Open Access

Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway

  • Authors:
    • Xiaoying Wang
    • Kai He
    • Linlin Ma
    • Lan Wu
    • Yan Yang
    • Yanfei Li
  • View Affiliations

  • Published online on: August 9, 2022     https://doi.org/10.3892/mmr.2022.12822
  • Article Number: 306
  • Copyright: © Wang 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

Myocardial hypertrophy (MH) is an independent risk factor for cardiovascular disease, which in turn lead to arrhythmia or heart failure. Therefore, attention must be paid to formulation of therapeutic strategies for MH. Puerarin is a key bioactive ingredient isolated from Pueraria genera of plants that is beneficial for the treatment of MH. However, its molecular mechanism of action has not been fully determined. In the present study, 40 µM puerarin was demonstrated to be a safe dose for human AC16 cells using Cell Counting Kit‑8 assay. The protective effects of puerarin against MH were demonstrated in AC16 cells stimulated with isoproterenol (ISO). These effects were characterized by a significant decrease in surface area of cells (assessed using fluorescence staining) and mRNA and protein expression levels of MH‑associated biomarkers, including atrial and brain natriuretic peptide, assessed using reverse transcription‑quantitative PCR and western blotting, as well as β‑myosin heavy chain mRNA expression levels. Mechanistically, western blotting demonstrated that puerarin inhibited activation of the Wnt signaling pathway. Puerarin also significantly decreased phosphorylation of p65; this was mediated via crosstalk between the Wnt and NF‑κB signaling pathways. An inhibitor (Dickkopf‑1) and activator (IM‑12) of the Wnt signaling pathway were used to demonstrate that puerarin‑mediated effects alleviated ISO‑induced MH via the Wnt signaling pathway. The results of the present study demonstrated that puerarin pre‑treatment may be a potential therapeutic strategy for preventing ISO‑induced MH and managing MH in the future

Introduction

Cardiovascular disease (CVD) is the leading cause of human mortality worldwide accounted for 32.9% of all deaths with a steadily reaching of the number of CVD deaths from 12.1 million in 1990 to 18.6 million in 2019 (1), and places a notable burden on human health (2,3). At present, management of patients with CVD is challenging due to the high mortality, disability rates and rising health care costs (4). According to the World Health Organization, three-quarters of CVD deaths could be prevented through early detection and lifestyle interventions (5). As a result, CVD has attracted the attention of research communities and healthcare professionals to ensure that measures, including developing the care of cardiovascular disease in the ambulatory setting, improving approaches to the prevention of cardiovascular disease, expanding digital health interventions and universal health coverage (6), have been taken. Pharmaceutical developments and technological improvements are used in the Western world to decrease CVD-associated mortality, alongside interventions to control risk factors for CVD (7). Among risk factors for CVD, pathological myocardial hypertrophy (MH) caused by long-term overload is an independent risk factor for adverse cardiovascular events (8). Therefore, reversing MH may minimize the risk of cardiovascular events.

MH refers to an adaptive and compensatory change in heart structure that maintains cardiac output during adverse stimulation (9). The hypertrophic process is categorized as physiological or pathological. Physiological hypertrophy is key to preservation of normal heart structure and function; it is a tightly regulated but reversible phenomenon, such as the hypertrophic process that occurs in the heart following continuous exercise (10). This is a compliant change (an adaptive response to stimuli) in the heart and causes negligible effects in the body. However, pathological hypertrophy is associated with fibrosis, increased proinflammatory cytokine production and cellular dysfunction (11). This type of maladaptive cardiac hypertrophy, typically caused by chronically high, continuous hemodynamic pressure, is irreversible and leads to heart failure in severe cases (12).

Angiotensin-converting enzyme inhibitors (13,14), angiotensin II receptor blockers (15,16), calcium channel antagonists, β-receptor blockers and diuretics (17,18) are currently the primary treatments for MH. They have all been reported to improve the clinical symptoms of patients to a certain extent; however, the single-target therapeutic efficacy of these agents remains unsatisfactory as none of them significantly decrease mortality rate (19). Therefore, MH needs to be addressed in the CVD research. Traditional Chinese medicine (TCM) has been reported to improve the symptoms and quality of life of patients with heart failure (20). It has been reported that TCM confers therapeutic benefits using natural, biologically active ingredients that have multiple targets (21). Furthermore, agents such as Radixastragali (Huangqi) and Ginseng (Renshen) (22,23) have gained attention due to their stable therapeutic effects by targeting multiple targets underlying the development of MH. Astragaloside IV(AS-IV) which is the main component of Radixastragali, improves cardiac function and ameliorate MH, which is mediated by attenuating inflammatory reaction (24) and inactivating TANK-binding kinase 1 (TBK1)/PI3K/AKT signaling pathway (25). The pharmacological properties of ginseng are mainly attributed to ginsenosides which plays a robust antihypertrophic role via inhibition of NHE-1-dependent calcineurin activation (26) and the suppression of MCT-activated calcineurin and ERK signaling pathways (27).

Based on clinical experience and systematic theory, advances have been achieved in the research of TCM formulas, extracts and compounds (28). For example, research and development of ‘compound Danshen dropping pill’ has contributed to treatment of coronary heart disease and angina pectoris (29). Numerous compounds isolated from TCM products have important properties, such as the antimalarial effects of artemisinin (30) and the anti-inflammatory properties of licorice extract, which contains 3 triterpenes and 13 flavonoids (31). Furthermore, the curative effects of novel TCM compounds that exert specific pharmacological effects may be enhanced by elucidating their mechanism individually whilst also characterizing their target profile. Ultimately, treatment strategies for MH may be developed using the multi-faceted profiles of TCM compounds combined with metabolomics and traditional pharmacological methods (32).

Puerarin (PU) is an isoflavone chemical component (33) found within the Pueraria genera of plants (34). PU has been reported to exert numerous therapeutic effects, including anti-cancer, vasodilatory and heart protective effects (35). Furthermore, it has been previously reported that PU ameliorates pressure overload-induced MH in ovariectomized rats via activation of the peroxisome proliferator-activated receptor-α/peroxisome proliferator-activated receptor γ-1 signaling pathway and regulation of energy metabolism remodeling (36). However, the multi-target characteristics of TCM monomers suggest that PU has multiple therapeutic targets (37). Numerous signaling pathways are associated with the pathophysiology of MH, including the 5′AMP-activated protein kinase/mTOR (38), PI3K/AKT/mTOR (39) and insulin-like growth factor 1/PI3K/AKT (40) signaling pathways. Therefore, the exploration of the mechanism of PU on MH to achieve the effect of precise treatment is urgent. Numerous studies have reported that the Wnt signaling pathway, which is activated in early embryonic development to promote cardiac development and silenced in adulthood to maintain normal cardiac function, serves an important role in CVD (41,42). As experimental data on the effects of PU on the Wnt signaling pathway in the context of MH remain incomplete, the present study was preformed to address this.

