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.
Keywords: EZH2, H3K27me3, NaV1.5, Na+ channel, IHD
1. 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[8–11]. We previously showed that enhanced β-catenin/T-cell factor 4 (TCF-4) signaling transcriptionally suppresses NaV1.5 expression[12–14] 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 Sen5a 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.
2. Materials and methods
2.1. 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 Laboratory Animals. Experimental procedures were approved by the Institutional Animal Care and Use Committee of the University of Washington. Detailed materials and methods in this study are included in the Online Data Supplement; the primers used in the studies are summarized in Supplemental Table 1.
2.2. Statistical analysis
Data are presented as mean±SEM (standard error of the mean). The unpaired 2-tailed Student’s t-test was used to analyze the difference between two groups; one-way ANOVA followed by post hoc Tukey’s test was used to compare the data from multiple groups. A value of P<0.05 was considered statistically significant. Statistical analysis was performed using Prism software (Graphpad, version 7.0)
3. Results
3.1. 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 decrease of NaV1.5 protein levels in IHD hearts, which was negatively correlated with EZH2 and H3K27me3 (Fig. 1A and B). One week after the ligation of the left anterior descending coronary artery (LAD) in MI mice, the peri-infarct zone (PIZ, 2.0 mm surrounding the infarcted zone) and the corresponding ventricular myocardium region of the sham-operated hearts were sampled for experiments. Western blotting demonstrated increased EZH2 and H3K27me3, and unchanged Histone H3 with decreased NaV1.5 protein in the PIZ compared to the sham control (Fig. 1C–D). Increased EZH2 and decreased NaV1.5 mRNA levels were observed in the PIZ (Fig. 1E). These results indicated that enhanced EZH2 expression and H3K27me3 levels are correlated with the downregulation of NaV1.5 in ischemic hearts.
Fig. 1. EZH2 expression and H3K27me3 levels are increased while NaV1.5 is decreased in the myocardium from patients with ischemic heart disease (IHD) and from the peri-infarct zone (PIZ) of mice with myocardial infarction (MI).

(A) Western blotting of the protein extract from the left ventricular myocardium of patients with IHD and those with non-failing hearts (NFHs) were collected. (B) Immunoblotted bands were normalized to GAPDH (n=3 for NFH and n=3 for IHD). *P<0.05, **P<0.01. (C) Western blots were performed on the protein extracts from the PIZs of mouse MI hearts and the areas of mouse hearts with sham procedure using antibodies against NaV1.5, EZH2, H3K27me3, Histone H3 and GAPDH. (D) Quantification of immunoblotted bands by densitometry normalized to GAPDH (n=3 for sham and n=3 for MI). (E) Quantitative real-time qRT-PCR analysis of NaV1.5 and EZH2 mRNA normalized to GAPDH mRNA of mouse PIZ and sham (n=4 for sham; n=3 for MI). Columns are mean±SEM and data were analyzed by Student’s t-test. *P<0.05, **P<0.01 (B, D and E).
3.2. Inhibition of EZH2 activity or decrease of EZH2 expression reduces H3K27me3 and increases expression of NaV1.5 in HL-1 cardiomyocytes
To understand whether the inhibition of EZH2 catalytic activity or decrease of EZH2 expression affects NaV1.5 expression, we first inhibited the activity of EZH2 histone methyltransferase using GSK126, a selective EZH2 inhibitor[21]. Inhibition of EZH2 was confirmed by a concentration-dependent decrease of H3K27me3 levels and unchanged histone H3 in HL-1 cells 72 hours (hrs) after treatment with 1 μM and 3 μM GSK126 (Fig. 2A and B). We found that protein levels of NaV1.5 were significantly elevated in HL-1 cardiomyocytes treated with GSK126 compared to cells with vehicle control (DMSO) (Fig. 2A and B). GSK126 produced a remarkable upregulation of NaV1.5 mRNA expression in HL-1 cells (Fig. 2C). The protein levels of EZH2 were not significantly different between DMSO- and GSK126-treated HL-1 cells (Fig. 2B). Immunofluorescence staining was performed on the HL-1 cells treated with 1 μM GSK126 using antibodies against NaV1.5, EZH2 and H3K27me3. Decreased nuclear H3K27me3 in HL-1 cardiomyocytes treated with GSK126 was observed without alteration in EZH2 nuclear levels (Fig. 3A and C). Fluorescence intensity quantification using imageJ (National Institutes of Health, NIH) showed that GSK126 significantly increased NaV1.5 signal (Fig. 3B); GSK126 significantly increased nuclear H3K27me3 fluorescence signal without altering EZH2 staining intensity (Fig. 3B and D). Therefore, the methyltransferase activity of EZH2 is indispensable for the suppression of NaV1.5.
