Open Access

Mechanistic analysis of resveratrol in cardiac hypertrophy by network pharmacology and animal experiments

  • Authors:
    • Shan Ren
    • Lei Shen
    • Song Lin
    • Dan Xiao
    • Wei Xiao
    • Pei-Mei Yan
    • Yan-Yan Zhang
    • Wei-Wei Jia
    • Yan Lin
  • View Affiliations

  • Published online on: September 1, 2022     https://doi.org/10.3892/mmr.2022.12840
  • Article Number: 324
  • Copyright: © Ren 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

Resveratrol (Res) serves a protective role in hepatic, cardiovascular and autoimmune hypertrophic disease. However, the mechanisms by which Res ameliorates cardiac hypertrophy have not yet been fully elucidated. In the present study, network pharmacology was used to construct a network and perform enrichment analysis to evaluate the effect of Res on cardiac hypertrophy. Experimental validation was performed using 40 Sprague‑Dawley rats administered intragastric 80 mg/kg/day Res and 20 mg/kg/day 3‑methyladenine (3‑MA) for 4 weeks. A total of 444 targets associated with cardiac hypertrophy and 229 potential disease‑associated targets of Res were identified, from which 8 overlapping genes were demonstrated. Gene Ontology function and ‘Kyoto Encyclopedia of Genes and Genomes’ pathway enrichment analysis demonstrated that Res affected STAT3 and was associated with autophagy signaling pathways, including ‘negative regulation of autophagy for hypertrophic cardiomyopathy’. Furthermore, Res ameliorated isoprenaline‑induced cardiac hypertrophy, significantly improving cardiac dysfunction in vivo experiment (echocardiography, the degree of ventricular hypertrophy, etc.); this effect may be associated with regulation of autophagy and apoptosis. The autophagy inhibitor 3‑MA markedly reversed the anti‑cardiac hypertrophy effects of Res. In conclusion, Res inhibited cardiac hypertrophy via downregulation of the apoptosis signaling pathway and upregulating the autophagy pathway.

Introduction

Myocardial hypertrophy is an independent risk factor for cardiovascular disease and a key cause of heart failure (1). The condition may be caused by valvular disease, hypertension, myocardial infarction and cardiomyopathy amongst other pathological mechanisms (1). Cardiomyocyte hypertrophy is an indicator of myocardial remodeling and induces decreased myocardial contractility, which progresses to heart failure (2). Therefore, it is necessary to determine factors offering a protective effect against cardiac hypertrophy to decrease the incidence of heart failure and subsequent mortality. Resveratrol (Res) is a natural non-flavonoid polyphenol with anti-inflammatory, cardioprotective, neuroprotective, antioxidant and anti-aging properties that is abundant in food such as grapes, mulberries and blueberries (3,4). Preclinical studies have reported the potential of Res as a treatment for severe cardiomyopathy (5,6). However, mechanisms underlying the cardioprotective action of Res remain unknown.

Network pharmacology is a powerful multi-component, multi-target tool which has been used to study therapeutic mechanisms of herbal medicines (7,8). The approach has been employed in mechanistic studies associated with numerous types of chronic disease, including obesity, diabetes, chronic obstructive pulmonary and cardio-cerebral vascular disease (9,10). Therefore, evaluation of the mechanistic actions of multi-target drugs in the treatment of complex diseases draws on abundant datasets to inform preliminary investigation.

The present study evaluated the pharmacological action of Res in cardiac hypertrophy using network pharmacology and animal experiments. Potential Res targets were predicted and cross-referenced with therapeutic targets associated with cardiomyopathy. Res targets associated with cardiac hypertrophy were integrated to establish a multi-target network. Cytoscape and Metascape were used to analyze significant genes in the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Significant targets and pathways obtained from network pharmacology were verified using western blotting. The process of network pharmacology and animal experiments is presented in Fig. 1.

Materials and methods

Construction of Res target library

The chemical structure of Res was downloaded from Pubchem (pubchem.ncbi.nlm.nih.gov) and Res targets were predicted and downloaded from the Traditional Chinese Medicine Systems (TCMSP; http://tcmsp-e.com/) and Swiss Target Prediction (swisstargetprediction.ch) databases (the default parameters of the websites were utilized), using the inclusion criteria of P≤0.5 (11). Genes corresponding to targets were identified from UniProt (12).

Acquisition of cardiac hypertrophy gene dataset

Target genes associated with cardiac hypertrophy were downloaded from the Gene Expression Omnibus database (GEO; ncbi.nlm.nih.gov/geo). The gene expression profiles of myocardial tissue removed surgically from 54 male and 52 female patients with hypertrophic cardiomyopathy were downloaded from the GSE36961 dataset (P<0.05; log2 fold change >1).

