MicroRNA-194 inhibits isoproterenol-induced chronic cardiac hypertrophy via targeting CnA/NFATc2 signaling in H9c2 cells
Original Article

MicroRNA-194 inhibits isoproterenol-induced chronic cardiac hypertrophy via targeting CnA/NFATc2 signaling in H9c2 cells

Jian Wang#, Yucheng An#, Jun Lin, Gang Tang

Department of Cardiology, The Third Affiliated Hospital of Chongqing Medical University (Jieer Hospital), Chongqing, China

Contributions: (I) Conception and design: J Wang, Y An; (II) Administrative support: G Tang; (III) Provision of study materials or patients: J Wang, Y An, J Lin; (IV) Collection and assembly of data: J Wang, Y An; (V) Data analysis and interpretation: J Wang, Y An; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Gang Tang. Department of Cardiology, The Third Affiliated Hospital of Chongqing Medical University (Jieer Hospital), No. 1, Shuanghu Branch Road, Yubei District, Chongqing 401120, China. Email: 650712@hospital.cqmu.edu.cn.

Background: This study explored the effects of microRNA(miR)-194 on chronic cardiac hypertrophy (CH) induced by isoproterenol (ISO). The potential mechanism through regulation of the calcineurin A (CnA)/nuclear factor of activated T cells (NFAT) c2 pathway was investigated in the rat cardiomyoblast cell line H9c2.

Methods: H9c2 cells were treated with ISO to induce cardiomyocyte hypertrophy to simulate CH in vitro. The cell surface area was assessed by phalloidin staining. The expression of miR-194, CnA mRNA, and CnA protein were assessed. Furthermore, the cellular localization of the NFATc2 protein after induction of CH was detected. The relationship between miR-194 and the CnA mRNA 3'-untranslated region (UTR) was verified by dual luciferase report assays. By constructing cardiomyocyte cell models with low expression of miR-194 and/or CnA, the effects of miR-194 and CnA on the localization of the NFATc2 protein and cell hypertrophy was investigated. Rescue experiments were conducted to analyze whether overexpression of miR-194 could alleviate the cell hypertrophy induced by ISO.

Results: The results demonstrated that induction with ISO significantly increased the surface area of H9c2 cells. After induction, the expression of miR-194 decreased, while both CnA mRNA and protein expression increased. Furthermore, the nuclear translocation of NFATc2 was obvious. MiR-194 bound to the 3'-UTR of CnA mRNA and inhibited CnA protein expression. Inhibition of miR-194 expression activated NFATc2 protein expression and increased the H9c2 cell surface area. After CnA expression was disturbed, hypertrophy induced by miR-194 down-regulation was blocked. In addition, overexpression of miR-194 significantly alleviated cell hypertrophy and activation of the CnA/NFATc2 pathway caused by ISO.

Conclusions: In conclusion, increasing the expression of miR-194 can alleviate CH by targeting can and inhibiting the CnA/NFATc2 pathway.

Keywords: Cardiac hypertrophy (CH); cardiomyocytes hypertrophy; microRNA-194; calcineurin A/nuclear factor of activated T cells c2 (CnA/NFATc2)


Submitted Feb 17, 2022. Accepted for publication Jun 14, 2022.

doi: 10.21037/atm-22-1894


Introduction

Cardiac hypertrophy (CH) is a common pathological reaction caused by multiple cardiovascular diseases (CVDs), such as hypertension (1) and pulmonary arterial hypertension (PAH) (2). It is the main pathological basis that ultimately leads to heart failure (3). Right ventricular hypertrophy often occurs in patients with PAH (4,5). The pulmonary vascular remodeling caused by PAH leads to an increase in vascular resistance, which in turn causes an increase in right ventricular afterload. Left ventricular hypertrophy is common in patients with hypertension (6,7). The pathogenesis of CH is complex and has not yet been fully elucidated. Therefore, identification of novel targets in the pathogenesis of CH is crucial for the prevention and treatment of CH and heart failure.

Calcineurin A (CnA) is one of the catalytic subunits of calcineurin. It is the only Ca2+/calmodulin-dependent serine/threonine (Ser/Thr) phosphoprotein phosphatase known thus far (8). In cells, CnA catalyzes the dephosphorylation of nuclear factor of activated T cells (NFAT) protein, which then enters the nucleus (9). It is showed that activation of NFATc2 and its downstream factors can induce CH in mice, and calcineurin-induced CH is blocked in NFATc2 knockout mice (10). Activation of the CnA/NFATc2 pathway promotes cell growth and hypertrophy, and affects the contractile function of cardiomyocytes (11,12). Therefore, the regulation of CnA/NFATc2 pathway may be a novel target for the treatment of CH (13).

