Graphical abstract graphic file with name fx1.jpg [31]Open in a new tab Highlights * • Nav1.5 deficiency enhances cardiac fibroblast activity and attenuates miR-452-5p * • MiR-452-5p regulates fibrogenesis by targeting the TGF-β/SMAD4 axis * • AAV miR-452-5p reduces cardiac fibrosis and improves heart failure in rats __________________________________________________________________ Cardiovascular medicine; Molecular biology; Cell biology Introduction Cardiac arrhythmia affects 33 million people globally and is the prevalent cause of myocardial fibrosis leading to heart failure (HF).[32]^1 Predominantly, myocardial fibrosis occurs due to abnormalities in ion channels and structural organization. The former contributes to the development of cardiac fibrosis by affecting the initiation and propagation of electrical stimuli, for instance, the reduced I[Na] significantly increased collagen deposition in SCN5A-knockout mice.[33]^2 SCN5A gene produces α-subunit of the cardiac sodium voltage-gated channel (Nav1.5) arbitrates the rapid Na^+ influx, which is crucial for the upstroke of action potential and excitation of cardiac cells.[34]^3^,[35]^4 Structural changes in Nav1.5 channel syndicate a spectrum of arrhythmic disorders that lead to sudden cardiac death. Studies reported that the loss of SCN5A gene expression in the human and heterozygous SCN5A mouse models favors the tissue remodeling vulnerability of the heart, and these structural abnormalities are secondary to cardiac sodium channelopathies.[36]^5^,[37]^6^,[38]^7^,[39]^8 In addition to this, under diminished expression of Nav1.5, distinctive fibrosis has been reported in calcineurin-induced murine cardiac hypertrophic ventricles and Cx43 knockout mice.[40]^9^,[41]^10 Sodium channel dysfunction-dependent onset of cardiac fibrosis and collagen production requires the activation of the transforming growth factor β (TGF-β) pathway. The inhibition of I[Na] triggered the abnormal upregulation of TGF-β receptors,[42]^11 and SCN5A-knockout mice also exhibited increased expression of TGF-β transcripts.[43]^12 Under activated TGF-β, fibroblasts differentiate into myofibroblasts which foster extracellular matrix (ECM) secretion. ECM coupled with paracrine factors (TGF-β) and electrical modulators further damages the myocyte-fibroblast’s communication, which then leads to irregular conduction propagation with more signal blockage.[44]^13 Furthermore, increased collagen release/deposition in ECM increases the myocardium’s proclivity to cardiac dysfunction and HF.[45]^14^,[46]^15^,[47]^16 Another study showed that SCN5A decisively contributes to the transdifferentiating of human atrial fibroblasts suggesting the ineluctable involvement of SCN5A mutation in fibroblast activity.[48]^17 Although the reduced expression of SCN5A plays an essential role in the development of cardiac fibrosis, there is no study on how the defective SCN5A increases the pro-fibrotic TGF-β signaling in human cardiac fibroblasts (HCF). MicroRNAs (miRNAs) contain 22 nucleotide sequences, crucial as profibrotic or anti-fibrotic by binding to 3ˊ untranslated region (3ˊUTR) of target mRNA.[49]^18^,[50]^19 The plethora of miRNAs has been associated with SCN5A channelopathies and HF, vital regulators in myocardial fibrosis by regulating ECM synthesis and cytokines secretion.[51]^20 For instance, miR-24 functionally engaged in the regulation of SCN5A expression in patients with HF and miR-210-5p reduces cardiac fibrosis by interfering with TGF-β type I receptor binding in rats.[52]^21^,[53]^22 In addition, miR-452-5p is a putative pathological factor, and significantly downregulated in the cardiac tissues of patients with hypertrophic cardiomyopathy and upregulated in aortic stenosis-induced HF rats.