Tetrodotoxin‐Sensitive Neuronal‐Type Na+ Channels: A Novel and Druggable Target for Prevention of Atrial Fibrillation

MA Munger, Y Olğar, ML Koleske… - Journal of the …, 2020 - Am Heart Assoc
Journal of the American Heart Association, 2020Am Heart Assoc
Background Atrial 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 Results We studied AF in 2 models of Ca2+‐dependent disorders,
a murine model of catecholaminergic polymorphic ventricular tachycardia and a canine …
Background
Atrial 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 Results
We 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.
Conclusions
These 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.
Am Heart Assoc