Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Pflugers Arch ; 474(1): 33-61, 2022 01.
Article in English | MEDLINE | ID: mdl-34978597

ABSTRACT

Diabetic cardiomyopathy is defined as the myocardial dysfunction that suffers patients with diabetes mellitus (DM) in the absence of hypertension and structural heart diseases such as valvular or coronary artery dysfunctions. Since the impact of DM on cardiac function is rather silent and slow, early stages of diabetic cardiomyopathy, known as prediabetes, are poorly recognized, and, on many occasions, cardiac illness is diagnosed only after a severe degree of dysfunction was reached. Therefore, exploration and recognition of the initial pathophysiological mechanisms that lead to cardiac dysfunction in diabetic cardiomyopathy are of vital importance for an on-time diagnosis and treatment of the malady. Among the complex and intricate mechanisms involved in diabetic cardiomyopathy, Ca2+ mishandling and mitochondrial dysfunction have been described as pivotal early processes. In the present review, we will focus on these two processes and the molecular pathway that relates these two alterations to the earlier stages and the development of diabetic cardiomyopathy.


Subject(s)
Calcium/metabolism , Diabetic Cardiomyopathies/etiology , Mitochondria, Heart/metabolism , Prediabetic State/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Cytosol/metabolism , Diabetic Cardiomyopathies/metabolism , Excitation Contraction Coupling , Humans
2.
J Physiol ; 599(14): 3477-3493, 2021 07.
Article in English | MEDLINE | ID: mdl-33932959

ABSTRACT

Metabolic diseases (MetDs) embrace a series of pathologies characterized by abnormal body glucose usage. The known diseases included in this group are metabolic syndrome, prediabetes and diabetes mellitus types 1 and 2. All of them are chronic pathologies that present metabolic disturbances and are classified as multi-organ diseases. Cardiomyopathy has been extensively described in diabetic patients without overt macrovascular complications. The heart is severely damaged during the progression of the disease; in fact, diabetic cardiomyopathies are the main cause of death in MetDs. Insulin resistance, hyperglycaemia and increased free fatty acid metabolism promote cardiac damage through mitochondria. These organelles supply most of the energy that the heart needs to beat and to control essential cellular functions, including Ca2+ signalling modulation, reactive oxygen species production and apoptotic cell death regulation. Several aspects of common mitochondrial functions have been described as being altered in diabetic cardiomyopathies, including impaired energy metabolism, compromised mitochondrial dynamics, deficiencies in Ca2+ handling, increases in reactive oxygen species production, and a higher probability of mitochondrial permeability transition pore opening. Therefore, the mitochondrial role in MetD-mediated heart dysfunction has been studied extensively to identify potential therapeutic targets for improving cardiac performance. Herein we review the cardiac pathology in metabolic syndrome, prediabetes and diabetes mellitus, focusing on the role of mitochondrial dysfunctions.


Subject(s)
Diabetic Cardiomyopathies , Insulin Resistance , Diabetic Cardiomyopathies/metabolism , Energy Metabolism , Humans , Mitochondria/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Dynamics
3.
Biochim Biophys Acta Mol Basis Dis ; 1867(1): 165984, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33002576

ABSTRACT

Diabetes mellitus-induced heart disease, including diabetic cardiomyopathy, is an important medical problem and is difficult to treat. Diabetes mellitus increases the risk for heart failure and decreases cardiac myocyte function, which are linked to changes in cardiac mitochondrial energy metabolism. The free mitochondrial calcium concentration ([Ca2+]m) is fundamental in activating the mitochondrial respiratory chain complexes and ATP production and is also known to regulate the activity of key mitochondrial dehydrogenases. The mitochondrial calcium uniporter complex (MCUC) plays a major role in mediating mitochondrial Ca2+ import, and its expression and function therefore may have a marked impact on cardiac myocyte metabolism and function. Here, we summarize the pathophysiological role of [Ca2+]m handling and MCUC in the diabetic heart. In addition, we evaluate potential therapeutic targets, directed to the machinery that regulates mitochondrial calcium handling, to alleviate diabetes-related cardiac disease.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Diabetic Cardiomyopathies/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Diabetic Cardiomyopathies/pathology , Humans , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology
4.
Drug Des Devel Ther ; 14: 3337-3350, 2020.
Article in English | MEDLINE | ID: mdl-32884238

