Sodium–glucose cotransporter 2 inhibitors (SGLT2i) have favorable cardiovascular outcomes in patients with diabetes. However, whether SGLT2i can improve obesity-related cardiac dysfunction is unknown. Sestrin2 is a novel stress-inducible protein that regulates AMPK–mammalian target of rapamycin (mTOR) and suppresses oxidative damage. The aim of this study was to determine whether empagliflozin (EMPA) improves obesity-related cardiac dysfunction via regulating Sestrin2-mediated pathways in diet-induced obesity. C57BL/6J mice and Sestrin2 knockout mice were fed a high-fat diet (HFD) for 12 weeks and then treated with or without EMPA (10 mg/kg) for 8 weeks. Treating HFD-fed C57BL/6J mice with EMPA reduced body weight and whole-body fat and improved metabolic disorders. Furthermore, EMPA improved myocardial hypertrophy/fibrosis and cardiac function and reduced cardiac fat accumulation and mitochondrial injury. Additionally, EMPA significantly augmented Sestrin2 levels and increased AMPK and endothelial nitric oxide synthase phosphorylation, but inhibited Akt and mTOR phosphorylation. These beneficial effects were partially attenuated in HFD-fed Sestrin2 knockout mice. Intriguingly, EMPA treatment enhanced the Nrf2/HO-1–mediated oxidative stress response, suggesting antioxidant and anti-inflammatory activity. Thus, EMPA improved obesity-related cardiac dysfunction via regulating Sestrin2-mediated AMPK-mTOR signaling and maintaining redox homeostasis. These findings provide a novel mechanism for the cardiovascular protection of SGLT2i in obesity.

Obesity is a growing health problem worldwide that is strongly linked to an increased risk of cardiovascular disease, cardiac dysfunction, and chronic heart failure (1,2). Thus, developing new strategies to prevent and treat obesity-related cardiac dysfunction is highly desirable. Numerous mechanisms such as insulin resistance, ectopic fat accumulation, lipotoxicity, and imbalanced adipocyte secretion have been shown to play a role in these pathophysiological processes. Moreover, these alterations can lead to progression of the pathological cardiac remodeling that manifests in cardiac hypertrophy and fibrosis (3). The process of cardiac hypertrophy is an adaptive response to multiple pathological stresses and is accompanied by excessive oxidative stress and chronic activation of mammalian target of rapamycin (mTOR) (4,5). AMPK is a major cellular sensor of energy availability. In obesity, AMPK phosphorylation in the heart is often downregulated due to overnutrition (6). Acting as a cardioprotective molecule, AMPK balances oxidative stress and inhibits mTOR signaling by directly phosphorylating the TSC1/2 complex (7,8). This AMPK-mediated mTOR regulation has been shown to have pleiotropic cardioprotective effects that restore cardiac dysfunction, including improving the energy supply and regulating other physiological processes (7).

Sestrin2 (Sesn2) is a novel stress-inducible protein that lacks kinase activity but has been shown to have two important functions: inhibition of mTOR by its COOH-terminal domain and suppression of oxidative stress through its N-terminal domain (9). Both functions are vital for preventing and treating cardiovascular disease, including ischemia-reperfusion injury and cardiomyocytes hypertrophy (10). For example, Sesn2 has been shown to attenuate phenylephrine-induced cardiac hypertrophy in neonatal rats (10). Our recent findings revealed that Sesn2 prevents age-related intolerance to myocardial infarction by activating AMPK (11). Sesn2 also promotes phosphorylation of AMPK through its upstream kinase liver kinase B1 (LKB1)–dependent way and mediates genotoxic stress-induced AMPK activation and mTOR inhibition (12,13). However, whether Sesn2 is also involved in obesity-related cardiac dysfunction remains unknown.

Sodium–glucose cotransporter 2 inhibitors (SGLT2i) are a novel class of hypoglycemic agents that selectively inhibit sodium and glucose reabsorption from the proximal renal tubule, increase the excretion of urine glucose, and thus decrease blood glucose levels (14). Several large randomized, controlled cardiovascular outcomes trials (EMPA-REG OUTCOME trial, CANVAS Program, and DECLARE‐TIMI 58) and a meta-analysis have demonstrated unprecedented favorable cardiovascular outcomes in patients with diabetes treated with empagliflozin (EMPA), canagliflozin, or dapagliflozin (1518). There are several mechanisms to explain these beneficial effects of SGLT2i on cardiovascular outcomes, such as weight loss, and improved glucose metabolism, blood pressure (BP), and sodium overload (19). However, in subjects without obvious diabetes and hypertension, SGLT2i also seemed to show cardiac protection (20). Additionally, the direct impact of SGLT2i on the heart remains controversial, as only SGLT1, and not SGLT2, is expressed in the myocardium (21). Hence, understanding the underlying pathophysiological mechanisms responsible for these positive effects requires further investigation.

While the consensus is that SGLT2i improves cardiac function in diabetes, no study has focused on whether SGLT2i could directly improve obesity-related cardiac dysfunction. In this study, we used the high-fat diet (HFD)–induced obesity model and Sesn2 knockout (KO) mice to test the hypothesis that EMPA improves obesity-related cardiac dysfunction via regulating Sesn2-mediated AMPK-mTOR signaling and the cellular antioxidant system.

Experimental Animals

Male 6-week-old wild-type (WT) C57BL/6J mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and randomly assigned to the normal control (NC), NC-EMPA (NC-E), HFD, and HFD-EMPA (HFD-E) groups. The NC and NC-E groups were fed a normal chow diet (17% energy from fat, 29% from protein, and 54% from carbohydrate; 300 kcal/100 g) (catalog number 8640; Envigo, Somerset, NJ), while the other two groups were fed an HFD (45% energy from fat, 19% from protein, and 36% from carbohydrate; 460 kcal/100 g) (catalog number TD.06415; Envigo) to induce obesity. After 12 weeks, the NC-E and HFD-E groups received EMPA (10 mg/kg) (Sigma-Aldrich, St. Louis, MO) by oral gavage once daily for another 8 weeks, whereas the other two control groups received saline. Sesn2 KO mice (C57BL/6J background) were generated as previously described (12,13). The KO mice and WT littermates were randomly assigned to normal diet and HFD with EMPA or saline treatment. All groups were fed an HFD for 12 weeks and then treated with EMPA (10 mg/kg) or without EMPA for another 8 weeks. All mice were caged in a temperature-controlled environment on a 12-h light/dark cycle. Body weight (BW) and food intake were measured weekly. After 8 weeks of EMPA treatment, body composition and metabolic profiles, including glucose homeostasis, plasma insulin, lipids, and adipokine, were performed. These are described in the Supplementary Materials and Methods. The study protocol was approved by the Institutional Animal Care and Use Committees of the University of Mississippi Medical Center, Affiliated Hospital of Weifang Medical University, and University of South Florida.

