KCa1.1

KCa1.1
  • 文章类型: Journal Article
    KCNMA1中的变体,编码电压和钙激活的K(BK)通道,与人类神经系统疾病相关。主要研究了在稳态电压和Ca2条件下诱发的BK通道电流的功能增益(GOF)和功能损失(LOF)变体的影响。然而,在他们的生理环境中,BK通道与电压门控Ca2通道共同存在,并响应细胞内Ca2(Ca2i)的动态变化。在这项研究中,存在于大脑中的L型电压门控Ca2+通道,CaV1.2与含有GOF的野生型和突变型BK通道共表达(D434G,N999S)和LOF(H444Q,D965V)HEK-293T细胞中的患者相关变体。在CaV1.2激活下,使用将Ca2+i限制为纳米或微米域的缓冲条件记录全细胞BK电流。两种条件都允许野生型BK电流激活以响应CaV1.2Ca2流入,但野生型和突变型BK通道之间的行为差异与之前的研究相比减少了。只有N999S突变使用平方电压命令在微域和纳米域中产生BK电流的增加,并且在微域内的神经元动作电位引起的BK电流中也可检测到。这些数据证实了N999S在动态电压和Ca2刺激下对BK通道活性的GOF效应,与其在神经系统疾病中的致病性一致。然而,患者相关突变D434G,H444Q,在CaV1.2介导的Ca2流入下,D965V对BK电流没有显着影响,与以前的稳态方案相反。这些结果证明了在不同电压和Ca2+条件下相比,KCNMA1变体致病性的不同电位。
    Variants in KCNMA1, encoding the voltage- and calcium-activated K+ (BK) channel, are associated with human neurological disease. The effects of gain-of-function (GOF) and loss-of-function (LOF) variants have been predominantly studied on BK channel currents evoked under steady-state voltage and Ca2+ conditions. However, in their physiological context, BK channels exist in partnership with voltage-gated Ca2+ channels and respond to dynamic changes in intracellular Ca2+ (Ca2+i). In this study, an L-type voltage-gated Ca2+ channel present in the brain, CaV1.2, was co-expressed with wild type and mutant BK channels containing GOF (D434G, N999S) and LOF (H444Q, D965V) patient-associated variants in HEK-293T cells. Whole-cell BK currents were recorded under CaV1.2 activation using buffering conditions that restrict Ca2+i to nano- or micro-domains. Both conditions permitted wild type BK current activation in response to CaV1.2 Ca2+ influx, but differences in behavior between wild type and mutant BK channels were reduced compared to prior studies in clamped Ca2+i. Only the N999S mutation produced an increase in BK current in both micro- and nano-domains using square voltage commands and was also detectable in BK current evoked by a neuronal action potential within a microdomain. These data corroborate the GOF effect of N999S on BK channel activity under dynamic voltage and Ca2+ stimuli, consistent with its pathogenicity in neurological disease. However, the patient-associated mutations D434G, H444Q, and D965V did not exhibit significant effects on BK current under CaV1.2-mediated Ca2+ influx, in contrast with prior steady-state protocols. These results demonstrate a differential potential for KCNMA1 variant pathogenicity compared under diverse voltage and Ca2+ conditions.
