1. Introduction
According to the data of the International Diabetes Federation, the prevalence of diabetes among people aged 20–79 in the world is 10.5%, and China is the country with the largest number of diabetes patients [
1]. Cardiovascular complications are a leading cause of death in diabetic patients. DCM was first proposed by Shirley Rubler in 1972, with altered myocardial structure and function independent of other cardiovascular diseases [
2,
3]. In diabetic patients, the main metabolic substrate switches from glucose to free fatty acid, a less efficient substrate than glucose in cardiac tissue, resulting in lower cardiac efficiency and further metabolic disorders [
4]. When fatty acid levels exceed mitochondrial utilization capacity, it leads to the accumulation of lipids and toxic metabolites, resulting in mitochondrial dysfunction and cardiac lipotoxicity [
5]. Lipotoxicity, directly impeding the metabolism of cardiomyocytes and promoting cells’ death [
4], plays an important role in the development of DCM. Clinical trials indicated that CAN, a sodium glucose cotransporter 2 inhibitor, could significantly reduce the cardiovascular risks in diabetic patients [
6,
7,
8]. Our previous study found that CAN improved heart failure caused by lipotoxicity and exerted anti-inflammatory effects [
9,
10,
11]. It suggested that CAN may be a potential ideal drug for the treatment of DCM, but the specific mechanism needs further study.
Inflammation is a key pathogenic feature of DCM, which occurs in various diseases. The activation of NF-κB (p65) leads to leukocyte infiltration and upregulation of inflammatory pathways, resulting in severe myocardial damage [
12]. Hyperglycemia, as well as increased free fatty acid metabolism, is associated with the upregulation of pro-inflammatory cytokines (e.g., IL-6, TNF-α) [
12]. Inflammatory responses are involved in various processes, such as cardiomyocyte apoptosis, pro-fibrosis, and cardiomyocyte hypertrophy, impair cardiomyocyte contractility, and directly or indirectly promote the development of DCM [
13]. Potential therapeutic strategies targeting inflammation are expected to show promising efficacy in future studies.
In 1981, Jerome L. Sullivan first proposed the hypothesis of “iron-derived heart disease”, but it was not until 2012 that Dixon discovered ferroptosis [
14]. Ferroptosis might be an important breakthrough in the pathogenesis of heart disease, though there are few studies on the mechanism of ferroptosis in cardiomyocytes. The main mechanism of ferroptosis is to catalyze the lipid peroxidation of fatty acids on the cell membrane under the action of ferrous iron or ester oxygenase, thereby inducing cell death [
15,
16]. Iron accumulation, ROS, increased fatty acid supply, and lipid peroxidation are key to inducing ferroptosis [
16]. Fang et al. observed the features of typical ferroptosis in doxorubicin-treated cardiomyocytes, revealing that ferroptosis might be a target for protection against cardiomyopathy [
17].
We found that inflammatory factors were upregulated in palmitic acid (PA)-induced HL-1 cells. COX-2 and iNOS screened by gene chip were suggested to be an important cause of myocardial lipotoxicity, using molecular biological techniques. Ferroptosis, related to lipid peroxidation, also existed in PA-induced cardiomyocytes. By comparing CAN with coxib and SMT, we unexpectedly found that anti-inflammatory treatments inhibited ferroptosis. It suggested that CAN regulated ferroptosis, possibly by inhibiting inflammation. In addition, COX-2 and iNOS were inhibited by Fer-1. That is to say, inflammation and ferroptosis might interact with each other. We speculated that CAN attenuated lipotoxicity in cardiomyocytes, possibly by inhibiting inflammation and ferroptosis. Therefore, in this study, we investigated the mechanisms underlying the anti-lipotoxic effects of CAN on inflammation and ferroptosis.
