Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Cardiovasc Diabetol ; 23(1): 164, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724987

ABSTRACT

Dynamin-related protein 1 (Drp1) is a crucial regulator of mitochondrial dynamics, the overactivation of which can lead to cardiovascular disease. Multiple distinct posttranscriptional modifications of Drp1 have been reported, among which S-nitrosylation was recently introduced. However, the detailed regulatory mechanism of S-nitrosylation of Drp1 (SNO-Drp1) in cardiac microvascular dysfunction in diabetes remains elusive. The present study revealed that mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) was consistently upregulated in diabetic cardiomyopathy (DCM) and promoted SNO-Drp1 in cardiac microvascular endothelial cells (CMECs), which in turn led to mitochondrial dysfunction and cardiac microvascular disorder. Further studies confirmed that MAP4K4 promoted SNO-Drp1 at human C644 (mouse C650) by inhibiting glutathione peroxidase 4 (GPX4) expression, through which MAP4K4 stimulated endothelial ferroptosis in diabetes. In contrast, inhibition of MAP4K4 via DMX-5804 significantly reduced endothelial ferroptosis, alleviated cardiac microvascular dysfunction and improved cardiac dysfunction in db/db mice by reducing SNO-Drp1. In parallel, the C650A mutation in mice abolished SNO-Drp1 and the role of Drp1 in promoting cardiac microvascular disorder and cardiac dysfunction. In conclusion, our findings demonstrate that MAP4K4 plays an important role in endothelial dysfunction in DCM and reveal that SNO-Drp1 and ferroptosis activation may act as downstream targets, representing potential therapeutic targets for DCM.


Subject(s)
Diabetic Cardiomyopathies , Dynamins , Endothelial Cells , Mice, Inbred C57BL , Signal Transduction , Animals , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Humans , Dynamins/metabolism , Dynamins/genetics , Male , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/enzymology , Endothelial Cells/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Ferroptosis/drug effects , Disease Models, Animal , Cells, Cultured , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/enzymology , Mice , Protein Processing, Post-Translational , Coronary Circulation , Intracellular Signaling Peptides and Proteins
2.
J Cardiovasc Pharmacol ; 79(1): e75-e86, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34740211

ABSTRACT

ABSTRACT: The present study was intended to evaluate the effect of polyherbal formulation (PHF) made with 3 nutraceuticals, such as Piper nigrum, Terminalia paniculata, and Bauhinia purpurea on inflammation and oxidative stress in diabetic cardiomyopathy (DCM), which is induced by streptozotocin and nicotinamide administration in rats. We supplemented DCM rats with PHF (250 and 500 mg/kg/BW) for 45 days and evaluated their effects on oxidative stress markers, proinflammatory cytokines, and messenger RNA expressions of the nuclear factor erythroid 2-related factor-2 (Nrf-2) and its linked genes [heme oxygenase-1 (HO-1), superoxide dismutase, catalase] along with inflammatory genes [tumour necrosis factor α and nuclear factor kappa B (NF-κB)]. Our study demonstrated that PHF successfully attenuated inflammation and oxidative stress via messenger RNA upregulation of Nrf-2, HO-1, superoxide dismutase, and catalase and concomitantly with downregulation of tumour necrosis factor α and NF-κB. Conversely, PHF also protected hyperglycemia-mediated cardiac damage, which was confirmed with histopathological and scanning electron microscopy analysis. In conclusion, our results suggested that PHF successfully ameliorated hyperglycemia-mediated inflammation and oxidative stress via regulation of NF-κB/Nrf-2/HO-1 pathway. Therefore, these results recommend that PHF may be a prospective therapeutic agent for DCM.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Diabetic Cardiomyopathies/prevention & control , Heme Oxygenase (Decyclizing)/metabolism , Hypoglycemic Agents/pharmacology , Inflammation Mediators/metabolism , Myocytes, Cardiac/drug effects , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Plant Preparations/pharmacology , Animals , Blood Glucose/metabolism , Cytokines/genetics , Cytokines/metabolism , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Gene Expression Regulation , Heme Oxygenase (Decyclizing)/genetics , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , Rats, Wistar , Signal Transduction
3.
Cardiovasc Res ; 118(1): 212-225, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33576380

ABSTRACT

AIMS: The glucose-driven enzymatic modification of myocardial proteins by the sugar moiety, ß-N-acetylglucosamine (O-GlcNAc), is increased in pre-clinical models of diabetes, implicating protein O-GlcNAc modification in diabetes-induced heart failure. Our aim was to specifically examine cardiac manipulation of the two regulatory enzymes of this process on the cardiac phenotype, in the presence and absence of diabetes, utilising cardiac-targeted recombinant-adeno-associated viral-vector-6 (rAAV6)-mediated gene delivery. METHODS AND RESULTS: In human myocardium, total protein O-GlcNAc modification was elevated in diabetic relative to non-diabetic patients, and correlated with left ventricular (LV) dysfunction. The impact of rAAV6-delivered O-GlcNAc transferase (rAAV6-OGT, facilitating protein O-GlcNAcylation), O-GlcNAcase (rAAV6-OGA, facilitating de-O-GlcNAcylation), and empty vector (null) were determined in non-diabetic and diabetic mice. In non-diabetic mice, rAAV6-OGT was sufficient to impair LV diastolic function and induce maladaptive cardiac remodelling, including cardiac fibrosis and increased Myh-7 and Nppa pro-hypertrophic gene expression, recapitulating characteristics of diabetic cardiomyopathy. In contrast, rAAV6-OGA (but not rAAV6-OGT) rescued LV diastolic function and adverse cardiac remodelling in diabetic mice. Molecular insights implicated impaired cardiac PI3K(p110α)-Akt signalling as a potential contributing mechanism to the detrimental consequences of rAAV6-OGT in vivo. In contrast, rAAV6-OGA preserved PI3K(p110α)-Akt signalling in diabetic mouse myocardium in vivo and prevented high glucose-induced impairments in mitochondrial respiration in human cardiomyocytes in vitro. CONCLUSION: Maladaptive protein O-GlcNAc modification is evident in human diabetic myocardium, and is a critical regulator of the diabetic heart phenotype. Selective targeting of cardiac protein O-GlcNAcylation to restore physiological O-GlcNAc balance may represent a novel therapeutic approach for diabetes-induced heart failure.