MicroRNA (miRNA) is a type of short, conserved endogenous RNA, consisting of about 22 nucleotides (14). MiRNAs efficiently bind to the 3'-untranslated region (UTR) of messenger RNAs (mRNAs). This kind of complementary base pairing can prevent translation or induce mRNA degradation (15). The role of miRNAs, such as miR-26a-5p (16), miR-146a (17), and miR-140-3p (18), in CH is gradually being revealed. Studies have shown that the levels of miR-194 are significantly reduced in children with dilated cardiomyopathy (19). It has also been suggested that miR-194 plays an antioxidant role by targeting MAPK1 in cardiomyocyte ischemia/reperfusion injury (20). Furthermore, the levels of miR-194 are also decreased in impaired renal function caused by hypertension (21). These data suggested that miR-194 may be involved in the occurrence of CH, but its expression characteristics and regulation mechanisms in CH are still unclear.

Therefore, this study analyzed the expression characteristics and effects of miR-194 in a CH cell model induced by isoproterenol (ISO), and explored the mechanisms based on the CnA/NFATc2 pathway. We present the following article in accordance with the MDAR reporting checklist (available at https://atm.amegroups.com/article/view/10.21037/atm-22-1894/rc).


Methods

Cell culture and treatment

The H9c2 cardiomyocytes (ATCC® CRL-1446) were provided by American Type Culture Collection (ATCC) (USA) and maintained in the RPMII640 complete medium containing 10% fetal bovine serum (FBS; Solarbio), 100 mg of streptomycin/mL (Solarbio), and 100 units of penicillin/mL (Solarbio). The cells were cultured in a 5% CO2 incubator at 37 ℃ and 95% humidity. To construct an in vitro CH cell model, cardiomyocyte hypertrophy was induced by ISO (JKLN013053, Jingke Chemical Technology Co., Ltd., Shanghai, China). The cells were seeded in a 24-well plate at 1×105 cells per well. The final concentration of ISO in the medium was 10 µmol/L, and the induction time was 48 hours.

Cell transfection

To analyze the influence of miR-194 and CnA in cardiomyocytes, miR-194 and/or CnA overexpression and/or silencing was performed. Transfectant sequences were showed in Table 1. Plasmids encoding the full-length human CnA (pcDNA3.1), small interfering (si) CnA, miR-194 mimic, miR-194 inhibitor, and corresponding negative control (NC) were purchased from GenePharma Co., Ltd. (Shanghai, China). The cells were transfected with 100 pmol pcDNA3.1 or 50 nM mimic (7 ℃, 5% CO2, 48 hours) using LipofectamineTM 2000 transfection reagent (Invitrogen Corporation, Carlsbad, CA, USA). The transfection efficiency was determined by quantitative reverse transcription polymerase chain reaction (RT-qPCR) or Western blot analysis.

Table 1

Transfectant sequences

Name Sequence (5'-3')
miR-194 mimic UGUAACAGCAACUCCAUGUGGA CACAUGGAGUUGCUGUUACAUU
Mimic NC UUCUCCGAACGUGUCACGUTT
miR-194 inhibitor UCCACAUGGAGUUGCUGUUACA
Inhibitor NC UACUCGGAUCUUGUCACTAAG
CnA CTAGAGAACCCACTGCTTAC
NC TAGAAGGCACAGTCGAGG
siCnA GAACCGCAAUUUUACCGUC
siNC UUCUCCGAACGUGUGUCACGU

NC, negative control; CnA, calcineurin A.

Assessment of CH

Cells were seeded in a 24-well plate at 5×103 cells per well. After 24 hours, the cells were fixed with 4% paraformaldehyde (P6148, Sigma-Aldrich, St. Louis, MO, USA). After permeabilization with 0.1% Triton X-100, cells were stained with DAPI and phalloidin (Life Technology, St. Louis, MO, USA) for 10 and 40 minutes, respectively, in the dark. After washing and removing the reagents, LSM800 confocal microscope (Zeiss, Germany) was used for imaging. According to the fluorescence range, ImageJ was applied to calculate the cell surface area.

RT-qPCR

H9c2 cells were harvested and RT-qPCR was performed for the detection of mRNA expression. Briefly, the total RNA in the cells was extracted using Trizol Reagent (Thermo Fisher Scientific, Waltham, MA, USA). Using the Primescript RT Reagent kit (Takara, Shiga, Japan), each total RNA sample (1 µg) was subjected to reverse transcription reaction to obtain the cDNA template. The qPCR amplification was performed with SYBR Green reagent (Takara) using the ABI 7500 fast real-time PCR System (Applied Biosystems, Foster City, CA, USA) under the following conditions: 95 ℃ for 10 seconds, followed by 40 cycles of 95 ℃ for 10 seconds, 50 ℃ for 30 seconds, and 72 ℃ for 30 seconds. The expression of mRNA was normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) using the 2−ΔΔCt method.

