Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
2,319
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      CVIA now indexed by SCOPUS from February 2024. CVIA received its first Journal Impact Factor (0.5) in the 2023 Journal Citation Reports Release. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Berberine Ameliorates Diabetic Cardiomyopathy in Mice by Decreasing Cardiomyocyte Apoptosis and Oxidative Stress

      Published
      research-article
      Bookmark

            Abstract

            Background: Diabetic cardiomyopathy is a multifaceted complication of diabetes that lacks effective treatments. Berberine (BBR), a bioactive compound from Rhizoma coptidis, has potential therapeutic implications, but its precise role in diabetic cardiomyopathy remains to be defined.

            Methods: In this study, a diabetic cardiomyopathy model was established by administration of a high-fat diet and streptozotocin injection to C57BL/6J mice. Concurrently, the mice received BBR treatment daily for a duration of 8 weeks. After the treatment period, myocardial injury, cardiac function, and the levels of oxidative stress and apoptosis were assessed.

            Results: BBR significantly ameliorated cardiac dysfunction and histopathological damage caused by diabetic cardiomyopathy. This treatment also elevated serum superoxide dismutase levels while decreasing malondialdehyde levels. The anti-apoptotic activity of BBR was evidenced by a decrease in TUNEL-positive cells and the percentage of apoptotic cells, as determined by flow cytometry, in conjunction with diminished levels of BCL2-associated X protein/B cell lymphoma 2 (BAX/BCL2) in heart tissues. Mechanistically, BBR was found to ameliorate diabetic cardiomyopathy by upregulating the expression of myocardial methionine sulfoxide reductase A (MsrA) and concurrently suppressing cardiac CaMKII oxidation.

            Conclusions: BBR alleviates diabetic cardiomyopathy by inhibiting myocardial apoptosis and oxidative stress through the MsrA and CaMKII signaling pathways.

            Main article text

            Background

            Diabetic cardiomyopathy (DCM), a major complication of diabetes, substantially contributes to the global public health burden [1, 2]. This multifaceted condition is a distinct clinical entity characterized by myocardial dysfunction and abnormal cardiac metabolism in people with diabetes, independently of coronary artery disease and hypertension [3]. The pathogenesis of DCM is a multifactorial process in which oxidative stress and apoptosis play crucial roles.

            Oxidative stress, characterized by an imbalance between the accumulation of reactive oxygen species (ROS) and the body’s antioxidative defenses, is exacerbated under diabetic conditions [4]. The elevated glucose in diabetes leads to excessive production of ROS. These reactive molecules consequently inflict considerable damage on crucial cellular components, including lipids, proteins, and DNA. This damage culminates in cell death through a process known as apoptosis, thus providing a critical pathophysiological link in the progression of DCM [5, 6].

            Investigations of the molecular mechanisms underlying DCM have focused on two key proteins: methionine sulfoxide reductase A (MsrA) and calcium/calmodulin-dependent protein kinase II (CaMKII). MsrA, an antioxidant enzyme, repairs proteins damaged by oxidative stress, thus preserving cellular function under adverse conditions [5, 6]. In contrast, CaMKII, particularly when oxidatively modified (ox-CaMKII), has been implicated in DCM pathogenesis by promoting myocardial apoptosis and fibrosis, thereby impairing cardiac function [7, 8]. Emerging evidence highlights a role of MsrA in mitigating ox-CaMKII activation in cardiovascular diseases, thereby suggesting a protective effect. However, the relevance of this pathway to DCM remains to be explored further [9].

            Berberine (BBR), an isoquinoline alkaloid extracted from Rhizoma coptidis, has received considerable attention for its potential cardioprotective properties. Accumulating evidence highlights its beneficial effects in alleviating oxidative stress, inflammation, and apoptosis in cardiovascular diseases [10, 11]. However, the specific role and underlying mechanisms of BBR in the context of DCM remain elusive.

            Therefore, the current study was aimed at exploring the effects and mechanistic action of BBR in DCM, specifically focusing on its influence on MsrA expression and cardiac CaMKII oxidation.

