Open Access

Calcium inhibitor inhibits high glucose‑induced hypertrophy of H9C2 cells

  • Authors:
    • Xiaohong Xu
    • Luoyang Ruan
    • Xiaohua Tian
    • Fengjuan Pan
    • Cailan Yang
    • Guosheng Liu
  • View Affiliations

  • Published online on: June 26, 2020     https://doi.org/10.3892/mmr.2020.11275
  • Pages: 1783-1792
  • Copyright: © Xu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to explore whether the hypertrophy of H9C2 cardiomyocytes was induced by high glucose, to investigate whether the calcium channel inhibitor (Norvasc) could inhibit this process and to clarify the possible signaling pathways. The morphology of H9C2 cells was observed under an optical microscope, and the cell surface area was measured by Image Pro Plus 6.1 software. Furthermore, fluorescence spectrophotometry was used to detect intracellular calcium concentration ([Ca2+]i). ELISA was performed to detect calcineurin (CaN) activity; reverse transcription‑quantitative PCR and western blotting were performed to detect the mRNA and protein expression levels of CaN Aβ subunit (CnAβ), nuclear factor of activated T cells 3 (NFAT3) and β type myosin heavy chain (β‑MHC). Cell size was increased with the increase in glucose concentration of culture medium at 48 and 72 h, respectively, and decreased with the addition of Norvasc compared with those without Norvasc (P<0.05). There was no significant difference in cell size with the addition of Norvasc compared with cells cultured with 5 mM glucose (P>0.05). The average [Ca2+]i activity of single cells in the 48‑ and 72‑h culture groups treated with 50 mM glucose was significantly higher than cells treated with 5 mM glucose (P<0.05); and the fluorescent value of average [Ca2+]i activity of single cells was lower, following the addition of Norvasc than that without Norvasc (P<0.05). CaN activity in the 48‑ and 72‑h culture group treated with 50 mM glucose was markedly higher than that treated with 5 mM glucose, and the activity of CaN notably decreased with the addition of Norvasc compared with those without Norvasc. The mRNA and protein expression levels of CnAβ, NFAT3 and β‑MHC in the 48‑ and 72‑h culture groups treated with 50 mM glucose were all significantly higher than those treated with 5 mM glucose (P<0.05). The mRNA and protein expression of CnAβ, NFAT3 and β‑MHC cultured with 50 mM glucose were significantly decreased following the addition of Norvasc (P<0.05). Thus, the calcium channel inhibitor Norvasc may inhibit high glucose‑induced hypertrophy of H9C2 cardiomyocytes by inhibiting the Ca2+‑CaN‑NFAT3 signaling pathway.

Introduction

China has a high incidence of diabetes mellitus, with the number of diabetic patients exceeding 90 million (1). At present, ~300 million individuals have been diagnosed with diabetes mellitus worldwide (2). Large-scale clinical studies have reported that patients with diabetes have a significantly higher risk of heart injury and cardiac insufficiency compared with those without diabetes (37). Diabetic cardiomyopathy is defined as a heart injury, which is independent of other diseases and is caused by diabetes mellitus itself (8), and manifests as left ventricular diastolic dysfunction at the early stage of onset and systolic dysfunction at the later stage. If not treated properly, diabetic cardiomyopathy can develop into heart failure, arrhythmia, cardiogenic shock and even sudden death in severe cases (9).

There are multiple clinical strategies to prevent and treat diabetic cardiomyopathy. However, the effect of reducing the incidence and mortality of cardiovascular complications requires improvement in patients with diabetes. Therefore, the study of diabetic cardiomyopathy requires extensive investigation. It is currently hypothesized that several mechanisms are involved in the pathogenesis of diabetic cardiomyopathy, including changes in myocardial energy metabolism and calcium signaling (1013).

As an important secondary messenger, calcium plays an important role in the excitation-contraction coupling of the heart and regulates the expression of cardiac-related genes. The role of Ca2+ signaling and its dependent signal transduction pathway in cardiac hypertrophy and myocardial apoptosis has been widely recognized by researchers. Ca2+ is ubiquitous in cells and plays an important role not only in the electrical activity of the heart but also as a direct activator of myofilament contraction. Calcium enters the cardiomyocytes through voltage-gated calcium channels which selectively permeate calcium ions into the cell when there is a change from a high voltage to a low voltage inside of the cell. Voltage-gated calcium ion channels are subdivided into six fundamental types according to their electrophysiology and sensitivity to certain drugs and toxins. These voltage-gated calcium ion channels are named L-, T-, N-, P-, Q- and R-channels: L-channels have a long activation and high conductance, and are mainly located in skeletal, cardiac and vascular muscle and are involved in its contraction; T-channels have a transient opening and are involved in calcium ion entry when the membrane is depolarized; N-channels are involved in neurotransmitter release; P/Q-channels are mainly localized in the nerve terminal of cells of the cerebellum and are involved in neurotransmitter release; and R-channels are mainly localized in cell bodies and are involved in Ca2+-dependent action potentials (14).

In the myocardium, the L-type calcium channel is the main pathway for calcium to enter the cell (15).

The role of the Ca2+-calcineurin (CaN)-nuclear factor of activated T cells 3 (NFAT3) signaling pathway in cardiac development has gained interest in recent years, and several classes of antihypertensive drugs, including angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and calcium channel blockers (CCBs), have been demonstrated to reverse hypertrophy in humans (16,17). CCBs were introduced for the treatment of hypertension in the 1980s. Their use was subsequently expanded to disorders such as angina pectoris, paroxysmal supraventricular tachycardia, hypertrophic cardiomyopathy, coronary spasm and cerebral vasospasm (18). Evidence from experimental studies indicate that CCBs nifedipine, nisoldipine and amlodipine additionally lead to regression of interstitial and perivascular myocardial fibrosis, which may contribute to the improvement of diastolic function and coronary reserve (19).

Norvasc is the brand name of amlodipine besylate. Amlodipine does not increase cardiovascular morbidity or mortality in patients with severe heart failure (20) and multiple studies have reported that amlodipine can reduce cardiac remodeling in spontaneously hypertensive rats and myocardial infarction in rats (2123). Therefore, Norvasc was selected for the current study to determine whether it could inhibit hypertrophy of H9C2 cardiomyocytes induced by high glucose.

