
Thymoquinone mitigates cardiac hypertrophy by activating adaptive autophagy via the PPAR‑γ/14‑3‑3γ pathway
- Authors:
- Published online on: February 7, 2025 https://doi.org/10.3892/ijmm.2025.5500
- Article Number: 59
-
Copyright: © Qiu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Myocardial hypertrophy represents a fundamental adaptive response of the heart to diverse stressors, frequently characterized by the enlargement of myocardial cells and increased fibrosis. Initially, this adaptation functions as a compensatory process aimed at preserving cardiac function. However, over time, sustained hypertrophy disrupts myocardial performance, precipitating heart failure (1). Myocardial hypertrophy is widely recognized as an independent predictor of cardiovascular-related morbidity and mortality, with prolonged hypertrophy driving structural remodeling, functional decline, heart failure progression and, in severe cases, sudden cardiac death (2). Epidemiological data demonstrate that myocardial hypertrophy is a leading cause of morbidity and mortality in heart failure cases (3). Cardiac hypertrophy typically manifests with cardiomyocyte enlargement, fibrosis, myofibrillar disarray, cell death, and increased protein synthesis (4). Underlying mechanisms often include mitochondrial dysfunction, cardiomyocyte fibrosis, apoptosis and the overproduction of reactive oxygen species (ROS) (5). Investigating pharmacological interventions that target these pathways may offer potential therapeutic strategies for controlling this pathological condition (6).
Thymoquinone (TQ), the major bioactive compound in Nigella sativa seeds, plays a significant role in mediating various biological activities of the plant (7). Studies have identified the therapeutic benefits of TQ across multiple pathological conditions, including tumorigenesis, autoimmune disorders, diabetes and neurodegenerative diseases, primarily through pathways involving immune modulation, suppression of apoptosis, attenuation of oxidative stress and neutralization of free radicals (8,9). However, the precise mechanism through which TQ exerts its protective effects on cardiac hypertrophy remains elusive.
Autophagy is a process that entails the engulfment of cytoplasmic proteins or organelles, their encapsulation into vesicles and subsequent fusion with lysosomes to form autolysosomes for degradation. In cardiomyocytes, adaptive autophagy helps eliminate damaged organelles, ensuring the preservation of cardiac function. However, both excessive activation and marked suppression of autophagy can compromise cardiac structure and function (10). Accumulating evidence indicates a key involvement of autophagy in cardiac hypertrophy. Xue et al (11) demonstrated that Sestrin 1 mitigated phenylephrine-induced cardiac hypertrophy by modulating the AMP-activated protein kinase (AMPK)/mTORC1 autophagy pathway. Furthermore, the deletion of ATPase inhibitory factor 1 enhances AMPK activity, boosts autophagy and alleviates transverse aortic constriction (TAC)-induced cardiac hypertrophy (12). Thus, targeting autophagy regulation offers potential therapeutic value for cardiac hypertrophy.
Peroxisome proliferator-activated receptor-γ (PPAR-γ) belongs to a family of ligand-induced transcription factors within the nuclear receptor superfamily (13). Activation of PPAR-γ can reduce inflammation, inhibit oxidative stress and enhance cardiomyocyte energy metabolism, thus inhibiting myocardial remodeling and mitigating myocardial hypertrophy (14). Research has demonstrated that thiazolidinediones exhibit efficacy in enhancing insulin sensitivity and mitigating hyperglycemia through their targeting of PPAR-γ, and they have gained widespread application in the treatment of type 2 diabetes (15). Since PPAR-γ targeted drugs have been poorly studied in cardiac hypertrophy, it is of clinical interest to investigate natural products that can effectively activate PPAR-γ to alleviate cardiac hypertrophy.
The objective of the present study was to validate the protective effect of TQ on cardiac hypertrophy both in vitro and in vivo, as well as to delve into the underlying mechanisms and provide key insights into the therapeutic potential of TQ in both preventing and managing cardiac hypertrophy.
Materials and methods
Reagents and chemicals
TQ (purity ≥99.59%), 3-Methyladenine (3-MA; purity ≥99.91%) and GW9662 (PPAR-γ antagonist; purity ≥99.87%) were sourced from MedChemExpress, while pAD/14-3-3γ-short hairpin (sh)RNA was procured from Cyagen Biosciences, Inc. AngII was acquired from GLPBIO Technology LLC. PPAR-γ (cat. no. YT3836) antibody was supplied by ImmunoWay Biotechnology Company. Primary antibodies for LC3 (cat. no. 381544), p62 (cat. no. 380612), 14-3-3γ (cat. no. R381405) and secondary antibodies (goat anti-mouse, cat. no. 511103; goat anti-rabbit, cat. no. 511203) were purchased from Chengdu Zen-Bioscience Co., Ltd. Proteintech Group, Inc. provided antibodies against collagen I (cat. no. 14695-1-AP), atrial natriuretic peptide (ANP; cat. no. 27426-1-AP) and β-actin (cat. no. 66009-1-Ig). ABclonal, Inc. provided antibodies against brain natriuretic peptide (BNP; cat. no. A2179).
Cell model of cardiac hypertrophy and treatment
The rat H9C2 cell line, obtained from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences, was maintained in H-DMEM (HyClone; Cytiva), supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.). The H9C2 cells were incubated at 37°C in a controlled environment (95% humidity, 21% O2, and 5% CO2) (16). The H9C2 cells were allocated into six distinct experimental groups: i) Control group, H9C2 cells were maintained in H-DMEM for 48 h under standard conditions; ii) AngII group: H9C2 cells were cultured in H-DMEM for 24 h, followed by exposure to 1 µM AngII for an additional 24 h (17); iii) AngII + TQ group: Cells were pretreated with 10 µM TQ for 24 h before subsequent exposure to 1 µM AngII for another 24 h; iv) AngII + TQ + 3-MA group: Cells were incubated with 10 µM TQ and 5 mM 3-MA (18) for 24 h, followed by 1 µM AngII treatment for 24 h; v) AngII + TQ + GW9662 group: Cells were pretreated with 10 µM TQ and 10 µM GW9662 (19) for 24 h, then exposed to 1 µM AngII for an additional 24 h; and vi) AngII + TQ + pAD/14-3-3γ-shRNA group: H9C2 cardiomyocytes were transfected with pAd/14-3-3γ-shRNA at 37°C for 48 h, incubated with 10 µM TQ for 24 h and subsequently treated with 1 µM AngII for another 24 h.