            Methods

            Mouse Model of Diabetic Cardiomyopathy

            The DCM model in mice was established as previously described [12]. The experimental animals were purchased from Beijing Weitong Lihua Experimental Animal Technology Co., Ltd., and the animal experiment was approved by the Ethics Committee of Tianjin First Central Hospital. Male C57 mice 8 weeks of age were divided into control, DCM, and BBR groups. Mice in the control group were maintained on a normal chow diet throughout the study. Mice in the DCM and BBR groups were subjected to a high-fat diet (45% fat, 0.25% cholesterol) for 6 weeks, then tested for glucose intolerance and insulin resistance. At 14 weeks, both the DCM and BBR groups exhibited elevated blood glucose levels after glucose and insulin injection, thus indicating impaired glucose tolerance and insulin sensitivity. Diabetes was induced with streptozotocin (75 mg/kg, Sigma, USA) injection. Mice with fasting blood glucose levels ≥11.1 mM were included in the DCM and BBR groups. Both the DCM and BBR groups continued to receive the high-fat diet for an additional 15 weeks, for a total of 22 weeks. After establishment of the DCM model, mice in the DCM group were administered physiological saline, whereas those in the BBR group were administered BBR (20 mg/kg × day) via gavage for 8 weeks. All mice were monitored until 30 weeks.

            Echocardiographic Measurement

            Mice were anesthetized with isoflurane, after which transthoracic echocardiography was conducted with a Vevo2100 imaging system (VisualSonics, Toronto, Canada). This procedure was performed by echocardiographers who were blinded to the group assignments of the mice. Measurements of left ventricular end-diastolic diameter (LVEDD), left ventricular ejection fraction (LVEF), fractional shortening (FS), left ventricular posterior wall thickness (LVPW), and interventricular septum thickness (IVS) were obtained through M-mode echocardiography in the left ventricular long-axis view. Pulsed Doppler imaging in four-chamber view was used to measure the early (E) and late (A) diastolic mitral flow velocities, from which the E/A ratio was calculated. Tissue Doppler imaging in the four-chamber view was used to measure the early (E´) and late (A´) diastolic mitral annular velocities, and the E´/A´ ratio was subsequently determined.

            Histology

            Heart weight (HW) and tibial length (TL) were measured in all mice and used to calculate the HW/TL ratio. Hearts were freshly excised and immediately fixed in 10% formalin. After fixation, the tissues were embedded in paraffin and sectioned into 4 μm thick slices. Hematoxylin and eosin (HE) staining was performed to visualize cardiomyocyte morphology. Masson’s trichrome staining was used to detect myocardial fibrosis, and the fibrotic area was calculated as a percentage of the total myocardial area. Wheat germ agglutinin staining was conducted to delineate the cell boundaries of cardiomyocytes, thus enabling calculation of the cross-sectional area of the cardiomyocytes.

            TUNEL Staining

            Myocardial sections (4 μm thick) underwent terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining. After deparaffinization and antigen retrieval with citrate buffer, sections were treated with 3% H2O2 for 10 min and blocked with 5% BSA for 30 min at 37 °C. Apoptosis was detected with an In Situ Cell Death Detection Kit and TMR red (Roche, Germany). After being washed with PBS, sections were stained with 4′,6-diamidino-2-phenylindole (DAPI) for 10 min at 37 °C. Images were captured with a fluorescence microscope (Ni-E, Nikon, Japan). The apoptosis ratio, expressed as the percentage of apoptotic cells to total myocardial cells, was calculated.

            Flow Cytometry Analysis of Cardiomyocyte Apoptosis

            Freshly excised mouse hearts were subjected to enzymatic digestion to isolate cardiomyocytes. The isolated cells were then stained with an apoptosis detection kit specifically designed for flow cytometry. The staining procedure involved a combination of fluorescently labeled annexin V and a propidium iodide solution. After staining, the cells were analyzed with a flow cytometer set up to detect the fluorescence of annexin V and propidium iodide. The flow cytometry data were analyzed with appropriate software to determine the percentage of apoptotic cells, calculated as the number of annexin V positive cells (early and late apoptotic cells) divided by the total number of cells.

