Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
1.
Cell Stress Chaperones ; 29(2): 272-284, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38485044

ABSTRACT

Long-term hyperglycemia can lead to diabetic cardiomyopathy (DCM), a main lethal complication of diabetes. However, the mechanisms underlying DCM development have not been fully elucidated. Heat shock protein A12A (HSPA12A) is the atypic member of the Heat shock 70kDa protein family. In the present study, we found that the expression of HSPA12A was upregulated in the hearts of mice with streptozotocin-induced diabetes, while ablation of HSPA12A improved cardiac systolic and diastolic dysfunction and increased cumulative survival of diabetic mice. An increased expression of HSPA12A was also found in H9c2 cardiac cells following treatment with high glucose (HG), while overexpression of HSPA12A-enhanced the HG-induced cardiac cell death, as reflected by higher levels of propidium iodide cells, lactate dehydrogenase leakage, and caspase 3 cleavage. Moreover, the HG-induced increase of oxidative stress, as indicated by dihydroethidium staining, was exaggerated by HSPA12A overexpression. Further studies demonstrated that the HG-induced increases of protein kinase B and forkhead box transcription factors 1 phosphorylation were diminished by HSPA12A overexpression, while pharmacologically inhibition of protein kinase B further enhanced the HG-induced lactate dehydrogenase leakage in HSPA12A overexpressed cardiac cells. Together, the results suggest that hyperglycemia upregulated HSPA12A expression in cardiac cells, by which induced cell death to promote DCM development. Targeting HSPA12A may serve as a potential approach for DCM management.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Hyperglycemia , Animals , Mice , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/metabolism , Heat-Shock Proteins/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Lactate Dehydrogenases/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats
2.
Ann Clin Lab Sci ; 54(1): 17-25, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38514055

ABSTRACT

OBJECTIVE: Diabetic cardiomyopathy (DCM) is the most common cardiovascular complication of type 2 diabetes mellitus (T2DM). Patients affected with DCM face a notably higher risk of progressing to congestive heart failure compared to other populations. Myocardial hypertrophy, a clearly confirmed pathological change in DCM, plays an important role in the development of DCM, with abnormal Ca2+ homeostasis serving as the key signal to induce myocardial hypertrophy. Therefore, investigating the mechanism of Ca2+ transport is of great significance for the prevention and treatment of myocardial hypertrophy in T2DM. METHODS: The rats included in the experiment were divided into wild type (WT) group and T2DM group. The T2DM rat model was established by feeding the rats with high-fat and high-sugar diets for three months combined with low dose of streptozotocin (100mg/kg). Afterwards, primary rat cardiomyocytes were isolated and cultured, and cardiomyocyte hypertrophy was induced through high-glucose treatment. Subsequently, mechanistic investigations were carried out through transfection with si-STIM1 and oe-STIM1. Western blot (WB) was used to detect the expression of the STIM1, Orai1 and p-CaMKII. qRT-PCR was used to detect mRNA levels of myocardial hypertrophy marker proteins. Cell surface area was detected using TRITC-Phalloidin staining, and intracellular Ca2+ concentration in cardiomyocytes was measured using Fluo-4 fluorescence staining. RESULTS: Through animal experiments, an upregulation of Orai1 and STIM1 was revealed in the rat model of myocardial hypertrophy induced by T2DM. Meanwhile, through cell experiments, it was found that in high glucose (HG)-induced hypertrophic cardiomyocytes, the expression of STIM1, Orai1, and p-CaMKII was upregulated, along with increased levels of store-operated Ca2+ entry (SOCE) and abnormal Ca2+ homeostasis. However, when STIM1 was downregulated in HG-induced cardiomyocytes, SOCE levels decreased and p-CaMKII was downregulated, resulting in an improvement in myocardial hypertrophy. To further elucidate the mechanism of action involving SOCE and CaMKII in T2DM-induced myocardial hypertrophy, high-glucose cardiomyocytes were respectively treated with BTP2 (SOCE blocker) and KN-93 (CaMKII inhibitor), and the results showed that STIM1 can mediate SOCE, thereby affecting the phosphorylation level of CaMKII and improving cardiomyocyte hypertrophy. CONCLUSION: STIM1/Orai1-mediated SOCE regulates p-CaMKII levels, thereby inducing myocardial hypertrophy in T2DM.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium , Cardiomegaly , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Glucose , ORAI1 Protein , Stromal Interaction Molecule 1 , Animals , Rats , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Diabetes Mellitus, Type 2/complications , Glucose/metabolism , Glucose/pharmacology , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Up-Regulation , Diabetic Cardiomyopathies/complications , Rats, Sprague-Dawley , Male
3.
Nutrients ; 16(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542759

