Calcium-activated potassium channel

钙激活钾通道
  • 文章类型: Journal Article
    自闭症谱系障碍(ASD)是一种复杂的神经发育障碍,其特征是社会交往和社会交往受损。限制和重复的行为,和利益。ASD的核心症状与从腹侧被盖区(VTA)投射到伏隔核(NAc)和内侧前额叶皮层(mPFC)的中皮层边缘多巴胺途径的缺陷有关。AST-001是目前正在进行3期临床试验的研究产品,用于治疗ASD的核心症状,以L-丝氨酸作为API(活性药物成分)。因为ASD的病因非常不同,单个遗传ASD模型不能代表所有自闭症模型。在本文中,我们使用了VPA暴露模型,比单个遗传模型更普遍和广泛使用,但这也是自闭症的动物模型之一。在这里,我们进行了实验,以证明AST-001作为L-丝氨酸的功效,通过抑制小电导Ca2激活的K通道(SK通道)来改变多巴胺神经元放电率的调节。通过这些行动,AST-001通过挽救暴露于VPA的ASD小鼠模型中多巴胺神经元的内在兴奋性来改善社交能力和社会新颖性,该模型显示出与ASD相关的行为异常。据认为,在暴露于VPA的ASD小鼠模型中改善社会缺陷的这种作用是由于AST-001使异常的SK通道活动正常化,从而减慢了VTA多巴胺神经元的放电。总的来说,这些结果表明,AST-001可能是ASD患者的潜在治疗剂,其作用机制可能涉及多巴胺神经元活性的调节和社会交往的改善。
    Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by impaired social communication and social interaction, restricted and repetitive behavior, and interests. The core symptoms of ASD are associated with deficits in mesocorticolimbic dopamine pathways that project from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC). AST-001 is an investigational product currently in a phase 3 clinical trial for treating the core symptoms of ASD, with L-serine as the API (active pharmaceutical ingredient). Because the causes of ASD are extremely heterogeneous, a single genetic ASD model cannot represent all autism models. In this paper, we used the VPA-exposed model, which is more general and widely used than a single genetic model, but this is also one of the animal models of autism. Herein, we conducted experiments to demonstrate the efficacy of AST-001 as L-Serine that alters the regulation of the firing rate in dopamine neurons by inhibiting small conductance Ca2+-activated K+ channels (SK channels). Through these actions, AST-001 improved sociability and social novelty by rescuing the intrinsic excitabilities of dopamine neurons in VPA-exposed ASD mouse models that showed ASD-related behavioral abnormalities. It is thought that this effect of improving social deficits in VPA-exposed ASD mouse models is due to AST-001 normalizing aberrant SK channel activities that slowed VTA dopamine neuron firing. Overall, these findings suggest that AST-001 may be a potential therapeutic agent for ASD patients, and that its mechanism of action may involve the regulation of dopamine neuron activity and the improvement of social interaction.
    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
    KCNMA1相关的信道病是一种以癫痫发作为特征的神经系统疾病,电机异常,和神经发育障碍。预测疾病机制是由KCNMA1编码的BKK通道活性的改变引起的;然而,仅对一部分患者相关变异进行了功能研究.尚未系统地评估这些变体在三级结构内的定位或通过致病性算法进行的评估。在这项研究中,在BK通道蛋白内定位了82种非同义患者相关的KCNMA1变体。位于cryo-EM分辨结构内的53个变体,包括21个在BK通道活动中被分类为功能获得(GOF)或功能丧失(LOF)。在孔中鉴定了LOF变体的簇,交流区域(RCK1),和附近的Ca2+碗(RCK2),与药理学或内源性调节位点重叠。然而,未发现GOF变异的聚类.为了进一步理解不确定意义(VUS)的变体,比较了多种标准致病性算法的评估结果,并从证实的GOF和LOF变异中建立了新的敏感性和特异性阈值.构建了集成算法(KCNMA1MetaScore),由这个训练的数据集的加权总和以及从Ca2+结合和未结合的BK通道导出的结构分量组成。KMS评估与10个VUS残基处性能最高的个体算法(REVEL)不同,通过电生理学在HEK293细胞中进一步研究了一个子集。M578T,E656A,和D965V(KMS+;REVEL-)被证实会改变电压钳记录中的BK通道特性,和D800Y(KMS-;REVEL+)在测试条件下被评估为良性的。然而,KMS未能准确评估K457E。这些综合结果揭示了BK通道功能域内潜在致病KCNMA1变体的分布和VUS的致病性评估,通过构建KMS等集成算法,提出在未来研究中改进信道级预测的策略。