            Detection of Superoxide Dismutase and Malondialdehyde in Mouse Blood

            Fresh blood samples were collected from the mice and immediately processed for the detection of superoxide dismutase (SOD) and malondialdehyde (MDA). For SOD detection, blood samples were subjected to a specific SOD assay measuring the dismutation of superoxide radicals, performed according to the manufacturer’s instructions. For MDA detection, a Bioxytech LPO-586 assay kit was used. This assay is based on the reaction of MDA, a biomarker of lipid peroxidation, with N-methyl-2-phenylindole, thus forming a stable chromophoric product. The absorbance of this product was measured at 586 nm with a spectrophotometer. The MDA concentration was calculated with a standard curve according to the manufacturer’s instructions.

            Western Blot Analysis

            Protein extracts from mouse hearts and cells were subjected to SDS-PAGE separation and subsequently transferred onto polyvinylidene fluoride membranes. The membranes were then incubated with various primary antibodies at 4 °C overnight and subsequently incubated with suitable secondary antibodies for 1 hour at room temperature. The relative protein expression levels were determined by normalization to GAPDH expression. This experiment used primary antibodies to BCL2-associated X protein (BAX) (Proteintech, 50599-2-Ig, 1:2000), B-cell lymphoma 2 (Bcl2) (Proteintech, 68103-1-Ig, 1:2000), MsrA (Proteintech, 14547-1-AP, 1:500), CAMKII (Proteintech, 12666-2-AP, 1:1000), and ox-CAMKII (Gene Tex, AB_2887447, 1:1000).

            Statistical Analysis

            The statistical results for all figures and tables are presented as mean ± SD. GraphPad Prism software (version 7.0, CA) was used for one-way or two-way ANOVA with post hoc Tukey’s test or unpaired test (two-tailed), when appropriate, or to calculate the Pearson correlation coefficient and the P-value (two tailed). Differences were considered significant at P < 0.05.

            Results

            Body Weight, Blood Glucose, and Serum Lipid Profiles

            Body weight (BW), fasting blood glucose (FBG), total cholesterol (TC), and triglycerides (TG) were significantly higher in the DCM group than the control group. However, no significant differences in BW, FBG, and serum lipid levels were observed between the BBR group and DCM group (Table 1). These findings suggested that the introduction of BBR did not significantly alter these metabolic parameters in the context of DCM.

            Table 1

            Body Weight, Blood Glucose, and Serum Lipid Profiles in Mice.

            Control (n = 6)DCM (n = 6)BBR (n = 6)
            BW (g)26.65 ± 1.2635.61 ± 2.31*34.65 ± 1.85
            FBG (mmol/L)7.51 ± 1.2129.41 ± 2.13*28.24 ± 5.21
            TC (mmol/L)3.15 ± 0.974.12 ± 1.04*4.31 ± 0.81
            TG (mmol/L)1.55 ± 0.281.99 ± 0.35*2.01 ± 0.42
            HDL-C (mmol/L)0.82 ± 0.620.80 ± 0.320.99 ± 0.25
            LDL-C (mmol/L)2.18 ± 0.232.34 ± 0.562.21 ± 0.47

            Data are shown as mean ± SD; *P < 0.05 vs. control group. BW, body weight; FBG, fasting blood glucose; TC, total cholesterol; TG, triglycerides; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol.

            Improved Cardiac Remodeling and Function in Berberine-Treated Diabetic Mice

            Echocardiographic evaluations indicated notable differences in cardiac remodeling and function among groups. In the DCM group, the LVEDD, LVPW, and IVS were significantly greater than those in the control group. In contrast, the LVEF, FS, E/A ratio, and E´/A´ ratio were significantly lower in the DCM group than the control group (Figure 1B–I).

            Figure 1

            Experiment timeline and echocardiographic measurements in three groups of mice.

            (A) Experiment timeline in three groups of mice. (B) Representative echocardiographic images in mice: (1) Two-dimensional echocardiogram showing left ventricular short-axis view; (2) M-mode echocardiogram showing left ventricular dimensions; (3) Pulse-wave Doppler echocardiogram depicting diastolic mitral flow; (4) Tissue Doppler echocardiogram displaying mitral annular velocities. (C) Measurements of left ventricular ejection fraction (LVEF) in mice. (D) Measurements of left ventricular fractional shortening (FS) in mice. (E) Measurements of left ventricular posterior wall (LVPW) thickness in mice. (F) Measurements of interventricular septum (IVS) thickness in mice. (G) Measurements of the ratio of early to late diastolic mitral flow velocities (E/A) in mice. (H) Measurements of the ratio of early to late diastolic mitral annular velocities (E´/A´) in mice. (I) Measurements of left ventricular end-diastolic diameter (LVEDD) in mice. *P < 0.05 vs. the control group; #P < 0.05 vs. the DCM group; n = 6.