Molecular docking and enrichment analysis

The structure of the signal transducer and activator of transcription (STAT3) protein was downloaded from the Protein Data Bank (rcsb.org/) and molecular docking performed using AutoDock Vina (version, 1.2.0) (13). The online Database for Annotation, Visualization and Integrated Discovery (david.ncifcrf.gov/home.jsp) enrichment analysis service was used to analyze enriched genes using GO terms and KEGG pathways. Venn diagrams were generated using Venny 2.1.0 (bioinfogp.cnb.csic.es/tools/venny/) to demonstrate potential associations between datasets. Functional GO and KEGG analysis of potential targets of Res for cardiac hypertrophy treatment was performed using Metascape (version, 3.5) (metascape.org) and pathways with P≤0.05 were considered to be statistically significant for cardiac hypertrophy treatment. A diagram of Res-associated signaling pathways based on GO and KEGG analyses was generated using the ‘Pathview’ package in the Toolkit for Biologists software (version, 1.098667) (14).

Protein-protein interaction (PPI) network

Interactions between potential Res targets associated with hypertrophic cardiomyopathy in both males and females were evaluated using a PPI network generated using the BisoGenet 1.0 plugin (apps.cytoscape.org/apps/bisogenet) for Cytoscape 3.9 (15). The Molecular Complex Detection (MCODE) plugin (version 2.0.2, apps.cytoscape.org/apps/mcode) for Cytoscape 3.9 was used to visualize protein-protein interactions with topology parameters evaluated using the Cytoscape network analyzer tool. Compound-target associations were presented in a pharmacological network map constructed using Cytoscape 3.9 (the default parameters were utilized). Core gene-encoding proteins were screened for using MCODE score >5.

Animal experiments

A total of 20 healthy adult female and 20 male Sprague Dawley rats (eight weeks of age, 200–250 g) were supplied by the Animal Center of Qiqihar Medical University. Rats were housed with temperature and humidity at 22±2°C and 45±15%, respectively, under a 12 h light-dark cycle, with free access to food and water. Rats were acclimated for 7 days prior to the experiment. Rats were randomly divided into four groups (each group contained 5 male and 5 female rats) as follows: i) Control; ii) isoprenaline (ISO); iii) Res and iv) Res + 3-Methyladenine (3-MA). Rats in the ISO group were administered 0.9% saline by gavage (in equal volume to Res group, about 2 ml) for 4 weeks and subcutaneous ISO injection (5 mg/kg/day) on days 22–28. The Res group were administered Res (80 mg/kg/day) by gavage for 4 weeks (16) and subcutaneous ISO injections (5 mg/kg/day) from day 22–28. The Res + 3-MA group was administered Res (80 mg/kg/day) by gavage, 3-MA (20 mg/kg/day) by intraperitoneal injection for 4 weeks and subcutaneous ISO injection (5 mg/kg/day) on days 22–28. Rats in the control group were administered 0.9% saline (in equal volume to Res group, about 2 ml) by gavage for 4 weeks and subcutaneous 0.9% saline (in equal volume to ISO, about 0.5 ml) injections on days 22–28. Rats were euthanized using intraperitoneal injection of 3% sodium pentobarbitone (240 mg/kg) following echocardiography and the ventricular myocardia was excised. The present study was approved by the Committee on the Ethics of Animal Experiments of Qiqihar Medical University (Qiqihar, heilingjiang, approval no. QMU-AECC-2019-53).

Echocardiography

Echocardiography was performed 24 h after the final treatment. Briefly, rats were anaesthetized with intraperitoneal injection of 3% sodium pentobarbitone (80 mg/kg) and the thoracic area was shaved. M-mode recording of the short-axis view was performed on the shaved chest wall. Left ventricular end-systolic dimension (LVESd), LV end-diastolic dimension (LVEDd), LV fractional shortening (FS) and ejection fraction (EF) were assessed by echocardiography using a V6 imaging system [VINNO Technology (Suzhou) Co., Inc.] with high-frequency ultrasound transducer.

Tissue collection and analysis

The heart and left ventricle of euthanized rats were isolated. The ratio of left ventricle weight: body weight (LVW/BW) and heart weight: BW (HW/BW) were calculated to evaluate the degree of ventricular hypertrophy.

Measurement of malondialdehyde (MDA) and lactate dehydrogenase (LDH)

MDA and LDH levels in heart homogenate was assessed using MDA assay kit and LDH assay kit (Nanjing Jiancheng Bioengineering Institute) according to the manufacturer's protocol and optical density at 530 and 450 nm was quantified using a microplate reader (Thermo Fisher Scientific, Inc.).

