ABCC9

ABCC9
  • 文章类型: Journal Article
    目标:坎图综合征(CS),具有复杂心血管表型的多系统疾病,由ATP敏感性钾(KATP)通道的Kir6.1/SUR2亚基中的GoF变体引起,其特点是全身血管阻力低,以及曲折,扩张的血管,脉搏波速度降低。因此,CS血管功能障碍是多因素的,同时具有肌强直和超弹性成分。为了剖析这种复杂性是否在血管平滑肌细胞(VSMC)内由细胞自主产生,或者作为对病理生理环境的二次反应,我们评估了人类诱导多能干细胞来源的VSMC(hiPSC-VSMC)的电特性和基因表达,从对照和CS患者来源的HiPSC分化,以及在本机鼠标控制和CSVSMC中。
    结果:从野生型(WT)和Kir6.1[V65M](CS)小鼠分离的主动脉和肠系膜动脉VSMC的全细胞电压钳显示电压门控K(Kv)或Ca2电流没有明显差异。Kv和Ca2+电流在从对照分化的验证的hiPSC-VSMC和CS患者来源的hiPSC之间也没有差异。虽然对照hiPSC-VSMC中的吡那地尔敏感的KATP电流与WT小鼠VSMC中的一致,它们在CShiPSC-VSMC中相当大。在电流钳位条件下,CShiPSC-VSMC也是超极化的,与基础钾电导增加一致,并为CS的音调降低和血管阻力降低提供了解释。在分离的CS小鼠主动脉中观察到顺应性增加,并与弹性蛋白mRNA表达增加有关。这与CShiPSC-VSMC中弹性蛋白mRNA的高水平一致,表明CS血管病变的超弹性成分是血管KATPGoF的细胞自主结果。
    结论:结果表明,hiPSC-VSMC重申了与初级VSMC相同的主要离子电流的表达,验证使用这些细胞来研究血管疾病。源自CS患者细胞的hiPSC-VSMC的结果表明,CS血管病变的肌强直和超弹性成分都是由VSMC内KATP过度活动驱动的细胞自主现象。
    Cantú syndrome (CS), a multisystem disease with a complex cardiovascular phenotype, is caused by gain-of-function (GoF) variants in the Kir6.1/SUR2 subunits of ATP-sensitive potassium (KATP) channels and is characterized by low systemic vascular resistance, as well as tortuous, dilated, vessels, and decreased pulse-wave velocity. Thus, CS vascular dysfunction is multifactorial, with both hypomyotonic and hyperelastic components. To dissect whether such complexities arise cell autonomously within vascular smooth muscle cells (VSMCs) or as secondary responses to the pathophysiological milieu, we assessed electrical properties and gene expression in human induced pluripotent stem cell-derived VSMCs (hiPSC-VSMCs), differentiated from control and CS patient-derived hiPSCs, and in native mouse control and CS VSMCs. Whole-cell voltage clamp of isolated aortic and mesenteric arterial VSMCs isolated from wild-type (WT) and Kir6.1[V65M] (CS) mice revealed no clear differences in voltage-gated K+ (Kv) or Ca2+ currents. Kv and Ca2+ currents were also not different between validated hiPSC-VSMCs differentiated from control and CS patient-derived hiPSCs. While pinacidil-sensitive KATP currents in control hiPSC-VSMCs were similar to those in WT mouse VSMCs, they were considerably larger in CS hiPSC-VSMCs. Under current-clamp conditions, CS hiPSC-VSMCs were also hyperpolarized, consistent with increased basal K conductance and providing an explanation for decreased tone and decreased vascular resistance in CS. Increased compliance was observed in isolated CS mouse aortae and was associated with increased elastin mRNA expression. This was consistent with higher levels of elastin mRNA in CS hiPSC-VSMCs and suggesting that the hyperelastic component of CS vasculopathy is a cell-autonomous consequence of vascular KATP GoF. The results show that hiPSC-VSMCs reiterate expression of the same major ion currents as primary VSMCs, validating the use of these cells to study vascular disease. Results in hiPSC-VSMCs derived from CS patient cells suggest that both the hypomyotonic and hyperelastic components of CS vasculopathy are cell-autonomous phenomena driven by KATP overactivity within VSMCs .