Materials and methods

Chemicals and reagents

PU (purity, 99.00%) was purchased from TargetMol Chemicals, Inc. Isoproterenol hydrochloride (ISO), Dickkopf-1 (DKK1) and IM-12 were purchased from MedChemExpress. Cell Counting Kit-8 (CCK-8) assay was purchased from Dojindo Molecular Technologies, Inc. PVDF membranes, RIPA buffer, Actin-Tracker Green (microfilament green fluorescent probe), DAPI Staining Solution and BCA Protein Assay kit were purchased from Beyotime Institute of Biotechnology. TransScript® Green Two-Step qRT-PCR SuperMix kit was purchased from TransGen Biotech Co., Ltd. Antibodies against low-density lipoprotein receptor-related protein 6 (LRP6; cat. no. 3395), phosphorylated (p-)p65 (cat. no. 3033), NF-κB p65 (cat. no. C22B4), Tubulin (cat. no. 2128S), β-catenin (cat. no. 8480), Wnt5a/b (cat. no. 2530), c-Myc (cat. no. 5605), glycogen synthase kinase-3β (GSK3β; cat. no. 12456) and p-GSK3β (cat. no. 9322) were purchased from Cell Signaling Technology, Inc. Atrial natriuretic peptide (ANP; cat. no. ab189921) and brain natriuretic peptide (BNP, cat. no. ab236101) were purchased from Abcam. GAPDH (cat. no. BM1623) and the secondary antibodies including HRP Conjugated AffiniPure Goat Anti-Rabbit IgG (cat. No. BA1054) and HRP Conjugated AffiniPure Mouse Anti-Human IgG (cat. No. BM2002) was from Boster.

Cells

The human cardiomyocyte AC16 cell line (MingzhouBio Inc., cat. no. MZ-4038) was used. Cells were cultured in culture flasks with DMEM (BasalMedia Inc.) at 37°C under 5% CO2 in a humidified atmosphere. When cells reached the logarithmic growth phase, they were evenly distributed into six-well plates. Experimental treatments were performed after 24 h.

In vitro cardiac hypertrophy model and drug treatment

Cells were randomly divided as follows: i) Control, cells treated with DMEM; ii) ISO, cells treated with 10 µM ISO for 24 h; iii) PU, cells treated with 40 µM PU for 6 h followed by 10 µM ISO for 24 h; iv) DKK1, cells treated with 780 nM DKK1 for 6 h followed by 10 µM ISO for 24 h; v) IM12, cells treated with 30 nM IM-12 for 24 h; vi) PU + IM12, cells treated with 40 µM PU for 6 h followed by 30 nM IM-12 for 24 h and vi) PU + ISO + IM12, cells treated with 40 µM PU for 6 h followed by 10 µM ISO for 24 h and 30 nM IM-12 for 6 h. A total of 1.5×105 cells were plated in culture flasks with DMEM at 37°C under 5% CO2 in a humidified atmosphere. The cells were washed three times with PBS before addition of new drugs to avoid mixing of different drug components.

CCK-8 assay

PU was dissolved in DMSO and DMEM to produce a range of concentrations (5, 10, 20, 40 and 80 µM) for subsequent use. The final DMSO content was 0.2%. Cells were evenly dispersed into six groups containing different concentrations of PU (0, 5, 10, 20, 40 and 80 µM) and inoculated into a 96-well plate (~5,000 cells/well). PU incubation was performed for 6 h at 37°C under 5% CO2 before 100 µl CCK-8 reagent was added, followed by incubation for 2 h at 37°C under 5% CO2 in a humidified atmosphere. The 96-well plate was assessed using a PT-3502PC microplate reader (Beijing Potenov Technology Co. Ltd.) to quantify the absorption value of each well at 450 nm. A line chart was constructed plotting optical density (OD) values against drug concentrations to assess the effect of PU on cell viability.

Cytoskeletal staining

The surface area of AC16 cardiomyocytes in the different treatment groups was assessed using cytoskeletal staining. Control, ISO and PU), cells were washed with PBS twice and fixed using 3.7% formaldehyde solution at room temperature for 10 min in the dark according to the manufacturer's protocol for the Actin-Tracker Green. PBS containing 0.1% Triton X-100 was added to permeabilize the fixed cells for 10 min at room temperature. The cells were stained with Actin-Tracker Green working solution diluted with PBS containing 1–5% BSA and 0.1% Triton X-100 at a ratio of 1:50 for 30 min at room temperature. Following rinsing with PBS, 5 mg/ml DAPI (sufficient to cover each section) was added to each well for 3–5 min at room temperature before imaging using a fluorescence microscope (Olympus Corporation). The magnification was 400×. A total of five non-overlapping fields located at the upper and lower left and right and center of each well was selected. The cells in each field were numbered and sampled by a random number generator. A total of four cells was randomly extracted from each field. Finally, the size of cells was quantified using ImageJ2 (version 1.53; National Institutes of Health). The mean area of the selected cells was determined for each well and normalized to the control.

Reverse transcription-quantitative (RT-q)PCR

RNA was extracted from AC16 cells (Control, ISO and PU) using TRIzol® (Thermo Fisher Scientific, Inc.). The concentration of RNA was detected using a NanoDrop® spectrophotometer (Thermo Fisher Scientific, Inc.). Complementary DNA synthesis, using incubation at 42°C for 15 min and heating at 85°C for 5 sec, was performed according to the manufacturer's protocols of the TransScript® Green Two-Step qRT-PCR SuperMix kit. The primer sequences were as follows: ANP forward (F), 5′-CAACGCAGACCTGATGGATTT-3′ and reverse (R), 5′-AGCCCCCGCTTCTTCATTC-3′; BNP F, 5′-TGGAAACGTCCGGGTTACAG-3′ and R, 5′-CTGATCCGGTCCATCTTCCT-3′; β-myosin heavy chain (β-MHC) F, 5′-TCACCAACAACCCCTACGATT-3′ and R, 5′-CTCCTCAGCGTCATCAATGGA-3′ and GAPDH F, 5′-CTGGGCTACACTGAGCACC-3′ and R, 5′-AAGTGGTCGTTGAGGGCAATG-3′. The reaction systems of each primer sequence were added into a 96-well fluorescent qPCR plate and CFX Connect Real-Time PCR Detection System (Bio-Rad Laboratories, Inc.). The thermocycling conditions were as follows: 94°C initial denaturation for 30 sec, followed by 40 cycles of 5 sec at 94°C and 30 sec at 60°C. The 2−ΔΔCq method (43) was used to assess mRNA expression levels.