3. Discussion
Although it has been 50 years since DCM was first discovered, many mechanisms are still unclear. Lipotoxicity plays a key role in the development of DCM, so studying the mechanisms of lipotoxicity is crucial for the treatment of DCM. Currently, there is still no specific treatment for DCM, so “reuse of old drugs” is a fast, economical, and safe way to solve it, which has a significant advantage over developing new drugs. Previous studies have demonstrated that SGLT2 inhibitors could inhibit lipotoxicity and inflammation. CAN significantly reduced the levels of IL-1, IL-6, and TNF-α in cardiomyocytes [
9]. This suggested that CAN alleviated lipotoxicity, possibly by reducing inflammation. Ferroptosis is closely related to lipid peroxidation [
16]. Therefore, we hypothesized that there was also some link between ferroptosis and DCM. Hyperglycemia and disorders of lipid metabolism are the main features of DCM. In this study, we used a high-glucose medium to maintain cell growth and added PA, a widely used lipotoxicity modeling agent, to construct a lipotoxicity model in HL-1 cells. Sequentially, we investigated the protective mechanisms of CAN against lipotoxicity from both inflammation and ferroptosis. Our results showed that CAN significantly inhibited the expression of COX-2 and iNOS as well as the key indicators of ferroptosis, possibly due to the activation of AMPK, which in turn ameliorated inflammation and ferroptosis, alleviating lipotoxicity.
The anti-inflammatory effect of CAN has been reported in many studies. Niu et al. first reported that CAN attenuated NLRP3 inflammasome-mediated inflammation in immune cells by inhibiting NF-κB signaling and upregulating Bif-1, providing a new idea for the anti-inflammatory effect of CAN [
10]. The discovery and selection of inflammatory targets might be accidental in many studies. In this study, we systematically screened the key inflammatory genes for further studies through bioinformatics analysis, which had better traceability and logic. Through subsequent repeated verification, we found that COX2 and iNOS screened from the gene chip might be important factors in mediating inflammation of cardiomyocytes. COX-2, an isoform of prostaglandin-endoperoxide synthase, acts in a pro-inflammatory manner, which is induced by multiple intracellular and extracellular stimuli [
18]. The iNOS, a member of the nitric oxide synthase family, is only expressed in induced or stimulated cells, especially by pro-inflammatory cytokines or lipopolysaccharide [
19]. The high expression of COX-2 and iNOS might be an important reason for the myocardial lipotoxicity. Targeting inflammation might be a reasonable strategy to attenuate lipotoxicity. In our study, we found that CAN had a significant regulatory effect on these inflammatory mediators. In addition, using specific inflammatory inhibitors and CAN, our study suggested that COX-2 had a greater effect on iNOS while, conversely, iNOS had a milder effect on COX-2. Therefore, we speculated that COX-2 might be an upstream protein of iNOS.
Several studies showed the existence of ferroptosis in cardiovascular diseases [
17,
20]. Ferroptosis, an iron-dependent, lipid peroxidation-driven programmed cell death, is different from apoptosis, autophagy, and pyroptosis [
15]. Therefore, we suspected that targeting ferroptosis might be an effective strategy for protection against PA-treated HL-1 cells. Ferroptosis is mainly characterized by abnormal iron metabolism, accumulation ofROS, lipid peroxidation, glutathione metabolism disorders, and mitochondrial membrane potential (MMP) hyperpolarization [
16,
21]. As one of the key indicators, iron ion content can indicate whether ferroptosis occurs in cardiomyocytes [
15]. An excess supply of fatty acids led to incomplete mitochondrial fatty acid oxidation, which caused an increase in redox pressure on the electron transport chain and ROS production, resulting in cellular oxidative stress and damage [
22,
23]. Lipid peroxidation plays a key role in cell damage and death in ferroptosis [
16]. MDA, a peroxidative intermediate in the process of ferroptosis lipid peroxidation, is a key markers of lipid peroxidative damage. GSH, an antioxidant in animal cells, plays an important role in the antioxidant system of ferroptosis [
15,
20]. Studies showed that the MMP undergoes morphological changes of hyperpolarization in ferroptosis [
21]. Therefore, based on the indicators, we evaluated whether ferroptosis was present in cardiomyocytes. Our results showed that Fer-1 corrected these indicators, indicating that ferroptosis did exist in cardiomyocytes. Recent studies showed that the SGLT2 inhibitor reduced ROS and had a significant inhibitory effect on MDA and SOD [
9,
22]. However, few studies systematically investigated the effect of CAN on ferroptosis. With this study, we were the first to discover that CAN could inhibit the indicators of ferroptosis in PA-treated HL-1 cells. Compared with specific inflammatory inhibitors, CAN was comparable in the regulation of ferroptosis. Anti-inflammation could inhibit the process of ferroptosis, which also suggested that CAN regulated ferroptosis, possibly mediated by inflammation inhibition. In addition, through assessing the effect of ferroptosis on COX-2 and iNOS, we found that ferroptosis mediates the occurrence of inflammation in PA-treated HL-1cells. That is, inflammation and ferroptosis interacted with each other. It suggested that CAN might ameliorate lipotoxicity by inhibiting inflammation and ferroptosis.