    KCNMA1-linked channelopathy is a neurological disorder characterized by seizures, motor abnormalities, and neurodevelopmental disabilities. The disease mechanisms are predicted to result from alterations in KCNMA1-encoded BK K+ channel activity; however, only a subset of the patient-associated variants have been functionally studied. The localization of these variants within the tertiary structure or evaluation by pathogenicity algorithms has not been systematically assessed. In this study, 82 nonsynonymous patient-associated KCNMA1 variants were mapped within the BK channel protein. Fifty-three variants localized within cryoelectron microscopy-resolved structures, including 21 classified as either gain of function (GOF) or loss of function (LOF) in BK channel activity. Clusters of LOF variants were identified in the pore, the AC region (RCK1), and near the Ca2+ bowl (RCK2), overlapping with sites of pharmacological or endogenous modulation. However, no clustering was found for GOF variants. To further understand variants of uncertain significance (VUSs), assessments by multiple standard pathogenicity algorithms were compared, and new thresholds for sensitivity and specificity were established from confirmed GOF and LOF variants. An ensemble algorithm was constructed (KCNMA1 meta score (KMS)), consisting of a weighted summation of this trained dataset combined with a structural component derived from the Ca2+-bound and unbound BK channels. KMS assessment differed from the highest-performing individual algorithm (REVEL) at 10 VUS residues, and a subset were studied further by electrophysiology in HEK293 cells. M578T, E656A, and D965V (KMS+;REVEL-) were confirmed to alter BK channel properties in voltage-clamp recordings, and D800Y (KMS-;REVEL+) was assessed as benign under the test conditions. However, KMS failed to accurately assess K457E. These combined results reveal the distribution of potentially disease-causing KCNMA1 variants within BK channel functional domains and pathogenicity evaluation for VUSs, suggesting strategies for improving channel-level predictions in future studies by building on ensemble algorithms such as KMS.
    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
    表达小白蛋白的中间神经元(PVIN)通过防止触摸输入激活疼痛回路在脊髓背角中起关键作用。在雄性和雌性小鼠中,神经损伤通过尚未完全了解的机制减少PVIN的输出。在这项研究中,我们表明,来自神经损伤的雄性小鼠的PVIN改变了它们的放电模式,从补品到适应性。为了检查导致产量下降的离子机制,我们采用了重新参数化的Hodgkin-Huxley(HH)型PVIN模型,预测(1)点火模式转变是由于小电导钙激活钾(SK)通道的贡献增加,通过(2)细胞内钙缓冲系统的损伤。分析HH型模型的动力学进一步表明,广义Hopf分叉区分了PVIN瞬态激发中观察到的两种状态转变。重要的是,当我们将PVINs模型嵌入脊髓背角的神经元回路模型中时,这种预测的机制成立.为了实验验证这个假设的机制,我们使用了SK通道的药理学调节剂,并证明(1)当暴露于SK通道激活剂时,来自幼稚雄性小鼠的强直放电PVIN变得适应性。和(2)适应神经损伤的雄性小鼠的PVIN在SK通道阻断后恢复到强直放电。我们的工作为神经损伤后脊髓背角PVIN输出减少的细胞机制提供了重要见解,并强调了神经性疼痛新的有效治疗方法的潜在药理靶点。重要声明:表达小白蛋白的中间神经元(PVIN)对脊髓背角的Aβ纤维介导的伤害性途径具有关键的抑制控制作用。它们抑制音调的丧失会导致神经性症状,比如机械性异常性疼痛,通过尚未完全理解的机制。这项研究确定了PVIN的内在兴奋性降低是其在神经损伤状况下抑制输出降低的潜在原因。结合计算和实验方法,我们预测在神经损伤后调节PVINs电活动的钙依赖性机制:胞浆钙缓冲液的消耗允许细胞内钙通过活性膜的快速积累,这反过来增强SK通道并阻碍尖峰产生。因此,我们的结果将SK通道确定为治疗神经性症状的潜在治疗靶标。
