[HTML][HTML] Post-transcriptional regulation of cardiac sodium channel gene SCN5A expression and function by miR-192-5p

Y Zhao, Y Huang, W Li, Z Wang, S Zhan, M Zhou… - … et Biophysica Acta (BBA …, 2015 - Elsevier
Y Zhao, Y Huang, W Li, Z Wang, S Zhan, M Zhou, Y Yao, Z Zeng, Y Hou, Q Chen, X Tu…
Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease, 2015Elsevier
The SCN5A gene encodes cardiac sodium channel Na v 1.5 and causes lethal ventricular
arrhythmias/sudden death and atrial fibrillation (AF) when mutated. MicroRNAs (miRNAs)
are important post-transcriptional regulators of gene expression, and involved in the
pathogenesis of many diseases. However, little is known about the regulation of SCN5A by
miRNAs. Here we reveal a novel post-transcriptional regulatory mechanism for expression
and function of SCN5A/Na v 1.5 via miR-192-5p. Bioinformatic analysis revealed that the 3 …
Abstract
The SCN5A gene encodes cardiac sodium channel Nav1.5 and causes lethal ventricular arrhythmias/sudden death and atrial fibrillation (AF) when mutated. MicroRNAs (miRNAs) are important post-transcriptional regulators of gene expression, and involved in the pathogenesis of many diseases. However, little is known about the regulation of SCN5A by miRNAs. Here we reveal a novel post-transcriptional regulatory mechanism for expression and function of SCN5A/Nav1.5 via miR-192-5p. Bioinformatic analysis revealed that the 3′-UTR of human and rhesus SCN5A, but not elephant, pig, rabbit, mouse, and rat SCN5A, contained a target binding site for miR-192-5p and dual luciferase reporter assays showed that the site was critical for down-regulation of human SCN5A. With Western blot assays and electrophysiological studies, we demonstrated that miR-192-5p significantly reduced expression of SCN5A and Nav1.5 as well as peak sodium current density INa generated by Nav1.5. Notably, in situ hybridization, immunohistochemistry and real-time qPCR analyses showed that miR-192-5p was up-regulated in tissue samples from AF patients, which was associated with down-regulation of SCN5A/Nav1.5. These results demonstrate an important post-transcriptional role of miR-192-5p in post-transcriptional regulation of Nav1.5, reveal a novel role of miR-192-5p in cardiac physiology and disease, and provide a new target for novel miRNA-based antiarrhythmic therapy for diseases with reduced INa.
Elsevier