Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 45(4); 2021 > Article
Original Article
Basic Research Role of Autophagy in Granulocyte-Colony Stimulating Factor Induced Anti-Apoptotic Effects in Diabetic Cardiomyopathy
Guang-Yin Shen1,2*orcid, Jeong-Hun Shin1*orcid, Yi-Sun Song3*orcid, Hyun-Woo Joo3, In-Hwa Park3, Jin-Hee Seong3, Na-Kyoung Shin3, A-Hyeon Lee3, Young Jong Cho4, Yonggu Lee1, Young-Hyo Lim1, Hyuck Kim5, Kyung-Soo Kim1,3orcid
Diabetes & Metabolism Journal 2021;45(4):594-605.
DOI: https://doi.org/10.4093/dmj.2020.0049
Published online: February 26, 2021
  • 7,064 Views
  • 147 Download
  • 3 Web of Science
  • 2 Crossref
  • 3 Scopus

1Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea

2Department of Cardiology, Jilin University Jilin Central Hospital, Jilin, China

3Graguate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea

4Laboratory Medicine, Kangwon National University School of Medicine, Chuncheon, Korea

5Department of Thoracic Surgery, Hanyang University College of Medicine, Seoul, Korea

Corresponding author: Kyung-Soo Kim orcid Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Korea E-mail: kskim@hanyang.ac.kr
*Guang-Yin Shen, Jeong-Hun Shin, and Yi-Sun Song contributed equally to this study as first authors.
• Received: February 28, 2020   • Accepted: September 27, 2020