The aim of the present study was to investigate high glucose-induced hypertrophy of H9C2 cells and its possible pathogenesis, from the perspective of the calcium signaling pathway, to provide a theoretical basis for the identification of potential targets for clinical prevention and treatment of this disease.

Materials and methods

Materials

Cell culture reagents: FBS (cat. no. SH30087.01; Hyclone; GE Healthcare Life Sciences); DMEM-high glucose culture medium (50 mM; cat. no. 11965-092; Gibco; Thermo Fisher Scientific, Inc.); DMEM-low glucose culture medium (5 mM; cat. no. 10567-014; Gibco; Thermo Fisher Scientific, Inc.). H9C2 cells were rat embryonic cardiomyocytes purchased from the Cell Resource Center of Shanghai Academy of Life Sciences, Chinese Academy of Sciences; Norvasc was obtained from Sigma-Aldrich (Merck KGaA); ELISA assay kit of CaN (cat. no. E-EL-R0134c) was purchased from Elabscience Biotechnology Co., Ltd.; Fluo-3 AM (calcium ion fluorescent probe; cat. no. S1056) was from Beyotime Institute of Biotechnology; DNase I (RNase-free) was purchased from Beijing Transgen Biotech Co., Ltd.; reverse transcription kit and real-time PCR kit were obtained from Vazyme; CnAβ, nuclear factor of activated T cells 3 (NFAT3), β type myosin heavy chain (β-MHC) and b-actin primary antibodies (cat. nos. ab3673, ab66781, ab207926 and ab8227, respectively) were all purchased from Abcam.

Cell culture

Following thawing, cells (1×104−1×105/ml) were cultured in DMEM containing 10% FBS in a 5% CO2 incubator, with saturated humidity at 37°C.

Experimental grouping

The experimental groups were as follows: i) A1, 48-h culture group treated with 5 mM glucose; ii) B1, 48-h culture group treated with 50 mM glucose; iii) C1, 48-h culture group treated with 50 mM glucose + 25 nmol/l Norvasc; iv) A2, 72-h culture group treated with 5 mM glucose; v) B2, 72-h culture group treated with 50 mM glucose; vi) C2, 72-h culture group treated with 50 mM glucose + 25 nmol/l Norvasc. All cell treatments used DMEM containing 10% FBS.

Determination of cell surface area

Cell morphology were observed using an optical microscope, and the cell surface area was measured by Image-Pro Plus 6.1 software (Media Cybernetics, Inc.). A total of 3 fields of view were observed at magnification ×20.

Determination of intracellular calcium ([Ca2+]i) activity

Cell culture media was removed and cells (1×104−1×105/ml) were washed with PBS. Fluo-3 (2 µM), diluted in serum-free media was added for 1.5 h at 37°C, 5% CO2. Fluo-3 was removed, followed by two PBS washes and the addition of cell culture media. Fluorescence microscopy (magnification, ×20) was performed to detect [Ca2+]i.

Determination of cellular CaN enzyme activity

Enzyme activity of CaN was measured using a rat CaN ELISA assay kit, according to the manufacturer's instructions.

Detection of CaN, NFAT3 and β-MHC mRNA expression in myocardial cells by reverse transcription-quantitative (RT-q)PCR

The extraction of RNA was achieved using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.). Next, the reaction mixture was prepared with RNase-free DNase І, followed by digestion for 30 min at 37°C and inactivation for 10 min at 65°C to remove DNA. Reverse transcription was conducted with the addition of template RNA and primer mixtures at 42°C for 1 h, then resting on ice for 2 min. This was followed by the addition of cDNA (10X) to the primer reaction mixture. The RNA concentration was measured with NanoDrop™ 1000 (Thermo Fisher Scientific, Inc.). The cycling conditions for qPCR were as follows: 95°C for 30 sec, 40 cycles at 95°C for 10 sec followed by 60°C for 34 sec. The melting curve was drawn within the temperature range of 60–95°C. The primer sequences were: Rat-GAPDH forward (F), 5′-CATCAACGACCCCTTCATTG-3′ and reverse (R), 5′-GAAGATGGTGATGGGTTTCC-3′; rat-CaN F, 5′-ATGTTGCCTAGTGGAGTGTT-3′ and R, 5′-GGAGAGTATCCTCGTATTGCTT-3′; NFAT3 F, 5′-CCACAAGGCATTGAGACACAT−3′ and R, 5′-TCACCAGCAGCAGCAGCAG-3′; and rat-β-MHC F, 5′-AATGAACACCGGAGCAAGG-3′; and R, 5′-CGGGTCAGCTGAGAGATAAGAGC-3′. mRNA levels were quantified by relative quantification and analyzed using the 2−ΔΔCq method (24).

Detection of CaN, NFAT3 and β-MHC protein expression in myocardial cells by western blotting

Culture plates of H9C2 cells (1×104−1×105/ml) were washed with pre-cooled PBS and treated with pre-cooled lysis buffer (CWBio) on ice for 30 min. The lysis products were centrifuged for 20 min at 8,300 × g at 4°C to collect the supernatant. Furthermore, 2 µl supernatant was used to determine the protein concentration by using a BCA protein quantitative kit. Protein samples (50 µg/well) were separated by SDS-PAGE on a 10% gel. Electrophoresis was performed for 20 min at a constant voltage of 100 V, and for 40 min at a constant voltage of 140 V. Subsequently, the proteins were transferred to a PVDF membrane, followed by three TBS washes. Following the removal of TBS, the membranes were blocked using TBS with Tween-20 (TBST) buffer (20 mM Tris-HCl, 150 mM NaCl and 0.1% Tween-20) containing 5% non-fat milk for 2 h at room temperature. The membrane was then incubated with TBST buffer (20 mM Tris-HCl, 150 mM NaCl and 0.1% Tween-20) containing 5% non-fat milk with the following primary antibodies, polyclonal rabbit anti-human CaN (1:1,000), polyclonal rabbit anti-human NFAT3 (1:1,000), polyclonal rabbit anti-human β-MHC (1:1,000) and polyclonal rabbit anti-human β-actin, overnight at 4°C. Following three washes with 1X PBS, the PVDF membrane was incubated with a horseradish peroxide-conjugated anti-rabbit secondary antibodies (1:10,000; cat. no. 111-035-003; Jackson ImmunoResearch Laboratories, Inc.) at room temperature for 2 h. The PVDF membrane was washed five times with PBS (15 min each), and developed using an ECL kit (Beijing ComWin Biotech Co., Ltd.).