Adenovirus transfection
H9C2 cardiomyocytes were pre-cultured in fresh H-DMEM supplemented with 10% FBS and then transfected with either pAd/14-3-3γ-shRNA or pAd/negative control-shRNA (Cyagen Biosciences, Inc.; concentration not measured) using HighGene plus Transfection reagent (cat. no. RM09014; ABclonal Biotech Co., Ltd.). The transfection efficiency was ~85% after 48 h at 37°C, when the subsequent experiments were carried out. The shRNA sequences are provided in Table I.
Cell viability assay
Cell viability was determined via the Cell Counting Kit-8 (CCK-8; GLPBIO Technology LLC). H9C2 cells were cultured in 96-well plates at a density of 2×104 cells/well and exposed to TQ at concentrations of 1, 5, 10 and 20 µM for 24 h. Subsequently, 10 µl CCK-8 reagent was introduced to each well for 1 h at 37°C, and the absorbance at 450 nm was recorded using a microplate reader (Thermo Fisher Scientific, Inc.).
Determination of oxidative stress and lysosome detection
ROS production levels were assessed via staining with dichlorofluorescein diacetate (DCFH-DA; Beyotime Institute of Biotechnology). H9C2 cells were exposed to 10 µM DCFH-DA for 20 min at 37°C in the dark, followed by three washes with H-DMEM lacking 10% FBS. Intracellular ROS was subsequently visualized using a fluorescence microscope (Olympus Corporation). Lyso-Tracker Red (Beyotime Institute of Biotechnology), a lysosomal red fluorescent probe, enables lysosome-specific staining in live cells by penetrating cell membranes. H9C2 cells were incubated with the Lyso-Tracker Red working solution for 30 min at 37°C in the dark, and fluorescence microscopy (Olympus Corporation) was employed for observation.
Transmission electron microscopy (TEM)
Cells were harvested and fixed in 2% glutaraldehyde at 25°C for 2 h, followed by sequential washing with PBS, dehydration, embedding, sectioning and staining. The sections were 60-80 nm thick and were embedded at 37°C for 5-8 h using EPON 812. Staining was performed using 2% uranyl acetate and 2.6% lead citrate at 37°C for 8 min. Autophagosome structures in H9C2 cells were analyzed via TEM (Hitachi 7800; Hitachi, Ltd.).
Western blot analysis
Total proteins were isolated from H9C2 cells and mouse myocardial tissue using RIPA lysis buffer (Beijing Solarbio Science & Technology Co., Ltd.). Protein concentrations were determined via the BCA Assay Kit (GLPBIO Technology LLC). Equivalent amounts of protein (30-40 µg) were resolved via 10-12.5% gradient SDS-PAGE and subsequently transferred to polyvinylidene fluoride membranes (Pall Corporation). Membranes were blocked with 5% skimmed milk at room temperature for 2 h, followed by overnight incubation at 4°C with primary antibodies against LC3 (1:1,000), p62 (1:1,000), 14-3-3γ (1:1,000), β-actin (1:1,000), collagen I (1:500) and PPAR-γ (1:500) on a shaker. After washing three times using TBST (0.05% Tween 20), the membranes were incubated with HRP-conjugated secondary antibodies (1:5,000) for 2 h at room temperature. β-actin served as a loading control. Finally, protein bands were visualized using the Ultra High Sensitivity ECL kit (cat. no. GK10008; GLPBIO Technology LLC) and protein band densities were semi-quantified using ImageJ software (National Institutes of Health; v1.8.0.345).
RNA isolation and reverse transcription-quantitative PCR (RT-qPCR)
RT-qPCR analysis was performed to quantify the mRNA expression levels of ANP, BNP, NADPH oxidase 4 (NOX4), superoxide dismutase 2 (SOD2) and collagen I. Total RNA was isolated with TRIzol reagent (Beijing Solarbio Science & Technology Co., Ltd.), followed by RT using the PrimeScript RT reagent kit (Monad Biotech Co., Ltd.) according to the manufacturer's instructions. qPCR was performed using the SYBR Green qPCR Master Mix (cat. no. B21203; Selleck Chemicals) on the BIO-RAD CFX Connect Real-Time PCR Detection System from Bio-Rad Laboratories, Inc. The qPCR protocol comprised an initial denaturation step at 95°C for 10 min, followed by 40 cycles of denaturation at 95°C for 15 sec and annealing plus extension at 60°C for 1 min. β-actin served as the internal reference and the relative mRNA expression levels of the target genes were calculated employing the 2−ΔΔCq method (20). The specific primers for each gene are detailed in Table II.
Immunofluorescence staining
LC3 expression was evaluated using immunofluorescence staining. After washing the cell samples with PBS fixation was carried out with 4% paraformaldehyde (Biosharp Life Sciences) at room temperature for 10 min, then permeabilization for 10 min at room temperature using 0.2% Triton X-100 (Beijing Solarbio Science & Technology Co., Ltd.), followed by blocking with 2% BSA (cat. no. 9048-46-8; Shanghai Yuanye Biotechnology Co., Ltd.) at room temperature for 30 min. The cells were subsequently incubated overnight at 4°C with rabbit anti-LC3 primary antibody (1:500). Following three washes with TBST (0.05% Tween 20), fluorescent secondary goat anti-rabbit antibody (1:200; cat.no. A32732; Thermo Fisher Scientific, Inc.) was applied for 1 h at room temperature. DAPI counterstaining was performed at room temperature for 5 min, and fluorescence microscopy was used for observation.
Luciferase activity assay
Luciferase activity was assessed using a dual-luciferase reporter assay system (Promega Corporation; cat. no. E1910 Transfection of pcDNA3.1-PPAR-γ and psiCheck2-14-3-3γ (Hunan Fenghui Biotechnology Co., Ltd.) constructs into cells was carried out with LipoFiter (cat. no. HB-LF-1000; Hanbio Biotechnology Co., Ltd.) for 6 h at 37°C. Subsequently, 100 µl each of firefly and Renilla luciferase detection reagents were applied, followed by the measurement of relative light units. The luciferase activity ratio between firefly and Renilla was then calculated.