Fig. 2. Inhibition of EZH2 methyltransferase activity by GSK126 reduces H3K27me3 level and increases NaV1.5 expression in HL-1 cardiomyocytes.

(A) Western blots were performed on the protein extract from HL-1 cardiomyocytes treated with the EZH2 inhibitor GSK126 (1 μM or 3 μM) or DMSO (vehicle) for 72 hours and the membranes were immunoblotted using antibodies against NaV1.5, EZH2, H3K27me3, Histone H3 and GAPDH. (B) Quantification of immunoblotted bands by densitometry normalized to GAPDH (n=3 for DMSO, n=3 for GSK126 1 μM and n=3 for GSK126 3 μM). (C) Quantitative real-time RT-PCR analysis of NaV1.5 mRNA normalized to GAPDH mRNA (n=10 for DMSO, n=10 for GSK126 1 μM and n=7 for GSK126 3 μM). Columns are mean±SEM and data were analyzed by one-way ANOVA with Tukey’s post hoc comparison test. *P<0.05, **P<0.01 (B and C).
Fig. 3. Inhibition of EZH2 methyltransferase activity by GSK126 increases the expression of NaV1.5 and reduces the nuclear enrichment of H3K27me3 in HL-1 cardiomyocytes.

(A) Representative immunofluorescence images of NaV1.5 (green), EZH2 (red), and 4′,6-diamidino-2-phenylindole (Hoechst 33342, blue) staining in HL-1 cardiomyocytes treated with GSK126 (1 μM) or DMSO for 72 hrs. (B) Quantitation of global NaV1.5 and nuclear EZH2 fluorescence intensity in GSK126 group normalized to DMSO’s in HL-1 cells using imageJ (NIH) (n=7 cells for DMSO and n=6 cells for GSK126). (C) Representative immunofluorescence images of H3K27me3 (green), EZH2 (red), and Hoechst 33342 (blue) staining in HL-1 cells treated with GSK126 (1 μM) or DMSO. (D) Quantitation of nuclear H3K27me3 and EZH2 fluorescence intensity normalized to DMSO’s in HL-1 cells (n=9 HPFs [high-power fields] for DMSO and n=10 HPFs for GSK126). Columns are mean±SEM and data were analyzed by Student’s t-test (B and D). *P<0.05, **P<0.01 (B and D).
To explore whether a decrease of EZH2 expression correlates with the upregulation of NaV1.5, we utilized siRNA to knockdown EZH2 in HL-1 cardiomyocytes. After 48 hrs transfection of siRNA, there was a significant reduction in EZH2 protein in HL-1 cells. Elevated NaV1.5 expression was observed and was accompanied by reduced H3K27me3 levels and unchanged histone H3 in HL-1 cells transfected with siRNA targeting EZH2, compared to those treated with the scrambled siRNA (SC siRNA) (Fig. 4A and B). Real time qRT-PCR demonstrated that EZH2 siRNA significantly reduced the mRNA levels of EZH2 but increased that of NaV1.5 (Fig. 4C). Immunofluorescence staining was performed on the HL-1 cells treated with SC siRNA and EZH2 siRNA for 48 hrs using antibodies against NaV1.5, EZH2 and H3K27me3. Images showed that EZH2 siRNA had an effect similar to GSK126 on NaV1.5 and H3K27me3 signals (Fig. 5A and C) and decreased nuclear EZH2 staining intensity (Fig. 5A and C). ImageJ (NIH) analyses showed that decreased EZH2 by siRNA (Fig. 5B and D) significantly increased NaV1.5 signal (Fig. 5B) and decreased H3K27me3 fluorescence intensity (Fig. 5D) compared to the data obtained from SC siRNA. The results showed that both expression and methyltransferase activity of EZH2 play a pivotal role in the regulation of NaV1.5 expression.
Fig. 4. Knockdown of EZH2 by siRNA reduces H3K27me3 levels and increases NaV1.5 expression in HL-1 cardiomyocytes.

