function variants in the KCNK4 gene were reported to cause a recognizable syndrome characterized by facial dysmorphism, hypertrichosis, epilepsy, intellectual/developmental delay, and gingival overgrowth (FHEIG, OMIM# 618381). FHEIG is extremely rare with only three reported cases in the literature. Herein, we describe the first inherited KCNK4 variant (c.730G>C, p.Ala244Pro) in an Egyptian boy and his mother. Variable phenotypic expressivity was noted as the patient presented with the full-blown picture of the syndrome while the mother presented only with hypertrichosis and gingival overgrowth without any neurological manifestations. The c.730G>C (p.Ala244Pro) variant was described before in a single patient and when comparing the phenotype with our patient, a phenotype-genotype correlation seems likely. Atrial fibrillation and joint laxity are new associated findings noted in our patient extending the clinical phenotype of the syndrome. Dental management was offered to the affected boy and a dramatic improvement was noted as the patient regained his smile, restored the mastication function, and resumed his psychological stability.
    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: Case Reports
    SCN8A的变体与几种疾病相关,包括发育性脑病和癫痫性脑病,中度癫痫或轻度至中度发育性和癫痫性脑病,自限性家族性婴儿癫痫,伴有全身性癫痫的神经发育迟缓,没有癫痫的神经发育障碍,低张力,和运动障碍。在这里,我们报道了一名8岁的摩洛哥男孩,患有不明原因的中度癫痫,智力残疾,自闭症谱系障碍,和多动症。患者表现出正常的46,XY核型和正常的比较基因组杂交谱。进行全外显子组测序,在KCNK4和SCN8A中鉴定出杂合变体。SCN8A变体[c.4499C>T(p。Pro1500Leu)]也在健康母亲中检测到,并被归类为具有不确定临床意义的变体。这种变异发生在一个高度保守的结构域,这可能会影响编码蛋白质的功能。需要更多的研究来证实这种新变体的致病性,以建立有效的护理,管理,和受影响个体的遗传咨询。
    Variants in SCN8A are associated with several diseases, including developmental and epileptic encephalopathy, intermediate epilepsy or mild-to-moderate developmental and epileptic encephalopathy, self-limited familial infantile epilepsy, neurodevelopmental delays with generalized epilepsy, neurodevelopmental disorder without epilepsy, hypotonia, and movement disorders. Herein, we report an 8-year-old Moroccan boy with intermediate epilepsy of unknown origin, intellectual disability, autism spectrum disorder, and hyperactivity. The patient presented a normal 46, XY karyotype and a normal comparative genomic hybridization profile. Whole-exome sequencing was performed, and heterozygous variants were identified in KCNK4 and SCN8A. The SCN8A variant [c.4499C > T (p.Pro1500Leu)] was also detected in the healthy mother and was classified as a variant of uncertain clinical significance. This variant occurs in a highly conserved domain, which may affect the function of the encoded protein. More studies are needed to confirm the pathogenicity of this novel variant to establish the effective care, management, and genetic counselling of affected individuals.