Total miRNA was extracted using miRNeasy Mini kit (GE Healthcare, USA), and the cDNA was generated using the TaqMan miRNA reverse transcription kit (DBI Bioscience, Germany). The TaqMan miRNA kit (DBI Bioscience, Germany) was applied to measure the expression levels of the miRNA, which was normalized to U6 using the 2−ΔΔCt method. Primer sequences were showed in Table 2.

Table 2

Primer sequences

Name Sequence (5'-3')
miR-194 (forward) CTAGTACCTAGAGGAACCTTTGAAGACTGTTACAGCTCAGCA
miR-194 (reverse) AGCTTGCTGAGCTGTAACAGTCTTCAAAGGTTCCTCTAGGTA
U6 (forward) ACCCTGAGAAATACCCTCACAT
U6 (reverse) GACGACTGAGCCCCTGATG
ANP (forward) GGCTCCTTCTCCATCACCAA
ANP (reverse) TGTTATCTTCGGTACCG
can (forward) CACTTCTTTTGCTGTAAGCCG
can (reverse) AAGGCAATTGATCCCAAGTT
GAPDH (forward) ACCACAGTCCATGCCATCAC
GAPDH (reverse) TCCACCACCCTGTTGCTGT

CnA, calcineurin A; ANP, atrial natriuretic peptide; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Western blot

H9c2 cells were harvested and incubated with RIPA lysis solution on ice for 30 minutes. Cell lysate samples were centrifuged for 20 minutes at 4 ℃, 12,000 ×g to obtain the supernatant. The concentration of the total proteins in the supernatant was determined using the BCA kit (Beyotime Biotechnology, Jiangsu, China). In addition, proteins in the cytoplasm and nucleus were separated using an Ambion PARISTM Kit (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was performed for the separation of proteins. After being transferred to a polyvinylidene fluoride membrane, proteins were blocked with 5% skimmed milk for 2 hours at room temperature. Membranes were incubated with rabbit anti-primary antibodies (1:1,000) overnight at 4 ℃. The primary antibodies used are as follows: anti-atrial natriuretic peptide (ANP; 1:1,000, sc-515701, Santa Cruz, CA, USA), anti-CnA (1:1,000, ab52761, Abcam, Cambridge, MA, USA), anti-NFATc2 (1:1,000, sc-7296), anti-GAPDH (1:1,000, ab8245), and anti-lamin B1 (1:1,000, ab229025). GAPDH was used as the control for cytoplasmic protein or total protein in the cells. Lamin B1 was used as the control for proteins in the nucleus. Thereafter, Tris-buffered saline/0.1% Tween (TBST) solution was used to wash the membranes twice. Horseradish peroxidase-labeled goat anti-rabbit IgG secondary antibody (1:2,000, ab6721, Abcam, Cambridge, MA, USA) was used to incubate the membranes for 2 hours at 37 ℃. Membranes were washed 3 times with TBST and the protein blots were visualized using enhanced chemiluminescence (ECL) (Solarbio). Image analysis software IPP6.0 was applied for the analysis of gray intensity.

Dual luciferase report

The 3'-UTR sequence of wild-type (wt-) CnA mRNA was amplified to the downstream site of the pGL4 luciferase vector (Promega, Madison, WI, USA). The rapid site-directed mutagenesis kit (D0206, Beyotime, Shanghai, China) was used to generate the mutated (mut-) CnA mRNA 3'-UTR. The H9c2 cells were seeded in a 24-well plate at a density of 3×104/well. After 24 hours, 1 µg of wt-CnA mRNA 3'-UTR or mut-CnA mRNA 3'-UTR luciferase plasmid, 50 nM miR-194 mimic or miR-194 NC, and 150 ng of Renilla luciferase plasmid (Beyotime, Shanghai, China) were transfected into cells via LipofectamineTM 2000. The cells were then incubated at 37 ℃ for 36 hours. The dual luciferase reporter gene detection kit (Promega, Madison, WI, USA) was used to detect luciferase activity according to the manufacturer’s protocol. All data were normalized to Renilla luciferase activity.

Statistical analysis

All experiments were performed independently 3 times. Each index was measured 3 times. Data are presented as mean ± standard deviation (SD) and analyzed using SPSS19.0 software (SPSS Inc., Chicago, IL, USA). The Student’s t-test was used for comparison between two groups. For comparison among at least three groups, one-way analysis of variance (ANOVA) was applied. A P value <0.05 was considered statistically significant.


Results

After ISO-induced hypertrophy of H9c2 cells, the levels of miR-194 expression decreases and the CnA/NFATc2 pathway was activated

H9c2 cells were treated with ISO to simulate an in vitro CH model. CH was identified by detecting the expression of the CH marker ANP and measuring cell surface area. The cell surface area increased to about 1.8 times the original after induction (P<0.05; Figure 1A). Furthermore, the mRNA and protein expression of ANP, a marker gene of CH, were significantly increased after ISO-induction (P<0.05; Figure 1B,1C). These results indicated that H9c2 hypertrophy was successfully induced in vitro. RT-qPCR results showed that the levels of miR-194 in hypertrophic H9c2 cells were reduced, while the levels of CnA mRNA were increased (P<0.05; Figure 1D). In addition, compared with non-induced cells, the levels of CnA protein in the induced H9c2 cells increased significantly, and a large amount of NFATc2 protein left the cytoplasm and entered the nucleus (P<0.05; Figure 1E-1I). In the hypertrophic cardiomyocytes induced by ISO, the levels of miR-194 decreased and the CnA/NFATc2 pathway was activated. The levels of CnA transcription and protein were elevated, and the levels of nuclear translocation of NFATc2 were also increased.

Figure 1 In H9c2 cells induced by ISO, the expression of miR-194 decreases and the CnA/NFATc2 pathway is activated. (A) The effects of ISO on cardiomyocyte hypertrophy (surface area) were assessed by immunofluorescence staining (×400). (B) The effects of ISO on the expression of ANP mRNA, a marker of cardiac hypertrophy in cardiomyocytes. (C) The effects of ISO on the expression of ANP protein expression. (D) The effects of ISO on the expression of miR-194. (E) The effects of ISO on the expression of CnA mRNA. (F-I) The effects of ISO on the expression of CnA protein and NFATc2 protein nuclear translocation. *, P<0.05 vs. control group. ISO, isoproterenol; miR, microRNA; CnA, calcineurin A; NFATc2, nuclear factor of activated T cells c2; ANP, atrial natriuretic peptide; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

MiR-194 targeting inhibits CnA protein expression

Sequence prediction analysis revealed the binding sites between miR-194 and the 3'-UTR of the CnA mRNA (Figure 2A). Dual luciferase reporter assays demonstrated that when miR-194 mimic and wt-CnA mRNA 3'-UTR were transfected into cells, the relative luciferase activity was significantly inhibited. When the CnA mRNA 3'-UTR was mutated, it could not bind to miR-194, and the luciferase activity remained unchanged compared to wt-CnA (Figure 2B). This suggested that miR-194 directly targeted and bound to the CnA mRNA 3'-UTR. In addition, overexpression of miR-194 was performed by transfecting cells with the miR-194 mimic (Figure 2C). When the levels of miR-194 were elevated, the expression of the CnA protein was significantly suppressed (P<0.05; Figure 2D), suggesting that miR-194 inhibits the expression of CnA protein by targeting the 3'-UTR of the CnA mRNA.

Figure 2 Targeting miR-194 inhibits calcineurin A protein expression. (A) The binding sites of miR-194 and the 3'-UTR of CnA mRNA. (B) Dual luciferase report assays verified the targeted binding of miR-194 to the 3'-UTR of CnA mRNA. (C) The levels of miR-194 in cells transfected with miR-194 NC and miR-194 mimic. (D) The expression of CnA protein in cells after overexpression of miR-194. *, P<0.05 vs. NC group. miR, microRNA; CnA, calcineurin A; UTR, untranslated region; WT, wild-type; mut, mutant; NC, negative control; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Decreasing the expression of miR-194 causes cardiomyocyte hypertrophy through the CnA/NFATc2 pathway

To analyze the effects of miR-194 and CnA on cardiomyocyte hypertrophy, H9c2 cells were divided into 4 groups as follows: NC, miR-194 inhibitor, siCnA, and miR-194 inhibitor + siCnA. Western blot analysis revealed that when the levels of miR-194 were suppressed, the levels of CnA protein were significantly elevated. The CnA protein levels in the siCnA group were significantly lower than that of the NC group (P<0.05; Figure 3A,3B). This not only indicated that the transfection experiment was successful, but also further verified that miR-194 could inhibit the expression of CnA protein. Consistent with the expression levels of the CnA protein, both the reduction of miR-194 and the overexpression of CnA protein promoted the nuclear translocation of NFATc2 protein. Moreover, interference with CnA blocked the entry of NFATc2 into the nucleus caused by miR-194 downregulation (P<0.05; Figure 3C-3E). The cell surface area was increased in the presence of the miR-194 inhibitor, while interference with CnA decreased the cell surface area. When the expression of CnA was interfered with, transfection of the miR-194 inhibitor could no longer increase the cell surface area (P<0.05; Figure 3F). This further verified that cardiomyocyte hypertrophy caused by a reduction in miR-194 expression is inseparable from the CnA/NFATc2 pathway.

Figure 3 Decreasing the expression of miR-194 causes cardiomyocyte hypertrophy through the CnA/NFATc2 pathway. (A) The levels of miR-194 in cells transfected with miR-194 NC and miR-194 inhibitor. (B) The expression levels of CnA protein in each group. (C-E) The nuclear translocation of the NFATc2 protein in each group of cells. (F) Comparison of the surface area in each group of cells by immunofluorescence staining (×400). *, P<0.05 vs. NC group; #, P<0.05 vs. miR-194 inhibitor group; +, P<0.05 vs. siCnA group. miR, microRNA; CnA, calcineurin A; NFATc2, nuclear factor of activated T cells c2; NC, negative control; si, small interfering; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Overexpression of miR-194 alleviates hypertrophy of H9c2 cells and activation of CnA/NFATc2 pathway induced by ISO

To further analyze whether miR-194 can alleviate the myocardial hypertrophy induced by ISO, rescue experiments were conducted. H9c2 cells were divided into 4 groups, namely, control, ISO, miR-194 mimic, and ISO + miR-194 mimic. Overexpression of miR-194 significantly inhibited the levels of CnA protein expression and inhibited nuclear translocation of NFATc2. Furthermore, increasing the levels of miR-194 partially blocked the activation of the CnA/NFATc2 pathway induced by ISO (P<0.05; Figure 4A-4E). Furthermore, increasing the levels of miR-194 reduced the cell surface area to about 70% of the area after ISO treatment. Overexpression of miR-194 significantly reduced the ISO-induced hypertrophy in cardiomyocytes (P<0.05; Figure 4F). The above experiments showed that overexpression of miR-194 not only alleviated the hypertrophy of cardiomyocytes induced by ISO, but also blocked the activation of the CnA/NFATc2 pathway caused by ISO.

Figure 4 Overexpression of miR-194 alleviates hypertrophy of H9c2 cells and activation of CnA/NFATc2 pathway induced by ISO. (A) The levels of miR-194 in cells transfected with miR-194 NC and miR-194 mimic. (B) The expression levels of CnA protein in each group. (C-E) The nuclear translocation of NFATc2 protein in each group of cells. (F) Comparison of the surface area of each group of cells by immunofluorescence staining (×400). *, P<0.05 vs. NC group; #, P<0.05 vs. miR-194 mimic group; +, P<0.05 vs. ISO group. ISO, isoproterenol; miR, microRNA; CnA, calcineurin A; NFATc2, nuclear factor of activated T cells c2; NC, negative control; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Discussion

CVD is characterized by high morbidity, disability, and mortality. Approximately 17 million people worldwide die from CVD each year, accounting for about one-third of patients who died of disease. It is estimated that by 2030, the number of deaths due to CVD every day will reach 65,000 (22,23). CVD, such as hypertension and myocardial infarction, can cause CH, and ultimately affect the ejection function of the heart. However, the mechanisms of CH remain to be fully elucidated.

The regulatory mechanisms of post-transcriptional non-coding RNAs (ncRNAs) is a new research focus. The post-transcriptional regulation of miRNAs directly affects the expression of proteins and thus participates in the process of CH, which makes miRNAs a potential target for the treatment of CH (24). For example, miR-26a-5p directly inhibits the expression of glycogen synthase kinase 3 beta (GSK3β), elevates the expression of ANP and other proteins, and increases the cell surface area (25). In ISO-induced CH, increasing the levels of miR-29a alleviates CH through targeted inhibition of peroxisome proliferator-activated receptor δ (26). In this study, ISO was successfully used to simulate in vitro CH, resulting in a 1.8-fold increase in the surface area of cardiomyocytes. After induction, the levels of miR-194 in the cells were significantly reduced. Furthermore, the levels of CnA mRNA and protein in the cells were elevated, and the downstream NFATc2 protein entered the nucleus. In addition, the dual luciferase reporter experiments showed that miR-194 directly targets and binds to the 3'-UTR of CnA mRNA. Overexpression of miR-194 significantly inhibited the expression of the CnA protein. This study demonstrated the alterations in miR-194 expression and the CnA/NFATc2 pathway in cardiomyocyte hypertrophy. Further research is warranted to elucidate the precise mechanisms involved.

While homeostasis of Ca2+ is the key to maintaining the normal morphology and function of cardiomyocytes, it is also involved in the process of β-adrenaline-induced CH (27). The stimulation of β-adrenaline affects the oscillation frequency of Ca2+, thereby activating CnA (8). This in turn activates the nuclear translocation of the NFATc2 protein (28). Indeed, activation of NFATc2 plays a crucial role in ISO-induced CH (8). The CnA/NFATc2 pathway can be targeted and inhibited by miRNAs. For example, Zhang et al. (29) reported that miR-137 inhibits NFAT by reducing the expression of CnA, thereby regulating the de-differentiation and proliferation of smooth muscle cells. CnA is also targeted and regulated by miR-206-3p and miR-381-3p, thereby affecting the transcription levels of inducible nitric oxide synthase (iNOS), which regulates NFATc2 and affects muscle cell function (30,31). MiR-30 also protects the morphology and function of podocytes by inhibiting the CnA/NFATc2 pathway (32). To analyze the effects of miR-194/CnA/NFATc2 on cardiomyocytes, cells models in which miR-194 and/or CnA expression were inhibited were constructed. The results showed that suppression of miR-194 increased the levels of CnA protein, enhanced nuclear translocation of NFATc2, and increased the cell surface area. Interference of CnA expression blocked the inhibitory effects of miR-194 on cardiomyocyte hypertrophy. This suggested that reduction of miR-194 expression causes cardiomyocyte hypertrophy by activating the CnA/NFATc2 pathway.

Previous clinical studies have shown that the levels of miR-194 in the plasma of children with idiopathic dilated cardiomyopathy is significantly reduced (19). Furthermore, in vitro studies have demonstrated that increasing the levels of miR-194 inhibited the proliferation and differentiation of rabbit skeletal muscle satellite cells (33). In addition, CH is a common complication of myocardial ischemia/reperfusion (34), and in vitro studies have reported that increasing the levels of miR-194 protected the human renal tubular epithelial cells from hypoxia/reperfusion injury (35). After the heart is damaged, fibrosis and hypertrophy can occur simultaneously (36). The hypoxia-induced mitogenic factor promotes the occurrence of CH by aggravating the inflammatory response, and the levels of intracellular fibrin inevitably increase in the process (37). A study has shown that miR-194 inhibits the expression of the Runx1 gene at the post-transcriptional level, thereby regulating the AKT pathway and inhibiting renal fibrosis (38). To further analyze the role of miR-194 in CH, rescue experiments were conducted. The data revealed that after elevating the levels of miR-194, the increased cells surface area induced by ISO was significantly inhibited. In addition, the increase in CnA protein expression caused by ISO and the nuclear translocation of NFATc2 were also alleviated. This suggested that ISO may promote the transcription of CnA and increase CnA protein levels, which in turn activates NFATc2 and induces nuclear translocation, thereby inducing cardiomyocyte hypertrophy. Therefore, increasing the levels of miR-194 may target and inhibit CnA expression at the post-transcriptional level, and alleviate cardiomyocyte hypertrophy.


Conclusions

In summary, increasing the expression of miR-194 can alleviate CH by targeting can and inhibiting the CnA/NFATc2 pathway. The characteristics of miR-194 in CH require further clinical research. Furthermore, the inhibitory effects of miR-194 on CH and the CnA/NFATc2 pathway warrant further verification through in vivo experiments.


Acknowledgments

Funding: The study was funded by General Project of Chongqing Natural Science Foundation (No. cstc2021jcyj-msxmX0307).


Footnote

Reporting Checklist: The authors have completed the MDAR reporting checklist. Available at https://atm.amegroups.com/article/view/10.21037/atm-22-1894/rc

Data Sharing Statement: Available at https://atm.amegroups.com/article/view/10.21037/atm-22-1894/dss

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-1894/coif). All authors report that the study was funded by General Project of Chongqing Natural Science Foundation (No. cstc2021jcyj-msxmX0307). The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Song XA, Jia LL, Cui W, et al. Inhibition of TNF-α in hypothalamic paraventricular nucleus attenuates hypertension and cardiac hypertrophy by inhibiting neurohormonal excitation in spontaneously hypertensive rats. Toxicol Appl Pharmacol 2014;281:101-8. [Crossref] [PubMed]
  2. Almodovar S, Swanson J, Giavedoni LD, et al. Lung Vascular Remodeling, Cardiac Hypertrophy, and Inflammatory Cytokines in SHIVnef-Infected Macaques. Viral Immunol 2018;31:206-22. [Crossref] [PubMed]
  3. Cluntun AA, Badolia R, Lettlova S, et al. The pyruvate-lactate axis modulates cardiac hypertrophy and heart failure. Cell Metab 2021;33:629-648.e10. [Crossref] [PubMed]
  4. Antigny F, Mercier O, Humbert M, et al. Excitation-contraction coupling and relaxation alteration in right ventricular remodelling caused by pulmonary arterial hypertension. Arch Cardiovasc Dis 2020;113:70-84. [Crossref] [PubMed]
  5. Shults NV, Kanovka SS, Ten Eyck JE, et al. Ultrastructural Changes of the Right Ventricular Myocytes in Pulmonary Arterial Hypertension. J Am Heart Assoc 2019;8:e011227. [Crossref] [PubMed]
  6. Petretta M, Bianchi V, Marciano F, et al. Influence of left ventricular hypertrophy on heart period variability in patients with essential hypertension. J Hypertens 1995;13:1299-306. [Crossref] [PubMed]
  7. Shenasa M, Shenasa H. Hypertension, left ventricular hypertrophy, and sudden cardiac death. Int J Cardiol 2017;237:60-3. [Crossref] [PubMed]
  8. Khalilimeybodi A, Daneshmehr A, Sharif Kashani B. Ca2+-dependent calcineurin/NFAT signaling in β-adrenergic-induced cardiac hypertrophy. Gen Physiol Biophys 2018;37:41-56. [Crossref] [PubMed]
  9. Zimmer J, Takahashi T, Hofmann AD, et al. Imbalance of NFATc2 and KV1.5 Expression in Rat Pulmonary Vasculature of Nitrofen-Induced Congenital Diaphragmatic Hernia. Eur J Pediatr Surg 2017;27:68-73. [Crossref] [PubMed]
  10. Bourajjaj M, Armand AS, da Costa Martins PA, et al. NFATc2 is a necessary mediator of calcineurin-dependent cardiac hypertrophy and heart failure. J Biol Chem 2008;283:22295-303. [Crossref] [PubMed]
  11. Zhang Y, Su SA, Li W, et al. Piezo1-Mediated Mechanotransduction Promotes Cardiac Hypertrophy by Impairing Calcium Homeostasis to Activate Calpain/Calcineurin Signaling. Hypertension 2021;78:647-60. [Crossref] [PubMed]
  12. Marunouchi T, Nakashima M, Ebitani S, et al. Hsp90 Inhibitor Attenuates the Development of Pathophysiological Cardiac Fibrosis in Mouse Hypertrophy via Suppression of the Calcineurin-NFAT and c-Raf-Erk Pathways. J Cardiovasc Pharmacol 2021;77:822-9. [Crossref] [PubMed]
  13. Sugiyama A, Okada M, Yamawaki H. Canstatin suppresses isoproterenol-induced cardiac hypertrophy through inhibition of calcineurin/nuclear factor of activated T-cells pathway in rats. Eur J Pharmacol 2020;871:172849. [Crossref] [PubMed]
  14. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281-97. [Crossref] [PubMed]
  15. Ambros V. The functions of animal microRNAs. Nature 2004;431:350-5. [Crossref] [PubMed]
  16. Shi H, Li H, Zhang F, et al. MiR-26a-5p alleviates cardiac hypertrophy and dysfunction via targeting ADAM17. Cell Biol Int 2021;45:2357-67. [Crossref] [PubMed]
  17. Oh JG, Watanabe S, Lee A, et al. miR-146a Suppresses SUMO1 Expression and Induces Cardiac Dysfunction in Maladaptive Hypertrophy. Circ Res 2018;123:673-85. [Crossref] [PubMed]
  18. Li H, Xu JD, Fang XH, et al. Circular RNA circRNA_000203 aggravates cardiac hypertrophy via suppressing miR-26b-5p and miR-140-3p binding to Gata4. Cardiovasc Res 2020;116:1323-34. [Crossref] [PubMed]
  19. Enes Coşkun M, Kervancıoğlu M, Öztuzcu S, et al. Plasma microRNA profiling of children with idiopathic dilated cardiomyopathy. Biomarkers 2016;21:56-61. [Crossref] [PubMed]
  20. Zhang Q, Wu X, Yang J. miR-194-5p protects against myocardial ischemia/reperfusion injury via MAPK1/PTEN/AKT pathway. Ann Transl Med 2021;9:654. [Crossref] [PubMed]
  21. Rudnicki M, Perco P, D, Haene B, et al. Renal microRNA- and RNA-profiles in progressive chronic kidney disease. Eur J Clin Invest 2016;46:213-26. [Crossref] [PubMed]
  22. Feigin VL, Forouzanfar MH, Krishnamurthi R, et al. Global and regional burden of stroke during 1990-2010: findings from the Global Burden of Disease Study 2010. Lancet 2014;383:245-54. [Crossref] [PubMed]
  23. Virani SS, Alonso A, Benjamin EJ, et al. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation 2020;141:e139-596. [Crossref] [PubMed]
  24. Bernardo BC, Ooi JY, Lin RC, et al. miRNA therapeutics: a new class of drugs with potential therapeutic applications in the heart. Future Med Chem 2015;7:1771-92. [Crossref] [PubMed]
  25. Tang L, Xie J, Yu X, et al. MiR-26a-5p inhibits GSK3β expression and promotes cardiac hypertrophy in vitro. PeerJ 2020;8:e10371. [Crossref] [PubMed]
  26. Zhang S, Yin Z, Dai FF, et al. miR-29a attenuates cardiac hypertrophy through inhibition of PPARδ expression. J Cell Physiol 2019;234:13252-62. [Crossref] [PubMed]
  27. Studer R, Reinecke H, Vetter R, et al. Expression and function of the cardiac Na+/Ca2+ exchanger in postnatal development of the rat, in experimental-induced cardiac hypertrophy and in the failing human heart. Basic Res Cardiol 1997;92:53-8. [Crossref] [PubMed]
  28. Park YJ, Yoo SA, Kim M, et al. The Role of Calcium-Calcineurin-NFAT Signaling Pathway in Health and Autoimmune Diseases. Front Immunol 2020;11:195. [Crossref] [PubMed]
  29. Zhang B, Chen L, Bai YG, et al. miR-137 and its target T-type CaV 3.1 channel modulate dedifferentiation and proliferation of cerebrovascular smooth muscle cells in simulated microgravity rats by regulating calcineurin/NFAT pathway. Cell Prolif 2020;53:e12774. [Crossref] [PubMed]
  30. Lin CC, Law BF, Hettick JM. Acute 4,4'-Methylene Diphenyl Diisocyanate Exposure-Mediated Downregulation of miR-206-3p and miR-381-3p Activates Inducible Nitric Oxide Synthase Transcription by Targeting Calcineurin/NFAT Signaling in Macrophages. Toxicol Sci 2020;173:100-13. [PubMed]
  31. Ahmed AM. Inhibition of inducible nitric oxide synthase (iNOS) by simvastatin attenuates cardiac hypertrophy in rats. Folia Morphol (Warsz) 2017;76:15-27. [Crossref] [PubMed]
  32. Lang Y, Zhao Y, Zheng C, et al. MiR-30 family prevents uPAR-ITGB3 signaling activation through calcineurin-NFATC pathway to protect podocytes. Cell Death Dis 2019;10:401. [Crossref] [PubMed]
  33. Shi Y, Mao X, Cai M, et al. miR-194-5p negatively regulates the proliferation and differentiation of rabbit skeletal muscle satellite cells. Mol Cell Biochem 2021;476:425-33. [Crossref] [PubMed]
  34. Chen J, Ma Q, King JS, et al. aYAP modRNA reduces cardiac inflammation and hypertrophy in a murine ischemia-reperfusion model. Life Sci Alliance 2020;3:e201900424. [Crossref] [PubMed]
  35. Shen Y, Zhao Y, Wang L, et al. MicroRNA-194 overexpression protects against hypoxia/reperfusion-induced HK-2 cell injury through direct targeting Rheb. J Cell Biochem 2018; [Epub ahead of print]. [PubMed]
  36. Rai V, Sharma P, Agrawal S, et al. Relevance of mouse models of cardiac fibrosis and hypertrophy in cardiac research. Mol Cell Biochem 2017;424:123-45. [Crossref] [PubMed]
  37. Kumar S, Wang G, Zheng N, et al. HIMF (Hypoxia-Induced Mitogenic Factor)-IL (Interleukin)-6 Signaling Mediates Cardiomyocyte-Fibroblast Crosstalk to Promote Cardiac Hypertrophy and Fibrosis. Hypertension 2019;73:1058-70. [Crossref] [PubMed]
  38. Cheng L, Tu C, Min Y, et al. MiR-194 targets Runx1/Akt pathway to reduce renal fibrosis in mice with unilateral ureteral obstruction. Int Urol Nephrol 2020;52:1801-8. [Crossref] [PubMed]
Cite this article as: Wang J, An Y, Lin J, Tang G. MicroRNA-194 inhibits isoproterenol-induced chronic cardiac hypertrophy via targeting CnA/NFATc2 signaling in H9c2 cells. Ann Transl Med 2022;10(14):780. doi: 10.21037/atm-22-1894

Download Citation