(A) Western blots were performed on the protein extract from HL-1 cardiomyocytes treated with the EZH2 siRNA (30 nM) or the scrambled (SC) siRNA for 48 hours, and the membranes were immunoblotted using antibodies against NaV1.5, EZH2, H3K27me3, Histone H3 and GAPDH. (B) Quantification of immunoblotted bands by densitometry normalized to GAPDH (n=3 for SC siRNA and n=3 for EZH2 siRNA). (C) Quantitative real-time qRT-PCR analysis of NaV1.5 mRNA normalized to GAPDH mRNA (n=6 for SC siRNA and n=6 for EZH2 siRNA). Columns are mean±SEM and data were analyzed by the Student’s t-test (B and C). **P<0.01 (B and C).
Fig. 5. Knockdown of EZH2 by siRNA increases the expression of NaV1.5 and reduces the nuclear enrichment of H3K27me3 in HL-1 cardiomyocytes.

(A) Representative immunofluorescence images of NaV1.5 (green), EZH2 (red), and Hoechst 33342 (blue) staining in HL-1 cardiomyocytes treated with EZH2 siRNA (30 nM) or the SC siRNA. (B) Quantitation of global NaV1.5 and nuclear EZH2 fluorescence intensity normalized to DMSO’s in HL-1 cells (n=8 cells for SC siRNA; n=9 cells for EZH2 siRNA). *P<0.05, **P<0.01. (C) Representative immunofluorescence images of H3K27me3 (green), EZH2 (red), and Hoechst 33342 (blue) staining in HL-1 cells treated with EZH2 siRNA or SC siRNA. (D) Quantitation of nuclear H3K27me3 (n=6 cells for EZH2 siRNA, n=6 cells For SC siRNA) and EZH2 fluorescence intensity normalized to DMSO’s in HL-1 cells (n=8 HPFs for EZH2 siRNA, n=8 HPFs for SC siRNA). Columns are mean±SEM and data were analyzed by Student’s t-test (B and D). *P<0.05, **P<0.01 (B and D).
3.3. EZH2 is recruited in the promoter of Scn5a and inhibits the promoter activity by increasing H3K27me3
We have shown that EZH2 expression is negatively correlated with NaV1.5 expression in ischemic hearts and HL-1 cardiomyocytes. These findings raise the possibility of EZH2 regulating the Scn5a promoter through H3K27me3. We screened for potential binding sites and confirmed the enrichment of EZH2 or H3K27me3 at conserved regions in the promoter region of human SCN5A and mouse Scn5a (Fig. 6A and B)[16,22,23]. Next, we determined the recruitment of EZH2 to the Scn5a promoter in HL-1 cardiomyocytes. ChIP-qPCR demonstrated the enrichment of EZH2 in the mouse Scn5a promoter region. A concomitant enrichment of H3K27me3 was detected and significantly reduced by the EZH2 inhibitor, GSK126, which did not alter the enrichment of EZH2 (Fig. 6C). Silencing of EZH2 expression by siRNA reduced the enrichment of both EZH2 and H3K27me3 in the Scn5a promoter region in HL-1 cells (Fig. 6D). A luciferase assay showed that inhibited EZH2 activity by GSK126 or decreased EZH2 expression by siRNA significantly increased the Scn5a promoter activity in HL-1 cells (Fig. 6E). These results suggest that EZH2 is a negative controller of Scn5a transcription.
Fig. 6. Enrichment of EZH2 and H3K27me3 in mouse Scn5a and human SCN5A promoter region and their regulation of Scn5a promoter activity in HL-1 cardiomyocytes.