    Parvalbumin-expressing interneurons (PVINs) play a crucial role within the dorsal horn of the spinal cord by preventing touch inputs from activating pain circuits. In both male and female mice, nerve injury decreases PVINs\' output via mechanisms that are not fully understood. In this study, we show that PVINs from nerve-injured male mice change their firing pattern from tonic to adaptive. To examine the ionic mechanisms responsible for this decreased output, we used a reparametrized Hodgkin-Huxley type model of PVINs, which predicted (1) the firing pattern transition is because of an increased contribution of small conductance calcium-activated potassium (SK) channels, enabled by (2) impairment in intracellular calcium buffering systems. Analyzing the dynamics of the Hodgkin-Huxley type model further demonstrated that a generalized Hopf bifurcation differentiates the two types of state transitions observed in the transient firing of PVINs. Importantly, this predicted mechanism holds true when we embed the PVIN model within the neuronal circuit model of the spinal dorsal horn. To experimentally validate this hypothesized mechanism, we used pharmacological modulators of SK channels and demonstrated that (1) tonic firing PVINs from naive male mice become adaptive when exposed to an SK channel activator, and (2) adapting PVINs from nerve-injured male mice return to tonic firing on SK channel blockade. Our work provides important insights into the cellular mechanism underlying the decreased output of PVINs in the spinal dorsal horn after nerve injury and highlights potential pharmacological targets for new and effective treatment approaches to neuropathic pain.SIGNIFICANCE STATEMENT Parvalbumin-expressing interneurons (PVINs) exert crucial inhibitory control over Aβ fiber-mediated nociceptive pathways at the spinal dorsal horn. The loss of their inhibitory tone leads to neuropathic symptoms, such as mechanical allodynia, via mechanisms that are not fully understood. This study identifies the reduced intrinsic excitability of PVINs as a potential cause for their decreased inhibitory output in nerve-injured condition. Combining computational and experimental approaches, we predict a calcium-dependent mechanism that modulates PVINs\' electrical activity following nerve injury: a depletion of cytosolic calcium buffer allows for the rapid accumulation of intracellular calcium through the active membranes, which in turn potentiates SK channels and impedes spike generation. Our results therefore pinpoint SK channels as potential therapeutic targets for treating neuropathic symptoms.
    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
    BKK+通道是神经元和肌肉兴奋性的关键调节因子,由来自KCNMA1基因的成孔α亚基和细胞和组织选择性β亚基(KCNMB1-4)的四聚体组成。KCNMA1的突变与神经系统疾病有关,包括自闭症。然而,关于神经元BK通道β亚基在人类神经病理学中的作用知之甚少。β2亚基在中枢神经元中表达,并使BK通道失活,以及改变激活和去激活门控。在这项研究中,我们报道了G124R的功能效应,从1例自闭症谱系障碍患者的全外显子组测序中获得的一种新的KCNMB2突变.残基G124位于TM1和TM2之间的胞外环中,在物种之间是保守的,G124R错义突变是用计算工具预测的。为了研究致病性潜力,BK通道在HEK293T细胞中与β2WT和β2G124R亚基共表达。在激活和失活过程中(电压依赖性和动力学),在生理K条件(140/6mMK和10μMCa2)下,从内向外的贴片评估BK/β2电流。使用缺乏失活的β2亚基(β2IR)表明,与来自BK/β2IRWT通道的电流相比,来自BK/β2IRG124R通道的电流激活快2倍,失活慢2倍,激活的电压依赖性没有变化(V1/2)。尽管BK通道的打开和关闭发生了变化,BK/β2G124R失活率(τinact和τrecovery),和失活的V1/2,在标准稳态电压方案下,与BK/β2WT通道相比没有改变。动作电位诱发的电流也没有变化。因此,突变表型表明β2G124RTM1-TM2胞外环可以调节BK通道的激活和失活动力学。然而,需要更多的证据来验证KCNMB2中这种患者相关变异体的致病性.
