当前位置: X-MOL 学术J. Am. Heart Assoc. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
Tetrodotoxin-Sensitive Neuronal-Type Na+ Channels: A Novel and Druggable Target for Prevention of Atrial Fibrillation.
Journal of the American Heart Association ( IF 5.0 ) Pub Date : 2020-05-29 , DOI: 10.1161/jaha.119.015119
Mark A Munger 1 , Yusuf Olğar 2, 3 , Megan L Koleske 2, 3 , Heather L Struckman 4 , Jessica Mandrioli 5 , Qing Lou 2, 6 , Ingrid Bonila 2, 6 , Kibum Kim 7 , Roberto Ramos Mondragon 8 , Silvia G Priori 9, 10 , Pompeo Volpe 11 , Héctor H Valdivia 8 , Joseph Biskupiak 7 , Cynthia A Carnes 2, 3 , Rengasayee Veeraraghavan 2, 4 , Sándor Györke 2, 6 , Przemysław B Radwański 2, 3, 6
Affiliation  

BackgroundAtrial fibrillation (AF) is a comorbidity associated with heart failure and catecholaminergic polymorphic ventricular tachycardia. Despite the Ca2+‐dependent nature of both of these pathologies, AF often responds to Na+ channel blockers. We investigated how targeting interdependent Na+/Ca2+ dysregulation might prevent focal activity and control AF.Methods and ResultsWe studied AF in 2 models of Ca2+‐dependent disorders, a murine model of catecholaminergic polymorphic ventricular tachycardia and a canine model of chronic tachypacing‐induced heart failure. Imaging studies revealed close association of neuronal‐type Na+ channels (nNav) with ryanodine receptors and Na+/Ca2+ exchanger. Catecholamine stimulation induced cellular and in vivo atrial arrhythmias in wild‐type mice only during pharmacological augmentation of nNav activity. In contrast, catecholamine stimulation alone was sufficient to elicit atrial arrhythmias in catecholaminergic polymorphic ventricular tachycardia mice and failing canine atria. Importantly, these were abolished by acute nNav inhibition (tetrodotoxin or riluzole) implicating Na+/Ca2+ dysregulation in AF. These findings were then tested in 2 nonrandomized retrospective cohorts: an amyotrophic lateral sclerosis clinic and an academic medical center. Riluzole‐treated patients adjusted for baseline characteristics evidenced significantly lower incidence of arrhythmias including new‐onset AF, supporting the preclinical results.ConclusionsThese data suggest that nNaVs mediate Na+‐Ca2+ crosstalk within nanodomains containing Ca2+ release machinery and, thereby, contribute to AF triggers. Disruption of this mechanism by nNav inhibition can effectively prevent AF arising from diverse causes.

中文翻译:

河豚毒素敏感的神经元型 Na+ 通道:预防心房颤动的新型药物靶点。

背景房颤 (AF) 是一种与心力衰竭和儿茶酚胺能多形性室性心动过速相关的合并症。尽管这两种病理都具有Ca 2+依赖性,但 AF 经常对 Na +通道阻滞剂有反应。我们研究了如何针对相互依赖的Na + /钙2+失调可能会阻止焦点活动和控制AF.Methods和ResultsWe研究AF 2种机型的Ca 2+依赖性疾病,儿茶酚胺多形性室性心动过速的小鼠模型和慢性犬模型快速起搏引起的心力衰竭。影像学研究揭示了神经元型 Na +通道 (nNa v) 与兰尼碱受体和 Na + /Ca 2+交换器。儿茶酚胺刺激仅在 nNa v活性的药理学增强期间在野生型小鼠中诱导细胞和体内房性心律失常。相比之下,单独的儿茶酚胺刺激足以引起儿茶酚胺能多形性室性心动过速小鼠和犬心房衰竭的房性心律失常。重要的是,这些被涉及 Na + /Ca 2+ 的急性 nNa v抑制(河豚毒素或利鲁唑)消除了AF 失调。然后在 2 个非随机回顾性队列中测试了这些发现:肌萎缩侧索硬化诊所和学术医疗中心。校正基线特征利鲁唑治疗的患者证明心律失常包括新发AF的显著发生率较低,支持临床前results.ConclusionsThese数据表明NNA V小号居间的Na + -Ca 2+含有Ca纳米畴内的串扰2+释放机械,因此,有助于 AF 触发。通过 nNa v抑制破坏这种机制可以有效防止多种原因引起的 AF。
更新日期:2020-05-29
down
wechat
bug