Copyright © 2021 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Background
    We previously, reported that granulocyte-colony stimulating factor (G-CSF) reduces cardiomyocyte apoptosis in diabetic cardiomyopathy. However, the underlying mechanisms are not yet fully understood. Therefore, we investigated whether the mechanisms underlying of the anti-apoptotic effects of G-CSF were associated with autophagy using a rat model of diabetic cardiomyopathy.
  • Methods
    Diabetic cardiomyopathy was induced in rats through a high-fat diet combined with low-dose streptozotocin and the rats were then treated with G-CSF for 5 days. Rat H9c2 cardiac cells were cultured under high glucose conditions as an in vitro model of diabetic cardiomyopathy. The extent of apoptosis and protein levels related to autophagy (Beclin-1, microtubule-binding protein light chain 3 [LC3]-II/LC3-I ratio, and P62) were determined for both models. Autophagy determination was performed using an Autophagy Detection kit.
  • Results
    G-CSF significantly reduced cardiomyocyte apoptosis in the diabetic myocardium in vivo and led to an increase in Beclin-1 level and the LC3-II/LC3-I ratio, and decreased P62 level. Similarly, G-CSF suppressed apoptosis, increased Beclin-1 level and LC3-II/LC3-I ratio, and decreased P62 level in high glucose-induced H9c2 cardiac cells in vitro. These effects of G-CSF were abrogated by 3-methyladenine, an autophagy inhibitor. In addition, G-CSF significantly increased autophagic flux in vitro.
  • Conclusion
    Our results suggest that the anti-apoptotic effect of G-CSF might be significantly associated with the up-regulation of autophagy in diabetic cardiomyopathy.
Diabetes mellitus increases the risk of developing diabetic cardiomyopathy, a specific cardiomyopathy first described by Rubler et al. [1]. Diabetic cardiomyopathy leads to heart failure in patients with diabetes that is independent of hypertension and coronary artery disease, and is characterized by diastolic dysfunction, ventricular hypertrophy, myocardial fibrosis, and cardiomyocyte apoptosis [1,2]. Although the mechanisms are not fully understood, overwhelming evidence indicates that cardiomyocyte apoptosis plays an important role in the development of diabetic cardiomyopathy [3,4].
Autophagy is a self-degradative process that removes protein aggregates and damaged organelles, and is important for balancing the sources of energy in development as well as in response to nutrient stress [5]. The majority of studies have indicated that autophagy is the most important regulatory target of cell survival, and it plays an important role in the development and prognosis of heart disease [6,7]. Zou and Xie [8] demonstrated that diabetes induces cardiomyocyte apoptosis and suppresses cardiac autophagy in diabetic mice. Moreover, recent studies have demonstrated, in experimental and clinical settings, that cardiomyocyte apoptosis is correlated with the inhibition of autophagy induced by hyperglycemia [9,10].
Granulocyte-colony stimulating factor (G-CSF) is a growth factor that mediates the proliferation, differentiation, and survival of hematopoietic progenitor cells as well as the mobilization of bone marrow cells [11]. Recent studies indicated that G-CSF improves cardiac function both after myocardial infarction and in dilated cardiomyopathy [12,13]. We previously reported that G-CSF treatment of diabetic rats improved cardiac diastolic dysfunction and attenuated cardiomyocyte apoptosis [14,15]. Therefore, in this study, we explored whether the mechanisms underlying the anti-apoptotic effects of G-CSF were associated with autophagy using a rat model of diabetic cardiomyopathy.
Animals
Male Sprague-Dawley rats (Koatech, Pyeongtaek, Korea), aged 7 weeks and weighing 210 to 230 g, were used in this study. A combination of a high-fat diet (HFD, 60.3% of total calories come from fat, D12492; Research Diets Inc., New Brunswick, NJ, USA) and a low-dose of streptozotocin (STZ; Sigma-Aldrich, St. Louis, MO, USA) was effectively used to induce a rat model of diabetic cardiomyopathy [16]. The rats were maintained in a specific pathogen-free facility at the Hanyang University Medical School Animal Experiment Center under controlled conditions: temperature, 23°C±2°C; humidity, 55%±5%; and with an alternating 12-hour light/dark cycle. The experiments were performed in compliance with the animal research: reporting of in vivo experiments (ARRIVE) guidelines on animal research [17], and the research protocol was approved by the Hanyang University Institutional Animal Care and Use Committee (HY-IACUC-16-0107).
In vivo experimental design and drug treatment
The experimental design is outlined in Fig. 1. Seven-week-old rats were randomly assigned to one of two dietary regimens, either normal chow diet (n=6) or HFD (n=16) for an initial period of 7 weeks. After 6 weeks (at 13 weeks of age), the HFD group received a single intraperitoneal injection of STZ 30 mg/kg in 0.1 mmol/L citrate buffer, and the normal chow group received an injection of an equivalent volume of citrate buffer vehicle. One week later (at 14 weeks of age), fasting blood glucose (FBG) was measured, and rats with blood glucose level ≥200 mg/dL (11.1 mmol/L) were considered to have diabetes mellitus [18]. At 15 weeks of age, the diabetic group rats were randomly divided into two subgroups: diabetic rats treated with saline (n=8) and diabetic rats treated with G-CSF (n=8). Rats in the G-CSF treatment group were injected intraperitoneally with recombinant human G-CSF (200 μg/kg/day, Leucostim; Dong-A Pharmacological, Seoul, Korea) for 5 days. Rats in the normal chow group and the diabetic group treated with saline were injected intraperitoneally with an equivalent volume of saline for 5 days. At 18 weeks of age, all rats were euthanized for laboratory analysis.
Body weight and biochemical analysis
Body weight, FBG, total cholesterol (TC), triglyceride (TG), and fasting insulin levels were measured. Blood samples were collected from tail veins after 8 hours of fasting. Serum glucose, TC, and TG were measured using an Olympus AU400 auto analyzer (Olympus GmbH, Hamburg, Germany). Fasting insulin was measured using an EZRMI 13K kit (Millipore, St. Charles, Mo, USA) according to the manufacturer’s instructions. Insulin resistance was estimated by the homeostasis model assessment of insulin resistance (HOMA-IR), using the following formula: HOMA-IR=fasting insulin (μU/mL)× fasting plasma glucose (mmol/L)/22.5 [19].
Echocardiography
Echocardiography was performed pre- and post-treatment. The rats were anesthetized by intramuscular injection of a mixture of zoletil 50 (30 mg/kg; Virbac SA, Carros, France) and rompun (10 mg/kg; Bayer Korea, Seoul, Korea) [20]. Serial echocardiographic examinations (VIVID E9; GE Healthcare with a 12 probe, Chicago, IL, USA) were performed by a single sonographer, with the rats in the left lateral decubitus position; the left side of the chest was shaved in order to obtain a clear image. The measurements included left ventricular ejection fraction (LVEF), early peak velocity of the early diastolic filling wave (E), and early mitral annulus velocity during the diastolic phase (E´) [14]. All measurements were based on the mean of five consecutive cardiac cycles; mean values were used in analyses.
Detection of myocardial apoptosis by TUNEL assay
Apoptotic cells in the myocardium were detected by the terminal deoxynucleotidyl transferase (TDT)–mediated dUTP–biotin nick end–labeling (TUNEL) assay in paraffin sections using an In situ Cell Death Detection kit (Roche, Mannheim, Germany). The stained sections were photographed using a light microscope (Leica DM 4000B; Leica Microsystems, Wetzlar, Germany). Five regions from each digitized image were selected at random, and the numbers of healthy and TUNEL-positive (apoptotic) nuclei were quantified. The apoptotic index was calculated as the number of TUNEL-positive nuclei/total number of nuclei [15]. All data were evaluated by an independent blinded investigator.
Cell culture
H9c2 cardiac cells (ATCC, Manassas, VA, USA) were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Life Technologies, New York, NY, USA) containing 5.5 mM glucose, 1% fetal bovine serum (Life Technologies), and 1% penicillin and streptomycin (Life Technologies) at 37°C in a humidified incubator containing 5% CO2 [21]. When cells reached 60% confluence they were divided into six treatment groups: (1) incubation with DMEM containing 5.5 mM glucose (normal); (2) incubation with DMEM containing 45 mM glucose (high glucose [HG]); (3) incubation with HG DMEM supplemented with 3 μg/mL G-CSF; (4) incubation with HG DMEM supplemented with 3 μg/mL G-CSF and 5 mM 3-methyladenine (3-MA; an autophagy inhibitor; Sigma-Aldrich, St. Louis, MO, USA); (5) incubation with HG DMEM supplemented with 5 mM 3-MA; (6) incubation with HG DMEM supplemented with 50 nM rapamycin (Sigma-Aldrich), an autophagy activator. After 36 hours, cells were harvested for flow cytometry and Western blot analysis of autophagy-related proteins. All tests were repeated at least three times.
Western blot analysis
The halves of the hearts were homogenized, and total protein was extracted using protein lysis buffer (Pro-prep; iNtRON, Seongnam, Korea). Cardiac tissue samples containing 60 μg total protein and H9c2 cell extracts containing 10 μg total proteins were boiled for 10 minutes and loaded onto sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) gels (8% stacking and 10%, 15% separating gels). Separated proteins were transferred to nitrocellulose membranes (NC, 0.45 μm pore size; Bio-Rad, Hercules, CA, USA) or Iimmobilon-P transfer membrane (PVDF, 0.45 μm pore size; Millipore, Billerica, MD, USA). After blocking in 5% bovine serum albumin solution (Sigma-Aldrich) or 5% skim milk solution (BD Biosciences, San Diego, CA, USA) for 60 minutes, the membranes were incubated with primary antibody overnight at 4°C. The primary antibodies used are specified in Supplementary Table 1. Blots were incubated with horseradish peroxidase-conjugated anti-rabbit antibody (1:2,000; Jackson Immunoresearch, West Grove, IA, USA) or anti-mouse antibody (1:2,000; Jackson Immunoresearch) for 1 hour at room temperature. Glyceraldehyde-3-phosphate dehydrogenase was used as a protein loading control. Positive protein bands were visualized using an ECL kit (GenDEPOT, Barker, NY, USA), and results were quantified with an image analyzer (Image lab 3.0; Bio-Rad).
Flow cytometry
H9c2 cardiac cells were cultured in 6-well plates and incubated with the appropriate drugs. Cells were resuspended in 500 μL of 1×binding buffer with 2 μL of Annexin V fluorescein isothiocyanate (FITC) and 2 μL of propidium iodide (FITC Annexin V Apoptosis Detection Kit I; BD Biosciences), and incubated at room temperature for 15 minutes in the dark. Cells stained with Annexin V-FITC and/or propidium iodide were analyzed by flow cytometry [22]. Three independent experiments were conducted for each condition investigated, with 1×104 cells analyzed per experiment.
Autophagic flux detection assay
Autophagy determination was performed using an Autophagy Detection kit (ab139484; Abcam, Cambridge, UK) according to the manufacturer’s protocol [23-25]. Autophagy Assay Kit ab139484 measures autophagic vacuoles and monitors autophagic flux in live cells using a dye that selectively labels autophagic vacuoles. The green dye accumulates in autophagy vacuoles based on the pH present in the vacuole. Moreover, it is pH clamed for pre-autophagosomes, autophagosomes, and autophagolysosomes. The quantity of stained vesicles reflects the degree of autophagy in the cell population. For flow cytometry, after treatment, cells underwent trypsinization and were pelleted. Cells were centrifuged at 125 rcf for 7 minutes to pellet the cells, then washed with 1x Assay buffer, and thereafter incubated with 250 μL of the diluted green stain solution for 30 minutes at 37°C in the dark. For fluorescence microscopy, cells were on 24-well plates on coverslips. After treatment, the medium was removed, and cells washed with 1x Assay buffer, following incubation with 100 μL microscopy dual detection reagent for 30 minutes at 37°C in the dark.
Statistical analyses
SPSS version 22.0 software (IBM Co., Armonk, NY, USA) was used for statistical analyses. All data are expressed as mean±standard deviation, except for histological and echocardiology data, which are expressed as mean±standard error. Data were analyzed using one-way analysis of variance (ANOVA) analysis (for multiple comparisons), and post hoc multiple comparisons were made with Tukey’s test (equal variances assumed) or Dunnett’s T3 test (equal variances not assumed). P values less than 0.05 were considered significant.
Body weight and biochemical analysis
At the end of the experiment, there were no significant differences in body weight between the diabetic and normal rats. The diabetic rats showed significantly higher FBG, TC, and TG levels compared with normal rats. The diabetic rats treated with saline also showed significantly higher HOMA-IR levels than normal rats, but there was no significant difference in the HOMA-IR level between the diabetic rats treated with G-CSF and normal rats (Supplementary Table 2). These results confirmed the successful development of a diabetic rat model using a combination of HFD and low-dose STZ.
Effect of G-CSF on cardiac diastolic dysfunction
Echocardiography was performed to assess cardiac function, pre- and post-treatment. At pre-treatment, LVEF was preserved, but the E’ velocity was significantly lower and the E/E’ ratio was significantly higher in diabetic rats than in normal rats, suggesting that the diabetic rats developed diastolic dysfunction. At post-treatment, echocardiography revealed that the E’ velocity was significantly higher and the E/E’ ratio was significantly lower in diabetic rats treated with G-CSF than in diabetic rats treated with saline, whereas LVEF and E velocity did not differ significantly between groups. In addition, there was a significant increase in E’ velocity (2.53±0.51 cm/sec vs. 4.00±0.55 cm/sec, P<0.05) and decrease in E/E’ ratio (27.35±5.01 vs. 17.19±2.13, P<0.05) in diabetic rats treated with G-CSF compared with pre-treatment (Supplementary Table 3). G-CSF significantly reduced the extent of fibrosis in the myocardium in diabetic rats, as observed in our previous study (Supplementary Fig. 1) [14,17]. Taken together, these results demonstrated that G-CSF has an ameliorative effect on diastolic dysfunction in a rat model of diabetic cardiomyopathy.