ABSTRACT

Previous studies have reported the therapeutic effects of oleuropein (OP) consumption on the early stage of diabetic nephropathy and diabetic cardiomyopathy. However, the efficacy of OP on the long-course of these diabetes complications has not been investigated. Therefore, in this study, to investigate the relieving effects of OP intake on these diseases, and to explore the underlying mechanisms, db/db mice (17-week-old) were orally administrated with OP (200 mg/kg bodyweight) for 15 weeks. We found that OP reduced expansion of the glomerular mesangial matrix, renal inflammation, renal fibrosis, and renal apoptosis. Meanwhile, OP treatment exerted cardiac anti-fibrotic, anti-inflammatory, and anti-apoptosis effects. Notably, transcriptomic and bioinformatic analyses indicated 290 and 267 differentially expressed genes in the kidney and heart replying to OP treatment, respectively. For long-course diabetic nephropathy, OP supplementation significantly upregulated the cyclic guanosine monophosphate-dependent protein kinase (cGMP-PKG) signaling pathway. For long-course diabetic cardiomyopathy, p53 and cellular senescence signaling pathways were significantly downregulated in response to OP supplementation. Furthermore, OP treatment could significantly upregulate the transcriptional expression of the ATPase Na+/K+ transporting subunit alpha 3, which was enriched in the cGMP-PKG signaling pathway. In contrast, OP treatment could significantly downregulate the transcriptional expressions of cyclin-dependent kinase 1, G two S phase expressed protein 1, and cyclin B2, which were enriched in p53 and cellular senescence signal pathways; these genes were confirmed by qPCR validation. Overall, our findings demonstrate that OP ameliorated long-course diabetic nephropathy and cardiomyopathy in db/db mice and highlight the potential benefits of OP as a functional dietary supplement in diabetes complications treatment.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Diabetic Nephropathies , Iridoid Glucosides , Mice , Animals , Diabetic Nephropathies/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/complications , Tumor Suppressor Protein p53/metabolism , Kidney/metabolism
4.
Molecules ; 28(14)2023 Jul 17.
Article in English | MEDLINE | ID: mdl-37513325

ABSTRACT

Diabetes mellitus is a chronic metabolic disorder defined as hyperglycemia and pancreatic ß-cell deterioration, leading to other complications such as cardiomyopathy. The current study assessed the therapeutic effects of phenolic acids extracted from Jasminum sambac phenols of leaves (JSP) against diabetes-induced cardiomyopathy in rats. The rats were divided into four groups, with each group consisting of 20 rats. The rats were given intraperitoneal injections of alloxan monohydrate (150 mg/kg) to induce diabetes. The diabetes-induced groups (III and IV) received treatment for six weeks that included 250 and 500 mg/kg of JSP extract, respectively. In the treated rats, the results demonstrated that JSP extract restored fasting glucose, serum glucose, and hyperlipidemia. Alloxan induced cardiomyopathy, promoted oxidative stress, and altered cardiac function biomarkers, including cardiac troponin I, proBNP, CK-MB, LDH, and IMA. The JSP extract-treated rats showed improved cardiac function indicators, apoptosis, and oxidative stress. In diabetic rats, the mRNA expression of caspase-3, BAX, and Bcl-2 was significantly higher, while Bcl-2, Nrf-2, and HO-,1 was significantly lower. In the treated groups, the expression levels of the BAX, Nrf-2, HO-1, Caspase-3, and Bcl-2 genes were dramatically returned to normal level. According to our findings, the JSP extract prevented cardiomyopathy and heart failure in the hyperglycemic rats by improving cardiac biomarkers and lowering the levels of hyperlipidemia, oxidative stress, apoptosis, hyperglycemia, and hyperlipidemia.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Hyperglycemia , Hyperlipidemias , Jasminum , Metabolic Diseases , Rats , Animals , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/complications , Alloxan , Caspase 3/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , bcl-2-Associated X Protein/metabolism , Oxidative Stress , Hyperglycemia/complications , Glucose/metabolism , Metabolic Diseases/complications , Phenols/pharmacology , Phenols/therapeutic use , Biomarkers/metabolism , Blood Glucose/metabolism
5.
Gac Med Mex ; 159(3): 255-261, 2023.
Article in English | MEDLINE | ID: mdl-37494725