    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
    由于死亡率高,卵巢癌严重威胁着女性的健康。广泛的腹部转移和化疗耐药是卵巢癌死亡的主要原因。通过lncRNA测序,我们之前的研究鉴定了lncRNASLC25A21-AS1,它在化疗耐药的卵巢癌细胞中显著下调.在这项研究中,我们旨在评估SLC25A21-AS1在卵巢癌中的作用和机制。通过qRT-PCR和在线数据库GEPIA分析SLC25A21-AS1的表达。用CCK-8、transwell、和流式细胞术。通过RNA测序分析其具体机制,RNA结合蛋白免疫沉淀,救援实验,和生物信息学分析。SLC25A21-AS1在卵巢癌组织和细胞系中降低。SLC25A21-AS1的过表达增强了卵巢癌细胞对紫杉醇和顺铂的敏感性,抑制细胞增殖,入侵,和移民,而SLC25A21-AS1沉默表现出相反的效果。钾通道亚家族K成员4(KCNK4)在SLC25A21-AS1强制表达时显著上调。过表达KCNK4抑制细胞增殖,入侵,迁移能力,并增强卵巢癌细胞对紫杉醇和顺铂的敏感性。同时,KNCK4过表达挽救了SLC25A21-AS1沉默对细胞增殖的促进作用,入侵和迁移。此外,SLC25A21-AS1可与Zeste同源物2(EZH2)的转录因子增强子相互作用,而EZH2敲除增加了一些卵巢癌细胞系中KCNK4的表达。SLC25A21-AS1增强化疗敏感性,抑制增殖,迁移,和卵巢癌细胞的侵袭能力至少部分通过阻断EZH2介导的KCNK4沉默。
    Owing to high mortality rate, ovarian cancer seriously threatens women\'s health. Extensive abdominal metastasis and chemoresistance are the leading causes of ovarian cancer deaths. Through lncRNA sequencing, our previous study identified lncRNA SLC25A21-AS1, which was significantly downregulated in chemoresistant ovarian cancer cells. In this study, we aimed to evaluate the role and mechanism of SLC25A21-AS1 in ovarian cancer. The expression of SLC25A21-AS1 was analyzed by qRT-PCR and online database GEPIA. The biological functions of SLC25A21-AS1 and KCNK4 were analyzed by CCK-8, transwell, and flow cytometry. The specific mechanism was analyzed by RNA-sequencing, RNA binding protein immunoprecipitation, rescue experiments, and bioinformatic analysis. SLC25A21-AS1 was decreased in ovarian cancer tissues and cell lines. Overexpression of SLC25A21-AS1 enhanced the sensitivity of ovarian cancer cells to paclitaxel and cisplatin, and inhibited cell proliferation, invasion, and migration, while SLC25A21-AS1-silencing showed the opposite effect. Potassium channel subfamily K member 4 (KCNK4) was significantly up-regulated upon enforced expression of SLC25A21-AS1. Overexpression of KCNK4 inhibited cell proliferation, invasion, migration ability, and enhanced the sensitivity of ovarian cancer cells to paclitaxel and cisplatin. Meanwhile, KNCK4-overexpression rescued the promotive effect of SLC25A21-AS1-silencing on cell proliferation, invasion and migration. In addition, SLC25A21-AS1 could interact with the transcription factor Enhancer of Zeste Homolog 2 (EZH2), while EZH2 knockdown increased the expression of KCNK4 in some of the ovarian cancer cell lines. SLC25A21-AS1 enhanced the chemosensitivity and inhibited the proliferation, migration, and invasion ability of ovarian cancer cells at least partially by blocking EZH2-mediated silencing of KCNK4.
    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
    KCNK4基因,主要分布在神经元中,在控制静息膜电位和调节细胞兴奋性方面发挥着重要作用。以前,只有两种变异被鉴定为与人类疾病相关,面部畸形,多毛症,癫痫,智力/发育迟缓,牙龈过度生长(FHEIG)综合征。在这项研究中,我们在一组癫痫患者中进行了基于三重的全外显子测序(WES).在两个具有异质表型的无关病例中发现了两个从头可能的致病变异,包括Rolandic癫痫和FHEIG综合征。预测这两种变体被大多数的计算机模拟算法破坏。这些变体在对照中未显示等位基因频率,并且在病例队列中呈现统计学上高于对照的频率。FHEIG综合征相关变异体均位于具有稳定导电构象的重要功能的区域,而Rolandic癫痫相关变异体分布在对导电构象影响较小的区域。本研讨扩大了KCNK4的遗传和表型谱。KCNK4的表型变异可能与分子亚区域效应有关。卡马西平/奥卡西平和丙戊酸钠可能是治疗KCNK4变异患者的有效抗癫痫药物。
    The KCNK4 gene, predominantly distributed in neurons, plays an essential role in controlling the resting membrane potential and regulating cellular excitability. Previously, only two variants were identified to be associated with human disease, facial dysmorphism, hypertrichosis, epilepsy, intellectual/developmental delay, and gingival overgrowth (FHEIG) syndrome. In this study, we performed trio-based whole exon sequencing (WES) in a cohort of patients with epilepsy. Two de novo likely pathogenic variants were identified in two unrelated cases with heterogeneous phenotypes, including one with Rolandic epilepsy and one with the FHEIG syndrome. The two variants were predicted to be damaged by the majority of in silico algorithms. These variants showed no allele frequencies in controls and presented statistically higher frequencies in the case cohort than that in controls. The FHEIG syndrome-related variants were all located in the region with vital functions in stabilizing the conductive conformation, while the Rolandic epilepsy-related variant was distributed in the area with less impact on the conductive conformation. This study expanded the genetic and phenotypic spectrum of KCNK4. Phenotypic variations of KCNK4 are potentially associated with the molecular sub-regional effects. Carbamazepine/oxcarbazepine and valproate may be effective antiepileptic drugs for patients with KCNK4 variants.