[54]^23^,[55]^24 However, the involvement of miR-452-5p in cardiac fibrosis has not been reported yet, and no research has been done to explore the regulatory function of miR-452-5p in SCN5A-dependent cardiac remodeling. This study aimed to investigate how the downregulation of SCN5A gene regulates fibrogenesis and clarifies the underlying mechanisms. In the current study, we identified miR-452-5p as a key regulator of SCN5A-induced cardiac fibrosis via miRNA sequencing. We further explored the effect of miR-452-5p on fibroblast differentiation, migration, and the potential molecular mechanism of TGF-β signaling mediation. Our study provides insight into the SCN5A knockdown-induced fibrosis and miR-452-5p as a potential therapeutic target. Results SCN5A knockdown promotes fibrosis in human cardiac fibroblasts After establishing the SCN5A knockdown cell model, the expression of Nav1.5 was measured via western blot. There was less protein expression of SCN5A by 50% than control HCF without affecting fibroblast morphology ([56]Figures 1A and 1B). To investigate the role of SCN5A knockdown in cardiac fibrosis, the expression level of known pathological cardiac fibrosis markers was checked in SCN5A knockdown and control HCF by western blot. SCN5A knockdown HCF significantly increased the protein expressions of pro-Collagen type 1A1, α-SMA, and fibronectin ([57]Figure 1C). Fibroblasts' ability to synthesize and secrete precursors of fibrillar collagen changes with cardiac remodeling.[58]^25 As excessive collagen secretion can lead to disproportionate fibrosis, we assessed the difference in collagen secretion capacity between SCN5A knockdown HCF and control HCF. The soluble collagen type-I level in conditioned media was 4-fold higher in SCN5A knockdown HCF compared to control HCF ([59]Figure 1D). These results divulge the crucial commitments of SCN5A knockdown in cardiac fibrosis. Figure 1. [60]Figure 1 [61]Open in a new tab SCN5A knockdown promotes the expression of fibrogenic signaling (A) Upper panel: Knockdown of SCN5A gene in human cardiac fibroblasts by targeting SCN5A gene using shRNA lentivirus. Lower panel: fibroblast grown after SCN5A gene knockdown and morphology analyzed microscopically, Scale bar 350 μm (representative pictures shown). (B) The protein expression of Nav1.5 after the knockdown of the SCN5A gene in HCF represents a 50% decrease in Nav1.5 protein expression. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, n = 5 independent experiments. (C) Representatives immunoblot and quantitative analysis showing the expression of pro-Col1agen 1A1, α-SMA, and fibronectin in SCN5A knockdown and control HCF normalized with the internal control group. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, ∗∗∗p < 0.001, n = 6 independent experiments. (D) There were higher soluble collagen-type 1 levels measured in a conditioned medium (serum-free) of SCN5A knockdown HCF than that from control cells Data are expressed as mean ± SEM, paired t-test, ∗∗∗p < 0.001, n = 6 independent experiments. SCN5A shRNA: SCN5A knockdown HCF. Differential expression of miR-452-5p, functional enrichment and validation in SCN5A knockdown HCF We performed hypothesis-free micro-RNA sequencing to identify the possible mechanistic link between the loss of SCN5A and the upregulation of fibrotic genes in HCF. The transcriptomic analysis revealed that a total of 410 microRNAs were expressed in SCN5A knockdown HCF, among which 53 were significantly expressed as shown in the heatmap ([62]Figure S1). A Total of 13 miRNAs were differentially expressed including three miRNAs markedly upregulated and ten were significantly downregulated ([63]Figure 2A). Next, we performed target prediction of upregulated and downregulated miRNAs using multiple tools and identified most common genes predicted by at least two tools ([64]Table S1). These genes were then subjected to GO analysis. ([65]Figure 2B). The KEGG pathway enrichment analysis revealed the most enriched pathways were TGF-β signaling pathway (path:hsa04350), cellular senescence (path:hsa04218), hippo signaling pathway (path:hsa04390), diabetic cardiomyopathy (path:hsa05415), and relaxin signaling pathway (path:hsa04926) ([66]Figure 2C). Furthermore, the differentially expressed miRNAs were verified through stem-loop quantitative real-time PCR. Overall, the top three upregulated and the top two downregulated miRNAs in SCN5A knockdown HCF were selected for validation ([67]Figure 2D). The expression of examined miRNAs showed partial agreement with the RNA-seq data. miR-34a-5p and miR-199b-5p were significantly upregulated, whereas miR-335-5p and miR-452-5p were strikingly lower in SCN5A knockdown HCF, with miR-452-5p being the significantly lowest expressed in SCN5A knockdown HCF. Consequently, we selected miR-452-5p for further functional analysis, and the miRNA-mRNA network was constructed using datasets comprising miRNA-target gene-binding data and miRNA-mRNA interactions using Cytoscape 3.9.1 ([68]Figure 2E). The network analysis unveiled a regulatory abundance of miR-452-5p, with numerous target genes displaying enrichment in TGF-β signaling pathway and genes related to ECM interaction. Specifically, SMAD2, SMAD4, TGF-βRI, and TGF-βRII were notably direct mediators of the TGF-β pathway. Further studies were performed to determine the role of miR-452-5p in SCN5A knockdown-induced fibrosis. Figure 2. [69]Figure 2 [70]Open in a new tab Maladaptive TGF-β signaling is upregulated in SCN5A knockdown HCF (A) Differentially expressed miRs against their enrichment score in SCN5A knockdown HCF. Data plotted against enrichment score, p values determined via DESeq2 R package. (B) GO enrichment of differentially expressed miRs target genes (including UP/Downregulated miRs) are presented, top 10 significantly enriched GO term (biological process, cellular component, and molecular function) branches are presented. The GO terms were plotted against fold enrichment values and -log p-values (Fisher’s exact test, p < 0.05). (C) Top 5 up-regulated (red bars) and top 3 down-regulated (blue bars) significantly enriched pathways in KEGG pathway analysis of differentially expressed genes plotted against -log FDR values. Their corresponding p-values calculated by the Fisher’s exact test are mentioned in parentheses in front of the bars. (D) qRT-PCR analysis of micro-RNAs (hsa-miR-1307-5p, hsa-miR-34a-5p, hsa-miR-199b-5p, hsa-miR-335, has-miR-452-5p) identified by small RNA sequencing analysis in SCN5A knockdown and control HCF. Shrek green represents upregulated miRNAs, blue bars represent downregulated miRNAs in SCN5A knockdown HCF, while black bars represent control cells. Data are expressed as mean ± SEM, one-way ANOVA, ∗p < 0.05, ∗∗∗p < 0.001, n = 7 independent biological repeats. SCN5A shRNA: SCN5A knockdown HCF. miR-NTC: miR negative control. (E) Rectangular nodes represent targets mRNA and Diamond nodes represent miRNAs. The network displays mRNAs with the highest predictive score. Edges with arrows indicate the inhibitory effect on targeted mRNA. Light blue nodes show mRNAs involved in the TGF-β signaling pathway. Green nodes illustrate genes related to ECM interaction. Blue and purple nodes show the genes involved in the FGF receptor signaling pathway and positive regulation of cell proliferation respectively. Gray nodes are for genes without specific interpretation. GO: Gene Ontology, KEGG: Kyoto Encyclopedia of Genes and Genome. miR-452-5p attenuated TGF-β signaling in SCN5A knockdown HCF Based on the bioinformatic study, we assessed TGF-β expression in SCN5A knockdown and control HCF. Our western blot and qRT-PCR analysis revealed a significant increase in both TGF-β protein expression and transcripts in SCN5A knockdown HCF compared to the control group ([71]Figures 3A and 3B). Furthermore, the ELISA consolidated the 4-fold increase in secretory form of TGF-β in conditioned media (24 h) from