Elevated EZH2 in ischemic heart disease epigenetically mediates suppression of NaV1.5 expression

https://doi.org/10.1016/j.yjmcc.2020.12.012Get rights and content

Highlights

  • EZH2 and H3K27me3 are increased while NaV1.5 expression is reduced in ischemic hearts.

  • Silencing expression or suppressing activity of EZH2 leads to a decrease of H3K27me3 and an elevation of NaV1.5.

  • Suppressing activity of EZH2 enhances Scn5a promoter activity by decreasing H3K27me3.

  • Suppressing activity of EZH2 increases Na+ channel activity.

  • EZH2-mediated suppression of NaV1.5 is one of the mechanisms underlying arrhythmias in patients with IHD.

Abstract

Suppression of the cardiac sodium channel NaV1.5 leads to fatal arrhythmias in ischemic heart disease (IHD). However, the transcriptional regulation of NaV1.5 in cardiac ischemia is still unclear. Our studies are aimed to investigate the expression of enhancer of zeste homolog 2 (EZH2) in IHD and regulation of cardiac NaV1.5 expression by EZH2. Human heart tissue was obtained from IHD and non-failing heart (NFH) patients; mouse heart tissue was obtained from the peri-infarct zone of hearts with myocardial infarction (MI) and hearts with a sham procedure. Protein and mRNA expression were measured by immunoblotting, immunostaining, and qRT-PCR. Protein-DNA binding and promoter activity were analyzed by ChIP-qPCR and luciferase assays, respectively. Na+ channel activity was assessed by whole-cell patch clamp recordings. EZH2 and H3K27me3 were increased while NaV1.5 expression was reduced in IHD hearts and in mouse MI hearts compared to the controls. Reduced NaV1.5 and increased EZH2 mRNA levels were observed in mouse MI hearts. A selective EZH2 inhibitor, GSK126 decreased H3K27me3 and elevated NaV1.5 in HL-1 cells. Silencing of EZH2 expression decreased H3K27me3 and increased NaV1.5 in these cells. EZH2 and H3K27me3 were enriched in the promoter regions of Scn5a and were decreased by treatment with EZH2 siRNA. GSK126 inhibited the enrichment of H3K27me3 in the Scn5a promoter and enhanced Scn5a transcriptional activity. GSK126 significantly increased Na+ channel activity. Taken together, EZH2 is increased in ischemic hearts and epigenetically suppresses Scn5a transcription by H3K27me3, leading to decreased NaV1.5 expression and Na+ channel activity underlying the pathogenesis of arrhythmias.

Introduction

Ischemic heart disease (IHD) is a leading cause of fatal cardiac arrhythmias including ventricular tachycardia (VT) and ventricular fibrillation (VF) [1]. Patients with severely depressed left ventricular ejection fraction are at a high risk of developing VT/VF and sudden cardiac death (SCD) [2]. In the ischemic myocardium, altered expression and function of ion channels affect cardiac electrical excitability, conduction, and automaticity [3]. In infarcted canine hearts, Na+ current density is significantly decreased in the epicardial border zone, leading to slow conduction velocity [4]. Encoded by SCN5A, the pore-forming NaV1.5 α-subunit of the cardiac Na+ channel governs cardiac depolarization [5]. Loss-of-function of NaV1.5 is associated with arrhythmogenicity in patients with cardiac ischemia and end-stage heart failure [6,7]. The decrease in Na+ current density is caused by reduced NaV1.5 protein levels on the cell membrane and/or altered Na+ channel kinetics [4]. The decrease of NaV1.5 expression in ischemic myocardium from mice with myocardial infarction (MI) suggests that the repression of Scn5a gene transcription contributes to the downregulation of NaV1.5 expression, suppression of Na+ channel activity, and cardiac arrhythmias [8].

Transcription factors, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), T-box transcription factor 5 (TBX5), and forkhead box protein O1 (FOXO1), are crucial regulators of NaV1.5 expression [[8], [9], [10], [11]]. We previously showed that enhanced β-catenin/T-cell factor 4 (TCF-4) signaling transcriptionally suppresses NaV1.5 expression [[12], [13], [14]] and increases arrhythmogenicity in mice [13]. High-throughput studies have revealed an essential role of epigenetic modulation, including DNA methylation and histone post-translational modification (PTM), underlying cardiovascular homeostasis [15]. Enhancer of zeste homolog 2 (EZH2), the functional enzymatic component of polycomb repressive complex 2 (PRC2), promotes heterochromatin formation via catalyzing trimethylation of histone 3 lysine 27 (H3K27me3), which leads to transcriptional suppression, serving as a potential barrier for cardiac regeneration [16,17]. In ischemic cardiomyopathy, EZH2 is elevated and regulates cardiac gene expression [18]. EZH2 functions as a methyltransferase to mediate Lamin A/C mutation-induced suppression of SCN5A transcription in human cardiac stem cells [19]. Interestingly, EZH2 interacts with β-catenin/TCF-4 to suppress target gene expressions, thereby regulating endothelial cell function [20]. Whether EZH2 epigenetically regulates Na+ channel expression in IHD warrants further investigation.