    BK K+ channels are critical regulators of neuron and muscle excitability, comprised of a tetramer of pore-forming αsubunits from the KCNMA1 gene and cell- and tissue-selective β subunits (KCNMB1-4). Mutations in KCNMA1 are associated with neurological disorders, including autism. However, little is known about the role of neuronal BK channel β subunits in human neuropathology. The β2 subunit is expressed in central neurons and imparts inactivation to BK channels, as well as altering activation and deactivation gating. In this study, we report the functional effect of G124R, a novel KCNMB2 mutation obtained from whole-exome sequencing of a patient diagnosed with autism spectrum disorder. Residue G124, located in the extracellular loop between TM1 and TM2, is conserved across species, and the G124R missense mutation is predicted deleterious with computational tools. To investigate the pathogenicity potential, BK channels were co-expressed with β2WT and β2G124R subunits in HEK293T cells. BK/β2 currents were assessed from inside-out patches under physiological K+ conditions (140/6 mM K+ and 10 μM Ca2+) during activation and inactivation (voltage-dependence and kinetics). Using β2 subunits lacking inactivation (β2IR) revealed that currents from BK/β2IRG124R channels activated 2-fold faster and deactivated 2-fold slower compared with currents from BK/β2IRWT channels, with no change in the voltage-dependence of activation (V1/2). Despite the changes in the BK channel opening and closing, BK/β2G124R inactivation rates (τinact and τrecovery), and the V1/2 of inactivation, were unaltered compared with BK/β2WT channels under standard steady-state voltage protocols. Action potential-evoked current was also unchanged. Thus, the mutant phenotype suggests the β2G124R TM1-TM2 extracellular loop could regulate BK channel activation and deactivation kinetics. However, additional evidence is needed to validate pathogenicity for this patient-associated variant in KCNMB2.
    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
    Apamin通常被认为是选择性作用于小电导Ca2激活的钾通道(KCa2)的少数物质之一。然而,已发表的药理学和结构数据仍然存在争议。这里,我们通过双电极电压钳在非洲爪的卵母细胞中和膜片钳在表达所研究的离子通道的HEK293,COS7和CHO细胞中研究了阿帕明的分子药理学,以及孤立的大鼠脑神经元。微量滴定肉汤稀释法用于抗菌活性筛选。通过NMR光谱确定了水溶液中阿帕明的空间结构。我们测试了阿帕明对42个离子通道(KCa,KV,NaV,nAChR,ASIC,和其他),并证实了其对KCa2通道的独特选择性。没有检测到阿帕明对革兰氏阳性或革兰氏阴性细菌的抗微生物活性。将阿帕明的NMR溶液结构保存在蛋白质数据库中。本文呈现的结果表明,阿帕明是选择性纳摩尔或甚至亚纳摩尔亲和力KCa2抑制剂,对其他分子靶标没有显著影响。本文提供的空间结构以及充足的功能数据支持使用阿帕明作为KCa2选择性药理学工具和作为药物设计的模板。
    Apamin is often cited as one of the few substances selectively acting on small-conductance Ca2+-activated potassium channels (KCa2). However, published pharmacological and structural data remain controversial. Here, we investigated the molecular pharmacology of apamin by two-electrode voltage-clamp in Xenopus laevis oocytes and patch-clamp in HEK293, COS7, and CHO cells expressing the studied ion channels, as well as in isolated rat brain neurons. The microtitre broth dilution method was used for antimicrobial activity screening. The spatial structure of apamin in aqueous solution was determined by NMR spectroscopy. We tested apamin against 42 ion channels (KCa, KV, NaV, nAChR, ASIC, and others) and confirmed its unique selectivity to KCa2 channels. No antimicrobial activity was detected for apamin against Gram-positive or Gram-negative bacteria. The NMR solution structure of apamin was deposited in the Protein Data Bank. The results presented here demonstrate that apamin is a selective nanomolar or even subnanomolar-affinity KCa2 inhibitor with no significant effects on other molecular targets. The spatial structure as well as ample functional data provided here support the use of apamin as a KCa2-selective pharmacological tool and as a template for drug design.