(A) The enrichment of EZH2 and H3K27me3 (blue bars) in mouse cardiac Scn5a (light yellow box) promoter region was obtained by data mining using the Cistrome Data Browser and WashU Epigenome Browser. The CpG island (orange box) and gene conservation with human SCN5A (violet bands) are displayed. The original datasets are GSM1902476 and GSM742287. (B) The enrichment of H3K27me3 (blue bars) in mouse Scn5a (light yellow box) promoter region was obtained by data mining using the Cistrome Data Browser and WashU Epigenome Browser. The CpG island (orange box) and gene conservation with human cardiac SCN5A (violet bands) are displayed. The original dataset is GSM621450. (C) ChIP-qPCR analysis of EZH2 and H3K27me3 in the promoter region of Scn5a in HL-1 cells treated with GSK126 (1 μM) or DMSO (n=3 for IgG, n=3 for DMSO and n=3 for GSK126). **P<0.01. (D) ChIP-qPCR analysis of EZH2 and H3K27me3 in the promoter region of Scn5a in HL-1 cells treated with EZH2 siRNA (30 nM) or SC siRNA (30 nM) (n=3 for IgG, n=3 for SC siRNA and n=3 for EZH2 siRNA). (E) Transcriptional activity of Scn5a promoter analyzed by the luciferase assay in HL-1 cells treated with GSK126 (1 μM) (n=5 for DMSO and n=6 for GSK126) or EZH2 siRNA (n=5 for SC siRNA and n=5 for EZH2 siRNA). Columns are mean±SEM and data were compared using Student’s t-test (C, D and E). *P<0.05, **P<0.01 (C, D, and E).
3.4. Inhibition of EZH2 activity increases Na+ channel activity in HL-1 cardiomyocytes
Whole-cell Na+ currents were recorded from HL-1 cells after treatment with GSK126 (1 μM) or DMSO (Fig. 7A) for 48 hrs. The values of cell capacitances were not significantly different between cells with DMSO treatment (34.9±1.9, n=14) and cells with GSK126 treatment (37.2±3.8, n=12). The current-voltage curve illustrated that inhibiting EZH2 activity by GSK126 markedly augmented the peak Na+ current densities at depolarization voltages from −45 mV to +35 mV (Fig. 7B). We did not detect a significant impact of GSK126 on Na+ channel kinetics (Fig. 8A–G and Supplemental Table 2). Taken together, an EZH2 inhibitor increases Na+ channel activity without affecting the channel’s kinetics.
Fig. 7. EZH2 inhibitor, GSK126 increases Na+ channel activity in HL-1 cardiomyocytes.

(A) Characteristic voltage-gated Na+ currents recorded at depolarization voltage levels from −80 mV to +50 mV using a whole-cell patch clamp in HL-1 cells treated with GSK126 (1 μM) or the vehicle (DMSO) for 72 hrs. (B) I-V curves of peak Na+ current densities obtained from the peak currents divided by individual capacitances in HL-1 cells (black: DMSO, n=13 and blue: GSK126, n=11). Peak current densities are mean±SEM and data were analyzed by Student’s t-test. *P<0.05, **P<0.01.
Fig. 8. Na+ channel kinetics is not changed by EZH2 inhibitor, GSK126 in HL-1 cardiomyocytes.

(A and B) Time constants of activation and inactivation wereobtained from single exponential fit on the individual current traces at the voltages from −40 mV to −10 mV, and they were not significantly different between DMSO (n = 13) and GSK126 (1 μM) (n = 11). (C, D and E). Voltage-dependent steady-state activation (C, DMSO, n=13 and GSK126, n=11) and inactivation (D and E, DMSO, n=9 and GSK126, n=11) of Na+ channel were not changed by GSK126, and V1/2 and k slope factor were not significantly different as described in table 1 in the supplemental data. (F) Typical Na+ current traces of recovery from inactivation at voltage levels from −120 mV to −40 mV in HL-1 cells treated with DMSO (black) and 1 μM GSK126 (blue) for 72 h, respectively. (G) Normalized recovery data for peak INa were plotted, and they were well described by a single exponential function. T and k slope were not significantly different between DMSO (n=17) and GSK126 (n=12) groups as described in table 1 in the supplemental data.
4. 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 NaV1.5 channel underlies cardiac arrhythmogenesis in patients with IHD.
We found that EZH2 expression was increased in the myocardium of IHD as reported by another group[18]. How EZH2 expression is regulated in ischemic hearts is unknown. EZH2 expression is regulated by various oncogenic transcription factors such as Myc and E2F and tumor suppressor miRNAs[24]. Recent work from Guo A. et al demonstrated that cardiac stress-induced N-terminal truncate of excitation-contraction (E-C) coupling protein junctoophilin-2 (JP2NT) functions as a transcription factor leading to cardioprotective transcriptional reprograming and specifically, it attenuates hypertrophic remodeling and the progression of heart failure. Overexpression of JP2NT significantly caused modulation of of multiple genes in adult mouse cardiomyocytes and among them. EZH2 expression was significantly decreased, indicating that JP2NT may regulate E2H2 transcription[25]. Further experiments are warranted to determine if transcription factors such as JP2NT and c-Myc mediate EZH2-suppressing NaV1.5 expression in the myocardium of IHD.