ABSTRACT

INTRODUCTION: Diabetic obese patients are susceptible to the development of cardiovascular disease, including hypertension and cardiac dysfunction culminating in diabetic cardiomyopathy (DC), which represents a life-threatening health problem with increased rates of morbidity and mortality. The aim of the study is to characterize the effects of a new benzofuran N-acylhydrazone compound, LASSBio-2090, on metabolic and cardiovascular alterations in Zucker diabetic fatty (ZDF) rats presenting DC. METHODS: Male non-diabetic lean Zucker rats (ZL) and ZDF rats treated with vehicle (dimethylsulfoxide) or LASSBio-2090 were used in this study. Metabolic parameters, cardiovascular function, left ventricle histology and inflammatory protein expression were analyzed in the experimental groups. RESULTS: LASSBio-2090 administration in ZDF rats reduced glucose levels to 85.0 ± 1.7 mg/dL (p < 0.05). LASSBio-2090 also lowered the cholesterol and triglyceride levels from 177.8 ± 31.2 to 104.8 ± 5.3 mg/dL and from 123.0 ± 11.4 to 90.9 ± 4.8 mg/dL, respectively, in obese diabetic rats (p < 0.05). LASSBio-2090 normalized plasma insulin, insulin sensitivity and endothelial function in aortas from diabetic animals (p < 0.05). It also enhanced systolic and diastolic left-ventricular function and reverted myocardial remodeling by blocking the threefold elevation of TNF-α levels in hearts from ZDF rats. CONCLUSION: LASSBio-2090 alleviates metabolic disturbance and cardiomyopathy in an obese and diabetic rat model, thus representing a novel strategy for the treatment of cardiovascular complications in obesity-associated type 2 diabetes mellitus.


Subject(s)
Benzofurans/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , Obesity/drug therapy , Animals , Benzofurans/administration & dosage , Benzofurans/chemistry , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Injections, Intraperitoneal , Male , Molecular Structure , Obesity/metabolism , Rats , Rats, Zucker
5.
Cardiovasc Pathol ; 45: 107181, 2020.
Article in English | MEDLINE | ID: mdl-31865268

ABSTRACT

The present study aimed to evaluate the effects of diabetes on quantitative parameters of right atrial cardiomyocytes of elderly rats. Wistar rats (14 months of age) were divided into two groups: streptozotocin-diabetic rats (DG) and control rats (CG). The groups were sacrificed at 16 months. Ultrafine sections of the right atrium were analyzed by electron microscopy. In elderly diabetic animals, histograms of the frequency distribution of natriuretic peptides according to their size showed increased number of small and medium peptides in relation to large peptides, which increased its numerical density leading to a decrease in the mean diameter of both natriuretic peptides. However, elderly diabetic animals remained normotensive. No significant difference was observed between the groups for the volume density of mitochondria, endoplasmic reticulum, and Golgi apparatus. In conclusion, elderly diabetic rats showed increased functional activity of atrial cardiomyocytes with greater production of natriuretic peptides in association with a quantitative maintenance of cytoplasmic components.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/pathology , Heart Atria/ultrastructure , Myocytes, Cardiac/ultrastructure , Age Factors , Animals , Atrial Natriuretic Factor/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Heart Atria/metabolism , Male , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/metabolism , Rats, Wistar , Streptozocin , Up-Regulation
6.
Oxid Med Cell Longev ; 2017: 5343972, 2017.
Article in English | MEDLINE | ID: mdl-28781721

ABSTRACT

Diabetic cardiomyopathy (DCM) is characterized by cardiac remodeling and impaired diastolic function that may lead to heart failure. The aim of this study was to evaluate oxidative stress, inflammatory cells, and fibrosis in both subendocardial (SEN) and interstitial (INT) areas of the myocardium. Male Wistar rats were allocated to 2 groups of 9 animals, a control (CT) group and streptozotocin-induced diabetes (DM). After 8 weeks, echocardiography morphometry, protein expression, and confocal microscopy in SEN and INT areas of the left ventricle (LV) were performed. The echocardiographic analysis showed that diabetes induction leads to cardiac dilation, hypertrophy, and LV diastolic dysfunction. As compared to CT, the induction of diabetes increased inflammatory cells and fibrosis in both SEN and INT areas of DM myocardium and increased ROS generation only in SEN. Comparing the SEN and INT areas in the DM group, inflammatory cells and fibrosis in SEN were greater than in INT. In conclusion, diabetic myocardium SEN area, wherein oxidative stress was more pronounced, is more susceptible to cardiac dysfunction than INT area. This finding can be important for the understanding of the heart remodeling process occurring in DCM and perhaps to engender targeted therapies to attenuate or revert DCM-related diastolic dysfunction.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Heart/physiology , Myocardium/metabolism , Oxidative Stress/physiology , Animals , Blotting, Western , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/immunology , Echocardiography , Male , Myocardium/immunology , Myocardium/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
7.
Oxid Med Cell Longev ; 2017: 7921363, 2017.
Article in English | MEDLINE | ID: mdl-28698769