ABSTRACT

Diabetic cardiomyopathy (DCM) is a serious complication of diabetes caused by oxidative stress, inflammation, insulin resistance, myocardial fibrosis, and lipotoxicity; its nature is insidious, complex and difficult to treat. NLRP3 inflammasome triggers the maturation and release of pro-inflammatory cytokines, participates in pathophysiological processes such as insulin resistance and myocardial fibrosis, in addition to being closely related to the development and progression of diabetic cardiomyopathy. The development of inhibitors targeting specific aspects of inflammation suggests that NLRP3 inflammasome can be used to treat diabetic cardiomyopathy. This paper aims to summarize NLRP3 inflammasome mechanism and therapeutic targets in diabetic cardiomyopathy, and to provide new suggestions for the treatment of this disease.


La cardiomiopatía diabética es una complicación grave de la diabetes causada por estrés oxidativo, inflamación, resistencia a la insulina, fibrosis miocárdica y lipotoxicidad. Se trata de un padecimiento insidioso, complejo y difícil de tratar. El inflamasoma NLRP3 desencadena la maduración y liberación de citoquinas proinflamatorias, participa en procesos fisiopatológicos como la resistencia a la insulina y la fibrosis miocárdica, además de estar estrechamente relacionado con la aparición y progresión de la cardiomiopatía diabética. El desarrollo de inhibidores dirigidos a aspectos específicos de la inflamación sugiere que el inflamasoma NLRP3 puede utilizarse para tratar la cardiomiopatía diabética. Este artículo pretende resumir el mecanismo y las dianas terapéuticas del inflamasoma NLRP3 en la cardiomiopatía diabética, así como aportar nuevas sugerencias para el tratamiento de esta enfermedad.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Insulin Resistance , Animals , Humans , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/complications , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Inflammation/etiology , Fibrosis
6.
Adv Sci (Weinh) ; 10(7): e2206171, 2023 03.
Article in English | MEDLINE | ID: mdl-36596646

ABSTRACT

Reactive oxygen species (ROS) are closely associated with the progression of diabetic cardiomyopathy (DCM) and can be regarded as one of its early biomarkers. Magnetic resonance imaging (MRI) is emerging as a powerful tool for the detection of cardiac abnormalities, but the sensitive and direct ROS-response MRI probe remains to be developed. This restricts the early diagnosis of DCM and prevents timely clinical interventions, resulting in serious and irreversible pathophysiological abnormalities. Herein, a novel ROS-response contrast-enhanced MRI nanoprobe (RCMN) is developed by multi-functionalizing fluorinated carbon nanosheets (FCNs) with multi-hydroxyl and 2,2,6,6-tetramethylpiperidin-1-oxyl groups. RCMNs capture ROS and then gather in the heart provisionally, which triggers MRI signal changes to realize the in vivo detection of ROS. In contrast to the clinical MRI agents, the cardiac abnormalities of disease mice is detected 8 weeks in advance with the assistance of RCMNs, which greatly advances the diagnostic window of DCM. To the best of the knowledge, this is the first ROS-response metal-free T2 -weighted MRI probe for the early diagnosis of DCM mice model. Furthermore, RCMNs can timely scavenge excessively produced ROS to alleviate oxidative stress.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Mice , Animals , Reactive Oxygen Species , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/complications , Diabetes Mellitus, Experimental/complications , Oxidative Stress , Disease Models, Animal , Magnetic Resonance Imaging , Early Diagnosis
7.
Appl Biochem Biotechnol ; 195(6): 3533-3545, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36622632