Statistical analysis

GraphPad Prism 7 (GraphPad Software, Inc.) was used for statistical analysis. Data were analyzed with two-way ANOVA adjusted for multiple comparisons. Multiple comparisons between the groups was performed using Bonferroni tests. The bilateral inspection level for α was 0.05, and P<0.05 was considered to indicate a statistically significant difference.

Results

Cell swelling analysis

The cell size in B1 and B2 groups was increased compared with cells in A1 and A2 groups after 48 and 72 h of culture under different conditions. Cell size was decreased following the addition of Norvasc compared with those without the addition of Norvasc under the same conditions. There was no significant difference between the C1 and C2 and A1 and A2 groups, respectively. Furthermore, there was no significant difference in cell size between the A1 and A2 groups, B1 and B2 groups, and C1 and C2 groups, respectively. The statistical analysis chart is presented in Fig. 1.

Determination of [Ca2+]i activity in each group

Following 48 and 72 h of culture, the fluorescent values of [Ca2+]i activity in B1, B2, C1 and C2 groups were increased compared with the A1 and A2 groups, respectively. Following the addition of Norvasc, the fluorescent value of [Ca2+]i activity in each group was notably lower than that without Norvasc under the same conditions. In addition, no obvious difference was found in the fluorescent values of intracellular [Ca2+]i activity between the A1 and A2 groups, B1 and B2 groups, and C1 and C2 groups, respectively. Images of fluorescent staining are presented in Fig. 2. The statistical analysis chart is presented in Fig. 3.

Cellular CaN enzyme activity

The CaN concentration was increased in the B1 and B2 groups compared with the A1 and A2 groups, following 48 and 72 h of culture, respectively. With the addition of Norvasc, the concentration of CaN in each group was significantly lower than that without Norvasc. The concentration standard curve is presented in Fig. 4A and the statistical analysis chart is presented in Fig. 4B.

CaN, NFAT3 and β-MHC mRNA expression levels of H9C2 cells determined by RT-qPCR
Changes in CaN mRNA expression in H9C2 cells

The mRNA expression of CaN in B1 and B2 groups was increased compared with that in the A1 and A2 groups, following 48 and 72 h of culture, respectively. Furthermore, the mRNA expression level of CaN was lower with the addition of Norvasc than that without Norvasc. The mRNA expression of CaN in the C1 and C2 groups had no significant difference between the A1 and A2 groups respectively. The statistical analysis chart is presented in Fig. 5.

Changes in NFAT3 mRNA expression in H9C2 cells

Following 48 and 72 h of culture, the mRNA expression of NFAT3 in the B1 and B2 groups was increased compared with the A1 and A2 groups, and expression in the C1 group was increased compared with the A1 group. The addition of Norvasc resulted in decreased NFAT3 mRNA expression than that without Norvasc. Additionally, no significant difference was observed in the mRNA expression level of NFAT3 in the C2 groups compared with that in the A2 groups. The statistical analysis chart is presented in Fig. 6.

Changes in β-MHC mRNA expression in H9C2 cells

A trend towards increased β-MHC mRNA expression was observed in B1 and B2 groups compared with the A1 and A2 groups following 48 and 72 h of culture, under different culture conditions. Following the addition of Norvasc, the mRNA expression of β-MHC was lower than that without Norvasc under the same conditions. The mRNA expression level of β-MHC in groups C1 and C2 were without any significant difference between groups A1 and A2, respectively. Furthermore, there was no statistical difference between the A1 and A2 groups or C1 and C2 groups. With the prolongation of culture time, a trend towards decreased β-MHC mRNA expression was observed in the B2 group compared with the B1 group. The statistical analysis chart is shown in Fig. 7.

Detection of CaN, NFAT3 and β-MHC protein expression in H9C2 cells by western blotting

Representative western blotting images are presented in Fig. 8.

Detection of CaN protein expression in H9C2 cells by western blotting

The protein expression of CaN in B1 and B2 groups was increased compared with the A1 and A2 groups, respectively, following 48 and 72 h of culture under different culture conditions. Protein expression of CaN was significantly decreased in groups C1 and C2 compared with the B1 and B2 groups, respectively, following the addition of Norvasc. There was no significant difference between the C1 and C2 groups and A1 and A2 groups, respectively. The statistical analysis chart is presented in Fig. 9.

Detection of NFAT3 protein expression in H9C2 cells by western blotting

The protein expression of NFAT3 was increased in the B1 and B2 groups compared with that of the A1 and A2 groups, respectively, following 48 and 72 h of culture under different culture conditions. However, the protein expression of NFAT3 was decreased with the addition of Norvasc compared to that without Norvasc, under the same conditions. The NFAT3 protein expression was also increased in the C1 and C2 groups compared with that in the A1 and A2 groups. The statistical analysis chart is presented in Fig. 10.

Detection of β-MHC protein expression in H9C2 cells by western blotting

The trend of increased protein expression of β-MHC was observed in the B1 and B2 groups compared with that in the A1 and A2 groups, respectively, following 48 and 72 h of culture under different culture conditions. The addition of Norvasc resulted in decreased β-MHC protein expression compared with that without the addition of Norvasc, under the same conditions. Additionally, the expression of β-MHC protein in the C1 and C2 groups was not significantly different with that in the A1 and A2 groups. There was no significant difference in the protein expression of β-MHC in group A and B and C with the prolongation of culture time. The statistical analysis chart is presented in Fig. 11.