Measurement of atrial natriuretic peptide (ANP) mRNA expression levels

Total RNA was isolated from left ventricle tissue with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). According to the manufacturer's protocols, total RNA was reverse transcribed with mRNA reverse transcription kit (cat. no. RR037A, Takara Bio, Inc.). The resulting cDNA was used as template for RT-qPCR using SYBR-Green Master Mix (cat. no. A25742, Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions, the PCR program was as follows: 95°C for 30 sec, followed by 40 cycles at 95°C for 5 sec and 60°C for 30 sec. Melting curves were used to ensure that only the correct product was amplified. Primer sequences were as follows: ANP forward (F), 5′-GGGAAGTCAACCCGTCTCA-3′ and reverse (R), 5′-GGGCTCCAATCCTGTCAAT-3′ and GAPDH F, 5′-GAGACAGCCGCATCTTCTTG-3′ and R, 5′-ATACGGCCAAATCCGTTCAC-3′. Data were normalized to expression of GAPDH mRNA as endogenous control and quantified using the 2−ΔΔCq method (17).

Western blotting

Myocardial tissue was homogenized with cold RIPA buffer with 1% phenylmethylsulfonyl fluoride (P0013C, Beyotime Institute of Biotechnology), Total protein concentrations were measured by a BCA kit (Beyotime Institute of Biotechnology). Equal amounts of protein (30 µg) was separated using 8 or 10% SDS-PAGE and electro-transferred to PVDF membranes (MilliporeSigma). Membranes were blocked with 5% blocking buffer for 1 h at room temperature and incubated with primary antibodies as follows: Anti-ANP (1:1,000; cat. no. sc-515701, Santa Cruz Biotechnology, Inc.), anti-Bcl-2 (1:1,000; cat. no. sc-7382, Santa Cruz Biotechnology, Inc.), anti-Bax (1:1,000; cat. no. sc-20067, Santa Cruz Biotechnology, Inc.), anti-cytochrome C (CytC; 1:1,000; cat. no. sc-13156, Santa Cruz Biotechnology, Inc.) and anti-autophagy-related gene 5 (Atg5; 1:1,000; cat. no.sc-133158, Santa Cruz Biotechnology, Inc.) at 4°C overnight. GAPDH (1:1,000; sc-166545, Santa Cruz Biotechnology, Inc) was used as the internal control. Membranes were incubated with Secondary goat antimouse antibody were used (diluted 1:5,000, cat. no. BA1050; Boster Biological Technology, Ltd.) for 1.5 h at room temperature. The membranes were washed three times in Wash Buffer and proteins were detected using ECL reagent (Beyotime Institute of Biotechnology). Relative protein expression levels were semi-quantified using ImageJ software (National Institutes of Health; version 1.48) and presented as fold-change compared with control.

Statistical analysis

All data are presented as mean ± SEM and represent at least three independent experiments. Statistical comparisons were made one-way ANOVA followed by a post hoc analysis (Tukey test) and GraphPad Prism software (GraphPad Software, Inc.,version 7.0). P<0.05 was considered to indicate a statistically significant difference.

Results

Res target characteristics for hypertrophic cardiomyopathy

The molecular structure of Res (C14H12O3) is presented in Fig. 2A. A total of 230 potential Res targets from the TCMSP and Swiss Target Prediction databases and 444 cardiac hypertrophy-associated targets from the GSE36961 dataset, in the form of differential gene expression profiles for male and female patients with hypertrophic cardiomyopathy, were screened to characterizing the potential association between Res and cardiac hypertrophy (Fig. 2B and C). A total of 8 proteins overlapped in the intersection of Res and cardiac hypertrophy-associated targets, of which 3 were identified in females only (Fig. 2D).

Bioinformatics analysis of target genes

Molecular docking analysis demonstrated the binding of Res to STAT3 (Fig. 3). Enrichment analysis demonstrated a further 10 pathways, including ‘IL-4 and IL-13 signaling’ and ‘TGF-β signaling pathway’, that were associated with Res treatment of cardiac hypertrophy. Further analysis demonstrated that Res intervening pathways, including ‘IL-5 signaling pathway’ and ‘lipid metabolism in senescent cells’ were associated with ‘apoptosis’, whereas ‘TGF-β signaling pathway’ and ‘positive regulation of glycolytic process’ were associated with ‘autophagy’. Therefore, apoptosis and autophagy may be potential therapeutic mechanistic targets of Res in hypertrophic cardiomyopathy.

PPI network of target genes

The BisoGenet 1.0 plugin for Cytoscape 3.9 was used to analyze the 129 Res targets associated with male patients with hypertrophic cardiomyopathy and a network of PPI associations with 1,206 nodes and 21,105 connections was constructed (Fig. 4A). A total of four modules of the core gene-encoded proteins had an MCODE score >5 as predicted using the MCODE plugin (version 2.0.2) in Cytoscape 3.9 (Fig. 4B). Analysis of the 215 Res targets associated with female patients with hypertrophic cardiomyopathy generated a PPI network with 3,350 nodes and 63,783 connections (Fig. 4C). A total of eight modules of the core gene-encoded proteins had an MCODE score >5, predicted using the MCODE plugin (version 2.0.2) in Cytoscape 3.9 (Fig. 4D). The female PPI network was more complex than the male network and featured more core proteins.