            However, these alterations in cardiac remodeling and function were significantly ameliorated in the BBR group. Specifically, the BBR group exhibited a reversal of the echocardiographic changes observed in the DCM group, with decreased LVEDD, LVPW, and IVS measurements, and increased LVEF, FS, E/A, and E´/A´ ratios (Figure 1B–I). These findings suggested that BBR treatment effectively improves cardiac remodeling and both systolic and diastolic function in diabetic mice.

            Alleviation of Cardiac Hypertrophy and Fibrosis in Berberine-Treated Diabetic Mice

            Histological examination of myocardial cells was conducted with HE staining. In the control group, the cardiomyocytes demonstrated a well-ordered arrangement, with uniformly distributed myocardial fibers. In contrast, the DCM group exhibited some thickened cardiomyocytes with irregular and deeply stained nuclei, thus indicating compensatory enlargement. In contrast, the BBR group displayed only slightly enlarged cardiomyocytes, and nuclei appearing relatively normal (Figure 2A).

            Figure 2

            Histological staining in three groups of mice.

            (A) Representative HE staining of myocardial cross-sections; (B) Representative Masson’s trichrome staining of cardiac short-axis cross sectional areas. The arrows indicate the myocardial cells. (C) Representative WGA staining of cardiac short-axis cross sectional areas; (D) Quantitative analysis of heart weight/tibial length (HW/TL) ratio in mice. (E) Quantification of the fibrosis area in mice. (F) Quantification of the cross-sectional area of cardiomyocyte in mice. *P < 0.05 vs. the control group; #P < 0.05 vs. the DCM group; n = 6.

            Measurements of HW and TL were collected from all mice, and the HW/TL ratio was calculated. In the DCM group, the HW/TL ratio was significantly greater than that in the control group, thus indicating cardiac hypertrophy (Figure 2D). This hypertrophy was accompanied by increases in myocardial fibrosis, as evidenced by Masson’s trichrome staining, and in the cross-sectional area of cardiomyocytes, as determined by wheat germ agglutinin staining (Figure 2E–F). However, these pathological changes were significantly ameliorated in the BBR group. Specifically, the BBR group exhibited a decrease in the HW/TL ratio, indicating diminished cardiac hypertrophy (Figure 2D). Furthermore, the extent of myocardial fibrosis and the cross-sectional area of cardiomyocytes were lower in the BBR group than the DCM group (Figure 2E, F). These findings suggested that BBR treatment effectively mitigates cardiac hypertrophy, myocardial fibrosis, and cardiomyocyte enlargement in mice with DCM.

            Berberine Alleviates Cardiomyocyte Apoptosis and Restores the BCL2/BAX Ratio

            We evaluated the effect of BBR on cardiomyocyte apoptosis in DCM mice with TUNEL staining and flow cytometry. We observed a significantly greater apoptosis ratio, indicating elevated cardiomyocyte apoptosis, in the DCM group than the control group (Figure 3A, B). However, the apoptosis ratio was significantly lower in the BBR group than the DCM group, thereby suggesting that BBR mitigated cardiomyocyte apoptosis in DCM mice (Figure 3A, B). Flow cytometry analysis further corroborated these findings, revealing a higher percentage of apoptotic cells in the DCM group than the control group (Figure 3C, D). However, the BBR group exhibited a lower percentage of apoptotic cells than the DCM group (Figure 3C, D) [13].

            Figure 3

            BBR reduces DCM-induced apoptosis.

            (A) Representative TUNEL-positive cardiomyocyte staining in mice. (B) Quantification of the TUNEL positive cardiomyocyte area in mice, n = 6. (C) Quantitative flow cytometry analysis of apoptotic cardiomyocytes, n = 3. (D) Representative scatter plots from flow cytometry analysis of isolated mouse cardiomyocytes. (E-G) Protein expression of BAX and BCL2 in heart tissues were determined by Western blot with quantitative analysis of band density, n = 3. *P < 0.05 vs. the control group; #P < 0.05 vs. the DCM group.