Molecular docking
The three-dimensional structures of TQ and PPAR-γ were obtained from PubChem (Compound CID: 10281) and the Protein Data Bank (PDB) (PDB ID: 3PRG), respectively. The CB-Dock2 (https://cadd.labshare.cn/cb-dock2/php/index.php) server was utilized to carry out molecular docking.
Animal model of cardiac hypertrophy and treatment
The experimental procedures followed the guidelines established by the National Institutes of Health and were approved by the Animal Experimentation Ethics Committee of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University (Nanchang, China; approval no. CDYFY-IACUC-202407QR115). Male C57BL/6 mice (22-24 g, 8 weeks old) were sourced from Changzhou Cavens Laboratory Animal Ltd. The mice were maintained in controlled environmental conditions, with a temperature of 23±1°C, a humidity of 40-50%, a 12-h light/dark cycle to simulate day and night, and ad libitum access to food and water to ensure their well-being and experimental accuracy.
The mice were randomly allocated into four groups: i) Sham operation, ii) TAC operation, iii) TAC operation with TQ treatment, and iv) TAC operation with TQ treatment alongside PPAR-γ antagonist (GW9662). Each group comprised 6 mice. The mouse model of pressure overload-induced cardiac hypertrophy was generated through TAC surgery, following the protocol outlined in a previous study (21). For the operation, animals were anesthetized in a chamber with isoflurane (induced with 2% and maintained with 1.5%), supplemented during surgery with 0.1 mg/kg buprenorphine HCl via subcutaneous injection. Post-surgery, the health and behavior of the mice were monitored twice daily. In the TQ treatment group, TQ (50 mg/kg, dissolved in corn oil) was administered via gavage for 6 weeks post-TAC (22). The PPAR-γ antagonist group received intraperitoneal injections of GW9662 (1 mg/kg) after TAC surgery, administered every 3 days for 6 weeks.
The animal was euthanized if predefined humane endpoints were met, including: i) Rapid weight loss of 15-20% of the original body weight (BW); ii) the inability to eat, drink or stand for up to 24 h; and iii) depression and hypothermia (<37°C) without anesthesia or sedation. No mice exhibited signs of reaching these endpoints during the experiment. Euthanasia was induced by an overdose of pentobarbital sodium (200 mg/kg). Animals were observed for 5-10 min post-injection to confirm death, assessed by respiratory and cardiac arrest, and loss of corneal reflex.
Histological analysis
Mouse heart weight (HW) and BW were recorded. Heart tissue samples were preserved in formalin for 12 h at room temperature and embedded in paraffin following established protocols (23). Paraffin-embedded specimens were sectioned at 5 µm thickness and subjected to hematoxylin and eosin staining for 5 min at room temperature as well as Masson's trichrome staining for 5 min at room temperature, using standard procedures (24,25). The myocardial cell size was observed from ventricular slices stained with fluorescein isothiocyanate-labeled wheat germ agglutinin (WGA) at 37°C in the dark for 2 h. Finally, the sections were examined under an inverted fluorescence microscope (Nikon Eclipse Ti-SR), and images were subsequently captured.
Echocardiography
After anesthetizing the mice with 1.5% isoflurane, echocardiography was performed. Cardiac function was assessed using a small animal ultrasound imaging system (VEVO2100; FUJIFILM VisualSonics, Inc.; FUJIFILM Sonosite, Inc.) featuring a 30-MHz probe.
Statistical analysis
Data analysis was conducted using GraphPad Prism 9.0 software (Dotmatics). For two-group comparisons, unpaired Student's t-test was applied, while one-way ANOVA followed by the Tukey's post hoc test was employed for multiple group comparisons. P<0.05 was considered to indicate a statistically significant difference.
Results
Effects of TQ on the viability of H9C2 cells
H9C2 cell viability under varying concentrations of TQ (0, 1, 5, 10 and 20 µM) and AngII (1 µM) was evaluated using the CCK-8 assay. Treatment with TQ at different concentrations did not significantly alter cell viability compared with the control (0 µM) group (Fig. 1A). By contrast, AngII (1 µM) significantly reduced cell viability, an effect that was attenuated by TQ pretreatment (Fig. 1B). As the 10 µM concentration demonstrated the most notable protective effect, it was selected for subsequent experiments. The chemical structure of TQ is illustrated in Fig. 1C and D.
TQ attenuates cardiomyocyte hypertrophy in vitro and in vivo
To assess the potential protective effects of TQ against AngII-induced hypertrophy in H9C2 cells, cells were pretreated with AngII (1 µM) for 24 h to induce hypertrophy, followed by evaluation of the cell surface area through WGA immunostaining. Pretreatment with TQ significantly decreased the cell surface area compared with the AngII group (Fig. 2A). In addition, analysis of the protein and mRNA expression levels of key hypertrophic markers, ANP and BNP, indicated that TQ significantly downregulated the expression of these markers (Fig. 2B-D). These results collectively indicated that TQ mitigated the hypertrophic response triggered by AngII in H9C2 cells.
To assess the cardioprotective effects of TQ on cardiac hypertrophy, a pressure overload hypertrophy mouse model was induced via TAC surgery. Cardiac function was analyzed through echocardiography (Fig. 3A). Mice in the TAC group exhibited a decline in left ventricular ejection fraction and fractional shortening, alongside an increase in left ventricular internal dimension diastole and posterior wall dimension, compared with the sham group (Fig. 3B-E). However, post-TAC administration of TQ improved cardiac function and attenuated hypertrophy in mice (Fig. 3A-E). Furthermore, pretreatment with TQ alleviated TAC-induced hypertrophy, as indicated by a reduction in cardiac size (Fig. 3G). Additionally, a marked increase in heart size and myofibril degeneration was observed in the TAC group, while the TQ-treated group exhibited reduced heart size and improved myofibril integrity, as shown by hematoxylin and eosin staining (Fig. 3H). The HW/BW ratio was significantly lower in the TAC + TQ group compared with the TAC group (Fig. 3F). Additionally, the protein and mRNA expression levels of ANP and BNP were notably down-regulated in the TAC + TQ group relative to the TAC group (Fig. 3I-L). These findings indicated that TQ attenuated pressure overload-induced cardiac hypertrophy.