Western blotting

The cells in each group (Control, ISO, PU, DKK1, IM12, PU + IM12, PU + ISO + IM12) were digested with 0.25% trypsin for 25 sec at 37°C under 5% CO2 in a humidified atmosphere, washed with PBS three times and lysed with RIPA lysis buffer containing protease and phosphate inhibitors at 4°C for 30 min. The supernatant was collected following centrifugation (12,000 × g at 4°C for 10 min), before protein concentration was quantified via BCA assay. An equal amount of protein (15 µg protein/lane) was separated by 10% SDS-PAGE and transferred onto PVDF membranes. PVDF membranes were blocked with 5% skimmed milk powder solution at room temperature for 1 h before being incubated at 4°C overnight with specific primary antibodies. The antibodies were as follows: LRP6 (1:1,000), p-p65 (1:1,000), p65 (1:1,000), Tubulin (1:10,000), β-catenin (1:1,000), Wnt5a/b (1:1,000), GAPDH (1:5,000), c-Myc (1:1,000), GSK3β (1:1,000), p-GSK3β (1:1,000), ANP (1:1,000) and BNP (1:1,000). After washing with TBST (0.1% Tween-20), membranes were incubated with anti-rabbit (1:5,000) and anti-mouse secondary antibodies (1:5,000) at room temperature for 1 h. The membranes were rinsed again and treated with BeyoECL Moon (cat. no. P0018FS, Beyotime Institute of Biotechnology) before bands were visualized using the Tanon-4600 Chemiluminescence Imaging System. Finally, blots were analyzed using ImageJ2 (version 1.53; National Institutes of Health) to semi-quantify the protein expression levels.

Statistical analysis

All statistical analysis was performed using GraphPad Prism (version, 9.1.1; GraphPad Software, Inc.). Comparisons between groups were performed using one-way ANOVA with Dunnett's (Fig. 1, Fig. 2, Fig. 3, Fig. 4, Fig. 5, Fig. 6, Fig. 7) or Tukey's post hoc test (Fig. 8). Data are presented as the mean ± SEM of ≥3 independent experimental repeats. P<0.05 was considered to indicate a statistically significant difference.

Results

PU is not toxic to cardiomyocytes

CCK-8 assay OD values were used to assess the effects of PU on AC16 cell viability. The results demonstrated no significant difference in cell viability between treatment groups (Fig. 1). These data demonstrated that the highest concentration of PU (80 µM) had little effect on cell viability after 6 h.

PU decreases ANP, BNP and β-MHC transcription

The mRNA expression levels of MH markers ANP, BNP and β-MHC were quantified using RT-qPCR. The results demonstrated significant increases in mRNA expression levels of ANP (P<0.05; Fig. 2A), BNP (P<0.01; Fig. 2B) and β-MHC (P<0.01; Fig. 2C) in the ISO group compared with the control. By contrast, treatment with PU significantly decreased mRNA expression levels of ANP (P<0.05), BNP (P<0.01) and β-MHC (P<0.05) compared with the ISO group. These results suggested that the ISO-induced MH model was successfully established and the effect of ISO was reversed by PU.

PU decreases protein expression levels of ANP and BNP

To assess the effect of PU on ISO-induced MH, western blotting was performed to determine protein expression levels of ANP and BNP. Following 24 h induction with 10 µM ISO, protein expression levels of ANP and BNP in the AC16 cardiomyocytes were significantly higher compared with those in the control (P<0.01; Fig. 3). However, expression levels of ANP and BNP protein in AC16 cardiomyocytes pretreated with PU were significantly lower compared with those in the ISO group (P<0.01). These results demonstrated that cells pretreated with PU were resistant to ISO-induced MH.

ISO-treated AC16 cell surface area decreases significantly following PU pretreatment

Changes in AC16 myocardial cell area were observed using fluorescence microscopy. The surface area of AC16 cells in the ISO group significantly increased compared with those in the control (P<0.01; Fig. 4A, B and D). However, PU pretreatment significantly decreased surface area of AC16 cells compared with those in the ISO group (P<0.05; Fig. 4C). These results suggested that MH was prevented by PU pretreatment.

PU inhibits the Wnt/β-catenin signaling pathway

To determine the potential anti-MH mechanism of PU, core components in the Wnt/β-catenin and NF-κB signaling pathways were assessed using western blotting. The protein expression levels of LRP6, β-catenin, Wnt-5a/b and c-Myc and phosphorylation levels of p65 and GSK3β, significantly increased in the ISO group compared with the control group (P<0.01; Fig. 5A-F and H-K). However, the expression of GSK3β was significantly decreased by ISO compared with the control group (P<0.01; Fig. 5G). There was a significant decrease in the expression of LRP6 (P<0.05), β-catenin (P<0.01), Wnt-5a/b (P<0.01) and c-Myc (P<0.01) as well as phosphorylation levels of p65 (P<0.01) and GSK3β (P<0.01) in the AC16 cells pretreated with PU compared with the ISO group. Furthermore, expression of GSK3β (P<0.01) was significantly increased by PU pretreatment compared with the ISO group. These results suggested that the protective effects of PU on MH may be mediated by Wnt and NF-κB signaling pathway activity.

PU serves as an inhibitor of the Wnt signaling pathway by antagonizing its activator

A Wnt inhibitor and activator were used to verify if PU regulated the Wnt signaling pathway. DKK1 is an inhibitor of the Wnt signaling pathway that has been reported to regulate embryonic development (44). Wnt signaling transduction is blocked by DKK1 binding, which induces LRP6 internalization (45).

After cells were pretreated with DKK1, protein expression levels of components in the Wnt signaling pathway, such as LRP6, β-catenin, Wnt5a/b and c-Myc, were significantly suppressed compared with ISO group (P<0.01; Fig. 6A-G); this effect was similar to the aforementioned effect mediated by PU. Furthermore, expression of ANP, a cardiac hypertrophy marker, was significantly decreased by DKK1 pretreatment (P<0.01; Fig. 6G), which suggested that inhibition of the Wnt signaling pathway may be beneficial for the treatment of MH.

IM-12 enhances Wnt signaling, primarily by inhibiting GSK-3β (46). In the present study, IM-12 significantly decreased GSK3β expression compared with the control (P<0.01; Fig. 7G). The protein expression levels of LRP6, β-catenin, Wnt-5a/b and c-Myc were significantly increased by IM-12 compared with the control (P<0.01; Fig. 7A-F). However, protein expression levels of LRP6 (P<0.05), β-catenin (P<0.01), Wnt-5a/b (P<0.01) and c-Myc (P<0.01) were significantly decreased by PU pretreatment. These results suggested that PU suppressed IM-12-induced activation of the Wnt/β-catenin signaling pathway.

The effect of IM-12 on expression of markers in the Wnt signaling pathway and ISO-induced MH in the presence of PU pretreatment were assessed (Fig. 8). Following treatment with IM-12, PU-induced inhibition of Wnt signaling pathway protein expression was partially reversed. With the exception of protein expression of GSK3β, which was increased by PU but significantly reversed by IM-12 compared with the PU + ISO group, expression levels of all proteins in the PU + ISO + IM12 group were significantly increased compared with PU group (P<0.01). Moreover, the inhibitory effects of PU on the MH marker ANP was partially but significantly reversed by IM-12 treatment compared with the PU + ISO group (P<0.01). These results demonstrated that the protective effects of PU on MH may be mediated by inhibition of the Wnt signaling pathway.