    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
    TRAAK (KCNK4, K2P4.1) is a mechanosensitive two-pore domain potassium (K2P) channel. Due to its expression within sensory neurons and genetic link to neuropathic pain it represents a promising potential target for novel analgesics. In common with many other channels in the wider K2P sub-family, there remains a paucity of small molecule pharmacological tools. Specifically, there is a lack of molecules selective for TRAAK over the other members of the TREK subfamily of K2P channels. We developed a thallium flux assay to allow high throughput screening of compounds and facilitate the identification of novel TRAAK activators. Using a library of ∼1200 drug like molecules we identified Aprepitant as a small molecule activator of TRAAK. Aprepitant is an NK-1 antagonist used to treat nausea and vomiting. Close structural analogues of Aprepitant and a range of NK-1 antagonists were also selected or designed for purchase or brief chemical synthesis and screened for their ability to activate TRAAK. Electrophysiology experiments confirmed that Aprepitant activates both the \'long\' and \'short\' transcript variants of TRAAK. We also demonstrated that Aprepitant is selective and does not activate other members of the K2P superfamily. This work describes the development of a high throughput assay to identify potential TRAAK activators and subsequent identification and confirmation of the novel TRAAK activator Aprepitant. This discovery identifies a useful tool compound which can be used to further probe the function of TRAAK K2P channels.
    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
    The two-pore domain K2P subunits form background (leak) potassium channels, which are characterized by constitutive, although not necessarily constant activity, at all membrane potential values. Among the fifteen pore-forming K2P subunits encoded by the KCNK genes, the three members of the TREK subfamily, TREK-1, TREK-2, and TRAAK are mechanosensitive ion channels. Mechanically induced opening of these channels generally results in outward K+ current under physiological conditions, with consequent hyperpolarization and inhibition of membrane potential-dependent cellular functions. In the past decade, great advances have been made in the investigation of the molecular determinants of mechanosensation, and members of the TREK subfamily have emerged among the best-understood examples of mammalian ion channels directly influenced by the tension of the phospholipid bilayer. In parallel, the crucial contribution of mechano-gated TREK channels to the regulation of membrane potential in several cell types has been reported. In this review, we summarize the general principles underlying the mechanical activation of K2P channels, and focus on the physiological roles of mechanically induced hyperpolarization.
    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 mechanosensitive two-pore domain (K2P) K+ channels (TREK-1, TREK-2, and TRAAK) are important for mechanical and thermal nociception. However, the mechanisms underlying their gating by membrane stretch remain controversial. Here we use molecular dynamics simulations to examine their behavior in a lipid bilayer. We show that TREK-2 moves from the \"down\" to \"up\" conformation in direct response to membrane stretch, and examine the role of the transmembrane pressure profile in this process. Furthermore, we show how state-dependent interactions with lipids affect the movement of TREK-2, and how stretch influences both the inner pore and selectivity filter. Finally, we present functional studies that demonstrate why direct pore block by lipid tails does not represent the principal mechanism of mechanogating. Overall, this study provides a dynamic structural insight into K2P channel mechanosensitivity and illustrates how the structure of a eukaryotic mechanosensitive ion channel responds to changes in forces within the bilayer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号