TQ inhibits cardiac fibrosis in vivo and in vitro
The inter-relation between cardiac fibrosis and hypertrophy has been well-established, with fibrosis progression impairing myocardial contractility, ultimately contributing to heart failure and potential mortality (26,27). Masson staining, immunoblotting and RT-qPCR were employed to assess the effects of TQ on cardiac fibrosis. Masson staining showed a marked increase in fibrosis in mice subjected to TAC surgery, which was markedly attenuated by TQ treatment (Fig. 4A). The protein and mRNA levels of type I collagen were significantly upregulated in the TAC group compared with the control but were reduced following TQ administration (Fig. 4B-D). Similarly, in vitro experiments using H9C2 cells revealed elevated expression of type I collagen in the AngII group, which was mitigated by TQ, aligning with the in vivo findings (Fig. 4E-G).
TQ attenuates oxidative stress levels in cardiac hypertrophy cells
Excessive generation of ROS is recognized as a key mechanism contributing to the progression of cardiac hypertrophy (5). Elevated ROS accumulation in cardiomyocytes has been shown to aggravate cardiac hypertrophy and myocardial fibrosis, ultimately leading to heart failure (28). To explore the potential antioxidative effect of TQ in cardiac hypertrophy, ROS levels were measured in treated cells. A notable increase in ROS levels were observed in the AngII group, while TQ treatment markedly reduced ROS levels (Fig. 5A). Additionally, RT-qPCR and western blot analysis indicated that TQ significantly downregulated the mRNA and protein expression of the oxidation gene, NOX4, and significantly upregulated the mRNA and protein expression of antioxidation gene, SOD2, in both the TAC-induced and AngII-induced groups (Fig. 5B-G).
TQ activates adaptive autophagy in H9C2 hypertrophy cardiomyocytes
Autophagy plays a central role in the pathogenesis of cardiac hypertrophy, with its dysregulation contributing to the worsening of the condition (29,30). To evaluate the effect of TQ pretreatment on adaptive autophagy in H9C2 cells, the expression levels of autophagy markers, LC3II and p62, as well as lysosome counts, were analyzed. As shown in Fig. 6A-C, TQ + AngII treatment significantly increased LC3II expression while reducing p62 levels compared with AngII alone. This modulation was attenuated by the autophagy inhibitor, 3-MA. These results suggested that autophagy was downregulated during the onset of hypertrophy in cardiac cells and that TQ pretreatment restored autophagic activity under these conditions. Furthermore, LysoTracker Red staining revealed a reduction in lysosome numbers following AngII-induced treatment of H9C2 cells, which was reversed upon TQ administration. Notably, the addition of 3-MA markedly reduced fluorescence intensity in H9C2 cells (Fig. 6D). Immunofluorescence analysis indicated an elevation in LC3 expression following TQ pretreatment, which was markedly reversed by 3-MA (Fig. 6E). Thus, we found that TQ activated adaptive autophagy in H9C2 hypertrophy cardiomyocyte.
TQ alleviates hypertrophy in cardiac cells by upregulating 14-3-3γ expression to activate adaptive autophagy
The 14-3-3 proteins, a group of highly conserved acidic proteins found in all eukaryotic cells, comprise seven isoforms (31). Research has established their role in regulating autophagy and their protective function in cardiomyocytes (16,32). To investigate whether TQ conferred protection via autophagy regulation mediated by 14-3-3γ, the expression of 14-3-3γ was knocked down in H9C2 cells, the success of which was verified by western blot analysis (Fig. S1). As shown in Fig. 7A and B, TQ pretreatment led to a significant upregulation of 14-3-3γ expression, an effect that was reversed by pAD/14-3-3γ shRNA. In addition, knockdown of 14-3-3γ significantly influenced autophagic activity (Fig. 7A, C and D). Specifically, pAD/14-3-3γ shRNA reduced LC3II expression while simultaneously increasing p62 expression compared with the TQ group. TEM further confirmed a reduction in autophagic vesicles and therefore a suppression of autophagy in H9C2 cells treated with pAD/14-3-3γ shRNA (Fig. 7E). Additionally, pAD/14-3-3γ shRNA upregulated the mRNA and protein expression levels of ANP and BNP compared with the AngII + TQ group (Fig. 7F-H). These experiments confirmed that TQ alleviated cardiac hypertrophy by upregulating 14-3-3γ.
TQ targets PPAR-γ and attenuates cardiac hypertrophy through activating adaptive autophagy
PPAR-γ, a protective regulator, is essential for maintaining cardiac homeostasis and preventing heart failure (33-35). We hypothesized that TQ could specifically target PPAR-γ and activate adaptive autophagy, thereby alleviating cardiac hypertrophy. To test this conjecture, the protein expression level of PPAR-γ was detected using western blot analysis. The results demonstrated that PPAR-γ expression in hypertrophic cardiac tissue revealed was significantly downregulated compared with the control group. However, TQ treatment effectively restored PPAR-γ levels. Notably, the co-administration of a PPAR-γ inhibitor (GW9662) reversed the TQ-mediated upregulation of PPAR-γ (Fig. 8A-D). Molecular docking techniques were employed to explore the regulatory interaction between TQ and PPAR-γ, simulating their binding. This method is widely used to evaluate compound interactions and activity (36). The analysis demonstrated that TQ formed hydrogen bonds with several key sites on PPAR-γ (Fig. 8F), suggesting that PPAR-γ modulation may be a potential mechanism of TQ's effects. Furthermore, similar results in AngII-pretreated H9C2 cells as those obtained from in vivo experiments were observed. The results indicated that the autophagy levels were suppressed as LC3II expression decreased and p62 expression increased in the AngII + TQ + GW9662 group treated compared with the AngII + TQ group (Fig. 8A-D). TEM further confirmed the reduction of autophagic vesicles in the PPAR-γ inhibitor-treated group, consistent with the western blot results (Fig. 8E). Additionally, PPAR-γ inhibition reversed the protective effects of TQ on cardiac hypertrophy, resulting in an enlarged cardiomyocyte surface area (Fig. 8G) and elevated the protein and mRNA expression of ANP and BNP (Fig. 8H-J).