Isolation of Cardiomyocytes

Cardiomyocytes were isolated enzymatically from NC and HFD mice as previously described by our group (22). Briefly, after mice were anesthetized, hearts were excised and fixed onto a cardiomyocyte’s perfusion apparatus (Radnoti Ltd., Monrovia, CA). Hearts were first perfused with Krebs-Henseleit buffer oxygenated with 95% O2/5% CO2 and then digested with a solution containing liberase blendzyme. After successful digestion, cardiomyocytes were incubated with EMPA (1 μmol/L) at 37°C for 2 h, and then total protein was extracted for Western blot analysis.

BP and Echocardiographic Evaluation

BP was measured by a noninvasive tail-cuff device (BP2000; VisiTech International, Sunderland, U.K.) at the end of the study period. Cardiac function was determined by high-frequency ultrasonography using a Vevo 3100 imaging system (VisualSonics Inc., Toronto, Ontario, Canada) as previously described (23). The following main variables were assessed: left ventricular (LV) ejection fraction (EF), LV fractional shortening (FS), LV mass, the ratio of early diastolic to late diastolic mitral inflow velocities (E/A), isovolumic relaxation time (IVRT), and myocardial performance index (MPI).

Histopathological Analysis

To assess morphological changes, LV tissues were fixed immediately in 10% neutral buffered formalin and embedded in paraffin after anesthesia. Then, 5-μm sections were cut from the paraffin blocks and stained with hematoxylin and eosin for histopathological examination. To evaluate perivascular/interstitial fibrosis, sections were stained with both Masson trichrome and picrosirius red (Sigma-Aldrich). To evaluate myocardial inflammation, sections were stained with CD68 antibody. The heart sections were also used to evaluate lipid accumulation by Oil Red O staining. Micrographs were acquired using light microscopy (Nikon, Tokyo, Japan).

Transmission Electron Microscopy

The cardiac ultrastructure was observed using transmission electron microscopy. Heart tissues were prepared as previously described by our group (23). Digital images were taken using an XR611 digital camera (Advanced Microscopy Techniques, Corp., Woburn, MA).

Mitochondrial Reactive Oxygen Species Measurement

Mitochondrial reactive oxygen species (ROS) production was evaluated by MitoSOX Red (Invitrogen, Carlsbad, CA) as previously described (24). Briefly, fresh-frozen sections of LV were washed with PBS and then incubated in PBS containing 1 μmol/L MitoSOX Red mitochondrial superoxide indicator (Invitrogen) for 10 min at 37°C, protected from light. Images were taken using a fluorescence microscope (excitation: 510 nm; and emission: 580 nm).

Western Blot Analysis

Equal amounts of total protein from LV myocardial tissues were electrophoresed on 7–15% SDS-PAGE gels and transferred to polyvinylidene difluoride membranes. The membranes were incubated with primary antibodies (Supplementary Materials and Methods) overnight at 4°C, washed, and then incubated with horseradish peroxidase–conjugated secondary antibodies for 1 h. Immunoreactive bands were detected with SuperSignal West Femto (Thermo Fisher Scientific, Waltham, MA) in a Western blotting detection system (Bio-Rad Laboratories, Hercules, CA). Results are expressed as density values normalized to β-tubulin or GAPDH.

Real-time Quantitative PCR Analysis

Total RNA from LV myocardial tissues was extracted using TRIzol (Invitrogen) and then synthesized into cDNA using the TaqMan High-Capacity RNA-to-cDNA Kit (#4387406; Applied Biosystems, Foster City, CA). Quantitative PCR was performed using iTaq Universal SYBR Green Supermix (#1725124; Bio-Rad Laboratories). Primer sequences are listed in the Supplementary Materials and Methods.

Statistical Analysis

All data are expressed as the mean ± SD. Differences among more than two groups were compared by one- or two-way ANOVA with multiple-comparisons and post hoc tests for selected comparisons. P < 0.05 was accepted as statistically significant. Statistical analyses were performed with SPSS 22.0 (IBM Corporation, Armonk, NY) or Prism 8 (GraphPad Software, San Diego, CA).

Data and Resource Availability

The data sets analyzed during the current study are available from the corresponding author on reasonable request.

EMPA Reduced BW Gain and Fat Mass and Improved Glucose Homeostasis and Adipokine Profiles Without Changing BP or Lipid Levels in HFD Mice

After 12 weeks, mice fed the HFD had significantly increased BW (32.6%) and fat mass (2.92-fold; P < 0.05). EMPA treatment of HFD mice for 8 weeks prevented further BW and fat mass gain (P < 0.05) (Fig. 1A–D). However, EMPA had no effect on lean mass (P > 0.05) (Supplementary Fig. 1A). Although HFD mice ingested fewer grams of food (2.8 vs. 4.3 g; P < 0.05) (Supplementary Fig. 1D), there was no significant difference in kilocalories of food intake between control and HFD mice (12.87 vs. 13.07 kcal; P > 0.05) (Supplementary Fig. 1E). EMPA had no effect on food intake (both by grams and kilocalories; P > 0.05) (Supplementary Fig. 1B–E). As expected, compared with control mice, HFD mice showed impaired glucose/insulin tolerance (higher glucose area under the curve), both of which were improved by EMPA treatment (P < 0.05) (Fig. 1E and F and Supplementary Fig. 1F and G). There was no significant difference in fasting blood glucose among the groups (P > 0.05) (Fig. 1G). Fasting insulin levels were higher in HFD mice compared with control mice (P < 0.05) (Fig. 1H). EMPA significantly decreased insulin levels by 58.27% (P < 0.05) but had no beneficial effects on free fatty acid (FFA), triglyceride, or cholesterol levels (P > 0.05) (Fig. 1I and J). To determine whether EMPA could improve adipokine profiles in HFD mice, we measured circulating adiponectin and leptin levels. EMPA treatment enhanced circulating adiponectin by 32.7% and reduced leptin levels by 75.36% in HFD mice (P < 0.05) (Fig. 1K and L). No significant differences in either systolic or diastolic BP were found among the groups (P > 0.05) (Supplementary Fig. 1H).