Discussion

Despite therapeutic advances, there is a lack of effective therapeutic treatment strategies for MH. Pathological cardiac hypertrophy is induced by various factors, including pressure (47) and volume overload (48) and pharmaceutical induction, such as induction using angiotensin II (49) and ISO (50). ISO is a β-receptor agonist that causes acute myocardial contraction, ischemia, hypoxia and cause damage to myocardial cells due to production of a large number of peroxynitrite and oxygen free radicals in rats (51). Prabhu et al (52) reported that activities of myocardial mitochondrial cathepsin-D and β-glucuronidase are significantly decreased by high-dose ISO and that ISO decreases stability of the myocardial mitochondrial membrane. ISO is widely used to induce MH (50,53). There is evidence demonstrating that ISO stimulation leads to MH (53). ISO activates MAPK in cardiomyocytes and phosphorylates Raf/MEK/ERK signaling pathway components (54). The increase of p-ERK1/2 protein expression levels is a downstream effect of ISO-induced protein kinase C ε activation, which leads to MH (55). A previous study reported that chronic infusion of ISO results in compensatory cardiac hypertrophy associated with protein kinase A activity and N-terminal cleaved histone deacetylase production (56). The mechanism of ISO-induced myocardial injury may be associated with oxidative stress, calcium overload, inflammation and lipid peroxidation (57). It has been confirmed that in ISO-induced MH, ISO treatment markedly increases reactive oxygen species levels in cardiomyocytes (58), activates the NF-κB signaling pathway in AC16 cells and induces an inflammatory response (59). Further studies are required to determine the mechanism underlying ISO-induced MH. Based on a previous study (60), 10 µM ISO was chosen as the induction concentration for the present study.

In failing human hearts, there is a significant increase in both mRNA and protein expression of ANP, BNP and β-MHC (61); whereas expression of these three genes is normally stable in normal cardiomyocytes (62,63). A previous study reported that PU decreases mRNA expression levels of MH markers ANP and BNP in a dose-dependent manner, with 40 µM being the optimal concentration (64). Combined with the results from the present CCK-8 assay, 40 µM was selected as the concentration to induce MH for subsequent experimentation in the present study. It has been reported that the early stage of MH is characterized by the increase of the surface area of cardiomyocytes (65). Changes in cell surface area (quantified and analyzed using immunofluorescence) directly reflect progression of cardiac hypertrophy; this method is used to assess MH. In a previous study, H9c2 cells were stained with crystal violet and surface area was observed as a measure of MH to confirm that the surface area of CD38 knockdown cells was not enlarged (66). Another study used wheat germ agglutinin staining to evaluate the size of cardiomyocytes from heart sections (67). A previous study used surface area of cells stained with gentian violet to assess MH (68). Studies have also aimed to use cell surface area to assess the role of miR-339-5p in cardiomyocyte hypertrophy (69) and determine the therapeutic potential of pyrroloquinoline quinone for ISO-induced cardiac hypertrophy (59). In the present study, pretreatment with PU was demonstrated to exert protective effects against ISO-induced MH, demonstrated by decreased area of cardiomyocytes and expression of markers of cardiac hypertrophy. Therefore, PU may be a promising clinical agent for the prevention and treatment of MH, although its mechanism remains to be fully elucidated.

The signal transduction mechanism underlying MH is complex. Inflammation (70) and oxidative stress (71) have both been previously demonstrated to cause alterations leading to MH and heart failure. A previous systematic study of Wnt on a genome-wide level reported an association between MH and the Wnt expression profile (72). The Wnt signaling pathway has also been reported to be associated with cardiac disease; excessive stimulation of Wnt signaling is detrimental to cardiovascular pathology (42). PU has been reported to demonstrate anti-inflammatory and antioxidant activities (73). However, the potential effects of PU on MH and the Wnt signaling pathway in cardiomyocytes remain poorly understood. Therefore, the present study aimed to explore if PU demonstrate effects against MH in an in vitro model, with specific focus on the Wnt/β-catenin signaling pathway.

The Wnt signal transduction pathway is divided into classical and non-classical branches (74). The present study verified the expression of associated proteins and demonstrated that PU inhibited ISO-induced MH by blocking the classical branch of the Wnt/β-catenin signaling pathway. Wnt signaling is typically activated by the membrane receptor complex consisting of frizzled (Fzd), LRP5/6 and disheveled (Dvl) (75). Following activation of the Wnt receptor, Fzd binds to Wnt protein and LRP5/6 to form a receptor complex (76). Dvl is recruited by the receptor complex and activated via phosphorylation before binding to the β-catenin degradation complex, which separates β-catenin from the degradation complex to promote stabilization and aggregation before β-catenin translocates into the nucleus (77). β-catenin is a core component of the Wnt/β-catenin signaling pathway and is reported to be upregulated in cardiomyocytes from patients with acute myocardial infarction and heart tissue of spontaneously hypertensive rats (78). However, GSK3β mediates both β-catenin-dependent and -independent cascades (79). A previous study reported that GSK3β exerts negative regulatory effects on the Wnt signaling pathway (80). In the nucleus, β-catenin binds to T cell factor/lymphoid enhancer factor (Tcf/Lef) to activate transcription of Wnt target genes (81). c-Myc and Tcf/Lef are downstream targets of the Wnt signaling pathway; a previous study reported that activity of these targets is inhibited by low expression of β-catenin (82).

In the present study, western blotting was used to assess expression of core proteins in the Wnt signaling pathway; expression levels of these proteins in the ISO group was significantly increased but significantly reversed by PU pretreatment. However, GSK3β protein expression levels exhibited an opposite trend, which is likely due to its negative regulatory mechanism of Wnt; following activation of Wnt signaling, activity of GSK3β is hampered (83). A previous study reported that decreased expression of GSK3β in cardiac fibroblasts that led to adverse ventricular remodeling (84). Therefore, inhibition of GSK3β may increase the activity of Wnt signaling, thereby leading to cardiac hypertrophy. PU induces expression of GSK3β, as demonstrated in the present study, and serves a protective role. Furthermore, numerous studies have demonstrated that the Wnt/β-catenin and NF-κB signaling pathways exhibit crosstalk and this interaction is enhanced by the inflammatory response (85,86). In the present study, it was demonstrated that, in the ISO group, the protein expression of Wnt5a/b and β-catenin and the phosphorylation levels of p65 were significantly increased compared with the control but significantly decreased by pretreatment with PU. These observations are in accordance with those in previous studies which suggested that there may be synergy between the two signaling pathways during the development of MH (87,88). However, more evidence is needed to support the synergistic effect.