SCN5A knockdown HCF as compared to control ([72]Figure 3C). To validate the functional relevance of miR-452-5p in TGF-β signaling, we treated the SCN5A knockdown HCF with miR-452-5p-mimic (10 nM) or miR negative control (miR-NTC) (10 nM) to increase miR-452-5p expression ([73]Figure 3D), and measured the TGF-βRI, TGF-βRII expression levels because these are TGF-β receptors and their activation leads to phosphorylation of downstream signaling mediators, such as SMAD2 and SMAD3 (canonical TGF-β signaling pathway),[74]^26 as predicted in miRNA-mRNA network. Interestingly, we found that TGF-β1 transcripts and expression levels of TGF-β1, TGF-βRI, and TGF-βRII, were significantly suppressed by the miR-452-5p mimic than the miR-NTC group which was initially activated in SCN5A knockdown HCF. In line with these results, the phospho-SMAD2/3 expression was noticeably reduced in the miR-452-5p mimic-treated group whereas the total smad2/3 remained unchanged ([75]Figures 3E and 3F). These findings suggest that miR-452-5p actively repressed the TGF-β signaling, which contributed to fibrosis. Figure 3. [76]Figure 3 [77]Open in a new tab miR-452-5p mimic regulates canonical TGF-β signaling pathway in SCN5A knockdown HCF (A) Representative immunoblot showing the expression of TGF-β1 expression in SCN5A knockdown and control HCF normalized with the internal control group. Data are expressed as mean ± SEM, paired t-test, ∗∗∗p < 0.001, n = 6 independent experiments. (B) The expression of TGF-β1 mRNA measured via qRT-PCR in SCN5A knockdown and control HCF normalized with the internal control. Data are expressed as mean ± SEM, paired t-test, ∗∗∗p < 0.001, n = 6 independent experiments. (C) ELISA was used to measure the total secreted TGF-β in a conditioned medium (serum-free) of SCN5A knockdown HCF than that from control cells. Data are expressed as mean ± SEM, paired t-test, ∗∗∗∗p < 0.0001, n = 6 independent experiments. (D) MiR-452-5p mimic was used to increase the expression of miR-452-5p in SCN5A knockdown HCF and miR-452-5p expression was measured through qRT-PCR. Data are expressed as mean ± SEM, paired t-test, ∗∗∗p < 0.001, n = 6 independent experiments. (E) TGF-β1 mRNA measured in SCN5A knockdown HCF treated with miR-452-5p and miR-NTC and control HCF via qRT-PCR. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, ∗∗∗∗p < 0.0001, n = 6 independent experiments. (F) Immunoblots and quantitative analysis of TGF-β1, TGF-βRI, TGF-βRII, total SMAD 2/3, and phospho-SMAD 2/3 in SCN5A knockdown HCF as compared to control and SCN5A knockdown HCF treated without or with miR-NTC and miR-452-5p mimic. n = 6 experiments. Data are expressed as mean ± SEM, paired t-test, ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, n = 6 independent experiments. SCN5A shRNA: SCN5A knockdown HCF. miR-NTC: miR negative control. miR-452-5p inhibited fibrosis-related genes, differentiation, and migration of SCN5A knockdown HCF but not restore sodium current (I[Na]) To assess the anti-fibrotic effect of miR-452-5p on SCN5A knockdown HCF, we performed immunoblot analysis of SCN5A knockdown HCF treated with miR-452-5p mimic (10 nM) and miR-NTC (10 nM). Our results showed a significant decrease in pro-Collagen type 1A1 and fibronectin levels in the miR-452-mimic treated SCN5A knockdown HCF compared to the miR-NTC group ([78]Figure 4A). In accordance, the concentration of soluble collagen type-I in the medium of miR-452-5p mimic treated SCN5A knockdown HCF reflected the downregulation at the protein level, unlike the miR-NTC group ([79]Figure 4B). Furthermore, our immunofluorescence analyses indicated that SCN5A knockdown dramatically promoted the formation of differentiation-related stress fibers, whereas the miR-452-5p mimic treatment showed a marked decrease in α-SMA fibers as compared to the miR-NTC group consistent with the α-SMA protein expression in miR-452-5p mimic