    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
    KCNMA1形成BK+通道的孔隙,调节神经元和肌肉的兴奋性。最近,遗传筛查在一组衰弱性阵发性非运动源性运动障碍患者中鉴定出杂合的KCNMA1变体,有或没有癫痫(PNKD3)。然而,KCNMA1突变的相关性和PNKD3临床异质性的基础尚未确定.这里,我们评估了BK通道中三种KCNMA1患者变异的相对严重程度,神经元,和老鼠。在异源细胞中,BKN999S和BKD434G通道显示功能增益(GOF)属性,而BKH444Q通道显示功能丧失(LOF)特性。通道活性的相对程度为BKN999S>BKD434G>WT>BKH444Q。BK电流和动作电位放电增加,癫痫发作阈值降低,在Kcnma1N999S/WT和Kcnma1D434G/WT转基因小鼠中,但不在Kcnma1H444Q/WT小鼠中。在一项新的阵发性运动障碍行为测试中,受影响更严重的Kcnma1N999S/WT小鼠在应激后变得不动。急性右旋苯丙胺治疗消除了这种情况,与受PNKD3影响的个体一致。纯合子Kcnma1D434G/D434G小鼠表现出相似的不动,但相比之下,纯合Kcnma1H444Q/H444Q小鼠表现出运动过度行为。这些数据建立了患者等位基因的相对致病潜力为N999S>D434G>H444Q,并验证了Kcnma1N999S/WT小鼠作为癫痫发作倾向增加的PNKD3模型。
    到目前为止,全球仅有70例患者被诊断出患有一种新发现的罕见综合征,即KCNMA1相关信道病.这种情况的特征是癫痫发作和异常运动,包括频繁的“跌落攻击”,肌肉控制的突然和衰弱的丧失,导致患者在没有警告的情况下跌倒。该疾病与KCNMA1基因的突变有关,KCNMA1是一类对控制神经细胞活性和脑功能重要的蛋白质的成员。然而,由于受影响的人数有限,很难将特定的突变与观察到的症状联系起来;因此,下降攻击的基础仍然未知。Park等人。在实验室中开始“模型”KCNMA1链接的信道作用,为了确定KCNMA1基因中的哪些突变引起了这些症状。三组小鼠均经过基因工程改造,以携带KCNMA1基因中两个最常见的突变之一,或与运动症状相关的非常罕见的突变。行为实验和神经细胞活性的研究表明,携带突变使KCNMA1蛋白更活跃的小鼠更容易发生癫痫发作,并被固定,显示鼠标版本的drop攻击。给这些老鼠服用右旋苯丙胺,对一些人类患者有效,完全停止了固定的攻击。这些结果首次显示了哪些特定的遗传变化引起了KCNMA1连锁信道病的主要症状。Park等人。希望这些知识将加深我们对这种疾病的理解,并帮助开发更好的治疗方法。
    KCNMA1 forms the pore of BK K+ channels, which regulate neuronal and muscle excitability. Recently, genetic screening identified heterozygous KCNMA1 variants in a subset of patients with debilitating paroxysmal non-kinesigenic dyskinesia, presenting with or without epilepsy (PNKD3). However, the relevance of KCNMA1 mutations and the basis for clinical heterogeneity in PNKD3 has not been established. Here, we evaluate the relative severity of three KCNMA1 patient variants in BK channels, neurons, and mice. In heterologous cells, BKN999S and BKD434G channels displayed gain-of-function (GOF) properties, whereas BKH444Q channels showed loss-of-function (LOF) properties. The relative degree of channel activity was BKN999S > BKD434G>WT > BKH444Q. BK currents and action potential firing were increased, and seizure thresholds decreased, in Kcnma1N999S/WT and Kcnma1D434G/WT transgenic mice but not Kcnma1H444Q/WT mice. In a novel behavioral test for paroxysmal dyskinesia, the more severely affected Kcnma1N999S/WT mice became immobile after stress. This was abrogated by acute dextroamphetamine treatment, consistent with PNKD3-affected individuals. Homozygous Kcnma1D434G/D434G mice showed similar immobility, but in contrast, homozygous Kcnma1H444Q/H444Q mice displayed hyperkinetic behavior. These data establish the relative pathogenic potential of patient alleles as N999S>D434G>H444Q and validate Kcnma1N999S/WT mice as a model for PNKD3 with increased seizure propensity.