TQ attenuates cardiac hypertrophy by activating adaptive autophagy through the PPAR-γ/14-3-3γ pathway
Inhibition of 14-3-3γ expression was observed upon treatment with the PPAR-γ inhibitor, GW9662. The suppression of PPAR-γ resulted in a corresponding reduction in 14-3-3γ levels (Fig. 9A and C). Similarly, knockdown of 14-3-3γ via pAD/14-3-3γ shRNA also diminished PPAR-γ expression (Fig. 9B and D). Moreover, the dual-luciferase assay demonstrated that PPAR-γ upregulated 14-3-3γ promoter activity (Fig. 9E). These results suggest that TQ mitigates cardiac hypertrophy by promoting adaptive autophagy through the PPAR-γ/14-3-3γ signaling axis (Fig. S2).
Discussion
Myocardial hypertrophy constitutes a significant risk factor for numerous cardiac pathologies. Prolonged hypertrophy often leads to a diminished left ventricular ejection fraction and progressive cardiac dysfunction, ultimately resulting in heart failure and increased mortality rates (37). At present, no specific pharmacological intervention exists for the effective treatment of cardiac hypertrophy in clinical practice. Thus, investigating the molecular mechanisms underlying the development of cardiac hypertrophy is essential for the discovery and advancement of novel therapeutic agents. In the present study, TQ treatment demonstrated a notable reduction in cardiac hypertrophy both in vitro, using H9C2 cells, and in vivo, utilizing a TAC-induced cardiac hypertrophy mouse model. TQ was found to markedly lower ROS levels, mitigate myocardial fibrosis, enhance autophagic activity in the context of cardiac hypertrophy and provide protection against hypertrophy-induced cardiac dysfunction. Additionally, TQ activated the PPAR-γ and 14-3-3γ signaling pathways, leading to enhanced autophagy and suppression of pathological cardiac hypertrophy. These results suggest that TQ holds considerable promise as a potential therapeutic agent for the treatment of cardiac hypertrophy and heart failure, with strong prospects for clinical application.
Chinese herbal medicines have gained prominence in the treatment of cardiovascular diseases (38). Baicalein, for instance, enhances catalase expression to eliminate ROS, binds to FOXO3a, promotes its transcriptional activity and activates autophagy, thereby alleviating cardiac hypertrophy (39). Similarly, Sophora effectively mitigates TAC-induced cardiac hypertrophy, protects against hypertrophy-related cardiac dysfunction, reduces myocardial fibrosis and activates AMPK/mTORC1-mediated autophagy to counteract hypertrophy (40). Berberine administration also ameliorates TAC-induced cardiac hypertrophy, reduces myocardial apoptosis, limits fibrosis and upregulates autophagy, providing cardioprotective effects in hypertrophy models (41). Despite these advances, research on the effects of TQ in cardiac hypertrophy remains limited. The present study highlights TQ's potential to attenuate cardiac hypertrophy, presenting a novel therapeutic strategy for managing the condition.
Oxidative stress arises from an imbalance between antioxidant defenses and ROS production, with excessive ROS leading to cellular damage. A major contributor to cardiac hypertrophy is oxidative stress driven by elevated ROS levels in cardiomyocytes (42). Persistent oxidative stress has been implicated in the progression of myocardial hypertrophy to heart failure (43). Evidence suggests that controlling oxidative stress can mitigate cardiac hypertrophy and hinder its transition to heart failure (44,45). In the present study, in the AngII-induced H9C2 cell hypertrophy model, ROS levels were assessed through DCFH-DA staining under fluorescence microscopy and the expression of ROS-related genes via qPCR. AngII markedly elevated ROS production in cells, while TQ pretreatment attenuated ROS levels, attributed to its potent antioxidant properties. However, a ROS inhibitor was not employed to further verify the impact of TQ on cardiac hypertrophy.
Autophagy is a conserved cellular mechanism responsible for degrading proteins and damaged organelles, playing a vital role in maintaining survival, development and homeostasis, and is therefore integral to human health and development (46). This homeostatic process ensures the degradation and recycling of cellular components under both physiological and stress conditions. Impaired autophagy results in disrupted ubiquitination, ROS accumulation and compromised mitochondrial function (47). Autophagy activation has been shown to alleviate cardiac hypertrophy and mitigate cardiac dysfunction in stress-induced hypertrophy models (48). The present study reinforces the hypothesis that enhancing autophagy can reduce the progression of cardiac hypertrophy. The present study utilized TEM and the Lyso-Tracker method to examine autophagic lysosomes, while western blotting was used to quantify the LC3 protein levels to evaluate autophagic activity. LC3, a widely recognized autophagy marker, is commonly used to assess autophagy, with LC3II levels closely linked to autophagosome numbers, and the LC3II/LC3I ratio serving as an indicator of autophagic flux (49). In the cardiac hypertrophy model, the protein expression level of LC3II was significantly reduced compared with the TQ-treated group, indicating that TQ mitigates cardiac hypertrophy and improves cardiac function by promoting autophagy. This observation is consistent with a prior study (48). Notably, to the best of our knowledge, the present study is the first to establish the role of TQ in attenuating cardiac hypertrophy through autophagy enhancement.
PPARs are part of the ligand-activated transcription factor family within the nuclear receptor superfamily, comprising three isoforms: PPAR-α, PPAR-δ and PPAR-γ (50). PPAR-γ, predominantly expressed in adipose tissue, governs adipocyte differentiation and the regulation of genes involved in lipid storage (13). PPAR-γ has been identified as a crucial regulator of adipose development and systemic metabolism, with therapeutic potential in enhancing insulin sensitivity in diabetic patients (51). Activation of PPAR-γ has been shown to prevent myocardial hypertrophy and attenuate post-myocardial infarction remodeling by reducing inflammation, oxidative stress, cell death and improving cardiomyocyte energy metabolism (13,50,52,53). In the present study, AngII exposure inhibited PPAR-γ expression in cultured H9C2 cells, while TQ treatment restored its activation. Notably, the cardioprotective effect of TQ disappeared upon administration of the PPAR-γ inhibitor, GW9662. To the best of our knowledge, the present study presents the first evidence that TQ mitigates cardiac hypertrophy via PPAR-γ activation.