treated SCN5A knockdown HCF ([80]Figures 4C and 4D). Increased myofibroblast differentiation is accompanied by fibroblast cell-migration. To corroborate the role of miR-452-5p in fibroblast migration, we treated SCN5A knockdown HCF with either miR-452-5p mimic or miR-NTC and assessed migration. The wound healing assay showed that SCN5A knockdown enhanced fibroblast migration, while treatment with the miR-452-5p mimic clearly restrained the migration of SCN5A knockdown HCF as compared to the miR-NTC group ([81]Figure 4E). We also investigated whether miR-452-5p could reverse the decreased sodium current (I[Na]) and late sodium current (I[Na-Late]) resulting from knockdown of SCN5A gene in HCF. Our findings indicate that miR-452-5p was unable to restore the decreased sodium currents ([82]Figure 5), suggesting that the anti-fibrotic potential of miR-452-5p is exclusively molecular in nature but does not have any effect on I[Na] under the loss of SCN5A activity. Figure 4. [83]Figure 4 [84]Open in a new tab MiR-452-5p mimic restored the fibrogenic phenotype in SCN5A knockdown HCF (A) Immunoblot and quantitative analysis of the expression in protein levels of pro-Collagen type 1A1 and fibronectin in control, and SCN5A knockdown HCF, miR-NTC, and miR-452-5p-mimic groups. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, n = 6 independent experiments). (B) Soluble collagen-type1 level measured in conditioned medium from SCN5A knockdown HCF as compared to control and SCN5A knockdown HCF treated without or with miR-NTC and miR-452-5p mimic. Data are expressed as mean ± SEM, paired t-test, ∗p < 0.05, ∗∗p < 0.01, n = 5 independent experiments. (C) Immunohistochemistry shows increased expression of α-SMA in SCN5A knockdown HCF and this expression was significantly reduced upon the treatment of miR-452-5p mimic as compared to both control and miR-NTC groups. α-SMA (green), DAPI (blue). Scale bar: 90 μm. (D) Immunoblots and quantitative analysis of α-SMA in SCN5A knockdown HCF as compared to control and SCN5A knockdown HCF, miR-NTC, and miR-452-5p-mimic groups. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01. ∗∗∗p < 0.001, n = 6 independent experiments. (E) Representative images of migration at 0 h and 10 h post-wounding. Scale bar: 300 μm. Graph showing the cell migration distances (μm) of control, SCN5A knockdown HCF, miR-NTC, and miR-452-5p mimic treated groups. Data are expressed as mean ± SEM, paired t-test, ∗∗∗p < 0.001, n = 10 independent experiments. SCN5A shRNA: SCN5A knockdown HCF. miR-NTC: miR negative control. Figure 5. [85]Figure 5 [86]Open in a new tab Effect of miR-452-5p on the sodium current (I[Na]) and late sodium current (I[Na-Late]) in SCN5A knockdown HCF (A) Representative current traces and current-voltage (I–V) relationships of I[Na] from control HCF (n = 9), SCN5A knockdown HCF (n = 10), and SCN5A knockdown HCF transfected with miR-452-5p mimic (n = 10). Data are expressed as mean ± SEM, unpaired t-test, ∗p < 0.05 Control versus SCN5A knockdown HCF, #p < 0.05, ##p < 0.01 Control versus miR-452-5p mimic-treated SCN5A knockdown HCF. (B) Representative trace and average data of I[Na-Late] from control HCF (n = 9), SCN5A knockdown HCF (n = 7), and miR-452-5p mimic transfected SCN5A knockdown HCF (n = 11). Data are represented mean ± SEM, unpaired t-test, ∗∗p < 0.01,∗p < 0.05. MiR-452-5p directly targets SMAD4 to mitigate fibrogenesis in SCN5A knockdown human cardiac fibroblasts To determine the mechanism through which miR-452-5p regulates the TGF-β signaling pathway, analysis using miRanda, predicted pivotal binding sites between miR-452-5p and the SMAD4 mRNA duplex ([87]Figure S2). In addition, the prediction of binding sites in the 3ˊUTR sequence substantiates that among miR-452-5p target genes, 3ˊUTR of SMAD4 contains the most and multiple conserved complementary sites for the seed region of miR-452-5p, corroborating the