To verify whether inhibition of the Wnt signaling pathway ameliorated ISO-induced MH, DKK1, an inhibitor of the Wnt signaling pathway, was used. DKK1 is a member of the DKK family that is reported to antagonize the Wnt/β-catenin signaling pathway by decreasing β-catenin and increasing OCT4 expression (89). The results of the present study demonstrated that the DKK1 group exhibited trends in the expression of protein in the Wnt signaling pathway that were comparable to those follow PU pretreatment, in that PU exerted inhibitory effects on Wnt signaling comparable with those mediated by the known Wnt inhibitor. IM-12, an activator of the Wnt signaling pathway, has been reported to increase expression of β-catenin and downstream proteins while suppressing expression of GSK-3β (46). The present study assessed whether IM-12 reversed the inhibitory effects of PU on ISO-induced MH, thus confirming whether PU exerted its effects via inhibition of the Wnt signaling pathway. The present study demonstrated that PU inhibited expression of Wnt signaling pathway proteins induced by IM-12 and increased expression of GSK-3β. Furthermore, IM-12, the specific activator of the Wnt signaling pathway, was demonstrated to eliminate the protective effects of PU on MH. In conclusion, PU served a cardioprotective role partially via inhibition of the Wnt signaling pathway.

However, in the present study only core proteins in the Wnt signaling pathway were assessed; therefore the detailed underlying molecular mechanism in the upstream regulation of Wnt/β-catenin signaling during MH was not fully elucidated. Since Wnt signaling induces nuclear localization of β-catenin (90), it is important to determine the localization and expression of β-catenin. These limitations of the present study require addressing in future. In future studies, the inhibitor-like mechanism of PU and its effects on other disease caused by aberrant activation of the Wnt signaling pathway, including colon cancer, hepatocellular carcinoma and pancreatic, lung and ovarian cancer (91), should be explored.

In summary, the present study demonstrated that pretreatment of AC16 cells with PU attenuated ISO-induced MH. The underlying mechanism was associated with inhibition of protein expression in the Wnt/β-catenin signaling pathway and NF-κB activity. These results suggested that PU may be a potential agent for treating MH. Based on the importance of the Wnt signaling pathway for initiation, maintenance, progression and relapse of MH, PU may serve an inhibitor-like role for the treatment of cardiovascular and associated disease caused by hyperactivation of the Wnt signaling pathway. Furthermore, the present study may provide a novel direction for development and use of agents for MH treatment.

Acknowledgements

Not applicable.

Funding

The present study was supported by Shanghai Special Project of Biomedical Science and Technology (grant no. 21S11901700).

Availability of data and materials

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

Authors' contributions

XW performed experiments, analyzed data and wrote the manuscript. KH performed experiments and data analysis. LM designed the study, analyzed data and revised the manuscript. LW performed data analysis. YY participated in discussions and analyzed data. YL designed the study and obtained funding. XW, KH, LM and YL confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for participation

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, Barengo NC, Beaton AZ, Benjamin EJ, Benziger CP, et al: Global burden of cardiovascular diseases and risk factors, 1990–2019: Update from the GBD 2019 study. J Am Coll Cardiol. 76:2982–3021. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Cho YS, Moon SC, Ryu KS and Ryu KH: A study on clinical and healthcare recommending service based on cardiovascula disease pattern analysis. Int J Biosci Biotechnol. 8:287–294. 2016.

3 

Nalban N, Sangaraju R, Alavala S, Mir SM, Jerald MK and Sistla R: Arbutin attenuates isoproterenol-induced cardiac hypertrophy by inhibiting TLR-4/NF-κB pathway in mice. Cardiovasc Toxicol. 20:235–248. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Roth GA, Mensah GA and Fuster V: The global burden of cardiovascular diseases and risks: A compass for global action. J Am Coll Cardiol. 76:2980–2981. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Oh T, Kim D, Lee S, Won C, Kim S, Yang JS, Yu J, Kim B and Lee J: Machine learning-based diagnosis and risk factor analysis of cardiocerebrovascular disease based on KNHANES. Sci Rep. 12:22502022. View Article : Google Scholar : PubMed/NCBI

6 

Leong DP, Joseph PG, McKee M, Anand SS, Teo KK, Schwalm JD and Yusuf S: Reducing the global burden of cardiovascular disease, part 2: Prevention and treatment of cardiovascular disease. Circ Res. 121:695–710. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Van Camp G: Cardiovascular disease prevention. Acta Clin Belg. 69:407–411. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Zhao Y, Jia WW, Ren S, Xiao W, Li GW, Jin L and Lin Y: Difluoromethylornithine attenuates isoproterenol-induced cardiac hypertrophy by regulating apoptosis, autophagy and the mitochondria-associated membranes pathway. Exp Ther Med. 22:8702021. View Article : Google Scholar : PubMed/NCBI

9 

Gallo S, Vitacolonna A, Bonzano A, Comoglio P and Crepaldi T: ERK: A key player in the pathophysiology of cardiac hypertrophy. Int J Mol Sci. 20:21642019. View Article : Google Scholar : PubMed/NCBI

10 

Ellison GM, Waring CD, Vicinanza C and Torella D: Physiological cardiac remodelling in response to endurance exercise training: Cellular and molecular mechanisms. Heart. 98:5–10. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Selvetella G, Hirsch E, Notte A, Tarone G and Lembo G: Adaptive and maladaptive hypertrophic pathways: Points of convergence and divergence. Cardiovasc Res. 63:373–380. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Shimizu I and Minamino T: Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol. 97:245–262. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Kurosawa Y, Kojima K, Kato M, Ohashi R, Minami K and Narita H: Protective action of angiotensin converting enzyme inhibitors on cardiac hypertrophy in the aortic-banded rat. Jpn Heart J. 40:645–654. 1999. View Article : Google Scholar : PubMed/NCBI

14 

A Romero C, Mathew S, Wasinski B, Reed B, Brody A, Dawood R, Twiner MJ, McNaughton CD, Fridman R, Flack JM, et al: Angiotensin-converting enzyme inhibitors increase anti-fibrotic biomarkers in African Americans with left ventricular hypertrophy. J Clin Hypertens (Greenwich). 23:1008–1016. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Liu Y, Shen HJ, Wang XQ, Liu HQ, Zheng LY and Luo JD: EndophilinA2 protects against angiotensin II-induced cardiac hypertrophy by inhibiting angiotensin II type 1 receptor trafficking in neonatal rat cardiomyocytes. J Cell Biochem. 119:8290–8303. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Walsh-Wilkinson É, Drolet MC, Le Houillier C, Roy ÈM, Arsenault M and Couet J: Sex differences in the response to angiotensin II receptor blockade in a rat model of eccentric cardiac hypertrophy. PeerJ. 7:e74612019. View Article : Google Scholar : PubMed/NCBI