ABSTRACT

Chronic hyperglycemia induces reactive oxygen species that have an essential function in tissue injuries in cases of diabetic cardiomyopathy. The mechanism of the absent in melanoma 2 (AIM2)-associated inflammasome response in diabetic cardiomyopathy is unknown. Therefore, this study was performed to investigate the role of AIM2 and its molecular mechanisms. Diabetic rats received 1 × 108 viral injections of 5'-GGTCACCAGTTCCTCAGTT-3' (n = 15) or 5'-TTCTCCGAACGTGTCACGT-3' (negative control group, n = 15). Normal rats (n = 15) and diabetic rats (n = 15) were also included in the experiment. Ex vivo study was performed on primary cardiomyocytes for different concentrations of glucose. AIM2 inhibition did not affect any of the metabolic parameters (p > 0.05 for all). AIM2 protein levels were significantly increased in rats with diabetes mellitus compared with those in the control group (p < 0.0001, q = 32.044). Also, viral injection (sequence: 5'-GGTCACCAGTTCCTCAGTT-3') decreased the diabetes mellitus-induced increase in expression of AIM2 protein levels (p < 0.0001, q = 27.129). Cardiac dysfunctions were reported in rats with diabetes mellitus characterized by several parameters (p < 0.01 for all). The diabetic myocardium of rats was reported to have higher deposits of extracellular matrix compared to the control rats (p < 0.001). These effects were downregulated by viral injection (sequence: 5'-GGTCACCAGTTCCTCAGTT-3'). Ex vivo research revealed that high glucose concentrations significantly increased AIM2 protein expression, reactive oxygen species, and cell death. AIM2 protein in diabetic cardiomyopathy is associated with reactive oxygen species production and cardiomyocyte death.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Rats , Animals , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/metabolism , Reactive Oxygen Species/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Gene Knockdown Techniques , Myocytes, Cardiac/metabolism , Glucose/pharmacology , Glucose/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics
8.
Peptides ; 159: 170918, 2023 01.
Article in English | MEDLINE | ID: mdl-36435275

ABSTRACT

Following an unprecedented rise in the number of the aged, the incidence of age-related diseases, such as diabetes and cardiovascular disease, is consequently increasing in the world. Type 2 diabetes mellitus (T2DM) is associated with excess cardiovascular morbidity and mortality. The diabetic heart is characterized by increased cardiomyocyte stiffness and fibrotic changes. Despite many factors resulting in cardiomyocyte injury and dysfunction in diabetes, insulin resistance is still a critical etiology of diabetic cardiomyopathy. Preclinical and clinical studies have revealed an intriguing role for galanin in the pathogenesis of insulin resistance and diabetic heart disease. A significant change in plasma galanin levels occurred in patients suffering from type 2 diabetes or cardiomyocyte injury. In turn, galanin may also distinctly mitigate hyperglycemia and insulin resistance in diabetes as well as increase glucose metabolism and mitochondrial biogenesis in cardiac muscle. Here, we critically review current data about the multivariate relationship among galanin, insulin resistance, and cardiac muscle to comprehensively evaluate the protective role of galanin and its receptors for the diabetic heart and to determine whether galanin receptor 2 agonists potentially represent a feasible way to treat diabetic cardiomyopathy in the future.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Insulin Resistance , Neuropeptides , Humans , Aged , Galanin/genetics , Galanin/therapeutic use , Insulin Resistance/genetics , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/complications , Insulin/metabolism
9.
Inflammation ; 46(1): 146-160, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35997998

ABSTRACT

A simultaneous increase in the prevalence of diabetes mellitus (DM), a risk factor for cardiovascular diseases (CVDs), has contributed to the escalation of CVD related mortalities. To date, oxidative stress and inflammation are increasingly recognized as significant drivers of cardiovascular complications in patients with diabetes. Therefore, this study aims to explore the correlation between oxidative stress, inflammation, and hematological indices in diabetic patients with CVDs. Patients were allocated into five groups: healthy controls; nondiabetic patients with myocardial infarction; diabetic patients with myocardial infarction; nondiabetic patients with heart failure; and diabetic patients with heart failure. The results revealed that the malondialdehyde levels were increased; whereas superoxide dismutase enzyme activities were markedly reduced in all CVD groups compared with those of healthy controls. Although the mRNA expression levels of interleukin (IL)-6, IL-18, and IL-38 were significantly increased, those of the anti-inflammatory cytokine, IL-35, have been reduced in all CVD groups compared with healthy controls. Regarding hematological indices, hematocrit, red blood cell distribution width, mean platelet (PLT) volume, plateletcrit, PLT distribution width, leukocyte count, and PLT-to-lymphocyte and neutrophil-to-lymphocyte ratios were markedly increased in the diabetic and nondiabetic CVD groups compared with those of the healthy controls. Oxidative stress and cytokine biomarkers may play a significant role in the complications of diabetic cardiomyopathy. Moreover, hematological indices are particularly sensitive to systemic inflammatory changes and are novel markers for the early detection of diabetic cardiomyopathy.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus , Diabetic Cardiomyopathies , Heart Failure , Myocardial Infarction , Humans , Cytokines , Diabetic Cardiomyopathies/complications , Oxidative Stress , Inflammation/complications , Interleukin-6 , Heart Failure/complications , Interleukins
10.
J Food Biochem ; 46(11): e14475, 2022 11.
Article in English | MEDLINE | ID: mdl-36219759