Discussion

The World Health Organization predicts that 300 million individuals worldwide will be diagnosed with diabetes mellitus by 2025 (25). Patients with diabetes are prone to cardiovascular diseases, and the probability of myocardial infarction is 10 times higher than that of non-diabetic patients (26,27). First proposed by Rubler et al (28) in 1972, diabetic cardiomyopathy is a type of heart disease that is independent of coronary heart disease, valve disease and hypertensive heart disease, and is mainly characterized by diastolic dysfunction in the early stage and systolic dysfunction at the late stage. The pathogenesis is complex, with the main pathological changes including myocardial inflammation, metabolic disorders, myocardial cell apoptosis and fibrosis (29,30). The duration of hyperglycemia serves as an important indicator in determining the severity of heart failure caused by diabetic cardiomyopathy (31). Therefore, hyperglycemia is studied as an independent risk factor of myocardial injury caused by diabetic cardiomyopathy (32,33).

High glucose will stimulate cardiac hypertrophy in a variety of ways (3437). In the present study, it was found that high glucose could lead to an increase in the average individual volume of H9C2 cells. Research investigating the aberrant molecular processes that occur during cardiac hypertrophy has used primary cardiomyocytes from neonatal rat hearts as the standard experimental in vitro system. In addition, some studies have made use of the H9C2 rat cardiomyoblast cell line (38,39), as it was found by Watkins et al (40) that the H9C2 cell line and primary neonatal cardiomyocyte cells exhibit similar hypertrophic responses in vitro. Therefore, the H9C2 cell line was selected for the present study, which also has the advantage of being an animal-free alternative. However, this is also a limitation of the present study because primary neonatal cardiomyocyte cells were not used. The role of the Ca2+-CaN-NFAT3 signaling pathway in cardiac development has become a research area of interest in recent years. The activation of Ca2+-dependent CaN A subunit (CnA) has been frequently observed in human heart hypertrophy and heart failure (41,42). In mice, increased intracellular calcium is known to activate CnA, which can bind and dephosphorylate the NFAT transcription factor family of activated T-nuclear factors. Subsequently, NFAT is transferred from the cytoplasm to the nucleus, activating the specific expression of various genes associated with cardiac hypertrophy, such as atrial natriuretic factor, brain natriuretic peptide (BNP), and β-MHC in the heart. This leads to increased nucleic acid synthesis of myocardial cell protein and increased cardiac cell volume, and finally cardiac hypertrophy (43,44). Transgenic mice overexpressing a consistently active form of CnA-specific cardiomyocytes, MHC-CnA, have been found to suffer from cardiac hypertrophy at 18 days following birth, with varying degrees of progression to heart failure and sudden death (45).

Several different studies have found that the Ca2+-CaN-NFAT3 signaling pathway plays a role in cardiac hypertrophy. Daskoulidou et al (46) found that increased Orai 1 expression could mediate an increase in the calcium current of the store-operated calcium entry channel, in order to regulate calcium in cardiac cells, which may be induced by hyperglycemia through the activation of the CaN/NFATc3 signaling pathway. Somvanshi et al (47) revealed that the activation of the somatostatin receptor 2 (SSTR2) not only inhibited the expression and activity of CaN phosphatase, but also hindered the dephosphorylation of NFAT and nuclear translocation, which provided evidence that SSTR2 could protect the heart by regulating the Ca2+-associated signaling pathway, leading to cardiac hypertrophy. The T-type calcium channel, Cav3.2 could be induced by Egr1 (early growth response 1), which is released at the early stage of myocardial hypertrophy due to the early pressure overload, to regulate cardiac hypertrophy through the CaN phosphatase-NFAT signaling pathway (48). A previous study also revealed that the activity of the CaN signaling pathway may be activated by the bacteria Porphyromonas gingivalis, and further lead to cardiomyocyte hypertrophy and cell death of cultured H9C2 cardiomyocytes (49). The Ca2+-CaN-NFAT3 pathway is considered to be the crucial mechanism in mediating the development of cardiac hypertrophy and therefore, the role of the Ca2+-CaN-NFAT3 signaling pathway in hyperglycemia-induced myocardial hypertrophy was investigated in the present study. Following the determination of [Ca2+]i and mRNA and protein expression levels of CaN, NFAT and β-MHC, it was revealed that high glucose could induce an increase in [Ca2+]i and CaN concentration. Furthermore, high glucose increased the mRNA and protein levels of CaN, NFAT and β-MHC, and in combination with the cell morphology data, suggest the involvement of the Ca2+-CaN-NFAT signaling pathway in hyperglycemia-induced H9C2 cells.

The activation of CaN mainly depends on the increase in intracellular Ca2+, and its activity is regulated by the change in intracellular Ca2+ concentration (50). The continuous maintenance of NFAT in the nucleus requires a continuous and oscillating increase in calcium (51) in order to maintain the activated form of CaN phosphatase. This CaN-dependent signal is sensitive to the inhibition of calcium channel blockers, such as verapamil, or CaN inhibitors, such as cyclosporine A (CSA) and tacrolimus (FK-506) (5254). L-type Ca2+ channels are widely distributed in the heart, smooth and skeletal muscles (55,56). In the heart and smooth muscle cells, the L-type Ca2+ channels are responsible for the inward movement of Ca2+, thereby causing contraction. In skeletal muscles, these channels act as voltage sensors in excitation-contraction coupling. L-type calcium channels consist of several subunits, namely, α1, α2, β, γ and δ (14). The α1 subunit consists of four homologous structural domains, and contains transmembrane pores, through which calcium ions can be obtained, as well as calcium antagonist binding sites adjacent to the pores, which are connected to each other and to the calcium channel via allosteric junctions (57,58).