    So far, only 70 patients around the world have been diagnosed with a newly identified rare syndrome known as KCNMA1-linked channelopathy. The condition is characterised by seizures and abnormal movements which include frequent ‘drop attacks’, a sudden and debilitating loss of muscle control that causes patients to fall without warning. The disease is associated with mutations in the gene for KCNMA1, a member of a class of proteins important for controlling nerve cell activity and brain function. However, due to the limited number of people affected by the condition, it is difficult to link a particular mutation to the observed symptoms; the basis for the drop attacks therefore remains unknown. Park et al. set out to ‘model’ KCNMA1-linked channelopathy in the laboratory, in order to determine which mutations in the KCNMA1 gene caused these symptoms. Three groups of mice were each genetically engineered to carry either one of the two most common mutations in the gene for KCNMA1, or a very rare mutation associated with the movement symptoms. Behavioural experiments and studies of nerve cell activity revealed that the mice carrying mutations that made the KCNMA1 protein more active developed seizures more easily and became immobilized, showing the mouse version of drop attacks. Giving these mice the drug dextroamphetamine, which works in some human patients, stopped the immobilizing attacks altogether. These results show for the first time which specific genetic changes cause the main symptoms of KCNMA1-linked channelopathy. Park et al. hope that this knowledge will deepen our understanding of this disease and help develop better treatments.
    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
    钙信号在各种细胞过程的调节中起着至关重要的作用,包括激活,扩散,和T淋巴细胞的分化,由ORAI1和钾(K+)通道介导。这些通道也已被确定为免疫相关疾病的极具吸引力的治疗靶标。LicochalconeA是一种甘草衍生的查耳酮,以其在药物治疗中的多方面有益作用而闻名。包括它的消炎药,抗哮喘,抗氧化剂,抗菌,和抗肿瘤特性。然而,其涉及淋巴细胞离子通道的抗炎作用尚不清楚.因此,本研究旨在研究甘草查尔酮A是否抑制T淋巴细胞的ORAI1和K+通道。我们的结果表明,甘草查尔酮A在浓度依赖性物质中抑制了所有三个通道(ORAI1,Kv1.3和KCa3.1),IC50值为2.97±1.217µM,0.83±1.222µM,和11.21±1.07µM,分别。值得注意的是,LichochalconeA对CD3和CD28抗体诱导的T细胞中IL-2的分泌和增殖具有抑制作用。这些结果表明,甘草查尔酮A的使用可能为炎症相关的免疫疾病提供有效的治疗策略。
    Calcium signaling plays a vital role in the regulation of various cellular processes, including activation, proliferation, and differentiation of T-lymphocytes, which is mediated by ORAI1 and potassium (K+) channels. These channels have also been identified as highly attractive therapeutic targets for immune-related diseases. Licochalcone A is a licorice-derived chalconoid known for its multifaceted beneficial effects in pharmacological treatments, including its anti-inflammatory, anti-asthmatic, antioxidant, antimicrobial, and antitumorigenic properties. However, its anti-inflammatory effects involving ion channels in lymphocytes remain unclear. Thus, the present study aimed to investigate whether licochalcone A inhibits ORAI1 and K+ channels in T-lymphocytes. Our results indicated that licochalcone A suppressed all three channels (ORAI1, Kv1.3, and KCa3.1) in a concentration-dependent matter, with IC50 values of 2.97 ± 1.217 µM, 0.83 ± 1.222 µM, and 11.21 ± 1.07 µM, respectively. Of note, licochalcone A exerted its suppressive effects on the IL-2 secretion and proliferation in CD3 and CD28 antibody-induced T-cells. These results indicate that the use of licochalcone A may provide an effective treatment strategy for inflammation-related immune diseases.
    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
    KCNMA1相关的信道病是一种新兴的神经系统疾病,其特征是运动障碍的异质性和重叠组合,癫痫发作,发育迟缓,智力残疾。KCNMA1编码BK+通道,这有助于兴奋性和抑制性神经元和肌肉活动。了解疾病的基础是积极调查的重要领域;然而,罕见的患病率阻碍了建立基因型-表型相关性所需的大型患者队列的发展.在这次审查中,我们总结了目前69例患者的37个KCNMA1等位基因,并评估了关键的诊断和临床标志.目前,3个变体被分类为关于BK通道活动的功能增益,14功能丧失,15个不确定意义的变体,和假定的良性/VUS。与这些变异相关的症状从患者提供的信息和先前的出版物中进行策划,以定义临床表型的谱。在这个新扩展的队列中,癫痫发作在携带GOF和LOF变异的患者之间没有差异分布,而运动障碍按突变类型分开。阵发性非运动源性运动障碍主要在具有BK通道的GOF等位基因的患者中观察到,虽然不完全如此,而在LOF变异的患者中观察到其他运动障碍。神经发育和脑结构异常在LOF突变患者中普遍存在。与突变相反,疾病相关的KCNMA1单核苷酸多态性并不主要与神经系统表型相关,但涵盖了更广泛的外周生理功能.一起,这篇综述提供了探索KCNMA1相关信道病的遗传和生化基础的更多证据,并总结了多种KCNMA1基因变异类型的患者症状的临床储存库.