Res reverses ISO-induced myocardial hypertrophy

The cardiac index and ANP mRNA and protein expression levels were assessed as cardiac hypertrophy indicators. HW/BW, LVW/BW, the levels of ANP mRNA and protein expression were increased in ISO group. Treatment with Res significantly decreased HW/BW and LVW/BW values and decreased the levels of ANP mRNA and protein expression compared with the ISO group. Moreover, Res + 3-MA significantly increased the levels of ANP mRNA and protein expression compared with Res-alone (Fig. 5A-D).

Res reverses ISO-induced cardiac dysfunction

Echocardiographic parameters were assessed in M-mode (Fig. 6A); treatment with Res produced significant increases in EF% and FS% compared with the ISO group (Fig. 6B and C). These results demonstrated the importance of Res in cardiac hypertrophy. Furthermore, both LVESd and LVEDd were significantly decreased in the Res group compared with the ISO group. No significant differences were demonstrated for EF%, FS% and LVEDd between the Res and Res + 3-MA groups; however, LVESd exhibited a significant increase in the Res + 3-MA group compared with the Res group (Fig. 6D and E). In summary, ISO and 3-MA treatment both led to progressive cardiac enlargement, however, Res improved cardiac function.

Res decreases ISO-induced oxidative stress in the heart

The effect of Res on oxidative stress was investigated. Treatment with ISO significantly increased cardiac MDA and LDH levels compared with the control and these increases were significantly reversed by treatment with Res (Fig. 7). However, levels of MDA (Fig. 7A) were markedly higher and levels of LDH (Fig. 7B) significantly higher in the Res + 3-MA compared with the Res group.

Res downregulates apoptosis and upregulates autophagy

The protein expression levels of Bcl-2 and Atg5 were significantly decreased by ISO treatment compared with the control. Res caused significant upregulation of Bcl-2 and Atg5 expression compared with the ISO group. Expression levels of pro-apoptosis proteins Bax and CytC significantly increased in the ISO group compared with control. However, this effect was significantly decreased by Res treatment (Fig. 8A-E). However, Res + 3-MA treatment significantly reversed changes in protein expression levels of CytC, Bcl-2 and Atg5 compared with the Res group (Fig. 8A-E).

Discussion

Cardiac hypertrophy is an adaptive response to pathological stimuli but prolonged hypertrophy results in cardiac dysfunction and heart failure. Volume overload or elevated pressure is a key cause and pathological remodeling is characterized by hypertrophied cardiomyocytes, interstitial fibrosis, perivascular fibrosis and decreased cardiac compliance leading to malignant arrhythmia, heart failure and potential sudden cardiac death (1820). A previous study demonstrated associations between cardiac hypertrophy and apoptosis, oxidative stress, autophagy and aging (21). These connections highlight the importance of elucidating the precise mechanism of cardiac hypertrophy.

Res is a plant-derived polyphenol present in grape skin, peanut, cranberries and veratrum (22). Numerous biological activities have been ascribed to Res, including potent anti-inflammatory, antioxidant, anti-aging, insulin sensitization and cardioprotective activities, which are reported to protect of nerve and heart tissue (16). Furthermore, anti-aging activity has been suggested due to upregulation of hydrogen peroxide-dependent autophagy (23). Res has also been reported to inhibit vascular smooth muscle remodeling and growth and proliferation of cardiac fibroblasts (24). Res (80 mg/kg) via intragastric administration has been reported to protect against L-arginine-induced ANP, which may be associated with enhancement of sirtuin 1-mediated deacetylation of p53 and heat shock transcription factor 1 (16). To the best of our knowledge, however, little detailed information regarding Res protective mechanisms has been reported.

Network pharmacology has allowed previous studies of the anti-cardiac hypertrophy mechanism of Res to be performed (2527) and may facilitate identification of potential drug treatments for complex disease (28,29). The present study demonstrated the presence of 230 Res targets in the Swiss Target Prediction and TCMSP databases and 444 sex-specific targets associated with cardiac hypertrophy in the GSE36961 dataset. A total of 8 Res targets were also associated with cardiac hypertrophy, 5 in males and 8 in females. The intersection of Res and cardiac hypertrophy targets was visualized using the ‘Res-target-disease’ network and GO functional and KEGG pathway enrichment analyses were performed. STAT3, Myc and apoptotic processes were keytargets of Res in hypertrophic cardiomyopathy. Molecular docking analysis demonstrated the potential regulatory effect of Res on STAT3, a protein which is known to inhibit apoptosis and numerous autophagy-associated proteins (30). STAT3 is reported to be a suppressor of autophagy, which suggested that the Res therapeutic mechanism may involve autophagy in both male and female patients. Moreover, recent evidence suggests that STAT3 may promote cardiomyocyte metabolism and function by an effect on endothelial autophagy (31). The effect of STAT3 on autophagy has also been reported in cardiac fibrosis (3234). In summary, effects of Res on apoptosis and autophagy may underlie the potential mechanisms of Res in treating cardiac hypertrophy.