Subject(s)
Antigens, Neoplasm/metabolism , Diabetic Cardiomyopathies/enzymology , Histone Acetyltransferases/metabolism , Hyaluronoglucosaminidase/metabolism , Myocytes, Cardiac/enzymology , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational , Ventricular Dysfunction, Left/enzymology , Ventricular Function, Left , Ventricular Remodeling , Aged , Animals , Antigens, Neoplasm/genetics , Cell Line , Class I Phosphatidylinositol 3-Kinases/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Glycosylation , Histone Acetyltransferases/genetics , Humans , Hyaluronoglucosaminidase/genetics , Male , Mice , Middle Aged , Myocytes, Cardiac/pathology , N-Acetylglucosaminyltransferases/genetics , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
4.
Biochem Biophys Res Commun ; 586: 8-13, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34818584

ABSTRACT

AIM: To evaluate the effects of exercise training (ET) on cardiac extracellular matrix (ECM) proteins homeostasis and cardiac dysfunction in mice with diabetic cardiomyopathy. METHODS: Thirty-six male C57BL/6 mice were randomized into 3 groups for 8 weeks (12mice/group): Diabetic control-DC: Diabetes was induced by single streptozotocin injection (200 mg/kg i.p.); Diabetic exercise-DE: Diabetic mice underwent ET program on motorized-treadmill (6-times/week, 60min/session); Non-diabetic control-NDC: Vehicle-treated, sedentary, non-diabetic mice served as controls. Before euthanasia, all groups underwent transthoracic echocardiography (TTE). Post-mortem, left-ventricle (LV) samples were histologically analysed for ECM proteins (collagen, elastin), matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs). RESULTS: DC group showed significantly higher cardiac contents of collagen and MMP-9 and lower elastic concentration than NDC (p < 0.001). The implementation of ET completely outweighed those diabetes-induced changes (DE vs NDC, p > 0.05). TIMP-1 levels significantly increased across all groups (DC: 18.98 ± 3.47%, DE: 24.24 ± 2.36%, NDC: 46.36 ± 5.91%; p < 0.05), while MMP-9/TIMP-1 ratio followed a reverse pattern. ET tended to increase MMP-2 concentrations versus DC (p = 0.055), but did not achieve non-diabetic levels (p < 0.05). TIMP-2 cardiac concentrations remained unaltered throughout the study (p > 0.05). Importantly, ET ameliorated both LV end-systolic internal diameter (LVESD) (p < 0.001) and the percentage of LV fractional shortening (FS%) (p = 0.006) compared to DC. Despite that favorable effect, the cardiac function level of DE group remained worse than NDC group (%FS: p = 0.002; LVESD: p < 0.001). CONCLUSION: Systemic ET may favorably change ECM proteins, MMP-9 and TIMP-1 cardiac concentrations in mice with diabetic cardiomyopathy. Those results were associated with partial improvement of echocardiography-assessed cardiac function, indicating a therapeutic effect of ET in diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Experimental/enzymology , Diabetic Cardiomyopathies/enzymology , Extracellular Matrix/enzymology , Matrix Metalloproteinase 9/genetics , Physical Conditioning, Animal/physiology , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Blood Glucose/metabolism , Collagen/genetics , Collagen/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/chemically induced , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Echocardiography , Elastin/genetics , Elastin/metabolism , Exercise Test , Extracellular Matrix/genetics , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Streptozocin/administration & dosage , Tissue Inhibitor of Metalloproteinase-1/metabolism
5.
Am J Physiol Heart Circ Physiol ; 321(5): H850-H864, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34477461