A recent study combining genome-wide methylation and RNA-seq analysis revealed that enhanced EZH2 epigenetically inhibits KLF15 expression by coordinating with differential DNA methylation in its promoter, contributing to myocardial metabolic reprogramming in human ischemic cardiomyopathy[18]. The emerging role of EZH2 led us to explore its potential effect on the epigenetic control of cardiac expression of NaV1.5, the downregulation of which predisposes to cardiac arrhythmias. We detected an increase in H3K27me3, an established marker of gene suppression[16]. In-silico promoter analysis unveiled the enrichment of EZH2 and H3K27me3 in the proximal region of human SCN5A and mouse Scn5a promoters [16,22]. NaV1.5 expression was decreased in human IHD. The increase of NaV1.5 expression and Na+ current density by inhibiting EZH2 suggested that histone methylation by EZH2 contributes to the suppression of Scn5a transcription. The dynamic control of SCN5A promoter activity by EZH2 may dictate the change of NaV1.5 and the development of arrhythmias in IHD.
We found that enhanced β-catenin/TCF-4 signaling transcriptionally suppress expression of NaV1.5 [12–14]. Interestingly, β-catenin contributes to the PRC2 complex to enable EZH2 binding to promoter regions of claudin-5. PTP and VWF thereby, affecting endothelial cell differentiation and vessel maturation[20]. Also. β-catenin/TCF-4 interacts with EZH2 to affect glioma tumorigenesis as well as survival and metastasis in chemoresistant triple-negative breast cancer[26,27]. These findings raise the possibility that β-catenin/TCF4 and EZH2 interplay to transcriptionally suppress NaV1.5 expression. In future research plans, we will use mouse models with cardiac-specific loss of EZH2 expression, gain of β-catenin function and/or heart infarction to obtain direct evidence.
Genetic analysis of the human SCN5A promoter revealed mutations and polymorphisms associated with various arrhythmia phenotypes[28]. Changes in epigenetic modulation, as well as transcription factor interaction, may contribute to the genetic disorders. A remote SNP rs6801957 in a cardiac enhancer that spatially interacts with the promoter of SCN5A is associated with reduced NaV1.5 expression and slowed conduction in Brugada syndrome. Decreased methylation of the SCN5A promoter has been associated with increased NaV1.5 providing a rescuing phenotype in the probands[29]. One study showed that a K219T-Lamin A/C mutation has been suggested to recruit PRC2 to the SCN5A promoter and inhibits NaV1.5 expression in a laminopathy[19]. By direct interaction with DNA methyltransferase, enriched EZH2 during cardiac ischemia may mediate the methylation of unmasked CpG island at the SCN5A promoter region, which is spatially overlapped with EZH2 enriched sites in the genome [16]. Further studies are necessary to determine the comprehensive role of EZH2 in regulating SCN5A promoter activity.
Compared to cancer studies, a much lower concentration of GSK126 was sufficient to increase Scn5a promoter activity, NaV1.5 expression, and Na+ channel function in our experiments. GSK126 was well tolerated by participants in the Phase I study[30]. Our findings raised a potential therapeutic implication of GSK126 for cardiac arrhythmias. GSK126 may be considered as an adjunct therapy in IHD patients, especially in certain scenarios when β-blockers and other antiarrhythmic medicine are contraindicated[31]. Interestingly, late embryonic deletion of EZH2 conveyed a mild effect in cardiac structure and function, indicating its safety as a therapeutic target in developed heart[16]. Our observation showed only a low level of EZH2 expression in the NFHs. These findings suggest that EZH2 as a feasible antiarrhythmic target by GSK126 in IHD. The role of EZH2 as a candidate antiarrhythmic target will be investigated in future studies using larger animal MI models.
Our studies indicate that suppression of SCN5A transcription is an important mechanism of decreased cardiac Na+ channel activity in ischemic hearts. However, mRNA splicing variants should not be neglected. In the human ischemic cardiomyopathy, splicing factors LUC7L3 and RBM25 are elevated[32] and they cause SCN5A mRNA splicing variants[32,33]. The two SCN5A mRNA variants reach greater that >50% of the total SCN5A mRNA and do not produce functional Na+ channels[32,33]; in fact, they function in a dominant negative manner[32,33]. Therefore, both decreased transcriptional regulation and mRNA splicing variants of SCN5A contribute to the decrease of Na+ channel activity in ischemic cardiomyopathy.