PPI networks demonstrated that Res action in female patients with hypertrophic cardiomyopathy was more complex and contained more core proteins than the equivalent action in male patients with hypertrophic cardiomyopathy. In both physiological and pathological scenarios and pre-menopausal, females exhibit enhanced protection against cardiac hypertrophy compared with males. Such protection disappears following the menopause; however it can be partially restored by estrogen replacement therapy. Estrogen antagonizes the pro-hypertrophy signaling pathway, whereas androgens have the opposing effect (35). The network pharmacology approach, encompassing various pharmacological effects of traditional drugs and the interaction between chemical molecules and target proteins at the molecular level, demonstrates interactions which support the suggestion that Res achieves its cardioprotective role in inhibiting cardiomyocyte apoptosis by increasing autophagy (36).

Pathological indicators, such as ANP, MDA and LDH, cardiac morphological indicators, such as HW/BW and LVW/BW, and cardiac structural indicators, such as EF%, FS%, LVESd and LVEDd, were analyzed in vivo to demonstrate the effects of Res. Res is demonstrated to reverse cardiac hypertrophy via complex mechanisms which remain to be clarified; however autophagy appears to be a key factor (37). However, the role of autophagy in myocardial hypertrophy is controversial. Previous studies have reported activation of general autophagy as having both protective and detrimental roles during cardiac hypertrophy and heart failure (38,39). Both Li et al (40) and Wang et al (41) reported that ISO-induced cardiac hypertrophy is accompanied by a significant decrease in autophagy and that enhanced autophagy may attenuate cardiac hypertrophy. The network pharmacology in the present study identified autophagy as a potential therapeutic Res target in cardiac hypertrophy. Furthermore, hypertrophic responses induced by ISO were alleviated by Res treatment via regulation of autophagy in the present study. Moreover, the autophagy inhibitor 3-MA stimulated hypertrophic responses in rats. However, HW/BW and LVW/BW, which demonstrated the therapeutic effect of Res on cardiac function, were not affected by 3-MA. However, 3-MA markedly decreased the reversal of Res-induced cardiac dysfunction, which suggested that the regulation of autophagy was involved. Apoptosis was also associated with the therapeutic effect of Res. Autophagy is reported to be associated with apoptosis, which affects homeostasis of the intracellular environment, initiated by both exogenous and endogenous pathways (42,43). The results of the present study demonstrated that apoptosis significantly increased following ISO treatment; this effect was significantly reversed by Res treatment. It could be hypothesized that 3-MA decreased the therapeutic effect of Res by suppressing autophagy and promoting apoptosis, two forms of programmed cell death. Autophagy may be regarded as a double-edged sword (44). Interactions between apoptosis and autophagy are highly complex and environmentally dependent and it can be concluded that both are mechanisms involved in the treatment of cardiac hypertrophy by Ras (45). Mitochondrial cytochrome C release and apoptosis are both inhibited by Bcl-2 binding to Bax (46). Bax is a proapoptotic member of the Bcl-2 family activated by apoptotic signaling, leading to cytochrome C release (47). This is stimulated by proapoptotic Bax, Bad, BH3-interacting domain death agonist and Bcl-2 antagonist/killer and inhibited by anti-apoptotic Bcl-xl and/or Bcl-2 (48). Res significantly reversed ISO-induced changes to Bcl-2, Bax and cytochrome C expression in the present study. 3-MA markedly reversed the regulation of apoptosis-associated proteins by Res, indicating that protection against myocardial hypertrophy involves autophagy and apoptosis. One potential limitation of this study would be that the mechanism of Res promoting the Atg5 require further study. Further investigation is therefore needed to understand the association between autophagy and the cardiac function following treatment with Res.

In conclusion, the results of the integrated network pharmacological and animal experiments in the present study demonstrated that Res ameliorated myocardial hypertrophy via regulation of autophagy and apoptosis-associated signaling pathways. These results may inform further studies on the use of Res in cardiac disease. Res may be an effective multi-target anti-myocardial hypertrophy drug.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. 82074148 and 31751004) and the Postdoctoral Scientific Research Foundation of Heilongjiang Province (grant no. LBH-Q18128).