ABSTRACT

Molecular mechanisms underlying cardiac dysfunction and subsequent heart failure in diabetic cardiomyopathy are incompletely understood. Initially we intended to test the role of G protein-coupled receptor kinase 2 (GRK2), a potential mediator of cardiac dysfunction in diabetic cardiomyopathy, but found that control animals on HFD did not develop cardiomyopathy. Cardiac function was preserved in both wild-type and GRK2 knockout animals fed high-fat diet as indicated by preserved left ventricular ejection fraction (LVEF) although heart mass was increased. The absence of cardiac dysfunction led us to rigorously evaluate the utility of diet-induced obesity to model diabetic cardiomyopathy in mice. Using pure C57BL/6J animals and various diets formulated with different sources of fat-lard (32% saturated fat, 68% unsaturated fat) or hydrogenated coconut oil (95% saturated fat), we consistently observed left ventricular hypertrophy, preserved LVEF, and preserved contractility measured by invasive hemodynamics in animals fed high-fat diet. Gene expression patterns that characterize pathological hypertrophy were not induced, but a modest induction of various collagen isoforms and matrix metalloproteinases was observed in heart with high-fat diet feeding. PPARα-target genes that enhance lipid utilization such as Pdk4, CD36, AcadL, and Cpt1b were induced, but mitochondrial energetics was not impaired. These results suggest that although long-term fat feeding in mice induces cardiac hypertrophy and increases cardiac fatty acid metabolism, it may not be sufficient to activate pathological hypertrophic mechanisms that impair cardiac function or induce cardiac fibrosis. Thus, additional factors that are currently not understood may contribute to the cardiac abnormalities previously reported by many groups.NEW & NOTEWORTHY Dietary fat overload (DFO) is widely used to model diabetic cardiomyopathy but the utility of this model is controversial. We comprehensively characterized cardiac contractile and mitochondrial function in C57BL6/J mice fed with lard-based or saturated fat-enriched diets initiated at two ages. Despite cardiac hypertrophy, contractile and mitochondrial function is preserved, and molecular adaptations likely limit lipotoxicity. The resilience of these hearts to DFO underscores the need to develop robust alternative models of diabetic cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies/etiology , Diet, High-Fat , Hypertrophy, Left Ventricular/etiology , Obesity/complications , Stroke Volume , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left , Age Factors , Animals , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Energy Metabolism , Female , Fibrosis , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocardium/enzymology , Myocardium/pathology , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling
6.
Cell Death Dis ; 12(7): 624, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135313

ABSTRACT

Proteasomal activity is compromised in diabetic hearts that contributes to proteotoxic stresses and cardiac dysfunction. Osteocrin (OSTN) acts as a novel exercise-responsive myokine and is implicated in various cardiac diseases. Herein, we aim to investigate the role and underlying molecular basis of OSTN in diabetic cardiomyopathy (DCM). Mice received a single intravenous injection of the cardiotrophic adeno-associated virus serotype 9 to overexpress OSTN in the heart and then were exposed to intraperitoneal injections of streptozotocin (STZ, 50 mg/kg) for consecutive 5 days to generate diabetic models. Neonatal rat cardiomyocytes were isolated and stimulated with high glucose to verify the role of OSTN in vitro. OSTN expression was reduced by protein kinase B/forkhead box O1 dephosphorylation in diabetic hearts, while its overexpression significantly attenuated cardiac injury and dysfunction in mice with STZ treatment. Besides, OSTN incubation prevented, whereas OSTN silence aggravated cardiomyocyte apoptosis and injury upon hyperglycemic stimulation in vitro. Mechanistically, OSTN treatment restored protein kinase G (PKG)-dependent proteasomal function, and PKG or proteasome inhibition abrogated the protective effects of OSTN in vivo and in vitro. Furthermore, OSTN replenishment was sufficient to prevent the progression of pre-established DCM and had synergistic cardioprotection with sildenafil. OSTN protects against DCM via restoring PKG-dependent proteasomal activity and it is a promising therapeutic target to treat DCM.


Subject(s)
Apoptosis/drug effects , Diabetic Cardiomyopathies/prevention & control , Muscle Proteins/pharmacology , Myocytes, Cardiac/drug effects , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/pharmacology , Animals , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/metabolism , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Forkhead Box Protein O1/metabolism , Male , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Phosphorylation , Proof of Concept Study , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Recombinant Proteins/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Cardiovasc Diabetol ; 20(1): 90, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33906662

ABSTRACT

BACKGROUND: Diabetic and obese patients are at higher risk of severe disease and cardiac injury in corona virus 2 (SARS-CoV-2) infections. Cellular entry of SARS-CoV-2 is mainly via the angiotensin-converting enzyme 2 (ACE2) receptor, which is highly expressed in normal hearts. There is a disagreement regarding the effect of factors such as obesity and diabetes on ACE2 expression in the human heart and whether treatment with renin-angiotensin system inhibitors or anti-diabetic medications increases ACE2 expression and subsequently the susceptibility to infection. We designed this study to elucidate factors that control ACE2 expression in human serum, human heart biopsies, and mice. METHODS: Right atrial appendage biopsies were collected from 79 patients that underwent coronary artery bypass graft (CABG) surgery. We investigated the alteration in ACE2 mRNA and protein expression in heart tissue and serum. ACE2 expression was compared with clinical risk factors: diabetes, obesity and different anti-hypertensive or anti-diabetic therapies. WT or db/db mice were infused with Angiotensin II (ATII), treated with different anti-diabetic drugs (Metformin, GLP1A and SGLT2i) were also tested. RESULTS: ACE2 gene expression was increased in diabetic hearts compared to non-diabetic hearts and was positively correlated with glycosylated hemoglobin (HbA1c), body mass index (BMI), and activation of the renin angiotensin system (RAS), and negatively correlated with ejection fraction. ACE2 was not differentially expressed in patients who were on angiotensin converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARBs) prior to the operation. We found no correlation between plasma free ACE2 and cardiac tissue ACE2 expression. Transmembrane serine protease 2 (TMPRSS2), metalloprotease ADAM10 and ADAM17 that facilitate viral-ACE2 complex entry and degradation were increased in diabetic hearts. ACE2 expression in mice was increased with ATII infusion and attenuated following anti-diabetic drugs treatment. CONCLUSION: Patients with uncontrolled diabetes or obesity with RAS activation have higher ACE2 expressions therefore are at higher risk for severe infection. Since ACEi or ARBs show no effect on ACE2 expression in the heart further support their safety.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Diabetes Mellitus, Type 2/enzymology , Diabetic Cardiomyopathies/enzymology , Myocardium/enzymology , Obesity/enzymology , Receptors, Virus/metabolism , Renin-Angiotensin System , SARS-CoV-2/pathogenicity , Aged , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/enzymology , Case-Control Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Female , Host-Pathogen Interactions , Humans , Hypoglycemic Agents/pharmacology , Male , Mice , Middle Aged , Obesity/complications , Obesity/physiopathology , Renin-Angiotensin System/drug effects , Risk Factors , SARS-CoV-2/metabolism , Up-Regulation
9.
Toxicol Appl Pharmacol ; 421: 115533, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33848515