Figure 1

Effects of EMPA treatment on BW, body composition, and biochemical parameters. A: Morphology and BW of mice. B: Weekly BWs (n = 6–8/group). Six-week-old male mice were randomized to either the control diet or HFD for 12 weeks and then treated with or without EMPA for another 8 weeks (end point at 20 weeks). *P < 0.05 vs. NC; #P < 0.05 vs. HFD. C: Body fat mass of the mice (n = 6–8/group). D: Body fat/BW (n = 6–8/group). E: Oral glucose tolerance test (n = 5/group). F: Insulin tolerance test (n = 5/group). G: Fasting blood glucose (FBG) levels (n = 6/group). H: Fasting plasma insulin levels (n = 6/group). I: Fasting plasma FFA levels (n = 6/group). J: Plasma lipid levels (n = 6/group). K: Plasma adiponectin levels (n = 6/group). L: Plasma leptin levels (n = 6/group). Data are means ± SD. *P < 0.05. w, weeks.

Figure 1

Effects of EMPA treatment on BW, body composition, and biochemical parameters. A: Morphology and BW of mice. B: Weekly BWs (n = 6–8/group). Six-week-old male mice were randomized to either the control diet or HFD for 12 weeks and then treated with or without EMPA for another 8 weeks (end point at 20 weeks). *P < 0.05 vs. NC; #P < 0.05 vs. HFD. C: Body fat mass of the mice (n = 6–8/group). D: Body fat/BW (n = 6–8/group). E: Oral glucose tolerance test (n = 5/group). F: Insulin tolerance test (n = 5/group). G: Fasting blood glucose (FBG) levels (n = 6/group). H: Fasting plasma insulin levels (n = 6/group). I: Fasting plasma FFA levels (n = 6/group). J: Plasma lipid levels (n = 6/group). K: Plasma adiponectin levels (n = 6/group). L: Plasma leptin levels (n = 6/group). Data are means ± SD. *P < 0.05. w, weeks.

Close modal

EMPA Treatment Attenuated LV Hypertrophy and Improved Cardiac Function in HFD Mice

There were significant increases in LV and heart mass in HFD mice compared with control mice (increased by 40.18% and 39.7%, respectively; P < 0.05). EMPA treatment significantly reduced LV/heart mass (reduced by 20.66% and 23.61%; P < 0.05) (Fig. 2A and B). Echocardiographic measurements of heart rates, LV volume, interventricular septal dimensions, LV internal dimensions, and LV posterior wall thickness were similar among the four groups (P > 0.05) (Supplementary Fig. 2). No significant differences were observed among groups regarding systolic function parameters (LVEF and LVFS, P > 0.05) (Fig. 2C and D). However, compared with NC mice, HFD mice exhibited impaired diastolic function, including E/A, IVRT, and MPI (P < 0.05) (Fig. 2E–H). EMPA treatment significantly improved diastolic function, which manifested as enhanced E/A and reduced changes in IVRT and MPI (P < 0.05) (Fig. 2E–H). We did not observe differences between the NC and NC-E groups (P > 0.05).

Figure 2

Effects of EMPA treatment on cardiac function. A: Heart morphology. B: LV/heart mass. C: Systolic function of the heart. D: EF and FS. E: Diastolic function of the heart. F: E/A. G: IVRT. H: MPI. n = 6–8/group. *P < 0.05.

Figure 2

Effects of EMPA treatment on cardiac function. A: Heart morphology. B: LV/heart mass. C: Systolic function of the heart. D: EF and FS. E: Diastolic function of the heart. F: E/A. G: IVRT. H: MPI. n = 6–8/group. *P < 0.05.

Close modal

EMPA Treatment Reduced Cardiac Perivascular/Interstitial Fibrosis and Lipid Accumulation in HFD Mice

In accordance with the observed morphometric changes, HFD mice exhibited obvious cardiac perivascular/interstitial fibrosis and increased fibrosis-related genes expression (FSP-1 and Postn) (P < 0.05) (Fig. 3A–C, E, and F and Supplementary Fig. 3). Additionally, Oil Red O staining revealed increased relative lipid contents in myocardial tissue from HFD mice (Fig. 3D). Interestingly, all of these abnormalities were ameliorated by EMPA treatment (Fig. 3). No significant differences were found between NC and NC-E mice (P > 0.05). To explore how EMPA markedly decreased lipid content, we detected acetyl CoA carboxylase (ACC) phosphorylation and found that EMPA could increase ACC phosphorylation (P < 0.05) (Supplementary Fig. 4).

Figure 3

Effects of EMPA treatment on fibrosis and lipid accumulation. A: Representative hematoxylin and eosin staining. B: Masson trichrome staining. C: Picrosirius red staining. D: Oil Red O staining. E: Collagen volume fraction (CVF). F: Picrosirius red staining, fibrotic area. Original magnification ×400; n = 4/group. *P < 0.05.

Figure 3

Effects of EMPA treatment on fibrosis and lipid accumulation. A: Representative hematoxylin and eosin staining. B: Masson trichrome staining. C: Picrosirius red staining. D: Oil Red O staining. E: Collagen volume fraction (CVF). F: Picrosirius red staining, fibrotic area. Original magnification ×400; n = 4/group. *P < 0.05.

Close modal

EMPA Treatment Attenuated Myocardial Mitochondrial Injury in HFD Mice

Compared with NC mice, HFD mice exhibited an altered mitochondrial cristae architecture, including swollen, distorted mitochondria, fission, and inner mitochondrial membrane disruption (Fig. 4A). However, EMPA treatment reduced myocardial mitochondria injury (Fig. 4A). To further determine whether EMPA could reduce mitochondrial injury, we measured mtDNA contents by RT-PCR and evaluated mitochondrial ROS through the MitoSOX Red probe, which specifically targets mitochondria. EMPA treatment of HFD mice significantly reduced mitochondrial ROS (P < 0.05) (Fig. 4B and C) and enhanced mtDNA contents (P < 0.05) (Fig. 4D).

Figure 4

Effects of EMPA treatment on mitochondrial alterations. A: Mitochondrial morphology, as examined by transmission electronic microscopy (original magnification ×3,000 and ×6,000). B: Representative images of mitochondrial superoxide production (MitoSOX, red fluorescence) in cardiomyocytes. C: Quantitative analysis of MitoSOX production. n = 5 from four mice/each group. D: Relative mtDNA content. n = 4–7/group. *P < 0.05.

Figure 4

Effects of EMPA treatment on mitochondrial alterations. A: Mitochondrial morphology, as examined by transmission electronic microscopy (original magnification ×3,000 and ×6,000). B: Representative images of mitochondrial superoxide production (MitoSOX, red fluorescence) in cardiomyocytes. C: Quantitative analysis of MitoSOX production. n = 5 from four mice/each group. D: Relative mtDNA content. n = 4–7/group. *P < 0.05.