ABSTRACT

One of the major complications of diabetes mellitus (DM) is diabetic cardiomyopathy (DCM) due to the multifaceted therapy involved. Here, we evaluated the combinatorial effect of Moringa leaf (ML) and seed (MS) supplemented diets plus acarbose (ACA) on cardiac acetylcholinesterase (AChE), adenosine triphosphatase (ATPase), adenosine deaminase (ADA), monoamine oxidase (MAO), arginase, angiotensin-I converting enzyme (ACE), and lactate dehydrogenase (LDH) activities, thiobarbituric acid reactive species (TBARS), and thiols levels. The diets and ACA (25 mg/kg) were administered for 14 days. The fasting blood glucose level (FBGL), cardiac AChE, ATPase, ADA, MAO, arginase, ACE, LDH activities, and TBARS and thiol levels were determined. Relative to the normal rats, the biomarkers were significantly increased in DM rats but were suppressed significantly in the diets plus ACA-treated rats while improving antioxidant status, with the 4% Moringa plus ACA proving outstanding compared to individual ML/MS and ACA. In addition, ML-supplemented diets with/without ACA had better effects compared to MS with/without ACA, respectively. In conclusion, the combination of ML/MS supplemented diets and ACA synergistically modulates the tested biochemicals. However, the effect on blood vessels and the nerves that control the heart, stiffness of left ventricular (LV) hypertrophy, fibrosis, cell signaling abnormalities, related gene expression, clinical trials, and echocardiology studies should be further investigated to affirm this claim. PRACTICAL APPLICATIONS: Moringa oleifera has been a vocal appetite in mitigating cardiovascular disease induced by diabetes, but the formulation of a medicinal diet as an ameliorative route of attention to the pathology is fairly addressed, not talking of its combination with the synthetic antidiabetic drug, such as ACA. Based on this experiment, it is imperative to explore such an idea. This research shows that co-administration of moringa leaf/seed formulated diets plus ACA exhibits a synergistic effect in DCM management. However, further research is needed in this field of experiment.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Dietary Supplements , Moringa , Animals , Rats , Acarbose/therapeutic use , Acetylcholinesterase/metabolism , Adenosine Triphosphatases/metabolism , Antioxidants/metabolism , Arginase , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/pathology , Diet , Monoamine Oxidase/metabolism , Moringa/chemistry , Rats, Wistar , Renin-Angiotensin System , Thiobarbituric Acid Reactive Substances
11.
Adv Sci (Weinh) ; 9(31): e2202590, 2022 11.
Article in English | MEDLINE | ID: mdl-36180407

ABSTRACT

Diabetes manifests as chronic inflammation and leads to the development diabetic cardiomyopathy (DCM). Targeting key proteins in inflammatory signaling may provide new therapy for DCM. In this study, the authors explore the pharmacological effects and mechanisms of Schisandrin B (Sch B), a natural compound with anti-inflammatory activity against DCM. It is shown that Sch B prevents high-level glucose (HG)-induced hypertrophic and fibrotic responses in cultured cardiomyocytes. RNA sequencing and inflammatory qPCR microarray show that Sch B mainly affects myeloid differentiation primary response 88 (MyD88)-dependent inflammatory gene expression in HG-challenged cardiomyocytes. Further studies indicate that Sch B directly binds to and inhibits MyD88 activation, but does not alter MyD88-independent Toll-like receptor signaling in vivo and in vitro. Inhibiting or silencing MyD88 is associated with reduced levels of HG-induced inflammatory cytokines and myocardial injuries in vitro. Treatment of type 1 and type 2 diabetic mice with Sch B protects heart function, reduces myocardial injuries, and decreases secretion of inflammatory cytokines. Cardiomyocyte-specific MyD88 knockout also protects mice against cardiac inflammation and injury in type 1 diabetic mice. In conclusion, these studies show that cardiomyocyte MyD88 plays an apathogenetic role in DCM and Sch B specifically targets MyD88 to reduce inflammatory DCM.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Animals , Mice , Cytokines/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/prevention & control , Inflammation/drug therapy , Inflammation/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/therapeutic use
12.
Biomed Pharmacother ; 153: 113382, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36076522