ABSTRACT

Decreased activity of AMP-activated protein kinase (AMPK) is implicated in the pathogenesis of diabetic cardiomyopathy (DCM). Recent evidence suggests a crosstalk between cinacalcet and AMPK activation. This study investigated the effects of cinacalcet on cardiac remodeling and dysfunction in type 2 diabetic rats (T2DM). High fat diet for 4 weeks combined with single intraperitoneal injection of streptozotocin (30 mg/kg) was used to induce type 2 diabetes in rats. Diabetic rats were either orally treated with vehicle, 5 or 10 mg/kg cinacalcet for 4 weeks. Control rats were fed standard chow diet and intraperitoneally injected with citrate buffer. T2DM rats showed lower body weight (BW), hyperglycemia and dyslipidemia, along with increased heart weight (HW) and HW/BW ratio. Masson's trichrome stained cardiac sections revealed massive fibrosis in T2DM rats. There were increased TGF-ß1 and hydroxyproline levels, coupled with up-regulation of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) in hearts of T2DM rats. These alterations were associated with redox imbalance and impaired cardiac functions. Decreased phosphorylation of AMPK at threonine172 residue was found in T2DM hearts. Cinacalcet for 4 weeks significantly activated AMPK and alleviated cardiac remodeling and dysfunction in a dose-dependent manner, without affecting blood glucose, serum calcium and phosphorus levels. Cinacalcet increased the mitochondrial DNA content, and expressions of PGC-1α, UCP-3, beclin-1 and LC3-II/LC3-I ratio. Cinacalcet decreased the pro-apoptotic Bax, while increased the anti-apoptotic Bcl-2 in cardiac tissue of T2DM rats. These findings might highlight cinacalcet as an alternative therapy to combat the development and progression of DCM.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Cinacalcet/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Ventricular Remodeling/drug effects , Animals , Apoptosis/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Fibrosis , Hemodynamics/drug effects , Male , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats, Wistar , Signal Transduction , Streptozocin
10.
Biosci Rep ; 41(9)2021 09 30.
Article in English | MEDLINE | ID: mdl-33682891

ABSTRACT

OBJECTIVE: Diabetic cardiomyopathy (DCM) is one of the complications experienced by patients with diabetes. In recent years, long noncoding RNAs (lncRNAs) have been investigated because of their role in the progression of various diseases, including DCM. The purpose of the present study was to explore the role of lncRNA GAS5 in high glucose (HG)-induced cardiomyocyte injury and apoptosis. MATERIALS AND METHODS: We constructed HG-induced AC16 cardiomyocytes and a streptozotocin (STZ)-induced rat diabetes model. GAS5 was overexpressed and knocked out at the cellular level, and GAS5 was knocked down by lentiviruses at the animal level to observe its effect on myocardial injury. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression of GAS5. Cell proliferation and apoptosis after GAS5 knockout were detected by CCK-8, TUNEL, and flow cytometry assays. ELISA was used to detect the changes in myocardial enzyme content in cells and animal myocardial tissues during the action of GAS5 on myocardial injury. RESULTS: GAS5 expression was up-regulated in HG-treated AC16 cardiomyocytes and the rat diabetic myocardial injury model. The down-regulation of GAS5 could inhibit HG-induced myocardial damage. This work proved that the down-regulation of GAS5 could reverse cardiomyocyte injury and apoptosis by targeting miR-138 to down-regulate CYP11B2. CONCLUSION: We confirmed for the first time that the down-regulation of GAS5 could reverse CYP11B2 via the miR-138 axis to reverse HG-induced cardiomyocyte injury. This research might provide a new direction for explaining the developmental mechanism of DCM and potential targets for the treatment of myocardial injury.