The L-type calcium channel is considered to be the main source of calcium that can activate CaN-NFAT3 signal transduction (59). Thus, L-type calcium channel inhibitors have become a research area of interest and an important therapeutic target for cardiac hypertrophy. Cardiac remodeling, which involves structural and functional changes at the molecular, cellular, tissue and whole-organ levels, can be used to determine the clinical course of heart failure (60). A number of studies have reported that amlodipine can reduce cardiac remodeling in spontaneously hypertensive rats and myocardial infarction in rats (61,62). Valsartan monotherapy as well as combination therapy with valsartan and either amlodipine or cilnidipine similarly attenuated hypertension and left ventricular hypertrophy (LVH) in hypertensive rats (22). Compared with amlodipine alone, amlodipine combined with atorvastatin had a greater beneficial effect on the myocardial hypertrophy. These benefits may be associated with the cumulative effect of the drug on inhibiting NADPH oxidase-mediated reactive oxygen species (63). Amlodipine has also been found to improve cardiomyocyte hypertrophy by inhibiting the phosphorylation of the epithelial growth factor receptor (64). Losartan, amlodipine, and particularly fosinopril can inhibit myocardial cell apoptosis, prevent myocardial fibrosis, and reverse cardiac hypertrophy; the inhibition of the cardiac renin-angiotensin-aldosterone system may be the mechanism of the cardioprotective effects of these three drugs (65). Meo et al (66) reported a decrease in L-type calcium current in cardiomyocytes in a glycosuria mouse model, induced by streptozotocin. In addition to the traditional L-type calcium channel, it is generally considered that Orai 1-mediated calcium store-operated calcium channel also participates in the calcium regulation of cardiomyocytes. In a diabetic model, Orai 1 expression was increased in both cardiomyocytes and smooth muscle cells (46,67). Amlodipine sulfonate is an L-type calcium channel blocker, which can selectively inhibit the transmembrane domain to inhibit calcium ions from entering cardiomyocytes. Several studies have demonstrated that amlodipine sulfonate could inhibit CaN-NFAT3 and thus inhibit cardiac hypertrophy (68,69). Therefore, the L-type calcium channel amlodipine bensulfonate, Norvasc, was selected to investigate whether intracellular influx of Ca2+ was observed in H9C2 cells cultured with high glucose, in order to induce changes in intracellular calcium concentration and the activation of CaN, thereby activating the signaling pathway, and ultimately causing cardiomyocyte hypertrophy.

The main purpose of the present study was to investigate whether Norvasc could induce the recovery of cardiomyocyte hypertrophy and inhibit Ca2+-CaN-NFAT3 signaling. Following the addition of Norvasc, the activity of [Ca2+]i, CaN concentration, CaN mRNA and protein expression in the corresponding groups were all significantly decreased, which indicated that Norvasc reduced CaN expression. Thus, intracellular calcium could activate CaN and promote its expression. The present study demonstrated that calcium channel inhibitors could alleviate cell hypertrophy and are more effective in inhibiting cell hypertrophy following prolonged action. Bugyei-Twum et al (70) also found that high glucose in vitro induced the activation of Smad in H9C2 cells and promoted cardiac fibrosis and hypertrophy through transcriptional coregulator p300, and Chen et al (71) revealed that lercanidipine may ameliorate cardiomyocyte hypertrophy, partially by blocking Cn-NFAT3 and CaMKII-HDAC4 signaling. These findings are consistent with the present study.

The signal transduction mechanism of myocardial hypertrophy is complex. In addition to Ca2+-CaN-NFAT3 signaling pathway, there may be other signaling pathways, which may have interactions with CaN-NFAT3. Thus, further studies are required, including the use of specific inhibitors, such as CSA, to identify other pathways. Liu et al (72) demonstrated that lipopolysaccharide (LPS) treatment leads to myocardial hypertrophy via the calcineurin/NFAT-3 signaling pathway in H9C2 cells. They further provided a link between the LPS-induced inflammatory response and the calcineurin/NFAT-3 signaling pathway that mediates the development of cardiac hypertrophy. LPS treatment was found to significantly promote the activation and nuclear translocation of NFAT3 and mediated the development of cardiac hypertrophy as a transcription factor. The cell size, actin fiber, atrial natriuretic peptide and BNP levels were assessed following the use of ERK1/2 inhibitors, p38 MAPK inhibitors, JNK 1/2 inhibitors, CaN inhibitors and NF-KB inhibitors, which demonstrated that only the CaN inhibitor could significantly inhibit NFAT3 nuclear localization. The present study provides findings that can be used to determine subsequent future studies.

In summary, the present experimental design and results suggest that, the calcium channel inhibitor Norvasc may inhibit hyperglycemia in H9C2 cells, which may result from the activation of the Ca2+-CaN-NFAT3 signaling pathway by high glucose levels.

Acknowledgements

The authors would like to thank Dr Shasha Han (Department of Pediatrics, The First Clinical Medical College of Jinan University, Guangzhou, China) for her technical support.

Funding

The present study was supported by NSFC (grant nos. 81741083, 81801492).

Availability of data and materials

The datasets used and/or analyzed are available from the corresponding author on reasonable request

Authors' contributions

XX, LR, XT, FP and GL made substantial contributions to conception and design of the study. XX, LR, XT, FP and CY provided resources, contributed to research and validation of the results, interpreted data and wrote the original draft preparation. XX, LR, FP and GL reviewed and edited the manuscript. XX, LR and GL supervised the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chan JC, Malik V, Jia W, Kadowaki T, Yajnik CS, Yoon KH and Hu FB: Diabetes in Asia: Epidemiology, risk factors, and pathophysiology. JAMA. 301:2129–2140. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U and Shaw JE: Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 103:137–149. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Lamblin N, Fertin M, De Groote P and Bauters C: Cardiac remodeling and heart failure after a first anterior myocardial infarction in patients with diabetes mellitus. J Cardiovasc Med (Hagerstown). 13:353–359. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Tochiya M, Makino H, Tamanaha T, Matsuo M, Hishida A, Koezuka R, Ohata Y, Tomita T, Son C, Miyamoto Y, et al: Effect of tofogliflozin on cardiac and vascular endothelial function in patients with type 2 diabetes and heart diseases: A pilot study. J Diabetes Investig. 11:400–404. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Sardu C, Barbieri M, Santamaria M, Giordano V, Sacra C, Paolisso P, Spirito A, Marfella R, Paolisso G and Rizzo MR: Multipolar pacing by cardiac resynchronization therapy with a defibrillators treatment in type 2 diabetes mellitus failing heart patients: Impact on responders rate, and clinical outcomes. Cardiovasc Diabetol. 16:752017. View Article : Google Scholar : PubMed/NCBI

6 

Kannel WB and McGee DL: Diabetes and cardiovascular disease: The Framingham study. JAMA. 241:2035–2038. 1979. View Article : Google Scholar : PubMed/NCBI

