KCNJ10

KCNJ10
  • 文章类型: Journal Article
    远曲小管(DCT)的基底外侧钾通道由向内整流钾通道4.1(Kir4.1)和Kir5.1组成。Kir4.1与Kir5.1相互作用形成40pSK通道,这是DCT基底外侧膜中表达的唯一K型通道。此外,Kir4.1/Kir5.1异四聚体在决定噻嗪敏感的Na-Cl共转运(NCC)的表达和活性中起关键作用。除Kir4.1/Kir5.1外,Kir1.1(ROMK)在晚期DCT(DCT2)的顶膜中表达,并在介导上皮Na通道(ENaC)依赖性K排泄中起关键作用。高膳食K+摄入量(HK)刺激ROMK并抑制DCT中的Kir4.1/Kir5.1。Kir4.1/Kir5.1的抑制对于HK诱导的NCC抑制是必需的,而ROMK的刺激对于在HK期间增加ENaC依赖性的K+排泄是重要的。我们现在已经使用膜片钳技术来检查K饮食的性别和Cl-含量是否影响HK诱导的基底外侧Kir4.1/Kir5.1的抑制和HK诱导的ROMK刺激。单通道记录显示,NPo定义的DCT的基底外侧40pSK通道(Kir4.1/Kir5.1)活性为1.34(1%KCl,正常K,NK),在雄性小鼠中为0.95(5%KCl)和1.03(5%K+-柠檬酸盐),而在雌性小鼠中为1.47、1.02和1.05。全细胞记录显示,早期DCT(DCT1)的Kir4.1/Kir5.1介导的K电流为1,170pA(NK),雄性小鼠的725pA(5%KCl)和700pA(5%K柠檬酸盐),而为1,125pA,雌性小鼠中的674pA和700pA。此外,DCT的K+电流(IK)反转电位(膜电位指数)为-63mV(NK),在雄性小鼠中-49mV(5%KCl)和-49mV(5%柠檬酸钾),而它是-63mV,雌性小鼠中的-50mV和-50mV。最后,DCT2/初始连接小管(CNT)中的TPNQ敏感的全细胞ROMK电流为910pA(NK),雄性小鼠的1,520pA(5%KCl)和1,540pA(5%柠檬酸钾),而ROMK介导的K电流为1,005pA,雌性小鼠中的1,590pA和1,570pA。我们得出的结论是,在雄性和雌性小鼠之间,HK摄入对DCT的Kir4.1/Kir5.1和DCT2/CNT的ROMK的影响相似。此外,HK饮食中的Cl-含量对HK诱导的DCT的Kir4.1/Kir5.1抑制和HK诱导的DCT2/CNT中的ROMK刺激没有影响。
    Basolateral potassium channels in the distal convoluted tubule (DCT) are composed of inwardly-rectifying potassium channel 4.1 (Kir4.1) and Kir5.1. Kir4.1 interacts with Kir5.1 to form a 40 pS K+ channel which is the only type K+ channel expressed in the basolateral membrane of the DCT. Moreover, Kir4.1/Kir5.1 heterotetramer plays a key role in determining the expression and activity of thiazide-sensitive Na-Cl cotransport (NCC). In addition to Kir4.1/Kir5.1, Kir1.1 (ROMK) is expressed in the apical membrane of the late DCT (DCT2) and plays a key role in mediating epithelial Na+ channel (ENaC)-dependent K+ excretion. High dietary-K+-intake (HK) stimulates ROMK and inhibits Kir4.1/Kir5.1 in the DCT. Inhibition of Kir4.1/Kir5.1 is essential for HK-induced suppression of NCC whereas the stimulation of ROMK is important for increasing ENaC-dependent K+ excretion during HK. We have now used the patch-clamp-technique to examine whether gender and Cl- content of K+-diet affect HK-induced inhibition of basolateral Kir4.1/Kir5.1 and HK-induced stimulation of ROMK. Single-channel-recording shows that basolateral 40 pS K+ channel (Kir4.1/Kir5.1) activity of the DCT defined by NPo was 1.34 (1% KCl, normal K, NK), 0.95 (5% KCl) and 1.03 (5% K+-citrate) in male mice while it was 1.47, 1.02 and 1.05 in female mice. The whole-cell recording shows that Kir4.1/Kir5.1-mediated-K+ current of the early-DCT (DCT1) was 1,170 pA (NK), 725 pA (5% KCl) and 700 pA (5% K+-citrate) in male mice whereas it was 1,125 pA, 674 pA and 700 pA in female mice. Moreover, K+-currents (IK) reversal potential of DCT (an index of membrane potential) was -63 mV (NK), -49 mV (5% KCl) and -49 mV (5% K-citrate) in the male mice whereas it was -63 mV, -50 mV and -50 mV in female mice. Finally, TPNQ-sensitive whole-cell ROMK-currents in the DCT2 /initial-connecting tubule (CNT) were 910 pA (NK), 1,520 pA (5% KCl) and 1,540 pA (5% K+-citrate) in male mice whereas the ROMK-mediated K+ currents were 1,005 pA, 1,590 pA and 1,570 pA in female mice. We conclude that the effect of HK intake on Kir4.1/Kir5.1 of the DCT and ROMK of DCT2/CNT is similar between male and female mice. Also, Cl- content in HK diets has no effect on HK-induced inhibition of Kir4.1/Kir5.1 of the DCT and HK-induced stimulation of ROMK in DCT2/CNT.