    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
    含肿瘤抑制基因F-box和WD重复结构域(FBXW)7通过促进多能干细胞标志物的蛋白质降解来降低癌症干性。我们最近通过几种癌细胞的三维(3D)球体形成证明了FBXW7的转录抑制。在本研究中,我们发现FBXW7的转录活性受到Ca2+激活的K+通道的抑制,KCa1.1,在人前列腺癌LNCaP细胞通过Akt-Nrf2信号通路的3D球体模型中。在LNCaP球体模型中转染miR223模拟物后,通过siRNA介导的CCAAT增强子结合蛋白C/EBPδ(CEBPD)的抑制,FBXW7的转录活性降低,提示在LNCaP球体模型中通过Akt-Nrf2-CEBPD-miR223转录轴对FBXW7进行转录调节。此外,KCa1.1抑制诱导的FBXW7的激活降低了(1)KCa1.1活性和质膜中的蛋白质水平,以及(2)癌症干细胞(CSC)标志物的蛋白质水平,c-Myc,在LNCaP球体模型中,它是被FBXW7降解的分子,表明KCa1.1抑制诱导的FBXW7激活抑制了KCa1.1阳性癌细胞中的CSC转化。
    The tumor suppressor gene F-box and WD repeat domain-containing (FBXW) 7 reduces cancer stemness properties by promoting the protein degradation of pluripotent stem cell markers. We recently demonstrated the transcriptional repression of FBXW7 by the three-dimensional (3D) spheroid formation of several cancer cells. In the present study, we found that the transcriptional activity of FBXW7 was promoted by the inhibition of the Ca2+-activated K+ channel, KCa1.1, in a 3D spheroid model of human prostate cancer LNCaP cells through the Akt-Nrf2 signaling pathway. The transcriptional activity of FBXW7 was reduced by the siRNA-mediated inhibition of the CCAAT-enhancer-binding protein C/EBP δ (CEBPD) after the transfection of miR223 mimics in the LNCaP spheroid model, suggesting the transcriptional regulation of FBXW7 through the Akt-Nrf2-CEBPD-miR223 transcriptional axis in the LNCaP spheroid model. Furthermore, the KCa1.1 inhibition-induced activation of FBXW7 reduced (1) KCa1.1 activity and protein levels in the plasma membrane and (2) the protein level of the cancer stem cell (CSC) markers, c-Myc, which is a molecule degraded by FBXW7, in the LNCaP spheroid model, indicating that KCa1.1 inhibition-induced FBXW7 activation suppressed CSC conversion in KCa1.1-positive cancer cells.
    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
    大电导Ca2+激活的K+通道,KCa1.1在癌症进展中起着关键作用,转移,以及化学抗性的获得。先前的研究表明,在人前列腺癌LNCaP的三维球体模型中,通过下调多药耐药相关蛋白,KCa1.1的药理学抑制克服了对多柔比星(DOX)的耐药性,骨肉瘤MG-63和软骨肉瘤SW-1353细胞。最近的研究集中在肿瘤内的关键作用,化学耐药中的药物代谢细胞色素P450酶(CYPs)。在本研究中,我们在癌症球体模型中检查了CYPs参与DOX抗性的获得及其通过抑制KCa1.1克服。在参与DOX代谢的CYP亚型中,CYP3A4通过球状体形成上调,并通过CCAAT/增强子结合蛋白的转录抑制而被KCa1.1的抑制显着抑制,CEBPB,它是Nrf2信号通路的下游转录因子。通过siRNA介导的CYP3A4抑制和用有效的CYP3A4抑制剂治疗克服了DOX抗性,酮康唑,在癌症球体模型中。抑制KCa1.1可显著降低癌球体模型中Akt的磷酸化水平,和KCa1.1诱导的CYP3A4的下调通过用Akt和Nrf2激活剂处理而逆转。总的来说,目前的结果表明,CYP3A4的上调负责在癌症球体模型中获得DOX抗性,KCa1.1的抑制主要通过Akt-Nrf2-CEBPB轴抑制CYP3A4转录来克服DOX抗性。
    The large-conductance Ca2+-activated K+ channel, KCa1.1, plays a pivotal role in cancer progression, metastasis, and the acquisition of chemoresistance. Previous studies indicated that the pharmacological inhibition of KCa1.1 overcame resistance to doxorubicin (DOX) by down-regulating multidrug resistance-associated proteins in the three-dimensional spheroid models of human prostate cancer LNCaP, osteosarcoma MG-63, and chondrosarcoma SW-1353 cells. Investigations have recently focused on the critical roles of intratumoral, drug-metabolizing cytochrome P450 enzymes (CYPs) in chemoresistance. In the present study, we examined the involvement of CYPs in the acquisition of DOX resistance and its overcoming by inhibiting KCa1.1 in cancer spheroid models. Among the CYP isoforms involved in DOX metabolism, CYP3A4 was up-regulated by spheroid formation and significantly suppressed by the inhibition of KCa1.1 through the transcriptional repression of CCAAT/enhancer-binding protein, CEBPB, which is a downstream transcription factor of the Nrf2 signaling pathway. DOX resistance was overcome by the siRNA-mediated inhibition of CYP3A4 and treatment with the potent CYP3A4 inhibitor, ketoconazole, in cancer spheroid models. The phosphorylation levels of Akt were significantly reduced by inhibiting KCa1.1 in cancer spheroid models, and KCa1.1-induced down-regulation of CYP3A4 was reversed by the treatment with Akt and Nrf2 activators. Collectively, the present results indicate that the up-regulation of CYP3A4 is responsible for the acquisition of DOX resistance in cancer spheroid models, and the inhibition of KCa1.1 overcame DOX resistance by repressing CYP3A4 transcription mainly through the Akt-Nrf2-CEBPB axis.