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy (DCM) is linked to disturbance in cardiac glucose handling and increased cardiac glycogen storage. This study tested the potential role of sacubitril/valsartan on the progression of DCM in high fat diet (HFD)/streptozotocin (STZ)-induced type 2 diabetic rats compared to valsartan alone, including their effects on the cardiac glycophagy process. MATERIALS AND METHODS: Rats were fed on HFD for 6 weeks followed by single low-dose STZ (35 mg/kg). After confirming hyperglycemia, diabetic rats were continued on HFD and divided into three subgroups: Untreated-diabetic, Valsartan-treated diabetic and Sacubitril/valsartan-treated diabetic groups; in addition to a control group. Changes in ECG, blood glucose, serum insulin, lipid profile, and Homeostasis model of assessment of insulin resistance (HOMA-IR) were assessed and the degree of cardiac fibrosis was examined. Cardiac glycogen content and glycophagy process were evaluated. RESULTS: Sacubitril/valsartan administration to diabetic rats resulted in improvement of metabolic changes more than valsartan alone. Also, sacubitril/valsartan effectively prevented diabetes-associated cardiac hypertrophy, QTc prolongation, and fibrosis. Finally, cardiac glycogen concentrations in diabetic rats were decreased by sacubitril/valsartan combination, coupled with significant induction of glycophagy process in the diabetic rats' heart. CONCLUSION: Sacubitril/valsartan therapy provides a more favorable metabolic and cardioprotective response compared to valsartan alone in a rat model of DCM. These findings may be due to a direct cardioprotective impact of sacubitril/valsartan and secondary beneficial effects of improved hyperglycemia and dyslipidemia. In addition, these beneficial cardiac effects could be attributed to the induction of the glycophagy process and alleviating cardiac glycogen overload.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Heart Failure , Hyperglycemia , Aminobutyrates , Animals , Biphenyl Compounds/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/prevention & control , Drug Combinations , Glycogen/pharmacology , Heart Failure/drug therapy , Hyperglycemia/drug therapy , Mice , Rats , Stroke Volume , Tetrazoles/pharmacology , Tetrazoles/therapeutic use , Valsartan/pharmacology , Valsartan/therapeutic use
13.
Biomed Pharmacother ; 154: 113583, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994819

ABSTRACT

The prevalence of cardiovascular complications in diabetes has become one of the major cause of diabetes related morbidity/mortality. The onset and progression of diabetic cardiomyopathy (DCM) has been majorly linked to lipid alterations, oxidative stress, inflammation and apoptosis. This present study investigated the cardioprotective role of Lycium chinense leaf extract (LCME) in fructose/streptozotocin induced diabetic rats. Diabetic animals were orally gavaged with LCME (100 and 400 mg/kg) for five weeks. The results indicated that diabetic rats showed increased blood glucose concentration, serum cardiac function markers (troponin T, creatine kinase-MB, aspartate aminotransferase and lactate dehydrogenase) and lipid profile (triglycerides and cholesterol). In addition, the cardiac tissues of diabetic rats showed increased levels of nuclear factor-κB (NF-κB), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL 1ß), interleukin 6 (IL-6), caspase-3 and malondialdehyde as well as significantly reduced activities of catalase, superoxide dismutase, reduced glutathione and glutathione peroxidase. LCME significantly ameliorated hyperglycemia and markedly decreased serum concentrations of troponin T, creatine kinase-MB, aspartate aminotransferase and lactate dehydrogenase, triglycerides and cholesterol. Furthermore, LCME notably suppressed cardiac oxido-inflammatory mediators and boosted cardiac antioxidant defense. Histopathologically, LCME restored cardiac structural alterations and also suppressed the immunohistochemical expression of collagen IV, smooth muscle alpha-actin (α-SMA) and p53, while Bcl2 expression was significantly increased. In conclusion, our result indicated that LCME protected against diabetic cardiomyopathy suppressing oxidative stress, inflammation, apoptosis and fibrosis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Lycium , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Aspartate Aminotransferases/metabolism , Biomarkers/metabolism , Creatine Kinase/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/prevention & control , Inflammation/pathology , Lactate Dehydrogenases/metabolism , Lipids , Lycium/chemistry , Oxidative Stress , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Triglycerides , Troponin T/metabolism
14.
Am J Physiol Heart Circ Physiol ; 323(1): H176-H200, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35657616