17 

Chang CS, Tsai PJ, Sung JM, Chen JY, Ho LC, Pandya K, Maeda N and Tsai YS: Diuretics prevent thiazolidinedione-induced cardiac hypertrophy without compromising insulin-sensitizing effects in mice. Am J Pathol. 184:442–453. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Okura T, Miyoshi K, Irita J, Enomoto D, Jotoku M, Nagao T, Watanabe K, Matsuokan H, Ashihara T, Higaki J, et al: Comparison of the effect of combination therapy with an angiotensin II receptor blocker and either a low-dose diuretic or calcium channel blocker on cardiac hypertrophy in patients with hypertension. Clin Exp Hypertens. 35:563–569. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Zhang X, Zhang MC and Wang CT: Loss of LRRC25 accelerates pathological cardiac hypertrophy through promoting fibrosis and inflammation regulated by TGF-β1. Biochem Biophys Res Commun. 506:137–144. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Zang Y, Wan J, Zhang Z, Huang S, Liu X and Zhang W: An updated role of astragaloside IV in heart failure. Biomed Pharmacother. 126:1100122020. View Article : Google Scholar : PubMed/NCBI

21 

Ma Y, Kang R and Liu X: Research progress in prevention and cure of fibrosis by traditional Chinese medicine. Mod Appl Sci. 2:127–132. 2008. View Article : Google Scholar

22 

Yang QY, Chen KJ, Lu S and Sun HR: Research progress on mechanism of action of Radix Astragalus in the treatment of heart failure. Chin J Integr Med. 18:235–240. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Karmazyn M and Gan XT: Treatment of the cardiac hypertrophic response and heart failure with ginseng, ginsenosides, and ginseng-related products. Can J Physiol Pharmacol. 95:1170–1176. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Yang J, Wang HX, Zhang YJ, Yang YH, Lu ML, Zhang J, Li ST, Zhang SP and Li G: Astragaloside IV attenuates inflammatory cytokines by inhibiting TLR4/NF-кB signaling pathway in isoproterenol-induced myocardial hypertrophy. J Ethnopharmacol. 150:1062–1070. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Liu ZH, Liu HB and Wang J: Astragaloside IV protects against the pathological cardiac hypertrophy in mice. Biomed Pharmacother. 97:1468–1478. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Guo J, Gan XT, Haist JV, Rajapurohitam V, Zeidan A, Faruq NS and Karmazyn M: Ginseng inhibits cardiomyocyte hypertrophy and heart failure via NHE-1 inhibition and attenuation of calcineurin activation. Circ Heart Fail. 4:79–88. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Qin N, Gong QH, Wei LW, Wu Q and Huang XN: Total ginsenosides inhibit the right ventricular hypertrophy induced by monocrotaline in rats. Biol Pharm Bull. 31:1530–1535. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Bu L, Dai O, Zhou F, Liu F, Chen JF, Peng C and Xiong L: Traditional Chinese medicine formulas, extracts, and compounds promote angiogenesis. Biomed Pharmacother. 132:1108552020. View Article : Google Scholar : PubMed/NCBI

29 

Luo J, Xu H and Chen K: Systematic review of compound danshen dropping pill: A chinese patent medicine for acute myocardial infarction. Evid Based Complement Alternat Med. 2013:8080762013. View Article : Google Scholar : PubMed/NCBI

30 

Tu Y: Artemisinin-A gift from traditional Chinese medicine to the world (nobel lecture). Angew Chem Int Ed Engl. 55:10210–10226. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Yang R, Yuan BC, Ma YS, Zhou S and Liu Y: The anti-inflammatory activity of licorice, a widely used Chinese herb. Pharm Biol. 55:5–18. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Mu F, Duan J, Bian H, Zhai X, Shang P, Lin R, Zhao M, Hu D, Yin Y, Wen A and Xi M: Metabonomic strategy for the evaluation of Chinese medicine Salvia miltiorrhiza and Dalbergia odorifera interfering with myocardial ischemia/reperfusion injury in rats. Rejuvenation Res. 20:263–277. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Wang S, Zhang S, Wang S, Gao P and Dai L: A comprehensive review on Pueraria: Insights on its chemistry and medicinal value. Biomed Pharmacother. 131:1107342020. View Article : Google Scholar : PubMed/NCBI

34 

Hou N, Huang Y, Cai SA, Yuan WC, Li LR, Liu XW, Zhao GJ, Qiu XX, Li AQ, Cheng CF, et al: Puerarin ameliorated pressure overload-induced cardiac hypertrophy in ovariectomized rats through activation of the PPARα/PGC-1 pathway. Acta Pharmacol Sin. 42:55–67. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Yuan G, Shi S, Jia Q, Shi J, Shi S, Zhang X, Shou X, Zhu X and Hu Y: Use of network pharmacology to explore the mechanism of Gegen (Puerariae lobatae Radix) in the treatment of type 2 diabetes mellitus associated with hyperlipidemia. Evid Based Complement Alternat Med. 2021:66334022021. View Article : Google Scholar : PubMed/NCBI

36 

Zhou YX, Zhang H and Peng C: Puerarin: A review of pharmacological effects. Phytother Res. 28:961–975. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Liu J, Zhang HJ, Ji BP, Cai SB, Wang RJ, Zhou F, Yang JS and Liu HJ: A diet formula of Puerariae radix, Lycium barbarum, Crataegus pinnatifida, and Polygonati rhizoma alleviates insulin resistance and hepatic steatosis in CD-1 mice and HepG2 cells. Food Funct. 5:1038–1049. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Liu B, Wu Z, Li Y, Ou C, Huang Z, Zhang J, Liu P, Luo C and Chen M: Puerarin prevents cardiac hypertrophy induced by pressure overload through activation of autophagy. Biochem Biophys Res Commun. 464:908–915. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Gao W, Guo N, Zhao S, Chen Z, Zhang W, Yan F, Liao H and Chi K: Carboxypeptidase A4 promotes cardiomyocyte hypertrophy through activating PI3K-AKT-mTOR signaling. Biosci Rep. 40:BSR202006692020. View Article : Google Scholar : PubMed/NCBI

40 

Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A and Blankesteijn WM: WNT signaling in cardiac and vascular disease. Pharmacol Rev. 70:68–141. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Weeks KL, Bernardo BC, Ooi JYY, Patterson NL and McMullen JR: The IGF1-PI3K-Akt signaling pathway in mediating exercise-induced cardiac hypertrophy and protection. Adv Exp Med Biol. 1000:187–210. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Fan J, Qiu L, Shu H, Ma B, Hagenmueller M, Riffel JH, Meryer S, Zhang M, Hardt SE, Wang L, et al: Recombinant frizzled1 protein attenuated cardiac hypertrophy after myocardial infarction via the canonical Wnt signaling pathway. Oncotarget. 9:3069–3080. 2018. View Article : Google Scholar : PubMed/NCBI

43 

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

44 

Lieven O, Knobloch J and Rüther U: The regulation of Dkk1 expression during embryonic development. Dev Biol. 340:256–268. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Li Y, Lu W, King TD, Liu CC, Bijur GN and Bu G: Dkk1 stabilizes Wnt co-receptor LRP6: Implication for Wnt ligand-induced LRP6 down-regulation. PLoS One. 5:e110142010. View Article : Google Scholar : PubMed/NCBI