Effect of G-CSF on cardiomyocyte apoptosis in cardiac tissue
The apoptotic index was significantly lower in diabetic rats treated with G-CSF than in diabetic rats treated with saline (25.12%±4.24% vs. 34.51%±3.93%, P<0.05). However, there was no significant difference in the apoptotic index between diabetic rats treated with G-CSF and normal rats (Fig. 2A and B). To understand the molecular basis of increased apoptosis in the myocardium of diabetic rats, we measured the level of anti-apoptotic protein, B-cell lymphoma 2 (Bcl-2). The Bcl-2 protein level was significantly higher in diabetic rats treated with G-CSF than in diabetic rats treated with saline (82.86%±14.76% vs. 52.99%±19.58%, P<0.05) (Fig. 2C and D). These results suggested that G-CSF has an anti-apoptotic effect on the diabetic myocardium.
Effect of G-CSF on autophagy in cardiac tissue
To clarify the effect of G-CSF on autophagy, we measured the cardiac Beclin-1 level, microtubule-binding protein light chain 3 (LC3)-II/LC3-I ratio, and P62 level, which are used as molecular markers of autophagy. Beclin-1 level was significantly higher in diabetic rats treated with G-CSF than in diabetic rats treated with saline (134.55%±25.46% vs. 70.08%±21.84%, P<0.05) (Fig. 3A and B). Moreover, the LC3-II/LC3-I ratio was also significantly higher (134.57±26.21 vs. 64.48±11.59, P<0.05) (Fig. 3A and C), whereas the P62 level was significantly lower (110.97%±13.85% vs. 169.56%±18.14%, P<0.05) (Fig. 3A and D), in diabetic rats treated with G-CSF than in diabetic rats treated with saline. These results suggest that the anti-apoptotic effect of G-CSF is significantly associated with upregulation of autophagy in the diabetic myocardium.
Effect of G-CSF on apoptosis in H9c2 cardiac cells
To clarify the effect of G-CSF on HG-induced apoptosis in H9c2 cardiac cells, we cultured H9c2 cardiac cells in HG media and measured the apoptosis rate by flow cytometry. HG media significantly increased the apoptosis rate of H9c2 cardiac cells compared with low glucose media (29.50%±3.90% vs. 16.50%±2.30%, P<0.05) (Fig. 4). Treatment with G-CSF significantly decreased the HG-induced apoptosis rate to 17.7% (Fig. 4). This effect was reversed to 33.1% by the autophagy inhibitor 3-MA (Fig. 4).
Effect of G-CSF on up-regulation of autophagic flux in H9c2 cardiac cells
To investigate the effect of G-CSF on autophagy, we measured the Beclin-1 protein level, the LC3-II/LC3-I ratio, and P62 level. Treatment with G-CSF significantly increased Beclin-1 level (75.00%±5.37% vs. 50.41%±7.86%, P<0.05) and the LC3-II/LC3-I ratio (119.36%±14.37% vs. 75.07%±5.41%, P<0.05) in H9c2 cardiac cells cultured in HG media (Fig. 5A-C); these increases were reduced upon treatment with 3-MA (36.99%±3.06% vs. 75.00%±5.37%, P<0.05; and 51.01%±7.41% vs. 119.36%±14.37%, P<0.05) (Fig. 5A-C). The levels of P62 in H9c2 cardiac cells was significantly lower in HG media supplemented with G-CSF alone (68.57%±5.31% vs. 134.46%±19.55%, P<0.05) (Fig. 5A and D), but was increased by 3-MA (68.57%±5.31% vs. 144.35%±5.40%, P<0.05) (Fig. 5A and D).
Furthermore, the apoptosis rate increased to 37.6% in H9c2 cardiac cells cultured in HG media supplemented with 3-MA, but decreased to 18.1% in cells cultured in HG media supplemented with rapamycin (Supplementary Fig. 2); this observation was consistent with the reduction in apoptosis in H9c2 cardiac cells induced by G-CSF under HG condition. In addition, the effect of G-CSF on autophagic flux in H9c2 cardiac cells was investigated using an Autophagy Detection kit. Using flow cytometry, we confirmed that treatment with G-CSF significantly increased autophagic flux (11.68% ±1.92% vs. 9.40%±0.45%, P<0.05) in H9c2 cardiac cells cultured in HG media (Fig. 6A and B). Additionally, using fluorescence microscopy, we showed that treatment with G-CSF increased the fluorescent signal, thus indicating enhanced autophagic flux in H9c2 cardiac cells cultured in HG media (Fig. 6C).
Taken together, these results also suggest that the anti-apoptotic effect of G-CSF in H9c2 cardiac cells under diabetic conditions is linked to the up-regulation of autophagy.
The present study demonstrated that G-CSF reduced cardiomyocyte apoptosis and up-regulated autophagy in the diabetic myocardium. In addition, the anti-apoptotic effect of G-CSF in H9c2 cardiac cells under diabetic conditions was counteracted by treatment with an autophagy inhibitor. Moreover, we confirmed that G-CSF increased autophagic flux in vitro. These data indicated that the anti-apoptotic effects of G-CSF in a rat model of diabetic cardiomyopathy may be mediated by upregulation of autophagy.
Cardiomyocyte apoptosis is an important mechanism in the development of diabetic cardiomyopathy, which is closely associated with cardiac dysfunction, hypertrophy, and fibrosis [26,27]. In addition, several studies revealed that the reduction of cardiomyocyte apoptosis prevents diabetic cardiomyopathy both in animal models and in vitro experiments [28,29]. In previous experimental studies, G-CSF treatment reduced cardiomyocyte apoptosis by modulating apoptosis-related proteins [15]; however, the mechanisms underlying this antiapoptotic effect of G-CSF in diabetic cardiomyopathy remain unclear.
Autophagy is an important mechanism for cell survival, as it maintains the quality of proteins and organelles [30]. Studies have suggested that down-regulation of autophagy and the resulting accumulation of abnormal proteins and organelles, leads to apoptosis and cardiac dysfunction in various cardiac diseases, such as ischemic heart disease, cardiac hypertrophy, and heart failure [31,32]. Sishi et al. [33] and Wang et al. [34] demonstrated that up-regulation of autophagy prevented cardiomyocyte apoptosis in doxorubicin-induced cardiomyopathy and hypertensive heart disease, respectively. Moreover, overwhelming evidence indicates that cardiomyocyte apoptosis, which plays an important role in the development of diabetic cardiomyopathy, is induced by the impairment of autophagy [35-37].