Close modal

EMPA Treatment Modulated Cardiac Sesn2-Mediated AMPK-mTOR Signaling in HFD Mice

As the AMPK-mTOR signaling pathway is essential for cardiac hypertrophy (25), we investigated whether EMPA treatment can ameliorate myocardial abnormalities via the AMPK-mTOR signaling cascade. The results demonstrated that EMPA treatment significantly enhanced AMPK phosphorylation and reduced phosphorylation of mTOR and phospho-S6 (P < 0.05) (Fig. 5A and B). We have previously reported that Sesn2 serves as a scaffold protein to modulate AMPK-mTOR signaling in the heart (11,26); thus, we next determined whether these beneficial effects of EMPA treatment involved Sesn2-mediated regulation. Intriguingly, we found that EMPA treatment augmented both Sesn2 protein and Sesn2 mRNA levels (P < 0.05) (Fig. 5A and B). To further explore whether EMPA could directly activate cardiac Sesn2-AMPK, we incubated isolated cardiomyocytes from normal diet and HFD mice with or without EMPA for 2 h. Interestingly, we found that EMPA treatment not only augmented Sesn2 protein but also triggered AMPK phosphorylation, indicating that EMPA could activate cardiomyocytes Sesn2-AMPK independent of beneficial whole-body metabolic changes (P < 0.05) (Fig. 5C).

Figure 5

Effects of EMPA treatment on the Sesn2-AMPK-mTOR and PI3K-Akt-eNOS signaling pathways. A and B: Western blot analysis of Sesn2, AMPK, mTOR, and p-S6 and RT-PCR analysis of Sesn2 mRNA from myocardial tissue. n = 3–6 mice/group. C: Western blot analysis of Sesn2 and AMPK in isolated cardiomyocytes treated with EMPA for 2 h. n = 6 mice/group. D and E: Western blot analysis of PI3K, Akt, and eNOS from myocardial tissue. n = 3–6 mice/group. *P < 0.05. p-, phosphorylated.

Figure 5

Effects of EMPA treatment on the Sesn2-AMPK-mTOR and PI3K-Akt-eNOS signaling pathways. A and B: Western blot analysis of Sesn2, AMPK, mTOR, and p-S6 and RT-PCR analysis of Sesn2 mRNA from myocardial tissue. n = 3–6 mice/group. C: Western blot analysis of Sesn2 and AMPK in isolated cardiomyocytes treated with EMPA for 2 h. n = 6 mice/group. D and E: Western blot analysis of PI3K, Akt, and eNOS from myocardial tissue. n = 3–6 mice/group. *P < 0.05. p-, phosphorylated.

Close modal

EMPA Treatment Inhibited Myocardial Phosphatidylinositol 3-Kinase–Akt but Activated Endothelial Nitric Oxide Synthase Pathway in HFD Mice

In addition to the important role of mTOR signaling in cardiac hypertrophy, phosphatidylinositol 3-kinase (PI3K)–Akt signaling is also critical for cardiac hypertrophy (25); thus, we investigated whether EMPA treatment could modulate the PI3K-Akt pathway. The results demonstrated that EMPA treatment inhibited phosphorylation of both PI3K and Akt triggered by HFD (P < 0.05) (Fig. 5D and E). Additionally, endothelial nitric oxide synthase (eNOS) phosphorylation was attenuated in HFD mice compared with NC mice (P < 0.05) (Fig. 5D and E). EMPA treatment increased phosphorylation of eNOS at Ser1177 with HFD, indicating the endothelial-protective effects of EMPA (P < 0.05) (Fig. 5D and E).

EMPA Treatment Reduced Myocardial Inflammation and Maintained Redox Homeostasis in HFD Mice

There is evidence that inflammation and oxidative stress are involved in HFD-induced cardiac dysfunction (27); thus, we examined the effects of EMPA treatment on HFD-induced cardiac functions. The results demonstrated that EMPA treatment significantly reduced myocardial nuclear factor-κB (NF-κB) p65 activity and MCP-1 protein levels caused by HFD (P < 0.05) (Fig. 6A). Moreover, EMPA treatment reduced macrophage marker (CD68) expression in the heart (Fig. 6B). Interestingly, EMPA treatment significantly upregulated the expression of oxidative signaling proteins and downstream antioxidant genes, including Nrf2/HO-1, catalase, and GCLM (P < 0.05) (Fig. 6A and C). The isolated cardiomyocytes from HFD mice were incubated with EMPA, augmented Nrf2/HO-1, and reduced NF-κB p65 activity (P < 0.05) (Supplementary Fig. 5).

Figure 6

Effects of EMPA treatment on redox homeostasis–related protein levels. A: Western blot analysis of Nrf2, HO-1, p–NF-κB p65, and MCP-1 from myocardial tissue. n = 3 mice/group. B: Immunohistochemistry for CD68 expression. n = 4 mice/group. C: Real-time PCR for Nrf2, HO-1, catalase, and GCLM. n = 4–8 mice/group. *P < 0.05.

Figure 6

Effects of EMPA treatment on redox homeostasis–related protein levels. A: Western blot analysis of Nrf2, HO-1, p–NF-κB p65, and MCP-1 from myocardial tissue. n = 3 mice/group. B: Immunohistochemistry for CD68 expression. n = 4 mice/group. C: Real-time PCR for Nrf2, HO-1, catalase, and GCLM. n = 4–8 mice/group. *P < 0.05.

Close modal

Sesn2 Is a Stress-Inducible Protein That Is Involved in the Beneficial Effects of EMPA Treatment on Obesity-Related Cardiac Dysfunction

We have previously identified Sesn2 as a critical scaffold protein that mediates cardiac AMPK signaling in response to stress conditions (13). To determine whether the beneficial effects of EMPA treatment on cardiac dysfunction induced by HFD are Sesn2 dependent, we compared the response of both WT littermates and Sesn2 KO mice to HFD with or without EMPA treatment. The results demonstrated that Sesn2 KO mice showed more obesity and metabolic disorders when exposed to HFD compared with WT littermates, as well as greater BW gain, fat mass, and glucose tolerance (P < 0.05) (Fig. 7A–F). Compared with Sesn2 KO mice with normal chow diet, Sesn2 KO mice with HFD (KO-HFD) mice showed both impaired systolic (reduced LVEF and LVFS) and diastolic function (P < 0.05) (Fig. 7G). However, no obvious improvement in cardiac function was observed in Sesn2 KO-HFD mice with EMPA (KO-HFD-E) treatment (P > 0.05) (Fig. 7G). Moreover, Masson staining and transmission electron microscopy data demonstrated that there was no improvement in cardiac interstitial fibrosis or myocardial mitochondria injury in KO-HFD-E treatment (Fig. 7H).