    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: Case Reports
    背景:KCNMA1连锁信道病是一种罕见的运动障碍,于2005年首次报道。KCNMA1相关的通道病的阵发性非运动障碍(PNKD)是具有KCNMA1-N999S突变的患者中最常见的症状。PNKD发作每天发生数百次,发病率显著,治疗选择有限。经常在癫痫的背景下。
    方法:我们报告了6例KCNMA1-N999S变体用右旋氨胺(0.7-1.25mg/kg/天)治疗,右旋苯丙胺的前药.数据是从访谈和图表审查中回顾性收集的。父母报告的每日PNKD发作计数在治疗下减少,范围从10倍减少到完整的分辨率。
    结论:我们的研究结果表明,右旋氨氟胺是治疗PNKD3(KCNMA1相关PNKD)的有效药物。治疗显著减少了使人衰弱的运动障碍发作,没有其他KCNMA1相关症状(如癫痫发作)的挑衅或恶化。
    BACKGROUND: KCNMA1-linked channelopathy is a rare movement disorder first reported in 2005. Paroxysmal non-kinesigenic dyskinesia (PNKD) in KCNMA1-linked channelopathy is the most common symptom in patients harboring the KCNMA1-N999S mutation. PNKD episodes occur up to hundreds of times daily with significant morbidity and limited treatment options, often in the context of epilepsy.
    METHODS: We report 6 cases with the KCNMA1-N999S variant treated with lisdexamfetamine (0.7-1.25 mg/kg/day), a pro-drug of dextroamphetamine. Data were collected retrospectively from interviews and chart review. Parent-reported daily PNKD episode counts were reduced under treatment, ranging from a 10-fold decrease to complete resolution.
    CONCLUSIONS: Our findings suggest that lisdexamfetamine is an effective therapy for PNKD3 (KCNMA1-associated PNKD). Treatment produced dramatic reductions in debilitating dyskinesia episodes, without provocation or exacerbation of other KCNMA1-associated symptoms such as seizures.
    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
    几种类型的K+通道在肿瘤发生中起着至关重要的作用,stemness,侵入性,和癌症的耐药性。具有超低附着表面培养物的人前列腺癌(PC)LNCaP细胞的球样形成诱导了癌症干细胞标志物的上调,比如NANOG,并通过下调E3泛素连接酶来降低Ca2激活的K通道KCa1.1的蛋白质降解,FBXW7,与LNCaP单层相比。因此,KCa1.1激活剂诱导的超极化反应在来自LNCaP球体的分离细胞中更大。KCa1.1的药理抑制作用克服了LNCaP球体对抗雄激素和多柔比星(DOX)的耐药性。LNCaP球状体的形成使雄激素受体(AR)的蛋白表达显着降低,而KCa1.1的抑制则使其逆转。MDM2的药理和遗传抑制可能与PC干细胞中AR蛋白降解有关,揭示了MDM2是LNCaP球体中获得抗雄激素抗性的原因,这被KCa1.1抑制所克服。此外,ABC转运蛋白的多药耐药相关蛋白亚家族成员,MRP5负责获得LNCaP球体中的DOX抗性,这也被KCa1.1抑制所克服。总的来说,目前的结果表明KCa1.1在LNCaP球体中的潜力,模拟PC干细胞,作为克服PC细胞抗雄激素和DOX抗性的治疗靶标。
    Several types of K+ channels play crucial roles in tumorigenicity, stemness, invasiveness, and drug resistance in cancer. Spheroid formation of human prostate cancer (PC) LNCaP cells with ultra-low attachment surface cultureware induced the up-regulation of cancer stem cell markers, such as NANOG, and decreased the protein degradation of the Ca2+-activated K+ channel KCa1.1 by down-regulating the E3 ubiquitin ligase, FBXW7, compared with LNCaP monolayers. Accordingly, KCa1.1 activator-induced hyperpolarizing responses were larger in isolated cells from LNCaP spheroids. The pharmacological inhibition of KCa1.1 overcame the resistance of LNCaP spheroids to antiandrogens and doxorubicin (DOX). The protein expression of androgen receptors (AR) was significantly decreased by LNCaP spheroid formation and reversed by KCa1.1 inhibition. The pharmacological and genetic inhibition of MDM2, which may be related to AR protein degradation in PC stem cells, revealed that MDM2 was responsible for the acquisition of antiandrogen resistance in LNCaP spheroids, which was overcome by KCa1.1 inhibition. Furthermore, a member of the multidrug resistance-associated protein subfamily of ABC transporters, MRP5 was responsible for the acquisition of DOX resistance in LNCaP spheroids, which was also overcome by KCa1.1 inhibition. Collectively, the present results suggest the potential of KCa1.1 in LNCaP spheroids, which mimic PC stem cells, as a therapeutic target for overcoming antiandrogen- and DOX-resistance in PC cells.