The major finding in this study was that G-CSF increased Beclin-1level and the LC3-II/LC3-I ratio and decreased P62 level in the diabetic myocardium. Beclin-1 is an essential inductor of autophagy activity that binds to class III phosphatidylinositol 3-kinase to form a kinase complex in mammals [38]. LC3 is the major regulatory protein that promotes the induction of the autophagosome membrane [39]. When autophagy is initiated, LC3-I is conjugated to phosphatidylethanolamine to form LC3-II, which is required for the formation of the autophagosome [40]. P62 is another major factor that targets specific cargo for autophagy; P62 accumulates when autophagy is inhibited, and its levels decrease when autophagy is induced [41]. Consistent with our data, Zhao et al. [42] reported that heme oxygenase-1 up-regulated the expression of Beclin-1 and LC3-II in diabetic mice and suggesting that heme oxygenase-1 prevents diabetic cardiomyopathy by up-regulation autophagy. Moreover, activation of AMP-activated protein kinase was shown to protect cardiac structure and function by up-regulation of Beclin-1 and LC3-II expression, suggesting that increasing cardiac autophagy protect would protect cardiac structure and function in the diabetic myocardium [43]. In this study we showed that G-CSF up-regulated cardiac autophagy, as indicated by the increase in Beclin-1 level and LC3-II/LC3-I ratio and decrease in P62 level in the diabetic myocardium.
In this study, to confirm the effect of G-CSF on HG-induced apoptosis in cardiac cells, we cultured H9c2 cardiac cells with HG media, to create a diabetic cardiomyopathy model [44], and measured the rate of apoptosis using these cells. H9c2 cardiac cells are a commercially available myogenic cell line derived from embryonic rat ventricular tissue [45], which show cardiac-specific characteristics, such as morphological, biochemical, and electrophysiological characterization [46]. H9c2 cardiac cells offer a unique in vitro model to study the metabolic activity of the heart [47]. Moreover, we used 3-MA (an autophagy inhibitor) and rapamycin (an autophagy inducer) to further confirm that apoptosis of H9c2 cardiac cells was related to autophagy under diabetic conditions. We found that G-CSF reduced apoptosis of H9c2 cardiac cells, concurrent with the up-regulation of autophagy; these effects were abrogated by 3-MA. We also confirmed that inhibition of autophagy by 3-MA increased H9c2 cardiac cell apoptosis under diabetic conditions, whereas up-regulation of autophagy by rapamycin reduced H9c2 cardiac cell apoptosis, under diabetic conditions. Jia et al. [48] previously showed that safflower extract reduced apoptosis of H9c2 cardiac cells treated with angiotensin II, by increasing LC3-II expression. They also reported that the anti-apoptotic effect of safflower was reversed by 3-MA and that rapamycin reduced apoptosis, suggesting that safflower inhibits apoptosis via the up-regulation of autophagy. Gao et al. [49] similarly reported that 3-MA abrogated the anti-apoptotic effect of fasudil, suggesting that fasudil protects H9c2 cardiac cells from apoptosis via increasing Beclin-1 level and LC3-II/LC3-I ratio and decreasing P62 level under diabetic conditions. Guo et al. [50] also demonstrated that G-CSF promoted autophagy and reduced neural tissue damage after spinal cord injury in mice. Inhibition of autophagy by 3-MA partially blocked the neuroprotective effect induced by G-CSF, suggesting that G-CSF reduced neural tissue damage through up-regulation of autophagy. Moreover, we confirmed that G-CSF reduced the up-regulated autophagic flux under diabetic conditions. Considering our data and previous studies, we suggest that the anti-apoptotic effect of G-CSF is potentially mediated by the up-regulation of autophagy in H9c2 cardiac cells, under diabetic condition.
This study does, however, have several limitations. First, we demonstrated the effects of 3-MA and rapamycin in in vitro diabetic condition experiments, but we did not confirm the systemic effects of 3-MA and rapamycin in diabetic rats. In addition, we did not perform genetic knockdown or gain-of-autophagy methods that are necessary to determine the functional role of autophagy in the anti-apoptotic effects of G-CSF and to rule out the potential nonspecific effects of 3-MA and rapamycin that are unrelated to autophagy. Second, we were unable to investigate the down-stream signaling of autophagy-related proteins such as Beclin-1, LC3, and P62. Additional studies are required to further elucidate the detailed mechanisms regarding effect of G-CSF on apoptosis linked to up-regulation of autophagy. Third, we cannot rule out the possibility that the anti-apoptotic effect of G-CSF is associated with any other previously postulated mechanism such as the action of G-CSF directly or through the G-CSF receptor-mediated signaling pathway, a systemic effect, mobilization or homing of bone marrow stem cells, or other paracrine effects; such as fibrosis, vascularization, oxidative stress, or endoplasmic reticulum stress. Further studies regarding the precise mechanism of the anti-apoptotic effect of G-CSF are also worth exploring.
In conclusion, the results of our study indicate that the antiapoptotic effect of G-CSF may be significantly associated with the up-regulation of autophagy in diabetic cardiomyopathy. These findings suggest that G-CSF could potentially be used a novel therapeutic drug for the treatment of patients with diabetic cardiomyopathy.
Supplementary materials related to this article can be found online at https://doi.org/10.4093/dmj.2020.0049.
Supplementary Table 1.
Primary antibodies for Western blotting analysis
dmj-2020-0049-suppl1.pdf
Supplementary Table 2.
Body weight and biochemical analysis
dmj-2020-0049-suppl2.pdf
Supplementary Table 3.
Echocardiographic data pre-treatment and post-treatment
dmj-2020-0049-suppl3.pdf
Supplementary Fig. 1.
Effect of granulocyte-colony stimulating factor (G-CSF) on myocardial fibrosis in diabetic myocardium. (A, B, C) Representative images of perivascular and interstitial fibrosis of rat myocardium demonstrated by blue Masson’s trichrome staining in each group (magnification ×400). (A) Normal rats, (B) diabetic rats treated with saline, (C) diabetic rats treated with G-CSF. (D) Quantitative analysis of the percentage of fibrosis area in the myocardium of each group. All data are expressed as mean±standard error of the mean. aP<0.05 vs. normal group, bP<0.05 vs. saline group (n=6–8 per group).
dmj-2020-0049-suppl4.pdf
Supplementary Fig. 2.
Effect of autophagy in high glucose-induced H9c2 cardiac cells. (A) Dot plots display the stages of apoptotic death of H9c2 cardiac cells: Annexin−/PI− (Q3), viable cells; Annexin+/PI− (Q4), cells undergoing apoptosis; Annexin+/PI+ (Q2), cells that are in end-stage apoptosis or are already dead; Annexin−/PI+ (Q1), cells that are in necrosis. (a) H9c2 cardiac cells cultured in low glucose media; (b) H9c2 cardiac cells cultured in high glucose media; (c) H9c2 cardiac cells cultured in high glucose media containing 3-methyladenine (3-MA; 5 mM); (d) H9c2 cardiac cells cultured in high glucose media containing rapamycin (RAPA; 50 nM). (B) Quantitative analysis of apoptotic cells (Q2+Q4). All data are expressed as mean±standard deviation. FITC, fluorescein isothiocyanate. aP<0.05 vs. H9c2 cardiac cells cultured in low glucose media, bP<0.05 vs. H9c2 cardiac cells cultured in high glucose media, cP<0.05 vs. H9c2 cardiac cells cultured in high glucose media containing 3-MA (n=5 per group).
dmj-2020-0049-suppl5.pdf

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

AUTHOR CONTRIBUTIONS

Conception or design: G.Y.S., J.H.S., Y.S.S.

Acquisition, analysis, or interpretation of data: G.Y.S., Y.S.S.,

H.W.J., I.H.P., J.H.S., N.K.S., A.H.L., Y.J.C.

Drafting the work or revising: Y.L., Y.H.L., H.K.

Final approval of the manuscript: K.S.K.

FUNDING

None

Acknowledgements
This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education (2016R1D1A1-B03931479).
Fig. 1.
Scheme of the animal experiment. Diabetes was induced in rats by feeding for 7 weeks with a high-fat diet and low-dose streptozotocin (30 mg/kg) injection. Rat were then randomized for treatment with granulocyte-colony stimulating factor (G-CSF) or saline administrated intraperitoneally, for 5 days. Body weight, biochemical analysis, and echocardiography were performed both pre- and post-treatment. At 18 weeks of age, all rats were euthanized for histology and protein analysis. SD, Sprague-Dawley.
dmj-2020-0049f1.jpg
Fig. 2.
Effect of granulocyte-colony stimulating factor (G-CSF) on myocardial apoptosis in the diabetic myocardium. (A) Representative images of terminal deoxynucleotidyl transferase (TDT)–mediated dUTP–biotin nick end–labeling (TUNEL) assay staining of myocardium for each group 4 weeks after treatment (magnification ×400). Apoptotic nuclei are stained brown and non-apoptotic nuclei are stained blue on TUNEL assay staining. (B) Quantitative analysis of apoptotic cells in the myocardium of each group. (C) Level of B-cell lymphoma 2 (Bcl-2) protein in cardiac tissue was detected by Western blotting. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a loading control. (D) Quantitative Western blot analysis of Bcl-2. The expression level was normalized by comparison with GAPDH expression. Protein levels are expressed as mean±standard deviation. Histology data are expressed as mean±standard error. aP<0.05 vs. normal group, bP<0.05 vs. saline group (n=6–8 per group).
dmj-2020-0049f2.jpg
Fig. 3.
Effect of granulocyte-colony stimulating factor (G-CSF) on autophagy in the diabetic myocardium. (A) Representative images showing the levels of autophagy-related proteins Beclin-1, the microtubule-binding protein light chain 3 (LC3)-II/LC3-I ratio, and P62 in the diabetic myocardium measured by Western blot at 18 weeks of age (4 weeks after G-CSF and saline treatment). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a loading control. (B, C, D) Quantitative Western blot analysis of Beclin-1, the LC3-II/LC3-I ratio, and P62. Protein levels were normalized by comparison with GAPDH expression. All data are expressed as mean±standard deviation. aP<0.05 vs. normal group, bP<0.05 vs. saline group (n=6–8 per group).
dmj-2020-0049f3.jpg
Fig. 4.
Effect of granulocyte-colony stimulating factor (G-CSF) on apoptosis in high glucose-induced H9c2 cardiac cells. (A) Dot plots displaying the stages of apoptotic death of H9c2 cardiac cells: Annexin−/PI− (Q3), viable cells; Annexin+/PI− (Q4), cells undergoing apoptosis; Annexin+/PI+ (Q2), cells in end-stage apoptosis or that are already dead; Annexin−/PI+ (Q1), cells that are in necrosis. (a) H9c2 cardiac cells cultured in low glucose media; (b) H9c2 cardiac cells cultured in high glucose media; (c) H9c2 cardiac cells cultured in high glucose media containing G-CSF (3 μg/mL); (d) H9c2 cardiac cells cultured in high glucose media containing G-CSF (3 μg/mL) and 3-methyladenine (3-MA; 5 mM). (B) Quantitative analysis of apoptotic cells (Q2+Q4). All data are expressed as mean±standard deviation. FITC, fluorescein isothiocyanate. aP<0.05 vs. H9c2 cardiac cells cultured in low glucose media, bP<0.05 vs. H9c2 cardiac cells cultured in high glucose media, cP<0.05 vs. H9c2 cardiac cells cultured in high glucose media containing G-CSF (n=5 per group).
dmj-2020-0049f4.jpg
Fig. 5.
Effect of granulocyte-colony stimulating factor (G-CSF) on autophagy in high glucose-induced H9c2 cardiac cells. (A) Representative images of levels of autophagy-related proteins Beclin-1, the microtubule-binding protein light chain 3 (LC3)-II/LC3-I ratio, and P62 in H9c2 cardiac cells measured by Western blot. (B, C, D) Quantitative Western blot analysis of Beclin-1, the LC3-II/LC3-I ratio, and P62. Protein levels were normalized by comparison with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) expression. GAPDH was used as a loading control. All data are expressed as mean±standard deviation. 3-MA, 3-methyladenine. aP<0.05 vs. H9c2 cardiac cells cultured in low glucose media, bP<0.05 vs. H9c2 cardiac cells cultured in high glucose media, cP<0.05 vs. H9c2 cardiac cells cultured in high glucose media containing G-CSF (n=5 per group).
dmj-2020-0049f5.jpg
Fig. 6.
Effect of granulocyte-colony stimulating factor (G-CSF) on autophagic flux in high glucose-induced H9c2 cardiac cells. Autophagic flux evaluated high glucose-induced H9c2 cardiac cells by flow cytometry and fluorescence microscopy. Representative histogram (A) and bar graph (B), where fluorescence increase of autophagy green indicates autophagic flux increase. (C) Autophagic flux evaluated by fluorescence microscopy (magnification ×400), photos are representative of four to five independent experiments. Arrows indicate autophagic vesicles. The inserts in (C) show higher magnification. Rapamycin was used as a positive control of autophagy. All data are expressed as mean±standard deviation. Normal, normal condition group. aP<0.05 vs. H9c2 cardiac cells cultured in normal condition, bP<0.05 vs. H9c2 cardiac cells cultured in high glucose media containing G-CSF (n=4–5 per group).
dmj-2020-0049f6.jpg
dmj-2020-0049f7.jpg
  • 1. Rubler S, Dlugash J, Yuceoglu YZ, Kumral T, Branwood AW, Grishman A. New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am J Cardiol 1972;30:595-602.ArticlePubMed
  • 2. Acar E, Ural D, Bildirici U, Sahin T, Yilmaz I. Diabetic cardiomyopathy. Anadolu Kardiyol Derg 2011;11:732-7.ArticlePubMed
  • 3. Cai L, Li W, Wang G, Guo L, Jiang Y, Kang YJ. Hyperglycemiainduced apoptosis in mouse myocardium: mitochondrial cytochrome C-mediated caspase-3 activation pathway. Diabetes 2002;51:1938-48.PubMed
  • 4. Frustaci A, Kajstura J, Chimenti C, Jakoniuk I, Leri A, Maseri A, et al. Myocardial cell death in human diabetes. Circ Res 2000;87:1123-32.ArticlePubMed
  • 5. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol 2010;221:3-12.ArticlePubMedPMC
  • 6. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008;132:27-42.ArticlePubMedPMC
  • 7. Orogo AM, Gustafsson AB. Therapeutic targeting of autophagy: potential and concerns in treating cardiovascular disease. Circ Res 2015;116:489-503.ArticlePubMedPMC
  • 8. Zou MH, Xie Z. Regulation of interplay between autophagy and apoptosis in the diabetic heart: new role of AMPK. Autophagy 2013;9:624-5.ArticlePubMedPMC
  • 9. Hsu HC, Chen CY, Lee BC, Chen MF. High-fat diet induces cardiomyocyte apoptosis via the inhibition of autophagy. Eur J Nutr 2016;55:2245-54.ArticlePubMedPDF
  • 10. Mellor KM, Reichelt ME, Delbridge LM. Autophagy anomalies in the diabetic myocardium. Autophagy 2011;7:1263-7.ArticlePubMed
  • 11. Demetri GD, Griffin JD. Granulocyte colony-stimulating factor and its receptor. Blood 1991;78:2791-808.ArticlePubMedPDF
  • 12. Deindl E, Zaruba MM, Brunner S, Huber B, Mehl U, Assmann G, et al. G-CSF administration after myocardial infarction in mice attenuates late ischemic cardiomyopathy by enhanced arteriogenesis. FASEB J 2006;20:956-8.ArticlePubMedPDF
  • 13. Huttmann A, Duhrsen U, Stypmann J, Noppeney R, Nuckel H, Neumann T, et al. Granulocyte colony-stimulating factor-induced blood stem cell mobilisation in patients with chronic heart failure: feasibility, safety and effects on exercise tolerance and cardiac function. Basic Res Cardiol 2006;101:78-86.ArticlePubMedPDF
  • 14. Lim YH, Joe JH, Jang KS, Song YS, So BI, Fang CH, et al. Effects of granulocyte-colony stimulating factor (G-CSF) on diabetic cardiomyopathy in Otsuka Long-Evans Tokushima fatty rats. Cardiovasc Diabetol 2011;10:92.ArticlePubMedPMCPDF
  • 15. Shin JH, Lim YH, Song YS, So BI, Park JY, Fang CH, et al. Granulocyte-colony stimulating factor reduces cardiomyocyte apoptosis and ameliorates diastolic dysfunction in Otsuka Long-Evans Tokushima Fatty rats. Cardiovasc Drugs Ther 2014;28:211-20.ArticlePubMedPDF
  • 16. Srinivasan K, Viswanad B, Asrat L, Kaul CL, Ramarao P. Combination of high-fat diet-fed and low-dose streptozotocintreated rat: a model for type 2 diabetes and pharmacological screening. Pharmacol Res 2005;52:313-20.ArticlePubMed
  • 17. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol 2010;8:e1000412.ArticlePubMedPMC
  • 18. Ti Y, Xie GL, Wang ZH, Bi XL, Ding WY, Wang J, et al. TRB3 gene silencing alleviates diabetic cardiomyopathy in a type 2 diabetic rat model. Diabetes 2011;60:2963-74.ArticlePubMedPMCPDF
  • 19. Song YS, Fang CH, So BI, Park JY, Lee Y, Shin JH, et al. Time course of the development of nonalcoholic Fatty liver disease in the Otsuka long-evans Tokushima Fatty rat. Gastroenterol Res Pract 2013;2013:342648.ArticlePubMedPMCPDF
  • 20. Song YS, Joo HW, Park IH, Shen GY, Lee Y, Shin JH, et al. Transplanted human amniotic epithelial cells secrete paracrine proangiogenic cytokines in rat model of myocardial infarction. Cell Transplant 2015;24:2055-64.ArticlePubMedPDF
  • 21. Liu L, Ding WY, Zhao J, Wang ZH, Zhong M, Zhang W, et al. Activin receptor-like kinase 7 mediates high glucose-induced H9c2 cardiomyoblast apoptosis through activation of Smad2/3. Int J Biochem Cell Biol 2013;45:2027-35.ArticlePubMed
  • 22. Song YS, Joo HW, Park IH, Shen GY, Lee Y, Shin JH, et al. Bone marrow mesenchymal stem cell-derived vascular endothelial growth factor attenuates cardiac apoptosis via regulation of cardiac miRNA-23a and miRNA-92a in a rat model of myocardial infarction. PLoS One 2017;12:e0179972.ArticlePubMedPMC
  • 23. Sharif T, Martell E, Dai C, Kennedy BE, Murphy P, Clements DR, et al. Autophagic homeostasis is required for the pluripotency of cancer stem cells. Autophagy 2017;13:264-84.ArticlePubMed
  • 24. Almasi S, Kennedy BE, El-Aghil M, Sterea AM, Gujar S, Partida-Sanchez S, et al. TRPM2 channel-mediated regulation of autophagy maintains mitochondrial function and promotes gastric cancer cell survival via the JNK-signaling pathway. J Biol Chem 2018;293:3637-50.ArticlePubMedPMC
  • 25. Perez-Arizti JA, Ventura-Gallegos JL, Galvan Juarez RE, Ramos-Godinez MDP, Colin-Val Z, Lopez-Marure R. Titanium dioxide nanoparticles promote oxidative stress, autophagy and reduce NLRP3 in primary rat astrocytes. Chem Biol Interact 2020;317:108966.ArticlePubMed
  • 26. Fang ZY, Prins JB, Marwick TH. Diabetic cardiomyopathy: evidence, mechanisms, and therapeutic implications. Endocr Rev 2004;25:543-67.ArticlePubMed