ABSTRACT

Diabetic cardiomyopathy refers to the manifestations in the heart as a result of altered glucose homeostasis, reflected as fibrosis, cellular hypertrophy, increased oxidative stress, and apoptosis, leading to ventricular dysfunction. Since physical exercise has been indicated as cardioprotective, we tested the hypothesis that high-intensity exercise training could reverse the cardiac maladaptations produced by diabetes. For this, diabetes was induced in rats by a single dose of alloxan. Diabetic rats were randomly assigned to a sedentary group or submitted to a program of exercise on a treadmill for 4 weeks at 80% of maximal performance. Another group of normoglycemic rats was used as control. Diabetic rat hearts presented cardiomyocyte hypertrophy and interstitial fibrosis. Chronic exercise reduced both parameters but increased apoptosis. Diabetes increased the myocardial levels of the mRNA and proteins of NADPH oxidases NOX2 and NOX4. These altered levels were not reduced by exercise. Diabetes also increased the level of uncoupled endothelial nitric oxide synthase (eNOS) that was not reversed by exercise. Finally, diabetic rats showed a lower degree of phosphorylated phospholamban and reduced levels of SERCA2 that were not restored by high-intensity exercise. These results suggest that high-intensity chronic exercise was able to reverse remodeling in the diabetic heart but was unable to restore the nitroso-redox imbalance imposed by diabetes.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Physical Conditioning, Animal/physiology , Animals , Apoptosis/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/physiopathology , Male , Myocardium/metabolism , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
8.
Cardiovasc Diabetol ; 15(1): 126, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27585437

ABSTRACT

PURPOSE: Although increased oxidative stress is a major component of diabetic hypertensive cardiomyopathy, research into the effects of antioxidants on cardiac remodeling remains scarce. The actions of antioxidant apocynin include inhibiting reactive oxygen species (ROS) generation by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and ROS scavenging. We evaluated the effects of apocynin on cardiac remodeling in spontaneously hypertensive rats (SHR) with diabetes mellitus (DM). METHODS: Male SHR were divided into four groups: control (SHR, n = 16); SHR treated with apocynin (SHR-APO; 16 mg/kg/day, added to drinking water; n = 16); diabetic SHR (SHR-DM, n = 13); and SHR-DM treated with apocynin (SHR-DM-APO, n = 14), for eight weeks. DM was induced by streptozotocin (40 mg/kg, single dose). Statistical analyzes: ANOVA and Tukey or Mann-Whitney. RESULTS: Echocardiogram in diabetic groups showed higher left ventricular and left atrium diameters indexed for body weight, and higher isovolumetric relaxation time than normoglycemic rats; systolic function did not differ between groups. Isolated papillary muscle showed impaired contractile and relaxation function in diabetic groups. Developed tension was lower in SHR-APO than SHR. Myocardial hydroxyproline concentration was higher in SHR-DM than SHR, interstitial collagen fraction was higher in SHR-DM-APO than SHR-APO, and type III collagen protein expression was lower in SHR-DM and SHR-DM-APO than their controls. Type I collagen and lysyl oxidase expression did not differ between groups. Apocynin did not change collagen tissue. Myocardial lipid hydroperoxide concentration was higher in SHR-DM than SHR and SHR-DM-APO. Glutathione peroxidase activity was lower and catalase higher in SHR-DM than SHR. Apocynin attenuated antioxidant enzyme activity changes in SHR-DM-APO. Advanced glycation end-products and NADPH oxidase activity did not differ between groups. CONCLUSION: Apocynin reduces oxidative stress independently of NADPH oxidase activity and does not change ventricular or myocardial function in spontaneously hypertensive rats with diabetes mellitus. The apocynin-induced myocardial functional impairment in SHR shows that apocynin actions need to be clarified during sustained chronic pressure overload.


Subject(s)
Acetophenones/pharmacology , Antioxidants/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/prevention & control , Hypertension/drug therapy , Hypertrophy, Left Ventricular/prevention & control , Myocardium/metabolism , Oxidative Stress/drug effects , Ventricular Remodeling/drug effects , Animals , Catalase/metabolism , Collagen Type III/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Glutathione Peroxidase/metabolism , Hypertension/complications , Hypertension/metabolism , Hypertension/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Lipid Peroxidation/drug effects , Lipid Peroxides/metabolism , Male , Myocardial Contraction/drug effects , Myocardium/pathology , Rats, Inbred SHR , Streptozocin , Ventricular Function, Left/drug effects
9.
J Diabetes Res ; 2016: 6143129, 2016.
Article in English | MEDLINE | ID: mdl-26770988

ABSTRACT

The prevalence of cardiac diabetic diseases has been increased around the world, being the most common cause of death and disability among diabetic patients. In particular, diabetic cardiomyopathy is characterized with a diastolic dysfunction and cardiac remodelling without signs of hypertension and coronary artery diseases. In an early stage, it is an asymptomatic disease; however, clinical studies demonstrate that diabetic myocardia are more vulnerable to injury derived by acute myocardial infarct and are the worst prognosis for rehabilitation. Currently, biochemical and imaging diagnostic methods are unable to detect subclinical manifestation of the disease (prior to diastolic dysfunction). In this review, we elaborately discuss the current scientific evidences to propose circulating microRNAs as promising biomarkers for early detection of diabetic cardiomyopathy and, then, to identify patients at high risk of diabetic cardiomyopathy development. Moreover, here we summarise the research strategies to identify miRNAs as potential biomarkers, present limitations, challenges, and future perspectives.