Subject(s)
Blood Glucose/metabolism , Cytochrome P-450 CYP11B2/metabolism , Diabetic Cardiomyopathies/prevention & control , Glucose/toxicity , MicroRNAs/metabolism , Myocytes, Cardiac/drug effects , RNA, Long Noncoding/metabolism , Animals , Apoptosis , Cell Line , Cell Proliferation , Cytochrome P-450 CYP11B2/genetics , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Down-Regulation , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , RNA, Long Noncoding/genetics , Signal Transduction
11.
Cell Biol Int ; 45(7): 1393-1403, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33595160

ABSTRACT

Farnesyl pyrophosphate synthase (FPPS)-catalyzed isoprenoid intermediates are involved in diabetic cardiomyopathy. This study investigated the specific role of FPPS in the development of diabetic cardiomyopathy. We demonstrated that FPPS expression was elevated in both in vivo and in vitro models of diabetic cardiomyopathy. FPPS inhibition decreased the expression of proteins related to cardiac fibrosis and cardiomyocytic hypertrophy, including collagen I, collagen III, connective tissue growth factor, natriuretic factor, brain natriuretic peptide, and ß-myosin heavy chain. Furthermore, FPPS inhibition and knockdown prevented phosphorylated c-Jun N-terminal kinase 1/2 (JNK1/2) activation in vitro. In addition, a JNK1/2 inhibitor downregulated high-glucose-induced responses to diabetic cardiomyopathy. Finally, immunofluorescence revealed that cardiomyocytic size was elevated by high glucose and was decreased by zoledronate, small-interfering farnesyl pyrophosphate synthase (siFPPS), and a JNK1/2 inhibitor. Taken together, our findings indicate that FPPS and JNK1/2 may be part of a signaling pathway that plays an important role in diabetic cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies/enzymology , Geranyltranstransferase/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Myocytes, Cardiac , Rats , Rats, Sprague-Dawley
12.
J Cell Physiol ; 236(6): 4290-4302, 2021 06.
Article in English | MEDLINE | ID: mdl-33421145

ABSTRACT

High-glucose (HG) suppresses mesenchymal stem cell (MSC) functions, resulting in a decrease in cardiac regenerative capability for MSC in diabetes mellitus (DM). Resveratrol enhances MSC functions under stress. This study explores if cardiac regenerative capability can be enhanced in MSCs pretreated with resveratrol in DM rats receiving MSCs. In vitro evidence confirms that HG decreases MSCs capability through suppression of survival markers, AMP-activated protein kinase (AMPK)/Sirtuin 1 (Sirt1) axis, and expression of apoptotic markers. All of these markers are improved when MSCs are cocultured with resveratrol. Wistar male rats were randomly divided into Sham, DM (DM rats), DM rats with autologous transplantation of adipose-derived stem cells (DM + ADSC), and DM rats with resveratrol pretreated ADSC (DM + RSVL-ADSC). Compared to the Sham, DM induces pathological pathways (including fibrosis, hypertrophy, and apoptosis) and suppresses survival as well as the AMPK/Sirt1 axis in the DM group. DM + ADSC slightly improves the above pathways whereas DM + RSVL-ADSC significantly improves the above pathways when compared to the DM group. These results illustrate that resveratrol pretreated with MSCs may show clinical potential in the treatment of heart failure in patients with DM.


Subject(s)
Antioxidants/pharmacology , Diabetic Cardiomyopathies/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Resveratrol/pharmacology , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/cytology , Animals , Cell Communication , Cell Line , Cell Proliferation , Coculture Techniques , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Male , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Signal Transduction , Ventricular Function, Left
13.
J Pineal Res ; 70(1): e12698, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33016468

ABSTRACT

Targeting mitochondrial quality control with melatonin has been found promising for attenuating diabetic cardiomyopathy (DCM), although the underlying mechanisms remain largely undefined. Activation of SIRT6 and melatonin membrane receptors exerts cardioprotective effects while little is known about their roles during DCM. Using high-fat diet-streptozotocin-induced diabetic rat model, we found that prolonged diabetes significantly decreased nocturnal circulatory melatonin and heart melatonin levels, reduced the expressions of cardiac melatonin membrane receptors, and decreased myocardial SIRT6 and AMPK-PGC-1α-AKT signaling. 16 weeks of melatonin treatment inhibited the progression of DCM and the following myocardial ischemia-reperfusion (MI/R) injury by reducing mitochondrial fission, enhancing mitochondrial biogenesis and mitophagy via re-activating SIRT6 and AMPK-PGC-1α-AKT signaling. After the induction of diabetes, adeno-associated virus carrying SIRT6-specific small hairpin RNA or luzindole was delivered to the animals. We showed that SIRT6 knockdown or antagonizing melatonin receptors abolished the protective effects of melatonin against mitochondrial dysfunction as evidenced by aggravated mitochondrial fission and reduced mitochondrial biogenesis and mitophagy. Additionally, SIRT6 shRNA or luzindole inhibited melatonin-induced AMPK-PGC-1α-AKT activation as well as its cardioprotective actions. Collectively, we demonstrated that long-term melatonin treatment attenuated the progression of DCM and reduced myocardial vulnerability to MI/R injury through preserving mitochondrial quality control. Melatonin membrane receptor-mediated SIRT6-AMPK-PGC-1α-AKT axis played a key role in this process. Targeting SIRT6 with melatonin treatment may be a promising strategy for attenuating DCM and reducing myocardial vulnerability to ischemia-reperfusion injury in diabetic patients.