  • 27. Huynh K, Bernardo BC, McMullen JR, Ritchie RH. Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther 2014;142:375-415.ArticlePubMed
  • 28. Fiordaliso F, Li B, Latini R, Sonnenblick EH, Anversa P, Leri A, et al. Myocyte death in streptozotocin-induced diabetes in rats in angiotensin II-dependent. Lab Invest 2000;80:513-27.PubMed
  • 29. Li K, Cui YC, Zhang H, Liu XP, Zhang D, Wu AL, et al. Glutamine reduces the apoptosis of H9C2 cells treated with high-glucose and reperfusion through an oxidation-related mechanism. PLoS One 2015;10:e0132402.ArticlePubMedPMC
  • 30. Lavandero S, Chiong M, Rothermel BA, Hill JA. Autophagy in cardiovascular biology. J Clin Invest 2015;125:55-64.ArticlePubMedPMC
  • 31. Yan L, Vatner DE, Kim SJ, Ge H, Masurekar M, Massover WH, et al. Autophagy in chronically ischemic myocardium. Proc Natl Acad Sci U S A 2005;102:13807-12.ArticlePubMedPMC
  • 32. Zhu H, Rothermel BA, Hill JA. Autophagy in load-induced heart disease. Methods Enzymol 2009;453:343-63.PubMedPMC
  • 33. Sishi BJ, Loos B, van Rooyen J, Engelbrecht AM. Autophagy upregulation promotes survival and attenuates doxorubic-ininduced cardiotoxicity. Biochem Pharmacol 2013;85:124-34.ArticlePubMed
  • 34. Wang ZV, Rothermel BA, Hill JA. Autophagy in hypertensive heart disease. J Biol Chem 2010;285:8509-14.ArticlePubMedPMC
  • 35. Cai L, Kang YJ. Cell death and diabetic cardiomyopathy. Cardiovasc Toxicol 2003;3:219-28.ArticlePubMed
  • 36. Xu X, Hua Y, Nair S, Zhang Y, Ren J. Akt2 knockout preserves cardiac function in high-fat diet-induced obesity by rescuing cardiac autophagosome maturation. J Mol Cell Biol 2013;5:61-3.ArticlePubMed
  • 37. Xu X, Kobayashi S, Chen K, Timm D, Volden P, Huang Y, et al. Diminished autophagy limits cardiac injury in mouse models of type 1 diabetes. J Biol Chem 2013;288:18077-92.ArticlePubMedPMC
  • 38. Kang R, Zeh HJ, Lotze MT, Tang D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ 2011;18:571-80.ArticlePubMedPMCPDF
  • 39. Kobayashi S, Liang Q. Autophagy and mitophagy in diabetic cardiomyopathy. Biochim Biophys Acta 2015;1852:252-61.ArticlePubMed
  • 40. Cherra SJ 3rd, Kulich SM, Uechi G, Balasubramani M, Mountzouris J, Day BW, et al. Regulation of the autophagy protein LC3 by phosphorylation. J Cell Biol 2010;Aug;190:533-9.ArticlePubMedPMCPDF
  • 41. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, Johansen T. Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol 2009;452:181-97.PubMed
  • 42. Zhao Y, Zhang L, Qiao Y, Zhou X, Wu G, Wang L, et al. Heme oxygenase-1 prevents cardiac dysfunction in streptozotocin-diabetic mice by reducing inflammation, oxidative stress, apoptosis and enhancing autophagy. PLoS One 2013;8:e75927.ArticlePubMedPMC
  • 43. Xie Z, Lau K, Eby B, Lozano P, He C, Pennington B, et al. Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 mice. Diabetes 2011;60:1770-8.ArticlePubMedPMCPDF
  • 44. Song H, Zandstra PW, Radisic M. Engineered heart tissue model of diabetic myocardium. Tissue Eng Part A 2011;17:1869-78.ArticlePubMed
  • 45. Kimes BW, Brandt BL. Properties of a clonal muscle cell line from rat heart. Exp Cell Res 1976;98:367-81.ArticlePubMed
  • 46. Hescheler J, Meyer R, Plant S, Krautwurst D, Rosenthal W, Schultz G. Morphological, biochemical, and electrophysiological characterization of a clonal cell (H9c2) line from rat heart. Circ Res 1991;69:1476-86.ArticlePubMed
  • 47. Watkins SJ, Borthwick GM, Arthur HM. The H9C2 cell line and primary neonatal cardiomyocyte cells show similar hypertrophic responses in vitro. In Vitro Cell Dev Biol Anim 2011;47:125-31.ArticlePubMedPDF
  • 48. Jia Z, Liu Y, Su H, Li M, Zhang M, Zhu Y, et al. Safflower extract inhibiting apoptosis by inducing autophagy in myocardium derived H9C2 cell. Int J Clin Exp Med 2015;8:20254-62.PubMedPMC
  • 49. Gao H, Hou F, Dong R, Wang Z, Zhao C, Tang W, et al. RhoKinase inhibitor fasudil suppresses high glucose-induced H9c2 cell apoptosis through activation of autophagy. Cardiovasc Ther 2016;34:352-9.ArticlePubMed
  • 50. Guo Y, Liu S, Zhang X, Wang L, Gao J, Han A, et al. G-CSF promotes autophagy and reduces neural tissue damage after spinal cord injury in mice. Lab Invest 2015;95:1439-49.ArticlePubMedPDF

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Ginkgo biloba extract protects against diabetic cardiomyopathy by restoring autophagy via adenosine monophosphate‐activated protein kinase/mammalian target of the rapamycin pathway modulation
      Xueyao Yang, Xin Zhao, Yanfei Liu, Yue Liu, Libo Liu, Ziyu An, Haoran Xing, Jinfan Tian, Xiantao Song
      Phytotherapy Research.2023; 37(4): 1377.     CrossRef
    • Perspectives for Forkhead box transcription factors in diabetic cardiomyopathy: Their therapeutic potential and possible effects of salvianolic acids
      Ronghui Han, Hemeng Huang, Weiyi Xia, Jingjin Liu, Hui Luo, Jing Tang, Zhengyuan Xia
      Frontiers in Cardiovascular Medicine.2022;[Epub]     CrossRef

    • PubReader PubReader
    • ePub LinkePub Link
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Role of Autophagy in Granulocyte-Colony Stimulating Factor Induced Anti-Apoptotic Effects in Diabetic Cardiomyopathy
      Diabetes Metab J. 2021;45(4):594-605.   Published online February 26, 2021
      Close
    • XML DownloadXML Download
    Figure

    Diabetes Metab J : Diabetes & Metabolism Journal