    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
    内向整流钾(Kir)通道在许多哺乳动物器官系统中广泛表达,它们有助于关键的生理功能。然而,Kir5.1通道(由KCNJ16基因编码)的重要性和功能尚未得到充分认识.这篇综述的重点是在了解Kir5.1通道在对钾稳态和神经系统疾病至关重要的基本生理系统中的表达模式和功能作用方面的最新进展。最近的研究已经描述了Kir5.1形成Kir通道在Kcnj16基因突变的小鼠和大鼠品系中的作用。动物研究揭示了不同的肾脏和神经表型,包括pH和电解质失衡,对高碳酸血症/缺氧的通气反应减弱,和癫痫症。此外,已证实,这些表型让人想起那些在KCNJ16基因突变的患者队列中,进一步表明Kir5.1通道在稳态/神经系统健康和疾病中的关键作用。未来的研究集中在这些渠道的许多功能作用,扩大对人类患者的基因筛查,以及Kir5.1通道的选择性小分子抑制剂的开发,将继续增加我们对这个独特的Kir频道家族成员的了解。
    Inwardly rectifying potassium (Kir) channels are broadly expressed in many mammalian organ systems, where they contribute to critical physiological functions. However, the importance and function of the Kir5.1 channel (encoded by the KCNJ16 gene) have not been fully recognized. This review focuses on the recent advances in understanding the expression patterns and functional roles of Kir5.1 channels in fundamental physiological systems vital to potassium homeostasis and neurological disorders. Recent studies have described the role of Kir5.1-forming Kir channels in mouse and rat lines with mutations in the Kcnj16 gene. The animal research reveals distinct renal and neurological phenotypes, including pH and electrolyte imbalances, blunted ventilatory responses to hypercapnia/hypoxia, and seizure disorders. Furthermore, it was confirmed that these phenotypes are reminiscent of those in patient cohorts in which mutations in the KCNJ16 gene have also been identified, further suggesting a critical role for Kir5.1 channels in homeostatic/neural systems health and disease. Future studies that focus on the many functional roles of these channels, expanded genetic screening in human patients, and the development of selective small-molecule inhibitors for Kir5.1 channels, will continue to increase our understanding of this unique Kir channel family member.