Subject(s)
Diabetic Cardiomyopathies/prevention & control , Melatonin/pharmacology , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Organelle Biogenesis , Sirtuins/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Forkhead Box Protein O3/metabolism , Male , Mitochondria, Heart/enzymology , Mitochondria, Heart/ultrastructure , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction , Sirtuins/genetics , Time Factors
14.
Eur J Pharmacol ; 881: 173206, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32442539

ABSTRACT

The human body has a mechanism for balancing the generation and neutralization of reactive oxygen species. The body is exposed to many agents that are responsible for the generation of reactive oxygen/nitrogen species, which leads to disruption of the balance between generation of these species and oxidative stress defence mechanisms. Diabetes is a chronic pathological condition associated with prolonged hyperglycaemia. Prolonged elevation of level of glucose in the blood leads to the generation of reactive oxygen species. This generation of reactive oxygen species is responsible for the development of diabetic vasculopathy, which includes micro- and macrovascular diabetic complications. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a membrane-bound enzyme responsible for the development of reactive oxygen species in hyperglycaemia. Phosphorylation of the cytosolic components of NOX, such as p47phox, p67phox, and RAC-1, in hyperglycaemia is one of the important causes of conversion of oxygen to reactive oxygen. Overexpression of NOX in pathological conditions is associated with activation of aldose reductase, advanced glycation end products, protein kinase C and the hexosamine pathway. In addition, NOX also promotes the activation of inflammatory cytokines, such as TGF-ß, TNF-α, NF-kß, IL-6, and IL-18, the activation of endothelial growth factors, such as VEGF and FGF, hyperlipidaemia, and the deposition of collagen. Thus, overexpression of NOX is linked to the development of diabetic complications. The present review focuses on the role of NOX, its associated pathways, and various NOX inhibitors in the management and treatment of diabetic complications, such as diabetic nephropathy, retinopathy, neuropathy and cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies/enzymology , Diabetic Nephropathies/enzymology , Diabetic Neuropathies/enzymology , Diabetic Retinopathy/enzymology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Diabetic Cardiomyopathies/drug therapy , Diabetic Nephropathies/drug therapy , Diabetic Neuropathies/drug therapy , Diabetic Retinopathy/drug therapy , Enzyme Inhibitors/therapeutic use , Humans , NADPH Oxidases/antagonists & inhibitors , Oxidative Stress , Signal Transduction
15.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32347295

ABSTRACT

Ischemic heart disease is the main cardiovascular complication of diabetes patients which is mainly caused by oxidative stress. DJ-1 is the key regulator for myocardial protection through inhibiting phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and activating Akt (also known as PKB or protein kinase B). This research is to investigate whether the antioxidant N-acetylcysteine (NAC) could alleviate diabetic myocardial ischemia/reperfusion (I/R) injury by the protective molecule DJ-1. DJ-1 in rat myocardial H9c2 cells and cardiac tissue was respectively knocked down by siRNA and adeno-associated virus (AAV). From the present study, it could be found that compared with high glucose (HG)-normal (N)/DM group, hypoxia/reoxygenation (H/R) or I/R injury can aggravate oxidative stress injury and apoptosis rate of myocardial cells, inhibit the expression of Bcl-2, activate the BAX and cleaved caspase-3 (c-caspase-3) protein and PTEN/Akt pathway. However, in the groups of HG-N, DM, HG-N+I/R and DM+I/R, NAC can significantly reduce oxidative stress injury and apoptosis rate of myocytes, promote the Bcl-2 and DJ-1 molecules, inhibit BAX and c-caspase-3 protein and PTEN/Akt pathway. Compared with HG-N+I/R+NAC and DM+I/R+NAC groups, the oxidative stress injury, apoptosis rate of myocardial cells and heart tissues increased after the knockdown of DJ-1, the expression of Bcl-2 and DJ-1 were inhibited, the BAX and c-caspase-3 expression was increased, and PTEN/Akt pathway was activated. Taken together, the findings suggest that NAC can reduce I/R injury in diabetic myocardium by up-regulating the PTEN/Akt pathway through the level of DJ-1.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Diabetic Cardiomyopathies/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , PTEN Phosphohydrolase/metabolism , Protein Deglycase DJ-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Male , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Protein Deglycase DJ-1/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction
16.
Cardiovasc Drugs Ther ; 34(4): 443-461, 2020 08.
Article in English | MEDLINE | ID: mdl-32335797

ABSTRACT

PURPOSE: Ticagrelor, a P2Y12 receptor antagonist, and dapagliflozin, a sodium-glucose-cotransporter-2 inhibitor, suppress the activation of the NLRP3 inflammasome. The anti-inflammatory effects of dapagliflozin depend on AMPK activation. Also, ticagrelor can activate AMPK. We assessed whether dapagliflozin and ticagrelor have additive effects in attenuating the progression of diabetic cardiomyopathy in T2DM mice. METHODS: Eight-week-old BTBR and wild-type mice received no drug, dapagliflozin (1.5 mg/kg/day), ticagrelor (100 mg/kg/day), or their combination for 12 weeks. Heart function was evaluated by echocardiography and heart tissue samples were assessed for fibrosis, apoptosis, qRT-PCR, and immunoblotting. RESULTS: Both drugs attenuated the progression of diabetic cardiomyopathy as evident by improvements in left ventricular end-systolic and end-diastolic volumes and left ventricular ejection fraction, which were further improved by the combination. Both drugs attenuated the activation of the NOD-like receptor 3 (NLRP3) inflammasome and fibrosis. The effect of the combination was significantly greater than each drug alone on myocardial tissue necrotic factorα (TNFα) and interleukin-6 (IL-6) levels, suggesting additive effects. The combination had also a greater effect on ASC, collagen-1, and collagen-3 mRNA levels than each drug alone. While both drugs activated adenosine mono-phosphate kinase (AMPK), only dapagliflozin activated mTOR and increased RICTOR levels. Moreover, only dapagliflozin decreased myocardial BNP and Caspase-1 mRNA levels, and the effects of dapagliflozin on NLRP3 and collagen-3 mRNA levels were significantly greater than those of ticagrelor. CONCLUSIONS: Both dapagliflozin and ticagrelor attenuated the progression of diabetic cardiomyopathy, the activation of the NLRP3 inflammasome, and fibrosis in BTBR mice with additive effects of the combination. While both dapagliflozin and ticagrelor activated AMPK, only dapagliflozin activated mTOR complex 2 (mTORC2) in hearts of BTBR mice.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Glucosides/pharmacology , Inflammasomes/metabolism , Myocytes, Cardiac/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , TOR Serine-Threonine Kinases/metabolism , Ticagrelor/pharmacology , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/enzymology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Disease Progression , Enzyme Activation , Fibrosis , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Signal Transduction , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
17.
Circ Res ; 126(10): e80-e96, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32134364