Subject(s)
Diabetic Cardiomyopathies/diagnosis , MicroRNAs/metabolism , Biomarkers/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Early Diagnosis , Humans , MicroRNAs/genetics
10.
Biochim Biophys Acta ; 1853(5): 1113-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25686534

ABSTRACT

Diabetic cardiomyopathy (DCM) is a common consequence of longstanding type 2 diabetes mellitus (T2DM) and encompasses structural, morphological, functional, and metabolic abnormalities in the heart. Myocardial energy metabolism depends on mitochondria, which must generate sufficient ATP to meet the high energy demands of the myocardium. Dysfunctional mitochondria are involved in the pathophysiology of diabetic heart disease. A large body of evidence implicates myocardial insulin resistance in the pathogenesis of DCM. Recent studies show that insulin signaling influences myocardial energy metabolism by impacting cardiomyocyte mitochondrial dynamics and function under physiological conditions. However, comprehensive understanding of molecular mechanisms linking insulin signaling and changes in the architecture of the mitochondrial network in diabetic cardiomyopathy is lacking. This review summarizes our current understanding of how defective insulin signaling impacts cardiac function in diabetic cardiomyopathy and discusses the potential role of mitochondrial dynamics.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Insulin/metabolism , Mitochondrial Dynamics , Signal Transduction , Animals , Diabetic Cardiomyopathies/pathology , Humans , Models, Biological , Myocardium/metabolism , Myocardium/pathology
11.
Acta Physiol (Oxf) ; 211(3): 491-500, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24837225

ABSTRACT

Diabetes mellitus is a serious health problem that can lead to several pathological complications in numerous organs and tissues. The most important and most prevalent organs affected by this disease are the heart and the kidneys, and these complications are the major causes of death in patients with diabetes. MicroRNAs (miRNAs), short non-coding RNAs, have been found to be functionally important in the regulation of several pathological processes, and they are emerging as an important therapeutic tool to avoid the complications of diabetes mellitus. This review summarizes the knowledge on the effects of miRNAs in diabetes. The use of miRNAs in diabetes from a clinical perspective is also discussed, focusing on their potential role to repair cardiovascular and renal complications.


Subject(s)
Diabetic Cardiomyopathies/therapy , Diabetic Retinopathy/therapy , Genetic Therapy/methods , Kidney/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Animals , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Gene Expression Regulation , Genetic Markers , Humans , Oligonucleotides, Antisense/therapeutic use , RNA Interference
12.
Am J Physiol Cell Physiol ; 306(9): C794-804, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24553187

ABSTRACT

Excess enzyme-mediated protein O-GlcNAcylation is known to occur with diabetes mellitus. A characteristic of diabetic cardiomyopathy is the development of myocardial fibrosis. The role that enhanced protein O-GlcNAcylation plays in modulating the phenotype of cardiac fibroblasts (CF) is unknown. To address this issue, rat CF were cultured in normal glucose (NG; 5 mM glucose) or high-glucose (HG; 25 mM) media for 48 h. Results demonstrate that CF cultured in HG have higher levels (~50%) of overall protein O-GlcNAcylation vs. NG cells. Key regulators of collagen synthesis such as transforming-growth factor-ß1 (TGF-ß1), SMADs 2/3, and SMAD 7 protein levels, including those of arginase I and II, were altered, leading to increases in collagen levels. The nuclear transcription factor Sp1 and arginase II evidence excess O-GlcNAcylation in HG cells. Expression in CF of an adenovirus coding for the enzyme N-acetylglucosaminidase, which removes O-GlcNAc moieties from proteins, decreased Sp1 and arginase II O-GlcNAcylation and restored HG-induced perturbations in CF back to NG levels. These findings may have important pathophysiological implications for the development of diabetes-induced cardiac fibrosis.


Subject(s)
Collagen/biosynthesis , Diabetic Cardiomyopathies/metabolism , Fibroblasts/metabolism , Glucose/metabolism , Myocardium/metabolism , Protein Processing, Post-Translational , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Animals , Arginase/metabolism , Cells, Cultured , Diabetic Cardiomyopathies/pathology , Fibroblasts/pathology , Glycosylation , Male , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins/metabolism , Sp1 Transcription Factor/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL