2.1 HFD/STZ-induced diabetic mouse models
C57BL/6 mice (body weight, 18±2 g; 4-weeks-old; male) were purchased from Guangzhou Medical Animal Centre (Guangzhou, China) and housed under controlled conditions (constant temperature: 22±2˚C; constant humidity: 60±5%; 12 h dark/light cycle). The study was performed in strict accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and the protocol was approved by the Bioethics Committee of Shenzhen International Graduate School, Tsinghua University, China (Ethics issue [2020] No. 9). The diabetic mouse models were fed with a HFD (41% energy from fat; Beijing HFK Bioscience, Beijing, China, H10141). The models were intraperitoneally injected with STZ (40 mg/kg; Sangon Biotech Co., Ltd., Shanghai, China), which was freshly dissolved in ice-cold 0.1 M citrate solution (pH 4.5), according to a previous study (12). After one week, the diabetic mice were divided into two groups: An untreated HFD/STZ control group and a CAN (ChenXi Biotech, Shanghai, China)-treated group (25 mg/kg/d; dissolved in 0.5% sodium salt of carboxymethyl cellulose [CMC-Na; Sangon Biotech, Shanghai, China]). The dose of CAN administered to the mice was converted according to the clinical dose in humans (100 mg/kg/day). The normal control and HFD/STZ control groups were orally administered with an equal volume of 0.5% CMC-Na daily. The body weight of the mice, and food and water uptake was monitored once a week. After 12 weeks of treatment, the mice were subjected to fasting for 6 h, anesthetized with urethane, which was dissolved in saline (intraperitoneal injection of urethane at a dose of 1000 mg kg-1; Sangon Biotech Co., Ltd., Shanghai, China), then sacrificed while under anesthesia. The blood was collected from the orbital plexus veins, and the serum was extracted from the blood samples using centrifugation (1000 rpm for 10 min at 4˚C) and stored at -20˚C for further analysis. Simultaneously, unrecovered anesthetized animals were sacrificed using cervical dislocation by skillful well-trained investigators, and the hearts and abdominal adipose tissues were removed and weighed. A portion of the heart tissue was immersed in 4% paraformaldehyde solution for regular pathological slicing, and hematoxylin and eosin (H&E) and Masson staining. The remaining samples were instantly frozen by using liquid nitrogen and stored at -80˚C for further analysis.
2.2 Biochemical analysis in animals
The levels of serum cholesterol, triglyceride, glucose, lactate dehydrogenase (LDH) and creatine kinase (CK) were measured using regular commercial kits (BioSino Bio-Technology & Science Inc, Beijing, China). B-type natriuretic peptide (BNP) was detected using an ELISA kit (YingXinBio, Shanghai, China). Total protein from the heart tissue was extracted using a cell lysis buffer, and the protein concentration was determined with a commercial kit (both from Beyotime Institute of Biotechnology, Shanghai, China). The cholesterol, triglyceride, and glycogen levels in the heart tissues were analyzed with commercial kits (Nanjing Jiancheng, Nanjing, China) and their levels were normalized to the protein concentration.
2.3 Cell culture
The HL-1 cells were purchased from Fenghui Biotechnology Company, (Changsha, China) and cultured in high glucose DMEM (Gibco®; Thermo Fisher Scientific, Inc., USA) supplemented with 10% premium fetal bovine serum (Pan Biotech, Germany) and 1% penicillin-streptomycin antibiotic (Gibco™; Thermo Fisher Scientific, Inc., USA), at 37˚C in a humidified atmosphere with 5% O2. The cells were seeded at a density of either 8 × 103 cells per well in 96-well plates, 2 × 104 cells per well in 24-well plates, or 3 × 105 cells in 6-well plates (all from Guangzhou Jet Bio-Filtration Co., Ltd.). The HL-1 cells were incubated with PA (0.25 mM; Sigma-Aldrich, USA) for 24 h. Different concentrations of CAN (0.625, 1.25 and 2.5 μg/ml), rapamycin (RAPA; 100, 200, and 400 nM; MedChemExpress, USA), and LW6 (5, 10 and 20 μM; MedChemExpress, USA) were added to investigate the anti-inflammatory activities on PA-induced HL-1 cells. PA was dissolved in 3% bovine serum albumin (BSA; Biofroxx, Germany) solution. CAN, RAPA, and LW6 were dissolved in dimethyl sulfoxide (DMSO; Beyotime Institute of Biotechnology, Shanghai, China). Blank control cells were treated with an equal volume of BSA or DMSO solution. After 24 h of treatment, oil red O (Sangon Biotech Co., Ltd., Shanghai, China) staining was performed to observe fat accumulation. Total protein and RNA was extracted, as described previously (13). The mRNA expression levels of Hif-1α, Il-1α, Il-6, and Tnf-α were analyzed using reverse transcription-quantitative PCR (RT-qPCR). The phosphorylation of HIF-1α and mTOR was measured using western blot analysis.
2.4 MTT assay
MTT (Sangon Biotech, Shanghai, China) was dissolved in phosphate buffer solution (PBS; 0.01 M; pH,7.0) at a concentration of 5 mg/ml and sterilized by filtration with 0.22 μm filters. The HL-1 cells (8 × 103 per well) were seeded into 96-well plates. The CAN, RAPA and LW6 reagents were dissolved in DMSO. The HL-1 cells were incubated with PA (0.25 mM) for 24 h to induce lipotoxicity. Different concentrations of CAN (0.625, 1.25 and 2.5 μg/ml), RAPA (100, 200 and 400 nM), and LW6 (5, 10 and 20 μM) were simultaneously added for 24 h to investigate the protective activity on PA-induced lipotoxicity. The blank or untreated control groups were treated with an equal volume of BSA solution. After 24 h of treatment, 20 μl MTT solution was added to each well and incubated for 4 h in the incubator. Following which, the cell medium was removed and 200 μl DMSO was added to dissolve the purple formazan crystals in each well. Optical density values at 490 nm (OD490) were analyzed using a microplate spectrophotometer. The following equation was used to determine the percentage cell viability: Cell viability % = (OD490 of each group/average OD490 of the blank control group) × 100%.
2.5 RT-qPCR assay
The mRNA expression level was analyzed, as described in a previous study (14). Approximately 500 ng total RNA was reverse transcribed into cDNA using the Evo M-MLV RT Premix for the qPCR kit (Accurate Biotechnology, Hunan, China). RT-qPCR was performed using SYBR Green Premix Pro Taq HS qPCR kit (Accurate Biotechnology, Hunan, China). The primers were synthesized by Genewiz, Inc., Suzhou, China (Table 1).
2.6 Western blot analysis
Western blot analysis was conducted according to a previous study, with a slight modification (14). The cell lysates were collected, and the protein samples were separated using 10% SDS-PAGE, then transferred to PVDF membranes (Pall, USA). Subsequently, the membranes were blocked with blocking buffer (5% g/ml skimmed milk power (Anchor, New Zealand), which was dissolved in PBS containing 0.5% g/ml Tween-20 (PBST; Sangon Biotech Co., Ltd., Shanghai, China) for 1 h, then incubated with primary antibodies dissolved in 3% BSA overnight at 4˚C. After the membrane was washed with PBST buffer (PBS added with 0.5% Tween-20) three times, the secondary antibodies (dissolved in blocking buffer) were added and incubated for 1 h. After washing again, the protein bands were visualized using enhanced chemiluminescence (Pierce™ ECL Western Blotting Substrate; Thermo Fisher Scientific, Inc., USA). The following primary antibodies were used: β-actin (mouse; 1:5000; A1978; Sigma-Aldrich), HIF-1α (mouse; 1:1000; 14179S; Cell Signaling Technology, Inc.), mTOR (mouse; 1:1000; 2983; Cell Signal Technology, Inc.), and phosphorylated (p)-mTOR (mouse; 1:1000; 5536; Cell Signal Technology Inc.).
2.7 ROS assay
The fluorescent probe, 2’, 7’-dichlorodihydrofluorescein diacetate (Beyotime Institute of Biotechnology) was used to detect reactive oxygen species (ROS) levels, as described in our previous study (13). The HL-1 cells (2 × 104 per well) were seeded into 24-well plates, then incubated with PA (0.25 mM) for 24 h to induce ROS generation. CAN (2.5 μg/ml) was simultaneously treated for 24 h to investigate the activity against ROS. The blank and untreated control cells were treated with an equal volume of DMSO or BSA solution. The fluorescent signal was measured using a fluorescence microscope (excitation wavelength of 485 nm; emission wavelength of 525 nm).
2.8 Immunofluorescence and confocal assay
The immunofluorescence assay in the HL-1 cells was performed as previously described (13). First, circular transparent glass slides (cat. no. 12-545-83; Fisherbrand™ microscope cover glass; Thermo Fisher Scientific, Inc. USA) were placed at the bottom of a 6-well plate. The cells, at a density of 2.5 × 105/well, were added to the surface of glass slides in a 6-well plate, then cultured in fresh medium. After 24 h of culture, the cells were divided into three groups: Normal untreated control group, PA (0.25 mM)- treated group, and PA (0.25 Mm)- and CAN (2.5 μg/ml)- treated group. After 24 h, the immobilized cells on the slides were washed with PBS, then fixed with 4% paraformaldehyde (Sangon Biotech, Shanghai, China) in PBS for 15 mins. The cells were washed with PBS three times, then incubated with 0.1% Triton X-100 in PBS for 15 min. After three washes with PBS, the cells were blocked with 3% BSA (Sangon Biotech, Shanghai, China) in PBS for 1 h. After blocking, the cells were incubated with p-mTOR (Ser2448) rabbit monoclonal antibody mAb (1:200; 5536S; Cell Signaling Technology, Inc., USA) and HIF-1α mouse monoclonal antibody (1:200; CST14179S; Cell Signaling Technology, Inc., USA) in 3% BSA for 1 h, then washed with PBS three times. Next, the cells were incubated with goat anti-rabbit IgG H&L (1:200; ab150077; Alexa Fluor® 488; Abcam, UK) or goat anti-mouse IgG H&L (1:100; ab150119; Alexa Fluor® 647; Abcam, UK) in 3% BSA for 1 h. The fluorescence in the cell was observed using confocal microscopy (Olympus, Corporation Japan) and analyzed using FV10-ASW Viewer v3.1 and ImageJ software.
2.9 Apoptosis assays
For the flow cytometry assays, the HL-1 cells (2 × 105/well) were cultured in 6-well plate. The cells were divided into three groups: Normal untreated control, PA (0.25 mM)- treated, and PA (0.25 Mm)- and CAN (2.5 μg/ml)-treated groups. After 24 h of treatment, the collected cells were fluorescently stained using the YF®488-Annexin V and PI Apoptosis commercial kit (Y6002; US Everbright Inc., Suzhou, Jiangsu, China). Flow cytometry was detected using a BD Accuri C6 flow cytometer (BD Biosciences, USA) and the results were analyzed using FlowJo v10.6.2 software.
For the confocal assay, the HL-1 cells (2 × 105/well) were seeded onto the surface of circular transparent glass slides in a 6-well plate and analyzed according to the confocal assay protocol, as described above. The cells on the slide were fluorescently stained using the YF®488-Annexin V and PI Apoptosis commercial kit (Y6002; US Everbright Inc, Suzhou, Jiangsu, China).
2.10 Pathway enrichment analyze assay
SwissTargetPrediction (http://www.swisstargetprediction.ch/) is a web server for small molecule drug target prediction (15). Using the web server, we obtained possible targets of CAN. ClueGo is a powerful tool for network analysis, annotation, and visualization. The predicted targets were analyzed using ClueGo 2.5.4 to enrich the pathway network for the Kyoto Encyclopedia of Genes and Genomes pathway database (16) (the threshold was set at P<0.05). GeneCards is a database containing details of disease-related genes and proteins (16). The key words “diabetes” and “HF” were used in GeneCards to search for disease-related genes. From the results of the diabetes- and HF-related genes, we selected the top 200 genes and analyzed them using ClueGo (the threshold was set at P<0.001). The enrichment results and intersection analysis were conducted using Origin Pro 2018c.
2.11 Molecular docking assay
SwissDock is an online sever for small molecule-protein docking (17). The mTOR crystal structure was obtained using the Protein Data Bank (PDB ID: 4DRI). The molecular docking model and affinity energy of CAN and RAPA with mTOR were analyzed using SwissDock (www.swissdock.ch) and the results were visualized using the software, Chimera v1.14.
2.12 Affinity chromatography
Affinity chromatography was conducted according to a previous study (18) with a slight modification.
Preparation of the drug: CAN (5 mg) was dissolved in 0.35 ml of 80% ethanol, while sucrose (3 mg) was dissolved in 0.35 ml of 40% ethanol. The two solutions were dropped into 0.25 ml of 8 mg/ml sodium periodate (Sangon Biotech Co., Ltd.) solution and left for 1 h for the reaction to occur. Following which, 0.5 ml of 100 mM sodium carbonate buffer (pH, 9) was added to the two solutions.
Preparation of the affinity chromatography column: Two 5 ml centrifuge chromatography tubes (Thermo Fisher Scientific, Inc., USA) were prepared, and 1 ml resin (Carboxylink™ Coupling Gel; Thermo Fisher Scientific, Inc., USA) was transferred into one of the tubes. The supernatants were removed using centrifugation at 1000 rpm for 20 s and washed with deionized water three times.
Linking the drug onto the column: 0.375 ml of 4 mg/ml sodium borohydride solution (Sangon Biotech Co., Ltd.) was added into the aforementioned two resin tubes, mixed in a shaker, and left to react for 4 h. The cover of the tube was opened every half an hour to release the gas generated in the reaction.
Affinity chromatography: The HL-1 cells were cultured in a 10-cm plate. The cell lysates were collected and loaded into the prepared chromatography resin tubes and incubated overnight at 4˚C. The proteins bound to the resin was separated using SDS-PAGE and identified using Coomassie brilliant blue staining and western blot analysis.
2.13 Statistical analysis
All the data are presented as the mean ± SD. Unpaired t-test (two tailed) was used when comparing two groups and GraphPad Prism v8 software. One-way ANOVA with Tukey’s post hoc test was used for multiple comparisons and SPSS v22 software. P<0.05 was considered to indicate a statistically significant difference.