    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
    向内整流钾通道(Kir)4.1(由KCNJ10编码)与Kir5.1(由KCNJ16编码)相互作用,在肾远曲小管(DCT)中形成主要的基底外侧K通道,连接管(CNT),和皮质集合管(CCD)。Kir4.1/Kir5.1异四聚体在DCT中调节Na+和K+转运中起重要作用,CNT,和CCD。该领域的最新发展已牢固地确立了DCT的Kir4.1/Kir5.1异四聚体在调节噻嗪敏感的Na-Cl协同转运蛋白(NCC)中的作用。DCT的Kir4.1/Kir5.1活性的变化是调节饮食K和Na摄入诱导的NCC表达/活性的重要步骤,并在通过2型血管紧张素II受体(AT2R)调节NCC中发挥作用,缓激肽II型受体(BK2R),和β-肾上腺素能受体。由于NCC活性决定了醛固酮敏感性远端肾单位(ASDN)的Na+递送率,从晚期DCT到CCD的远端肾单位段,Kir4.1/Kir5.1活性不仅在调节肾脏Na吸收中起关键作用,而且在调节肾脏K排泄和维持K稳态中起关键作用。因此,Kir4.1/Kir5.1活性是肾脏K+传感机制的重要组成部分。这篇综述的主要重点是概述DCT和CCD的Kir4.1和Kir5.1在调节肾脏K排泄和Na吸收中的作用。
    The inwardly rectifying potassium channel (Kir) 4.1 (encoded by KCNJ10) interacts with Kir5.1 (encoded by KCNJ16) to form a major basolateral K+ channel in the renal distal convoluted tubule (DCT), connecting tubule (CNT), and the cortical collecting duct (CCD). Kir4.1/Kir5.1 heterotetramer plays an important role in regulating Na+ and K+ transport in the DCT, CNT, and CCD. A recent development in the field has firmly established the role of Kir4.1/Kir5.1 heterotetramer of the DCT in the regulation of thiazide-sensitive Na-Cl cotransporter (NCC). Changes in Kir4.1/Kir5.1 activity of the DCT are an essential step for the regulation of NCC expression/activity induced by dietary K+ and Na+ intakes and play a role in modulating NCC by type 2 angiotensin II receptor (AT2R), bradykinin type II receptor (BK2R), and β-adrenergic receptor. Since NCC activity determines the Na+ delivery rate to the aldosterone-sensitive distal nephron (ASDN), a distal nephron segment from late DCT to CCD, Kir4.1/Kir5.1 activity plays a critical role not only in the regulation of renal Na+ absorption but also in modulating renal K+ excretion and maintaining K+ homeostasis. Thus, Kir4.1/Kir5.1 activity serves as an important component of renal K+ sensing mechanism. The main focus of this review is to provide an overview regarding the role of Kir4.1 and Kir5.1 of the DCT and CCD in the regulation of renal K+ excretion and Na+ absorption.
    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
    2009年,两组独立地将向内整流K通道Kir4.1(基因名称KCNJ10)中的人类突变与影响中枢神经系统(CNS)的综合征联系起来。听力,和肾小管盐重吸收。常染色体隐性综合征被命名为EAST(癫痫,共济失调,感觉神经性耳聋,和肾小管病)或SeSAME综合征(癫痫发作,感觉神经性耳聋,共济失调,智力残疾,和电解质不平衡),因此。EAST/SeSAME患者的肾功能障碍导致Na+的损失,K+,和尿液中的Mg2+,激活肾素-血管紧张素-醛固酮系统,和低钾血症代谢性碱中毒。Kir4.1在受影响的器官中高度表达:CNS,内耳,还有肾.在肾脏,它主要与Kir5.1(KCNJ16)形成异聚通道。Kir5.1的双等位基因功能丧失突变也可能具有疾病意义,但临床症状与EAST/SeSAME综合征的症状有很大不同:虽然感觉神经性听力损失和低钾血症被复制,没有碱中毒,而是不同严重程度的酸中毒;与EAST/SeSAME综合征相反,中枢神经系统不受影响。这篇综述为理解这些差异提供了一个框架,并将指导读者通过关于Kir4.1和Kir5.1的越来越多的文献,从分子和系统生理学的角度讨论复杂的疾病机制和疾病症状的可变表达。了解这些疾病的病理生理学及其多方面的临床谱是做出正确诊断的重要前提,并为个性化治疗奠定了基础。
    In 2009, two groups independently linked human mutations in the inwardly rectifying K+ channel Kir4.1 (gene name KCNJ10) to a syndrome affecting the central nervous system (CNS), hearing, and renal tubular salt reabsorption. The autosomal recessive syndrome has been named EAST (epilepsy, ataxia, sensorineural deafness, and renal tubulopathy) or SeSAME syndrome (seizures, sensorineural deafness, ataxia, intellectual disability, and electrolyte imbalance), accordingly. Renal dysfunction in EAST/SeSAME patients results in loss of Na+, K+, and Mg2+ with urine, activation of the renin-angiotensin-aldosterone system, and hypokalemic metabolic alkalosis. Kir4.1 is highly expressed in affected organs: the CNS, inner ear, and kidney. In the kidney, it mostly forms heteromeric channels with Kir5.1 (KCNJ16). Biallelic loss-of-function mutations of Kir5.1 can also have disease significance, but the clinical symptoms differ substantially from those of EAST/SeSAME syndrome: although sensorineural hearing loss and hypokalemia are replicated, there is no alkalosis, but rather acidosis of variable severity; in contrast to EAST/SeSAME syndrome, the CNS is unaffected. This review provides a framework for understanding some of these differences and will guide the reader through the growing literature on Kir4.1 and Kir5.1, discussing the complex disease mechanisms and the variable expression of disease symptoms from a molecular and systems physiology perspective. Knowledge of the pathophysiology of these diseases and their multifaceted clinical spectrum is an important prerequisite for making the correct diagnosis and forms the basis for personalized therapies.