The 14-3-3 proteins are ubiquitously expressed across animal and plant tissues, serving critical roles in cellular biology and signal transduction pathways (54). Numerous studies have identified 14-3-3 proteins as endogenous cardioprotective agents, offering protection in various heart injury models. For instance, curcumin has been shown to mitigate doxorubicin (DOX)-induced cardiotoxicity by upregulating 14-3-3γ, which in turn reduces serum lactate dehydrogenase (LDH) activity, inhibits apoptosis and limits mitochondrial damage (55). Quercetin has been shown to enhance cardiomyocyte viability, elevate SOD and catalase activity, and decrease LDH, ROS and malondialdehyde levels through the upregulation of 14-3-3γ, thereby mitigating DOX-induced cardiotoxicity. The protective effect is diminished when 14-3-3γ expression is knocked down (56). However, the role of 14-3-3γ in cardiac hypertrophy remains uncertain. In the present study, 14-3-3γ expression was reduced in the cardiac hypertrophy model but increased in the TQ-treated group. Knockdown of 14-3-3γ using pAD/14-3-3γ in the TQ group weakened the protective effects of TQ and suppressed autophagy. To the best of our knowledge, the present study is the first to demonstrate that TQ alleviates cardiac hypertrophy by upregulating 14-3-3γ and promoting autophagy.
Research has demonstrated that PPAR-γ plays a central role in regulating lipid metabolism and inflammation (57). PPAR-γ is crucial for maintaining cellular energy homeostasis, modulating inflammatory responses and controlling fibrosis (58). As a transcription factor, PPAR-γ regulates TGF-β1 expression and Smad2/3 phosphorylation, thereby inhibiting hepatic stellate cell activation and mitigating liver fibrosis (59). PPAR-γ activation also suppresses platelet derived growth factor and tissue inhibitory of metalloproteinase-2 expression in non-alcoholic fatty liver disease, reducing inflammation and fibrosis (60). We hypothesize that PPAR-γ may bind to the 14-3-3γ promoter to alleviate cardiac hypertrophy. Notably, in the present study, both 14-3-3γ and PPAR-γ protein expression increased in the TQ group but declined following PPAR-γ inhibitor (GW9662) treatment. A dual-luciferase reporter assay confirmed that PPAR-γ enhanced 14-3-3γ promoter activity. However, the absence of chromatin immunoprecipitation, Baf A1-induced LC3-I/II conversion and mRFP-LC3 tandem fluorescence assays in the present study limits its conclusiveness, necessitating further investigation. Additionally, primary cardiomyocytes, which were not used in the present study, may better mimic the in vivo environment and produce more convincing findings.
In conclusion, TQ appears to play a notable role in preventing pressure overload-induced cardiac hypertrophy by reducing oxidative stress and inhibiting fibrosis. TQ alleviates cardiac hypertrophy by modulating 14-3-3γ expression and enhancing autophagy through the activation of PPAR-γ transcriptional activity. This mechanism offers a potential therapeutic strategy for the clinical treatment of cardiac hypertrophy. Nonetheless, the present study contains certain limitations. First, while TQ exhibited a protective effect, its efficacy remains limited, necessitating further investigation across additional cardiac hypertrophy models. Second, the precise mechanism by which TQ induces autophagy through the PPAR-γ/14-3-3γ pathway requires further elucidation.
Supplementary Data
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
RBQ conducted the cell experiments and analyzed and mapped the experimental data. STZ provided the experimental design and data for analysis. ZQX conducted animal experiments and analyzed the results. RYZ, ZCQ, HZP and LFZ contributed to the cell experiments. LJH enhanced the language of the article and analyzed the data. YPC and LW designed the experiments and provided financial support. RYZ, ZCQ and HZP confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
Ethics approval and consent to participate
Animal experiments followed the guidelines of the National Institutes of Health and were authorized by the Animal Experimentation Ethics Committee of the First Affiliated Hospital, Jiangxi Medical College, Nanchang University (Nanchang, China; approval no. CDYFY-IACUC-202407QR115).
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgments
Not applicable.
Funding
This study was funded by the Natural Science Foundation of Jiangxi Province (grant no. 20212ACB206011) and the National Natural Science Foundation of China (grant nos. 82460057, 81860082 and 82260059).
References
Frey N and Olson EN: Cardiac hypertrophy: The good, the bad, and the ugly. Annu Rev Physiol. 65:45–79. 2003. View Article : Google Scholar : PubMed/NCBI | |
Xu M, Wan CX, Huang SH, Wang HB, Fan D, Wu HM, Wu QQ, Ma ZG, Deng W and Tang QZ: Oridonin protects against cardiac hypertrophy by promoting P21-related autophagy. Cell Death Dis. 10:4032019. View Article : Google Scholar : PubMed/NCBI | |
Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, et al: The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med. 13:619–624. 2007. View Article : Google Scholar : PubMed/NCBI | |
Ritterhoff J and Tian R: Metabolic mechanisms in physiological and pathological cardiac hypertrophy: New paradigms and challenges. Nat Rev Cardiol. 20:812–829. 2023. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Maillet M, Van Berlo JH and Molkentin JD: Molecular basis of physiological heart growth: Fundamental concepts and new players. Nat Rev Mol Cell Biol. 14:38–48. 2013. View Article : Google Scholar | |
Gali-Muhtasib H, Roessner A and Schneider-Stock R: Thymoquinone: A promising anti-cancer drug from natural sources. Int J Biochem Cell Biol. 38:1249–1253. 2006. View Article : Google Scholar | |
Darakhshan S, Bidmeshki Pour A, Hosseinzadeh Colagar A and Sisakhtnezhad S: Thymoquinone and its therapeutic potentials. Pharmacol Res. 95-96:138–158. 2015. View Article : Google Scholar : PubMed/NCBI | |