ABSTRACT

Diabetes is a major risk factor for cardiovascular diseases, including diabetic cardiomyopathy, atherosclerosis, myocardial infarction, and heart failure. As cardiovascular disease represents the number one cause of death in people with diabetes, there has been a major emphasis on understanding the mechanisms by which diabetes promotes cardiovascular disease, and how antidiabetic therapies impact diabetic heart disease. With a wide array of models to study diabetes (both type 1 and type 2), the field has made major progress in answering these questions. However, each model has its own inherent limitations. Therefore, the purpose of this guidelines document is to provide the field with information on which aspects of cardiovascular disease in the human diabetic population are most accurately reproduced by the available models. This review aims to emphasize the advantages and disadvantages of each model, and to highlight the practical challenges and technical considerations involved. We will review the preclinical animal models of diabetes (based on their method of induction), appraise models of diabetes-related atherosclerosis and heart failure, and discuss in vitro models of diabetic heart disease. These guidelines will allow researchers to select the appropriate model of diabetic heart disease, depending on the specific research question being addressed.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Heart Failure , Myocardial Infarction , Animals , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/complications , Heart Failure/etiology , Humans , Hypoglycemic Agents , Myocardial Infarction/complications
15.
Chem Biodivers ; 19(3): e202100591, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35119190

ABSTRACT

Diabetic cardiomyopathy is one of the major complications in type 2 diabetes associated with myocardial structure abnormality and major cause of morbidity in type 2 diabetic patients. Biochanin A is a methylated isoflavone present in flowering tops of Trifolium pratense reported for anti-inflammatory, anti-oxidant, anti-infective, anti-cancer and anti-diabetic activity. The study was designed to assess the efficacy of Biochanin A in type 2 diabetic cardiomyopathy. Type 2 diabetes was induced in rats feeding high fat diet for two weeks and administration of single low dose of streptozotocin. Biochanin A was administered for 16 weeks orally once in a day (10, 20 and 40 mg/kg of body weight). Various parameters such as blood glucose, cardiac markers, oxidative stress and hemodynamic parameters, immunohistochemical, histopathological investigation and SIRT1 expression were measured at the end of the study. Biochanin A treatment resulted into reduction in plasma concentration of cardiac markers along with reduction in hyperglycemia, hyperlipidemia and oxidative stress in cardiac tissue. Biochanin A treated animals also demonstrated improvement in hemodynamic parameters. Diabetic animals treated with different doses of Biochanin A shown increased SIRT1 expression in cardiac tissue, and also confirmed reduced cardiac hypertrophy and cardiac protection in histopathological study. Outcome of the study indicates that Biochanin A is the potential candidate to control hyperglycemia, oxidative stress and improve SIRT1 expression in cardiac tissue. Biochanin A might be considered as potential candidate to control progression of cardiomyopathy in type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/drug therapy , Genistein , Humans , Oxidative Stress , Rats , Sirtuin 1/metabolism
16.
Rev Clin Esp (Barc) ; 222(2): 100-111, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35115137

ABSTRACT

The relationship between diabetes and heart failure is complex and bidirectional. Nevertheless, the existence of a cardiomyopathy attributable exclusively to diabetes has been and is still the subject of controversy, due, among other reasons, to a lack of a consensus definition. There is also no unanimous agreement in terms of the physiopathogenic findings that need to be present in the definition of diabetic cardiomyopathy or on its classification, which, added to the lack of diagnostic methods and treatments specific for this disease, limits its general understanding. Studies conducted on diabetic cardiomyopathy, however, suggest a unique physiopathogenesis different from that of other diseases. Similarly, new treatments have been shown to play a potential role in this disease. The following review provides an update on diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Heart Failure , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/therapy , Heart Failure/diagnosis , Heart Failure/etiology , Heart Failure/therapy , Humans
17.
Diabetes ; 71(2): 298-314, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34844991

ABSTRACT

Cardiometabolic diseases, including diabetes and its cardiovascular complications, are the global leading causes of death, highlighting a major unmet medical need. Over the past decade, mitsugumin 53 (MG53), also called TRIM72, has emerged as a powerful agent for myocardial membrane repair and cardioprotection, but its therapeutic value is complicated by its E3 ligase activity, which mediates metabolic disorders. Here, we show that an E3 ligase-dead mutant, MG53-C14A, retains its cardioprotective function without causing metabolic adverse effects. When administered in normal animals, both the recombinant human wild-type MG53 protein (rhMG53-WT) and its E3 ligase-dead mutant (rhMG53-C14A) protected the heart equally from myocardial infarction and ischemia/reperfusion (I/R) injury. However, in diabetic db/db mice, rhMG53-WT treatment markedly aggravated hyperglycemia, cardiac I/R injury, and mortality, whereas acute and chronic treatment with rhMG53-C14A still effectively ameliorated I/R-induced myocardial injury and mortality or diabetic cardiomyopathy, respectively, without metabolic adverse effects. Furthermore, knock-in of MG53-C14A protected the mice from high-fat diet-induced metabolic disorders and cardiac damage. Thus, the E3 ligase-dead mutant MG53-C14A not only protects the heart from acute myocardial injury but also counteracts metabolic stress, providing a potentially important therapy for the treatment of acute myocardial injury in metabolic disorders, including diabetes and obesity.


