Abstract

Loss-of-function mutations of the SCN5A gene encoding for the sodium channel α-subunit NaV1.5 result in the autosomal dominant hereditary disease Brugada Syndrome (BrS) with a high risk of sudden cardiac death in the adult. We here engineered human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) carrying the CRISPR/Cas9 introduced BrS-mutation p.A735V-NaV1.5 (g.2204C > T in exon 14 of SCN5A) as a novel model independent of patient´s genetic background. Recent studies raised concern regarding the use of hiPSC-CMs for studying adult-onset hereditary diseases due to cells’ immature phenotype. To tackle this concern, long-term cultivation of hiPSC-CMs on a stiff matrix (27–42 days) was applied to promote maturation. Patch clamp recordings of A735V mutated hiPSC-CMs revealed a substantially reduced upstroke velocity and sodium current density, a prominent rightward shift of the steady state activation curve and decelerated recovery from inactivation as compared to isogenic hiPSC-CMs controls. These observations were substantiated by a comparative study on mutant A735V-NaV1.5 channels heterologously expressed in HEK293T cells. In contrast to mutated hiPSC-CMs, a leftward shift of sodium channel inactivation was not observed in HEK293T, emphasizing the importance of investigating mechanisms of BrS in independent systems. Overall, our approach supports hiPSC-CMs’ relevance for investigating channelopathies in a dish.

Highlights

  • A pathophysiological reduction of the NaV1.5 current density associated with a prominent transient outward potassium current (Ito) is thought to cause the typical action potentials (APs) notch during early repolarization phase in myocardial cells

  • Low levels of Kir channel expression were found for hiPSC-CMs resulting in more depolarized resting membrane potentials poorly resembling the properties of native cardiomyocytes[17]

  • Equivalent cardiac differentiation of WT versus mutant clones was further confirmed by qRT-PCR for MYH6 (α-myosin heavy chain (MyHC) isoform; Fig. 1d, lower left) and SCN5A expression (Fig. 1d, lower right), suggesting that the g.2204C > T mutation did not impact on SCN5A mRNA abundance

Read more

Summary

Introduction

A pathophysiological reduction of the NaV1.5 current density associated with a prominent transient outward potassium current (Ito) is thought to cause the typical AP notch during early repolarization phase (phase 1) in myocardial cells. Extensive electrophysiological investigations of a disease-causing A735V-NaV1.5 mutation introduced into hiPSC-CMs were performed in comparison to both isogenic and non-genetically related hiPSC-CM controls (“wild type” WT) on the single cell level. To bridge the gap to such non-mammalian model, we introduced the A735V-NaV1.5 mutation into another heterologous system that is HEK293T cells. This cell line is well established for investigating channelopathies and provides a relevant comparison to our hiPSC-CM approach. Combining these technologies, we present a novel hiPSC-CM disease model for A735V-NaV1.5 mutation-based BrS, revealing the causative effect of such point mutation irrespective of patients genetic background

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call