首页> 外文期刊>Oxidative Medicine and Cellular Longevity >Thioridazine Induces Cardiotoxicity via Reactive Oxygen Species-Mediated hERG Channel Deficiency and L-Type Calcium Channel Activation
【24h】

Thioridazine Induces Cardiotoxicity via Reactive Oxygen Species-Mediated hERG Channel Deficiency and L-Type Calcium Channel Activation

机译:硫嗪通过反应性氧物种介导的HERG通道缺乏和L型钙通道激活诱导心脏毒性

获取原文
           

摘要

Thioridazine (THIO) is a phenothiazine derivative that is mainly used for the treatment of psychotic disorders. However, cardiac arrhythmias especially QT interval prolongation associated with the application of this compound have received serious attention after its introduction into clinical practice, and the mechanisms underlying the cardiotoxicity induced by THIO have not been well defined. The present study was aimed at exploring the long-term effects of THIO on the hERG and L-type calcium channels, both of which are relevant to the development of QT prolongation. The hERG current (IhERG) and the calcium current (ICa‐L) were measured by patch clamp techniques. Protein levels were analyzed by Western blot, and channel-chaperone interactions were determined by coimmunoprecipitation. Reactive oxygen species (ROS) were determined by flow cytometry and laser scanning confocal microscopy. Our results demonstrated that THIO induced hERG channel deficiency but did not alter channel kinetics. THIO promoted ROS production and stimulated endoplasmic reticulum (ER) stress and the related proteins. The ROS scavenger N-acetyl cysteine (NAC) significantly attenuated hERG reduction induced by THIO and abolished the upregulation of ER stress marker proteins. Meanwhile, THIO increased the degradation of hERG channels via disrupting hERG-Hsp70 interactions. The disordered hERG proteins were degraded in proteasomes after ubiquitin modification. On the other hand, THIO increased ICa‐L density and intracellular Ca2+ ([Ca2+]i) in neonatal rat ventricular cardiomyocytes (NRVMs). The specific CaMKII inhibitor KN-93 attenuated the intracellular Ca2+ overload, indicating that ROS-mediated CaMKII activation promoted calcium channel activation induced by THIO. Optical mapping analysis demonstrated the slowing effects of THIO on cardiac repolarization in mouse hearts. THIO significantly prolonged APD50 and APD90 and increased the incidence of early afterdepolarizations (EADs). In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), THIO also resulted in APD prolongation. In conclusion, dysfunction of hERG channel proteins and activation of L-type calcium channels via ROS production might be the ionic mechanisms for QT prolongation induced by THIO.
机译:硫嗪(Thio)是一种吩噻嗪衍生物,主要用于治疗精神病疾病。然而,心律失常尤其是与应用该化合物的应用相关的QT间隔延长,在引入临床实践后受到严重关注,并且硫虫诱导的心脏毒性的机制尚未明确定义。本研究旨在探索硫虫对HERG和L型钙通道的长期影响,这两者都与QT延长的发展有关。通过膜片钳技术测量Herg电流(Iherg)和钙电流(ICA-1)。通过蛋白质印迹分析蛋白质水平,通过CoimMunopectipation测定通道 - 伴侣相互作用。通过流式细胞术和激光扫描共聚焦显微镜测定反应性氧物质(ROS)。我们的结果表明,Thio诱发了Herg频道缺乏,但没有改变通道动力学。 Thio促进ROS生产和刺激的内质网(ER)应力和相关蛋白质。 ROS清除剂N-乙酰半胱氨酸(NAC)显着减弱了硫脲诱导的HERG减少,并废除了ER应激标记蛋白的上调。同时,硫醚通过破坏HERG-HSP70相互作用来增加HERG通道的降解。在泛素改性后,在蛋白质体中,无序的疱疹蛋白质在蛋白质体中降解。另一方面,硫醚在新生大鼠心室心肌细胞(NRVMS)中增加了ICA-L密度和细胞内Ca2 +([Ca2 +] I)。特异性Camkii抑制剂KN-93衰减细胞内Ca2 +过载,表明ROS介导的CAMKII活化促进了硫粒诱导的钙通道激活。光学映射分析表明了蒂奥对小鼠心中心脏复极性的效果。 Thio显着延长了APD50和APD90,增加了早期后掺杂的发病率(EADS)。在人诱导的多能干细胞衍生的心肌细胞(HIPSC-CM)中,硫族也导致APD延长。总之,HERG通道蛋白质的功能障碍和L型钙通道的活性通过ROS产生可能是硫氧化物诱导的QT延长的离子机制。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

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