ABSTRACT

RATIONALE: Diabetes mellitus is a complex, multisystem disease, affecting large populations worldwide. Chronic CaMKII (Ca2+/calmodulin-dependent kinase II) activation may occur in diabetes mellitus and be arrhythmogenic. Diabetic hyperglycemia was shown to activate CaMKII by (1) O-linked attachment of N-acetylglucosamine (O-GlcNAc) at S280 leading to arrhythmia and (2) a reactive oxygen species (ROS)-mediated oxidation of CaMKII that can increase postinfarction mortality. OBJECTIVE: To test whether high extracellular glucose (Hi-Glu) promotes ventricular myocyte ROS generation and the role played by CaMKII. METHODS AND RESULTS: We tested how extracellular Hi-Glu influences ROS production in adult ventricular myocytes, using DCF (2',7'-dichlorodihydrofluorescein diacetate) and genetically targeted Grx-roGFP2 redox sensors. Hi-Glu (30 mmol/L) significantly increased the rate of ROS generation-an effect prevented in myocytes pretreated with CaMKII inhibitor KN-93 or from either global or cardiac-specific CaMKIIδ KO (knockout) mice. CaMKII KO or inhibition also prevented Hi-Glu-induced sarcoplasmic reticulum Ca2+ release events (Ca2+ sparks). Thus, CaMKII activation is required for Hi-Glu-induced ROS generation and sarcoplasmic reticulum Ca2+ leak in cardiomyocytes. To test the involvement of O-GlcNAc-CaMKII pathway, we inhibited GlcNAcylation removal by Thiamet G (ThmG), which mimicked the Hi-Glu-induced ROS production. Conversely, inhibition of GlcNAcylation (OSMI-1 [(αR)-α-[[(1,2-dihydro-2-oxo-6-quinolinyl)sulfonyl]amino]-N-(2-furanylmethyl)-2-methoxy-N-(2-thienylmethyl)-benzeneacetamide]) prevented ROS induction in response to either Hi-Glu or ThmG. Moreover, in a CRSPR-based knock-in mouse in which the functional GlcNAcylation site on CaMKIIδ was ablated (S280A), neither Hi-Glu nor ThmG induced myocyte ROS generation. So CaMKIIδ-S280 is required for the Hi-Glu-induced (and GlcNAc dependent) ROS production. To identify the ROS source(s), we used different inhibitors of NOX (NADPH oxidase) 2 (Gp91ds-tat peptide), NOX4 (GKT137831), mitochondrial ROS (MitoTempo), and NOS (NO synthase) pathway inhibitors (L-NAME, L-NIO, and L-NPA). Only NOX2 inhibition or KO prevented Hi-Glu/ThmG-induced ROS generation. CONCLUSIONS: Diabetic hyperglycemia induces acute cardiac myocyte ROS production by NOX2 that requires O-GlcNAcylation of CaMKIIδ at S280. This novel ROS induction may exacerbate pathological consequences of diabetic hyperglycemia.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Diabetic Cardiomyopathies/etiology , Glucose/toxicity , Hyperglycemia/complications , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/physiopathology , Enzyme Activation , Glutaredoxins/genetics , Glutaredoxins/metabolism , Glycosylation , Humans , Hyperglycemia/enzymology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/enzymology , NADPH Oxidase 2/deficiency , NADPH Oxidase 2/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology
18.
J Diabetes Res ; 2019: 4857921, 2019.
Article in English | MEDLINE | ID: mdl-31871948