5. 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.
Supplementary Material
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 in the Scn5a promoter.
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.
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.
Sources of Funding
This work was supported by the National Institutes of Health, Unites States (grants R01HL122793 [to Dr Xu] and R01HL111480 [to Dr Li]), American Heart Association, Unites States (grant 19TPA34910069 [to Dr Xu]) and the Department of Laboratory Medicine and Pathology (to Dr Xu).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of interest statement: The authors have no conflicts to disclose
Conflict of Interest
None declared
References
- [1].Zipes DP, Wellens HJJ. Sudden cardiac death. Circulation. 1998; 98: 2334–2351. [DOI] [PubMed] [Google Scholar]
- [2].Rouleau JL, Talajic M, Sussex B, Potvin L, Warnica W, Davies RF, et al. Myocardial infarction patients in the 1990s - Their risk factors, stratification and survival in Canada: The Canadian Assessment of Myocardial Infarction (CAMI) study. J Am Coll Cardiol. 1996; 27: 1119–1127. [DOI] [PubMed] [Google Scholar]
- [3].Carmeliet E Cardiac ionic currents and acute ischemia: From channels to arrhythmias. Physiol Rev. 1999; 79: 917–1017. [DOI] [PubMed] [Google Scholar]
- [4].Baba S, Dun W, Cabo C, Boyden PA. Remodeling in cells from different regions of the reentrant circuit during ventricular tachycardia. Circulation. 2005; 112:2386–2396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Remme CA, Bezzina CR. Sodium channel (Dys)function and cardiac arrhythmias. Cardiovasc Ther. 2010; 28: 287–294. [DOI] [PubMed] [Google Scholar]
- [6].Beltran-Alvarez P, Tarradas A, Chiva C, Pérez-Serra A, Batlle M, Pérez-Villa F, et al. Identification of N-terminal protein acetylation and arginine methylation of the voltage-gated sodium channel in end-stage heart failure human heart. Curr Ther Res - Clin Exp. 2014; 76: 126–129. [DOI] [PubMed] [Google Scholar]
- [7].Hu D, Viskin S, Oliva A, Carrier T, Cordeiro JM, Barajas-Martinez H, et al. Novel mutation in the SCN5A gene associated with arrhythmic storm development during acute myocardial infarction. Hear Rhythm. 2007; 4: 1072–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Cai B, Wang N, Mao W, You T, Lu Y, Li X, et al. Deletion of FoxO1 leads to shortening of QRS by increasing Na+ channel activity through enhanced expression of both cardiac NaV1.5 and β3 subunit. J Mol Cell Cardiol. 2014; 74: 297–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Shang LL, Sanyal S, Pfahnl AE, Jiao Z, Allen J, Liu H, et al. NF-κB-dependent transcriptional regulation of the cardiac scn5a sodium channel by angiotensin II. Am J Physiol - Cell Physiol. 2008; 294:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Arnolds DE, Liu F, Fahrenbach JP, Kim GH, Schillinger KJ, Smemo S, et al. TBX5 drives Scn5a expression to regulate cardiac conduction system function. J Clin Invest. 2012; 122: 2509–2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Mao W, You T, Ye B, Li X, Dong HH, Hill JA, et al. Reactive oxygen species suppress cardiac Na V1.5 expression through Foxo1. PLoS One. 2012; 7: e32738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Zhao L, Sun L, Lu Y, Li F, Xu H. A small-molecule LF3 abrogates β-catenin/TCF4-mediated suppression of NaV1.5 expression in HL-1 cardiomyocytes. J Mol Cell Cardiol. 2019; 135: 90–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Wang N, Huo R, Cai B, Lu Y, Ye B, Li X, et al. Activation of Wnt/β-catenin signaling by hydrogen peroxide transcriptionally inhibits NaV1.5 expression. Free Radic Biol Med. 2016; 96: 34–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Huo R, Hu C, Zhao L, Sun L, Wang N, Lu Y, et al. Enhancement of β-catenin/T-cell factor 4 signaling causes susceptibility to cardiac arrhythmia by suppressing NaV1.5 expression in mice. Hear Rhythm. 2019; 16: 1720–1728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Liu CF, Tang WHW. Epigenetics in Cardiac Hypertrophy and Heart Failure. JACC Basic to Transl Sci. 2019; 4: 976–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].He A, Ma Q, Cao J, Von Gise A, Zhou P, Xie H, et al. Polycomb repressive complex 2 regulates normal development of the mouse heart. Circ Res. 2012; 110: 406–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Talkhabi M, Zonooz ER, Baharvand H. Boosters and barriers for direct cardiac reprogramming. Life Sci. 2017; 178: 70–86. [DOI] [PubMed] [Google Scholar]
