首页> 美国卫生研究院文献>Proceedings of the National Academy of Sciences of the United States of America >PNAS Plus: Structure-guided examination of the mechanogating mechanism of PIEZO2
【2h】

PNAS Plus: Structure-guided examination of the mechanogating mechanism of PIEZO2

机译:PNAS Plus:PIEZO2机械化机制的结构指导检查

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Piezo channels are mechanically activated ion channels that confer mechanosensitivity to a variety of different cell types. Piezos oligomerize as propeller-shaped homotrimers that are thought to locally curve the membrane into spherical domes that project into the cell. While several studies have identified domains and amino acids that control important properties such as ion permeability and selectivity as well as inactivation kinetics and voltage sensitivity, only little is known about intraprotein interactions that govern mechanosensitivity—the most unique feature of PIEZOs. Here we used site-directed mutagenesis and patch-clamp recordings to investigate the mechanogating mechanism of PIEZO2. We demonstrate that charged amino acids at the interface between the beam domain—i.e., a long α-helix that protrudes from the intracellular side of the “propeller” blade toward the inner vestibule of the channel—and the C-terminal domain (CTD) as well as hydrophobic interactions between the highly conserved Y2807 of the CTD and pore-lining helices are required to ensure normal mechanosensitivity of PIEZO2. Moreover, single-channel recordings indicate that a previously unrecognized intrinsically disordered domain located adjacent to the beam acts as a cytosolic plug that limits ion permeation possibly by clogging the inner vestibule of both PIEZO1 and PIEZO2. Thus, we have identified several intraprotein domain interfaces that control the mechanical activation of PIEZO1 and PIEZO2 and which might thus serve as promising targets for drugs that modulate the mechanosensitivity of Piezo channels.
机译:压电通道是机械活化的离子通道,可赋予多种不同细胞类型机械敏感性。压电低聚体为螺旋桨状的均三聚体,被认为可以将膜局部弯曲成伸入细胞的球形圆顶。尽管几项研究已经确定了控制重要特性(例如离子渗透性和选择性以及失活动力学和电压敏感性)的结构域和氨基酸,但对于控制机械敏感性(PIEZO的最独特特征)的蛋白内相互作用知之甚少。在这里,我们使用定点诱变和膜片钳记录来研究PIEZO2的机械化机制。我们证明了束域(即从“螺旋桨”叶片的细胞内侧向通道内前庭突出的长α螺旋)与C端域(CTD)之间的界面处带电氨基酸为了确保PIEZO2的正常机械敏感性,需要高度保守的CTD Y2807和孔衬螺旋之间的疏水作用以及疏水作用。此外,单通道记录表明,先前无法识别的与电子束相邻的固有无序结构域充当了胞质栓塞,可能通过堵塞PIEZO1和PIEZO2的内部前庭来限制离子渗透。因此,我们已经确定了几个蛋白内结构域接口,这些接口控制PIEZO1和PIEZO2的机械激活,因此可以作为调节压电通道机械敏感性的药物的有希望的靶标。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号