Availability of data and materials

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

Authors' contributions

SR, LS and YL designed the experiments and wrote and revised the manuscript. WWJ, SL and WX performed the experiments and analyzed the data. PMY, YYZ and DX performed the experiments. SR and LS confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The experimental protocols were approved by the Committee on the Ethics of Animal Experiments of Qiqihar Medical University (Qiqihar, heilingjiang, approval no. QMU-AECC-2019-53).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Nakamura M and Sadoshima J: Mechanisms of physiological and pathological cardiac hypertrophy. Nat Rev Cardiol. 15:387–407. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Ott C, Jung T, Brix S, John C, Betz IR, Foryst-Ludwig A, Deubel S, Kuebler WM, Grune T, Kintscher U and Grune J: Hypertrophy-reduced autophagy causes cardiac dysfunction by directly impacting cardiomyocyte contractility. Cells. 10:8052021. View Article : Google Scholar : PubMed/NCBI

3 

Shaito A, Posadino AM, Younes N, Hasan H, Halabi S, Alhababi D, Al-Mohannadi A, Abdel-Rahman WM, Eid AH, Nasrallah GK and Pintus G: Potential adverse effects of resveratrol: A literature review. Int J Mol Sci. 21:20842020. View Article : Google Scholar : PubMed/NCBI

4 

Tian B and Liu J: Resveratrol: A review of plant sources, synthesis, stability, modification and food application. J Sci Food Agric. 100:1392–1404. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Peñalver P, Belmonte-Reche E, Adán N, Caro M, Mateos-Martín ML, Delgado M, González-Rey E and Morales JC: Alkylated resveratrol prodrugs and metabolites as potential therapeutics for neurodegenerative diseases. Eur J Med Chem. 146:123–138. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Chen C, Zou LX, Lin QY, Yan X, Bi HL, Xie X, Wang S, Wang QS, Zhang YL and Li HH: Resveratrol as a new inhibitor of immunoproteasome prevents PTEN degradation and attenuates cardiac hypertrophy after pressure overload. Redox Biol. 20:390–401. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Wang Y, Yang SH, Zhong K, Jiang T, Zhang M, Kwan HY and Su T: Network pharmacology-based strategy for the investigation of the anti-obesity effects of an ethanolic extract of zanthoxylum bungeanum maxim. Front Pharmacol. 11:5723872020. View Article : Google Scholar : PubMed/NCBI

8 

Lin X, Shao T, Huang L, Wen X, Wang M, Wen C and He Z: Simiao decoction alleviates gouty arthritis by modulating proinflammatory cytokines and the gut ecosystem. Front Pharmacol. 11:9552020. View Article : Google Scholar : PubMed/NCBI

9 

Wang W, Liu T, Yang L, Ma Y, Dou F, Shi L, Wen A and Ding Y: Study on the multi-targets mechanism of triphala on cardio-cerebral vascular diseases based on network pharmacology. Biomed Pharmacother. 116:1089942019. View Article : Google Scholar : PubMed/NCBI

10 

Wang J, Peng L, Jin L, Fu H and Shou Q: Network pharmacology analysis of the identification of phytochemicals and therapeutic mechanisms of paeoniae radix alba for the treatment of asthma. J Immunol Res. 2021:96593042021. View Article : Google Scholar : PubMed/NCBI

11 

Daina A, Michielin O and Zoete V: SwissTargetPrediction: Updated data and new features for efficient prediction of protein targets of small molecules. Nucleic Acids Res. 47((W1)): W357–W364. 2019. View Article : Google Scholar : PubMed/NCBI

12 

UniProt Consortium: UniProt: The universal protein knowledgebase in 2021. Nucleic Acids Res. 49(D1): D480–D489. 2021. View Article : Google Scholar : PubMed/NCBI

13 

Trott O and Olson AJ: AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 31:455–461. 2010.PubMed/NCBI

14 

Chen C, Chen H, Zhang Y, Thomas HR, Frank MH, He Y and Xia R: TBtools: An integrative toolkit developed for interactive analyses of big biological data. Mol Plant. 13:1194–1202. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Martin A, Ochagavia ME, Rabasa LC, Miranda J, Fernandez-de-Cossio J and Bringas R: BisoGenet: A new tool for gene network building, visualization and analysis. BMC Bioinformatics. 11:912010. View Article : Google Scholar : PubMed/NCBI

16 

Wang N, Zhang F, Yang L, Zou J, Wang H, Liu K, Liu M, Zhang H, Xiao X and Wang K: Resveratrol protects against L-arginine-induced acute necrotizing pancreatitis in mice by enhancing SIRT1-mediated deacetylation of p53 and heat shock factor 1. Int J Mol Med. 40:427–437. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Arocho A, Chen B, Ladanyi M and Pan Q: Validation of the 2-DeltaDeltaCt calculation as an alternate method of data analysis for quantitative PCR of BCR-ABL P210 transcripts. Diagn Mol Pathol. 15:56–61. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Peng S, Lu XF, Qi YD, Li J, Xu J, Yuan TY, Wu XY, Ding Y, Li WH, Zhou GQ, et al: LCZ696 ameliorates oxidative stress and pressure overload-induced pathological cardiac remodeling by regulating the Sirt3/MnSOD pathway. Oxid Med Cell Longev. 2020:98150392020. View Article : Google Scholar : PubMed/NCBI