Figure 7

Effects of EMPA treatment on BW, body composition, biochemical parameters, and cardiac function in Sesn2 KO-HFD mice. A: Morphology and BW of mice. B: Weekly BWs (n = 5/group). Six-week-old male mice were fed the HFD for 12 weeks and then treated with or without EMPA for another 8 weeks (end point at 20 weeks). C: Body fat mass of the mice (n = 5/group). D: Body fat/BW (n = 5/group). E: Oral glucose tolerance test (n = 4–6/group). F: Insulin tolerance test (n = 4–6/group). G: Cardiac function: EF, FS, E/A, IVRT, and MPI. n = 6 to 7/group. H: Masson trichrome staining and mitochondrial morphology, as examined by transmission electronic microscopy (original magnification ×3,000 and ×6,000). KO-HFD-E, KO-HFD and EMPA; KO-NC, Sesn2 KO mice with normal chow diet; WT-HFD-E, WT-HFD and EMPA. *P < 0.05.

Figure 7

Effects of EMPA treatment on BW, body composition, biochemical parameters, and cardiac function in Sesn2 KO-HFD mice. A: Morphology and BW of mice. B: Weekly BWs (n = 5/group). Six-week-old male mice were fed the HFD for 12 weeks and then treated with or without EMPA for another 8 weeks (end point at 20 weeks). C: Body fat mass of the mice (n = 5/group). D: Body fat/BW (n = 5/group). E: Oral glucose tolerance test (n = 4–6/group). F: Insulin tolerance test (n = 4–6/group). G: Cardiac function: EF, FS, E/A, IVRT, and MPI. n = 6 to 7/group. H: Masson trichrome staining and mitochondrial morphology, as examined by transmission electronic microscopy (original magnification ×3,000 and ×6,000). KO-HFD-E, KO-HFD and EMPA; KO-NC, Sesn2 KO mice with normal chow diet; WT-HFD-E, WT-HFD and EMPA. *P < 0.05.

Close modal

Sesn2 Mediated Cardiac AMPK-mTOR Signaling in Response to HFD-Induced Obesity

The immunoblotting results demonstrated that EMPA treatment led to the upregulation of AMPK phosphorylation and reduction of mTOR phosphorylation in WT littermates with HFD (WT-HFD) (Fig. 8A). However, these effects of EMPA treatment were not observed in Sesn2 KO-HFD mice (Fig. 8A). Additionally, compared with WT-HFD mice, EMPA treatment had no beneficial effects on HO-1 protein (Fig. 8B), Nrf2 mRNA levels (Fig. 8C), or NF-κB activation in Sesn2 KO-HFD mice (P > 0.05) (Fig. 8B). Similarly, no enhancement was observed in myocardial mtDNA in KO-HFD-E treatment (P > 0.05) (Fig. 8C).

Figure 8

Effects of EMPA treatment on the Sesn2-AMPK-mTOR pathway and redox homeostasis in Sesn2 KO-HFD mice. A: Western blot analysis of AMPK and mTOR from myocardial tissue. n = 3–6 mice/group. B: Western blot analysis of HO-1 and p65 from myocardial tissue. n = 3 mice/group. C: Real-time PCR for mtDNA and Nrf2. n = 4–6 mice/group. KO-HFD-E, KO-HFD and EMPA; KO-NC, Sesn2 KO mice with normal chow diet; WT-HFD-E, WT-HFD and EMPA. *P < 0.05. p-, phosphorylated.

Figure 8

Effects of EMPA treatment on the Sesn2-AMPK-mTOR pathway and redox homeostasis in Sesn2 KO-HFD mice. A: Western blot analysis of AMPK and mTOR from myocardial tissue. n = 3–6 mice/group. B: Western blot analysis of HO-1 and p65 from myocardial tissue. n = 3 mice/group. C: Real-time PCR for mtDNA and Nrf2. n = 4–6 mice/group. KO-HFD-E, KO-HFD and EMPA; KO-NC, Sesn2 KO mice with normal chow diet; WT-HFD-E, WT-HFD and EMPA. *P < 0.05. p-, phosphorylated.

Close modal

The experiments described above elucidated the effect of EMPA treatment on obesity-related cardiac dysfunction in obese mice. We found for the first time that EMPA treatment attenuated myocardial hypertrophy and improved cardiac function via regulating cardiac Sesn2-mediated AMPK-mTOR signaling and maintaining cardiac redox homeostasis. Further studies found that Sesn2 is a stress-inducible protein that is required for the effects of EMPA in obesity-related cardiac dysfunction. This study demonstrated that the protective role of EMPA in obesity-related cardiac dysfunction is via regulating Sesn2.

The incidence of cardiac dysfunction has risen in parallel with increasing obesity-related metabolic disorders. Obese subjects are more likely to display the phenotype of heart failure with preserved ejection fraction, without significant reduction of LVEF or LVFS (28). The worldwide obesity-related cardiac dysfunction epidemic has heightened the need to find an effective therapy that can improve cardiac dysfunction and reduce the risk of cardiovascular disease. Although some drugs such as ACE receptor and angiotensin receptor blockers as well as mineralocorticoid receptor antagonists have been used to treat cardiac dysfunction, few effective therapeutic strategies have been found to prevent or attenuate the progression of cardiac dysfunction.

Recently, SGLT2i has been shown to have beneficial effects on cardiovascular disease outcomes by improving cardiometabolic profiles. The Dapagliflozin and Prevention of Adverse-Outcomes in Heart Failure (DAPA-HF) study demonstrated the incremental efficacy and safety of SGLT2i in patients with heart failure and reduced ejection fraction without diabetes (29). Our study found that HFD mice exhibited impairments in diastolic function, including reduced E/A, prolonged IVRT, and increased MPI, without significant alterations in LVEF and LVFS. However, 8 weeks of EMPA treatment improved LV hypertrophy and diastolic function. Other studies have also demonstrated that EMPA can improve cardiac dysfunction in other models, such as leptin-deficient and nondiabetic mice (2931). In addition to improving cardiac function, EMPA also reduced cardiac fibrosis and lipid accumulation and attenuated myocardial mitochondrial injury. Our results are the first to demonstrate that EMPA can improve obesity-related cardiac structure and function in HFD mice.

To explore the potential mechanism of these cardioprotective effects of EMPA, we assessed metabolic profiles. The results demonstrated that EMPA prevented BW gain, reduced whole-body fat, and improved glucose homeostasis. We did not observe significant changes in BP or lipid levels. Thus, the cardiac function improvements of EMPA-treated HFD mice are independent of changes in BP and lipid levels. It is well known that altered adipokine secretion is a link between obesity and the increased prevalence of obesity-associated diseases. The high ratio of leptin/adiponectin appears to be associated with high risk of cardiovascular diseases in obesity (32). Our data found that EMPA treatment significantly ameliorated these disorders along with reducing leptin and enhancing adiponectin, which were also confirmed by a recent meta-analysis (33). These alterations are possibly due to the reduction of BW and whole fat mass.