AMPK, known as a “metabolic master switch”, has many beneficial effects on cardio-metabolic abnormalities, such as myocardial inflammation, oxidative stress, and altered substrate utilization [
3]. CAN exerts an anti-inflammatory effect by activating AMPK and inhibiting the NF-κB (p65) pathway [
10]. P65 is essential for the expression of COX-2, by mediating the activity of a COX-2 promoter [
24]. The iNOS induced by inflammatory signals is also regulated by p65 through directly binding to an
NOS2 promoter [
19,
25]. In addition, our studies above suggested that COX-2 might regulate the expression of iNOS. Therefore, we speculated that CAN inhibited inflammation in cardiomyocytes by inhibiting the inflammatory pathway of p-p65/COX-2/iNOS through activating AMPK. Our study demonstrated that the inactivation of AMPK indeed significantly weakened the inhibitory effect of CAN on inflammatory pathways, consistent with our hypotheses. AMPK also plays a role in the ferroptosis pathway. The biosynthesis of fatty acids is restrained and ferroptosis is inhibited when phosphorylated AMPK is activated by energy stress [
26]. Collectively, our findings suggested that CAN alleviated lipotoxicity by inhibiting inflammation and ferroptosis, possibly due to the activation of AMPK.
Pro-inflammatory chemokines are a class of chemotactic cytokines that regulate cell migration and localization in inflammation [
27]. In this study, we found that some chemokines were significantly upregulated in gene chips. Additionally, CAN had a significant inhibitory effect on the mRNA levers of
Ccl2,
Ccl5, and
Cxcl1 in PA-treated HL-1 cells. Previous studies discussed the importance of the chemokines above in acute inflammatory response and that
Ccl2 may be an effective target for the treatment of heart failure [
27,
28]. Therefore, we speculated that pro-inflammatory chemokines might play a role in the inflammation of DCM. In addition, our study only selected part of the results in the gene chips for detailed research; there are still many key genes that need to be further studied, such as
Serpine1,
Vegfa, etc. (
Figure S1A,B). A variety of bioinformatics technologies can be adopted to optimize the screening methods in the future, which will contribute to obtaining more accurate and comprehensive results through systematic research. Regarding changes of MMP in ferroptosis, MMP hyperpolarization and the loss of MMP were the main opinions [
17]. We speculated that MMP was hyperpolarized in the early stage of ferroptosis, as we demonstrated before, and dissipated in the later stage, leading to mitochondrial dysfunction and severe cell damage. Our study demonstrated the existence of ferroptosis in PA-treated HL-1 cells, mainly through testing the key indicators of ferroptosis. Previous studies suggested that apoptosis was a pattern of death in cardiomyocytes [
9], whereas we found that ferroptosis also occurred in cardiomyocytes. In addition, cell viability was only partially improved by reducing inflammation and ferroptosis in our study, probably because there are other ways for cell death, such as apoptosis, autophagy, pyroptosis, etc. Therefore, we speculated that there might be multiple patterns of death in cardiomyocytes. It still requires more data to further support our findings and hypotheses.