ABSTRACT

Although the underlying mechanisms of diabetes-induced myocardial injury have not been fully illuminated, the inflammation reaction has been reported intently linked with diabetes. The nucleotide binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome, the key component of pyroptosis, is involved in inflammation reaction, which may be one of the important mechanisms in diabetes-induced myocardial injury. The purpose of this study was to investigate the changes of NLRP3 inflammasome and pyroptosis in high glucose-induced H9C2 cardiac cell injury and investigate whether overexpression of mitochondrial aldehyde dehydrogenase 2 (ALDH2) can reduce the occurrence of pyroptosis. The H9C2 cardiac cells were exposed to 35 mM glucose for 24 h to induce cytotoxicity. Mitochondrial ALDH2 overexpression cardiac cell line was constructed. The results showed in high glucose condition that ALDH2 overexpression significantly increased H9C2 cardiac cell viability, increased mitochondrial ALDH2 activity and protein expression, and reduced mitochondrial reactive oxygen species (ROS) production, 4-hydroxynonenal (4-HNE), and lactate dehydrogenase (LDH) levels; meanwhile, the pyroptosis key components-NLRP3 inflammasome-related proteins, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), cysteine-containing aspartate specific protease 1 (Caspase-1), and interleukin-18 (IL-18) protein expressions-were significantly decreased, and IL-18 and interleukin-1ß (IL-1ß) levels were also decreased. In high glucose-induced cardiac cell injury, ALDH2 overexpression may reduce ROS production, thereby inhibiting the activation of NLRP3 inflammation and cell pyroptosis. ALDH2 gene might play the potential role in the treatment of high glucose-induced H9C2 cardiac cell injury.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/biosynthesis , Diabetic Cardiomyopathies/prevention & control , Glucose/toxicity , Inflammasomes/drug effects , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Cardiotoxicity , Cell Line , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/immunology , Enzyme Induction , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mitochondria, Heart/enzymology , Mitochondria, Heart/immunology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/immunology , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
19.
J Diabetes Res ; 2019: 8905917, 2019.
Article in English | MEDLINE | ID: mdl-31886288

ABSTRACT

Diabetic cardiomyopathy is a common cardiac condition in patients with diabetes mellitus, which results in cardiac hypertrophy and subsequent heart failure. Chronic inflammation in the diabetic heart results in loss of cardiomyocytes and subsequentially cardiac dysfunction. Accumulated evidence implicated pyroptosis as a vital contributor to the hyperglycemia-induced cardiac inflammatory response. Exendin-4, a GLP analog, promotes survival of cardiomyocytes in cardiovascular diseases, including diabetic cardiomyopathy. However, the role of Exendin-4 in cardiac pyroptosis remains to be elucidated. Our study revealed that Exendin-4 treatment protected against heart remolding and dysfunction and attenuated cardiac inflammation in high-fat diet-fed rats. The activity of caspase-1 and production of pyroptotic cytokines were significantly inhibited by Exendin-4 treatment in the diabetic heart and in high glucose-treated cardiomyocytes as well. In an effort to understand the signaling mechanisms underlying the antipyroptotic property of Exendin-4, we found that blockade of AMPK, an oxidative stress sensor, activity diminished the antipyroptotic property of Exendin-4. Phosphorylation of AMPK resulted in degeneration of TXNIP that promoted the activation of the NLRP3 inflammasome. Exendin-4 treatment decreased the protein level of TXNIP. Moreover, RNA silencing of TXNIP mimicked the antipyroptotic actions of Exendin-4. These findings promoted us to propose a new signaling pathway mediating cardioprotective effect of Exendin-4 under hyperglycemic conditions: Exendin-4 → ROS↓ → pAMPK↑ → TXNIP↓ → caspase-1↓ → IL-1ß and IL-18↓ → pyroptosis↓. In general, our study identified Exendin-4 as a pyroptotic inhibitor protecting against hyperglycemia-induced cardiomyocyte pyroptosis via the AMPK-TXNIP pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Blood Glucose/drug effects , Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Exenatide/pharmacology , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Myocytes, Cardiac/drug effects , Pyroptosis/drug effects , Thioredoxins/metabolism , Animals , Biomarkers/blood , Blood Glucose/metabolism , Carrier Proteins/genetics , Caspase 1/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphorylation , Proteolysis , Reactive Oxygen Species/metabolism , Signal Transduction , Thioredoxins/genetics
20.
J Cardiovasc Pharmacol ; 74(3): 235-245, 2019 09.
Article in English | MEDLINE | ID: mdl-31306370

ABSTRACT

A plethora of studies have demonstrated that cardiomyopathy represents a serious source of morbidity and mortality in patients with diabetes. Yet, the underlying mechanisms of diabetic cardiomyopathy are still poorly understood. Of interest, cytochrome P450 2J (CYP2J) and soluble epoxide hydrolase (sEH) are known to control the maintenance of cardiovascular health through the regulation of cardioprotective epoxyeicosatrienoic acids (EETs) and its less active products, dihydroxyeicosatrienoic acids (DHETs). Therefore, we examined the role of the aforementioned pathway in the development of diabetic cardiomyopathy. Our diabetic model initiated cardiomyopathy as indexed by the increase in the expression of hypertrophic markers such as NPPA. Furthermore, diabetic cardiomyopathy was associated with a low level of cardiac EETs and an increase of the DHETs/EETs ratio both in vivo and in cardiac cells. The modulation in EETs and DHETs was attributed to the increase of sEH and the decrease of CYP2J. Interestingly, the reduction of sEH attenuates cardiotoxicity mediated by high glucose in cardiac cells. Mechanistically, the beneficial effect of sEH reduction might be due to the decrease of phosphorylated ERK1/2 and p38. Overall, the present work provides evidence that diabetes initiates cardiomyopathy through the increase in sEH, the reduction of CYP2J, and the decrease of cardioprotective EETs.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Diabetic Cardiomyopathies/enzymology , Eicosanoids/metabolism , Epoxide Hydrolases/metabolism , Myocytes, Cardiac/enzymology , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blood Glucose/metabolism , Cell Line , Cytochrome P-450 Enzyme System/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diet, High-Fat , Epoxide Hydrolases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Male , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Obesity/complications , Obesity/enzymology , Phosphorylation , Signal Transduction , Streptozocin , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...