recruitment of SMAD4 via miR-452-5p ([88]Figure 6A). To verify this hypothesis, we evaluated the expression of SMAD4 in our SCN5A knockdown cell model. The western blot results disclosed that the SMAD4 expression was significantly higher in SCN5A knockdown HCF ([89]Figure 6B). To identify the substantial binding and influence of miR-452-5p on target, we performed 3′UTR reporter assay ([90]Figure 6C). Overexpression of miR-452-5p dramatically amortized the luciferase activity of the SMAD4 3ˊUTR construct and diminished SMAD4 protein level ([91]Figures 6D and 6E), indicating that miR-452-5p may interfere with the nuclear translocation of SMAD4. To confirm this, we performed the nuclear and cytosolic fractionation assay from SCN5A knockdown HCF, miR-452-5p mimic, and miR-NTC treated HCF. As shown in [92]Figure 6F, SMAD4 was translocated into the nucleus of SCN5A knockdown HCF as compared to the control, while the nuclear accumulation of SMAD4 was attenuated in the miR-452-5p mimic group, when compared with the miR-NTC group. Figure 6. [93]Figure 6 [94]Open in a new tab SMAD4: a direct target of miR-452-5p (A) Sequence was predicted from the online database (Target Scan) and complementary targets of 3ˊUTR of human SMAD4 gene with hsa-miR-452-5p. (B) Quantitative analysis and immunoblot representing the protein expression level of SMAD4 in SCN5A knockdown and control HCF. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, n = 5 independent experiments. (C) Profile of pEZX-MT06 plasmid for dual-luciferase reporter assay to detect the interaction of miR-452-5p and 3ˊUTR of SMAD4 in SCN5A knockdown vs. control HCF and miR-NTC vs. miR-452-5P mimic in SCN5A knockdown HCF. (D) The luciferase activity was assessed in SCN5A knockdown and control HCF co-transfected without or with miR-NTC and miR-452-5p mimic (10 nM). Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, n = 5 independent experiments. (E and F) Representative immunoblots and quantitative analysis showing the expression of SMAD4 in whole cell lysate, nuclear, and cytoplasmic fractions from SCN5A knockdown HCF as compared to control and miR-NTC vs. miR-452-5p mimic groups revealed that SMAD4 nuclear translocation blocked by miR-452-5p mimic. Lamin B and GAPDH indicate nuclear and cytoplasmic fractions, respectively. Data are represented as mean ± SEM, paired t-test. ∗∗p < 0.01, ∗∗∗p < 0.001, n = 5–6 experiments. SCN5A shRNA: SCN5A knockdown HCF. miR-NTC: miR negative control, fLuc: firefly luciferase gene, rLuc: Renilla luciferase gene, CMV-promotor: cytomegalovirus promotor, Amp^R: ampicillin resistance gene, ori: origin of replication. Treatment with miR-452-5p mimic mitigates fibrosis in isoproterenol-induced HF HF is a common heart disease with enhanced cardiac fibrosis and acquired down-regulation of Nav1.5.[95]^21^,[96]^27^,[97]^28^,[98]^29 To examine whether miR-452-5p could rescue the fibrogenic phenotype under HF conditions, we generated HF rat model by subcutaneous injection of isoproterenol (100 mg/kg) at the age of 10 weeks. After 2 weeks rats were given the intravenous injection (tail vein) of AAV9-miR-452-5p (3.0 x10^10 genome copies per rat, once a week for 2 weeks) and saline as negative control once a week. After 2 weeks, rats were sacrificed, and the heart and the serum were collected ([99]Figure 7A). We found that AAV miR452 markedly recovered the fibrotic area in the left ventricular region of the heart as well as the heart weight/body weight ratio ([100]Figures 7B and 7C). Improved cardiac function including restored mean blood pressure, ejection fraction, fraction shortening, left ventricular diameter both in systole and diastole, and thickness of intraventricular septum was observed in AAV miR452 rats compared to HF rats ([101]Figure 7D). As shown in [102]Figure 8A, the expression of Nav1.5 in the ventricular tissues of HF rats were significantly reduced. We