    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
    位于远端肾单位上皮细胞基底外侧膜上的向内整流K(Kir)通道在K处理和BP控制中起着至关重要的作用。使这些通道成为治疗高血压的有吸引力的目标。本研究的目的是确定基底外侧Kir4.1/Kir5.1异聚K通道的抑制如何影响皮质集合管(CCD)主要细胞中上皮钠通道(ENaC)介导的Na转运。
    氟西汀的作用,阿米替林和最近开发的Kir抑制剂,VU0134992,对Kir4.1,Kir4.1/Kir5.1和ENaC的活性使用电生理学方法在用各自通道亚基转染的CHO细胞中进行测试,培养的极化上皮mCCDcl1细胞和新鲜分离的大鼠和人CCD小管。为了测试药理Kir4.1/Kir5.1抑制对体内电解质稳态的影响以及远端小管运输的相应变化,Dahl盐敏感大鼠注射阿米替林(15mg·kg-1·day-1)3天。
    我们发现抑制Kir4.1/Kir5.1,但不抑制Kir4.1通道,使细胞膜去极化,诱导细胞内Ca2+浓度升高并抑制ENaC活性。此外,我们证明阿米替林给药导致血浆K+水平显著下降,引发钠排泄和利尿。
    目前的数据揭示了Kir4.1/Kir5.1通道在调节ENaC活性中的特定作用,并强调了使用Kir4.1/Kir5.1抑制剂调节电解质稳态和BP的潜力。
    Inwardly rectifying K+ (Kir ) channels located on the basolateral membrane of epithelial cells of the distal nephron play a crucial role in K+ handling and BP control, making these channels an attractive target for the treatment of hypertension. The purpose of the present study was to determine how the inhibition of basolateral Kir 4.1/Kir 5.1 heteromeric K+ channel affects epithelial sodium channel (ENaC)-mediated Na+ transport in the principal cells of cortical collecting duct (CCD).
    The effect of fluoxetine, amitriptyline and recently developed Kir inhibitor, VU0134992, on the activity of Kir 4.1, Kir 4.1/Kir 5.1 and ENaC were tested using electrophysiological approaches in CHO cells transfected with respective channel subunits, cultured polarized epithelial mCCDcl1 cells and freshly isolated rat and human CCD tubules. To test the effect of pharmacological Kir 4.1/Kir 5.1 inhibition on electrolyte homeostasis in vivo and corresponding changes in distal tubule transport, Dahl salt-sensitive rats were injected with amitriptyline (15 mg·kg-1 ·day-1 ) for 3 days.
    We found that inhibition of Kir 4.1/Kir 5.1, but not the Kir 4.1 channel, depolarizes the cell membrane, induces the elevation of intracellular Ca2+ concentration and suppresses ENaC activity. Furthermore, we demonstrate that amitriptyline administration leads to a significant drop in plasma K+ level, triggering sodium excretion and diuresis.
    The present data uncover a specific role of the Kir 4.1/Kir 5.1 channel in the modulation of ENaC activity and emphasize the potential for using Kir 4.1/Kir 5.1 inhibitors to regulate electrolyte homeostasis and BP.