            Moreover, we investigated the effects of BBR on the protein expression of BCL2-associated X protein (BAX), a proapoptotic factor, and B cell lymphoma 2 (BCL2), an anti-apoptotic factor. In the DCM group [14], we observed elevated BAX protein expression and diminished BCL2 expression, thus leading to an elevated BAX/BCL2 ratio indicative of the activation of the mitochondrial cell death pathway (Figure 3E–G). Remarkably, BBR substantially reversed these changes, thereby demonstrating the potential of this treatment to modulate the expression of key proteins involved in apoptosis (Figure 3E–G).

            Berberine Alleviates Oxidative Stress in Diabetic Cardiomyopathy

            We further explored the effects of BBR on oxidative stress in DCM mice. In the DCM group, we observed diminished expression of the antioxidant enzyme SOD and elevated expression of MDA, a marker of lipid peroxidation (Figure 4A, B). These findings indicated elevated oxidative stress in DCM mice. However, BBR treatment significantly increased SOD expression and decreased MDA expression, thus suggesting that BBR alleviated oxidative stress in DCM mice (Figure 4A, B). DHE staining indicated that BBR substantially inhibited DCM-induced ROS levels (Figure 4G–H).

            Figure 4

            BBR concurrently inhibits ox-CaMKII levels and enhances MsrA expression, reducing DCM-induced oxidative stress.

            (A) Serum malondialdehyde (MDA) levels, n = 6. (B) Serum superoxide dismutase (SOD) levels, n = 6. (C-E) Protein expression of MsrA, ox-CaMKII and total-CaMKII in heart tissues were determined by Western blot with quantitative analysis of band density, n = 3. (F, G) Representative DHE staining and quantitative analysis of DHE MFI of myocardial cross-sections, n = 6. *P < 0.05 vs. the control group; #P < 0.05 vs. the DCM group.

            Importantly, BBR treatment increased the expression of MsrA, an enzyme that reduces oxidized CaMKII, thereby potentially mitigating the harmful effects of oxidative stress. This finding was in line with those from recent studies highlighting the role of MsrA in modulating the oxidative state of CaMKII, a key player in oxidative stress and cardiac dysfunction [15]. Furthermore, following BBR treatment, we observed a decrease in the expression of ox-CaMKII, a protein associated with oxidative stress and cardiac dysfunction [16] (Figure 4C–F). These changes suggested a complex interplay of stress responses in the context of BBR treatment.

            Discussion

            DCM, a consequence of dysregulated glucose and lipid metabolism in diabetes mellitus, remains a major challenge, because of the complexity of its pathogenesis and the difficulty in developing targeted interventions [1, 17]. BBR, a naturally occurring isoquinoline alkaloid, has been demonstrated to have substantial cardioprotective potential, by markedly decreasing myocardial infarct size and mitigating ventricular arrhythmia, according to animal studies [18]. In this study, we extended these findings by showing that BBR treatment significantly improved cardiac function and decreased cardiac remodeling in mice with DCM, thus revealing its potential as an effective therapeutic approach for DCM. Although previous research had established the role of BBR in DCM [19], we performed deeper investigation of the mechanistic action of BBR. We explored how BBR modulates the MsrA and CaMKII pathways, thereby influencing cardiomyocyte apoptosis and oxidative stress in the context of DCM. This in-depth mechanistic investigation enhances understanding of BBR’s therapeutic potential and provides valuable insights that may guide the development of novel therapeutic strategies for DCM.

            Oxidative stress, characterized by an imbalance between the production of ROS and the body’s antioxidant defenses, is a key player in the pathogenesis of DCM [20]. In diabetes, elevated glucose levels lead to excess production of ROS, thus resulting in oxidative stress, which in turn substantially damages essential cellular components. This oxidative stress instigates cell death through a process known as apoptosis. Moreover, in diabetes, ROS further exacerbate apoptosis through several pathways [21], including the activation of complex metabolic routes, such as the DAG-PKC-NADPH oxidase pathway, direct oxidative damage to cellular components, triggering of inflammatory responses, and dysregulation of cellular processes such as autophagy [22, 23]. Our findings indicated that BBR effectively decreases oxidative stress and apoptosis in cardiomyocytes affected by DCM, thus reinforcing earlier research that established the beneficial effects of BBR on cardiac function [24, 25].

            Our study demonstrated that BBR increases the expression of MsrA in the myocardium, and decreases the oxidation of CaMKII, myocardial oxidative stress, and apoptosis in a mouse model of DCM. These findings contribute to the growing body of evidence indicating the role of CaMKII, a serine/threonine kinase, in various cardiovascular diseases, particularly in the context of diabetes [2628]. Previous studies have underscored the importance of CaMKII in endothelium-derived relaxation and eNOS activity in type 2 diabetes, and its role in hyperglycemia-induced apoptosis of retinal capillary endothelial cells [29, 30]. Moreover, the absence of CaMKII after cardiac injury has been associated with diminished cardiac remodeling and with reversal of symptoms in heart failure; therefore, CaMKII inhibition may be a potential treatment [3133]. Cardiac-specific CaMKIIδ gene deletion has been associated with a decrease in ischemia–reperfusion induced infarct formation and myocardial cell death via the NFκB-signaling pathway [34, 35]. However, the effects of CaMKII activity on oxidative stress and apoptosis in DCM have not been extensively explored.

            Our findings suggest a novel mechanism of action of BBR in the context of DCM. MsrA, a key component of the antioxidant defense system, counteracts oxidative stress by reducing methionine sulfoxide back to methionine [36]. Dysregulation of MsrA has been implicated in the pathogenesis of cardiovascular disorders, which are associated with elevated oxidative stress [37]. Previous studies have indicated that MsrA, a ubiquitous protein oxidation repair enzyme, decreases CaMKII oxidation, thereby inhibiting transformation of CaMKII into a perpetually active state, decreasing its overactivation, and mitigating its contribution to cellular damage [9, 38]. In a mouse model deficient in MsrA, cardiomyocytes have been found to exhibit elevated sensitivity to oxidative stress, thus leading to cardiac dysfunction and perturbations in calcium dynamics [39]. In this study, BBR appeared to exert its cardioprotective effects by increasing MsrA expression, and consequently decreasing CaMKII oxidation and inhibiting CaMKII oxidation-associated myocardial cell apoptosis. The role of BBR in modulating MsrA expression and CaMKII oxidation presents a promising avenue for further investigation. Future studies should be aimed at elucidating the precise molecular pathways involved, and assessing the potential therapeutic implications of these findings in the treatment of DCM.

            Conclusion

            In conclusion, our study demonstrated the cardioprotective effects of BBR in a mouse model of DCM, through increasing MsrA expression, and decreasing CaMKII oxidation, myocardial oxidative stress, and apoptosis. These findings suggest a novel mechanism of BBR’s action, thereby potentially offering a new therapeutic approach for DCM.

            Declarations

            Data Availability Statement

            All authors confirm that the data supporting the findings of the study are available on reasonable request.

            Ethics Statement

            The animal study was reviewed and approved by the Animal Experimental Ethics Association of Tianjin First Centre Hospital.

            Author Contributions

            Xiaoqiang Sun, Zhuqing Li, and Li Wang performed experiments. Xiaoqiang Sun performed data collection and analyzed related indexes. Zhuqing Li and Li Wang assisted in laboratory experiments. Xiaoqiang Sun assisted in statistical analyses. Li Wang assisted in data analysis. Xiaoqiang Sun, Zhuqing Li, and Yanxin Wang wrote the manuscript. Xiaoqiang Sun conceptualized and designed the study. Chengzhi Lu and Yanxin Wang contributed to supervision of the study. The final version of this manuscript was reviewed and revised by all authors, and all authors approved its submission.

            Consent for Publication

            Not applicable.

            Conflict of Interest

            The authors declare no conflicts of interest.

            Citation Information

            References

            1. , , , , , , et al. Diabetic cardiomyopathy - A comprehensive updated review. Prog Cardiovasc Dis 2019;62(4):315–26.

            2. , , , , , . Diabetic cardiomyopathy: current and future therapies. Beyond glycemic control. Front Physiol 2018;9:1514.