Sestrins are a novel family of stress-inducible proteins that are involved in AMPK-mTOR signaling, redox, and autophagy. Our laboratory and others (13) have highlighted that Sestrins are vital for cardiac homeostasis. Sesn2 protects cardiomyocytes from ischemia-reperfusion–induced injury by initiating AMPK activation (11). However, whether Sesn2 is also involved in obesity-related cardiac function has not been studied. Our data showed that both Sesn2 protein and Sesn2 mRNA levels were lower in HFD cardiac tissue and that EMPA treatment augmented both Sesn2 protein and mRNA levels. To further explore whether these alterations were dependent on whole-body metabolic changes, we incubated the cardiomyocytes with EMPA for 2 h and found that EMPA treatment triggered Sesn2 protein levels, indicating that EMPA can induce cardiomyocytes Sesn2 upregulation independent of beneficial whole-body metabolic changes. Next, we examined whether the cardioprotective effect of EMPA on cardiac dysfunction was Sesn2 dependent. Sesn2 KO mice were fed an HFD for 12 weeks and then treated with EMPA for another 8 weeks. Although Sesn2 KO mice were more prone to developing obesity and metabolic disorders compared with WT littermates, EMPA could not attenuate these adverse alterations in the mutants. Sesn2 KO mice displayed not only diastolic dysfunction but also adverse systolic function, with reduced LVEF and LVFS. The results indicated that Sesn2 is a critical protein that mediates the beneficial effects of EMPA on obesity-related cardiac dysfunction. HFD decreases Sesn2 levels in the C57BL/6J mouse hearts and consequently decreases downstream AMPK phosphorylation that modulates mTOR activation (11,25). Although the isolated cardiomyocytes from HFD hearts did not show significant alterations in Sesn2 levels, the downstream AMPK phosphorylation was downregulated in the HFD cardiomyocytes. These observations indicate two potential underlying mechanisms of observed cardiac dysfunction in HFD mice. First, the Sesn2-AMPK signaling cascade, rather than the levels of Sesn2 protein alone, plays a role in the HFD-induced diastolic function. Second, a reduced expression of Sesn2 in noncardiomyocytes, such as endothelial cells, contributes to the observed reduction of Sesn2 in HFD hearts. A reduced Sesn2 in endothelial cells could affect cardiac diastolic function by regulating the nitric oxide pathway (34,35). In agreement, we found that EMPA treatment modulates the phosphorylation of eNOS at Ser1177 in the mouse hearts with HFD, indicating the endothelial-protective effects of EMPA. Future studies are needed to explore the difference of Sesn2 signaling response between cardiomyocytes and noncardiomyocytes in HFD heart.

mTOR is a central regulator of biogenesis that responds to the nutritional status of the cell. In obesity, activated mTOR can phosphorylate p70 S6 kinase, which is involved in myocardial proliferation and hypertrophy (36,37). AMPK inhibits protein synthesis and myocardial proliferation by inhibiting the mTOR-p70S6 kinase pathway (36). Sesn2 has two main functions: modulating AMPK-mTOR and antioxidative capability. We first detected whether the upregulation of Sesn2 by EMPA regulated the AMPK-mTOR pathway and found that EMPA treatment significantly enhanced AMPK phosphorylation and reduced phosphorylation of mTOR and p70 S6 kinase, indicating that EMPA could alleviate myocardial hypertrophy through the Sesn2-AMPK-mTOR pathway in HFD mice.

In obesity, there is increased FFA-induced myocardial lipid accumulation, which can increase the risk of heart failure in obesity by the imbalance of oxidative/antioxidant and inflammatory cell infiltration (38). Nrf2/HO-1 is the primary endogenous antioxidant defense pathway and counteracts oxidative stress-related cardiac injury, contributes to myocardia hypertrophy, and participates in protecting cardiac function (39,40). Our results showed that EMPA treatment inactivated NF-κB p65 activity and MCP-1 protein, as well as effectively enhancing oxidative signaling proteins, including the Nrf2/HO-1 system and its downstream antioxidant genes catalase and GCLM. Bae et al. (41) reported that Sesn2 induces Nrf2 activation by enhancing the autophagic degradation of Keap1. We also found that EMPA activated Nrf2/HO-1 by Sesn2, because treatment of KO-HFD-E did not upregulate Nrf2 mRNA or HO-1 protein, nor did it promote NF-κB activation. These findings indicated that Sesn2 is an essential factor for the EMPA-induced Nrf2/HO-1 antioxidative system. EMPA treatment activates the AMPK-signaling pathway, which can phosphorylate downstream ACC. The ACC phosphorylation leads to an augmented fatty acid oxidation that occurred in the HFD hearts. It could be a reason that EMPA treatment decreased lipid content in the HFD hearts.

The endothelial dysfunction induced by eNOS uncoupling initiates atherosclerosis, which is the leading cause of cardiovascular disease and often observed in obesity. We found that EMPA treatment triggered eNOS phosphorylation at Ser1177, indicating endothelial-protective effects of EMPA, which is consistent with a previous study (42). The PI3K/Akt pathway is also involved in cell proliferation, and activation of this pathway stimulates cell cycle progression and cardiac remodeling via the mTOR pathway. Akt activation in myocardial cells leads to hypertrophy and cardiac dysfunction in HFD mice with heart failure (43). We found that EMPA treatment inhibited phosphorylation of PI3K/Akt, indicating that EMPA could also inhibit the cardiac hypertrophy caused by the PI3K/Akt/mTOR pathway. We reported that EMPA triggers AMPK phosphorylation through AMPK upstream LKB1 activation (44). We also revealed that Sesn2 serves as a scaffold protein to modulate AMPK activation via recruiting AMPK upstream LKB1 to the Sesn2-AMPK complex (13). Therefore, Sesn2 KO limits LKB1 access to AMPK to phosphorylate AMPK by EMPA treatment.

The absence of data on the inhibition of Sesn2 by siRNA or overexpress Sesn2 in cardiomyocytes is a limitation of our study. In this regard, we used Sesn2 KO-HFD mice and treated them with EMPA, which better reflects the beneficial effects of EMPA for obesity-related cardiac dysfunction in vivo. Therefore, it is possible that the notable improvements in cardiac structure and function observed in our study could be due to Sesn2-mediated metabolic alterations. Additionally, some studies found that Sesn2 activation is dependent on hematopoietic progenitor kinase-germinal cell kinase-like kinase or protein kinase-like ER kinase (45,46). Further studies are needed to determine whether other SGLT2i can protect the heart via Sesn2 and whether EMPA activates Sesn2 through hematopoietic progenitor kinase-germinal cell kinase-like kinase or other signaling pathways.