also identified the downregulation of miR-452-5p in the HF rats, and it was increased after the delivery of miR-452-5p mimic via AAV9 ([103]Figure 8B). Consistent with in vitro findings, over-expression of miR452-5p reversed fibrosis in AAV miR452 group ([104]Figure 8C). Impaired TGF-β signaling in HF was significantly retrieved after treatment with AAV-miR452, SMAD4 expression was also attenuated in AAV9 miR-452 group ([105]Figure 8D). We conducted liver and kidney function tests at the end of the experiments to examine the potential toxicity of the viral vector. There were similar ALT, AST, and BUN in different groups, which implies the non-toxicity of AAV9 ([106]Figure S3). Collectively, these data illustrate the notable significance of in vivo administered miR-452 in ameliorating the pathophysiology of the HF animal model. Figure 7. [107]Figure 7 [108]Open in a new tab Effect of AAV miR452 on cardiac function in isoproterenol-induced HF (A) Schematic illustration of in vivo induction of HF via subcutaneous injection of isoproterenol 100 mg/kg (once a week for 2 weeks) and treatment with AAV miR452 (3.0 x10^10 genome copies per rat via tail vein, once a week for 2 weeks), control rats received normal saline for 2 weeks. (B) Cardiac pictures from control, HF, and AAV miR452 groups. (C) The ratio of heart to body weight (mg/g) in control, HF, and AAV miR452 groups. Data are presented as mean ± SEM, one-way ANOVA followed by Tukey’s multiple comparison test, ∗p < 0.05, ∗∗p < 0.01, n = 5-6 independent experiments. (D) Upper Panel: Representative M-mode echocardiographic images. Lower Panel: Averaged data presenting the results of mean blood pressure (BP, mmHg), percentage of ejection fraction (EF%), left ventricle fractional shortening (FS%), left ventricular internal diameter in systole (LVIDs, mm), left ventricular internal diameter in diastole (LVIDd, mm), and intraventricular septal diameter (IVSd, mm) in control, HF, and AAV miR452 groups. Data are represented as mean ± SEM, one-way ANOVA followed by Tukey’s multiple comparison test, ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, n = 5–6 independent experiments. Figure 8. [109]Figure 8 [110]Open in a new tab Systemic delivery of AAV miR452 ameliorates fibrosis in isoproterenol-induced HF rats (A) Nav1.5 protein expression in left ventricular tissues of HF and control rats. Data are expressed as mean ± SEM, paired t-test, ∗∗p < 0.01, n = 3 independent experiments. (B) MiR-452-5p mimic delivery through AAV9 increased the miR-452 expression in left ventricular tissues which was initially reduced after HF development as compared to control, measured via qRT-PCR. Data are expressed as mean ± SEM, one-way ANOVA followed by Tukey’s multiple comparison test, ∗∗∗p < 0.001, n = 5–6 independent experiments. (C) Immunoblots and quantitative analysis of pro-collagen type 1a1, fibronectin, α-SMA in LV tissues of HF, and AAV miR452 compared with control. Data are expressed as mean ± SEM, one-way ANOVA followed by Tukey’s multiple comparison test, ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, n = 5–6 independent experiments. (D) Immunoblots and quantitative analysis of TGF-β signaling including TGF-β1, TGF-βRI, RII, p-SMAD2/3, SMAD4 in LV tissues of HF (induced by subcutaneous injection of isoproterenol 100 mg/kg once a week for 2 weeks) and HF rats treated with AAV miR452 (3.0 x10^10 genome copies per rat via tail vein, once a week for 2 weeks) compared with control (received normal saline). Data are represented as mean ± SEM, one-way ANOVA followed by Tukey’s multiple comparison test, ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, n = 5–6 independent experiments. Discussion Cardiac fibrosis provokes pathological changes thereby promoting arrhythmia and HF.[111]^30 Despite the quintessential advancement in the acknowledgment of cardiac fibrosis and SCN5A channelopathies, the