            3. , , . Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res 2018;122(4):624–38.

            4. , , , . Molecular mechanisms and epigenetic regulation in diabetic cardiomyopathy. Front Cardiovasc Med 2021;8:725532.

            5. , , , , , . Methionine sulfoxide reductase (MsrA) is a regulator of antioxidant defense and lifespan in mammals. Proc Natl Acad Sci U S A 2001;98(23):12920–5.

            6. , , . Mammals reduce methionine-S-sulfoxide with MsrA and are unable to reduce methionine-R-sulfoxide, and this function can be restored with a yeast reductase. J Biol Chem 2008;283(42):28361–9.

            7. , , . CaMKII inhibition is a novel therapeutic strategy to prevent diabetic cardiomyopathy. Front Pharmacol 2021;12:695401.

            8. , , , , , , et al. CaMKII activation in early diabetic hearts induces altered sarcoplasmic reticulum-mitochondria signaling. Sci Rep 2021;11(1):20025.

            9. , , , , , , et al. Oxidation of CaMKII determines the cardiotoxic effects of aldosterone. Nat Med 2011;17(12):1610–8.

            10. , , , , , , et al. Promising antioxidative effect of berberine in cardiovascular diseases. Front Pharmacol 2022;13:865353.

            11. , , , , , . Inflammatory factors driving atherosclerotic plaque progression new insights. J Transl Intern Med 2022;10:36–47.

            12. , , , , , , et al. Cardiomyocyte-specific knockout of ADAM17 ameliorates left ventricular remodeling and function in diabetic cardiomyopathy of mice. Signal Transduct Target Ther 2022;7(1):259.

            13. , , , , . Research progress on N6-adenosylate methylation RNA modification in heart failure remodeling. J Transl Intern Med 2022;10:340–8.

            14. , , , , , , et al. Amount of spontaneous apoptosis detected by Bax/Bcl-2 ratio predicts outcome in acute myeloid leukemia (AML). Blood 2003;101(6):2125–31.

            15. , , , , , . Redox modification of cell signaling in the cardiovascular system. J Mol Cell Cardiol 2012;52(3):550–8.

            16. . Oxidant stress promotes disease by activating CaMKII. J Mol Cell Cardiol 2015;89(Pt B):160–7.

            17. , , , , , , et al. Novel insights into the pathogenesis of diabetic cardiomyopathy and pharmacological strategies. Front Cardiovasc Med 2021;8:707336.

            18. , , , , , , et al. Berberine protects rat heart from ischemia/reperfusion injury via activating JAK2/STAT3 signaling and attenuating endoplasmic reticulum stress. Acta Pharmacol Sin 2016;37(3):354–67.

            19. , , , , , , et al. Berberine could ameliorate cardiac dysfunction via interfering myocardial lipidomic profiles in the rat model of diabetic cardiomyopathy. Front Physiol 2018;9:1042.

            20. , . Oxidative stress and diabetic complications. Circ Res 2010;107(9):1058–70.

            21. , , , . The impact of glucose exposure on bioenergetics and function in a cultured endothelial cell model and the implications for cardiovascular health in diabetes. Sci Rep 2020;10(1):19547.

            22. , . NAD(P)H oxidase activation: a potential target mechanism for diabetic vascular complications, progressive beta-cell dysfunction and metabolic syndrome. Curr Drug Targets 2005;6(4):495–501.

            23. , , , , . Systematic understanding of pathophysiological mechanisms of oxidative stress-related conditions-diabetes mellitus, cardiovascular diseases, and ischemia-reperfusion injury. Front Cardiovasc Med 2021;8:649785.

            24. , , , , , , et al. Berberine protects human and rat cardiomyocytes from hypoxia/reoxygenation-triggered apoptosis. Am J Transl Res 2021;13(2):659–71. Retraction in: Am J Transl Res 2021;13(12):14241.

            25. , , , , , , et al. Berberine protects cardiac cells against ferroptosis. Tzu Chi Med J 2022;34(3):310–7.

            26. , , , . CaMKII in the cardiovascular system: sensing redox states. Physiol Rev 2011;91(3):889–915.

            27. , . Role of CaMKII in cardiac arrhythmias. Trends Cardiovasc Med 2015;25(5):392–7.