This work clarified the critical functions of Sesn2 in obesity-related cardiac dysfunction and revealed that the molecular mechanism underlying the cardioprotective effects of EMPA on obesity-induced cardiac dysfunction was through regulating Sesn2-mediated AMPK-mTOR signaling and maintaining redox homeostasis. These promising findings will have an important positive impact in the field of heart failure research, as they provide potential therapeutic approaches for obesity-related cardiac dysfunction.

This article contains supplementary material online at https://doi.org/10.2337/db20-4567/suppl.12023325.

Acknowledgments. The authors thank Haiyan Zhang for measurement of the ELISA kit, Glenn Hoskins for transmission electron microscopy analysis, and Joshua Jefferson and Jesus Monico for tissue preparation and specific staining (all from University of Mississippi Medical Center).

Funding. The current study was supported by the National Natural Science Foundation of China (81870593), the American Diabetes Association (1-17-IBS-296), and the Overseas Study Program from the Government in Shandong Province.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. J.L. conceived and designed the study and participated in the revision and final approval of the manuscript. X.S. and F.H. participated in the study design, data collection, and writing of the draft manuscript. Q.L., X.L., D.R., J.Z., Y.H., and Y.K.X. participated in the partial data collection. All authors read and approved the final manuscript. J.L. is the guarantor of this work and, as such, had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in oral form at the 55th Annual Meeting of the European Association for the Study of Diabetes, Barcelona, Spain, 16–20 September 2019.