    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
    ABCC9的功能缺失突变,该基因编码ATP敏感钾(KATP)通道的SUR2亚基,最近与常染色体隐性遗传ABCC9相关的智力障碍和肌病综合征(AIMS)相关。在这里,我们确定了九个额外的主题,来自七个不相关的家庭,在ABCC9中具有不同的纯合LoF变体,并具有保守的临床特征。预测所有变体导致SUR2内的严重截短或框内缺失,导致非功能性SUR2依赖性KATP通道的产生。受影响的个体表现出不同严重程度的精神运动延迟和智力残疾,小头畸形,call体和白质异常,癫痫发作,痉挛,身材矮小,肌肉疲劳,和弱点。杂合的父母没有显示任何保守的临床病理,但报告多次发生宫内胎儿死亡,在本研究中纳入的第八个家庭中也观察到了这一点。斑马鱼abcc9LoF的体内研究揭示了对戊四唑的加剧的运动反应,一种促惊厥的药物,与癫痫发作易感性增加相关的神经发育受损一致。我们的发现定义了ABCC9LoF相关表型,扩展AIMS的基因型和表型谱,并揭示由KATP通道功能障碍引起的新型人类病理。
    Loss-of-function mutation of ABCC9, the gene encoding the SUR2 subunit of ATP sensitive-potassium (KATP) channels, was recently associated with autosomal recessive ABCC9-related intellectual disability and myopathy syndrome (AIMS). Here we identify nine additional subjects, from seven unrelated families, harbouring different homozygous loss-of-function variants in ABCC9 and presenting with a conserved range of clinical features. All variants are predicted to result in severe truncations or in-frame deletions within SUR2, leading to the generation of non-functional SUR2-dependent KATP channels. Affected individuals show psychomotor delay and intellectual disability of variable severity, microcephaly, corpus callosum and white matter abnormalities, seizures, spasticity, short stature, muscle fatigability and weakness. Heterozygous parents do not show any conserved clinical pathology but report multiple incidences of intra-uterine fetal death, which were also observed in an eighth family included in this study. In vivo studies of abcc9 loss-of-function in zebrafish revealed an exacerbated motor response to pentylenetetrazole, a pro-convulsive drug, consistent with impaired neurodevelopment associated with an increased seizure susceptibility. Our findings define an ABCC9 loss-of-function-related phenotype, expanding the genotypic and phenotypic spectrum of AIMS and reveal novel human pathologies arising from KATP channel dysfunction.
    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
    暂无摘要。
    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
    ABCC9相关的智力障碍和肌病综合征(AIMS)源于ABCC9基因的功能丧失(LoF)突变,它编码ATP敏感性钾(KATP)通道的SUR2亚基。KATP通道遍布心血管系统和骨骼肌,并将细胞代谢与兴奋性联系起来。AIMS个人表现出易疲劳,肌肉痉挛,和心脏功能障碍。我们发现在ABCC9中带有过早停止密码子的AIMS小鼠模型中运动表现降低。鉴于KATP通道在所有肌肉中的作用,我们试图通过组织选择性抑制KATP来确定肌病是如何发生的,并发现骨骼肌中的LoF,具体来说,是肌病的基础.在孤立的肌肉中,SUR2LoF导致未刺激力的异常产生,可能解释AIMS中痛苦的痉挛。我们试图确定通过CaV1.1通道的过量Ca2流入是否与肌肉病理学有关,但发现Ca2通道阻断剂维拉帕米意外地导致AIMS小鼠过早死亡,并且使CaV1.1通道不可渗透的突变无法逆转病理;结果警告在AIMS中使用钙通道阻断剂。
    ABCC9-related intellectual disability and myopathy syndrome (AIMS) arises from loss-of-function (LoF) mutations in the ABCC9 gene, which encodes the SUR2 subunit of ATP-sensitive potassium (KATP ) channels. KATP channels are found throughout the cardiovascular system and skeletal muscle and couple cellular metabolism to excitability. AIMS individuals show fatigability, muscle spasms, and cardiac dysfunction. We found reduced exercise performance in mouse models of AIMS harboring premature stop codons in ABCC9. Given the roles of KATP channels in all muscles, we sought to determine how myopathy arises using tissue-selective suppression of KATP and found that LoF in skeletal muscle, specifically, underlies myopathy. In isolated muscle, SUR2 LoF results in abnormal generation of unstimulated forces, potentially explaining painful spasms in AIMS. We sought to determine whether excessive Ca2+ influx through CaV 1.1 channels was responsible for myopathology but found that the Ca2+ channel blocker verapamil unexpectedly resulted in premature death of AIMS mice and that rendering CaV 1.1 channels nonpermeable by mutation failed to reverse pathology; results which caution against the use of calcium channel blockers in AIMS.
    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
    未经证实:我们假设ATP敏感性K+通道(KATP)调节亚基(ABCC9)参与PAH发病机制。ABCC9基因编码KATP通道的两个调节亚基:SUR2A和SUR2B蛋白。在KATP通道中,SUR2亚基与K+通道Kir6.1相关。我们研究了SUR2/Kir6.1通道如何促进PAH发病机理及其作为PAH治疗靶点的潜力。
    未经评估:使用体外,离体,和体内方法,我们分析了SUR2A的定位和表达,SUR2B,在实验性肺动脉高压(PH)大鼠模型中,对照组和PAH患者的肺血管中的Kir6.1及其对PAH病理生理学的贡献。最后,我们破译了在野百合碱(MCT)诱导的PH模型中SUR2/Kir6.1体内活化的后果。我们发现SUR2A,SUR2B,和Kir6.1在PAH和MCT诱导的PH大鼠模型的对照组和患者的肺中表达。器官浴研究表明,吡那地尔激活的SUR2可诱导大鼠和人的肺动脉舒张。对照组和PAH患者的人肺动脉平滑肌细胞和内皮细胞(hPASMCs和hPAECs)的体外实验显示,SUR2激活后细胞增殖和迁移减少。我们证明,通过膜片钳,大鼠右心室(RV)心肌细胞中的SUR2激活减少了RV动作电位的持续时间。对照大鼠的慢性吡那地尔给药增加了心率,而血液动力学参数没有变化。最后,SUR2对MCT和慢性缺氧(CH)诱导的PH大鼠的体内药理激活显示出改善的PH。
    未经批准:我们证明了SUR2A,SUR2B,和Kir6.1出现在对照组和PAH患者的hPASMC和hPAECs中。体内SUR2活化降低了MCT诱导和CH诱导的PH表型,提示SUR2激活应考虑用于治疗PAH。
    UNASSIGNED: We hypothesized that the ATP-sensitive K+ channels (KATP) regulatory subunit (ABCC9) contributes to PAH pathogenesis. ABCC9 gene encodes for two regulatory subunits of KATP channels: the SUR2A and SUR2B proteins. In the KATP channel, the SUR2 subunits are associated with the K+ channel Kir6.1. We investigated how the SUR2/Kir6.1 channel contributes to PAH pathogenesis and its potential as a therapeutic target in PAH.
    UNASSIGNED: Using in vitro, ex vivo, and in vivo approaches, we analyzed the localization and expression of SUR2A, SUR2B, and Kir6.1 in the pulmonary vasculature of controls and patients with PAH as in experimental pulmonary hypertension (PH) rat models and its contribution to PAH physiopathology. Finally, we deciphered the consequences of in vivo activation of SUR2/Kir6.1 in the monocrotaline (MCT)-induced PH model. We found that SUR2A, SUR2B, and Kir6.1 were expressed in the lungs of controls and patients with PAH and MCT-induced PH rat models. Organ bath studies showed that SUR2 activation by pinacidil induced relaxation of pulmonary arterial in rats and humans. In vitro experiments on human pulmonary arterial smooth muscle cells and endothelial cells (hPASMCs and hPAECs) in controls and PAH patients showed decreased cell proliferation and migration after SUR2 activation. We demonstrated that SUR2 activation in rat right ventricular (RV) cardiomyocytes reduced RV action potential duration by patch-clamp. Chronic pinacidil administration in control rats increased heart rate without changes in hemodynamic parameters. Finally, in vivo pharmacological activation of SUR2 on MCT and Chronic-hypoxia (CH)-induced-PH rats showed improved PH.
    UNASSIGNED: We showed that SUR2A, SUR2B, and Kir6.1 are presented in hPASMCs and hPAECs of controls and PAH patients. In vivo SUR2 activation reduced the MCT-induced and CH-induced PH phenotype, suggesting that SUR2 activation should be considered for treating PAH.
    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
    背景由左心室肥厚引起的心脏肥大是充血性心力衰竭发展的危险因素。与收缩和/或舒张心室功能下降有关。人们对左心室肥厚的表型关注较少,心室功能增强,心输出量增加,这可能与高输出心力衰竭有关。缺乏识别可能会造成诊断歧义和管理复杂性。方法和结果我们试图系统地描述Cantu综合征(CS)受试者的高输出心脏肥大,由ABCC9中的功能获得变体引起,ABCC9编码心血管KATP(ATP敏感性钾)通道亚基。我们纵向研究了31例具有证实ABCC9变异的CS患者的心血管表型(中位[四分位距]8岁[3-32岁],体重指数19.9[16.5-22.9],16名男性受试者)。患有CS的受试者表现出显著的左心室肥厚(CS中左心室质量指数86.7[57.7-103.0]g/m2,n=30;对照组为26.6[24.1-32.8]g/m2,n=17;P<0.0001)和低血压(CS收缩压94.5[90-103]mmHg,n=17;对照中109[98-115]mmHg,n=17;P=0.0301;CS舒张压60[56-66]mmHg,n=17;对照为69[65-72]mmHg,n=17;P=0.0063)。大多数(21/31)CS患者表现出偏心肥大,左心室壁厚度正常。在长期随访中,5名年龄>40岁的CS受试者中有4名出现充血性心力衰竭症状。结论数据定义了CS患者的全身血管阻力降低导致高输出心脏肥大的自然史。由低全身血管阻力引起的高输出肥大的长期后果的定义人群,以及进展为高输出心力衰竭的可能性。
    Background Cardiomegaly caused by left ventricular hypertrophy is a risk factor for development of congestive heart failure, classically associated with decreased systolic and/or diastolic ventricular function. Less attention has been given to the phenotype of left ventricular hypertrophy with enhanced ventricular function and increased cardiac output, which is potentially associated with high-output heart failure. Lack of recognition may pose diagnostic ambiguity and management complexities. Methods and Results We sought to systematically characterize high-output cardiac hypertrophy in subjects with Cantu syndrome (CS), caused by gain-of-function variants in ABCC9, which encodes cardiovascular KATP (ATP-sensitive potassium) channel subunits. We studied the cardiovascular phenotype longitudinally in 31 subjects with CS with confirmed ABCC9 variants (median [interquartile range] age 8 years [3-32 years], body mass index 19.9 [16.5-22.9], 16 male subjects). Subjects with CS presented with significant left ventricular hypertrophy (left ventricular mass index 86.7 [57.7-103.0] g/m2 in CS, n=30; 26.6 [24.1-32.8] g/m2 in controls, n=17; P<0.0001) and low blood pressure (systolic 94.5 [90-103] mm Hg in CS, n=17; 109 [98-115] mm Hg in controls, n=17; P=0.0301; diastolic 60 [56-66] mm Hg in CS, n=17; 69 [65-72] mm Hg in control, n=17; P=0.0063). Most (21/31) subjects with CS exhibited eccentric hypertrophy with normal left ventricular wall thickness. Congestive heart failure symptoms were evident in 4 of the 5 subjects with CS aged >40 years on long-term follow-up. Conclusions The data define the natural history of high-output cardiac hypertrophy resulting from decreased systemic vascular resistance in subjects with CS, a defining population for long-term consequences of high-output hypertrophy caused by low systemic vascular resistance, and the potential for progression to high-output heart failure.
    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
    Kir6.1和SUR2是在多种组织中表达的ATP敏感性钾(KATP)通道的亚基。广泛的研究暗示了这些通道亚基在多种生理功能中的作用。它们一起在血管平滑肌中产生主要的KATP电导,并且是血管舒张药物的靶标。基于动物模型的研究和人类遗传发现,已经提出了Kir6.1/SUR2功能障碍在疾病中的作用。近年来,很明显,这两个基因的功能获得(GoF)突变会导致Cantú综合征(CS)-一种复杂的疾病,多系统疾病。目前尚无针对CS的靶向治疗,但是对该疾病小鼠模型的研究表明,通过施用KATP通道抑制剂可以实现心血管和胃肠道病理的药理学可逆性,格列本脲.在这里我们回顾一下函数,结构,以及Kir6.1/SUR2B通道的生理和病理作用,专注于CS。最近的研究使人们对潜在的病理和治疗潜力有了很大的了解,
    Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?
    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
    ATP敏感性钾通道(K-ATP)编码基因的功能缺失或获得突变,KCNJ8和ABCC9,引起人类中枢神经系统紊乱,发病机制不明。这里,用老鼠,斑马鱼,和细胞培养模型,我们研究了由K-ATP通道功能改变引起的脑功能障碍的细胞和分子原因。我们表明,含有KCNJ8/ABCC9的K-ATP通道功能的遗传/化学抑制或激活导致脑选择性抑制或促进动脉/小动脉血管平滑肌细胞(VSMC)分化,分别。我们进一步表明,大脑VSMC是从含有KCNJ8/ABCC9的K-ATP通道表达壁系细胞祖细胞发展而来的,并且K-ATP通道细胞通过电压依赖性钙通道调节细胞内Ca2振荡来自主调节VSMC分化。与有缺陷的VSMC开发一致,Kcnj8基因敲除小鼠表现出血管收缩能力和神经元诱发的血管舒张功能不足,导致局部充血。我们的结果证明了含有KCNJ8/ABCC9的K-ATP通道在脑VSMC分化中的作用。这反过来又是微调脑血流所必需的。
    Loss- or gain-of-function mutations in ATP-sensitive potassium channel (K-ATP)-encoding genes, KCNJ8 and ABCC9, cause human central nervous system disorders with unknown pathogenesis. Here, using mice, zebrafish, and cell culture models, we investigated cellular and molecular causes of brain dysfunctions derived from altered K-ATP channel function. We show that genetic/chemical inhibition or activation of KCNJ8/ABCC9-containing K-ATP channel function leads to brain-selective suppression or promotion of arterial/arteriolar vascular smooth muscle cell (VSMC) differentiation, respectively. We further show that brain VSMCs develop from KCNJ8/ABCC9-containing K-ATP channel-expressing mural cell progenitor and that K-ATP channel cell autonomously regulates VSMC differentiation through modulation of intracellular Ca2+ oscillation via voltage-dependent calcium channels. Consistent with defective VSMC development, Kcnj8 knockout mice showed deficiency in vasoconstrictive capacity and neuronal-evoked vasodilation leading to local hyperemia. Our results demonstrate a role for KCNJ8/ABCC9-containing K-ATP channels in the differentiation of brain VSMC, which in turn is necessary for fine-tuning of cerebral blood flow.
    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
    ABCC9基因的遗传变异,编码来自KATP通道的SUR2辅助亚基,以前与各种遗传性疾病有关。这种广泛的先天性疾病包括多系统和心血管疾病。功能获得突变导致Cantu综合征,肢端巨大样面部外观,多毛症,和肢端类面部特征。ABCC9基因的功能缺失突变与Brugada综合征有关,早期复极综合征,和扩张型心肌病.这里,我们报道了一名ABCC9基因功能缺失变异的患者,通过靶高通量测序鉴定。女性先证者因情绪压力而出现多次室颤和低钾血症。该病例揭示了心血管系统中KATP通道功能障碍的后果,并强调了ABCC9相关疾病临床表现的复杂性。
    Genetic variants in the ABCC9 gene, encoding the SUR2 auxiliary subunit from KATP channels, were previously linked with various inherited diseases. This wide range of congenital disorders includes multisystem and cardiovascular pathologies. The gain-of-function mutations result in Cantu syndrome, acromegaloid facial appearance, hypertrichosis, and acromegaloid facial features. The loss-of-function mutations in the ABCC9 gene were associated with the Brugada syndrome, early repolarization syndrome, and dilated cardiomyopathy. Here, we reported a patient with a loss-of-function variant in the ABCC9 gene, identified by target high-throughput sequencing. The female proband presented with several episodes of ventricular fibrillation and hypokalemia upon emotional stress. This case sheds light on the consequences of KATP channel dysfunction in the cardiovascular system and underlines the complexity of the clinical presentation of ABCC9-related 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
    ATP敏感性钾通道(KATP通道)是异源八聚体核苷酸门控离子通道,可将细胞代谢与各种组织中的兴奋性耦合。在心中,KATP通道在缺血期间和潜在的肾上腺素能刺激期间被激活。在脉管系统中,它们通常在低水平活跃,降低血管张力,但是,这些通道的普遍存在的性质导致复杂和知之甚少的信道病作为增益或功能丧失突变的结果。这些通道病的斑马鱼(ZF)模型可能为分子功能障碍和复杂的病理生理学之间的联系提供了见解。但这需要了解通道活性和亚基特异性的组织依赖性。到目前为止,ZFKATP表达和功能特性的直接分析仅在胰腺β细胞中进行。使用转基因鱼类的综合组合,电生理学和基因表达分析,我们证明ZF心肌细胞(CM)和血管平滑肌(VSM)表达相似亚基组成的功能性KATP通道,对哺乳动物的结构和代谢敏感性。然而,与哺乳动物心血管KATP通道相反,ZF通道对钾通道开放剂药物不敏感(吡那地尔,米诺地尔)在心脏的两个腔室和VSM中。结果提供了鱼类KATP通道分子特性的首次表征,并验证了将此类转基因鱼类用作人类Cantú综合征和ABCC9相关的智力障碍和肌病综合征的模型。关键点:斑马鱼心肌细胞(CM)和血管平滑肌(VSM)表达相似亚基组成的功能KATP通道,对哺乳动物的结构和代谢敏感性。与哺乳动物心血管KATP通道相反,斑马鱼通道对钾通道开放剂药物不敏感(吡那地尔,米诺地尔)在心脏的两个腔室和VSM中。我们对鱼类KATP通道的分子特性进行了首次表征,并验证了将此类转基因鱼类用作人类Cantú综合征和ABCC9相关的智力障碍和肌病综合征的模型。
    ATP-sensitive potassium channels (KATP channels) are hetero-octameric nucleotide-gated ion channels that couple cellular metabolism to excitability in various tissues. In the heart, KATP channels are activated during ischaemia and potentially during adrenergic stimulation. In the vasculature, they are normally active at a low level, reducing vascular tone, but the ubiquitous nature of these channels leads to complex and poorly understood channelopathies as a result of gain- or loss-of-function mutations. Zebrafish (ZF) models of these channelopathies may provide insights to the link between molecular dysfunction and complex pathophysiology, but this requires understanding the tissue dependence of channel activity and subunit specificity. Thus far, direct analysis of ZF KATP expression and functional properties has only been performed in pancreatic β-cells. Using a comprehensive combination of genetically modified fish, electrophysiology and gene expression analysis, we demonstrate that ZF cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. However, in contrast to mammalian cardiovascular KATP channels, ZF channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. The results provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome. KEY POINTS: Zebrafish cardiac myocytes (CM) and vascular smooth muscle (VSM) express functional KATP channels of similar subunit composition, structure and metabolic sensitivity to their mammalian counterparts. In contrast to mammalian cardiovascular KATP channels, zebrafish channels are insensitive to potassium channel opener drugs (pinacidil, minoxidil) in both chambers of the heart and in VSM. We provide a first characterization of the molecular properties of fish KATP channels and validate the use of such genetically modified fish as models of human Cantú syndrome and ABCC9-related Intellectual Disability and Myopathy syndrome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号