    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
    结性神经节中有髓鞘的Ah型压力感受器神经元(BRN)的女性特异性亚群是血压调节的性二态自主神经控制的神经解剖学基础,KCa1.1是调节结节性神经节神经兴奋的关键因素。在这项研究中,我们研究了在存在或不存在雌激素的情况下KCa1.1介导的有髓鞘Ah型BRNs神经兴奋的确切机制。从成年卵巢完整(OVI)或卵巢切除(OVX)雌性大鼠中分离BRN,并通过电生理和荧光鉴定。使用全细胞记录记录动作电位(AP)和钾电流。始终如一,有髓鞘的Ah型BRN显示出特征性的放电模式,并且在OVX后兴奋性显着降低,AP持续时间变窄,复极化更快,这主要是由于艾贝毒素(IbTX)敏感成分上调;G1(雌激素膜受体GPR30,100nM的选择性激动剂)和/或IbTX(100nM)逆转了OVX雌性大鼠的AP波形变化和Ah型重复放电。此外,G15(雌激素膜受体GPR30的选择性拮抗剂,3μM)可以完全阻断G1对重复放电的影响。这些数据表明,通过去除卵巢来缺乏雌激素会上调Ah型BRNs中的KCa1.1通道蛋白,并随后通过雌激素膜受体信号传导增加AP复极化并钝化神经兴奋。有趣的是,这种在电生理上预测的KCa1.1上调通过雌激素治疗消除的平均荧光强度增加得到证实.这些电生理学发现与免疫染色和药理学操作相结合,揭示了KCa1.1在调节神经兴奋中的关键作用,尤其是在有髓鞘的Ah型BRN的女性特异性亚群中,并扩展了我们目前对BP调节的神经控制的性二态性的理解。
    Female-specific subpopulation of myelinated Ah-type baroreceptor neurons (BRNs) in nodose ganglia is the neuroanatomical base of sexual-dimorphic autonomic control of blood pressure regulation, and KCa1.1 is a key player in modulating the neuroexcitation in nodose ganglia. In this study we investigated the exact mechanisms underlying KCa1.1-mediated neuroexcitation of myelinated Ah-type BRNs in the presence or absence of estrogen. BRNs were isolated from adult ovary intact (OVI) or ovariectomized (OVX) female rats, and identified electrophysiologically and fluorescently. Action potential (AP) and potassium currents were recorded using whole-cell recording. Consistently, myelinated Ah-type BRNs displayed a characteristic discharge pattern and significantly reduced excitability after OVX with narrowed AP duration and faster repolarization largely due to an upregulated iberiotoxin (IbTX)-sensitive component; the changes in AP waveform and repetitive discharge of Ah-types from OVX female rats were reversed by G1 (a selective agonist for estrogen membrane receptor GPR30, 100 nM) and/or IbTX (100 nM). In addition, the effect of G1 on repetitive discharge could be completely blocked by G15 (a selective antagonist for estrogen membrane receptor GPR30, 3 μM). These data suggest that estrogen deficiency by removing ovaries upregulates KCa1.1 channel protein in Ah-type BRNs, and subsequently increases AP repolarization and blunts neuroexcitation through estrogen membrane receptor signaling. Intriguingly, this upregulated KCa1.1 predicted electrophysiologically was confirmed by increased mean fluorescent intensity that was abolished by estrogen treatment. These electrophysiological findings combined with immunostaining and pharmacological manipulations reveal the crucial role of KCa1.1 in modulation of neuroexcitation especially in female-specific subpopulation of myelinated Ah-type BRNs and extend our current understanding of sexual dimorphism of neurocontrol of BP regulation.