Woo CC, Kumar AP, Sethi G and Tan KH: Thymoquinone: Potential cure for inflammatory disorders and cancer. Biochem Pharmacol. 83:443–451. 2012. View Article : Google Scholar | |
Mialet-Perez J and Vindis C: Autophagy in health and disease: Focus on the cardiovascular system. Essays Biochem. 61:721–732. 2017. View Article : Google Scholar : PubMed/NCBI | |
Xue R, Zeng J, Chen Y, Chen C, Tan W, Zhao J, Dong B, Sun Y, Dong Y and Liu C: Sestrin 1 ameliorates cardiac hypertrophy via autophagy activation. J Cell Mol Med. 21:1193–1205. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yang K, Long Q, Saja K, Huang F, Pogwizd SM, Zhou L, Yoshida M and Yang Q: Knockout of the ATPase inhibitory factor 1 protects the heart from pressure overload-induced cardiac hypertrophy. Sci Rep. 7:105012017. View Article : Google Scholar : PubMed/NCBI | |
Kelly DP: PPARs of the heart: Three is a crowd. Circ Res. 92:482–484. 2003. View Article : Google Scholar : PubMed/NCBI | |
Majdalawieh A and Ro HS: PPARgamma1 and LXRalpha face a new regulator of macrophage cholesterol homeostasis and inflammatory responsiveness, AEBP1. Nucl Recept Signal. 8:e0042010. View Article : Google Scholar : PubMed/NCBI | |
Kung J and Henry RR: Thiazolidinedione safety. Expert Opin Drug Saf. 11:565–579. 2012. View Article : Google Scholar : PubMed/NCBI | |
Peng Y, Wang L, Zhang Z, He X, Fan Q, Cheng X, Qiao Y, Huang H, Lai S, Wan Q, et al: Puerarin activates adaptive autophagy and protects the myocardium against doxorubicin-induced cardiotoxicity via the 14-3-3γ/PKCε pathway. Biomed Pharmacother. 153:1134032022. View Article : Google Scholar | |
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 | |
He H, Wang L, Qiao Y, Yang B, Yin D and He M: Epigallocatechin-3-gallate pretreatment alleviates doxorubicin-induced ferroptosis and cardiotoxicity by upregulating AMPKα2 and activating adaptive autophagy. Redox Biol. 48:1021852021. View Article : Google Scholar | |
Lu Q, Hu S, Guo P, Zhu X, Ren Z, Wu Q and Wang X: PPAR-γ with its anti-fibrotic action could serve as an effective therapeutic target in T-2 toxin-induced cardiac fibrosis of rats. Food Chem Toxicol. 152:1121832021. View Article : Google Scholar | |
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 | |
Rockman HA, Ross RS, Harris AN, Knowlton KU, Steinhelper ME, Field LJ, Ross J Jr and Chien KR: Segregation of atrial-specific and inducible expression of an atrial natriuretic factor transgene in an in vivo murine model of cardiac hypertrophy. Proc Natl Acad Sci USA. 88:8277–8281. 1991. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Zhuo C, Zu A, Yuan S, Zhang H, Zhao J and Zheng L: Thymoquinone ameliorates pressure overload-induced cardiac hypertrophy by activating the AMPK signalling pathway. J Cell Mol Med. 26:855–867. 2022. View Article : Google Scholar | |
Ji YX, Zhang P, Zhang XJ, Zhao YC, Deng KQ, Jiang X, Wang PX, Huang Z and Li H: The ubiquitin E3 ligase TRAF6 exacerbates pathological cardiac hypertrophy via TAK1-dependent signalling. Nat Commun. 7:112672016. View Article : Google Scholar : PubMed/NCBI | |
Li HH, Kedar V, Zhang C, McDonough H, Arya R, Wang DZ and Patterson C: Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex. J Clin Invest. 114:1058–1071. 2004. View Article : Google Scholar : PubMed/NCBI | |
Xie X, Bi HL, Lai S, Zhang YL, Li N, Cao HJ, Han L, Wang HX and Li HH: The immunoproteasome catalytic β5i subunit regulates cardiac hypertrophy by targeting the autophagy protein ATG5 for degradation. Sci Adv. 5:eaau04952019. View Article : Google Scholar | |
Gyöngyösi M, Winkler J, Ramos I, Do QT, Firat H, McDonald K, González A, Thum T, Díez J, Jaisser F, et al: Myocardial fibrosis: Biomedical research from bench to bedside. Eur J Heart Fail. 19:177–191. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bacmeister L, Schwarzl M, Warnke S, Stoffers B, Blankenberg S, Westermann D and Lindner D: Inflammation and fibrosis in murine models of heart failure. Basic Res Cardiol. 114:192019. View Article : Google Scholar : PubMed/NCBI | |
Doroszko A, Dobrowolski P, Radziwon-Balicka A and Skomro R: New insights into the role of oxidative stress in onset of cardiovascular disease. Oxid Med Cell Longev. 2018:95638312018. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Song Y, Liu L, Hou N, An X, Zhan D, Li Y, Zhou L, Li P, Yu L, et al: miR-199a impairs autophagy and induces cardiac hypertrophy through mTOR activation. Cell Death Differ. 24:1205–1213. 2017. View Article : Google Scholar : | |
Simonson B, Subramanya V, Chan MC, Zhang A, Franchino H, Ottaviano F, Mishra MK, Knight AC, Hunt D, Ghiran I, et al: DDiT4L promotes autophagy and inhibits pathological cardiac hypertrophy in response to stress. Sci Signal. 10:eaaf59672017. View Article : Google Scholar : PubMed/NCBI | |
Obsil T and Obsilova V: Structural basis of 14-3-3 protein functions. Semin Cell Dev Biol. 22:663–672. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wu W, Yang B, Qiao Y, Zhou Q, He H and He M: Kaempferol protects mitochondria and alleviates damages against endotheliotoxicity induced by doxorubicin. Biomed Pharmacother. 126:1100402020. View Article : Google Scholar : PubMed/NCBI | |
Diebold I, Hennigs JK, Miyagawa K, Li CG, Nickel NP, Kaschwich M, Cao A, Wang L, Reddy S, Chen PI, et al: BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab. 21:596–608. 2015. View Article : Google Scholar : PubMed/NCBI | |
Caglayan E, Stauber B, Collins AR, Lyon CJ, Yin F, Liu J, Rosenkranz S, Erdmann E, Peterson LE, Ross RS, et al: Differential roles of cardiomyocyte and macrophage peroxisome proliferator-activated receptor gamma in cardiac fibrosis. Diabetes. 57:2470–2479. 2008. View Article : Google Scholar : PubMed/NCBI | |