46 

Wang T, Duan YM, Fu Q, Liu T, Yu JC, Sui ZY, Huang L and Wen GQ: IM-12 activates the Wnt-β-catenin signaling pathway and attenuates rtPA-induced hemorrhagic transformation in rats after acute ischemic stroke. Biochem Cell Biol. 97:702–708. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Cheng Y, Shen A, Wu X, Shen Z, Chen X, Li J, Liu L, Lin X, Wu M, Chen Y, et al: Qingda granule attenuates angiotensin II-induced cardiac hypertrophy and apoptosis and modulates the PI3K/AKT pathway. Biomed Pharmacother. 133:1110222021. View Article : Google Scholar : PubMed/NCBI

48 

Guo Y, Yu ZY, Wu J, Gong H, Kesteven S, Iismaa SE, Chan AY, Holman S, Pinto S, Pironet A, et al: The Ca2+-activated cation channel TRPM4 is a positive regulator of pressure overload-induced cardiac hypertrophy. Elife. 10:e665822021. View Article : Google Scholar : PubMed/NCBI

49 

Schnelle M, Chong M, Zoccarato A, Elkenani M, Sawyer GJ, Hasenfuss G, Ludwig C and Shah AM: In vivo [U-13C]glucose labeling to assess heart metabolism in murine models of pressure and volume overload. Am J Physiol Heart Circ Physiol. 319:H422–H431. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Ma X, Song Y, Chen C, Fu Y, Shen Q, Li Z and Zhang Y: Distinct actions of intermittent and sustained β-adrenoceptor stimulation on cardiac remodeling. Sci China Life Sci. 54:493–501. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Ribeiro DA, Buttros JB, Oshima C, Bergamaschi CT and Campos RR: Ascorbic acid prevents acute myocardial infarction induced by isoproterenol in rats: Role of inducible nitric oxide synthase production. J Mol Histol. 40:99–105. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Prabhu S, Narayan S and Devi CS: Mechanism of protective action of mangiferin on suppression of inflammatory response and lysosomal instability in rat model of myocardial infarction. Phytother Res. 23:756–760. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Xu H, Wang Z, Chen M, Zhao W, Tao T, Ma L, Ni Y and Li W: YTHDF2 alleviates cardiac hypertrophy via regulating Myh7 mRNA decoy. Cell Biosci. 11:1322021. View Article : Google Scholar : PubMed/NCBI

54 

Zhang GX, Kimura S, Murao K, Yu X, Obata K, Matsuyoshi H and Takaki M: Effects of angiotensin type I receptor blockade on the cardiac Raf/MEK/ERK cascade activated via adrenergic receptors. J Pharmacol Sci. 113:224–233. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Li L, Cai H, Liu H and Guo T: β-Adrenergic stimulation activates protein kinase Cε and induces extracellular signal-regulated kinase phosphorylation and cardiomyocyte hypertrophy. Mol Med Rep. 11:4373–4380. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Werhahn SM, Kreusser JS, Hagenmüller M, Beckendorf J, Diemert N, Hoffmann S, Schultz JH, Backs J and Dewenter M: Adaptive versus maladaptive cardiac remodelling in response to sustained β-adrenergic stimulation in a new ‘ISO on/off model’. PLoS One. 16:e02489332021. View Article : Google Scholar : PubMed/NCBI

57 

Garg M and Khanna D: Exploration of pharmacological interventions to prevent isoproterenol-induced myocardial infarction in experimental models. Ther Adv Cardiovasc Dis. 8:155–169. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Liu BY, Li L, Liu GL, Ding W, Chang WG, Xu T, Ji XY, Zheng XX, Zhang J and Wang JX: Baicalein attenuates cardiac hypertrophy in mice via suppressing oxidative stress and activating autophagy in cardiomyocytes. Acta Pharmacol Sin. 42:701–714. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Wen J, Shen J, Zhou Y, Zhao X, Dai Z and Jin Y: Pyrroloquinoline quinone attenuates isoproterenol hydrochloride-induced cardiac hypertrophy in AC16 cells by inhibiting the NF-κB signaling pathway. Int J Mol Med. 45:873–885. 2020.PubMed/NCBI

60 

Zhao Y, Jiang Y, Chen Y, Zhang F, Zhang X, Zhu L and Yao X: Dissection of mechanisms of Chinese medicinal formula Si-Miao-Yong-an decoction protects against cardiac hypertrophy and fibrosis in isoprenaline-induced heart failure. J Ethnopharmacol. 248:1120502020. View Article : Google Scholar : PubMed/NCBI

61 

Zhang C, Wang Y, Ge Z, Lin J, Liu J, Yuan X and Lin Z: GDF11 attenuated ANG II-induced hypertrophic cardiomyopathy and expression of ANP, BNP and beta-MHC through down-regulating CCL11 in mice. Curr Mol Med. 18:661–671. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Cameron VA, Rademaker MT, Ellmers LJ, Espiner EA, Nicholls MG and Richards AM: Atrial (ANP) and brain natriuretic peptide (BNP) expression after myocardial infarction in sheep: ANP is synthesized by fibroblasts infiltrating the infarct. Endocrinology. 141:4690–4697. 2000. View Article : Google Scholar : PubMed/NCBI

63 

Edwards JG: Cardiac MHC gene expression: More complexity and a step forward. Am J Physiol Heart Circ Physiol. 294:H14–H15. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Yuan Y, Zong J, Zhou H, Bian ZY, Deng W, Dai J, Gan HW, Yang Z, Li H and Tang QZ: Puerarin attenuates pressure overload-induced cardiac hypertrophy. J Cardiol. 63:73–81. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Yeh YL, Tsai HI, Cheng SM, Pai P, Ho TJ, Chen RJ, Lai CH, Huang PJ, Padma VV and Huang CY: Mechanism of Taiwan Mingjian Oolong tea to inhibit isoproterenol-induced hypertrophy and apoptosis in cardiomyoblasts. Am J Chin Med. 44:77–86. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Guan XH, Hong X, Zhao N, Liu XH, Xiao YF, Chen TT, Deng LB, Wang XL, Wang JB, Ji GJ, et al: CD38 promotes angiotensin II-induced cardiac hypertrophy. J Cell Mol Med. 21:1492–1502. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Hu H, Jiang M, Cao Y, Zhang Z, Jiang B, Tian F, Feng J, Dou Y, Gorospe M, Zheng M, et al: HuR regulates phospholamban expression in isoproterenol-induced cardiac remodelling. Cardiovasc Res. 116:944–955. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Huo S, Shi W, Ma H, Yan D, Luo P, Guo J, Li C, Lin J, Zhang C, Li S, et al: Alleviation of inflammation and oxidative stress in pressure overload-induced cardiac remodeling and heart failure via IL-6/STAT3 inhibition by raloxifene. Oxid Med Cell Longev. 2021:66990542021. View Article : Google Scholar : PubMed/NCBI