7 

Lind M, Bounias I, Olsson M, Gudbjörnsdottir S, Svensson AM and Rosengren A: Glycaemic control and incidence of heart failure in 20,985 patients with type 1 diabetes: An observational study. Lancet. 378:140–146. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Bayeva M, Sawicki KT and Ardehali H: Taking diabetes to heart-deregulation of myocardial lipid metabolism in diabetic cardiomyopathy. J Am Heart Assoc. 2:e4332013. View Article : Google Scholar

9 

Xu Y, Wang L, He J, Bi Y, Li M, Wang T, Wang L, Jiang Y, Dai M, Lu J, et al: Prevalence and control of diabetes in Chinese adults. JAMA. 310:948–959. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Kim HW, Ch YS, Lee HR, Park SY and Kim YH: Diabetic alterations in cardiac sarcoplasmic reticulum Ca2+-ATPase and phospholamban protein expression. Life Sci. 70:367–379. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Cai L, Li W, Wang G, Guo L, Jiang Y and Kang YJ: Hyperglycemia-induced apoptosis in mouse myocardium: Mitochondrial cytochrome C-mediated caspase-3 activation pathway. Diabetes. 51:1938–1948. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Way KJ, Isshiki K, Suzuma K, Yokota T, Zvagelsky D, Schoen FJ, Sandusky GE, Pechous PA, Vlahos CJ, Wakasaki H and King GL: Expression of connective tissue growth factor is increased in injured myocardium associated with protein kinase C beta2 activation and diabetes. Diabetes. 51:2709–2718. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Candido R, Forbes JM, Thomas MC, Thallas V, Dean RG, Burns WC, Tikellis C, Ritchie RH, Twigg SM, Cooper ME and Burrell LM: A breaker of advanced glycation end products attenuates diabetes-induced myocardial structural changes. Circ Res. 92:785–792. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Catterall WA: Structure and regulation of voltage-gated Ca2+ channels. Annu Rev Cell Dev Biol. 16:521–555. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Dolphin AC: Voltage-gated calcium channels: Their discovery, function and importance as drug targets. Brain Neurosci Adv. 2:23982128187948052018. View Article : Google Scholar : PubMed/NCBI

16 

Klingbeil AU, Schneider M, Martus P, Messerli FH and Schmieder RE: A meta-analysis of the effects of treatment on left ventricular mass in essential hypertension. Am J Med. 115:41–46. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Fagard RH, Celis H, Thijs L and Wouters S: Regression of left ventricular mass by antihypertensive treatment: A meta-analysis of randomized comparative studies. Hypertension. 54:1084–1091. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Puscas L, Gilau L, Coltau M, Pasca R, Domuta G, Baican M and Hecht A: Calcium channel blockers reduce blood pressure in part by inhibiting vascular smooth muscle carbonic anhydrase I. Cardiovasc Drug Ther. 14:523–528. 2000. View Article : Google Scholar

19 

Bangalore S, Fakheri R, Toklu B and Messerli FH: Diabetes mellitus as a compelling indication for use of renin angiotensin system blockers: Systematic review and meta-analysis of randomized trials. BMJ. 352:i4382016. View Article : Google Scholar : PubMed/NCBI

20 

Packer M, O'Connor CM, Ghali JK, Pressler ML, Carson PE, Belkin RN, Miller AB, Neuberg GW, Frid D, Wertheimer JH, et al: Effect of amlodipine on morbidity and mortality in severe chronic heart failure. Prospective randomized amlodipine survival evaluation study group. N Engl J Med. 335:1107–1114. 1996. View Article : Google Scholar : PubMed/NCBI

21 

Lu J, Hao J, Du H, Xiao B, Li Y, Yang X and Cui W: Amlodipine and atorvastatin improved hypertensive cardiac remodeling through regulation of MMPs/TIMPs in SHR RATS. Cell Physiol Biochem. 39:47–60. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Nagasawa K, Takahashi K, Matsuura N, Takatsu M, Hattori T, Watanabe S, Harada E, Niinuma K, Murohara T and Nagata K: Comparative effects of valsartan in combination with cilnidipine or amlodipine on cardiac remodeling and diastolic dysfunction in Dahl salt-sensitive rats. Hypertens Res. 38:39–47. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Whittaker P, Zhang HP and Kloner RA: Biphasic survival response to amlodipine after myocardial infarction in rats: Association with cardiac vascular remodeling. Cardiovasc Pathol. 2:85–93. 2000. View Article : Google Scholar

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta CT) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Verkhratsky A and Fernyhough P: Mitochondrial malfunction and Ca2+ dyshomeostasis drive neuronal pathology in diabetes. Cell Calcium. 1:112–122. 2008. View Article : Google Scholar

26 

Jouven X, Lemaître RN, Rea TD, Sotoodehnia N, Empana JP and Siscovick DS: Diabetes, glucose level, and risk of sudden cardiac death. Eur Heart J. 26:2142–2147. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Yeung EH, Pankow JS, Astor BC, Powe NR, Saudek CD and Kao WH: Increased risk of type 2 diabetes from a family history of coronary heart disease and type 2 diabetes. Diabetes Care. 30:154–156. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Rubler S, Dlugash J, Yuceoglu YZ, Kumral T, Branwood AW and Grishman A: New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am J Cardiol. 30:595–602. 1972. View Article : Google Scholar : PubMed/NCBI

29 

Bergner DW and Goldberger JJ: Diabetes mellitus and sudden cardiac death: What are the data? Cardiol J. 17:117–129. 2010.PubMed/NCBI