19 

Wolf CM: Hypertrophic cardiomyopathy: Genetics and clinical perspectives. Cardiovasc Diagn Ther. 9 (Suppl 2):S388–S415. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Halliday BP, Cleland JGF, Goldberger JJ and Prasad SK: Personalizing Risk Stratification for sudden death in dilated cardiomyopathy: The past, present, and future. Circulation. 136:215–231. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Qiao Y, Zhu B, Tian A and Li Z: PEG-coated gold nanoparticles attenuate β-adrenergic receptor-mediated cardiac hypertrophy. Int J Nanomedicine. 12:4709–4719. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Galiniak S, Aebisher D and Bartusik-Aebisher D: Health benefits of resveratrol administration. Acta Biochim Pol. 66:13–21. 2019.PubMed/NCBI

23 

Du L, Chen E, Wu T, Ruan Y and Wu S: Resveratrol attenuates hydrogen peroxide-induced aging through upregulation of autophagy in human umbilical vein endothelial cells. Drug Des Devel Ther. 13:747–755. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Zhang X, Lu H, Xie S, Wu C, Guo Y, Xiao Y, Zheng S, Zhu H, Zhang Y and Bai Y: Resveratrol suppresses the myofibroblastic phenotype and fibrosis formation in kidneys via proliferation-related signalling pathways. Br J Pharmacol. 176:4745–4759. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Li W, Yuan G, Pan Y, Wang C and Chen H: Network pharmacology studies on the bioactive compounds and action mechanisms of natural products for the treatment of diabetes mellitus: A review. Front Pharmacol. 8:742017.PubMed/NCBI

26 

Zhang W, Bai Y, Wang Y and Xiao W: Polypharmacology in drug discovery: A review from systems pharmacology perspective. Curr Pharm Des. 22:3171–3181. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Liu TH, Tu WQ, Tao WC, Liang QE, Xiao Y and Chen LG: Verification of resveratrol inhibits intestinal aging by downregulating ATF4/Chop/Bcl-2/Bax signaling pathway. Based on network pharmacology and animal Front Pharmacol. 11:10642020.PubMed/NCBI

28 

Liu B, Palmfeldt J, Lin L, Colaço A, Clemmensen KKB, Huang J, Xu F, Liu X, Maeda K, Luo Y and Jäättelä M: STAT3 associates with vacuolar H+-ATPase and regulates cytosolic and lysosomal pH. Cell Res. 28:996–1012. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Wang W, Wang S, Liu T, Ma Y, Huang S, Lei L, Wen A and Ding Y: Resveratrol: Multi-targets mechanism on neurodegenerative diseases based on network pharmacology. Front Pharmacol. 11:6942020. View Article : Google Scholar : PubMed/NCBI

30 

You L, Wang Z, Li H, Shou J, Jing Z, Xie J, Sui X, Pan H and Han W: The role of STAT3 in autophagy. Autophagy. 11:729–739. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Cao X, Li B, Han X, Zhang X, Dang M, Wang H, Du F, Zeng X and Guo C: Soluble receptor for advanced glycation end-products promotes angiogenesis through activation of STAT3 in myocardial ischemia/reperfusion injury. Apoptosis. 25:341–353. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Lu X, Zhu Z, Jiang L, Sun X, Jia Z, Qian S, Li J and Ma L: Matrine increases NKG2D ligand ULBP2 in K562 cells via inhibiting JAK/STAT3 pathway: A potential mechanism underlying the immunotherapy of matrine in leukemia. Am J Transl Res. 7:1838–1849. 2015.PubMed/NCBI

33 

Zhang YG, Zhu X, Lu R, Messer JS, Xia Y, Chang EB and Sun J: Intestinal epithelial HMGB1 inhibits bacterial infection via STAT3 regulation of autophagy. Autophagy. 15:1935–1953. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Wang N, Xin H, Xu P, Yu Z and Shou D: Erxian decoction attenuates TNF-α induced osteoblast apoptosis by modulating the Akt/Nrf2/HO-1 signaling pathway. Front Pharmacol. 10:9882019. View Article : Google Scholar : PubMed/NCBI

35 

Goncalves GK, Scalzo S, Alves AP, Agero U, Guatimosim S and Reis AM: Neonatal cardiomyocyte hypertrophy induced by endothelin-1 is blocked by estradiol acting on GPER. Am J Physiol Cell Physiol. 314:C310–C322. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Wang XQ, Xu ZT, Zhang GP, Hou N, Mo QX, Wei J, Jiang X, Liu Y and Luo JD: Endophilin A2 attenuates cardiac hypertrophy induced by isoproterenol through the activation of autophagy. Am J Transl Res. 11:5065–5075. 2019.PubMed/NCBI