69 

Bi X, Zhang Y, Yu Y, Yuan J, Xu S, Liu F, Ye J and Liu P: MiRNA-339-5p promotes isoproterenol-induced cardiomyocyte hypertrophy by targeting VCP to activate the mTOR signaling. Cell Biol Int. 46:288–299. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Han B, Xu J, Shi X, Zheng Z, Shi F, Jiang F and Han J: DL-3-n-butylphthalide attenuates myocardial hypertrophy by targeting gasdermin D and inhibiting gasdermin D mediated inflammation. Front Pharmacol. 12:6881402021. View Article : Google Scholar : PubMed/NCBI

71 

Shah AK, Bhullar SK, Elimban V and Dhalla NS: Oxidative stress as a mechanism for functional alterations in cardiac hypertrophy and heart failure. Antioxidants (Basel). 10:9312021. View Article : Google Scholar : PubMed/NCBI

72 

Gai Z, Wang Y, Tian L, Gong G and Zhao J: Whole genome level analysis of the Wnt and DIX gene families in mice and their coordination relationship in regulating cardiac hypertrophy. Front Genet. 12:6089362021. View Article : Google Scholar : PubMed/NCBI

73 

Qin H, Zhang Y, Wang R, Du X, Li L and Du H: Puerarin suppresses Na+-K+-ATPase-mediated systemic inflammation and CD36 expression, and alleviates cardiac lipotoxicity in vitro and in vivo. J Cardiovasc Pharmacol. 68:465–472. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Moon RT, Kohn AD, De Ferrari GV and Kaykas A: WNT and beta-catenin signalling: Diseases and therapies. Nat Rev Genet. 5:691–701. 2004. View Article : Google Scholar : PubMed/NCBI

75 

Agostino M and Pohl SÖ: The structural biology of canonical Wnt signalling. Biochem Soc Trans. 48:1765–1780. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Hua Y, Yang Y, Li Q, He X, Zhu W, Wang J and Gan X: Oligomerization of Frizzled and LRP5/6 protein initiates intracellular signaling for the canonical WNT/β-catenin pathway. J Biol Chem. 293:19710–19724. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Gao C and Chen YG: Dishevelled: The hub of Wnt signaling. Cell Signal. 22:717–727. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Zeng KW, Wang JK, Wang LC, Guo Q, Liu TT, Wang FJ, Feng N, Zhang XW, Liao LX, Zhao MM, et al: Small molecule induces mitochondrial fusion for neuroprotection via targeting CK2 without affecting its conventional kinase activity. Signal Transduct Target Ther. 6:712021. View Article : Google Scholar : PubMed/NCBI

79 

Ríos JA, Godoy JA and Inestrosa NC: Wnt3a ligand facilitates autophagy in hippocampal neurons by modulating a novel GSK-3β-AMPK axis. Cell Commun Signal. 16:152018. View Article : Google Scholar : PubMed/NCBI

80 

Barker N, Morin PJ and Clevers H: The Yin-Yang of TCF/beta-catenin signaling. Adv Cancer Res. 77:1–24. 2000. View Article : Google Scholar : PubMed/NCBI

81 

Piazza F, Manni S, Tubi LQ, Montini B, Pavan L, Colpo A, Gnoato M, Cabrelle A, Adami F, Zambello R, et al: Glycogen synthase kinase-3 regulates multiple myeloma cell growth and bortezomib-induced cell death. BMC Cancer. 10:5262010. View Article : Google Scholar : PubMed/NCBI

82 

Guo Y, Gupte M, Umbarkar P, Singh AP, Sui JY, Force T and Lal H: Entanglement of GSK-3β, β-catenin and TGF-β1 signaling network to regulate myocardial fibrosis. J Mol Cell Cardiol. 110:109–120. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Guan X, He Y, Wei Z, Shi C, Li Y, Zhao R, Pan L, Han Y, Hou T and Yang J: Crosstalk between Wnt/β-catenin signaling and NF-κB signaling contributes to apical periodontitis. Int Immunopharmacol. 98:1078432021. View Article : Google Scholar : PubMed/NCBI

84 

Jia D, Yang W, Li L, Liu H, Tan Y, Ooi S, Chi L, Filion LG, Figeys D and Wang L: β-Catenin and NF-κB co-activation triggered by TLR3 stimulation facilitates stem cell-like phenotypes in breast cancer. Cell Death Differ. 22:298–310. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Shang S, Hua F and Hu ZW: The regulation of β-catenin activity and function in cancer: Therapeutic opportunities. Oncotarget. 8:33972–33989. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Gitau SC, Li X, Zhao D, Guo Z, Liang H, Qian M, Lv L, Li T, Xu B, Wang Z, et al: Acetyl salicylic acid attenuates cardiac hypertrophy through Wnt signaling. Front Med. 9:444–456. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Olsen NT, Dimaano VL, Fritz-Hansen T, Sogaard P, Chakir K, Eskesen K, Steenbergen C, Kass DA and Abraham TP: Hypertrophy signaling pathways in experimental chronic aortic regurgitation. J Cardiovasc Transl Res. 6:852–860. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Liu JJ, Shentu LM, Ma N, Wang LY, Zhang GM, Sun Y, Wang Y, Li J and Mu YL: Inhibition of NF-κB and Wnt/β-catenin/GSK3β signaling pathways ameliorates cardiomyocyte hypertrophy and fibrosis in streptozotocin (STZ)-induced type 1 diabetic rats. Curr Med Sci. 40:35–47. 2020. View Article : Google Scholar : PubMed/NCBI

89 

Ou L, Fang L, Tang H, Qiao H, Zhang X and Wang Z: Dickkopf Wnt signaling pathway inhibitor 1 regulates the differentiation of mouse embryonic stem cells in vitro and in vivo. Mol Med Rep. 13:720–730. 2016. View Article : Google Scholar : PubMed/NCBI

90 

Kim S, Song G, Lee T, Kim M, Kim J, Kwon H, Kim J, Jeong W, Lee U, Na C, et al: PARsylated transcription factor EB (TFEB) regulates the expression of a subset of Wnt target genes by forming a complex with β-catenin-TCF/LEF1. Cell Death Differ. 28:2555–2570. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Zhang L, Guo Z, Wang Y, Geng J and Han S: The protective effect of kaempferol on heart via the regulation of Nrf2, NF-κβ, and PI3K/Akt/GSK-3β signaling pathways in isoproterenol-induced heart failure in diabetic rats. Drug Dev Res. 80:294–309. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2022
Volume 26 Issue 4

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
Wang X, He K, Ma L, Wu L, Yang Y and Li Y: Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway. Mol Med Rep 26: 306, 2022
APA
Wang, X., He, K., Ma, L., Wu, L., Yang, Y., & Li, Y. (2022). Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway. Molecular Medicine Reports, 26, 306. https://doi.org/10.3892/mmr.2022.12822
MLA
Wang, X., He, K., Ma, L., Wu, L., Yang, Y., Li, Y."Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway". Molecular Medicine Reports 26.4 (2022): 306.
Chicago
Wang, X., He, K., Ma, L., Wu, L., Yang, Y., Li, Y."Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway". Molecular Medicine Reports 26, no. 4 (2022): 306. https://doi.org/10.3892/mmr.2022.12822