Duan SZ, Ivashchenko CY, Russell MW, Milstone DS and Mortensen RM: Cardiomyocyte-specific knockout and agonist of peroxisome proliferator-activated receptor-gamma both induce cardiac hypertrophy in mice. Circ Res. 97:372–379. 2005. View Article : Google Scholar : PubMed/NCBI | |
Li T, Guo R, Zong Q and Ling G: Application of molecular docking in elaborating molecular mechanisms and interactions of supramolecular cyclodextrin. Carbohydr Polym. 276:1186442022. View Article : Google Scholar | |
Oka T, Akazawa H, Naito AT and Komuro I: Angiogenesis and cardiac hypertrophy: Maintenance of cardiac function and causative roles in heart failure. Circ Res. 114:565–571. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zhang C, Liu J, Pan H, Yang X and Bian K: Mitochondrial dysfunction induced by excessive ROS/RNS-metabolic cardiovascular disease and traditional Chinese medicines intervention. Zhongguo Zhong Yao Za Zhi. 36:2423–2428. 2011.In Chinese. PubMed/NCBI | |
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 : | |
Gao M, Hu F, Hu M, Hu Y, Shi H, Zhao GJ, Jian C, Ji YX, Zhang XJ, She ZG, et al: Sophoricoside ameliorates cardiac hypertrophy by activating AMPK/mTORC1-mediated autophagy. Biosci Rep. 40:BSR202006612020. View Article : Google Scholar : PubMed/NCBI | |
Li MH, Zhang YJ, Yu YH, Yang SH, Iqbal J, Mi QY, Li B, Wang ZM, Mao WX, Xie HG and Chen SL: Berberine improves pressure overload-induced cardiac hypertrophy and dysfunction through enhanced autophagy. Eur J Pharmacol. 728:67–76. 2014. View Article : Google Scholar : PubMed/NCBI | |
Togliatto G, Lombardo G and Brizzi MF: The future challenge of reactive oxygen species (ROS) in hypertension: From bench to bed side. Int J Mol Sci. 18:19882017. View Article : Google Scholar : PubMed/NCBI | |
Dhalla AK, Hill MF and Singal PK: Role of oxidative stress in transition of hypertrophy to heart failure. J Am Coll Cardiol. 28:506–514. 1996. View Article : Google Scholar : PubMed/NCBI | |
Qin F, Lennon-Edwards S, Lancel S, Biolo A, Siwik DA, Pimentel DR, Dorn GW, Kang YJ and Colucci WS: Cardiac-specific overexpression of catalase identifies hydrogen peroxide-dependent and -independent phases of myocardial remodeling and prevents the progression to overt heart failure in G(alpha)q-overexpressing transgenic mice. Circ Heart Fail. 3:306–313. 2010. View Article : Google Scholar | |
Liu C, Wu QQ, Cai ZL, Xie SY, Duan MX, Xie QW, Yuan Y, Deng W and Tang QZ: Zingerone attenuates aortic banding-induced cardiac remodelling via activating the eNOS/Nrf2 pathway. J Cell Mol Med. 23:6466–6478. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang Z and Klionsky DJ: Eaten alive: A history of macroautophagy. Nat Cell Biol. 12:814–822. 2010. View Article : Google Scholar : PubMed/NCBI | |
Hansen M, Rubinsztein DC and Walker DW: Autophagy as a promoter of longevity: Insights from model organisms. Nat Rev Mol Cell Biol. 19:579–593. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sun M, Ouzounian M, de Couto G, Chen M, Yan R, Fukuoka M, Li G, Moon M, Liu Y, Gramolini A, et al: Cathepsin-L ameliorates cardiac hypertrophy through activation of the autophagy-lysosomal dependent protein processing pathways. J Am Heart Assoc. 2:e0001912013. View Article : Google Scholar : PubMed/NCBI | |
Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y and Yoshimori T: LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19:5720–5728. 2000. View Article : Google Scholar : PubMed/NCBI | |
Drosatos K, Khan RS, Trent CM, Jiang H, Son NH, Blaner WS, Homma S, Schulze PC and Goldberg IJ: Peroxisome proliferator-activated receptor-γ activation prevents sepsis-related cardiac dysfunction and mortality in mice. Circ Heart Fail. 6:550–562. 2013. View Article : Google Scholar : PubMed/NCBI | |
Desvergne B and Wahli W: Peroxisome proliferator-activated receptors: Nuclear control of metabolism. Endocr Rev. 20:649–688. 1999.PubMed/NCBI | |
Herrmann JE, Heale J, Bieraugel M, Ramos M, Fisher RL and Vickers AE: Isoproterenol effects evaluated in heart slices of human and rat in comparison to rat heart in vivo. Toxicol Appl Pharmacol. 274:302–312. 2014. View Article : Google Scholar | |
Yang J, Liu Y, Fan X, Li Z and Cheng Y: A pathway and network review on beta-adrenoceptor signaling and beta blockers in cardiac remodeling. Heart Fail Rev. 19:799–814. 2014. View Article : Google Scholar | |
Yamaguchi O: Autophagy in the heart. Circ J. 83:697–704. 2019. View Article : Google Scholar : PubMed/NCBI | |
He H, Luo Y, Qiao Y, Zhang Z, Yin D, Yao J, You J and He M: Curcumin attenuates doxorubicin-induced cardiotoxicity via suppressing oxidative stress and preventing mitochondrial dysfunction mediated by 14-3-3γ. Food Funct. 9:4404–4418. 2018. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Peng X, Luo Y, You J, Yin D, Xu Q, He H and He M: Quercetin protects cardiomyocytes against doxorubicin-induced toxicity by suppressing oxidative stress and improving mitochondrial function via 14-3-3γ. Toxicol Mech Methods. 29:344–354. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Tan H, Wan J, Zeng Y, Wang J, Wang H and Lu X: PPAR-γ signaling in nonalcoholic fatty liver disease: Pathogenesis and therapeutic targets. Pharmacol Ther. 245:1083912023. View Article : Google Scholar | |
Botta M, Audano M, Sahebkar A, Sirtori CR, Mitro N and Ruscica M: PPAR agonists and metabolic syndrome: An established role? Int J Mol Sci. 19:11972018. View Article : Google Scholar : PubMed/NCBI | |
NiX X, Li XY, Wang Q and Hua J: Regulation of peroxisome proliferator-activated receptor-gamma activity affects the hepatic stellate cell activation and the progression of NASH via TGF-β1/Smad signaling pathway. J Physiol Biochem. 77:35–45. 2021. View Article : Google Scholar | |
Deng W, Meng Z, Sun A and Yang Z: Pioglitazone suppresses inflammation and fibrosis in nonalcoholic fatty liver disease by down-regulating PDGF and TIMP-2: Evidence from in vitro study. Cancer Biomark. 20:411–415. 2017. View Article : Google Scholar |