30 

Aharinejad S, Andrukhova O, Lucas T, Zuckermann A, Wieselthaler G, Wolner E and Grimm M: Programmed cell death in idiopathic dilated cardiomyopathy is mediated by suppression of the apoptosis inhibitor apollon. Ann Thorac Surg. 86:109–114. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Fang ZY, Schull-Meade R, Leano R, Mottram PM, Prins JB and Marwick TH: Screening for heart disease in diabetic subjects. Am Heart J. 149:349–354. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Boyer JK, Thanigaraj S, Schechtman KB and Pérez JE: Prevalence of ventricular diastolic dysfunction in asymptomatic, normotensive patients with diabetes mellitus. Am J Cardiol. 93:870–875. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Boudina S and Abel ED: Diabetic cardiomyopathy, causes and effects. Rev Endocr Metab Disord. 11:31–39. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Kanai M, Otsuka Y, Otsuka K, Sato M, Nishimura T, Mori Y, Kawaguchi M, Hatano E, Kodama Y, Matsumoto S, et al: A phase I study investigating the safety and pharmacokinetics of highly bioavailable curcumin (Theracurmin) in cancer patients. Cancer Chemother Pharmacol. 71:1521–1530. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Sasaki H, Sunagawa Y, Takahashi K, Imaizumi A, Fukuda H, Hashimoto T, Wada H, Katanasaka Y, Kakeya H, Fujita M, et al: Innovative preparation of curcumin for improved oral bioavailability. Biol Pharm Bull. 34:660–665. 2011. View Article : Google Scholar : PubMed/NCBI

36 

van Heerebeek L, Hamdani N, Handoko ML, Falcao-Pires I, Musters RJ, Kupreishvili K, Ijsselmuiden AJ, Schalkwijk CG, Bronzwaer JG, Diamant M, et al: Diastolic stiffness of the failing diabetic heart: Importance of fibrosis, advanced glycation end products, and myocyte resting tension. Circulation. 117:43–51. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Gonzalez-Quesada C, Cavalera M, Biernacka A, Kong P, Lee DW, Saxena A, Frunza O, Dobaczewski M, Shinde A and Frangogiannis NG: Thrombospondin-1 induction in the diabetic myocardium stabilizes the cardiac matrix in addition to promoting vascular rarefaction through angiopoietin-2 upregulation. Circ Res. 113:1331–1344. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Li MD, Cheng WP, Shi MX, Ge TD, Zheng XL, Wu DY, Hu XY, Luo JC, Li FL and Li H: Role of tRNA selenocysteine 1 associated protein 1 in the proliferation and apoptosis of cardiomyocyte-like H9c2 cells. Mol Med Rep. 15:988–994. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Saito M, Sakiyama K, Shiota T and Ito M: Isoproterenol produces a rapid increase in sialidase activity in rat heart tissue and cardiomyocyte-derived H9c2 cells in culture. FEBS Lett. 542:105–108. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Watkins SJ, Borthwick GM and Arthur HM: The H9C2 cell line and primary neonatal cardiomyocyte cells show similar hypertrophic responses in vitro. In Vitro Cell Dev Biol Anim. 47:125–131. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Diedrichs H, Chi M, Boelck B, Mehlhorn U and Schwinger RH: Increased regulatory activity of the calcineurin/NFAT pathway in human heart failure. Eur J Heart Fail. 6:3–09. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Haq S, Choukroun G, Lim H, Tymitz KM, del Monte F, Gwathmey J, Grazette L, Michael A, Hajjar R, Force T and Molkentin JD: Differential activation of signal transduction pathways in human hearts with hypertrophy versus advanced heart failure. Circulation. 103:670–677. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Zhu SJ, Yang YJ, Yu LJ and Huang L: CaN-NFAT3 signal pathway: A crucial hinge relates Ca2+ signal with cardiomyocyte hypertrophy. Zhonghua Nei Ke Za Zhi. 43:19–21. 2004.(In Chinese). PubMed/NCBI

44 

Wilkins BJ, Dai YS, Bueno OF, Parsons SA, Xu J, Plank DM, Jones F, Kimball TR and Molkentin JD: Calcineurin/NFAT coupling participates in pathological, but not physiological, cardiac hypertrophy. Circ Res. 94:110–118. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Molkentin JD, Lu JR, Antos CL, Markham B, Richardson J, Robbins J, Grant SR and Olson EN: A calcineurin-dependent transcriptional pathway for cardiac hypertrophy. Cell. 93:215–228. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Daskoulidou N, Zeng B, Berglund LM, Jiang H, Chen GL, Kotova O, Bhandari S, Ayoola J, Griffin S, Atkin SL, et al: High glucose enhances store-operated calcium entry by upregulating ORAI/STIM via calcineurin-NFAT signalling. J Mol Med (Berl). 93:511–521. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Somvanshi RK, Zou S, Qiu X and Kumar U: Somatostatin receptor-2 negatively regulates β-adrenergic receptor mediated Ca(2+) dependent signaling pathways in H9c2 cells. Biochim Biophys Acta. 1843:735–745. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Hsu SC, Chang YT and Chen CC: Early growth response 1 is an early signal inducing Cav3.2 T-type calcium channels during cardiac hypertrophy. Cardiovasc Res. 100:222–230. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Lee SD, Kuo WW, Lin DY, Chen TH, Kuo WH, Hsu HH, Chen JZ, Liu JY, Yeh YL and Huang CY: Role of calcineurin in Porphyromonas gingivalis-induced myocardial cell hypertrophy and apoptosis. J Biomed Sci. 13:251–260. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Strack S, Wadzinski BE and Ebner FF: Localization of the calcium/calmodulin-dependent protein phosphatase, calcineurin, in the hindbrain and spinal cord of the rat. J Comp Neurol. 375:66–76. 1996. View Article : Google Scholar : PubMed/NCBI

51 

Smedler E and Uhlén P: Frequency decoding of calcium oscillations. Biochim Biophys Acta. 1840:964–969. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Fric J, Zelante T, Wong AY, Mertes A, Yu HB and Ricciardi-Castagnoli P: NFAT control of innate immunity. Blood. 120:1380–1389. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Sommerer C, Meuer S, Zeier M and Giese T: Calcineurin inhibitors and NFAT-regulated gene expression. Clin Chim Acta. 413:1379–1386. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Maguire O, Tornatore KM, O'loughlin KL, Venuto RC and Minderman H: Nuclear translocation of nuclear factor of activated T cells (NFAT) as a quantitative pharmacodynamic parameter for tacrolimus. Cytometry A. 83:1096–1104. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Hess P: Calcium channels in vertebrate cells. Annu Rev Neurosci. 13:337–356. 1990. View Article : Google Scholar : PubMed/NCBI

56 

Rampe D and Triggle DJ: New synthetic ligands for L-type voltage-gated calcium channels. Prog Drug Res. 40:191–238. 1993.PubMed/NCBI