Subject(s)
Membrane Proteins/genetics , Metabolic Syndrome/genetics , Myocardial Reperfusion Injury/prevention & control , Animals , Cells, Cultured , Cytoprotection/genetics , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Diet, High-Fat , Female , Heart/physiopathology , Humans , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Signal Transduction/genetics
18.
Molecules ; 26(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34885867

ABSTRACT

Metabolic disorders often lead to cardiac complications. Metabolic deregulations during diabetic conditions are linked to mitochondrial dysfunctions, which are the key contributing factors in cardiac hypertrophy. However, the underlying mechanisms involved in diabetes-induced cardiac hypertrophy are poorly understood. In the current study, we initially established a diabetic rat model by alloxan-administration, which was validated by peripheral glucose measurement. Diabetic rats displayed myocardial stiffness and fibrosis, changes in heart weight/body weight, heart weight/tibia length ratios, and enhanced size of myocytes, which altogether demonstrated the establishment of diabetic cardiac hypertrophy (DCH). Furthermore, we examined the expression of genes associated with mitochondrial signaling impairment. Our data show that the expression of PGC-1α, cytochrome c, MFN-2, and Drp-1 was deregulated. Mitochondrial-signaling impairment was further validated by redox-system dysregulation, which showed a significant increase in ROS and thiobarbituric acid reactive substances, both in serum and heart tissue, whereas the superoxide dismutase, catalase, and glutathione levels were decreased. Additionally, the expression levels of pro-apoptotic gene PUMA and stress marker GATA-4 genes were elevated, whereas ARC, PPARα, and Bcl-2 expression levels were decreased in the heart tissues of diabetic rats. Importantly, these alloxan-induced impairments were rescued by N-acetyl cysteine, ascorbic acid, and selenium treatment. This was demonstrated by the amelioration of myocardial stiffness, fibrosis, mitochondrial gene expression, lipid profile, restoration of myocyte size, reduced oxidative stress, and the activation of enzymes associated with antioxidant activities. Altogether, these data indicate that the improvement of mitochondrial dysfunction by protective agents such as N-acetyl cysteine, selenium, and ascorbic acid could rescue diabetes-associated cardiac complications, including DCH.


Subject(s)
Acetylcysteine/therapeutic use , Ascorbic Acid/therapeutic use , Cardiomegaly/drug therapy , Diabetic Cardiomyopathies/drug therapy , Mitochondria, Heart/metabolism , Selenium/therapeutic use , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Biomarkers/blood , Blood Glucose/metabolism , Body Weight/drug effects , Calcium/blood , Cardiomegaly/blood , Cardiomegaly/complications , Cardiomegaly/pathology , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cytochromes c/metabolism , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Down-Regulation , GATA4 Transcription Factor/metabolism , Lipid Peroxidation/drug effects , Lipids/blood , Mitochondria, Heart/drug effects , Myocardium/pathology , Oxidation-Reduction , Oxidative Stress , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Selenium/pharmacology
19.
Nutrients ; 13(11)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34836405

ABSTRACT

Cardiovascular disease (CVD) is the most common cause of morbidity and mortality in developed countries. The prevalence of CVD is much higher in patients with type 2 diabetes mellitus (T2DM), who may benefit from lifestyle changes, which include adapted diets. In this review, we provide the role of different groups of nutrients in patients with T2DM and CVD, as well as dietary approaches that have been associated with better and worse outcomes in those patients. Many different diets and supplements have proved to be beneficial in T2DM and CVD, but further studies, guidelines, and dietary recommendations are particularly required for patients with both diseases.


Subject(s)
Cardiovascular Diseases/diet therapy , Diabetes Mellitus, Type 2/diet therapy , Diabetic Cardiomyopathies/diet therapy , Diet/methods , Nutrition Therapy/methods , Cardiovascular Diseases/complications , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/complications , Dietary Supplements , Humans , Nutrients/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...