    KCNMA1-linked channelopathy is an emerging neurological disorder characterized by heterogeneous and overlapping combinations of movement disorder, seizure, developmental delay, and intellectual disability. KCNMA1 encodes the BK K+ channel, which contributes to both excitatory and inhibitory neuronal and muscle activity. Understanding the basis of the disorder is an important area of active investigation; however, the rare prevalence has hampered the development of large patient cohorts necessary to establish genotype-phenotype correlations. In this review, we summarize 37 KCNMA1 alleles from 69 patients currently defining the channelopathy and assess key diagnostic and clinical hallmarks. At present, 3 variants are classified as gain-of-function with respect to BK channel activity, 14 loss-of-function, 15 variants of uncertain significance, and putative benign/VUS. Symptoms associated with these variants were curated from patient-provided information and prior publications to define the spectrum of clinical phenotypes. In this newly expanded cohort, seizures showed no differential distribution between patients harboring GOF and LOF variants, while movement disorders segregated by mutation type. Paroxysmal non-kinesigenic dyskinesia was predominantly observed among patients with GOF alleles of the BK channel, although not exclusively so, while additional movement disorders were observed in patients with LOF variants. Neurodevelopmental and structural brain abnormalities were prevalent in patients with LOF mutations. In contrast to mutations, disease-associated KCNMA1 single nucleotide polymorphisms were not predominantly related to neurological phenotypes but covered a wider set of peripheral physiological functions. Together, this review provides additional evidence exploring the genetic and biochemical basis for KCNMA1-linked channelopathy and summarizes the clinical repository of patient symptoms across multiple types of KCNMA1 gene variants.
    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
    Carbon monoxide (CO) is a cardioprotectant and potential cardiovascular therapeutic agent. Human cardiac fibroblasts (HCFs) are important determinants of myocardial structure and function. Large-conductance Ca2+-activated K+ (BK) channel is a potential therapeutic target for cardiovascular disease. We investigated whether CO modulates BK channels and the signaling pathways in HCFs using whole-cell mode patch-clamp recordings. CO-releasing molecules (CORMs; CORM-2 and CORM-3) significantly increased the amplitudes of BK currents (IBK). The CO-induced stimulating effects on IBK were blocked by pre-treatment with specific nitric oxide synthase (NOS) blockers (L-NG-monomethyl arginine citrate and L-NG-nitroarginine methyl ester). 8-bromo-cyclic GMP increased IBK. KT5823 (inhibits PKG) or ODQ (inhibits soluble guanylate cyclase) blocked the CO-stimulating effect on IBK. Moreover, 8-bromo-cyclic AMP also increased IBK, and pre-treatment with KT5720 (inhibits PKA) or SQ22536 (inhibits adenylate cyclase) blocked the CO effect. Pre-treatment with Nethylmaleimide (a thiol-alkylating reagent) also blocked the CO effect on IBK, and DLdithiothreitol (a reducing agent) reversed the CO effect. These data suggest that CO activates IBK through NO via the NOS and through the PKG, PKA, and S-nitrosylation pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    The diagnosis of a paroxysmal dyskinesia is difficult and status dystonicus is a rare life threatening movement disorder characterised by severe, frequent or continuous episodes of dystonic spasms. A 25 year old woman with chronic ataxia and paroxysmal dyskinesia presented with facial twitching, writhing of arms, oculogyric crisis and visual and auditory hallucinations. She developed respiratory failure and was ventilated. No cause was found so whole exome sequencing was performed and this revealed a novel, non-synonymous heterozygous variant in exon 11 of the KCNMA1 gene, K457E (c 1369A>G) in the patient but not her parents. This variant has not been previously reported in gnomAD or ClinVar. The finding of a de novo variant in a potassium channel gene guided a trial of the potassium channel antagonist 3,4 diaminopyridine resulting in significant improvement, discharge from the intensive care unit and ultimately home.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号