57 

Katz AM: Calcium channel diversity in the cardiovascular system. J Am Coll Cardiol. 28:522–529. 1996. View Article : Google Scholar : PubMed/NCBI

58 

Triggle DJ: Calcium-channel antagonists: Mechanisms of action, vascular selectivities, and clinical relevance. Cleve Clin J Med. 59:617–627. 1992. View Article : Google Scholar : PubMed/NCBI

59 

Wild AR, Sinnen BL, Dittmer PJ, Kennedy MJ, Sather WA and Dell'Acqua ML: Synapse-to-nucleus communication through NFAT is mediated by L-type Ca2+ channel Ca2+ spike propagation to the Soma. Cell Rep. 26:3537–3550.e4. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Cohn JN, Ferrari R and Sharpe N: Cardiac remodeling-concepts and clinical implications: A consensus paper from an international forum on cardiac remodeling. Behalf of an international forum on cardiac remodeling. J Am Coll Cardiol. 35:569–582. 2000. View Article : Google Scholar : PubMed/NCBI

61 

Yamazaki T, Komuro I, Zou Y, Kudoh S, Shiojima I, Mizuno T, Hiroi Y, Nagai R and Yazaki Y: Efficient inhibition of the development of cardiac remodeling by a long-acting calcium antagonist amlodipine. Hypertension. 31:32–38. 1998. View Article : Google Scholar : PubMed/NCBI

62 

Sandmann S, Claas R, Cleutjens JP, Daemen MJ and Unger T: Calcium channel blockade limits cardiac remodeling and improves cardiac function in myocardial infarction-induced heart failure in rats. J Cardiovasc Pharmacol. 37:64–77. 2001. View Article : Google Scholar : PubMed/NCBI

63 

Lu JC, Cui W, Zhang HL, Liu F, Han M, Liu DM, Yin HN, Zhang K and Du J: Additive beneficial effects of amlodipine and atorvastatin in reversing advanced cardiac hypertrophy in elderly spontaneously hypertensive rats. Clin Exp Pharmacol Physiol. 36:1110–1119. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Liao Y, Asakura M, Takashima S, Kato H, Asano Y, Shintani Y, Minamino T, Tomoike H, Hori M and Kitakaze M: Amlodipine ameliorates myocardial hypertrophy by inhibiting EGFR phosphorylation. Biochem Biophys Res Commun. 327:1083–1087. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Yu GL, Liang XQ and Zheng JQ: Contrast of losartan, fosinopril and amlodipine on cardiomyocyte apoptosis and left ventricular remolding in hypertensive rats. Hunan Yi Ke Da Xue Xue Bao. 26:405–408. 2001.(In Chinese). PubMed/NCBI

66 

Meo M, Meste O, Signore S, Sorrentino A, Cannata A, Zhou Y, Matsuda A, Luciani M, Kannappan R, Goichberg P, et al: Reduction in Kv current enhances the temporal dispersion of the action potential in diabetic myocytes: Insights from a novel repolarization algorithm. J Am Heart Assoc. 5:e0030782016. View Article : Google Scholar : PubMed/NCBI

67 

Sung HH, Kam SC, Lee JH, Chae MR, Hong C, Ko M, Han DH, So I and Lee SW: Molecular and functional characterization of ORAI and STIM in human corporeal smooth muscle cells and effects of the transfer of their dominant-negative mutant genes into diabetic rats. J Urol. 187:1903–1910. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Liu CZ, Tan JX, Wang Y, Huang YG and Huang DL: L-type calcium channel blocker suppresses calcineurin signal pathway and development of right ventricular hypertrophy. J Formos Med Assoc. 104:798–803. 2005.PubMed/NCBI

69 

Zou Y, Yamazaki T, Nakagawa K, Yamada H, Iriguchi N, Toko H, Takano H, Akazawa H, Nagai R and Komuro I: Continuous blockade of L-type Ca2+ channels suppresses activation of calcineurin and development of cardiac hypertrophy in spontaneously hypertensive rats. Hypertens Res. 25:117–124. 2002. View Article : Google Scholar : PubMed/NCBI

70 

Bugyei-Twum A, Advani A, Advani SL, Zhang Y, Thai K, Kelly DJ and Connelly KA: High glucose induces Smad activation via the transcriptional coregulator p300 and contributes to cardiac fibrosis and hypertrophy. Cardiovasc Diabetol. 13:892014. View Article : Google Scholar : PubMed/NCBI

71 

Chen Y, Yuan J, Jiang G, Zhu J, Zou Y and Lv Q: Lercanidipine attenuates angiotensin II-induced cardiomyocyte hypertrophy by blocking calcineurin-NFAT3 and CaMKII-HDAC4 signaling. Mol Med Rep. 16:4545–4552. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Liu CJ, Cheng YC, Lee KW, Hsu HH, Chu CH, Tsai FJ, Tsai CH, Chu CY, Liu JY, Kuo WW and Huang CY: Lipopolysaccharide induces cellular hypertrophy through calcineurin/NFAT-3 signaling pathway in H9c2 myocardiac cells. Mol Cell Biochem. 313:167–178. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 22 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu X, Ruan L, Tian X, Pan F, Yang C and Liu G: Calcium inhibitor inhibits high glucose‑induced hypertrophy of H9C2 cells. Mol Med Rep 22: 1783-1792, 2020
APA
Xu, X., Ruan, L., Tian, X., Pan, F., Yang, C., & Liu, G. (2020). Calcium inhibitor inhibits high glucose‑induced hypertrophy of H9C2 cells. Molecular Medicine Reports, 22, 1783-1792. https://doi.org/10.3892/mmr.2020.11275
MLA
Xu, X., Ruan, L., Tian, X., Pan, F., Yang, C., Liu, G."Calcium inhibitor inhibits high glucose‑induced hypertrophy of H9C2 cells". Molecular Medicine Reports 22.3 (2020): 1783-1792.
Chicago
Xu, X., Ruan, L., Tian, X., Pan, F., Yang, C., Liu, G."Calcium inhibitor inhibits high glucose‑induced hypertrophy of H9C2 cells". Molecular Medicine Reports 22, no. 3 (2020): 1783-1792. https://doi.org/10.3892/mmr.2020.11275