37 

Shirakabe A, Zhai P, Ikeda Y, Saito T, Maejima Y, Hsu CP, Nomura M, Egashira K, Levine B and Sadoshima J: Drp1-dependent mitochondrial autophagy plays a protective role against pressure overload-induced mitochondrial dysfunction and heart failure. Circulation. 133:1249–1263. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Zhang Y, Ding Y, Li M, Yuan J, Yu Y, Bi X, Hong H, Ye J and Liu P: MicroRNA-34c-5p provokes isoprenaline-induced cardiac hypertrophy by modulating autophagy via targeting ATG4B. Acta Pharm Sin B. 12:2374–2390. 2022. View Article : Google Scholar : PubMed/NCBI

39 

Luan Y, Luan Y, Feng Q, Chen X, Ren KD and Yang Y: Emerging role of mitophagy in the heart: Therapeutic potentials to modulate mitophagy in cardiac diseases. Oxid Med Cell Longev. 2021:32599632021. View Article : Google Scholar : PubMed/NCBI

40 

Li Y, Chen X, Li P, Xiao Q, Hou D and Kong X: CD47 antibody suppresses isoproterenol-induced cardiac hypertrophy through activation of autophagy. Am J Transl Res. 12:5908–5923. 2020.PubMed/NCBI

41 

Wang SY, Ni X, Hu KQ, Meng FL, Li M, Ma XL, Meng TT, Wu HH, Ge D, Zhao J, et al: Cilostazol alleviate nicotine induced cardiomyocytes hypertrophy through modulation of autophagy by CTSB/ROS/p38MAPK/JNK feedback loop. Int J Biol Sci. 16:2001–2013. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Xie C, Liu S, Wu B, Zhao Y, Chen B, Guo J, Qiu S and Cao YM: miR-19 promotes cell proliferation, invasion, migration, and EMT by inhibiting SPRED2-mediated autophagy in osteosarcoma cells. Cell Transplant. 29:9636897209624602020. View Article : Google Scholar : PubMed/NCBI

43 

Tang Z, Takahashi Y, Chen C, Liu Y, He H, Tsotakos N, Serfass JM, Gebru MT, Chen H, Young MM and Wang HG: Atg2A/B deficiency switches cytoprotective autophagy to non-canonical caspase-8 activation and apoptosis. Cell Death Differ. 24:2127–2138. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Feng Q, Wang H, Pang J, Ji L, Han J, Wang Y, Qi X, Liu Z and Lu L: Prevention of wogonin on colorectal cancer tumorigenesis by regulating p53 nuclear translocation. Front Pharmacol. 9:13562018. View Article : Google Scholar : PubMed/NCBI

45 

Mariño G, Niso-Santano M, Baehrecke EH and Kroemer G: Self-consumption: The interplay of autophagy and apoptosis. Nat Rev Mol Cell Biol. 15:81–94. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Valcourt DM, Dang MN, Scully MA and Day ES: Nanoparticle-mediated co-delivery of notch-1 antibodies and ABT-737 as a potent treatment strategy for triple-negative breast cancer. ACS Nano. 14:3378–3388. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Cusack CL, Swahari V, Hampton Henley W, Michael Ramsey J and Deshmukh M: Distinct pathways mediate axon degeneration during apoptosis and axon-specific pruning. Nat Commun. 4:18762013. View Article : Google Scholar : PubMed/NCBI

48 

Acuner Ozbabacan SE, Keskin O, Nussinov R and Gursoy A: Enriching the human apoptosis pathway by predicting the structures of protein-protein complexes. J Struct Biol. 179:338–346. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 26 Issue 5

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
Ren S, Shen L, Lin S, Xiao D, Xiao W, Yan P, Zhang Y, Jia W and Lin Y: Mechanistic analysis of resveratrol in cardiac hypertrophy by network pharmacology and animal experiments. Mol Med Rep 26: 324, 2022
APA
Ren, S., Shen, L., Lin, S., Xiao, D., Xiao, W., Yan, P. ... Lin, Y. (2022). Mechanistic analysis of resveratrol in cardiac hypertrophy by network pharmacology and animal experiments. Molecular Medicine Reports, 26, 324. https://doi.org/10.3892/mmr.2022.12840
MLA
Ren, S., Shen, L., Lin, S., Xiao, D., Xiao, W., Yan, P., Zhang, Y., Jia, W., Lin, Y."Mechanistic analysis of resveratrol in cardiac hypertrophy by network pharmacology and animal experiments". Molecular Medicine Reports 26.5 (2022): 324.
Chicago
Ren, S., Shen, L., Lin, S., Xiao, D., Xiao, W., Yan, P., Zhang, Y., Jia, W., Lin, Y."Mechanistic analysis of resveratrol in cardiac hypertrophy by network pharmacology and animal experiments". Molecular Medicine Reports 26, no. 5 (2022): 324. https://doi.org/10.3892/mmr.2022.12840