    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
    一氧化氮(NO)生成酶,一氧化氮合酶-1β(NOS1β),对于钠(Na+)稳态和血压控制至关重要。我们先前表明,在高Na摄入期间,收集导管主细胞NOS1β对于抑制上皮钠通道(ENaC)至关重要。先前对新鲜分离的皮质集合管(CCD)的研究表明,外源性NO通过基底外侧通道促进基底外侧钾(K)电导,大概是基尔4.1(Kcnj10)和基尔5.1(Kcnj16)。我们,因此,研究了NOS1β敲除对Kir4.1/Kir5.1通道活性的影响。的确,在过表达NOS1β和Kir4.1/Kir5.1的CHO细胞中,NO信号传导的抑制降低了通道活性。雄性同窝对照和主细胞NOS1β敲除小鼠(CDNOS1KO)在7天,4%NaCl日粮(HSD)检测基底外侧K+电导的变化。我们先前证明,尽管高盐饮食和适当抑制肾素,CDNOS1KO小鼠仍具有高循环醛固酮。我们在CDNOS1KO小鼠的主要细胞中观察到更高的Kir4.1皮质丰度和更高的Kir4.1/Kir5.1单通道活性。此外,与对照组相比,体内螺内酯治疗阻断醛固酮作用导致CDNOS1KO小鼠中Kir4.1丰度降低,血浆K增加。降低HSD中的K含量可防止CDNOS1KO小鼠的高醛固酮和更高的血浆Na,并使Kir4.1丰度正常化。我们得出结论,在慢性HSD期间,缺乏NOS1β导致血浆K+增加,增强循环醛固酮,并激活ENaC和Kir4.1/Kir5.1通道。因此,主要细胞NOS1β是肾脏调节Na和K所必需的。
    The nitric oxide (NO)-generating enzyme, NO synthase-1β (NOS1β), is essential for sodium (Na+ ) homeostasis and blood pressure control. We previously showed that collecting duct principal cell NOS1β is critical for inhibition of the epithelial sodium channel (ENaC) during high Na+ intake. Previous studies on freshly isolated cortical collecting ducts (CCD) demonstrated that exogenous NO promotes basolateral potassium (K+ ) conductance through basolateral channels, presumably Kir 4.1 (Kcnj10) and Kir 5.1 (Kcnj16). We, therefore, investigated the effects of NOS1β knockout on Kir 4.1/Kir 5.1 channel activity. Indeed, in CHO cells overexpressing NOS1β and Kir 4.1/Kir 5.1, the inhibition of NO signaling decreased channel activity. Male littermate control and principal cell NOS1β knockout mice (CDNOS1KO) on a 7-day, 4% NaCl diet (HSD) were used to detect changes in basolateral K+ conductance. We previously demonstrated that CDNOS1KO mice have high circulating aldosterone despite a high-salt diet and appropriately suppressed renin. We observed greater Kir 4.1 cortical abundance and significantly greater Kir 4.1/Kir 5.1 single-channel activity in the principal cells from CDNOS1KO mice. Moreover, blocking aldosterone action with in vivo spironolactone treatment resulted in lower Kir 4.1 abundance and greater plasma K+ in the CDNOS1KO mice compared to controls. Lowering K+ content in the HSD prevented the high aldosterone and greater plasma Na+ of CDNOS1KO mice and normalized Kir 4.1 abundance. We conclude that during chronic HSD, lack of NOS1β leads to increased plasma K+ , enhanced circulating aldosterone, and activation of ENaC and Kir 4.1/Kir 5.1 channels. Thus, principal cell NOS1β is required for the regulation of both Na+ and K+ by the kidney.
    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
    精子向卵母细胞游动并使其受精的能力取决于精确的K通透性变化。Kir5.1是一种向内整流钾(Kir)通道,对细胞内H(pHi)和细胞外K浓度[K]o具有很高的敏感性,因此提供了pHi和[K+]o变化和膜电位之间的联系。Kir5.1的固有pHi敏感性表明该通道在受精过程中发生的pHi依赖性过程中可能发挥作用。然而,尽管Kir5.1在小鼠精子中定位,它的表达随着年龄和性成熟而增加,通道在精子形态中的作用,成熟,运动性,生育能力是未知的。这里,我们证实了精子中Kir5.1的存在,并在附睾管衬里的平滑肌和上皮细胞中显示出Kir4.1通道的强表达。相比之下,在睾丸中未检测到Kir4.2表达。为了检查Kir5.1在精子生理中的可能作用,我们饲养了Kcnj16(Kir5.1)基因缺失的小鼠,观察到20%的Kir5.1敲除雄性小鼠不育。此外,超过3个月的敲除小鼠中有50%无法繁殖。相比之下,100%的野生型(WT)小鼠是可育的。与WT同窝动物相比,Kcnj16的遗传失活还导致睾丸更小,鞭毛折叠的精子百分比更高。然而,来自突变动物的异常精子表现出增加的进行性运动性。因此,Kcnj16基因的消融将Kir5.1通道识别为有助于睾丸发育的重要元素,精子鞭毛形态,运动性,和生育能力。这些发现可能与理解不同精子离子通道之间复杂的phi和[K]o依赖性相互作用有关,并深入了解它们在受精和不育中的作用。
    The ability of spermatozoa to swim towards an oocyte and fertilize it depends on precise K+ permeability changes. Kir5.1 is an inwardly-rectifying potassium (Kir) channel with high sensitivity to intracellular H+ (pHi) and extracellular K+ concentration [K+]o, and hence provides a link between pHi and [K+]o changes and membrane potential. The intrinsic pHi sensitivity of Kir5.1 suggests a possible role for this channel in the pHi-dependent processes that take place during fertilization. However, despite the localization of Kir5.1 in murine spermatozoa, and its increased expression with age and sexual maturity, the role of the channel in sperm morphology, maturity, motility, and fertility is unknown. Here, we confirmed the presence of Kir5.1 in spermatozoa and showed strong expression of Kir4.1 channels in smooth muscle and epithelial cells lining the epididymal ducts. In contrast, Kir4.2 expression was not detected in testes. To examine the possible role of Kir5.1 in sperm physiology, we bred mice with a deletion of the Kcnj16 (Kir5.1) gene and observed that 20% of Kir5.1 knock-out male mice were infertile. Furthermore, 50% of knock-out mice older than 3 months were unable to breed. By contrast, 100% of wild-type (WT) mice were fertile. The genetic inactivation of Kcnj16 also resulted in smaller testes and a greater percentage of sperm with folded flagellum compared to WT littermates. Nevertheless, the abnormal sperm from mutant animals displayed increased progressive motility. Thus, ablation of the Kcnj16 gene identifies Kir5.1 channel as an important element contributing to testis development, sperm flagellar morphology, motility, and fertility. These findings are potentially relevant to the understanding of the complex pHi- and [K+]o-dependent interplay between different sperm ion channels, and provide insight into their role in fertilization and infertility.
    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
    电解质的跨上皮运输,溶质,肾脏中的水是一个精心策划的过程,涉及许多膜转运系统。肾小管细胞中的基底外侧钾通道不仅介导钾再循环以获得适当的Na+,K+-ATP酶功能,但也参与钾和pH传感。KCNJ10基因缺陷导致EAST/SeSAME综合征,以肾脏盐消耗与癫痫相关的低钾性碱中毒为特征,共济失调,和感觉神经性耳聋.
    候选基因方法和全外显子组测序确定了8名具有新疾病表型的患者的潜在遗传缺陷,这些疾病表型包括低钾性肾小管病伴有肾盐消耗,酸碱稳态受干扰,和感觉神经性耳聋.电生理研究和表面表达实验研究了新鉴定的基因变体的功能后果。
    我们鉴定了编码KCNJ16的KCNJ16基因中的突变,该基因与KCNJ15和KCNJ10一起构成了近端和远端小管的主要基底外侧钾通道,分别。突变体KCNJ16与KCNJ15或KCNJ10在非洲爪狼卵母细胞中的共表达显着降低了电流。
    KCNJ16的双等位基因变异在具有新疾病表型的患者中被发现,该表型包括与耳聋相关的可变近端和远端肾小管病。变体影响异聚钾通道的功能,干扰近端管状碳酸氢盐处理以及远端管状盐重吸收。
    The transepithelial transport of electrolytes, solutes, and water in the kidney is a well-orchestrated process involving numerous membrane transport systems. Basolateral potassium channels in tubular cells not only mediate potassium recycling for proper Na+,K+-ATPase function but are also involved in potassium and pH sensing. Genetic defects in KCNJ10 cause EAST/SeSAME syndrome, characterized by renal salt wasting with hypokalemic alkalosis associated with epilepsy, ataxia, and sensorineural deafness.