            28. , , . Physiological and unappreciated roles of CaMKII in the heart. Basic Res Cardiol 2018;113(4):29.

            29. , , , , , . Involvement of CaM kinase II in the impairment of endothelial function and eNOS activity in aortas of Type 2 diabetic rats. Clin Sci (Lond) 2012;123(6):375–86.

            30. , , , , . Calcium entry mediates hyperglycemia-induced apoptosis through Ca(2+)/calmodulin-dependent kinase II in retinal capillary endothelial cells. Mol Vis 2012;18:2371–9.

            31. , , , , , , et al. Inducible cardiomyocyte-specific deletion of CaM kinase II protects from pressure overload-induced heart failure. Basic Res Cardiol 2016;111(6):65.

            32. , , . CaMKII in myocardial hypertrophy and heart failure. J Mol Cell Cardiol 2011;51(4):468–73.

            33. , , , , , . The role of CaMKII in diabetic heart dysfunction. Heart Fail Rev 2015;20(5):589–600.

            34. , , , , , , et al. Ca2+/Calmodulin-dependent protein kinase II δ mediates myocardial ischemia/reperfusion injury through nuclear factor-κB. Circ Res 2013;112(6):935–44.

            35. , , , , . Myocardial death and dysfunction after ischemia-reperfusion injury require CaMKIIδ oxidation. Sci Rep 2019;9(1):9291.

            36. , , . Methionine sulfoxide reductase (Msr) dysfunction in human brain disease. Free Radic Res 2019;53(11–12):1144–54.

            37. , , , , , , et al. Distinctive under-expression profile of inflammatory and redox genes in the blood of elderly patients with cardiovascular disease. J Inflamm Res 2021;14:429–42.

            38. , , , , . Suppression of oxidative stress and apoptosis in electrically stimulated neonatal rat cardiomyocytes by resveratrol and underlying mechanisms. J Cardiovasc Pharmacol 2017;70(6):396–404.

            39. , , , , , , et al. Deficiency of methionine sulfoxide reductase A causes cellular dysfunction and mitochondrial damage in cardiac myocytes under physical and oxidative stresses. Biochem Biophys Res Commun 2010;402(4):608–13.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            03 November 2023
            : 8
            : 1
            : e948
            Affiliations
            [1] 1Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
            [2] 2Department of Cardiology, Tianjin Fourth Central Hospital, Tianjin 300140, China
            Author notes
            Correspondence: Chengzhi Lu, MD, PhD, Department of Cardiology, Tianjin First Central Hospital, No.24 Fukang Road, Nankai District, Tianjin 300192, China, E-mail: 5020200072@ 123456nankai.edu.cn
            Article
            cvia.2023.0064
            10.15212/CVIA.2023.0064
            734b2af7-a0a2-4f86-8803-5d893bcbb1ae
            Copyright © 2023 Cardiovascular Innovations and Applications

            This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 Unported License (CC BY-NC 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc/4.0/.

            History
            : 19 June 2023
            : 30 August 2023
            : 14 September 2023
            Page count
            Figures: 4, Tables: 1, References: 39, Pages: 12
            Funding
            Funded by: Traditional Chinese Medicine and Integrative Medicine Research Project of Tianjin Administration of Traditional Chinese Medicine
            Award ID: 2021192
            Funded by: National Natural Science Foundation of China
            Award ID: 81970303
            Funded by: Medical Health Science and Technology Project of the Health Commission of Tianjin
            Award ID: TJWJ2023QN026
            Funded by: Natural Science Foundation of Tianjin
            Award ID: 22JCQNJC01130
            This research was funded by the Traditional Chinese Medicine and Integrative Medicine Research Project of Tianjin Administration of Traditional Chinese Medicine (2021192), the National Natural Science Foundation of China (NSFC, No. 81970303), the Medical Health Science and Technology Project of the Health Commission of Tianjin (TJWJ2023QN026), the Natural Science Foundation of Tianjin (22JCQNJC01130), and the Tianjin Key Medical Discipline (Specialty) Construction Project.
            Categories
            Research Article

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            apoptosis,oxidative stress,Berberine,diabetic cardiomyopathy

            Comments

            Comment on this article