- [18].Pepin ME, Ha CM, Crossman DK, Litovsky SH, Varambally S, Barchue JP, et al. Genome-wide DNA methylation encodes cardiac transcriptional reprogramming in human ischemic heart failure. Lab Investig. 2019; 99: 371–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Salvarani N, Crasto S, Miragoli M, Bertero A, Paulis M, Kunderfranco P, et al. The K219T-Lamin mutation induces conduction defects through epigenetic inhibition of SCN5A in human cardiac laminopathy. Nat Commun. 2019; 10: 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Morini MF, Giampietro C, Corada M, Pisati F, Lavarone E, Cunha SI, et al. VE-cadherin-mediated epigenetic regulation of endothelial gene expression. Circ Res. 2018; 122: 231–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].McCabe MT, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012; 492: 108–112. [DOI] [PubMed] [Google Scholar]
- [22].Ai S, Peng Y, Li C, Gu F, Yu X, Yue Y, et al. EED orchestration of heart maturation through interaction with HDACS is H3K27ME3-independent. Elife. 2017; 6:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Bernstein BE, Stamatoyannopoulos JA, Costello JF, Ren B, Milosavljevic A, Meissner A, et al. The NIH roadmap epigenomics mapping consortium. Nat Biotechnol. 2010; 28: 1045–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Yamaguchi H, Hung MC. Regulation and role of EZH2 in cancer. Cancer Res Treat. 2014; 46: 209–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Guo A, Wang Y, Chen B, Wang Y, Yuan J, Zhang L, et al. E-C coupling structural protein junctophilin-2 encodes a stress-adaptive transcription regulator. Science (80-). 2018; 362:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Ma L, Lin K, Chang G, Chen Y, Yue C, Guo Q, et al. Aberrant activation of b-catenin signaling drives glioma tumorigenesis via USP1-mediated stabilization of EZH2. Cancer Res. 2019; 79: 72–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Ayachi I El, Fatima I, Wend P, Alva-Ornelas JA, Runke S, Kuenzinger WL, et al. The WNT10B network is associated with survival and metastases in chemoresistant triple-negative breast cancer. Cancer Res. 2019; 79: 982–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Yagihara N, Watanabe H, Barnett P, Duboscq-Bidot L, Thomas AC, Yang P, et al. Variants in the SCN5A Promoter Associated With Various Arrhythmia Phenotypes. J Am Heart Assoc. 2016; 5:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Matsumura H, Nakano Y, Ochi H, Onohara Y, Sairaku A, Tokuyama T, et al. H558R, a common SCN5A polymorphism, modifies the clinical phenotype of Brugada syndrome by modulating DNA methylation of SCN5A promoters. J Biomed Sci. 2017; 24:. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Yap TA, Winter JN, Giulino-Roth L, Longley J, Lopez J, Michot JM, et al. Phase I study of the novel enhancer of zeste homolog 2 (EZH2) inhibitor GSK2816126 in patients with advanced hematologic and solid tumors. Clin Cancer Res. 2019; 25: 7331–7339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Chen J, Radford MJ, Wang Y, Marciniak TA, Krumholz HM. Effectiveness of beta-blocker therapy after acute myocardial infarction in elderly patients with chronic obstructive pulmonary disease or asthma. J Am Coll Cardiol. 2001; 37: 1950–1956. [DOI] [PubMed] [Google Scholar]
- [32].Shang LL, Pfahnl AE, Sanyal S, Jiao Z, Allen J, Banach K, et al. Human heart failure is associated with abnormal C-terminal splicing variants in the cardiac sodium channel. Circ Res. 2007; 101: 1146–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Gao G, Xie A, Huang SC, Zhou A, Zhang J, Herman AM, et al. Role of RBM25/LUC7L3 in abnormal cardiac sodium channel splicing regulation in human heart failure. Circulation. 2011; 124: 1124–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