    A candidate gene approach and whole-exome sequencing determined the underlying genetic defect in eight patients with a novel disease phenotype comprising a hypokalemic tubulopathy with renal salt wasting, disturbed acid-base homeostasis, and sensorineural deafness. Electrophysiologic studies and surface expression experiments investigated the functional consequences of newly identified gene variants.
    We identified mutations in the KCNJ16 gene encoding KCNJ16, which along with KCNJ15 and KCNJ10, constitutes the major basolateral potassium channel of the proximal and distal tubules, respectively. Coexpression of mutant KCNJ16 together with KCNJ15 or KCNJ10 in Xenopus oocytes significantly reduced currents.
    Biallelic variants in KCNJ16 were identified in patients with a novel disease phenotype comprising a variable proximal and distal tubulopathy associated with deafness. Variants affect the function of heteromeric potassium channels, disturbing proximal tubular bicarbonate handling as well as distal tubular salt reabsorption.
    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
    We first enrolled the available case-control studies to investigate the genetic association between three polymorphisms (rs1130183, rs1890532, and rs2486253) of KCNJ10 (the potassium voltage-gated channel subfamily J member 10) gene and the susceptibility towards clinical epilepsy. We utilized the meta-analysis, FPRP (false-positive report probability) test, and the TSA (trial sequential analysis) for the data pooling and the evaluation of statistical power. Totally, eight eligible articles were finally included. For KCNJ10 rs1130183, compared with population-based controls, a reduced epilepsy risk in cases was observed in models of allelic T vs. C, heterozygotic CT vs. CC, dominant CT + TT vs. CC, carrier T vs. C [all OR (odds ratio) <1, P < 0.05, Benjamini & Hochberg-adjusted P < 0.05, bonferroni-adjusted P < 0.05]. There were similar results in the subgroup analysis of \"Caucasian\". The positive conclusion was also statistically supported by the result of the FPRP test and TSA. Nevertheless, no statistically significant differences between epilepsy cases and negative controls were detected in any comparison of KCNJ101890532 and rs2486253. In summary, it is possible that the CT genotype of KCNJ10 rs1130183 is related to a reduced clinical epilepsy susceptibility, especially in Caucasians. However, more sample sizes are still required for a more robust conclusion in different populations, and more adjusted factors should be considered.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    KCNJ10 encodes the inward-rectifying potassium channel (Kir4.1) that is expressed in the brain, inner ear, and kidney. Loss-of-function mutations in KCNJ10 gene cause a complex syndrome consisting of epilepsy, ataxia, intellectual disability, sensorineural deafness, and tubulopathy (EAST/SeSAME syndrome). Patients with EAST/SeSAME syndrome display renal salt wasting and electrolyte imbalance that resemble the clinical features of impaired distal tubular salt transport in Gitelman\'s syndrome. A key distinguishing feature between these two conditions is the additional neurological (extrarenal) manifestations found in EAST/SeSAME syndrome. Recent reports have further expanded the clinical and mutational spectrum of KCNJ10-related disorders including non-syndromic early-onset cerebellar ataxia. Here, we describe a kindred of three affected siblings with early-onset ataxia, deafness, and progressive spasticity without other prominent clinical features. By using targeted next-generation sequencing, we have identified two novel missense variants, c.488G>A (p.G163D) and c.512G>A (p.R171Q), in the KCNJ10 gene that, in compound heterozygosis, cause this distinctive EAST/SeSAME phenotype in our family. Electrophysiological characterization of these two variants confirmed their pathogenicity. When expressed in CHO cells, the R171Q mutation resulted in 50% reduction of currents compared to wild-type KCNJ10 and G163D showed a complete loss of function. Co-expression of G163D and R171Q had a more pronounced effect on currents and membrane potential than R171Q alone but less severe than single expression of G163D. Moreover, the effect of the mutations seemed less pronounced in the presence of Kir5.1 (encoded by KCNJ16), with whom the renal Kir4.1 channels form heteromers. This partial functional rescue by co-expression with Kir5.1 might explain the lack of renal symptoms in the patients. This report illustrates that a spectrum of disorders with distinct clinical symptoms may result from mutations in different parts of KCNJ10, a gene initially associated only with the EAST/SeSAME syndrome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号