    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
    BK钙激活的钾通道具有复杂的动力学,因为它们被电压和细胞质钙激活。动作电位过程中BK激活和失活的时机决定了它们在调节点火模式中的功能作用,但难以先验预测。我们使用动作电位钳表征了男女小鼠浦肯野神经元动作电位期间电压依赖性钙电流和BK电流的动力学,使用可在37°C下实现快速电压钳的急性解离神经元。同时具有去极化电压阶跃和动作电位波形,BK电流完全依赖于钙通过电压依赖性钙通道进入。随着电压阶跃,BK电流大大超过触发钙电流,只有一个简短的,在Ca激活的BK电流之前,小的净向内钙电流主导了总的Ca依赖性电流。在动作电位波形期间,虽然BK电流在钙电流后仅有短暂(~100μs)延迟激活,这两个电流在很大程度上是分开的,钙电流在动作电位下降阶段流动,大部分BK电流在复极化后的几毫秒内流动。逐步去极化既激活了对伊贝毒素敏感的BK成分,又具有快速的激活和失活动力学,同时激活了对伊贝毒素敏感的BK成分。在动作电位激发期间,然而,几乎所有的BK电流都来自更快的门控艾贝毒素敏感通道,即使在动作电位爆发期间。抑制BK电流对动作电位宽度或快速后超极化几乎没有影响,但将介质后超极化转换为后去极化,并且可以将单个动作电位的强直发射转换为爆发发射。BK钙激活钾通道广泛表达于中枢神经元。BK通道的功能改变与癫痫和其他神经元疾病有关。包括小脑共济失调.BK在调节神经元放电模式中的功能作用高度依赖于其他通道的环境,并且在不同类型的神经元之间差异很大。最常见的是,BK通道在动作电位期间被激活,并有助于产生快速的后超极化。我们发现,在Purkinje神经元中,BK电流主要在快速的后超极化之后流动,并有助于防止后期的后极化产生快速的爆发放电,实现典型的定期补品发射。
    BK calcium-activated potassium channels have complex kinetics because they are activated by both voltage and cytoplasmic calcium. The timing of BK activation and deactivation during action potentials determines their functional role in regulating firing patterns but is difficult to predict a priori. We used action potential clamp to characterize the kinetics of voltage-dependent calcium current and BK current during action potentials in Purkinje neurons from mice of both sexes, using acutely dissociated neurons that enabled rapid voltage clamp at 37°C. With both depolarizing voltage steps and action potential waveforms, BK current was entirely dependent on calcium entry through voltage-dependent calcium channels. With voltage steps, BK current greatly outweighed the triggering calcium current, with only a brief, small net inward calcium current before Ca-activated BK current dominated the total Ca-dependent current. During action potential waveforms, although BK current activated with only a short (∼100 μs) delay after calcium current, the two currents were largely separated, with calcium current flowing during the falling phase of the action potential and most BK current flowing over several milliseconds after repolarization. Step depolarizations activated both an iberiotoxin-sensitive BK component with rapid activation and deactivation kinetics and a slower-gating iberiotoxin-resistant component. During action potential firing, however, almost all BK current came from the faster-gating iberiotoxin-sensitive channels, even during bursts of action potentials. Inhibiting BK current had little effect on action potential width or a fast afterhyperpolarization but converted a medium afterhyperpolarization to an afterdepolarization and could convert tonic firing of single action potentials to burst firing.SIGNIFICANCE STATEMENT BK calcium-activated potassium channels are widely expressed in central neurons. Altered function of BK channels is associated with epilepsy and other neuronal disorders, including cerebellar ataxia. The functional role of BK in regulating neuronal firing patterns is highly dependent on the context of other channels and varies widely among different types of neurons. Most commonly, BK channels are activated during action potentials and help produce a fast afterhyperpolarization. We find that in Purkinje neurons BK current flows primarily after the fast afterhyperpolarization and helps to prevent a later afterdepolarization from producing rapid burst firing, enabling typical regular tonic firing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号