In this study, we report that EZH2 expression is increased in human IHD and mouse MI hearts. This upregulation correlates with increased H3K27me3 and decreased NaV1.5 expression. Inhibition of EZH2 methyltransferase activity or knockdown of EZH2 increases NaV1.5 expression at mRNA and protein levels in HL-1 cardiomyocytes. Also, we found that EZH2 is enriched in the promoter of Scn5a and is associated with a high level of H3K27me3. Both EZH2 expression and function are required to suppress the promoter activity of Scn5a. Moreover, GSK126, a selective EZH2 inhibitor, significantly increases Na+ channel activity in HL-1 cardiomyocytes. These findings demonstrate the indispensable role of EZH2 in the epigenetic downregulation of NaV1.5, highlighting EZH2 inhibition as a potential antiarrhythmic therapy in IHD.

Section snippets

Experimental protocol

Human heart tissue samples were composed of failing hearts with end-stage IHD hearts (n = 3) and non-failing hearts (n = 3). These samples were kindly offered by Dr. Shin Lin (Division of Cardiology, Department of Medicine, University of Washington). We obtained the institutional review board (IRB) approval at the University of Washington at Seattle. The patients' information was de-identified. Mice were handled according to the National Institutes of Health's Guide for the Care and Use of

NaV1.5 expression is decreased while EZH2 expression and H3K27me3 levels are increased in human hearts with IHD and mouse hearts with MI

We first measured the expression of EZH2, H3K27me3, and NaV1.5 in the hearts from patients with end-stage heart failure secondary to IHD and in human samples from non-failing hearts (NFHs). Consistent with previous reports [18], we found that the protein levels of EZH2 were increased in IHD hearts compared to NFHs. In IHD hearts, we also found increased H3K27me3 levels, a repressive chromatin mark of genes. Histone H3 expression was similar between IHD and NFH groups. We detected a significant

Discussion

In this study, we showed that elevated EZH2 expression and H3K27me3 levels were associated with decreased NaV1.5 expression in human IHD and mouse MI hearts. Inhibiting EZH2 activity or decreasing EZH2 expression increased NaV1.5 expression in HL-1 cardiomyocytes. EZH2 and H3K27me3 were enriched in the Scn5a promoter region, suppressing the promoter activity of Scn5a. GSK126 significantly increased the Na+ channel activity in HL-1 cells. The findings indicated that EZH2 mediated suppression of

Conclusions

Here we show that EZH2 is increased in IHD and plays an important role in the regulation of cardiac NaV1.5 expression and Na+ channel function. Our findings may shed light on the epigenetic regulation of NaV1.5 expression underlying the development of arrhythmias related to cardiac ischemia, thereby providing a new candidate molecular target for antiarrhythmic therapy.

Sources of funding

This work was supported by the National Institutes of Health, United States (grants R01HL122793 [to Dr. Xu] and R01HL111480 [to Dr. Li]), American Heart Association, United States (grant 19TPA34910069 [to Dr. Xu]) and the Department of Laboratory Medicine and Pathology (to Dr. Xu).

Declaration of Competing Interest

None declared.

Acknowledgments

We thank Drs. William C. Claycomb at Louisiana State University Health Science Center and Hideko Kasahara at the Department of Physiology and Functional Genomics, University of Florida, United States for kindly providing HL-1 cells and Scn5a promoter-Luc plasmid, respectively.

References (33)

  • J. Chen et al.

    Effectiveness of beta-blocker therapy after acute myocardial infarction in elderly patients with chronic obstructive pulmonary disease or asthma

    J. Am. Coll. Cardiol.

    (2001)
  • D.P. Zipes et al.

    Sudden cardiac death

    Circulation.

    (1998)
  • E. Carmeliet

    Cardiac ionic currents and acute ischemia: from channels to arrhythmias

    Physiol. Rev.

    (1999)
  • S. Baba et al.

    Remodeling in cells from different regions of the reentrant circuit during ventricular tachycardia

    Circulation.

    (2005)
  • C.A. Remme et al.

    Sodium channel (Dys)function and cardiac arrhythmias

    Cardiovasc. Ther.

    (2010)
  • L.L. Shang et al.

    NF-κB-dependent transcriptional regulation of the cardiac scn5a sodium channel by angiotensin II

    Am. J. Phys. Cell Phys.

    (2008)
  • Cited by (7)

    • Role of transcriptional cofactors in cardiovascular diseases

      2024, Biochemical and Biophysical Research Communications
    • Targeting miR-30d reverses pathological cardiac hypertrophy

      2022, eBioMedicine
      Citation Excerpt :

      In the current study, we found that EZH2 was upregulated in pathological cardiac hypertrophy models and served as an epigenetic inhibitor of miR-30d. It should be noted that EZH2 has been found to be up‐regulated in multiple types of cardiovascular diseases, such as dilated cardiomyopathy, pathological cardiac hypertrophy, myocardial ischemia-reperfusion, coronary heart disease, atrial fibrosis, ischemic heart disease.44-49 H3K27me3 enrichment within the miR-30d promotor region has been reported in breast cancer cell, malignant peripheral nerve sheath tumors and brain tissues.50-52

    • The Roles of Histone Lysine Methyltransferases in Heart Development and Disease

      2023, Journal of Cardiovascular Development and Disease
    View all citing articles on Scopus
    1

    L Zhao and T You equally contributed to this work.

    View full text