1.
Marinou
K
,
Tousoulis
D
,
Antonopoulos
AS
,
Stefanadi
E
,
Stefanadis
C
.
Obesity and cardiovascular disease: from pathophysiology to risk stratification
.
Int J Cardiol
2010
;
138
:
3
8
2.
Halade
GV
,
Kain
V
.
Obesity and cardiometabolic defects in heart failure pathology
.
Compr Physiol
2017
;
7
:
1463
1477
3.
Wang
Z
,
Li
L
,
Zhao
H
,
Peng
S
,
Zuo
Z
.
Chronic high fat diet induces cardiac hypertrophy and fibrosis in mice
.
Metabolism
2015
;
64
:
917
925
4.
Buss
SJ
,
Muenz
S
,
Riffel
JH
, et al
.
Beneficial effects of mammalian target of rapamycin inhibition on left ventricular remodeling after myocardial infarction
.
J Am Coll Cardiol
2009
;
54
:
2435
2446
5.
Steven
S
,
Frenis
K
,
Oelze
M
, et al
.
Vascular inflammation and oxidative stress: major triggers for cardiovascular disease
.
Oxid Med Cell Longev
2019
;
2019
:
7092151
6.
Kuwabara
Y
,
Horie
T
,
Baba
O
, et al
.
MicroRNA-451 exacerbates lipotoxicity in cardiac myocytes and high-fat diet-induced cardiac hypertrophy in mice through suppression of the LKB1/AMPK pathway
.
Circ Res
2015
;
116
:
279
288
7.
Li
X
,
Liu
J
,
Lu
Q
, et al
.
AMPK: a therapeutic target of heart failure-not only metabolism regulation
.
Biosci Rep
2019
;
39
:
BSR20181767
8.
Li
X
,
Liu
J
,
Hu
H
, et al
.
Dichloroacetate ameliorates cardiac dysfunction caused by ischemic insults through AMPK signal pathway-not only shifts metabolism
.
Toxicol Sci
2019
;
167
:
604
617
9.
Kim
H
,
An
S
,
Ro
SH
, et al
.
Janus-faced Sestrin2 controls ROS and mTOR signalling through two separate functional domains
.
Nat Commun
2015
;
6
:
10025
10.
Dong
B
,
Xue
R
,
Sun
Y
,
Dong
Y
,
Liu
C
.
Sestrin 2 attenuates neonatal rat cardiomyocyte hypertrophy induced by phenylephrine via inhibiting ERK1/2
.
Mol Cell Biochem
2017
;
433
:
113
123
11.
Quan
N
,
Wang
L
,
Chen
X
, et al
.
Sestrin2 prevents age-related intolerance to post myocardial infarction via AMPK/PGC-1α pathway
.
J Mol Cell Cardiol
2018
;
115
:
170
178
12.
Budanov
AV
,
Karin
M
.
p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling
.
Cell
2008
;
134
:
451
460
13.
Morrison
A
,
Chen
L
,
Wang
J
, et al
.
Sestrin2 promotes LKB1-mediated AMPK activation in the ischemic heart
.
FASEB J
2015
;
29
:
408
417
14.
Thomas
MC
,
Cherney
DZI
.
The actions of SGLT2 inhibitors on metabolism, renal function and blood pressure
.
Diabetologia
2018
;
61
:
2098
2107
15.
Fitchett
D
,
Inzucchi
SE
,
Cannon
CP
, et al
.
Empagliflozin reduced mortality and hospitalization for heart failure across the spectrum of cardiovascular risk in the EMPA-REG OUTCOME trial
.
Circulation
2019
;
139
:
1384
1395
16.
Kondo
H
,
Takahashi
N
.
Reduced hospitalization for heart failure using anti-diabetic drug dapagliflozin: implications of DECLARE-TIMI 58 for the basic science community
.
Cardiovasc Res
2019
;
115
:
e54
e57
17.
Mahaffey
KW
,
Neal
B
,
Perkovic
V
, et al.;
CANVAS Program Collaborative Group
.
Canagliflozin for primary and secondary prevention of cardiovascular events: results from the CANVAS Program (Canagliflozin Cardiovascular Assessment Study)
.
Circulation
2018
;
137
:
323
334
18.
Wu
JH
,
Foote
C
,
Blomster
J
, et al
.
Effects of sodium-glucose cotransporter-2 inhibitors on cardiovascular events, death, and major safety outcomes in adults with type 2 diabetes: a systematic review and meta-analysis
.
Lancet Diabetes Endocrinol
2016
;
4
:
411
419
19.
Martens
P
,
Mathieu
C
,
Verbrugge
FH
.
Promise of SGLT2 inhibitors in heart failure: diabetes and beyond
.
Curr Treat Options Cardiovasc Med
2017
;
19
:
23
20.
Yurista
SR
,
Silljé
HHW
,
Oberdorf-Maass
SU
, et al
.
Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction
.
Eur J Heart Fail
2019
;
21
:
862
873
21.
Di Franco
A
,
Cantini
G
,
Tani
A
, et al
.
Sodium-dependent glucose transporters (SGLT) in human ischemic heart: a new potential pharmacological target
.
Int J Cardiol
2017
;
243
:
86
90
22.
Xue
M
,
Chen
X
,
Guo
Z
, et al
.
L-carnitine attenuates cardiac dysfunction by ischemic insults through Akt signaling pathway
.
Toxicol Sci
2017
;
160
:
341
350
23.
Wang
L
,
Quan
N
,
Sun
W
, et al
.
Cardiomyocyte-specific deletion of Sirt1 gene sensitizes myocardium to ischaemia and reperfusion injury
.
Cardiovasc Res
2018
;
114
:
805
821
24.
Zanoli
L
,
Granata
A
,
Lentini
P
, et al
.
Sodium-glucose linked transporter-2 inhibitors in chronic kidney disease
.
ScientificWorldJournal
2015
;
2015
:
317507
25.
Li
Y
,
Chen
C
,
Yao
F
, et al
.
AMPK inhibits cardiac hypertrophy by promoting autophagy via mTORC1
.
Arch Biochem Biophys
2014
;
558
:
79
86
26.
Quan
N
,
Sun
W
,
Wang
L
, et al
.
Sestrin2 prevents age-related intolerance to ischemia and reperfusion injury by modulating substrate metabolism
.
FASEB J
2017
;
31
:
4153
4167
27.
Xu
W
,
Wang
C
,
Liang
M
, et al
.
A20 prevents obesity-induced development of cardiac dysfunction
.
J Mol Med (Berl)
2018
;
96
:
159
172
28.
Obokata
M
,
Reddy
YNV
,
Pislaru
SV
,
Melenovsky
V
,
Borlaug
BA
.
Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction
.
Circulation
2017
;
136
:
6
19
29.
McMurray
JJV
,
DeMets
DL
,
Inzucchi
SE
, et al.;
DAPA-HF Committees and Investigators
.
The Dapagliflozin And Prevention of Adverse-outcomes in Heart Failure (DAPA-HF) trial: baseline characteristics
.
Eur J Heart Fail
2019
;
21
:
1402
1411
30.
Li
C
,
Zhang
J
,
Xue
M
, et al
.
SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart
.
Cardiovasc Diabetol
2019
;
18
:
15
31.
Ye
Y
,
Bajaj
M
,
Yang
HC
,
Perez-Polo
JR
,
Birnbaum
Y
.
SGLT-2 inhibition with dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the development of diabetic cardiomyopathy in mice with type 2 diabetes. Further augmentation of the effects with saxagliptin, a DPP4 inhibitor
.
Cardiovasc Drugs Ther
2017
;
31
:
119
132
32.
Ghantous
CM
,
Azrak
Z
,
Hanache
S
,
Abou-Kheir
W
,
Zeidan
A
.
Differential role of leptin and adiponectin in cardiovascular system
.
Int J Endocrinol
2015
;
2015
:
534320
33.
Wu
P
,
Wen
W
,
Li
J
, et al
.
Systematic review and meta-analysis of randomized controlled trials on the effect of SGLT2 inhibitor on blood leptin and adiponectin level in patients with type 2 diabetes
.
Horm Metab Res
2019
;
51
:
487
494
34.
Sorop
O
,
Olver
TD
,
van de Wouw
J
, et al
.
The microcirculation: a key player in obesity-associated cardiovascular disease
.
Cardiovasc Res
2017
;
113
:
1035
1045
35.
Yang
JH
,
Obokata
M
,
Reddy
YNV
,
Redfield
MM
,
Lerman
A
,
Borlaug
BA
.
Endothelium-dependent and independent coronary microvascular dysfunction in patients with heart failure with preserved ejection fraction
.
Eur J Heart Fail
2020
;
22
:
432
441
36.
Chan
AY
,
Dyck
JR
.
Activation of AMP-activated protein kinase (AMPK) inhibits protein synthesis: a potential strategy to prevent the development of cardiac hypertrophy
.
Can J Physiol Pharmacol
2005
;
83
:
24
28
37.
Jia
G
,
Aroor
AR
,
Martinez-Lemus
LA
,
Sowers
JR
.
Overnutrition, mTOR signaling, and cardiovascular diseases
.
Am J Physiol Regul Integr Comp Physiol
2014
;
307
:
R1198
R1206
38.
Sun
X
,
Pan
H
,
Tan
H
,
Yu
Y
.
High free fatty acids level related with cardiac dysfunction in obese rats
.
Diabetes Res Clin Pract
2012
;
95
:
251
259
39.
Erkens
R
,
Kramer
CM
,
Lückstädt
W
, et al
.
Left ventricular diastolic dysfunction in Nrf2 knock out mice is associated with cardiac hypertrophy, decreased expression of SERCA2a, and preserved endothelial function
.
Free Radic Biol Med
2015
;
89
:
906
917
40.
Bai
T
,
Wang
F
,
Zheng
Y
, et al
.
Myocardial redox status, mitophagy and cardioprotection: a potential way to amend diabetic heart
?
Clin Sci (Lond)
2016
;
130
:
1511
1521
41.
Bae
SH
,
Sung
SH
,
Oh
SY
, et al
.
Sestrins activate Nrf2 by promoting p62-dependent autophagic degradation of Keap1 and prevent oxidative liver damage
.
Cell Metab
2013
;
17
:
73
84
42.
Zhou
H
,
Wang
S
,
Zhu
P
,
Hu
S
,
Chen
Y
,
Ren
J
.
Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission
.
Redox Biol
2018
;
15
:
335
346
43.
O’Neill
BT
,
Abel
ED
.
Akt1 in the cardiovascular system: friend or foe
?
J Clin Invest
2005
;
115
:
2059
2064
44.
Lu
Q
,
Liu
J
,
Li
X
, et al
.
Empagliflozin attenuates ischemia and reperfusion injury through LKB1/AMPK signaling pathway
.
Mol Cell Endocrinol
2020
;
501
:
110642
45.
Yen
JH
,
Huang
ST
,
Huang
HS
, et al
.
HGK-sestrin 2 signaling-mediated autophagy contributes to antitumor efficacy of Tanshinone IIA in human osteosarcoma cells
.
Cell Death Dis
2018
;
9
:
1003
46.
Kim
HJ
,
Joe
Y
,
Kim
SK
, et al
.
Carbon monoxide protects against hepatic steatosis in mice by inducing sestrin-2 via the PERK-eIF2α-ATF4 pathway
.
Free Radic Biol Med
2017
;
110
:
81
91
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.