4. Materials and Methods
4.1. Cell Culture and Treatment
HL-1 cells (CL0683) were purchased from Fenghui Biotechnology Co., Ltd., Changsha, China. Cells were cultured in high-glucose DMEM (Gibco, Thermo Fisher Scientific, Waltham, MA, USA), supplemented with 10% fetal bovine serum (FBS; Gibco, Thermo Fisher Scientific, Waltham, MA, USA) and 1% penicillin-streptomycin antibiotic (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) in an incubator of 5% CO2 at 37 °C. For inducing a lipotoxic cardiomyocyte model, cells were seeded into cell culture dishes or plates at an appropriate density and stimulated with 0.1 mM palmitic acid (PA; Sigma-Aldrich, Saint Louis, MO, USA) for 24 h in the presence or absence of CAN (5 μg/mL, BiochemPartner, Shanghai, China). Bovine serum albumin (BSA; 5%, g/mL, BIOFROXX, Einhausen, Germany) used to liquefy PA, was also added to blank control cells. COX-2 inhibitor celecoxib (5 μg/mL, TCI, Shanghai, China), iNOS inhibitor SMT (10 μM, Beyotime, Shanghai, China), and the ferroptosis inhibitor Fer-1 (5 μM, ABclonal Technology, Wuhan, China) were added simultaneously with PA. AMPK inhibitor Compound C (5 μg/mL, BiochemPartner, Shanghai, China) was added with PA and CAN.
4.2. Western Blotting Analysis
The whole cell lysates from HL-1 cells were collected, and the protein samples were separated using 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE; Epizyme Biotech, Shanghai, China). Then, they were transferred to nitrocellulose filter membranes (Pall, New York, NY, USA). Subsequently, the membranes were blocked with a fresh blocking buffer (5% g/mL nonfat milk powder (Epizyme Biotech, Shanghai, China), which was dissolved in tris-buffered saline with 0.2% Tween-20 (TBST)) for 2 h and then incubated with primary antibodies diluted with 1.5% BSA overnight at 4 °C. After a third rinse with TBST, the membranes were incubated with the secondary antibodies for 1 h at room temperature and then rinsed three more times with TBST. After that, the protein bands were visualized with a chemiluminescence solution (Thermo Fisher Scientific, Waltham, MA, USA). Finally, the ImageJ 1.48 software (National Institutes of Health, Bethesda, MD, USA) was used to quantify the relative gray density values of the protein bands.
The following were the specific primary antibodies used in the study: β-actin (1:5000, Mouse, A1978, Sigma-Aldrich, Saint Louis, MO, USA), COX-2 (1:1000, ET1610-23, HUABIO, Hangzhou, China), iNOS (1:1000, Rabbit, A3200, Abclonal Technology, Wuhan, China), LKB1 (1:1000, Rabbit, A2122, Abclonal Technology, Wuhan, China), TAK1 (1:1000, Rabbit, ET1705-14, HUABIO, Hangzhou, China), CaMKK2 (1:1000, Rabbit, A9899, Abclonal Technology, Wuhan, China), Phospho-CaMKK2 (Ser511) (1:1000, Rabbit, BD-PP1258, Biodragon, Suzhou, China), AMPKα (1:1000, Rabbit, 2532S, Cell Signaling Technology, Boston, MA, USA), Phospho-AMPKα (Thr172) (1:1000, Rabbit, 2535S, Cell Signaling Technology, Boston, MA, USA), NF-κB p65 (1:1000, Mouse, 6956S, Cell Signaling Technology, Boston, MA, USA), and Phospho-NF-κB p65 (Ser536) (1:1000, Rabbit, 3033S, Cell Signaling Technology, Boston, MA, USA). The following secondary antibodies were used: goat polyclonal antibody to rabbit IgG H&L HRP (1:5000, 7074P2, Cell Signaling Technology, Boston, MA, USA) and goat polyclonal antibody to mouse IgG H&L HRP (1:5000, 7076S, Cell Signaling Technology, Boston, MA, USA).
4.3. Quantitative Real-Time Polymerase Chain Reaction (RT-qPCR) Assay
Total RNA from HL-1 was extracted by AG RNAex Pro Reagent (Accurate Biology, Changsha, China) according to the manufacturer’s instructions. The concentration of the total RNA was measured by a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA), and approximately 500 ng of total RNA was used for reverse transcription. Subsequently, cDNA was synthesized with Evo M-MLV RT Premix for qPCR (Accurate Biology, Changsha, China) and then quantitatively analyzed with SYBR Green Premix Pro Taq HS qPCR Kit (Accurate Biology, Changsha, China). Finally, the 2−ΔΔCt method was used to calculate the relative gene expression.
The following were the sequences of primers used for RT-qPCR, which were synthesized by GENEWIZ, China: 5′-GGCTGTATTCCCCTCCATCG-3′ and 5′-CCAGTTGGTAACAATGCCATGT-3′ for β-actin, 5′-TGAGCAACTATTCCAAACCAGC-3′ and 5′-GCACGTAGTCTTCGATCACTATC-3′ for Ptgs2, 5′-GTTCTCAGCCCAACAATACAAGA-3′ and 5′-GTGGACGGGTCGATGTCAC-3′ for Nos2, 5′-GCTGCTTTGCCTACCTCTCC-3′ and 5′-TCGAGTGACAAACACGACTGC-3′ for Ccl5, 5′-TTAAAAACCTGGATCGGAACCAA-3′ and 5′-GCATTAGCTTCAGATTTACGGGT-3′ for Ccl2, and 5′-CTGGGATTCACCTCAAGAACATC-3′ and 5′-CAGGGTCAAGGCAAGCCTC-3′ for Cxcl1.
4.4. Thiazolyl Blue Tetrazolium Bromide (MTT) Assay
The cell viability was analyzed by MTT (Sangon Biotech, Shanghai, China) assay. Each well of cells was added 20 µL MTT (5 mg/mL) solution, and the cells were incubated for 2 h in an incubator of 5% CO2 at 37 °C. Then, we removed the cell medium and added 200 µL dimethyl sulfoxide (DMSO; Sangon Biotech, Shanghai, China) solution to thoroughly dissolve the purple formazan crystals. Optical density values at 490 nm (OD490) were analyzed by an Epoch microplate spectrophotometer (Bio-Tek, Winooski, VT, USA).
4.5. NO, MDA, and GSH Assays
Cell culture supernatants were collected and assessed by a Total Nitric Oxide Assay kit (S0023, Beyotime, Shanghai, China) for NO detection. The supernatants were collected after repeated freeze–thaw cycles of cells at −80 °C and assessed by GSH and GSSG Assay kit (S0053, Beyotime, Shanghai, China) for GSH detection. The whole cell lysates were collected and assessed by a Lipid Peroxidation MDA Assay Kit (S0131S, Beyotime, Shanghai, China) for MDA. They were all conducted according to the manufacturer’s instructions, and the protein concentration was determined by a Detergent Compatible Bradford Protein Assay Kit (P0006C, Beyotime, Shanghai, China).
4.6. Intracellular Ferrous Ion (Fe2+) Measurement
Intracellular Fe2+ was detected by a FeRhoNox-1 (MX4558, MKBio, Shanghai, China) fluorescent probe according to the manufacturer’s instructions. After one rinse with PBS buffer, the cells were incubated with 5 µM FeRhoNox-1 for 1 h in an incubator of 5% CO2 at 37 °C. Then, the fluorescence of Fe2+ under a Cy3 excitation filter was observed by a fluorescence microscope after two rinses with PBS buffer. Finally, ImageJ software was used to measure the fluorescence intensity of Fe2+.
4.7. ROS Assay
ROS was detected by a DCFH-DA (S0033S, Beyotime, Shanghai, China) fluorescent probe according to the manufacturer’s instructions. After one rinse with PBS buffer, the cells were incubated with 10 µM DCFH-DA (1:1000 diluted in serum-free culture medium) for 20 min at 37 °C. The fluorescence intensity was observed and measured after three rinses with a serum-free culture medium.
4.8. MMP Assay
MMP was detected by a mitochondrial membrane potential assay kit with JC-1 (C2006, Beyotime, Shanghai, China) according to the manufacturer’s instructions. The cells were collected and incubated with a JC-1 working solution for 25 min at 37 °C. After two rinses with JC-1 buffer (1×), the cells were resuspended with JC-1 buffer (1×) and filtered with 200-mesh cell sieves. Finally, the fluorescence signals were detected by flow cytometry and quantified by FlowJo_v10.8.1 software (BD, Ashland, OR, USA).
4.9. Statistical Analysis
All data were presented as the mean ± standard deviation (SD) in this study. Statistical differences among the different groups were analyzed by ANOVA followed by Tukey’s post hoc test. Statistical differences were considered significant at p < 0.05 and highly significant at p < 0.01.